1
|
Poletto Bonetto JH, Deprez A, Wolf D, Oliveira Fernandes R, Casali K, Sonea A, Flahault A, Siqueira Flores M, He Y, Belló-Klein A, Ravizzoni Dartora D, Nuyt AM. Impact of neonatal hyperoxia on adult cardiac autonomic function in rats: Role of angiotensin II type 1 receptor activation. Eur J Pharmacol 2024; 984:177026. [PMID: 39396751 DOI: 10.1016/j.ejphar.2024.177026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 09/27/2024] [Accepted: 10/01/2024] [Indexed: 10/15/2024]
Abstract
Individuals born preterm present altered cardiac autonomic function, a risk factor to heart diseases. Neonatal renin-angiotensin-system activation contributes to adult cardiomyopathy in rats exposed to neonatal hyperoxia, a well-established model of preterm birth-related conditions. Central angiotensin II receptor activation is a key modulator of the autonomic drive to the heart. Whether neonatal hyperoxia leads to alteration of the cardiac autonomic function through activation of the angiotensin II receptor type 1 (AT1) is unknown and was examined in the present study. Sprague-Dawley pups were exposed to hyperoxia or room air from postnatal days 3-10. AT1 antagonist losartan or water was given orally postnatal days 8-10. Blood pressure, autonomic function, left ventricular sympathetic innervation, β-adrenergic-receptors expression, and AT1 expression in the solitary-tract-nucleus were examined in adult rats. Neonatal hyperoxia led to loss of day-night blood pressure variation, decreased heart rate variability, increased sympathovagal balance, increased AT1 expression in the solitary-tract, decreased left ventricle sympathetic innervation, and increased β1-adrenergic-receptor protein expression. Losartan prevented the autonomic changes and AT1 expression in the solitary-tract but did not impact the loss of circadian blood pressure variation nor the changes in sympathetic innervation and in β1-adrenergic-receptor expression. In conclusion, neonatal hyperoxia leads to both central autonomic and cardiac sympathetic changes, partly programmed by neonatal activation of the renin-angiotensin system.
Collapse
Affiliation(s)
- Jéssica Hellen Poletto Bonetto
- Sainte-Justine University Hospital and Research Center, Université de Montréal, Québec, Canada; Laboratory of Cardiovascular Physiology, Federal University of Rio Grande do Sul (UFRGS), Rio Grande do Sul, Brazil
| | - Alyson Deprez
- Sainte-Justine University Hospital and Research Center, Université de Montréal, Québec, Canada
| | - Daniele Wolf
- Sainte-Justine University Hospital and Research Center, Université de Montréal, Québec, Canada
| | | | - Karina Casali
- Institute of Science and Technology, Federal University of São Paulo (USP), São Paulo, Brazil
| | - Aurélie Sonea
- Sainte-Justine University Hospital and Research Center, Université de Montréal, Québec, Canada
| | - Adrien Flahault
- Sainte-Justine University Hospital and Research Center, Université de Montréal, Québec, Canada
| | - Marina Siqueira Flores
- Laboratory of Cardiovascular Physiology, Federal University of Rio Grande do Sul (UFRGS), Rio Grande do Sul, Brazil
| | - Ying He
- Sainte-Justine University Hospital and Research Center, Université de Montréal, Québec, Canada
| | - Adriane Belló-Klein
- Laboratory of Cardiovascular Physiology, Federal University of Rio Grande do Sul (UFRGS), Rio Grande do Sul, Brazil
| | | | - Anne Monique Nuyt
- Sainte-Justine University Hospital and Research Center, Université de Montréal, Québec, Canada.
| |
Collapse
|
2
|
Zhong J, Doughty R, Thymann T, Sangild PT, Nguyen DN, Muk T. Insulin-like growth factor-1 effects on kidney development in preterm piglets. Pediatr Res 2024; 96:1655-1665. [PMID: 38762663 PMCID: PMC11772250 DOI: 10.1038/s41390-024-03222-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 04/03/2024] [Accepted: 04/04/2024] [Indexed: 05/20/2024]
Abstract
BACKGROUND Preterm birth disrupts fetal kidney development, potentially leading to postnatal acute kidney injury. Preterm infants are deficient in insulin-like growth factor 1 (IGF-1), a growth factor that stimulates organ development. By utilizing a preterm pig model, this study investigated whether IGF-1 supplementation enhances preterm kidney maturation. METHODS Cesarean-delivered preterm pigs were treated systemically IGF-1 or vehicle control for 5, 9 or 19 days after birth. Blood, urine, and kidney tissue were collected for biochemical, histological and gene expression analyses. Age-matched term-born pigs were sacrificed at similar postnatal ages and served as the reference group. RESULTS Compared with term pigs, preterm pigs exhibited impaired kidney maturation, as indicated by analyses of renal morphology, histopathology, and inflammatory and injury markers. Supplementation with IGF-1 reduced signs of kidney immaturity, particularly in the first week of life, as indicated by improved morphology, upregulated expression of key developmental genes, reduced severity and incidence of microscopic lesions, and decreased levels of inflammatory and injury markers. No association was seen between the symptoms of necrotizing enterocolitis and kidney defects. CONCLUSION Preterm birth in pigs impairs kidney maturation and exogenous IGF-1 treatment partially reverses this impairment. Early IGF-1 supplementation could support the development of preterm kidneys. IMPACT Preterm birth may disrupt kidney development in newborns, potentially leading to morphological changes, injury, and inflammation. Preterm pigs have previously been used as models for preterm infants, but not for kidney development. IGF-1 supplementation promotes kidney maturation and alleviates renal impairments in the first week of life in preterm pigs. IGF-1 may hold potential as a supportive therapy for preterm infants sensitive to acute kidney injury.
Collapse
Affiliation(s)
- Jingren Zhong
- Section for Comparative Paediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Richard Doughty
- Department of Pathology, Akershus University Hospital, Lørenskog, Norway
| | - Thomas Thymann
- Section for Comparative Paediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Per Torp Sangild
- Section for Comparative Paediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
- Department of Pediatrics, Odense University Hospital, Odense, Denmark
- Department of Neonatology, Rigshospitalet, Copenhagen, Denmark
| | - Duc Ninh Nguyen
- Section for Comparative Paediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Tik Muk
- Section for Comparative Paediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark.
| |
Collapse
|
3
|
Zhou M, Wang J, Cao R, Zhang F, Luo X, Liao Y, Chen W, Ding H, Tan X, Qiao Z, Yang K. Hypoxia-Induced Differences in the Expression of Pyruvate Dehydrogenase Kinase 1-Related Factors in the Renal Tissues and Renal Interstitial Fibroblast-like Cells of Yak (Bos Grunniens). Animals (Basel) 2024; 14:3110. [PMID: 39518833 PMCID: PMC11545261 DOI: 10.3390/ani14213110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 10/01/2024] [Accepted: 10/09/2024] [Indexed: 11/16/2024] Open
Abstract
Hypoxia is one of the factors severely affect renal function, and, in severe cases, it can lead to renal fibrosis. Although much progress has been made in identifying the molecular mediators of fibrosis, the mechanisms that govern renal fibrosis remain unclear, and there have been no effective therapeutic anti-fibrotic strategies to date. Mammals exposed to low oxygen in the plateau environment for a long time are prone to high-altitude disease, while yaks have been living in the plateau for generations do not develop kidney fibrosis caused by low oxygen. It has been suggested that metabolic reprogramming occurs in renal fibrosis and that pyruvate dehydrogenase kinase 1 (PDK1) plays a crucial role in metabolic reprogramming as an important node between glycolysis and the tricarboxylic acid cycle. The aim of this study was to investigate the effects of hypoxia on the renal tissues and renal interstitial fibroblasts of yaks. We found that, at the tissue level, HIF-1α, PDK1, TGF-β1, Smad2, Smad3, and α-SMA were mainly distributed and expressed in tubular epithelial cells but were barely present in the renal mesenchymal fibroblasts of healthy cattle and yak kidneys. Anoptical density analysis showed that in healthy cattle kidneys, TGF-β1, Smad2, and Smad3 expression was significantly higher than in yak kidneys (p < 0.05), and HIF-1α and PDK1 expression was significantly lower than in yak kidneys (p < 0.05). The results at the protein and gene levels showed the same trend. At the cellular level, prolonged hypoxia significantly elevated PDK1 expression in the renal mesangial fibroblasts of cattle and yak kidneys compared with normoxia (p < 0.05) and was proportional to the degree of cellular fibrosis. However, PDK1 expression remained stable in yaks compared with renal interstitial fibroblast-like cells in cattle during the same hypoxic time period. At the same time, prolonged hypoxia also promoted changes in cellular phenotype, promoting the proliferation, activation, glucose consumption, lactate production, and anti-apoptosis in the both of cattle and yaks renal interstitial fibroblasts The differences in kidney structure and expression of PDK1 and HIF-1α in kidney tissue and renal interstitial fibroblasts induced by different oxygen concentrations suggest that there may be a regulatory relationship between yak kidney adaptation and hypoxic environment at high altitude. This provides strong support for the elucidation of the regulatory relationship between PDK1 and HIF-1α, as well as a new direction for the treatment or delay of hypoxic renal fibrosis; additionally, these findings provide a basis for further analysis of the molecular mechanism of hypoxia adaptation-related factors and the adaptation of yaks to plateau hypoxia.
Collapse
Affiliation(s)
- Manlin Zhou
- Engineering Research Center of Key Technology and Industrialization of Cell-Based Vaccine, Ministry of Education, Northwest Minzu University, Lanzhou 730030, China; (M.Z.); (F.Z.); (Z.Q.)
- Gansu Tech Innovation Center of Animal Cell, Biomedical Research Center, Northwest Minzu University, Lanzhou 730030, China
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Affairs Commission, Biomedical Research Center, Northwest Minzu University, Lanzhou 730030, China
- College of Life Science and Engineering, Northwest Minzu University, Lanzhou 730030, China; (J.W.); (R.C.); (X.L.); (Y.L.); (H.D.); (X.T.)
| | - Jun Wang
- College of Life Science and Engineering, Northwest Minzu University, Lanzhou 730030, China; (J.W.); (R.C.); (X.L.); (Y.L.); (H.D.); (X.T.)
| | - Ruirui Cao
- College of Life Science and Engineering, Northwest Minzu University, Lanzhou 730030, China; (J.W.); (R.C.); (X.L.); (Y.L.); (H.D.); (X.T.)
| | - Fan Zhang
- Engineering Research Center of Key Technology and Industrialization of Cell-Based Vaccine, Ministry of Education, Northwest Minzu University, Lanzhou 730030, China; (M.Z.); (F.Z.); (Z.Q.)
- Gansu Tech Innovation Center of Animal Cell, Biomedical Research Center, Northwest Minzu University, Lanzhou 730030, China
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Affairs Commission, Biomedical Research Center, Northwest Minzu University, Lanzhou 730030, China
- College of Life Science and Engineering, Northwest Minzu University, Lanzhou 730030, China; (J.W.); (R.C.); (X.L.); (Y.L.); (H.D.); (X.T.)
| | - Xuehui Luo
- College of Life Science and Engineering, Northwest Minzu University, Lanzhou 730030, China; (J.W.); (R.C.); (X.L.); (Y.L.); (H.D.); (X.T.)
| | - Yiyuan Liao
- College of Life Science and Engineering, Northwest Minzu University, Lanzhou 730030, China; (J.W.); (R.C.); (X.L.); (Y.L.); (H.D.); (X.T.)
| | - Weiji Chen
- College of Life Science and Engineering, Northwest Minzu University, Lanzhou 730030, China; (J.W.); (R.C.); (X.L.); (Y.L.); (H.D.); (X.T.)
| | - Haie Ding
- College of Life Science and Engineering, Northwest Minzu University, Lanzhou 730030, China; (J.W.); (R.C.); (X.L.); (Y.L.); (H.D.); (X.T.)
| | - Xiao Tan
- College of Life Science and Engineering, Northwest Minzu University, Lanzhou 730030, China; (J.W.); (R.C.); (X.L.); (Y.L.); (H.D.); (X.T.)
| | - Zilin Qiao
- Engineering Research Center of Key Technology and Industrialization of Cell-Based Vaccine, Ministry of Education, Northwest Minzu University, Lanzhou 730030, China; (M.Z.); (F.Z.); (Z.Q.)
- Gansu Tech Innovation Center of Animal Cell, Biomedical Research Center, Northwest Minzu University, Lanzhou 730030, China
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Affairs Commission, Biomedical Research Center, Northwest Minzu University, Lanzhou 730030, China
| | - Kun Yang
- Engineering Research Center of Key Technology and Industrialization of Cell-Based Vaccine, Ministry of Education, Northwest Minzu University, Lanzhou 730030, China; (M.Z.); (F.Z.); (Z.Q.)
- Gansu Tech Innovation Center of Animal Cell, Biomedical Research Center, Northwest Minzu University, Lanzhou 730030, China
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Affairs Commission, Biomedical Research Center, Northwest Minzu University, Lanzhou 730030, China
- College of Life Science and Engineering, Northwest Minzu University, Lanzhou 730030, China; (J.W.); (R.C.); (X.L.); (Y.L.); (H.D.); (X.T.)
| |
Collapse
|
4
|
DeFreitas MJ, Shelton EL, Schmidt AF, Ballengee S, Tian R, Chen P, Sharma M, Levine A, Katz ED, Rojas C, Abitbol CL, Hunter J, Kulandavelu S, Wu S, Young KC, Benny M. Neonatal hyperoxia exposure leads to developmental programming of cardiovascular and renal disease in adult rats. Sci Rep 2024; 14:16742. [PMID: 39033222 PMCID: PMC11271593 DOI: 10.1038/s41598-024-65844-1] [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: 01/17/2024] [Accepted: 06/25/2024] [Indexed: 07/23/2024] Open
Abstract
Premature infants are often exposed to hyperoxia. However, there is limited data regarding the mechanistic underpinnings linking neonatal hyperoxia exposure and its contribution to cardio-renal dysfunction in adults born preterm. Our objective was to determine whether neonatal hyperoxia induces systemic vascular stiffness and cardio-renal dysfunction in adulthood. Newborn rats were randomly assigned to room air (RA) or hyperoxia (85% O2) from postnatal day 1 to 14, then recovered in RA until 1 year of life. Arterial stiffness, cardio-renal histomorphometry, and fibrosis in the aorta, heart, and kidney were assessed. RNA-sequencing (RNA-seq) of the aorta and kidney was also done. Adult rats exposed to neonatal hyperoxia had increased aortic and mesenteric artery stiffness as demonstrated by wire and pressure myography. They also had cardiomyocyte hypertrophy, glomerulomegaly, and tubular injury. Hyperoxia exposure altered the transcriptome profile associated with fibrosis and matrix remodeling in the aorta and kidney. There was also increased TGF-β1 levels and fibrosis in the aorta, left ventricle, and kidney. In conclusion, neonatal hyperoxia exposure was associated with systemic vascular and cardio-renal alterations in 1-year-old rats. Further studies to determine how targeted therapies could reprogram cardio-renal injury after neonatal hyperoxia exposure are indicated.
Collapse
Affiliation(s)
- Marissa J DeFreitas
- Department of Pediatrics/Division of Nephrology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Elaine L Shelton
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Augusto F Schmidt
- Department of Pediatrics/Division of Neonatology, Batchelor Children's Research Institute, Miller School of Medicine, University of Miami, P.O. Box 016960 (R-131), Miami, FL, 33101, USA
| | - Sydne Ballengee
- Department of Pediatrics/Division of Neonatology, Batchelor Children's Research Institute, Miller School of Medicine, University of Miami, P.O. Box 016960 (R-131), Miami, FL, 33101, USA
| | - Runxia Tian
- Department of Pediatrics/Division of Neonatology, Batchelor Children's Research Institute, Miller School of Medicine, University of Miami, P.O. Box 016960 (R-131), Miami, FL, 33101, USA
| | - PingPing Chen
- Department of Pediatrics/Division of Neonatology, Batchelor Children's Research Institute, Miller School of Medicine, University of Miami, P.O. Box 016960 (R-131), Miami, FL, 33101, USA
| | - Mayank Sharma
- Department of Pediatrics/Division of Neonatology, Batchelor Children's Research Institute, Miller School of Medicine, University of Miami, P.O. Box 016960 (R-131), Miami, FL, 33101, USA
| | - Amanda Levine
- Department of Pediatrics/Division of Neonatology, Batchelor Children's Research Institute, Miller School of Medicine, University of Miami, P.O. Box 016960 (R-131), Miami, FL, 33101, USA
| | - Emily Davidovic Katz
- Department of Pediatrics/Division of Neonatology, Batchelor Children's Research Institute, Miller School of Medicine, University of Miami, P.O. Box 016960 (R-131), Miami, FL, 33101, USA
| | - Claudia Rojas
- Department of Pathology, Memorial Healthcare Systems, Hollywood, FL, USA
| | - Carolyn L Abitbol
- Department of Pediatrics/Division of Nephrology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Juanita Hunter
- Department of Pediatrics/Division of Cardiology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Shathiyah Kulandavelu
- Department of Pediatrics/Division of Nephrology, University of Miami Miller School of Medicine, Miami, FL, USA
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Shu Wu
- Department of Pediatrics/Division of Neonatology, Batchelor Children's Research Institute, Miller School of Medicine, University of Miami, P.O. Box 016960 (R-131), Miami, FL, 33101, USA
| | - Karen C Young
- Department of Pediatrics/Division of Neonatology, Batchelor Children's Research Institute, Miller School of Medicine, University of Miami, P.O. Box 016960 (R-131), Miami, FL, 33101, USA
| | - Merline Benny
- Department of Pediatrics/Division of Neonatology, Batchelor Children's Research Institute, Miller School of Medicine, University of Miami, P.O. Box 016960 (R-131), Miami, FL, 33101, USA.
| |
Collapse
|
5
|
Shen Y, Yuan Y, Dong W. The Mechanism of Hyperoxia-Induced Neonatal Renal Injury and the Possible Protective Effect of Resveratrol. Am J Perinatol 2024; 41:1126-1133. [PMID: 35381611 DOI: 10.1055/a-1817-5357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
With recent advances in neonatal intensive care, preterm infants are surviving into adulthood. Nonetheless, epidemiological data on the health status of these preterm infants have begun to reveal a worrying theme; prematurity and the supplemental oxygen therapy these infants receive after birth appear to be risk factors for kidney disease in adulthood, affecting their quality of life. As the incidence of chronic kidney disease and the survival time of preterm infants both increase, the management of the hyperoxia-induced renal disease is becoming increasingly relevant to neonatologists. The mechanism of this increased risk is currently unknown, but prematurity itself and hyperoxia exposure after birth may predispose to disease by altering the normal trajectory of kidney maturation. This article reviews altered renal reactivity due to hyperoxia, the possible mechanisms of renal injury due to hyperoxia, and the role of resveratrol in renal injury. KEY POINTS: · Premature infants commonly receive supplementary oxygen.. · Hyperoxia can cause kidney damage via signal pathways.. · We should reduce the occurrence of late sequelae..
Collapse
Affiliation(s)
- Yunchuan Shen
- Division of Neonatology, Department of Pediatrics, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Yuan Yuan
- Division of Neonatology, Department of Pediatrics, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Wenbin Dong
- Division of Neonatology, Department of Pediatrics, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
6
|
Iacobelli S, Lapillonne A, Boubred F. Early postnatal nutrition and renal consequences in preterm infants. Pediatr Res 2024:10.1038/s41390-024-03080-z. [PMID: 38374220 DOI: 10.1038/s41390-024-03080-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 01/06/2024] [Accepted: 01/20/2024] [Indexed: 02/21/2024]
Abstract
Perinatal nutritional factors may lead to decreased nephron endowment, decreased kidney function, and long-term development of chronic kidney disease and non-communicable diseases. At the same time, optimal postnatal nutrition and catch-up growth are associated with better neurodevelopmental outcomes in preterm infants. Therefore, nutritional management of preterm infants is a major challenge for neonatologists. In this context, the Section of Nutrition, Gastroenterology and Metabolism reviewed the current knowledge on nutritional issues related to kidney function. This narrative review discusses the clinical impact of early postnatal nutrition on long-term kidney function. In preterm infants, data are largely lacking to determine the extent to which early nutrition contributes to nephrogenesis and nephron endowment. However, some nutritional principles may help clinicians better protect the developing kidney in preterm infants. IMPACT: Clinical data show that preterm infants are an emerging population at high risk for chronic kidney disease. Both undernutrition and overnutrition can alter long-term kidney function. In preterm infants, data are largely lacking to determine the extent to which early postnatal nutrition contributes to nephrogenesis, nephron endowment and increased risk for chronic kidney disease. Some nutritional principles may help clinicians better protect the developing kidney in preterm infants: avoiding extrauterine growth restriction; providing adequate protein and caloric intakes; limiting exposure to high and prolonged hyperglycaemia; avoiding micronutrient deficiencies and maintaining acid-base and electrolyte balance.
Collapse
Affiliation(s)
- Silvia Iacobelli
- Réanimation Néonatale et Pédiatrique, CHU La Réunion, Saint-Pierre, France.
- Centre d'Études Périnatales de l'Océan Indien (UR7388), Université de La Réunion, de La Réunion, France.
| | - Alexandre Lapillonne
- Service de Médecine néonatale, CHU La Conception, APHM, Marseille, France
- Aix-Marseille Université, C2VN, INRAe, INSERM, Marseille, France
| | - Farid Boubred
- Department of Neonatology, APHP, Necker-Enfants Malades University Hospital, EHU 7328 Paris Cite University Paris, Paris, France
- CNRC Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
7
|
Carpenter J, Yarlagadda S, VandenHeuvel KA, Ding L, Schuh MP. Human Nephrogenesis can Persist Beyond 40 Postnatal Days in Preterm Infants. Kidney Int Rep 2024; 9:436-450. [PMID: 38344733 PMCID: PMC10851065 DOI: 10.1016/j.ekir.2023.10.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 10/13/2023] [Accepted: 10/30/2023] [Indexed: 02/15/2024] Open
Abstract
Introduction Human nephrogenesis is typically completed by 36 weeks gestation; however, it is impacted by preterm birth. Early studies suggested that nephrogenesis persisted for ≤40 postnatal days in preterm infants. However, the postmenstrual age (PMA) of the preterm infants who survived >40 days was uncertain. In this study, we sought to reexamine postnatal kidney development in preterm infants surviving >40 days. Methods Human kidney samples were obtained from an institutional biobank. Samples were considered controls if survival was ≤4 days after birth with PMA of 30 to ≤36 weeks. Kidneys from preterm neonates with postnatal survival >40 days and PMA of 30 to ≤36 weeks were compared to controls. We counted glomerular generations, measured nephrogenic zone widths (NZW), and performed immunofluorescence (IF) with SIX1 and RET. We compared kidney weights and quantified the cross-sectional area of proximal (lotus tetragonolobus lectin [LTL], SL22A2), distal (SLC12A3, KCNJ10), and glomerular (nephrin) markers using IF. Results Seven preterm infants surviving >40 days and 8 controls were analyzed. Four of 7 preterm infants had histologic and molecular evidence of nephrogenesis. Cessation of nephrogenesis in preterm infants occurred 2 weeks earlier than PMA-matched controls with attenuated expression of both SIX1 and RET. We found increased kidney weight-to-body weight ratio, increased distal tubular cross-sectional staining in the superficial nephrons, and distal tubular hypertrophy and hyperplasia in the preterm infant kidneys. Conclusion Our study supports that nephrogenesis in preterm infants persists longer than previously thought with evidence of early nephron stress, placing importance on the neonatal environment.
Collapse
Affiliation(s)
- James Carpenter
- Division of Nephrology and Hypertension, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Sunitha Yarlagadda
- Division of Nephrology and Hypertension, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Katherine A. VandenHeuvel
- Division of Pathology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Lili Ding
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Biostatistics and Epidemiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Meredith P. Schuh
- Division of Nephrology and Hypertension, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| |
Collapse
|
8
|
Cai Y, Luo Y, Dai N, Yang Y, He Y, Chen H, Zhao M, Fu X, Chen T, Xing Z. Functional metagenomic and metabolomics analysis of gut dysbiosis induced by hyperoxia. Front Microbiol 2023; 14:1197970. [PMID: 37840730 PMCID: PMC10569423 DOI: 10.3389/fmicb.2023.1197970] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 09/11/2023] [Indexed: 10/17/2023] Open
Abstract
Background Inhaled oxygen is the first-line therapeutic approach for maintaining tissue oxygenation in critically ill patients, but usually exposes patients to damaging hyperoxia. Hyperoxia adversely increases the oxygen tension in the gut lumen which harbors the trillions of microorganisms playing an important role in host metabolism and immunity. Nevertheless, the effects of hyperoxia on gut microbiome and metabolome remain unclear, and metagenomic and metabolomics analysis were performed in this mouse study. Methods C57BL/6 mice were randomly divided into a control (CON) group exposed to room air with fractional inspired oxygen (FiO2) of 21% and a hyperoxia (OXY) group exposed to FiO2 of 80% for 7 days, respectively. Fecal pellets were collected on day 7 and subjected to metagenomic sequencing. Another experiment with the same design was performed to explore the impact of hyperoxia on gut and serum metabolome. Fecal pellets and blood were collected and high-performance liquid chromatography with mass spectrometric analysis was carried out. Results At the phylum level, hyperoxia increased the ratio of Firmicutes/Bacteroidetes (p = 0.049). At the species level, hyperoxia reduced the abundance of Muribaculaceae bacterium Isolate-037 (p = 0.007), Isolate-114 (p = 0.010), and Isolate-043 (p = 0.011) etc. Linear discriminant analysis effect size (LEfSe) revealed that Muribaculaceae and Muribaculaceae bacterium Isolate-037, both belonging to Bacteroidetes, were the marker microbes of the CON group, while Firmicutes was the marker microbes of the OXY group. Metagenomic analysis using Kyoto Encyclopedia of Genes and Genomes (KEGG) and Carbohydrate-Active enZYmes (CAZy) revealed that hyperoxia provoked disturbances in carbohydrate and lipid metabolism. Fecal metabolomics analysis showed hyperoxia reduced 11-dehydro Thromboxane B2-d4 biosynthesis (p = 1.10 × 10-11). Hyperoxia blunted fecal linoleic acid metabolism (p = 0.008) and alpha-linolenic acid metabolism (p = 0.014). We showed that 1-docosanoyl-glycer-3-phosphate (p = 1.58 × 10-10) was the most significant differential serum metabolite inhibited by hyperoxia. In addition, hyperoxia suppressed serum hypoxia-inducible factor-1 (HIF-1, p = 0.007) and glucagon signaling pathways (p = 0.007). Conclusion Hyperoxia leads to gut dysbiosis by eliminating beneficial and oxygen strictly intolerant Muribaculaceae with genomic dysfunction of carbohydrate and lipid metabolism. In addition, hyperoxia suppresses unsaturated fatty acid metabolism in the gut and inhibits the HIF-1 and glucagon signaling pathways in the serum.
Collapse
Affiliation(s)
- Yulan Cai
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Department of Endocrinology and Metabolism, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Kweichow Moutai Hospital, Renhuai, China
| | - Yanhong Luo
- The First Clinical College, Zunyi Medical University, Zunyi, China
| | - Ninan Dai
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yan Yang
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Department of Endocrinology and Metabolism, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Ying He
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Huajun Chen
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Manlu Zhao
- The First Clinical College, Zunyi Medical University, Zunyi, China
| | - Xiaoyun Fu
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Tao Chen
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Zhouxiong Xing
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| |
Collapse
|
9
|
Sutherland MR, Black MJ. The impact of intrauterine growth restriction and prematurity on nephron endowment. Nat Rev Nephrol 2023; 19:218-228. [PMID: 36646887 DOI: 10.1038/s41581-022-00668-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/04/2022] [Indexed: 01/18/2023]
Abstract
In humans born at term, maximal nephron number is reached by the time nephrogenesis is completed - at approximately 36 weeks' gestation. The number of nephrons does not increase further and subsequently remains stable until loss occurs through ageing or disease. Nephron endowment is key to the functional capacity of the kidney and its resilience to disease; hence, any processes that impair kidney development in the developing fetus can have lifelong adverse consequences for renal health and, consequently, for quality and length of life. The timing of nephrogenesis underlies the vulnerability of developing human kidneys to adverse early life exposures. Indeed, exposure of the developing fetus to a suboptimal intrauterine environment during gestation - resulting in intrauterine growth restriction (IUGR) - and/or preterm birth can impede kidney development and lead to reduced nephron endowment. Furthermore, emerging research suggests that IUGR and/or preterm birth is associated with an elevated risk of chronic kidney disease in later life. The available data highlight the important role of early life development in the aetiology of kidney disease and emphasize the need to develop strategies to optimize nephron endowment in IUGR and preterm infants.
Collapse
Affiliation(s)
- Megan R Sutherland
- Department of Anatomy and Developmental Biology and Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Mary Jane Black
- Department of Anatomy and Developmental Biology and Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia.
| |
Collapse
|
10
|
Exposure to high levels of oxygen in neonatal rats induce a decrease in hemoglobin levels. Pediatr Res 2022; 92:430-435. [PMID: 34718354 DOI: 10.1038/s41390-021-01802-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 08/26/2021] [Accepted: 10/10/2021] [Indexed: 11/08/2022]
Abstract
BACKGROUND Anemia of prematurity is common in extremely preterm neonates, and oxygen exposure may participate to anemia by inhibiting erythropoietin secretion. We aimed to determine whether hyperoxia exerts an independent role in the occurrence of the anemia of prematurity. METHODS Sprague-Dawley pups were exposed to 80% oxygen or room air from days 3 to 10 of life. Main outcome was the difference in hemoglobin and circulating erythropoietin levels in animals exposed to hyperoxia at 10 days of life. We performed a complete blood count analysis using fluorescent laser flow cytometry and measured circulating erythropoietin levels using ELISA. RESULTS We found lower hemoglobin in the hyperoxia group, compared to the normoxia group, both in males (70 ± 3 versus 78 ± 2 g/l) and in females (71 ± 2 versus 81 ± 3 g/l) at 10 days of life. Reticulocyte count was not increased in the hyperoxia group. Circulating erythropoietin levels were lower at 10 days of life in the animals exposed to hyperoxia, both in males (33 ± 7 versus 73 ± 6 pg/ml) and in females (37 ± 5 versus 66 ± 3 pg/ml), but were similar at 28 days of life. CONCLUSION Neonatal exposure to hyperoxia decreases hematopoiesis in rats. IMPACT Mechanisms leading to anemia of prematurity are not well known and their study in humans is complicated due to multiple confounders. This study shows for the first time that exposure to high concentrations of oxygen in the neonatal period decreases hematopoiesis in rats, providing insight on the pathophysiological mechanisms of the anemia of prematurity. This research paves the way for future therapeutic developments aiming to reduce the burden of anemia of prematurity and the necessity of red blood cell transfusions in extremely preterm neonates.
Collapse
|
11
|
Bertagnolli M, Dartora DR, Lamata P, Zacur E, Mai-Vo TA, He Y, Beauchamp L, Lewandowski AJ, Cloutier A, Sutherland MR, Santos RAS, Nuyt AM. Reshaping the Preterm Heart: Shifting Cardiac Renin-Angiotensin System Towards Cardioprotection in Rats Exposed to Neonatal High-Oxygen Stress. Hypertension 2022; 79:1789-1803. [PMID: 35588210 PMCID: PMC9278707 DOI: 10.1161/hypertensionaha.122.19115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Approximately 10% of infants are born preterm. Preterm birth leads to short and long-term changes in cardiac shape and function. By using a rat model of neonatal high-oxygen (80%O2) exposure, mimicking the premature hyperoxic transition to the extrauterine environment, we revealed a major role of the renin-angiotensin system peptide Angio II (angiotensin II) and its receptor AT1 (angiotensin receptor type 1) on neonatal O2-induced cardiomyopathy. Here, we tested whether treatment with either orally active compounds of the peptides Angio-(1-7) or alamandine included in cyclodextrin could prevent postnatal cardiac remodeling and the programming of cardiomyopathy induced by neonatal high-O2 exposure. METHODS Sprague-Dawley pups were exposed to room air or 80% O2 from postnatal day 3 (P3) to P10. Neonatal rats were treated orally from P3 to P10 and assessed at P10 and P28. Left ventricular (LV) shapes were characterized by tridimensional computational atlases of ultrasound images in addition to histomorphometry. RESULTS At P10, high O2-exposed rats presented a smaller, globular and hypertrophied LV shape versus controls. Treatment with cyclodextrin-Angio-(1-7) significantly improved LV function in the O2-exposed neonatal rats and slightly changed LV shape. Cyclodextrin-alamandine and cyclodextrin-Angio-(1-7) treatments similarly reduced hypertrophy at P10 as well as LV remodeling and dysfunction at P28. Both treatments upregulated cardiac angiotensin-converting enzyme 2 in O2-exposed rats at P10 and P28. CONCLUSIONS Our findings demonstrate LV remodeling changes induced by O2-stress and the potential benefits of treatments targeting the cardioprotective renin-angiotensin system axis, supporting the neonatal period as an important window for interventions aiming at preventing cardiomyopathy in people born preterm.
Collapse
Affiliation(s)
- Mariane Bertagnolli
- Sainte-Justine University Hospital Research Center, Université de Montréal, Canada (M.B., D.R.D., T.-A.M.-V., Y.H., L.B., A.C., M.R.S., A.M.N.).,Research Center of the Hospital Sacré-Coeur, CIUSSS Nord-de-l'Île-de-Montréal, Canada (M.B.).,School of Physical and Occupational Therapy, Faculty of Medicine, McGill University, Montréal, Canada (M.B.)
| | - Daniela R Dartora
- Sainte-Justine University Hospital Research Center, Université de Montréal, Canada (M.B., D.R.D., T.-A.M.-V., Y.H., L.B., A.C., M.R.S., A.M.N.).,Instituto de Cardiologia de Porto Alegre, Fundação Universitária de Cardiologia, Brazil (D.R.D.)
| | - Pablo Lamata
- Department of Biomedical Engineering, Division of Imaging Sciences and Biomedical Engineering, King's College London, United Kingdom (P.L., E.Z.)
| | - Ernesto Zacur
- Department of Biomedical Engineering, Division of Imaging Sciences and Biomedical Engineering, King's College London, United Kingdom (P.L., E.Z.)
| | - Thuy-An Mai-Vo
- Sainte-Justine University Hospital Research Center, Université de Montréal, Canada (M.B., D.R.D., T.-A.M.-V., Y.H., L.B., A.C., M.R.S., A.M.N.)
| | - Ying He
- Sainte-Justine University Hospital Research Center, Université de Montréal, Canada (M.B., D.R.D., T.-A.M.-V., Y.H., L.B., A.C., M.R.S., A.M.N.)
| | - Léonie Beauchamp
- Sainte-Justine University Hospital Research Center, Université de Montréal, Canada (M.B., D.R.D., T.-A.M.-V., Y.H., L.B., A.C., M.R.S., A.M.N.)
| | - Adam J Lewandowski
- Oxford Cardiovascular Clinical Research Facility, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, United Kingdom (A.J.L.)
| | - Anik Cloutier
- Sainte-Justine University Hospital Research Center, Université de Montréal, Canada (M.B., D.R.D., T.-A.M.-V., Y.H., L.B., A.C., M.R.S., A.M.N.)
| | - Megan R Sutherland
- Sainte-Justine University Hospital Research Center, Université de Montréal, Canada (M.B., D.R.D., T.-A.M.-V., Y.H., L.B., A.C., M.R.S., A.M.N.).,Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia (M.R.S.)
| | - Robson A S Santos
- Department of Physiology, Instituto Nacional de Ciência e Tecnologia - Nanobiofar, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil (R.A.S.S.)
| | - Anne Monique Nuyt
- Sainte-Justine University Hospital Research Center, Université de Montréal, Canada (M.B., D.R.D., T.-A.M.-V., Y.H., L.B., A.C., M.R.S., A.M.N.)
| |
Collapse
|
12
|
Kidney Injuries and Evolution of Chronic Kidney Diseases Due to Neonatal Hyperoxia Exposure Based on Animal Studies. Int J Mol Sci 2022; 23:ijms23158492. [PMID: 35955627 PMCID: PMC9369080 DOI: 10.3390/ijms23158492] [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: 06/15/2022] [Revised: 07/22/2022] [Accepted: 07/26/2022] [Indexed: 02/04/2023] Open
Abstract
Preterm birth interrupts the development and maturation of the kidneys during the critical growth period. The kidneys can also exhibit structural defects and functional impairment due to hyperoxia, as demonstrated by various animal studies. Furthermore, hyperoxia during nephrogenesis impairs renal tubular development and induces glomerular and tubular injuries, which manifest as renal corpuscle enlargement, renal tubular necrosis, interstitial inflammation, and kidney fibrosis. Preterm birth along with hyperoxia exposure induces a pathological predisposition to chronic kidney disease. Hyperoxia-induced kidney injuries are influenced by several molecular factors, including hypoxia-inducible factor-1α and interleukin-6/Smad2/transforming growth factor-β, and Wnt/β-catenin signaling pathways; these are key to cell proliferation, tissue inflammation, and cell membrane repair. Hyperoxia-induced oxidative stress is characterized by the attenuation or the induction of multiple molecular factors associated with kidney damage. This review focuses on the molecular pathways involved in the pathogenesis of hyperoxia-induced kidney injuries to establish a framework for potential interventions.
Collapse
|
13
|
DeFreitas MJ, Katsoufis CP, Benny M, Young K, Kulandavelu S, Ahn H, Sfakianaki A, Abitbol CL. Educational Review: The Impact of Perinatal Oxidative Stress on the Developing Kidney. Front Pediatr 2022; 10:853722. [PMID: 35844742 PMCID: PMC9279889 DOI: 10.3389/fped.2022.853722] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 06/13/2022] [Indexed: 01/01/2023] Open
Abstract
Oxidative stress occurs when there is an imbalance between reactive oxygen species/reactive nitrogen species and antioxidant systems. The interplay between these complex processes is crucial for normal pregnancy and fetal development; however, when oxidative stress predominates, pregnancy related complications and adverse fetal programming such as preterm birth ensues. Understanding how oxidative stress negatively impacts outcomes for the maternal-fetal dyad has allowed for the exploration of antioxidant therapies to prevent and/or mitigate disease progression. In the developing kidney, the negative impact of oxidative stress has also been noted as it relates to the development of hypertension and kidney injury mostly in animal models. Clinical research addressing the implications of oxidative stress in the developing kidney is less developed than that of the neurodevelopmental and respiratory conditions of preterm infants and other vulnerable neonatal groups. Efforts to study the oxidative stress pathway along the continuum of the perinatal period using a team science approach can help to understand the multi-organ dysfunction that the maternal-fetal dyad sustains and guide the investigation of antioxidant therapies to ameliorate the global toxicity. This educational review will provide a comprehensive and multidisciplinary perspective on the impact of oxidative stress during the perinatal period in the development of maternal and fetal/neonatal complications, and implications on developmental programming of accelerated aging and cardiovascular and renal disease for a lifetime.
Collapse
Affiliation(s)
- Marissa J. DeFreitas
- Division of Pediatric Nephrology, Department of Pediatrics, University of Miami, Miami, FL, United States
- Department of Pediatrics, Batchelor Children’s Research Institute, University of Miami, Miami, FL, United States
| | - Chryso P. Katsoufis
- Division of Pediatric Nephrology, Department of Pediatrics, University of Miami, Miami, FL, United States
- Department of Pediatrics, Batchelor Children’s Research Institute, University of Miami, Miami, FL, United States
| | - Merline Benny
- Department of Pediatrics, Batchelor Children’s Research Institute, University of Miami, Miami, FL, United States
- Division of Neonatology, Department of Pediatrics, University of Miami, Miami, FL, United States
| | - Karen Young
- Department of Pediatrics, Batchelor Children’s Research Institute, University of Miami, Miami, FL, United States
- Division of Neonatology, Department of Pediatrics, University of Miami, Miami, FL, United States
| | - Shathiyah Kulandavelu
- Division of Pediatric Nephrology, Department of Pediatrics, University of Miami, Miami, FL, United States
- Interdisciplinary Stem Cell Institute, University of Miami, Miami, FL, United States
| | - Hyunyoung Ahn
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Miami, Miami, FL, United States
| | - Anna Sfakianaki
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Miami, Miami, FL, United States
| | - Carolyn L. Abitbol
- Division of Pediatric Nephrology, Department of Pediatrics, University of Miami, Miami, FL, United States
- Department of Pediatrics, Batchelor Children’s Research Institute, University of Miami, Miami, FL, United States
| |
Collapse
|
14
|
Iacobelli S, Guignard JP. When the progresses in neonatology lead to severe congenital nephron deficit: is there a pilot in the NICU? Pediatr Nephrol 2022; 37:1277-1284. [PMID: 34761299 DOI: 10.1007/s00467-021-05338-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 10/10/2021] [Accepted: 10/14/2021] [Indexed: 12/19/2022]
Abstract
Advances in the care of neonates to the extreme limits of viability have increased the risk of severe comorbidities in surviving preemies. The respiratory and the neurodevelopmental consequences of premature birth and/or intra-uterine growth retardation have been well described. Because of the usual clinical silence of the kidney, the long-term renal consequences of low birth weight have not been as well studied. A case report illustrates the risk factors associated with low birth weight and prematurity and discusses the pathogenesis of the late consequences of the congenital nephron deficit associated with a low birth weight. Practical recommendations are given for a tight follow-up of these newly born preemies.
Collapse
Affiliation(s)
- Silvia Iacobelli
- Réanimation Néonatale et Pédiatrique, Néonatologie, CHU La Réunion, Site Sud, Saint Pierre, France. .,Centre d'Études Périnatales de l'Océan Indien UR 7388, Université de la Réunion, Saint Pierre, France.
| | | |
Collapse
|
15
|
SAITOH MASAHITO, ENDO AMANE, MATSUDA AKINA, MIYANO HIROKI, GONDA YUSUKE, MIZUTANI AKIRA, HARA TAICHI, NAKAGAWA MAYU, SAKURAYA KOJI, MURANO YAYOI, NISHIZAKI NAOTO, FUJINAGA SHUICHIRO, OHTOMO YOSHIYUKI, SHIMIZU TOSHIAKI. Protective Effects of Hydrogen-rich Water Intake on Renal Injury in Neonatal Rats with High Oxygen Loading. JUNTENDO IJI ZASSHI = JUNTENDO MEDICAL JOURNAL 2022; 68:235-241. [PMID: 39021721 PMCID: PMC11250013 DOI: 10.14789/jmj.jmj21-0048-oa] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 02/02/2022] [Indexed: 07/20/2024]
Abstract
Objectives This study aimed to investigate the protective effects of hydrogen-rich water (HW) intake on renal injury in neonatal rats with high oxygen loading. Materials We used pregnant and newborn Sprague-Dawley rats. Methods Four groups were set up, with mother and newborn rats immediately after delivery as one group: RA-PW (room air and purified water), RA-HW (room air and HW), O2-PW (80% oxygen and purified water), and O2-HW (80% oxygen and HW). The newborn rats were maintained in either a normoxic (room air, 21% oxygen) or controlled hyperoxic (80% oxygen) environment from birth. Then, HW (O2-HW and RA-HW groups) or PW (O2-PW and RA-PW groups) was administered to parents of each group. Results The number of immature glomeruli significantly increased in the O2-PW group (exposed to hyperoxia). Conversely, the O2-HW group had significantly fewer immature glomeruli than O2-PW group. In the RT-PCR analysis of kidney tissue, α-SMA, TGF-β, and TNF-α levels were significantly higher in the O2-PW group than in the RA-PW group and significantly lower in the O2-HW group than in the O2-PW group. Conclusions HW intake can potentially reduce oxidative stress and prevent renal injury in neonates with high oxygen loading.
Collapse
Affiliation(s)
| | - AMANE ENDO
- Corresponding author: Amane Endo, Department of Pediatrics and Adolescent Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan, TEL: +81-3-3813-3111 ext. 5670 FAX: +81-3-5800-1580 E-mail:
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Voggel J, Mohr J, Nüsken KD, Dötsch J, Nüsken E, Alejandre Alcazar MA. Translational insights into mechanisms and preventive strategies after renal injury in neonates. Semin Fetal Neonatal Med 2022; 27:101245. [PMID: 33994314 DOI: 10.1016/j.siny.2021.101245] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Adverse perinatal circumstances can cause acute kidney injury (AKI) and contribute to chronic kidney disease (CKD). Accumulating evidence indicate that a wide spectrum of perinatal conditions interferes with normal kidney development and ultimately leads to aberrant kidney structure and function later in life. The present review addresses the lack of mechanistic knowledge with regard to perinatal origins of CKD and provides a comprehensive overview of pre- and peri-natal insults, including genetic predisposition, suboptimal nutritional supply, obesity and maternal metabolic disorders as well as placental insufficiency leading to intrauterine growth restriction (IUGR), prematurity, infections, inflammatory processes, and the need for life-saving treatments (e.g. oxygen supplementation, mechanical ventilation, medications) in neonates. Finally, we discuss future preventive, therapeutic, and regenerative directions. In summary, this review highlights the perinatal vulnerability of the kidney and the early origins of increased susceptibility toward AKI and CKD during postnatal life. Promotion of kidney health and prevention of disease require the understanding of perinatal injury in order to optimize perinatal micro- and macro-environments and enable normal kidney development.
Collapse
Affiliation(s)
- Jenny Voggel
- University of Cologne, Faculty of Medicine, University Hospital Cologne, Department of Pediatric and Adolescent Medicine, Germany; University of Cologne, Faculty of Medicine, University Hospital Cologne, Center for Molecular Medicine Cologne (CMMC), Germany
| | - Jasmine Mohr
- University of Cologne, Faculty of Medicine, University Hospital Cologne, Translational Experimental Pediatrics - Experimental Pulmonology, Department of Pediatric and Adolescent Medicine, Germany; University of Cologne, Faculty of Medicine, University Hospital Cologne, Center for Molecular Medicine Cologne (CMMC), Germany
| | - Kai-Dietrich Nüsken
- University of Cologne, Faculty of Medicine, University Hospital Cologne, Department of Pediatric and Adolescent Medicine, Germany
| | - Jörg Dötsch
- University of Cologne, Faculty of Medicine, University Hospital Cologne, Department of Pediatric and Adolescent Medicine, Germany
| | - Eva Nüsken
- University of Cologne, Faculty of Medicine, University Hospital Cologne, Department of Pediatric and Adolescent Medicine, Germany
| | - Miguel A Alejandre Alcazar
- University of Cologne, Faculty of Medicine, University Hospital Cologne, Translational Experimental Pediatrics - Experimental Pulmonology, Department of Pediatric and Adolescent Medicine, Germany; University of Cologne, Faculty of Medicine, University Hospital Cologne, Center for Molecular Medicine Cologne (CMMC), Germany; Excellence Cluster on Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Faculty of Medicine, University Hospital Cologne Cologne, Germany; Institute for Lung Health, University of Giessen and Marburg Lung Centre (UGMLC), Member of the German Centre for Lung Research (DZL), Gießen, Germany.
| |
Collapse
|
17
|
Anti-Tn Monoclonal Antibody Ameliorates Hyperoxia-Induced Kidney Injury by Suppressing Oxidative Stress and Inflammation in Neonatal Mice. Mediators Inflamm 2021; 2021:1180543. [PMID: 34720748 PMCID: PMC8553484 DOI: 10.1155/2021/1180543] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Accepted: 10/09/2021] [Indexed: 01/27/2023] Open
Abstract
The Tn antigen, an N-acetylgalactosamine structure linked to serine or threonine, has been shown to induce high-specificity, high-affinity anti-Tn antibodies in mice. Maternal immunization with the Tn vaccine increases serum anti-Tn antibody titers and attenuates hyperoxia-induced kidney injury in neonatal rats. However, immunizing mothers to treat neonatal kidney disease is clinically impractical. This study is aimed at determining whether anti-Tn monoclonal antibody treatment ameliorates hyperoxia-induced kidney injury in neonatal mice. Newborn BALB/c mice were exposed to room air (RA) or normobaric hyperoxia (85% O2) for 1 week. On postnatal days 2, 4, and 6, the mice were injected intraperitoneally with PBS alone or with anti-Tn monoclonal antibodies at 25 μg/g body weight in 50 μL phosphate-buffered saline (PBS). The mice were divided into four study groups: RA + PBS, RA + anti-Tn monoclonal antibody, O2 + PBS, and O2 + anti-Tn monoclonal antibody. The kidneys were excised for histology, oxidative stress, cytokine, and Western blot analyses on postnatal day 7. The O2 + PBS mice exhibited significantly higher kidney injury scores, 8-hydroxy-2'-deoxyguanosine (8-OHdG) and nuclear factor-κB (NF-κB) expression, and cytokine levels than did the RA + PBS mice or RA + anti-Tn mice. Anti-Tn monoclonal antibody treatment reduced kidney injury and cytokine levels to normoxic levels. The attenuation of kidney injury was accompanied by a reduction of oxidative stress and NF-κB expression. Therefore, we propose that anti-Tn monoclonal antibody treatment ameliorates hyperoxia-induced kidney injury by suppressing oxidative stress and inflammation in neonatal mice.
Collapse
|
18
|
Zangaladze A, Cai CL, Marcelino M, Aranda JV, Beharry KD. Renal biomarkers of acute kidney injury in response to increasing intermittent hypoxia episodes in the neonatal rat. BMC Nephrol 2021; 22:299. [PMID: 34481475 PMCID: PMC8418040 DOI: 10.1186/s12882-021-02507-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 08/19/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND We tested the hypotheses that: 1) early exposure to increasing episodes of clinically relevant intermittent hypoxia (IH) is detrimental to the developing kidneys; and 2) there is a critical number of daily IH episodes which will result in irreparable renal damage that may involve angiotensin (Ang) II and endothelin (ET)-1. METHODS At birth (P0), neonatal rat pups were exposed to brief IH episodes from the first day of life (P0) to P7 or from P0-P14. Pups were either euthanized immediately or placed in room air (RA) until P21. RA littermates served as controls. Kidneys were harvested at P7, P14, and P21 for histopathology; angiotensin converting enzyme (ACE), ACE-2, ET-1, big ET-1, and malondialdehyde (MDA) levels; immunoreactivity of ACE, ACE-2, ET-1, ET-2, ET receptors (ETAR, ETBR), and hypoxia inducible factor (HIF)1α; and apoptosis (TUNEL stain). RESULTS Histopathology showed increased renal damage with 8-12 IH episodes/day, and was associated with Ang II, ACE, HIF1α, and apoptosis. ACE-2 was not expressed at P7, and minimally increased at P14. However, a robust ACE-2 response was seen during recovery with maximum levels noted in the groups recovering from 8 IH episodes/day. ET-1, big ET-1, ETAR, ETBR, and MDA increased with increasing levels of neonatal IH. CONCLUSIONS Chronic neonatal IH causes severe damage to the developing kidney with associated elevations in vasoconstrictors, suggesting hypertension, particularly with 8 neonatal IH episodes. ACE-2 is not activated in early postnatal life, and this may contribute to IH-induced vasoconstriction. Therapeutic targeting of ACE and ET-1 may help decrease the risk for kidney injury in the developing neonate to prevent and/or treat neonatal acute kidney injury and/or chronic kidney disease.
Collapse
Affiliation(s)
- Anano Zangaladze
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, State University of New York, Downstate Medical Center, Brooklyn, NY, USA
| | - Charles L Cai
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, State University of New York, Downstate Medical Center, Brooklyn, NY, USA
| | - Matthew Marcelino
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, State University of New York, Downstate Medical Center, Brooklyn, NY, USA
| | - Jacob V Aranda
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, State University of New York, Downstate Medical Center, Brooklyn, NY, USA
- Department of Ophthalmology, State University of New York, Downstate Medical Center, Brooklyn, NY, USA
- SUNY Eye Institute, New York, NY, USA
| | - Kay D Beharry
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, State University of New York, Downstate Medical Center, Brooklyn, NY, USA.
- Department of Ophthalmology, State University of New York, Downstate Medical Center, Brooklyn, NY, USA.
- SUNY Eye Institute, New York, NY, USA.
- Department of Pediatrics & Ophthalmology, Neonatal-Perinatal Medicine Clinical & Translational Research Labs, State University of New York, Downstate Medical Center, 450 Clarkson Avenue, Box 49, Brooklyn, NY, 11203, USA.
| |
Collapse
|
19
|
Immunization with anti-Tn immunogen in maternal rats protects against hyperoxia-induced kidney injury in newborn offspring. Pediatr Res 2021; 89:476-482. [PMID: 32311698 DOI: 10.1038/s41390-020-0894-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 02/27/2020] [Accepted: 03/30/2020] [Indexed: 11/08/2022]
Abstract
BACKGROUND Neonatal hyperoxia increases oxidative stress and adversely disturbs glomerular and tubular maturity. Maternal Tn immunization induces anti-Tn antibody titer and attenuates hyperoxia-induced lung injury in neonatal rats. METHODS We intraperitoneally immunized female Sprague-Dawley rats (6 weeks old) with Tn immunogen (50 μg/dose) or carrier protein five times at biweekly intervals on 8, 6, 4, 2, and 0 weeks before the delivery day. The pups were reared for 2 weeks in either room air (RA) or in 85% oxygen-enriched atmosphere (O2), thus generating four study groups, namely carrier protein + RA, Tn vaccine + RA, carrier protein + O2, and Tn vaccine + O2. On postnatal day 14, the kidneys were harvested for the oxidative stress marker 8-hydroxy-2'-deoxyguanosine (8-OHdG), nuclear factor-κB (NF-κB), and collagen expression and histological analyses. RESULTS Hyperoxia reduced body weight, induced tubular and glomerular injuries, and increased 8-OHdG and NF-κB expression and collagen deposition in the kidneys. By contrast, maternal Tn immunization reduced kidney injury and collagen deposition in neonatal rats. Furthermore, kidney injury attenuation was accompanied by a reduction in 8-OHdG and NF-κB expression. CONCLUSION Maternal Tn immunization protects against hyperoxia-induced kidney injury in neonatal rats by attenuating oxidative stress and NF-κB activity. IMPACT Hyperoxia increased nuclear factor-κB (NF-κB) activity and collagen deposition in neonatal rat kidney. Maternal Tn immunization reduced kidney injury as well as collagen deposition in neonatal rats. Maternal Tn immunization reduced kidney injury and was associated with a reduction in 8-hydroxy-2'-deoxyguanosine and NF-κB activity. Tn vaccine can be a promising treatment modality against hyperoxia-induced kidney injury in neonates.
Collapse
|
20
|
Kumar A, Dey AD, Behl T, Chadha S, Aggarwal V. Exploring the multifocal therapeutic approaches in COVID-19: A ray of hope. Int Immunopharmacol 2021; 90:107156. [PMID: 33189613 PMCID: PMC7608010 DOI: 10.1016/j.intimp.2020.107156] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 09/24/2020] [Accepted: 10/21/2020] [Indexed: 12/28/2022]
Abstract
The ongoing global pandemic of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) is marked as one of the most challenging infectious diseases in the history of mankind with affliction of ~29,737,453 confirmed cases globally. Looking at the present scenario where there is a parallel increment in curve with time, there is an utmost emergency to discover a perennial solution to this life-threatening virus which has led the Human race in an unusual state of affair. The entire health care fraternity is engaged in endeavouring an ultimate way out to hit this pandemic but no such research made till now has been approved yet, to have the potential to bring an end to this fatal situation. Although a few possible treatment choices exist at the moment yet the requirement to search for a still better therapeutic option remains persistent. Global laboratories are working day and night in search for an effective vaccine, many are undergoing clinical trials but their commercialization is no less than a year away. The present review highlights the current potential therapies viz., vaccines, immunotherapies, convulsant plasma therapies, corticosteroids, antithrombotic, intravenous immunoglobulins, nocturnal oxygen therapy etc. that may prove beneficial in attenuating the pandemic situation. However, comparison and presentation of collective data on the therapeutic advancements in mitigating the pandemic situation needs further clinical investigations in order to prove boon to mankind.
Collapse
Affiliation(s)
- Arun Kumar
- Chitkara College of Pharmacy, Chitkara University, Punjab, India.
| | - Asmita Deka Dey
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Swati Chadha
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Vishal Aggarwal
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| |
Collapse
|
21
|
Neonatal Hyperoxia Downregulates Claudin-4, Occludin, and ZO-1 Expression in Rat Kidney Accompanied by Impaired Proximal Tubular Development. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:2641461. [PMID: 33343804 PMCID: PMC7725566 DOI: 10.1155/2020/2641461] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 10/25/2020] [Accepted: 11/09/2020] [Indexed: 11/21/2022]
Abstract
Hyperoxia is essential to manage in preterm infants but causes injury to immature kidney. Previous study indicates that hyperoxia causes oxidative damage to neonatal kidney and impairs renal development. However, the underlying mechanisms by which neonatal hyperoxia effects on immature kidney still need to be elucidated. Tight junction, among which the representative proteins are claudin-4, occludin, and ZO-1, plays a crucial role in nephrogenesis and maintaining renal function. Inflammatory cytokines are involved in the pleiotropic regulation of tight junction proteins. Here, we investigated how neonatal hyperoxia affected the expression of key tight junction proteins and inflammatory factors (IL-6 and TNF-α) in the developing rat kidneys and elucidated their correlation with renal injury. We found claudin-4, occludin, and zonula occludens-1 (ZO-1) expression in proximal tubules was significantly downregulated after neonatal hyperoxia. The expression of these tight junction proteins was positively correlated with that of IL-6 and TNF-α, while claudin-4 expression was positively correlated with injury score of proximal tubules in mature kidneys. These findings indicated that impaired expression of tight junction proteins in kidney might be a potential mechanism of hyperoxia-induced nephrogenic disorders. It provides new insights to further study oxidative renal injury and development disorders and will be helpful for seeking potential therapeutics for hyperoxia-induced renal injury in the future.
Collapse
|
22
|
Bae J, Kim J, Lee S, Ju JW, Cho YJ, Kim TK, Jeon Y, Nam K. Association Between Intraoperative Hyperoxia and Acute Kidney Injury After Cardiac Surgery: A Retrospective Observational Study. J Cardiothorac Vasc Anesth 2020; 35:2405-2414. [PMID: 33342731 DOI: 10.1053/j.jvca.2020.11.054] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 11/22/2020] [Accepted: 11/23/2020] [Indexed: 11/11/2022]
Abstract
OBJECTIVE Optimal oxygen management during cardiac surgery has not been established, and studies on the effects of perioperative hyperoxia on postoperative acute kidney injury (AKI) are scarce. The association between intraoperative hyperoxia and AKI after cardiac surgery involving cardiopulmonary bypass was evaluated for the present study. DESIGN Retrospective observational study. SETTING A tertiary teaching hospital. PARTICIPANTS Adult patients who underwent cardiac surgery with cardiopulmonary bypass from November 2006-December 2018. INTERVENTIONS None. MEASUREMENTS AND MAIN RESULTS The area above arterial oxygen partial pressure (PaO2) threshold of 300 mmHg (AOT300, mmHg × h) was used as a metric of intraoperative hyperoxia and was associated with postoperative AKI, using the logistic regression analysis. Data also were fitted using the restricted cubic spline model. Sensitivity analyses were conducted using different PaO2 thresholds (150, 200, 250, and 350 mmHg). A total of 2,926 patients were analyzed. Intraoperative AOT300 independently was associated with the risk of AKI (odds ratio 1.0009; 95% confidence interval 1.0002-1.0015). A PaO2 increment of 100 mmHg above PaO2 300 mmHg for an hour was associated with an increased risk of AKI by 9.4% (1.0009100 ≈ 1.094). In the spline model, the log-odds of AKI increased as AOT300 increased. In the sensitivity analyses, AOT250 and AOT350 also significantly were associated with the risk of AKI, whereas AOT150 and AOT200 were not. As the PaO2 threshold increased from 150 to 350 mmHg, the odds ratio gradually increased. CONCLUSIONS Intraoperative hyperoxia significantly was associated with the risk of AKI after cardiac surgery involving cardiopulmonary bypass.
Collapse
Affiliation(s)
- Jinyoung Bae
- Department of Anesthesiology and Pain Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Jay Kim
- Department of Anesthesiology and Pain Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Seohee Lee
- Department of Anesthesiology and Pain Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Jae-Woo Ju
- Department of Anesthesiology and Pain Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Youn Joung Cho
- Department of Anesthesiology and Pain Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Tae Kyong Kim
- Department of Anesthesiology and Pain Medicine, Seoul Metropolitan Government-Seoul National University Boramae Medical Centre, Seoul National University College of Medicine, Seoul, Korea
| | - Yunseok Jeon
- Department of Anesthesiology and Pain Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Karam Nam
- Department of Anesthesiology and Pain Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea.
| |
Collapse
|
23
|
Effects of Klotho supplementation on hyperoxia-induced renal injury in a rodent model of postnatal nephrogenesis. Pediatr Res 2020; 88:565-570. [PMID: 32059229 PMCID: PMC8226112 DOI: 10.1038/s41390-020-0803-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 12/18/2019] [Accepted: 02/01/2020] [Indexed: 01/28/2023]
Abstract
BACKGROUND Hyperoxia (HO) causes kidney injury in preterm infants; however, whether these effects are modifiable is unknown. We hypothesized that administration of exogenous soluble Klotho, a kidney-derived antioxidant, would attenuate HO-induced kidney injury during postnatal nephrogenesis in rats. METHODS Sprague Dawley neonatal rats assigned to normoxia (21% O2) or HO (85% O2) groups from postnatal day (P) 1 to 21 were randomly assigned to receive alternate day intraperitoneal injections of recombinant Klotho or placebo for 3 weeks. They were recovered in normoxia for an additional 3 weeks and sacrificed at 6 weeks. Renal artery resistance and pulsatility indices, tubular injury scores, glomerular area, and renal antioxidant capacity were assessed. RESULTS Rodents exposed to HO during postnatal nephrogenesis had reduced kidney Klotho expression, glomerulomegaly, and higher tubular injury scores. Exogenous Klotho administration improved renal perfusion as indicated by decreases in both resistance and pulsatility indices and increased antioxidant enzyme expression. CONCLUSIONS HO exposure during postnatal nephrogenesis in rodents results in a decline in kidney Klotho expression, decreased renal perfusion, enlarged glomerular size, and tubular injury. The exogenous administration of Klotho attenuated HO-induced kidney injury and augmented antioxidant capacity.
Collapse
|
24
|
Harer MW, Charlton JR, Tipple TE, Reidy KJ. Preterm birth and neonatal acute kidney injury: implications on adolescent and adult outcomes. J Perinatol 2020; 40:1286-1295. [PMID: 32277164 DOI: 10.1038/s41372-020-0656-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 03/02/2020] [Accepted: 03/11/2020] [Indexed: 02/06/2023]
Abstract
As a result of preterm birth, immature kidneys are exposed to interventions in the NICU that promote survival, but are nephrotoxic. Furthermore, the duration of renal development may be truncated in these vulnerable neonates. Immaturity and nephrotoxic exposures predispose preterm newborns to acute kidney injury (AKI), particularly in the low birth weight and extremely preterm gestational age groups. Several studies have associated preterm birth as a risk factor for future chronic kidney disease (CKD). However, only a few publications have investigated the impact of neonatal AKI on CKD development. Here, we will review the evidence linking preterm birth and AKI in the NICU to CKD and highlight the knowledge gaps and opportunities for future research. For neonatal intensive care studies, we propose the inclusion of AKI as an important short-term morbidity outcome and CKD findings such as a reduced glomerular filtration rate in the assessment of long-term outcomes.
Collapse
Affiliation(s)
- Matthew W Harer
- Department of Pediatrics, Division of Neonatology, University of Wisconsin-Madison, Madison, WI, USA
| | - Jennifer R Charlton
- Department of Pediatrics, Division of Nephrology, University of Virginia Children's Hospital, Box 800386, Charlottesville, VA, USA.
| | - Trent E Tipple
- Department of Pediatrics, Section of Neonatal-Perinatal Medicine, University of Oklahoma College of Medicine, Oklahoma City, OK, USA
| | - Kimberly J Reidy
- Department of Pediatrics, Division of Pediatric Nephrology, Children's Hospital at Montefiore, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
25
|
Shen C, Yue X, Wang J, Shi C, Li W. Nocturnal oxygen therapy as an option for early COVID-19. Int J Infect Dis 2020; 98:176-179. [PMID: 32599285 PMCID: PMC7319633 DOI: 10.1016/j.ijid.2020.06.080] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 06/20/2020] [Accepted: 06/23/2020] [Indexed: 01/04/2023] Open
Abstract
Oxygen supplementation may disrupt virus replication. Oxygen therapy can improve the antiviral immune response. An oxygen-rich environment may down-regulate ACE2 expression. Nocturnal oxygen therapy may delay the progression of COVID-19.
There is currently no effective antiviral therapy or immune-based treatment for coronavirus disease (COVID-19). The urgent challenge is to prevent the transition of COVID-19 from mild to severe infection. This paper discussed nocturnal oxygen therapy as a new option for people with COVID-19 under home quarantine. It suggested that nocturnal oxygen therapy in the early stages may be helpful in preventing disease progression by inhibiting the rapid replication of the virus and improving the body's antiviral ability.
Collapse
Affiliation(s)
- Chongxing Shen
- Department of Urology, The Third Affiliated Hospital (General Hospital) of Chongqing Medical University, Chongqing, China
| | - Xiaofeng Yue
- Department of Urology, The Third Affiliated Hospital (General Hospital) of Chongqing Medical University, Chongqing, China
| | - Jianwu Wang
- Department of Urology, The Third Affiliated Hospital (General Hospital) of Chongqing Medical University, Chongqing, China
| | - Chunmeng Shi
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University, Chongqing, China.
| | - Weibing Li
- Department of Urology, The Third Affiliated Hospital (General Hospital) of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
26
|
Charlton JR, Baldelomar EJ, deRonde KA, Cathro HP, Charlton NP, Criswell SJ, Hyatt DM, Nam S, Pearl V, Bennett KM. Nephron loss detected by MRI following neonatal acute kidney injury in rabbits. Pediatr Res 2020; 87:1185-1192. [PMID: 31805577 PMCID: PMC7255918 DOI: 10.1038/s41390-019-0684-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 10/14/2019] [Accepted: 10/18/2019] [Indexed: 12/16/2022]
Abstract
BACKGROUND Acute kidney injury affects nearly 30% of preterm neonates in the intensive care unit. We aimed to determine whether nephrotoxin-induced AKI disrupted renal development assessed by imaging (CFE-MRI). METHODS Neonatal New Zealand rabbits received indomethacin and gentamicin (AKI) or saline (control) for four days followed by cationic ferritin (CF) at six weeks. Ex vivo images were acquired using a gradient echo pulse sequence on 7 T MRI. Glomerular number (Nglom) and apparent glomerular volume (aVglom) were determined. CF toxicity was assessed at two and 28 days in healthy rabbits. RESULTS Nglom was lower in the AKI group as compared to controls (74,034 vs 198,722, p < 0.01). aVglom was not different (AKI: 7.3 × 10-4 vs control: 6.2 × 10-4 mm3, p = 0.69). AKI kidneys had a band of glomeruli distributed radially in the cortex that were undetectable by MRI. Following CF injection, there was no difference in body or organ weights except for the liver, and transient changes in serum iron, platelets and white blood cell count. CONCLUSIONS Brief nephrotoxin exposure during nephrogenesis results in fewer glomeruli and glomerular maldevelopment in a unique pattern detectable by MRI. Whole kidney evaluation by CFE-MRI may provide an important tool to understand the development of CKD following AKI.
Collapse
Affiliation(s)
- Jennifer R Charlton
- Department of Pediatrics, Division of Nephrology, University of Virginia, Charlottesville, VA, USA.
| | | | - Kimberly A deRonde
- Department of Pediatrics, Division of Nephrology, University of Virginia, Charlottesville, VA, USA
| | - Helen P Cathro
- Department of Pathology, University of Virginia, Charlottesville, VA, USA
| | - Nathan P Charlton
- Department of Emergency Medicine, Division of Medical Toxicology, University of Virginia, Charlottesville, VA, USA
| | - Stacey J Criswell
- Department of Cell Biology, University of Virginia, Charlottesville, VA, USA
| | - Dylan M Hyatt
- School of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Sejin Nam
- Department of Physics, University of Hawaii, Honolulu, HI, USA
| | - Valeria Pearl
- Department of Pediatrics, Division of Nephrology, University of Virginia, Charlottesville, VA, USA
| | | |
Collapse
|
27
|
Ruetzler K, Cohen B, Leung S, Mascha EJ, Knotzer J, Kurz A, Sessler DI, Turan A. Supplemental Intraoperative Oxygen Does Not Promote Acute Kidney Injury or Cardiovascular Complications After Noncardiac Surgery. Anesth Analg 2020; 130:933-940. [DOI: 10.1213/ane.0000000000004359] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
28
|
Abstract
Aim: Supplemental oxygen is often used to treat neonates with respiratory disorders. Human and animal studies have demonstrated that neonatal hyperoxia increases oxidative stress and induces damage and collagen deposition in kidney during the perinatal period. Cathelicidin LL-37 is one important group of human antimicrobial peptides which exhibits antioxidant activity and its overexpression resists hyperoxia-induced oxidative stress. This study was designed to evaluate the protective effects of cathelicidin in hyperoxia-induced kidney injury in newborn rats. Methods: Sprague-Dawley rat pups were reared in either room air (RA) or hyperoxia (85% O2) and were randomly treated with low-dose (4 mg/kg) and high-dose (8 mg/kg) cathelicidin in normal saline (NS) administered intraperitoneally on postnatal days 1–6. The following six groups were obtained: RA + NS, RA + low-dose cathelicidin, RA + high-dose cathelicidin, O2 + NS, O2 + low-dose cathelicidin, and O2 + high-dose cathelicidin. Kidneys were taken for Western blot and histological analyses on postnatal day 7. Results: The hyperoxia-reared rats exhibited significantly lower body weights and anti-inflammatory M2 macrophages, but the kidney injury scores, oxidative stress marker 8-hydroxy-2'-deoxyguanosine (8-OHdG)-positive cells, pro-inflammatory M1 macrophages, collagen deposition, and NF-κB expression were higher than did the RA-reared rats. Conclusions: Cathelicidin treatment attenuated kidney injury as evidenced by lower kidney injury scores, 8-OHdG-positive cells, collagen deposition, and reversion of hyperoxia-induced M1/M2 macrophage polarization. The role of Cathelicidin in ameliorates kidney injury of the hyperoxia newborn rats was accompanied by decreased NF-κB expression, which probably through the modulating NF-κB activity in the kidney.
Collapse
Affiliation(s)
- Hsiu-Chu Chou
- Department of Anatomy and Cellular Biology, School of Medicine, College of Medicine, Taipei Medical University , Taipei , Taiwan
| | - Chung-Ming Chen
- Department of Pediatrics, School of Medicine, College of Medicine, Taipei Medical University , Taipei , Taiwan
| |
Collapse
|
29
|
Mohamed T, Abdul-Hafez A, Gewolb IH, Uhal BD. Oxygen injury in neonates: which is worse? hyperoxia, hypoxia, or alternating hyperoxia/hypoxia. JOURNAL OF LUNG, PULMONARY & RESPIRATORY RESEARCH 2020; 7:4-13. [PMID: 34337150 PMCID: PMC8320601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
Premature birth results in an increased risk of respiratory distress and often requires oxygen therapy. While the supplemental oxygen has been implicated as a cause of bronchopulmonary dysplasia (BPD), in clinical practice this supplementation usually only occurs after the patient's oxygen saturation levels have dropped. The effect of hyperoxia on neonates has been extensively studied. However, there is an unanswered fundamental question: which has the most impact-hyperoxia, hypoxia or fluctuating oxygen levels? In this review, we will summarize the reported effect of hypoxia, hyperoxia or a fluctuation of oxygen levels (hypoxia/hyperoxia cycling) in preterm neonates, with special emphasis on the lungs.
Collapse
Affiliation(s)
- Tarek Mohamed
- Department of Pediatrics and Human Development, Michigan State University, USA
| | - Amal Abdul-Hafez
- Department of Pediatrics and Human Development, Michigan State University, USA
| | - Ira H Gewolb
- Department of Pediatrics and Human Development, Michigan State University, USA
| | - Bruce D Uhal
- Department of Physiology, Michigan State University, USA
| |
Collapse
|
30
|
Proximal Tubular Development Is Impaired with Downregulation of MAPK/ERK Signaling, HIF-1 α, and Catalase by Hyperoxia Exposure in Neonatal Rats. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:9219847. [PMID: 31558952 PMCID: PMC6735195 DOI: 10.1155/2019/9219847] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 06/04/2019] [Indexed: 12/22/2022]
Abstract
Supplemental oxygen therapy (hyperoxia) is a widely used treatment for alveolar hypoxia in preterm infants. Despite being closely monitored, hyperoxia exposure is believed to undermine neonatal nephrogenesis and renal function caused by elevated oxidative stress. Previous studies have mostly focused on the hyperoxia-induced impairment of glomerular development, while the long-term impact of neonatal hyperoxia on tubular development and the regulatory component involved in this process remain to be clarified. Here, we examined tubular histology and apoptosis, along with the expression profile of mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) signaling, hypoxia-inducible factor 1α (HIF-1α), and catalase, following hyperoxia exposure in neonatal rats. Hematoxylin and eosin (H&E) staining revealed the early disappearance of the nephrogenic zone, as well as dilated lumens and reduced epithelial cells, of mature proximal tubules following neonatal hyperoxia. A robust increase in tubular cell apoptosis caused by neonatal hyperoxia was found using a TUNEL assay. Moreover, neonatal hyperoxia altered renal MAPK/ERK signaling activity and downregulated the expression of HIF-1α and catalase in the proximal tubules throughout nephrogenesis from S-shaped bodies to mature proximal tubules. Cell apoptosis in the proximal tubules was positively correlated with HIF-1α expression on the 14th postnatal day. Our data indicates that proximal tubular development is impaired by neonatal hyperoxia, which is accompanied by altered MAPK/ERK signaling as well as downregulated HIF-1α and catalase. Therapeutic management that targets MAPK/ERK signaling, HIF-1α, or catalase may serve as a protective agent against hyperoxia-induced oxidative damage to neonatal proximal tubules.
Collapse
|
31
|
Ischemia-reperfusion injury in renal transplantation: 3 key signaling pathways in tubular epithelial cells. Kidney Int 2019; 95:50-56. [PMID: 30606429 DOI: 10.1016/j.kint.2018.10.009] [Citation(s) in RCA: 107] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 09/25/2018] [Accepted: 10/02/2018] [Indexed: 01/09/2023]
Abstract
Renal ischemia-reperfusion injury (IRI) is a significant clinical challenge faced by clinicians perioperatively in kidney transplantation. Recent work has demonstrated the key importance of transmembrane receptors in the injured tubular epithelial cell, most notably Toll-like receptors, activated by exogenous and endogenous ligands in response to external and internal stresses. Through sequential protein-protein interactions, the signal is relayed deep into the core physiological machinery of the cell, having numerous effects from upregulation of pro-inflammatory gene products through to modulating mitochondrial respiration. Inter-pathway cross talk facilitates a co-ordinated response at an individual cellular level, as well as modulating the surrounding tissue's microenvironment through close interactions with the endothelium and circulating leukocytes. Defining the underlying cellular cascades involved in IRI will assist the identification of novel interventional targets to attenuate IRI with the potential to improve transplantation outcomes. We present a focused review of 3 key cellular signalling pathways in the injured tubular epithelial cell that have been the focus of much research over the past 2 decades: toll-like receptors, sphingosine-1-phosphate receptors and hypoxia inducible factors. We provide a unique perspective on the potential clinical translations of this recent work in the transplant setting. This is particularly timely with the recent completion of phase I and ongoing phase 2 clinical trials of inhibitors targeting specific components of these signaling cascades.
Collapse
|
32
|
Paquette K, Fernandes RO, Xie LF, Cloutier A, Fallaha C, Girard-Bock C, Mian MOR, Lukaszewski MA, Mâsse B, El-Jalbout R, Lapeyraque AL, Santos RA, Luu TM, Nuyt AM. Kidney Size, Renal Function, Ang (Angiotensin) Peptides, and Blood Pressure in Young Adults Born Preterm. Hypertension 2019; 72:918-928. [PMID: 30354721 DOI: 10.1161/hypertensionaha.118.11397] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Preterm birth incurs a higher risk for adult cardiovascular diseases, including hypertension. Because preterm birth may impact nephrogenesis, study objectives were to assess renal size and function of adults born preterm versus full term and to examine their relationship with blood pressure (BP; 24-hour ambulatory BP monitoring) and circulating renin-Ang (angiotensin) system peptides. The study included 92 young adults born (1987-1997) preterm (≤29 weeks of gestation) and term (n=92) matched for age, sex, and race. Young adults born preterm had smaller kidneys (80±17 versus 90±18 cm3/m2; P<0.001), higher urine albumin-to-creatinine ratio (0.70; interquartile range, 0.47-1.14 versus 0.58, interquartile range 0.42 to 0.78 mg/mmol, P=0.007), higher 24-hour systolic (121±9 versus 116±8 mm Hg; P=0.001) and diastolic (69±5 versus 66±6 mm Hg; P=0.004) BP, but similar estimated glomerular filtration rate. BP was inversely correlated with kidney size in preterm participants. Plasma Ang I was higher in preterm versus term participants (36.3; interquartile range, 13.2-62.3 versus 19.4; interquartile range, 9.9-28.1 pg/mL; P<0.001). There was no group difference in renin, Ang II, Ang (1-7), and alamandine. In the preterm, but not in the term group, higher BP was significantly associated with higher renin and alamandine and lower birth weight and gestational age with smaller adult kidney size. Young adults born preterm have smaller kidneys, higher urine albumin-to-creatinine ratio, higher BP, and higher circulating Ang I levels compared with term controls. Preterm young adults with smaller kidneys have higher BP. Clinical Trial Registration- URL: http://www.clinicaltrials.gov . Unique identifier: NCT03261609.
Collapse
Affiliation(s)
- Katryn Paquette
- From the Department of Pediatrics, Sainte-Justine University Hospital Research Center (K.P., R.O.F., L.F.X., A.C., C.F., C.G.-B., M.O.R.M., M.-A.L., A.-L.L., T.M.L., A.M.N.), University of Montreal, Quebec, Canada
| | - Rafael Oliveira Fernandes
- From the Department of Pediatrics, Sainte-Justine University Hospital Research Center (K.P., R.O.F., L.F.X., A.C., C.F., C.G.-B., M.O.R.M., M.-A.L., A.-L.L., T.M.L., A.M.N.), University of Montreal, Quebec, Canada
| | - Li Feng Xie
- From the Department of Pediatrics, Sainte-Justine University Hospital Research Center (K.P., R.O.F., L.F.X., A.C., C.F., C.G.-B., M.O.R.M., M.-A.L., A.-L.L., T.M.L., A.M.N.), University of Montreal, Quebec, Canada
| | - Anik Cloutier
- From the Department of Pediatrics, Sainte-Justine University Hospital Research Center (K.P., R.O.F., L.F.X., A.C., C.F., C.G.-B., M.O.R.M., M.-A.L., A.-L.L., T.M.L., A.M.N.), University of Montreal, Quebec, Canada
| | - Catherine Fallaha
- From the Department of Pediatrics, Sainte-Justine University Hospital Research Center (K.P., R.O.F., L.F.X., A.C., C.F., C.G.-B., M.O.R.M., M.-A.L., A.-L.L., T.M.L., A.M.N.), University of Montreal, Quebec, Canada
| | - Camille Girard-Bock
- From the Department of Pediatrics, Sainte-Justine University Hospital Research Center (K.P., R.O.F., L.F.X., A.C., C.F., C.G.-B., M.O.R.M., M.-A.L., A.-L.L., T.M.L., A.M.N.), University of Montreal, Quebec, Canada
| | - Muhammad Oneeb Rehman Mian
- From the Department of Pediatrics, Sainte-Justine University Hospital Research Center (K.P., R.O.F., L.F.X., A.C., C.F., C.G.-B., M.O.R.M., M.-A.L., A.-L.L., T.M.L., A.M.N.), University of Montreal, Quebec, Canada
| | - Marie-Amélie Lukaszewski
- From the Department of Pediatrics, Sainte-Justine University Hospital Research Center (K.P., R.O.F., L.F.X., A.C., C.F., C.G.-B., M.O.R.M., M.-A.L., A.-L.L., T.M.L., A.M.N.), University of Montreal, Quebec, Canada
| | - Benoit Mâsse
- Department of Social and Preventive Medicine, School of Public Health (B.M.), University of Montreal, Quebec, Canada
| | - Ramy El-Jalbout
- Department of Medical Imaging, Sainte-Justine University Hospital (R.E.-J.), University of Montreal, Quebec, Canada
| | - Anne-Laure Lapeyraque
- From the Department of Pediatrics, Sainte-Justine University Hospital Research Center (K.P., R.O.F., L.F.X., A.C., C.F., C.G.-B., M.O.R.M., M.-A.L., A.-L.L., T.M.L., A.M.N.), University of Montreal, Quebec, Canada
| | - Robson A Santos
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Brazil (R.A.S.)
| | - Thuy Mai Luu
- From the Department of Pediatrics, Sainte-Justine University Hospital Research Center (K.P., R.O.F., L.F.X., A.C., C.F., C.G.-B., M.O.R.M., M.-A.L., A.-L.L., T.M.L., A.M.N.), University of Montreal, Quebec, Canada
| | - Anne Monique Nuyt
- From the Department of Pediatrics, Sainte-Justine University Hospital Research Center (K.P., R.O.F., L.F.X., A.C., C.F., C.G.-B., M.O.R.M., M.-A.L., A.-L.L., T.M.L., A.M.N.), University of Montreal, Quebec, Canada
| |
Collapse
|
33
|
Mohr J, Voggel J, Vohlen C, Dinger K, Dafinger C, Fink G, Göbel H, Liebau MC, Dötsch J, Alejandre Alcazar MA. IL-6/Smad2 signaling mediates acute kidney injury and regeneration in a murine model of neonatal hyperoxia. FASEB J 2019; 33:5887-5902. [PMID: 30721632 DOI: 10.1096/fj.201801875rr] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Prematurity is linked to incomplete nephrogenesis and risk of chronic kidney diseases (CKDs). Oxygen is life-saving in that context but induces injury in numerous organs. Here, we studied the structural and functional impact of hyperoxia on renal injury and its IL-6 dependency. Newborn wild-type (WT) and IL-6 knockout (IL-6-/-) mice were exposed to 85% O2 for 28 d, followed by room air until postnatal d (P) 70. Controls were in room air throughout life. At P28, hyperoxia reduced estimated kidney cortex area (KCA) in WT; at P70, KCA was greater, number of glomeruli was fewer, fractional potassium excretion was higher, and glomerular filtration rate was slightly lower than in controls. IL-6-/- mice were protected from these changes after hyperoxia. Mechanistically, the acute renal injury phase (P28) showed in WT but not in IL-6-/- mice an activation of IL-6 (signal transducer and activator of transcription 3) and TGF-β [mothers against decapentaplegic homolog (Smad)2] signaling, increased inflammatory markers, disrupted mitochondrial biogenesis, and reduced tubular proliferation. Regenerative phase at P70 was characterized by tubular proliferation in WT but not in IL-6-/- mice. These data demonstrate that hyperoxia increases the risk of CKD through a novel IL-6-Smad2 axis. The amenability of these pathways to pharmacological approaches may offer new avenues to protect premature infants from CKD.-Mohr, J., Voggel, J., Vohlen, C., Dinger, K., Dafinger, C., Fink, G., Göbel, H., Liebau, M. C., Dötsch, J., Alejandre Alcazar, M. A. IL-6/Smad2 signaling mediates acute kidney injury and regeneration in a murine model of neonatal hyperoxia.
Collapse
Affiliation(s)
- Jasmine Mohr
- Translational Experimental Pediatrics, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany.,Department of Pediatrics and Adolescent Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany.,Nephrology Research Laboratory, Department of Medicine II, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Jenny Voggel
- Department of Pediatrics and Adolescent Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Christina Vohlen
- Translational Experimental Pediatrics, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany.,Department of Pediatrics and Adolescent Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany.,Nephrology Research Laboratory, Department of Medicine II, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Katharina Dinger
- Translational Experimental Pediatrics, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany.,Department of Pediatrics and Adolescent Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Claudia Dafinger
- Department of Pediatrics and Adolescent Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany.,Nephrology Research Laboratory, Department of Medicine II, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany.,Nephrology Research Laboratory, Department of Medicine II, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Gregor Fink
- Department of Pediatrics and Adolescent Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Heike Göbel
- Institute of Pathology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Max C Liebau
- Department of Pediatrics and Adolescent Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany.,Nephrology Research Laboratory, Department of Medicine II, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany.,Nephrology Research Laboratory, Department of Medicine II, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Jörg Dötsch
- Department of Pediatrics and Adolescent Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Miguel A Alejandre Alcazar
- Translational Experimental Pediatrics, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany.,Department of Pediatrics and Adolescent Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany.,Nephrology Research Laboratory, Department of Medicine II, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| |
Collapse
|
34
|
Li B, Zhu Y, Chen H, Gao H, He H, Zuo N, Pei L, Xie W, Chen L, Ao Y, Wang H. Decreased H3K9ac level of AT2R mediates the developmental origin of glomerulosclerosis induced by prenatal dexamethasone exposure in male offspring rats. Toxicology 2018; 411:32-42. [PMID: 30359671 DOI: 10.1016/j.tox.2018.10.013] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 10/04/2018] [Accepted: 10/20/2018] [Indexed: 12/11/2022]
Abstract
This study aimed to demonstrate that prenatal dexamethasone exposure (PDE) can induce kidney dysplasia in utero and adult glomerulosclerosis in male offspring, and to explore the underlying intrauterine programming mechanisms. Pregnant rats were subcutaneously administered dexamethasone 0.2 mg/kg.d from gestational day (GD) 9 to GD20. The male fetus on GD20 and the adult offspring at age of postnatal week 28 were analyzed. The adult offspring kidneys in the PDE group displayed glomerulosclerosis, elevated levels of serum creatinine and urine protein, ultrastructural damage of podocytes, the reduced expression levels of podocyte marker genes, nephrin and podocin. The histone 3 lysine 9 acetylation (H3K9ac) level in the promoter of renal angiotensin II receptor type 2 (AT2R) and its expression were reduced, whereas the angiotensin II receptor type 1a (AT1aR)/AT2R expression ratio was increased. The fetal kidneys in the PDE group displayed an enlarged Bowman's space and a shrunken glomerular tuft, a reduced cortex width and an increase in the nephrogenic zone/cortical zone ratio, reduced the expression level of glial-cell-line derived neurotrophic factor/c-Ret tyrosine kinase receptor (GDNF/c-Ret) signal pathway and podocyte marker genes. Moreover, the H3K9ac and H3K27ac levels of AT2R as well as the gene and protein expression levels of AT2R in fetal kidneys were inhibited by PDE. In vitro, primary metanephric mesenchyme stem cells (MMSCs) were treated with dexamethasone. Overexpression of AT2R reversed the inhibited expression of GDNF/c-Ret and podocin/nephrin induced by dexamethasone, and glucocorticoids receptor antagonist abolished the decreased H3K9ac level and gene expression of AT2R. In conclusion, PDE induced the offspring's kidney dysplasia as well as adult glomerulosclerosis, which was mediated by a sustained decrease in renal AT2R expression via decreasing the H3 K9ac level.
Collapse
Affiliation(s)
- Bin Li
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan, 430071, China; Department of Orthopaedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Yanan Zhu
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan, 430071, China
| | - Haiyun Chen
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan, 430071, China
| | - Hui Gao
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan, 430071, China; Department of Orthopaedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Hangyuan He
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan, 430071, China; Department of Orthopaedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Na Zuo
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan, 430071, China
| | - Linguo Pei
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan, 430071, China
| | - Wen Xie
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Liaobin Chen
- Department of Orthopaedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China
| | - Ying Ao
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan, 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China.
| | - Hui Wang
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan, 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China.
| |
Collapse
|
35
|
Time Dependent Pathway Activation of Signalling Cascades in Rat Organs after Short-Term Hyperoxia. Int J Mol Sci 2018; 19:ijms19071960. [PMID: 29973540 PMCID: PMC6073502 DOI: 10.3390/ijms19071960] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 06/22/2018] [Accepted: 06/29/2018] [Indexed: 01/04/2023] Open
Abstract
Administration of oxygen is one of the most common interventions in medicine. Previous research showed that differential regulated proteins could be linked to hyperoxia-associated signaling cascades in different tissues. However, it still remains unclear which signaling pathways are activated by hyperoxia. The present study analyses hyperoxia-induced protein alterations in lung, brain, and kidney tissue using a proteomic and bioinformatic approach. Pooled data of 36 Wistar rats exposed to hyperoxia were used. To identify possible hyperoxia biomarkers, and to evaluate the relationship between protein alterations in hyperoxia affected organs and blood, proteomics data from brain, lung, and kidney were analyzed. Functional network analyses (IPA®, PathwaysStudio®, and GENEmania®) in combination with hierarchical cluster analysis (Perseus®) was used to identify relevant pathways and key proteins. Data of 54 2D-gels with more than 2500 significantly regulated spots per gel were collected. Thirty-eight differentially expressed proteins were identified and consecutively analyzed by bioinformatic methods. Most differences between hyperoxia and normoxia (21 proteins up-regulated, 17 proteins down-regulated) were found immediately after hyperoxia (15 protein spots), followed by day 3 (13 spots), and day 7 (10 spots). A highly significant association with inflammation and the inflammatory response was found. Cell proliferation, oxidative stress, apoptosis and cell death as well as cellular functions were revealed to be affected. Three hours of hyperoxia resulted in significant alterations of protein expression in different organs (brain, lung, kidney) up to seven days after exposure. Further studies are required to interpret the relevance of protein alterations in signaling cascades during/after hyperoxia.
Collapse
|
36
|
Iwuagwu BU. Regulation of HIF1 alpha in hyperoxia and hyperglycaemia. FASEB J 2018. [DOI: 10.1096/fasebj.2018.32.1_supplement.670.54] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
37
|
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
|
38
|
Kumar VH, Wang H, Kishkurno S, Paturi BS, Nielsen L, Ryan RM. Long-Term Effects of Neonatal Hyperoxia in Adult Mice. Anat Rec (Hoboken) 2018; 301:717-726. [DOI: 10.1002/ar.23766] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 08/02/2017] [Accepted: 10/04/2017] [Indexed: 12/22/2022]
Affiliation(s)
| | - Huamei Wang
- Department of Pediatrics; University at Buffalo; Buffalo New York
| | - Sergei Kishkurno
- Department of Pediatrics; University at Buffalo; Buffalo New York
| | - Babu S Paturi
- Department of Pediatrics; University at Buffalo; Buffalo New York
| | - Lori Nielsen
- Department of Pediatrics; University at Buffalo; Buffalo New York
| | - Rita M. Ryan
- Department of Pediatrics; Medical University of South Carolina; Charleston South Carolina
| |
Collapse
|
39
|
Wurm S, Steege A, Rom-Jurek EM, van Roeyen CR, Kurtz A, Banas B, Banas MC. CCR7 Is Important for Mesangial Cell Physiology and Repair. J Histochem Cytochem 2017; 66:7-22. [PMID: 29077526 DOI: 10.1369/0022155417737975] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The homeostatic chemokine receptor CCR7 serves as key molecule in lymphocyte homing into secondary lymphoid tissues. Previous experiments from our group identified CCR7 also to be expressed by human mesangial cells. Exposing cultured human mesangial cells to the receptor ligand CCL21 revealed a positive effect on these cells regarding proliferation, migration, and survival. In the present study, we localized CCR7 and CCL21 during murine nephrogenesis. Analyzing wild-type and CCR7 deficient (CCR7-/-) mice, we observed a retarded glomerulogenesis during renal development and a significantly decreased mesangial cellularity in adult CCR7-/- mice, as a consequence of less mesangial cell proliferation between embryonic day E17.5 and week 5 postpartum. Cell proliferation assays and cell-wounding experiments confirmed reduced proliferative and migratory properties of mesangial cells cultured from CCR7-/- kidneys. To further emphasize the role of CCR7 as important factor for mesangial biology, we examined the chemokine receptor expression in rats after induction of a mesangioproliferative glomerulonephritis. Here, we demonstrated for the first time that extra- and intraglomerular mesangial cells that were CCR7-negative in control rats exhibited a strong CCR7 expression during the phase of mesangial repopulation and proliferation.
Collapse
Affiliation(s)
- Simone Wurm
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - Andreas Steege
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - Eva-Maria Rom-Jurek
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany.,Department of Gynaecology and Obstetrics, University Hospital Regensburg, Regensburg, Germany
| | - Claudia R van Roeyen
- Division of Nephrology and Immunology, Rheinisch-Westfälische Technische Hochschule Aachen University, Aachen, Germany
| | - Armin Kurtz
- Institute of Physiology, University of Regensburg, Regensburg, Germany
| | - Bernhard Banas
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - Miriam C Banas
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
40
|
Effects of preterm birth and ventilation on glomerular capillary growth in the neonatal lamb kidney. J Hypertens 2017; 34:1988-97. [PMID: 27428042 DOI: 10.1097/hjh.0000000000001028] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
OBJECTIVES Preterm birth is linked to the development of hypertension later in life. This may relate to impaired glomerular capillary growth following preterm birth. The aim of this study was to determine the effects of preterm birth, and/or ventilation, on glomerular capillary growth in the neonatal lamb kidney. METHODS Four experimental groups were analysed: preterm lambs delivered at 130 days gestation (term = 147 days) and mechanically ventilated for 3 days (preterm ventilated: n = 9), 133 days gestational controls (gestational control: n = 5), term controls, unassisted breathing for 3 days (term control: n = 8), and term lambs ventilated for 3 days (term ventilated: n = 5). In perfusion-fixed kidneys, total nephron number, average total capillary length, and surface area per renal corpuscle were stereologically assessed, and total renal filtration surface area (TRFSA) was calculated. RESULTS In comparison with term controls, preterm lambs had significantly reduced glomerular capillary length, surface area, and TRFSA, indicative of a low renal functional capacity. Term-ventilated lambs exhibited significantly reduced glomerular capillary length and surface area compared with term controls, indicating that ventilation impairs glomerular capillary growth independently of preterm birth. CONCLUSION Impaired glomerular capillary growth and subsequent reduced TRFSA following preterm birth may mediate the increased predisposition to hypertension later in life.
Collapse
|
41
|
Nakagawa M, Nishizaki N, Endo A, Someya T, Saito Y, Mizutani A, Hara T, Murano Y, Sakuraya K, Hara S, Umino D, Hirano D, Fujinaga S, Ohtomo Y, Shimizu T. Impaired nephrogenesis in neonatal rats with oxygen-induced retinopathy. Pediatr Int 2017; 59:704-710. [PMID: 28207964 DOI: 10.1111/ped.13264] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 02/10/2017] [Accepted: 02/13/2017] [Indexed: 12/20/2022]
Abstract
BACKGROUND Preterm neonates are born while nephrogenesis is ongoing, and are commonly exposed to factors in a hyperoxic environment that can impair renal development. Oxidative stress has also been implicated in the development of retinopathy of prematurity (ROP). The rat model of oxygen-induced retinopathy (OIR) is the most clinically relevant model of ROP because its biologic features closely resemble those of ROP in preterm infants. We investigated impaired renal development in a rat model of OIR. METHODS Newborn Sprague-Dawley rats were maintained in either a normoxic (room air, 21% O2 ; control group) or a controlled hyperoxic (80% O2 ; OIR group) environment from birth to postnatal day (P) 12. All pups were then raised in room air from P12 to P19. RESULTS The hyperoxic environment led to significantly higher urinary excretion of 8-hydroxy-2'-deoxyguanosine, a marker of oxidative DNA damage, and a reduction in nephrogenic zone width at P5 in OIR pups. Additionally, glomerular count was significantly reduced by 20% in the OIR group, and avascular and neovascular changes in the retina were observed only in the OIR group at P19. Messenger RNA levels of vascular endothelial growth factor-A (VEGF-A) and platelet-derived growth factor-β, essential angiogenic cytokines for glomerulogenesis, in the renal cortex were significantly lower at P5 and significantly higher at P19 in the OIR group compared with controls. CONCLUSION Renal impairment was caused by exposure to a hyperoxic environment during nephrogenesis, and the pathology of the impaired nephrogenesis in this OIR model reflects the characteristics of ROP observed in preterm infants.
Collapse
Affiliation(s)
- Mayu Nakagawa
- Department of Pediatrics and Adolescent Medicine, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Naoto Nishizaki
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine, Juntendo University, Tokyo, Japan
| | - Amane Endo
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine, Juntendo University, Tokyo, Japan
| | - Tomonosuke Someya
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine, Juntendo University, Tokyo, Japan
| | - Yuta Saito
- Department of Ophthalmology, Showa University School of Medicine, Tokyo, Japan
| | - Akira Mizutani
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine, Juntendo University, Tokyo, Japan
| | - Taichi Hara
- Department of Pediatrics and Adolescent Medicine, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Yayoi Murano
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine, Juntendo University, Tokyo, Japan
| | - Koji Sakuraya
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine, Juntendo University, Tokyo, Japan
| | - Satoshi Hara
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine, Juntendo University, Tokyo, Japan
| | - Daisuke Umino
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine, Juntendo University, Tokyo, Japan
| | - Daishi Hirano
- Department of Pediatrics, Jikei University School of Medicine, Tokyo, Japan
| | - Shuichiro Fujinaga
- Department of Nephrology, Saitama Children's Medical Center, Saitama, Japan
| | - Yoshiyuki Ohtomo
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine, Juntendo University, Tokyo, Japan
| | - Toshiaki Shimizu
- Department of Pediatrics and Adolescent Medicine, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| |
Collapse
|
42
|
Nuyt AM, Lavoie JC, Mohamed I, Paquette K, Luu TM. Adult Consequences of Extremely Preterm Birth: Cardiovascular and Metabolic Diseases Risk Factors, Mechanisms, and Prevention Avenues. Clin Perinatol 2017; 44:315-332. [PMID: 28477663 DOI: 10.1016/j.clp.2017.01.010] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Extremely preterm babies are exposed to various sources of injury during critical stages of development. The extremely preterm infant faces premature transition to ex utero physiology and undergoes adaptive mechanisms that may be deleterious in the long term because of permanent alterations in organ structure and function. Perinatal events can also directly cause structural injury. These disturbances induce morphologic and functional changes in their organ systems that might heighten their risks for later adult chronic diseases. This review examines the pathophysiology of programming of long-term health and diseases after preterm birth and associated perinatal risk factors.
Collapse
Affiliation(s)
- Anne Monique Nuyt
- Division of Neonatology, Department of Pediatrics, Faculty of Medicine, Research Center, Centre Hospitalier Universitaire Sainte-Justine, Université de Montréal, 3175 chemin de la Côte-Sainte-Catherine, Montreal, Quebec H3T 1C5, Canada.
| | - Jean-Claude Lavoie
- Division of Neonatology, Department of Pediatrics, Faculty of Medicine, Research Center, Centre Hospitalier Universitaire Sainte-Justine, Université de Montréal, 3175 chemin de la Côte-Sainte-Catherine, Montreal, Quebec H3T 1C5, Canada; Department of Nutrition, Faculty of Medicine, Research Center, Centre Hospitalier Universitaire Sainte-Justine, Université de Montréal, 3175 chemin de la Côte-Sainte-Catherine, Montreal, Quebec H3T 1C5, Canada
| | - Ibrahim Mohamed
- Division of Neonatology, Department of Pediatrics, Faculty of Medicine, Research Center, Centre Hospitalier Universitaire Sainte-Justine, Université de Montréal, 3175 chemin de la Côte-Sainte-Catherine, Montreal, Quebec H3T 1C5, Canada
| | - Katryn Paquette
- Division of Neonatology, Department of Pediatrics, Faculty of Medicine, Research Center, Centre Hospitalier Universitaire Sainte-Justine, Université de Montréal, 3175 chemin de la Côte-Sainte-Catherine, Montreal, Quebec H3T 1C5, Canada
| | - Thuy Mai Luu
- Division of General Pediatrics, Department of Pediatrics, Faculty of Medicine, Research Center, Centre Hospitalier Universitaire Sainte-Justine, Université de Montréal, 3175 chemin de la Côte-Sainte-Catherine, Montreal, Quebec H3T 1C5, Canada
| |
Collapse
|
43
|
Sutherland MR, Béland C, Lukaszewski MA, Cloutier A, Bertagnolli M, Nuyt AM. Age- and sex-related changes in rat renal function and pathology following neonatal hyperoxia exposure. Physiol Rep 2017; 4:4/15/e12887. [PMID: 27528005 PMCID: PMC4985552 DOI: 10.14814/phy2.12887] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2016] [Accepted: 07/18/2016] [Indexed: 12/04/2022] Open
Abstract
Preterm neonates are prematurely exposed to high oxygen levels at birth which may adversely impact ongoing renal development. The aim of this study was to determine the effects of neonatal hyperoxia exposure on renal function and morphology with aging. Sprague Dawley rat pups were raised in a hyperoxic environment (80% oxygen) from P3 to P10 during ongoing postnatal nephrogenesis. Control litters were kept in room air (n = 6–8 litters/group; one male, one female/litter/age). Kidney function (urine and plasma creatinine, sodium, and protein) and morphology (renal corpuscle size, glomerulosclerosis, fibrosis, and glomerular crescents) were assessed at 1, 5, and 11 months of age. Neonatal hyperoxia exposure had no impact on body or kidney weights. Creatinine clearance was significantly reduced following hyperoxia exposure at 5 months; there was no significant effect on renal function at 1 or 11 months. The percentage of crescentic glomeruli (indicative of glomerular injury) was markedly increased in 11 month hyperoxia‐exposed males. Renal corpuscle size, glomerulosclerosis index, and renal fibrosis were not affected. Findings suggest that exposure to high oxygen levels during development may impact renal functional capacity and increase susceptibility to renal disease in adulthood depending on age and sex.
Collapse
Affiliation(s)
- Megan R Sutherland
- Sainte-Justine University Hospital and Research Center, Montreal, Quebec, Canada Department of Pediatrics, Université de Montréal, Montreal, Quebec, Canada
| | - Chanel Béland
- Sainte-Justine University Hospital and Research Center, Montreal, Quebec, Canada Department of Pediatrics, Université de Montréal, Montreal, Quebec, Canada
| | - Marie-Amélie Lukaszewski
- Sainte-Justine University Hospital and Research Center, Montreal, Quebec, Canada Department of Pediatrics, Université de Montréal, Montreal, Quebec, Canada
| | - Anik Cloutier
- Sainte-Justine University Hospital and Research Center, Montreal, Quebec, Canada Department of Pediatrics, Université de Montréal, Montreal, Quebec, Canada
| | - Mariane Bertagnolli
- Sainte-Justine University Hospital and Research Center, Montreal, Quebec, Canada Department of Pediatrics, Université de Montréal, Montreal, Quebec, Canada
| | - Anne Monique Nuyt
- Sainte-Justine University Hospital and Research Center, Montreal, Quebec, Canada Department of Pediatrics, Université de Montréal, Montreal, Quebec, Canada
| |
Collapse
|
44
|
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
|
45
|
Low functional programming of renal AT 2 R mediates the developmental origin of glomerulosclerosis in adult offspring induced by prenatal caffeine exposure. Toxicol Appl Pharmacol 2015; 287:128-138. [DOI: 10.1016/j.taap.2015.05.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 04/05/2015] [Accepted: 05/07/2015] [Indexed: 12/22/2022]
|
46
|
Wang HH. Oxidative Stress and Potential Renal Damage in Neonates. Pediatr Neonatol 2015; 56:209-10. [PMID: 25910519 DOI: 10.1016/j.pedneo.2015.03.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 03/17/2015] [Indexed: 11/29/2022] Open
Affiliation(s)
- Hsin-Hui Wang
- Department of Pediatrics, Division of Pediatric Immunology and Nephrology, Taipei Veterans General Hospital, Taipei, Taiwan; Department of Pediatrics, Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan; Institute of Emergency and Critical Care Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan.
| |
Collapse
|
47
|
Schley G, Scholz H, Kraus A, Hackenbeck T, Klanke B, Willam C, Wiesener MS, Heinze E, Burzlaff N, Eckardt KU, Buchholz B. Hypoxia inhibits nephrogenesis through paracrine Vegfa despite the ability to enhance tubulogenesis. Kidney Int 2015. [PMID: 26200943 DOI: 10.1038/ki.2015.214] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Reduced nephron number predisposes to hypertension and kidney disease. Interaction of the branching ureteric bud and surrounding mesenchymal cells determines nephron number. Since oxygen supply may be critical for intrauterine development, we tested whether hypoxia and hypoxia-inducible factor-1α (HIF-1α) influence nephrogenesis. We found that HIF-1α is required for branching of MDCK cells. In addition, culture of metanephric mouse kidneys with ureteric bud cell-specific stabilization or knockout of HIF-1α revealed a positive impact of HIF-1α on nephrogenesis. In contrast, widespread stabilization of HIF-1α in metanephric kidneys through hypoxia or HIF stabilizers impaired nephrogenesis, and pharmacological HIF inhibition enhanced nephrogenesis. Several lines of evidence suggest an inhibitory effect through the hypoxia response of mesenchymal cells. HIF-1α was expressed in mesenchymal cells during nephrogenesis. Expression of the anti-branching factors Bmp4 and Vegfa, secreted by mesenchymal cells, was increased upon HIF stabilization. The conditioned medium from hypoxic metanephric kidneys inhibited MDCK branching, which was partially rescued by Vegfa antibodies. Thus, the effect of HIF-1α on nephrogenesis appears context dependent. While HIF-1α in the ureteric bud is of importance for proper branching morphogenesis, the net effect of hypoxia-induced HIF activation in the embryonic kidney appears to be mesenchymal cell-dependent inhibition of ureter branching.
Collapse
Affiliation(s)
- Gunnar Schley
- Department of Nephrology and Hypertension, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Holger Scholz
- Department of Physiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Andre Kraus
- Department of Nephrology and Hypertension, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Thomas Hackenbeck
- Department of Nephrology and Hypertension, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Bernd Klanke
- Department of Nephrology and Hypertension, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Carsten Willam
- Department of Nephrology and Hypertension, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Michael S Wiesener
- Department of Nephrology and Hypertension, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Eva Heinze
- Department of Chemistry and Pharmacy, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Nicolai Burzlaff
- Department of Chemistry and Pharmacy, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Kai-Uwe Eckardt
- Department of Nephrology and Hypertension, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Bjoern Buchholz
- Department of Nephrology and Hypertension, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| |
Collapse
|
48
|
Hyperoxia-Induced Protein Alterations in Renal Rat Tissue: A Quantitative Proteomic Approach to Identify Hyperoxia-Induced Effects in Cellular Signaling Pathways. DISEASE MARKERS 2015; 2015:964263. [PMID: 26106253 PMCID: PMC4461769 DOI: 10.1155/2015/964263] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/01/2015] [Revised: 04/04/2015] [Accepted: 04/20/2015] [Indexed: 12/15/2022]
Abstract
Introduction. In renal tissue as well as in other organs, supranormal oxygen pressure may lead to deleterious consequences on a cellular level. Additionally, hyperoxia-induced effect in cells and related free radicals may potentially contribute to renal failure. The aim of this study was to analyze time-dependent alterations of rat kidney protein expression after short-term normobaric hyperoxia using proteomics and bioinformatic approaches. Material and Methods. N = 36 Wistar rats were randomized into six different groups: three groups with normobaric hyperoxia (exposure to 100% oxygen for 3 h) and three groups with normobaric normoxia (NN; room air). After hyperoxia exposure, kidneys were removed immediately, after 3 days and after 7 days. Kidney lysates were analyzed by two-dimensional gel electrophoresis followed by peptide mass fingerprinting using tandem mass spectrometry. Statistical analysis was performed with DeCyder 2D software (p < 0.01). Biological functions of differential regulated proteins were studied using functional network analysis (Ingenuity Pathways Analysis and PathwayStudio). Results. Expression of 14 proteins was significantly altered (p < 0.01): eight proteins (MEP1A_RAT, RSSA_RAT, F16P1_RAT, STML2_RAT, BPNT1_RAT, LGMN_RAT, ATPA_RAT, and VDAC1_RAT) were downregulated and six proteins (MTUS1_RAT, F16P1_RAT, ACTG_RAT, ACTB_RAT, 2ABA_RAT, and RAB1A_RAT) were upregulated. Bioinformatic analyses revealed an association of regulated proteins with inflammation. Conclusions. Significant alterations in renal protein expression could be demonstrated for up to 7 days even after short-term hyperoxia. The identified proteins indicate an association with inflammation signaling cascades. MEP1A and VDAC1 could be promising candidates to identify hyperoxic injury in kidney cells.
Collapse
|
49
|
Sun Z, Hu S, Zuo N, Yang S, He Z, Ao Y, Wang H. Prenatal nicotine exposure induced GDNF/c-Ret pathway repression-related fetal renal dysplasia and adult glomerulosclerosis in male offspring. Toxicol Res (Camb) 2015. [DOI: 10.1039/c5tx00040h] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Prenatal nicotine exposure could induce fetal renal dysplasia associated with the suppression of the GDNF/c-Ret pathway and adult glomerulosclerosis in male offspring, which might be mediated by alterations in angiotensin II receptors.
Collapse
Affiliation(s)
- Zhaoxia Sun
- Department of Pharmacology
- Wuhan University School of Basic Medical Sciences
- Wuhan 430071
- China
| | - Shuangshuang Hu
- Department of Pharmacology
- Wuhan University School of Basic Medical Sciences
- Wuhan 430071
- China
| | - Na Zuo
- Department of Pharmacology
- Wuhan University School of Basic Medical Sciences
- Wuhan 430071
- China
| | - Shuailong Yang
- Department of Pharmacology
- Wuhan University School of Basic Medical Sciences
- Wuhan 430071
- China
| | - Zheng He
- Department of Pharmacology
- Wuhan University School of Basic Medical Sciences
- Wuhan 430071
- China
| | - Ying Ao
- Department of Pharmacology
- Wuhan University School of Basic Medical Sciences
- Wuhan 430071
- China
- Hubei Provincial Key Laboratory of Developmentally Originated Disorder
| | - Hui Wang
- Department of Pharmacology
- Wuhan University School of Basic Medical Sciences
- Wuhan 430071
- China
- Hubei Provincial Key Laboratory of Developmentally Originated Disorder
| |
Collapse
|
50
|
Davies J, Karmouty-Quintana H, Le TT, Chen NY, Weng T, Luo F, Molina J, Moorthy B, Blackburn MR. Adenosine promotes vascular barrier function in hyperoxic lung injury. Physiol Rep 2014; 2:2/9/e12155. [PMID: 25263205 PMCID: PMC4270235 DOI: 10.14814/phy2.12155] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Hyperoxic lung injury is characterized by cellular damage from high oxygen concentrations that lead to an inflammatory response in the lung with cellular infiltration and pulmonary edema. Adenosine is a signaling molecule that is generated extracellularly by CD73 in response to injury. Extracellular adenosine signals through cell surface receptors and has been found to be elevated and plays a protective role in acute injury situations. In particular, ADORA2B activation is protective in acute lung injury. However, little is known about the role of adenosine signaling in hyperoxic lung injury. We hypothesized that hyperoxia-induced lung injury leads to CD73-mediated increases in extracellular adenosine, which is protective through ADORA2B signaling pathways. To test this hypothesis, we exposed C57BL6, CD73(-/-), and Adora2B(-/-) mice to 95% oxygen or room air and examined markers of pulmonary inflammation, edema, and monitored lung histology. Hyperoxic exposure caused pulmonary inflammation and edema in association with elevations in lung adenosine levels. Loss of CD73-mediated extracellular adenosine production exacerbated pulmonary edema without affecting inflammatory cell counts. Furthermore, loss of the ADORA2B had similar results with worsening of pulmonary edema following hyperoxia exposure without affecting inflammatory cell infiltration. This loss of barrier function correlated with a decrease in occludin in pulmonary vasculature in CD73(-/-) and Adora2B(-/-) mice following hyperoxia exposure. These results demonstrate that exposure to a hyperoxic environment causes lung injury associated with an increase in adenosine concentration, and elevated adenosine levels protect vascular barrier function in hyperoxic lung injury through the ADORA2B-dependent regulation of occludin.
Collapse
Affiliation(s)
- Jonathan Davies
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Harry Karmouty-Quintana
- Department of Biochemistry and Molecular Biology, The University of Texas - Houston Medical School, Houston, Texas
| | - Thuy T Le
- Department of Biochemistry and Molecular Biology, The University of Texas - Houston Medical School, Houston, Texas
| | - Ning-Yuan Chen
- Department of Biochemistry and Molecular Biology, The University of Texas - Houston Medical School, Houston, Texas
| | - Tingting Weng
- Department of Biochemistry and Molecular Biology, The University of Texas - Houston Medical School, Houston, Texas
| | - Fayong Luo
- Department of Biochemistry and Molecular Biology, The University of Texas - Houston Medical School, Houston, Texas
| | - Jose Molina
- Department of Biochemistry and Molecular Biology, The University of Texas - Houston Medical School, Houston, Texas
| | - Bhagavatula Moorthy
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Michael R Blackburn
- Department of Biochemistry and Molecular Biology, The University of Texas - Houston Medical School, Houston, Texas
| |
Collapse
|