1
|
Wu W, Zhang B, Zhao J, Hu W, Li Y, Feng Y, Zhang Y, Wang S. Relationship between apoptosis gene DNA methylation and fetal growth and development. Gene 2025; 941:149224. [PMID: 39788353 DOI: 10.1016/j.gene.2025.149224] [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: 10/22/2024] [Revised: 12/14/2024] [Accepted: 01/06/2025] [Indexed: 01/12/2025]
Abstract
OBJECTIVE To investigate the relationship between DNA methylation of cord blood apoptosis genes and low birth weight (LBW). METHODS A case-control study was conducted on 50 pairs of LBW neonates and normal birth weight. Genome-wide methylation assay was performed using Illumina Human Methylation EPIC microarray to analyze the methylation sites of apoptosis-related genes BCL-2, CASP3, and CASP8. The mRNA level of apoptosis gene was verified by RT-qPCR. RESULTS Three CpG sites of BCL-2 and three CpG sites of CASP3 were different between the LBW and NBW groups. Logistic regression showed that higher methylation of BCL-2 CpG sites cg12459502 (OR = 1.208, 95% CI:1.029, 1.418) and cg25059899 (OR = 1.193, 95% CI:1.019, 1.395) increased LBW risk, while cg22152050 was protective (OR = 0.589, 95% CI:0.424, 0.820). Stratified analysis confirmed this. Maternal pre-pregnancy BMI positively correlated with BCL-2 methylation (cg12459502, cg25059899) (b = 0.431, 95% CI: 0.027, 0.835; b = 0.494, 95% CI: 0.141, 0.848), while excessive pregnancy weight gain negatively correlated with cg12459502 methylation (b = -0.269, 95% CI: -0.525, -0.013). The results showed that the mRNA level of BCL-2 in NBW group was significantly higher than that in LBW group (P-value < 0.0001). CONCLUSION The DNA methylation levels of BCL-2 and CASP3 genes are associated with fetal growth and development. Additionally, maternal pre-pregnancy BMI and weight gain during pregnancy were found to correlate with BCL-2 methylation, indicating potential epigenetic mechanisms influencing fetal growth.
Collapse
Affiliation(s)
- Weiwei Wu
- Department of Epidemiology, School of Public Health, Center of Clinical Epidemiology and Evidence Based Medicine, Shanxi Medical University, Taiyuan, China; MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Shanxi Medical University, Taiyuan, Shanxi, China.
| | - Bole Zhang
- Department of Epidemiology, School of Public Health, Center of Clinical Epidemiology and Evidence Based Medicine, Shanxi Medical University, Taiyuan, China; MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jing Zhao
- Department of Epidemiology, School of Public Health, Center of Clinical Epidemiology and Evidence Based Medicine, Shanxi Medical University, Taiyuan, China; MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Weixuan Hu
- Department of Epidemiology, School of Public Health, Center of Clinical Epidemiology and Evidence Based Medicine, Shanxi Medical University, Taiyuan, China; MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yulin Li
- Department of Epidemiology, School of Public Health, Center of Clinical Epidemiology and Evidence Based Medicine, Shanxi Medical University, Taiyuan, China; MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yongliang Feng
- Department of Epidemiology, School of Public Health, Center of Clinical Epidemiology and Evidence Based Medicine, Shanxi Medical University, Taiyuan, China; MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yawei Zhang
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Suping Wang
- Department of Epidemiology, School of Public Health, Center of Clinical Epidemiology and Evidence Based Medicine, Shanxi Medical University, Taiyuan, China; MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Shanxi Medical University, Taiyuan, Shanxi, China.
| |
Collapse
|
2
|
Huang M, Zeng X, Dai Z, Huang Y, Luo C, Tan X, Jiang Z, Fang X, Xu Y. Association between early exposure to famine and risk of renal impairment in adulthood: a systematic review and meta-analysis. Nutr Diabetes 2024; 14:84. [PMID: 39384564 PMCID: PMC11464504 DOI: 10.1038/s41387-024-00342-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 09/10/2024] [Accepted: 09/20/2024] [Indexed: 10/11/2024] Open
Abstract
Malnutrition early in life increases the later-life risk of noncommunicable diseases, and previous epidemiologic studies have found a link between famine and renal impairment, but no consensus has been reached. This meta-analysis and systematic review were conducted to assess the correlation between early-life famine exposure and the risk of developing renal impairment. Search in Embase, Scopus, Web of Science, PubMed, and Cochrane using keywords that report the correlation between early famine exposure and renal function indicators. RevMan and Stata software were used for data analysis. This meta-analysis contained twelve observational studies. The findings demonstrated a link between prenatal famine exposure and a higher risk of developing chronic kidney disease (CKD) (odds ratio (OR) = 1.73, 95% confidence interval (CI): 1.25, 2.39), a decreased estimated glomerular filtration rate (eGFR) (mean difference (MD) = -10.05, 95% CI: -11.64, -8.46), and increased serum creatinine (Scr) (MD = 0.02, 95% CI: 0.01, 0.03) compared to unexposed individuals. Famine exposure in childhood was associated with decreased eGFR (MD = -9.43, 95% CI: -12.01, -6.84) and increased Scr (MD = 0.03, 95% CI: 0.01, 0.04), but not with CKD (OR = 0.980, 95% CI: 0.53, 1.81). Famine exposure in adolescence and adulthood was associated with decreased eGFR (MD = -20.73, 95% CI: -22.40, -19.06). Evidence certainty was deemed to be of low or extremely low quality. Famine exposure early in life could pose a greater risk of developing renal impairment in adulthood, but this outcome may be driven by uncontrolled age differences between famine-births and post-famine-births (unexposed).
Collapse
Affiliation(s)
- Mengting Huang
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Southwest Medical University, Luzhou, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, China
- Sichuan Clinical Research Center for Diabetes and Metabolic Diseases, Luzhou, Sichuan, China
| | - Xin Zeng
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Southwest Medical University, Luzhou, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, China
- Sichuan Clinical Research Center for Diabetes and Metabolic Diseases, Luzhou, Sichuan, China
| | - Zhuojun Dai
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Southwest Medical University, Luzhou, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, China
- Sichuan Clinical Research Center for Diabetes and Metabolic Diseases, Luzhou, Sichuan, China
| | - Yuqing Huang
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Southwest Medical University, Luzhou, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, China
- Sichuan Clinical Research Center for Diabetes and Metabolic Diseases, Luzhou, Sichuan, China
| | - Changfang Luo
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Southwest Medical University, Luzhou, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, China
- Sichuan Clinical Research Center for Diabetes and Metabolic Diseases, Luzhou, Sichuan, China
| | - Xiaozhen Tan
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Southwest Medical University, Luzhou, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, China
- Sichuan Clinical Research Center for Diabetes and Metabolic Diseases, Luzhou, Sichuan, China
| | - Zongzhe Jiang
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Southwest Medical University, Luzhou, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, China
- Sichuan Clinical Research Center for Diabetes and Metabolic Diseases, Luzhou, Sichuan, China
| | - Xia Fang
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Southwest Medical University, Luzhou, China.
- Sichuan Clinical Research Center for Nephropathy, Luzhou, China.
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, China.
- Sichuan Clinical Research Center for Diabetes and Metabolic Diseases, Luzhou, Sichuan, China.
| | - Yong Xu
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Southwest Medical University, Luzhou, China.
- Sichuan Clinical Research Center for Nephropathy, Luzhou, China.
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, China.
- Sichuan Clinical Research Center for Diabetes and Metabolic Diseases, Luzhou, Sichuan, China.
| |
Collapse
|
3
|
Xiong H, Liu D, Tang D, Ma F. Exposure to Chinese famine and the risk of hyperuricemia in later life: a population-based cross-sectional study. Front Nutr 2024; 11:1266817. [PMID: 38298423 PMCID: PMC10828035 DOI: 10.3389/fnut.2024.1266817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 01/02/2024] [Indexed: 02/02/2024] Open
Abstract
Background Limited studies have investigated the relationship between famine exposure and the risk of hyperuricemia in later life. Consequently, the primary purpose of the current study was to examine the potential association between exposure to Chinese famine and hyperuricemia, as well as any gender disparities in this relationship. Method The data were obtained from the China PEACE (China Patient-Centered Evaluative Assessment of Cardiac Events) Million Persons Project in Rongchang. The study participants were enrolled into different cohorts based on their birthdates: the fetal-exposed cohort (born between 1959 and 1962), the childhood-exposed cohort (born between 1949 and 1958), the adolescence-exposed cohort (born between 1941 and 1948), and the non-exposed cohorts (born between 1963 and 1974). The potential association between famine exposure and hyperuricemia was assessed using binary logistic regression models. Results A total of 6,916 individuals were enrolled in the current study with an average age of 60.11 ± 9.22 years, out of which 3,544 were women. After adjusting for confounding factors, fetal (OR = 0.530, 95% CI: 0.411-0.0.683), childhood (OR = 0.642, 95% CI: 0.494-0.833) exposure to the Chinese famine for men was negatively associated with hyperuricemia. Conversely, exposure to the Chinese famine during fetal (OR = 2.144, 95% CI: 1.622-2.834), childhood (OR = 1.485, 95% CI: 1.105-1.997), and adolescence (OR = 1.967, 95% CI: 1.465-2.641) for women was positively associated with hyperuricemia. Furthermore, the impact of famine on hyperuricemia that has been observed in exposed women might be intensified by the presence of dyslipidemia, abdominal obesity, and overweight/obesity. Conclusion Women exposed to the Chinese famine during fetal, childhood, and adolescence were positively associated with hyperuricemia, while men exhibited a negative association during fetal and childhood. Additionally, the effect of famine on hyperuricemia in exposed women appears to be intensified by the presence of dyslipidemia, abdominal obesity, and overweight/obesity.
Collapse
Affiliation(s)
- Huali Xiong
- Department of Public Health, Health Commission of Rongchang District, Chongqing, China
| | - Daiqiang Liu
- Department of Hospital Information, The People's Hospital of Rongchang District, Chongqing, China
| | - Dayi Tang
- First Clinical College, Mudanjiang Medical College, Mudanjiang, Heilongjiang, China
| | - Fengxun Ma
- Department of Public Health, Health Commission of Rongchang District, Chongqing, China
| |
Collapse
|
4
|
Martinez GJ, Appleton M, Kipp ZA, Loria AS, Min B, Hinds TD. Glucocorticoids, their uses, sexual dimorphisms, and diseases: new concepts, mechanisms, and discoveries. Physiol Rev 2024; 104:473-532. [PMID: 37732829 PMCID: PMC11281820 DOI: 10.1152/physrev.00021.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 08/07/2023] [Accepted: 09/10/2023] [Indexed: 09/22/2023] Open
Abstract
The normal stress response in humans is governed by the hypothalamic-pituitary-adrenal (HPA) axis through heightened mechanisms during stress, raising blood levels of the glucocorticoid hormone cortisol. Glucocorticoids are quintessential compounds that balance the proper functioning of numerous systems in the mammalian body. They are also generated synthetically and are the preeminent therapy for inflammatory diseases. They act by binding to the nuclear receptor transcription factor glucocorticoid receptor (GR), which has two main isoforms (GRα and GRβ). Our classical understanding of glucocorticoid signaling is from the GRα isoform, which binds the hormone, whereas GRβ has no known ligands. With glucocorticoids being involved in many physiological and cellular processes, even small disruptions in their release via the HPA axis, or changes in GR isoform expression, can have dire ramifications on health. Long-term chronic glucocorticoid therapy can lead to a glucocorticoid-resistant state, and we deliberate how this impacts disease treatment. Chronic glucocorticoid treatment can lead to noticeable side effects such as weight gain, adiposity, diabetes, and others that we discuss in detail. There are sexually dimorphic responses to glucocorticoids, and women tend to have a more hyperresponsive HPA axis than men. This review summarizes our understanding of glucocorticoids and critically analyzes the GR isoforms and their beneficial and deleterious mechanisms and the sexual differences that cause a dichotomy in responses. We also discuss the future of glucocorticoid therapy and propose a new concept of dual GR isoform agonist and postulate why activating both isoforms may prevent glucocorticoid resistance.
Collapse
Affiliation(s)
- Genesee J Martinez
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, Kentucky, United States
| | - Malik Appleton
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, Kentucky, United States
| | - Zachary A Kipp
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, Kentucky, United States
| | - Analia S Loria
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, Kentucky, United States
- Barnstable Brown Diabetes Center, University of Kentucky College of Medicine, Lexington, Kentucky, United States
| | - Booki Min
- Department of Microbiology and Immunology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States
| | - Terry D Hinds
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, Kentucky, United States
- Barnstable Brown Diabetes Center, University of Kentucky College of Medicine, Lexington, Kentucky, United States
- Markey Cancer Center, University of Kentucky, Lexington, Kentucky, United States
| |
Collapse
|
5
|
Oliveira CA, Mercês ÉAB, Portela FS, De Benedictis JM, De Benedictis LM, da Silva AVB, Campanati JDAG, de Melo FF, Oliveira MV, de Magalhães ACM, Soares TDJ, Amaral LSDB. Benefits of high-intensity interval training compared to continuous training to reduce apoptotic markers in female rats with cisplatin nephrotoxicity - possible modulatory role of IL-11. Apoptosis 2023; 28:566-575. [PMID: 36653732 DOI: 10.1007/s10495-023-01816-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/08/2023] [Indexed: 01/20/2023]
Abstract
Apoptotic signaling pathways are involved in acute kidney injury (AKI) induced by the antineoplastic drug cisplatin (Cis). Mechanical stress is known to increase interleukin (IL) -11, a pleiotropic cytokine with antiapoptotic and antinecrotic effects. We compared the impact of high-intensity interval training (HIIT) with low-intensity continuous training (LICT) and moderate-intensity continuous training (MICT) on renal levels of IL-11 and the expression of apoptotic markers in female rats with nephrotoxicity induced by Cis. For that, the animals were divided into five groups (n = 7): control and sedentary (C + S); Cis and sedentary (Cis + S); Cis and LICT (Cis + LICT); Cis and MICT (Cis + MICT) and Cis and HIIT (Cis + HIIT). At the end of 8 weeks of treadmill running, the rats received a single injection of Cis (5 mg/kg), and 7 days later they were euthanized. Serum and kidney samples were collected to assess the blood urea nitrogen (BUN), gene expression of TNF receptor 1 (TNFR1) and 2 (TNFR2), caspase-3, (p38) MAPK (MAPK14), p53, Bax, Bak, Bcl-2, and Bcl-xL, renal levels of IL-11, IL-8, and p53, and immunolocalization of cleaved caspase-3, Bax, Bcl-2, and (p38) MAPK in renal tissue. Our data indicate that all trained groups showed a significant intensity-dependent increase in renal levels of IL-11 associated with reduced local expression of proapoptotic and increased antiapoptotic markers, but these effects were more pronounced with HIIT. So, HIIT appears to provide superior renoprotection than traditional continuous training by modulating apoptotic signaling pathways, and this effect can be related to the increase in renal levels of IL-11.
Collapse
Affiliation(s)
- Caroline Assunção Oliveira
- Instituto Multidisciplinar em Saúde, Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Universidade Federal da Bahia, Vitória da Conquista, Rua Rio de Contas, 58, Candeias, 45029-094, Bahia, Brazil
| | - Érika Azenathe Barros Mercês
- Instituto Multidisciplinar em Saúde, Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Universidade Federal da Bahia, Vitória da Conquista, Rua Rio de Contas, 58, Candeias, 45029-094, Bahia, Brazil
| | - Fernanda Santos Portela
- Instituto Multidisciplinar em Saúde, Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Universidade Federal da Bahia, Vitória da Conquista, Rua Rio de Contas, 58, Candeias, 45029-094, Bahia, Brazil
| | - Júlia Mafra De Benedictis
- Instituto Multidisciplinar em Saúde, Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Universidade Federal da Bahia, Vitória da Conquista, Rua Rio de Contas, 58, Candeias, 45029-094, Bahia, Brazil
| | - Laís Mafra De Benedictis
- Instituto Multidisciplinar em Saúde, Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Universidade Federal da Bahia, Vitória da Conquista, Rua Rio de Contas, 58, Candeias, 45029-094, Bahia, Brazil
| | - Antônio Victor Brito da Silva
- Instituto Multidisciplinar em Saúde, Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Universidade Federal da Bahia, Vitória da Conquista, Rua Rio de Contas, 58, Candeias, 45029-094, Bahia, Brazil
| | - João de Assis Gonçalves Campanati
- Instituto Multidisciplinar em Saúde, Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Universidade Federal da Bahia, Vitória da Conquista, Rua Rio de Contas, 58, Candeias, 45029-094, Bahia, Brazil
| | - Fabrício Freire de Melo
- Instituto Multidisciplinar em Saúde, Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Universidade Federal da Bahia, Vitória da Conquista, Rua Rio de Contas, 58, Candeias, 45029-094, Bahia, Brazil
| | - Márcio Vasconcelos Oliveira
- Instituto Multidisciplinar em Saúde, Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Universidade Federal da Bahia, Vitória da Conquista, Rua Rio de Contas, 58, Candeias, 45029-094, Bahia, Brazil
| | - Amélia Cristina Mendes de Magalhães
- Instituto Multidisciplinar em Saúde, Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Universidade Federal da Bahia, Vitória da Conquista, Rua Rio de Contas, 58, Candeias, 45029-094, Bahia, Brazil
| | - Telma de Jesus Soares
- Instituto Multidisciplinar em Saúde, Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Universidade Federal da Bahia, Vitória da Conquista, Rua Rio de Contas, 58, Candeias, 45029-094, Bahia, Brazil
| | - Liliany Souza de Brito Amaral
- Instituto Multidisciplinar em Saúde, Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Universidade Federal da Bahia, Vitória da Conquista, Rua Rio de Contas, 58, Candeias, 45029-094, Bahia, Brazil.
| |
Collapse
|
6
|
Fukunaga S, Fujita Y. Low glomerular number at birth can lead to the development of chronic kidney disease. Front Endocrinol (Lausanne) 2023; 14:1120801. [PMID: 36777357 PMCID: PMC9909536 DOI: 10.3389/fendo.2023.1120801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Accepted: 01/13/2023] [Indexed: 01/27/2023] Open
Abstract
Chronic kidney disease (CKD) prevalence is increasing worldwide, and reducing the number of patients with CKD is of utmost importance. The environment during the fetal, perinatal, and early childhood stages may influence CKD development (developmental origins of health and disease). Under conditions of maternal malnutrition, the glomerular number of infants reduces, and the risk of developing CKD may increase. Nephron progenitor cells and ureteric buds interact with each other to form glomeruli at the tip of the ureteric bud. Thus, the number of glomeruli is determined by the number of ureteric bud branches, which are reportedly decreased due to maternal malnutrition, in turn reducing the glomerular number. Four possible mechanisms can explain the low glomerular number resulting from maternal malnutrition: 1) suppression of c-Ret expression, 2) suppression of nephron formation by renin-angiotensin-aldosterone system inhibition, 3) exposure to excess glucocorticoids, and 4) promotion of apoptosis. Additionally, nephron formation does not continue after birth in humans. Therefore, a low glomerular number at birth is a lifelong burden on the glomeruli and increases the risk of developing CKD. Therefore, it is important to maintain the glomerular number at birth. Accurate glomerular counts are essential for conducting studies on the glomerular number. The dissector/fractionator method is the gold standard; however, it can only be performed at some institutions. Recently, methods have been developed to measure the glomerular number by combining computed tomography and pathological examination and measure the glomerular count using magnetic resonance imaging. Models of decreased and increased glomerular numbers have been developed. Moreover, research regarding the causes of decreased glomerular number and its relationship with development of lifestyle-related diseases and renal dysfunction has significantly progressed, furthering our understanding of the importance of glomerular number.
Collapse
Affiliation(s)
- Shohei Fukunaga
- Division of Nephrology, Shimane University Hospital, Izumo, Shimane, Japan
- *Correspondence: Shohei Fukunaga, ; Yuki Fujita,
| | - Yuki Fujita
- Department of Developmental Biology, Shimane University Faculty of Medicine, Izumo, Shimane, Japan
- *Correspondence: Shohei Fukunaga, ; Yuki Fujita,
| |
Collapse
|
7
|
Voggel J, Fink G, Zelck M, Wohlfarth M, Post JM, Bindila L, Rauh M, Amann K, Alejandre Alcázar MA, Dötsch J, Nüsken KD, Nüsken E. Elevated n-3/n-6 PUFA ratio in early life diet reverses adverse intrauterine kidney programming in female rats. J Lipid Res 2022; 63:100283. [PMID: 36152882 PMCID: PMC9619183 DOI: 10.1016/j.jlr.2022.100283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/30/2022] [Accepted: 09/11/2022] [Indexed: 11/27/2022] Open
Abstract
Intrauterine growth restriction (IUGR) predisposes to chronic kidney disease via activation of proinflammatory pathways, and omega-3 PUFAs (n-3 PUFAs) have anti-inflammatory properties. In female rats, we investigated 1) how an elevated dietary n-3/n-6 PUFA ratio (1:1) during postnatal kidney development modifies kidney phospholipid (PL) and arachidonic acid (AA) metabolite content and 2) whether the diet counteracts adverse molecular protein signatures expected in IUGR kidneys. IUGR was induced by bilateral uterine vessel ligation or intrauterine stress through sham operation 3.5 days before term. Control (C) offspring were born after uncompromised pregnancy. On postnatal (P) days P2–P39, rats were fed control (n-3/n-6 PUFA ratio 1:20) or n-3 PUFA intervention diet (N3PUFA; ratio 1:1). Plasma parameters (P33), kidney cortex lipidomics and proteomics, as well as histology (P39) were studied. We found that the intervention diet tripled PL-DHA content (PC 40:6; P < 0.01) and lowered both PL-AA content (PC 38:4 and lyso-phosphatidylcholine 20:4; P < 0.05) and AA metabolites (HETEs, dihydroxyeicosatrienoic acids, and epoxyeicosatrienoic acids) to 25% in all offspring groups. After ligation, our network analysis of differentially expressed proteins identified an adverse molecular signature indicating inflammation and hypercoagulability. N3PUFA diet reversed 61 protein alterations (P < 0.05), thus mitigating adverse IUGR signatures. In conclusion, an elevated n-3/n-6 PUFA ratio in early diet strongly reduces proinflammatory PLs and mediators while increasing DHA-containing PLs regardless of prior intrauterine conditions. Counteracting a proinflammatory hypercoagulable protein signature in young adult IUGR individuals through early diet intervention may be a feasible strategy to prevent developmentally programmed kidney damage in later life.
Collapse
Affiliation(s)
- Jenny Voggel
- Clinic and Polyclinic for Pediatric and Adolescent Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Gregor Fink
- Clinic and Polyclinic for Pediatric and Adolescent Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Germany
| | - Magdalena Zelck
- Clinic and Polyclinic for Pediatric and Adolescent Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Germany
| | - Maria Wohlfarth
- Clinic and Polyclinic for Pediatric and Adolescent Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Germany
| | - Julia M Post
- Clinical Lipidomics Unit, Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University of Mainz, Mainz, Germany
| | - Laura Bindila
- Clinical Lipidomics Unit, Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University of Mainz, Mainz, Germany
| | - Manfred Rauh
- Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, Erlangen, Germany
| | - Kerstin Amann
- Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-University Erlangen, Erlangen, Germany
| | - Miguel A Alejandre Alcázar
- Clinic and Polyclinic for Pediatric and Adolescent Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany; Institute for Lung Health, University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Gießen, Germany
| | - Jörg Dötsch
- Clinic and Polyclinic for Pediatric and Adolescent Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Germany
| | - Kai-Dietrich Nüsken
- Clinic and Polyclinic for Pediatric and Adolescent Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Germany
| | - Eva Nüsken
- Clinic and Polyclinic for Pediatric and Adolescent Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Germany.
| |
Collapse
|
8
|
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
|
9
|
Doan TNA, Akison LK, Bianco-Miotto T. Epigenetic Mechanisms Responsible for the Transgenerational Inheritance of Intrauterine Growth Restriction Phenotypes. Front Endocrinol (Lausanne) 2022; 13:838737. [PMID: 35432208 PMCID: PMC9008301 DOI: 10.3389/fendo.2022.838737] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 03/02/2022] [Indexed: 12/20/2022] Open
Abstract
A poorly functioning placenta results in impaired exchanges of oxygen, nutrition, wastes and hormones between the mother and her fetus. This can lead to restriction of fetal growth. These growth restricted babies are at increased risk of developing chronic diseases, such as type-2 diabetes, hypertension, and kidney disease, later in life. Animal studies have shown that growth restricted phenotypes are sex-dependent and can be transmitted to subsequent generations through both the paternal and maternal lineages. Altered epigenetic mechanisms, specifically changes in DNA methylation, histone modifications, and non-coding RNAs that regulate expression of genes that are important for fetal development have been shown to be associated with the transmission pattern of growth restricted phenotypes. This review will discuss the subsequent health outcomes in the offspring after growth restriction and the transmission patterns of these diseases. Evidence of altered epigenetic mechanisms in association with fetal growth restriction will also be reviewed.
Collapse
Affiliation(s)
- Thu Ngoc Anh Doan
- School of Agriculture, Food and Wine, Waite Research Institute, University of Adelaide, Adelaide, SA, Australia
- Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia
| | - Lisa K. Akison
- School of Biomedical Sciences, University of Queensland, Brisbane, QLD, Australia
| | - Tina Bianco-Miotto
- School of Agriculture, Food and Wine, Waite Research Institute, University of Adelaide, Adelaide, SA, Australia
- Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia
- *Correspondence: Tina Bianco-Miotto,
| |
Collapse
|
10
|
Niu Y, Zhao Y, He J, Yun Y, Shen M, Gan Z, Zhang L, Wang T. Dietary dihydroartemisinin supplementation alleviates intestinal inflammatory injury through TLR4/NOD/NF-κB signaling pathway in weaned piglets with intrauterine growth retardation. ACTA ACUST UNITED AC 2021; 7:667-678. [PMID: 34430721 PMCID: PMC8361298 DOI: 10.1016/j.aninu.2020.12.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/16/2020] [Accepted: 12/20/2020] [Indexed: 01/10/2023]
Abstract
The aim of present study was to evaluate whether diets supplemented with dihydroartemisinin (DHA) could alleviate intestinal inflammatory injury in weaned piglets with intrauterine growth retardation (IUGR). Twelve normal birth weight (NBW) piglets and 12 piglets with IUGR were fed a basal diet (NBW-CON and IUCR-CON groups), and another 12 piglets with IUGR were fed the basal diet supplemented with DHA at 80 mg/kg (IUGR-DHA group) from 21 to 49 d of age. At 49 d of age, 8 piglets with similar body weight in each group were sacrificed. The jejunal and ileal samples were collected for further analysis. The results showed that IUGR impaired intestinal morphology, increased intestinal inflammatory response, raised enterocyte apoptosis and reduced enterocyte proliferation and activated transmembrane toll-like receptor 4 (TLR4)/nucleotide-binding and oligomerization domain (NOD)/nuclear factor-κB (NF-κB) signaling pathway. Dihydroartemisinin inclusion ameliorated intestinal morphology, indicated by increased villus height, villus height-to-crypt depth ratio, villus surface area and decreased villus width of piglets with IUGR (P < 0.05). Compared with NBW piglets, IUGR piglets supplemented with DHA exhibited higher apoptosis index and caspase-3 expression, and lower proliferation index and proliferating cell nuclear antigen expression in the intestine (P < 0.05). Dihydroartemisinin supplementation attenuated the intestinal inflammation of piglets with IUGR, indicated by increased concentrations of intestinal inflammatory cytokines and lipopolysaccharides (P < 0.05). In addition, DHA supplementation down-regulated the related mRNA expressions of TLR4/NOD/NF-κB signaling pathway and upregulated mRNA expressions of negative regulators of TLR4 and NOD signaling pathway in the intestine of piglets with IUGR (P < 0.05). Piglets in the IUGR-DHA group showed lower protein expressions of TLR4, phosphorylated NF-κB (pNF-κB) inhibitor α, nuclear pNF-κB, and higher protein expression of cytoplasmic pNF-κB in the intestine than those in the IUGR-CON group (P < 0.05). In conclusion, DHA supplementation could improve intestinal morphology, regulate enterocyte proliferation and apoptosis, and alleviate intestinal inflammation through TLR4/NOD/NF-κB signaling pathway in weaned piglets with IUGR.
Collapse
Affiliation(s)
- Yu Niu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Yongwei Zhao
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Jintian He
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Yang Yun
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Mingming Shen
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Zhending Gan
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Lili Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Tian Wang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| |
Collapse
|
11
|
Voggel J, Lubomirov L, Lechner F, Fink G, Nüsken E, Wohlfarth M, Pfitzer G, Shah-Hosseini K, Hellmich M, Alejandre Alcázar MA, Dötsch J, Nüsken KD. Vascular tone regulation in renal interlobar arteries of male rats is dysfunctional after intrauterine growth restriction. Am J Physiol Renal Physiol 2021; 321:F93-F105. [PMID: 34056927 DOI: 10.1152/ajprenal.00653.2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Intrauterine growth restriction (IUGR) due to an adverse intrauterine environment predisposes to arterial hypertension and loss of kidney function. Here, we investigated whether vascular dysregulation in renal interlobar arteries (RIAs) may contribute to hypertensive glomerular damage after IUGR. In rats, IUGR was induced by bilateral uterine vessel ligation. Offspring of nonoperated rats served as controls. From postnatal day 49, blood pressure was telemetrically recorded. On postnatal day 70, we evaluated contractile function in RIAs and mesenteric arteries. In addition, blood, urine, and glomerular parameters as well as renal collagen deposition were analyzed. IUGR RIAs not only showed loss of stretch activation in 9 of 11 arteries and reduced stretch-induced myogenic tone but also showed a shift of the concentration-response relation of acetylcholine-induced relaxation toward lower concentrations. However, IUGR RIAs also exhibited augmented contractions through phenylephrine. Systemic mean arterial pressure [mean difference: 4.8 mmHg (daytime) and 5.7 mmHg (night)], mean glomerular area (IUGR: 9,754 ± 338 µm2 and control: 8,395 ± 227 µm2), and urinary protein-to-creatinine ratio (IUGR: 1.67 ± 0.13 g/g and control: 1.26 ± 0.10 g/g) were elevated after IUGR. We conclude that male IUGR rat offspring may have increased vulnerability toward hypertensive glomerular damage due to loss of myogenic tone and augmented endothelium-dependent relaxation in RIAs.NEW & NOTEWORTHY For the first time, our study presents wire myography data from renal interlobar arteries (RIAs) and mesenteric arteries of young adult rat offspring after intrauterine growth restriction (IUGR). Our data indicate that myogenic tone in RIAs is dysfunctional after IUGR. Furthermore, IUGR offspring suffer from mild arterial hypertension, glomerular hypertrophy, and increased urinary protein-to-creatinine ratio. Dysregulation of vascular tone in RIAs could be an important variable that impacts upon vulnerability toward glomerular injury after IUGR.
Collapse
Affiliation(s)
- Jenny Voggel
- Department of Pediatrics and Adolescent Medicine, Medical Faculty and University Hospital Cologne, University of Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Lubomir Lubomirov
- Institute of Vegetative Physiology, University of Cologne, Cologne, Germany
| | - Felix Lechner
- Department of Pediatrics and Adolescent Medicine, Medical Faculty and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Gregor Fink
- Department of Pediatrics and Adolescent Medicine, Medical Faculty and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Eva Nüsken
- Department of Pediatrics and Adolescent Medicine, Medical Faculty and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Maria Wohlfarth
- Department of Pediatrics and Adolescent Medicine, Medical Faculty and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Gabriele Pfitzer
- Institute of Vegetative Physiology, University of Cologne, Cologne, Germany
| | - Kija Shah-Hosseini
- Institute of Medical Statistics and Computational Biology, Medical Faculty and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Martin Hellmich
- Institute of Medical Statistics and Computational Biology, Medical Faculty and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Miguel A Alejandre Alcázar
- Department of Pediatrics and Adolescent Medicine, Medical Faculty and University Hospital Cologne, University of Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany.,Faculty of Medicine and University Hospital Cologne, Cologne Excellence Cluster for Stress Responses in Ageing-Associated Diseases, University of Cologne, Cologne, Germany.,Institute for Lung Health (ILH), University of Giessen and Marburg Lung Center, Member of the German Center for Lung Research (DZL), Gießen, Germany
| | - Jörg Dötsch
- Department of Pediatrics and Adolescent Medicine, Medical Faculty and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Kai-Dietrich Nüsken
- Department of Pediatrics and Adolescent Medicine, Medical Faculty and University Hospital Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
12
|
Alshamrani A, Aldahmash W, Falodah F, Arafah M, Harrath AH, Alwasel S. Long-Term but Not Short-Term Maternal Fasting Reduces Nephron Number and Alters the Glomerular Filtration Barrier in Rat Offspring. Life (Basel) 2021; 11:life11040318. [PMID: 33917410 PMCID: PMC8067523 DOI: 10.3390/life11040318] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 03/28/2021] [Accepted: 04/02/2021] [Indexed: 01/04/2023] Open
Abstract
The present study examined the effects of maternal Ramadan-type fasting during selected days in the first, second, or third trimester, or during the entire pregnancy, on the kidney structure of male rat offspring. Pregnant rats were provided with food ad libitum during pregnancy (control group, C), or they were exposed to 16 h of fasting/day for three consecutive days in the middle of the first (FT1), second (FT2), or third trimester (FT3), or during whole pregnancy (FWP). Our results showed that dams in the FWP group demonstrated lower food intake and body weight during gestation. Litter size was unaltered by fasting in all groups; however, litter weight was significantly reduced only in the FWP group. Nephron number was decreased in the FWP group, but it remained unchanged in the other fasting groups. The ultrastructure of the glomerular filtration barrier indicated that the kidneys of offspring of the FWP group demonstrated wider diameters of fenestrations and filtration slits and smaller diameters of basement membranes. This was reflected by a significant increase in proteinuria in FWP only. These results suggest that, unlike with short-term fasting, which seems to be safe, maternal long-term fasting induces structural changes that were non-reversible, and that may contribute to impaired renal function, leading to chronic diseases in later life.
Collapse
Affiliation(s)
- Abdullah Alshamrani
- Zoology Department, College of Science, King Saud University, Riyadh 11451, Saudi Arabia; (A.A.); (W.A.); (F.F.); (A.H.H.)
| | - Waleed Aldahmash
- Zoology Department, College of Science, King Saud University, Riyadh 11451, Saudi Arabia; (A.A.); (W.A.); (F.F.); (A.H.H.)
| | - Fawaz Falodah
- Zoology Department, College of Science, King Saud University, Riyadh 11451, Saudi Arabia; (A.A.); (W.A.); (F.F.); (A.H.H.)
| | - Maria Arafah
- Department of Pathology, College of Medicine, King Saud University, Riyadh 11451, Saudi Arabia;
| | - Abdel Halim Harrath
- Zoology Department, College of Science, King Saud University, Riyadh 11451, Saudi Arabia; (A.A.); (W.A.); (F.F.); (A.H.H.)
| | - Saleh Alwasel
- Zoology Department, College of Science, King Saud University, Riyadh 11451, Saudi Arabia; (A.A.); (W.A.); (F.F.); (A.H.H.)
- Correspondence:
| |
Collapse
|
13
|
Block LN, Bowman BD, Schmidt JK, Keding LT, Stanic AK, Golos TG. The promise of placental extracellular vesicles: models and challenges for diagnosing placental dysfunction in utero†. Biol Reprod 2021; 104:27-57. [PMID: 32856695 PMCID: PMC7786267 DOI: 10.1093/biolre/ioaa152] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 08/04/2020] [Accepted: 08/26/2020] [Indexed: 12/14/2022] Open
Abstract
Monitoring the health of a pregnancy is of utmost importance to both the fetus and the mother. The diagnosis of pregnancy complications typically occurs after the manifestation of symptoms, and limited preventative measures or effective treatments are available. Traditionally, pregnancy health is evaluated by analyzing maternal serum hormone levels, genetic testing, ultrasonographic imaging, and monitoring maternal symptoms. However, researchers have reported a difference in extracellular vesicle (EV) quantity and cargo between healthy and at-risk pregnancies. Thus, placental EVs (PEVs) may help to understand normal and aberrant placental development, monitor pregnancy health in terms of developing placental pathologies, and assess the impact of environmental influences, such as infection, on pregnancy. The diagnostic potential of PEVs could allow for earlier detection of pregnancy complications via noninvasive sampling and frequent monitoring. Understanding how PEVs serve as a means of communication with maternal cells and recognizing their potential utility as a readout of placental health have sparked a growing interest in basic and translational research. However, to date, PEV research with animal models lags behind human studies. The strength of animal pregnancy models is that they can be used to assess placental pathologies in conjunction with isolation of PEVs from fluid samples at different time points throughout gestation. Assessing PEV cargo in animals within normal and complicated pregnancies will accelerate the translation of PEV analysis into the clinic for potential use in prognostics. We propose that appropriate animal models of human pregnancy complications must be established in the PEV field.
Collapse
Affiliation(s)
- Lindsey N Block
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, USA
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Brittany D Bowman
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Jenna Kropp Schmidt
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Logan T Keding
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Aleksandar K Stanic
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI, USA
| | - Thaddeus G Golos
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, USA
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI, USA
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
14
|
Echocardiographic assessment of fetal cardiac function in the uterine artery ligation rat model of IUGR. Pediatr Res 2021; 90:801-808. [PMID: 33504964 PMCID: PMC8566221 DOI: 10.1038/s41390-020-01356-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 10/31/2020] [Accepted: 12/18/2020] [Indexed: 01/30/2023]
Abstract
BACKGROUND Intrauterine growth restriction (IUGR) leads to cardiac dysfunction and adverse remodeling of the fetal heart, as well as a higher risk of postnatal cardiovascular diseases. The rat model of IUGR, via uterine artery ligation, is a popular model but its cardiac sequelae is not well investigated. Here, we performed an echocardiographic evaluation of its cardiac function to determine how well it can represent the disease in humans. METHODS Unilateral uterine artery ligation was performed at embryonic day 17 (E17) and echocardiography was performed at E19 and E20. RESULTS Growth-restricted fetuses were significantly smaller and lighter, and had an higher placenta-to-fetus weight ratio. Growth-restricted fetal hearts had reduced wall thickness-to-diameter ratio, indicating left ventricular (LV) dilatation, and they had elevated trans-mitral and trans-tricuspid E/A ratios and reduced left and right ventricular fractional shortening (FS), suggesting systolic and diastolic dysfunction. These were similar to human IUGR fetuses. However, growth-restricted rat fetuses did not demonstrate head-sparing effect, displayed a lower LV myocardial performance index, and ventricular outflow velocities were not significantly reduced, which were dissimilar to human IUGR fetuses. CONCLUSIONS Despite the differences, our results suggest that this IUGR model has significant cardiac dysfunction, and could be a suitable model for studying IUGR cardiovascular physiology. IMPACT Animal models of IUGR are useful, but their fetal cardiac function is not well studied, and it is unclear if they can represent human IUGR fetuses. We performed an echocardiographic assessment of the heart function of a fetal rat model of IUGR, created via maternal uterine artery ligation. Similar to humans, the model displayed LV dilatation, elevated E/A ratios, and reduced FS. Different from humans, the model displayed reduced MPI, and no significant outflow velocity reduction. Despite differences with humans, this rat model still displayed cardiac dysfunction and is suitable for studying IUGR cardiovascular physiology.
Collapse
|
15
|
Epigenetic mechanisms involved in intrauterine growth restriction and aberrant kidney development and function. J Dev Orig Health Dis 2020; 12:952-962. [PMID: 33349286 DOI: 10.1017/s2040174420001257] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Intrauterine growth restriction (IUGR) due to uteroplacental insufficiency results in a placenta that is unable to provide adequate nutrients and oxygen to the fetus. These growth-restricted babies have an increased risk of hypertension and chronic kidney disease later in life. In rats, both male and female growth-restricted offspring have nephron deficits but only males develop kidney dysfunction and high blood pressure. In addition, there is transgenerational transmission of nephron deficits and hypertension risk. Therefore, epigenetic mechanisms may explain the sex-specific programming and multigenerational transmission of IUGR-related phenotypes. Expression of DNA methyltransferases (Dnmt1and Dnmt3a) and imprinted genes (Peg3, Snrpn, Kcnq1, and Cdkn1c) were investigated in kidney tissues of sham and IUGR rats in F1 (embryonic day 20 (E20) and postnatal day 1 (PN1)) and F2 (6 and 12 months of age, paternal and maternal lines) generations (n = 6-13/group). In comparison to sham offspring, F1 IUGR rats had a 19% decrease in Dnmt3a expression at E20 (P < 0.05), with decreased Cdkn1c (19%, P < 0.05) and increased Kcnq1 (1.6-fold, P < 0.01) at PN1. There was a sex-specific difference in Cdkn1c and Snrpn expression at E20, with 29% and 34% higher expression in IUGR males compared to females, respectively (P < 0.05). Peg3 sex-specific expression was lost in the F2 IUGR offspring, only in the maternal line. These findings suggest that epigenetic mechanisms may be altered in renal embryonic and/or fetal development in growth-restricted offspring, which could alter kidney function, predisposing these offspring to kidney disease later in life.
Collapse
|
16
|
Folic acid supplementation alleviates reduced ureteric branching, nephrogenesis, and global DNA methylation induced by maternal nutrient restriction in rat embryonic kidney. PLoS One 2020; 15:e0230289. [PMID: 32251454 PMCID: PMC7135271 DOI: 10.1371/journal.pone.0230289] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 02/25/2020] [Indexed: 01/21/2023] Open
Abstract
We previously reported that maternal nutrient restriction (NR) inhibited ureteric branching, metanephric growth, and nephrogenesis in the rat. Here we examined whether folic acid, a methyl-group donor, rescues the inhibition of kidney development induced by NR and whether DNA methylation is involved in it. The offspring of dams given food ad libitum (CON) and those subjected to 50% food restriction (NR) were examined. NR significantly reduced ureteric tip number at embryonic day 14, which was attenuated by folic acid supplementation to nutrient restricted dams. At embryonic day 18, glomerular number, kidney weight, and global DNA methylation were reduced by NR, and maternal folic acid supplementation again alleviated them. Among DNA methyltransferases (DNMTs), DNMT1 was strongly expressed at embryonic day 15 in CON but was reduced in NR. In organ culture, an inhibitor of DNA methylation 5-aza-2 '-deoxycytidine as well as medium lacking methyl donors folic acid, choline, and methionine, significantly decreased ureteric tip number and kidney size mimicking the effect of NR. In conclusion, global DNA methylation is necessary for normal kidney development. Folic acid supplementation to nutrient restricted dams alleviated the impaired kidney development and DNA methylation in the offspring.
Collapse
|
17
|
Zhang W, Luan R. Early-life exposure to the Chinese famine of 1959-61 and risk of Hyperuricemia: results from the China health and retirement longitudinal study. BMC Public Health 2020; 20:15. [PMID: 31906901 PMCID: PMC6945412 DOI: 10.1186/s12889-019-8017-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 11/28/2019] [Indexed: 01/14/2023] Open
Abstract
Background Short-term starvation has been related to hyperuricemia. However, little is known about the long-term effect of early-life exposure to famine on hyperuricemia risk in adulthood. Methods The analysis included 2383 participants from the China Health and Retirement Longitudinal Study in 2015. Hyperuricemia was diagnosed as serum uric acid ≥7 mg/dL in men and serum uric acid ≥6 mg/dL in women. Famine exposure subgroups were defined as unexposed (born between October 1, 1962, and September 30, 1964), fetal-exposed (born between October 1, 1959, and September 30, 1961), and early-childhood exposed (born between October 1, 1956, and September 1, 1958). The association between early-life famine exposure and hyperuricemia risk was assessed using multivariate logistic regression. Results The prevalence of hyperuricemia in the unexposed, fetal-exposed, and early-childhood exposed participants was 10.7, 14.1, 11.1%, respectively. Compared with unexposed and early-childhood exposed participants combined as an age-balanced control, fetal-exposed participants showed an increased risk of hyperuricemia in adulthood (OR = 1.41; 95% CI: 1.06–1.88), after adjusting for gender, marital status, famine severity, residence, smoking, drinking, BMI, hypertension, and diabetes. The famine effect on hyperuricemia was accentuated by overweight or obesity (P for interaction = 0.042). Compared with unexposed and BMI < 24 kg/m2 participants, the OR (95%CI) of hyperuricemia was 3.66 (2.13–6.30) for fetal-exposed and overweight/obesity participants. However, combined unexposed and early-childhood exposed participants as an age-balanced control, the interaction of famine exposure and BMI was not statistically significant (P for interaction = 0.054). Conclusion Famine exposure in the fetal stage was associated with an increased risk of hyperuricemia in adulthood.
Collapse
Affiliation(s)
- Wenqiang Zhang
- West China School of Public Health and West China Fourth Hospital, Sichuan University, No. 16, Section 3, Ren Min Nan Road, Chengdu, 610041, Sichuan, China
| | - Rongsheng Luan
- West China School of Public Health and West China Fourth Hospital, Sichuan University, No. 16, Section 3, Ren Min Nan Road, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
18
|
|
19
|
Intapad S, Dasinger JH, Johnson JM, Brown AD, Ojeda NB, Alexander BT. Male and Female Intrauterine Growth-Restricted Offspring Differ in Blood Pressure, Renal Function, and Glucose Homeostasis Responses to a Postnatal Diet High in Fat and Sugar. Hypertension 2019; 73:620-629. [PMID: 30636548 DOI: 10.1161/hypertensionaha.118.12134] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
It is well established that inadequate nutrition during fetal life followed by postnatal overabundance programs adiposity and glucose intolerance. Studies addressing sexual dimorphism in developmental responses to a dietary mismatch are limited; the effect on blood pressure and renal function is understudied. Therefore, this study tested the hypothesis that a mismatch of prenatal and postnatal nutrition heightens cardiorenal and metabolic risk, outcomes that may vary by sex. Male and female offspring from sham-operated (control) or reduced uterine perfusion dams (growth restricted) were fed regular chow or a diet high in fat and sugar (enriched diet) from weaning until 6 months of age. Male and female offspring were assessed separately; 2-way ANOVA was used to investigate interactions between intrauterine growth-restricted and enriched-diet. Blood pressure was increased in all enriched-diet groups but did not differ in enriched-diet male or female growth-restricted versus same-sex control counterparts. Glomerular filtration rate was reduced in male growth-restricted regardless of diet; a decrease exacerbated by the enriched-diet suggesting the pathogenesis of increased blood pressure induced via an enriched-diet differs between male growth-restricted versus male control. An enriched diet was associated with glucose intolerance in male and female control but not male growth-restricted; the enriched diet exacerbated glucose intolerance in female growth-restricted. Thus, these findings indicate male growth-restricted are resistant to impaired glucose homeostasis, whereas female growth-restricted are susceptible to metabolic dysfunction regardless of postnatal diet. Hence, moderation of fat and sugar intake may be warranted in those born low birth weight to ensure minimal risk for chronic disease.
Collapse
Affiliation(s)
- Suttira Intapad
- From the Department of Physiology and Biophysics (S.I., J.H.D., J.M.J., A.D.B., B.T.A.), University of Mississippi Medical Center, Jackson.,Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA (S.I.)
| | - John Henry Dasinger
- From the Department of Physiology and Biophysics (S.I., J.H.D., J.M.J., A.D.B., B.T.A.), University of Mississippi Medical Center, Jackson
| | - Jeremy M Johnson
- From the Department of Physiology and Biophysics (S.I., J.H.D., J.M.J., A.D.B., B.T.A.), University of Mississippi Medical Center, Jackson
| | - Andrew D Brown
- From the Department of Physiology and Biophysics (S.I., J.H.D., J.M.J., A.D.B., B.T.A.), University of Mississippi Medical Center, Jackson
| | - Norma B Ojeda
- Department of Pediatrics (N.B.O.), University of Mississippi Medical Center, Jackson
| | - Barbara T Alexander
- From the Department of Physiology and Biophysics (S.I., J.H.D., J.M.J., A.D.B., B.T.A.), University of Mississippi Medical Center, Jackson
| |
Collapse
|
20
|
Lopez-Tello J, Arias-Alvarez M, Gonzalez-Bulnes A, Sferuzzi-Perri AN. Models of Intrauterine growth restriction and fetal programming in rabbits. Mol Reprod Dev 2019; 86:1781-1809. [PMID: 31538701 DOI: 10.1002/mrd.23271] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 09/02/2019] [Indexed: 12/23/2022]
Abstract
Intrauterine growth restriction (IUGR) affects approximately 10% of human pregnancies globally and has immediate and life-long consequences for offspring health. However, the mechanisms underlying the pathogenesis of IUGR and its association with later health and disease outcomes are poorly understood. To address these knowledge gaps, the use of experimental animals is critically important. Since the 50's different environmental, pharmacological, and surgical manipulations have been performed in the rabbit to improve our knowledge of the control of fetal growth, fetal responses to IUGR, and mechanisms by which offspring may be programmed by an adverse gestational environment. The purpose of this review is therefore to summarize the utility of the rabbit as a model for IUGR research. It first summarizes the knowledge of prenatal and postnatal development in the rabbit and how these events relate to developmental milestones in humans. It then describes the methods used to induce IUGR in rabbits and the knowledge gained about the mechanisms determining prenatal and postnatal outcomes of the offspring. Finally, it discusses the application of state of the art approaches in the rabbit, including high-resolution ultrasound, magnetic resonance imaging, and gene targeting, to gain a deeper integrative understanding of the physiological and molecular events governing the development of IUGR. Overall, we hope to engage and inspire investigators to employ the rabbit as a model organism when studying pregnancy physiology so that we may advance our understanding of mechanisms underlying IUGR and its consequences in humans and other mammalian species.
Collapse
Affiliation(s)
- Jorge Lopez-Tello
- Department of Physiology, Development, and Neuroscience, Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
| | - Maria Arias-Alvarez
- Department of Animal Production. Veterinary Faculty, Complutense University of Madrid, Ciudad Universitaria, Madrid, Spain
| | | | - Amanda N Sferuzzi-Perri
- Department of Physiology, Development, and Neuroscience, Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
| |
Collapse
|
21
|
Stewart T, Kallash M, Vehaskari VM, Hodgeson SM, Aviles DH. Increased Autophagy and Apoptosis in the Kidneys of Intrauterine Growth Restricted Rats. Fetal Pediatr Pathol 2019; 38:185-194. [PMID: 30741571 DOI: 10.1080/15513815.2018.1564160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
BACKGROUND IUGR has been associated with nephron loss and chronic kidney disease (CKD). MATERIALS AND METHODS We examined autophagy and apoptosis markers in the kidneys of IUGR Sprague Dawley rats induced by maternal low protein diet (LP), comparing them to controls. The autophagy marker LC3B, the pro-apoptotic protein Bax, and the anti-apoptotic protein Bcl-2 were determined by quantitative immunoblotting. Immunohistochemical expressions of LC3B, Bax, and Bcl-2 were evaluated at 4 weeks age. Glomerular counts (by maceration techniques) were performed at 5 weeks. RESULTS The LP diet offspring were lighter (P < 0.05). In IUGR kidneys, LC3B and Bax were increased at birth (p < 0.05, p < 0.001) and at 4 weeks (p < 0.0142, p < 0.0001), Bcl-2 was decreased at birth (p < 0.05), and there were less glomeruli (p < 0.01) at 5 weeks. CONCLUSIONS Autophagy and apoptosis may have a role in IUGR associated decreased nephron number in Sprague rats.
Collapse
Affiliation(s)
- Tyrus Stewart
- a Louisiana State University, Health Sciences Center , New Orleans , Louisiana , USA
| | - Mahmoud Kallash
- b Nationwide Children's Hospital , Columbus , Ohio , USA.,c University at Buffalo , Buffalo , New York , USA
| | - V Matti Vehaskari
- a Louisiana State University, Health Sciences Center , New Orleans , Louisiana , USA
| | - Sydney M Hodgeson
- a Louisiana State University, Health Sciences Center , New Orleans , Louisiana , USA
| | - Diego H Aviles
- a Louisiana State University, Health Sciences Center , New Orleans , Louisiana , USA.,d Children's Hospital , New Orleans , Louisiana , USA
| |
Collapse
|
22
|
Yeung KR, Sunderland N, Lind JM, Heffernan S, Pears S, Xu B, Hennessy A, Makris A. Increased salt sensitivity in offspring of pregnancies complicated by experimental preeclampsia. Clin Exp Pharmacol Physiol 2018; 45:1302-1308. [PMID: 29992611 DOI: 10.1111/1440-1681.13008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 06/25/2018] [Accepted: 06/26/2018] [Indexed: 01/28/2023]
Abstract
Preeclampsia is a hypertensive disorder of pregnancy known to increase the risk of cardiovascular disease in mothers and offspring. Offspring exposed to a suboptimal intrauterine environment may experience altered fetal programming and subsequent long-term cardiovascular changes. This study investigated changes in the vascular response in offspring from experimental preeclampsia (EPE) induced by uterine artery ligation, in the absence of fetal growth restriction, compared to normal baboon pregnancies (controls), following a high salt diet challenge. After 1 week of standard diet (containing <1% salt), animals were fed a high salt diet (6%) for 2 weeks. Systolic and diastolic blood pressure (SBP, DBP), aldosterone, renin and creatinine clearance were evaluated in EPE (n = 6, 50% male) and control (n = 6, 50% male) offspring. A repeated measures analysis was performed, and P < 0.05 was considered significant. At baseline, there were no differences between the groups in any parameter (EPE, mean age and weight 3.2 ± 1.2 years, 6.8 ± 1.0 kg, respectively; Control, 2.9 ± 0.8 years, 7.1 ± 1.5 kg). After salt loading the EPE group had significantly higher SBP (92 ± 5 mm Hg) compared to the control group (83 ± 4 mm Hg, P = 0.03). Aldosterone concentration was higher in the EPE group despite the same salt excretion and no difference in renal function. Salt sensitivity may differ in offspring from hypertensive pregnancies due to fetal programming. This could have long-term consequences for cardiovascular health of EPE offspring and further research is required to determine the exact pathological mechanisms.
Collapse
Affiliation(s)
- Kristen R Yeung
- Western Sydney University, Sydney, NSW, Australia.,Heart Research Institute, Sydney, NSW, Australia
| | | | | | | | - Suzanne Pears
- Heart Research Institute, Sydney, NSW, Australia.,Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Bei Xu
- Western Sydney University, Sydney, NSW, Australia.,Heart Research Institute, Sydney, NSW, Australia
| | - Annemarie Hennessy
- Western Sydney University, Sydney, NSW, Australia.,Heart Research Institute, Sydney, NSW, Australia
| | - Angela Makris
- Western Sydney University, Sydney, NSW, Australia.,Heart Research Institute, Sydney, NSW, Australia.,Nephrology Department, Liverpool Hospital, Liverpool, NSW, Australia
| |
Collapse
|
23
|
Effect of early postnatal nutrition on chronic kidney disease and arterial hypertension in adulthood: a narrative review. J Dev Orig Health Dis 2018; 9:598-614. [PMID: 30078383 DOI: 10.1017/s2040174418000454] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Intrauterine growth restriction (IUGR) has been identified as a risk factor for adult chronic kidney disease (CKD), including hypertension (HTN). Accelerated postnatal catch-up growth superimposed to IUGR has been shown to further increase the risk of CKD and HTN. Although the impact of excessive postnatal growth without previous IUGR is less clear, excessive postnatal overfeeding in experimental animals shows a strong impact on the risk of CKD and HTN in adulthood. On the other hand, food restriction in the postnatal period seems to have a protective effect on CKD programming. All these effects are mediated at least partially by the activation of the renin-angiotensin system, leptin and neuropeptide Y (NPY) signaling and profibrotic pathways. Early nutrition, especially in the postnatal period has a significant impact on the risk of CKD and HTN at adulthood and should receive specific attention in the prevention of CKD and HTN.
Collapse
|
24
|
Briffa JF, Wlodek ME, Moritz KM. Transgenerational programming of nephron deficits and hypertension. Semin Cell Dev Biol 2018; 103:94-103. [PMID: 29859996 DOI: 10.1016/j.semcdb.2018.05.025] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 05/28/2018] [Accepted: 05/29/2018] [Indexed: 01/16/2023]
Abstract
Exposure to a sub-optimal environment in the womb can result in poor fetal growth and impair the normal development of organs. The kidney, specifically the process of nephrogenesis, has been shown to be impacted by many common pregnancy exposures including an inadequate diet, poor placental function, maternal stress as well as maternal smoking and alcohol consumption. This can result in offspring being born with a reduced nephron endowment, which places these individuals at increased risk of hypertension and chronic kidney disease (CKD). Of recent interest is whether this disease risk can be passed on to subsequent generations and, if so, what are the mechanisms and pathways involved. In this review, we highlight the growing body of evidence that a low birth weight and hypertension, which are both major risk factors for cardiovascular and CKD, can be transmitted across generations. However, as yet there is little data as to whether a low nephron endowment contributes to this disease transmission. The emerging data suggests transmission can occur both through both the maternal and paternal lines, which likely involves epigenetic mechanisms such chromatin remodelling (DNA methylation and histone modification) and non-coding RNA modifications. In addition, females who were born small and/or have a low nephron endowment are at an increased risk for pregnancy complications, which can influence the growth and development of the next generation. Future animal studies in this area should include examining nephron endowment across multiple generations and determining adult renal function. Clinically, long term follow-up studies of large birth cohorts need to be undertaken to more clearly determine the impact a sub-optimal environment in one generation has on the health outcomes in the second, and subsequent, generation.
Collapse
Affiliation(s)
- Jessica F Briffa
- Department of Physiology, The University of Melbourne, Parkville, VIC, Australia
| | - Mary E Wlodek
- Department of Physiology, The University of Melbourne, Parkville, VIC, Australia
| | - Karen M Moritz
- Child Health Research Centre and School of Biomedical Sciences, The University of Queensland, St. Lucia, QLD, Australia.
| |
Collapse
|
25
|
Li Y, Zhang H, Su W, Ying Z, Chen Y, Zhang L, Lu Z, Wang T. Effects of dietary Bacillus amyloliquefaciens supplementation on growth performance, intestinal morphology, inflammatory response, and microbiota of intra-uterine growth retarded weanling piglets. J Anim Sci Biotechnol 2018; 9:22. [PMID: 29564121 PMCID: PMC5848560 DOI: 10.1186/s40104-018-0236-2] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 01/16/2018] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND The focus of recent research has been directed toward the probiotic potential of Bacillus amyloliquefaciens (BA) on the gut health of animals. However, little is known about BA's effects on piglets with intra-uterine growth retardation (IUGR). Therefore, this study investigated the effects of BA supplementation on the growth performance, intestinal morphology, inflammatory response, and microbiota of IUGR piglets. METHODS Eighteen litters of newborn piglets were selected at birth, with one normal birth weight (NBW) and two IUGR piglets in each litter (i.e., 18 NBW and 36 IUGR piglets in total). At weaning, the NBW piglet and one of the IUGR piglets were assigned to groups fed a control diet (i.e., the NBW-CON and IUGR-CON groups). The other IUGR piglet was assigned to a group fed the control diet supplemented with 2.0 g BA per kg of diet (i.e., IUGR-BA group). The piglets were thus distributed across three groups for a four-week period. RESULTS IUGR reduced the growth performance of the IUGR-CON piglets compared with the NBW-CON piglets. It was also associated with decreased villus sizes, increased apoptosis rates, reduced goblet cell numbers, and an imbalance between pro- and anti-inflammatory cytokines in the small intestine. Supplementation with BA improved the average daily weight gain and the feed efficiency of the IUGR-BA group compared with the IUGR-CON group (P < 0.05). The IUGR-BA group exhibited increases in the ratio of jejunal villus height to crypt depth, in ileal villus height, and in ileal goblet cell density. They also exhibited decreases in the numbers of jejunal and ileal apoptotic cells and ileal proliferative cells (P < 0.05). Supplementation with BA increased interleukin 10 content, but it decreased tumor necrosis factor alpha level in the small intestines of the IUGR-BA piglets (P < 0.05). Furthermore, compared with the IUGR-CON piglets, the IUGR-BA piglets had less Escherichia coli in their jejunal digesta, but more Lactobacillus and Bifidobacterium in their ileal digesta (P < 0.05). CONCLUSIONS Dietary supplementation with BA improves morphology, decreases inflammatory response, and regulates microbiota in the small intestines of IUGR piglets, which may contribute to improved growth performance during early life.
Collapse
Affiliation(s)
- Yue Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095 China
| | - Hao Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095 China
| | - Weipeng Su
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095 China
| | - Zhixiong Ying
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095 China
| | - Yueping Chen
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095 China
| | - Lili Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095 China
| | - Zhaoxin Lu
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095 China
| | - Tian Wang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095 China
| |
Collapse
|
26
|
Cuffe JSM, Briffa JF, Rosser S, Siebel AL, Romano T, Hryciw DH, Wlodek ME, Moritz KM. Uteroplacental insufficiency in rats induces renal apoptosis and delays nephrogenesis completion. Acta Physiol (Oxf) 2018; 222. [PMID: 29047216 DOI: 10.1111/apha.12982] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2017] [Revised: 10/08/2017] [Accepted: 10/10/2017] [Indexed: 12/28/2022]
Abstract
AIM Uteroplacental insufficiency in rats reduces nephron endowment, leptin concentrations and programmes cardiorenal disease in offspring. Cross-fostering growth-restricted (Restricted) offspring onto a mother with normal lactation restores leptin concentrations and nephron endowment. This study aimed to determine whether the reduced nephron endowment in Restricted offspring is due to delayed glomerular formation and dysregulation of renal genes regulating branching morphogenesis, apoptosis or leptin signalling. Furthermore, we aimed to investigate whether cross-fostering Restricted offspring onto Control mothers could improve glomerular maturation and restore renal gene abundance. METHODS Uteroplacental insufficiency was induced by bilateral uterine vessel ligation (Restricted) or sham (Control) surgery on gestation day 18 (E18). Kidneys were collected at E20, postnatal day 1 (PN1) and PN7. An additional cohort was cross-fostered onto separate mothers at birth and kidneys collected at PN7. RESULTS Kidneys were lighter in the Restricted group, but weight was restored with cross-fostering. At E20, abundance of Bax, Flt1 and Vegfa was increased in Restricted offspring, while Ret and Bcl2 transcripts were increased only in Restricted females. At PN7, abundance of Gdnf and Ret was higher in Restricted offspring, as was Casp3. Restricted offspring had a wider nephrogenic zone with more immature glomeruli suggesting a delayed or extended nephrogenic period. Cross-fostering had subtle effects on gene abundance and glomerular maturity. CONCLUSION Uteroplacental insufficiency induced apoptosis in the developing kidney and delayed and extended nephrogenesis. Cross-fostering Restricted offspring onto Control mothers had beneficial effects on kidney growth and renal maturity, which may contribute to the restoration of nephron endowment.
Collapse
Affiliation(s)
- J. S. M. Cuffe
- School of Biomedical Sciences; The University of Queensland; St. Lucia Qld Australia
- School of Medical Science; Menzies Health Institute Queensland; Griffith University; Southport Qld Australia
| | - J. F. Briffa
- Department of Physiology; The University of Melbourne; Parkville Vic. Australia
| | - S. Rosser
- School of Biomedical Sciences; The University of Queensland; St. Lucia Qld Australia
| | - A. L. Siebel
- Department of Physiology; The University of Melbourne; Parkville Vic. Australia
| | - T. Romano
- Department of Physiology, Anatomy and Microbiology; La Trobe University; Bundoora Vic. Australia
| | - D. H. Hryciw
- Department of Physiology; The University of Melbourne; Parkville Vic. Australia
| | - M. E. Wlodek
- Department of Physiology; The University of Melbourne; Parkville Vic. Australia
| | - K. M. Moritz
- School of Biomedical Sciences; The University of Queensland; St. Lucia Qld Australia
| |
Collapse
|
27
|
Condon DE, Tran PV, Lien YC, Schug J, Georgieff MK, Simmons RA, Won KJ. Defiant: (DMRs: easy, fast, identification and ANnoTation) identifies differentially Methylated regions from iron-deficient rat hippocampus. BMC Bioinformatics 2018; 19:31. [PMID: 29402210 PMCID: PMC5800085 DOI: 10.1186/s12859-018-2037-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2017] [Accepted: 01/23/2018] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Identification of differentially methylated regions (DMRs) is the initial step towards the study of DNA methylation-mediated gene regulation. Previous approaches to call DMRs suffer from false prediction, use extreme resources, and/or require library installation and input conversion. RESULTS We developed a new approach called Defiant to identify DMRs. Employing Weighted Welch Expansion (WWE), Defiant showed superior performance to other predictors in the series of benchmarking tests on artificial and real data. Defiant was subsequently used to investigate DNA methylation changes in iron-deficient rat hippocampus. Defiant identified DMRs close to genes associated with neuronal development and plasticity, which were not identified by its competitor. Importantly, Defiant runs between 5 to 479 times faster than currently available software packages. Also, Defiant accepts 10 different input formats widely used for DNA methylation data. CONCLUSIONS Defiant effectively identifies DMRs for whole-genome bisulfite sequencing (WGBS), reduced-representation bisulfite sequencing (RRBS), Tet-assisted bisulfite sequencing (TAB-seq), and HpaII tiny fragment enrichment by ligation-mediated PCR-tag (HELP) assays.
Collapse
Affiliation(s)
- David E Condon
- Department of Genetics, The Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Phu V Tran
- Department of Pediatrics, University of Minnesota, 2450 Riverside Avenue, Minneapolis, MN, 55454, USA
| | - Yu-Chin Lien
- Center for Research on Reproduction and Women's Health, University of Pennsylvania, 421 Curie Blvd, Philadelphia, PA, 19104, USA
| | - Jonathan Schug
- Department of Genetics, The Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Michael K Georgieff
- Department of Pediatrics, University of Minnesota, 2450 Riverside Avenue, Minneapolis, MN, 55454, USA
| | - Rebecca A Simmons
- Center for Research on Reproduction and Women's Health, University of Pennsylvania, 421 Curie Blvd, Philadelphia, PA, 19104, USA
| | - Kyoung-Jae Won
- Department of Genetics, The Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA. .,Biotech Research and Innovation Centre (BRIC), University of Copenhagen, 2200, Copenhagen, Denmark.
| |
Collapse
|
28
|
A new model for fetal programming: maternal Ramadan-type fasting programs nephrogenesis. J Dev Orig Health Dis 2018; 9:287-298. [PMID: 29317010 DOI: 10.1017/s204017441700109x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The effect of maternal Ramadan-type fasting (RTF) on the outcome of pregnancy, kidney development and nephron number in male rat offspring was investigated in current study. Pregnant rats were given food and water ad libitum during pregnancy (control) or restricted for 16 h per day (RTF). Kidney structure was examined during fetal life, at birth, and in early and late adulthood. Maternal body weight, food intake, relative food intake and plasma glucose levels were significantly lower (P<0.001) in the RTF group. Litter and pup weights also were significantly lower (P<0.05) in the RTF group at birth, with no difference in the litter size. The RTF group had a longer gestation, delayed nephrogenesis with less well-differentiated glomeruli, more connective tissue, fewer medullary rays, an increase in the nephrogenic zone/cortical zone ratio, and significant increase (P<0.001) in kidney apoptosis at birth. On the other hand, maternal fasting reduced nephron number (by ~31%) with unchanged kidney and total glomerular volumes. Mean glomerular volume was significantly higher in RTF offspring. Assessment of renal structure revealed mild glomerulosclerosis with enlarged lobulated glomeruli in the renal cortex and high interstitial fibrosis in the medulla of RTF kidneys. Taken together, gestational fasting delays nephrogenesis and reduces nephron number in the kidneys of the offspring, that could be partially owing to increased apoptosis.
Collapse
|
29
|
Chehade H, Simeoni U, Guignard JP, Boubred F. Preterm Birth: Long Term Cardiovascular and Renal Consequences. Curr Pediatr Rev 2018; 14:219-226. [PMID: 30101715 PMCID: PMC6416185 DOI: 10.2174/1573396314666180813121652] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 07/26/2018] [Accepted: 07/27/2018] [Indexed: 12/31/2022]
Abstract
BACKGROUND Cardiovascular and chronic kidney diseases are a part of noncommunicable chronic diseases, the leading causes of premature death worldwide. They are recognized as having early origins through altered developmental programming, due to adverse environmental conditions during development. Preterm birth is such an adverse factor. Rates of preterm birth increased in the last decades, however, with the improvement in perinatal and neonatal care, a growing number of preterm born subjects has now entered adulthood. Clinical and experimental evidence suggests that preterm birth is associated with impaired or arrested structural or functional development of key organs/systems making preterm infants vulnerable to cardiovascular and chronic renal diseases at adulthood. This review analyzes the evidence of such cardiovascular and renal changes, the role of perinatal and neonatal factors such as antenatal steroids and potential pathogenic mechanisms, including developmental programming and epigenetic alterations. CONCLUSION Preterm born subjects are exposed to a significantly increased risk for altered cardiovascular and renal functions at young adulthood. Adequate, specific follow-up measures remain to be determined. While antenatal steroids have considerably improved preterm birth outcomes, repeated therapy should be considered with caution, as antenatal steroids induce long-term cardiovascular and metabolic alterations in animals' models and their involvement in the accelerated cellular senescence observed in human studies cannot be excluded.
Collapse
Affiliation(s)
- Hassib Chehade
- Division of Pediatrics and DOHaD Lab, CHUV-UNIL, Lausanne, CH, Switzerland
| | - Umberto Simeoni
- Division of Pediatrics and DOHaD Lab, CHUV-UNIL, Lausanne, CH, Switzerland
| | | | - Farid Boubred
- Medecine Neonatale, Hopital La Conception, APHM, Aix-Marseille Universite, Marseille, France
| |
Collapse
|
30
|
Abstract
Hypertension (HT) is among the major components of the metabolic syndrome, i.e., obesity, dyslipidemia, and hyperglycemia/insulin resistance. It represents a significant health problem with foremost risks for chronic cardiovascular disease and a significant cause of morbidity and mortality worldwide. Therefore, it is not surprising that this disorder constitutes a serious public health concern. Although multiple studies have stressed the multifactorial nature of HT, the pathogenesis remains largely unknown. However, if we want to reduce the global prevalence of HT, restrain the number of deaths (currently 9.4 million/year in the world), and alleviate the socio-economic burden, a deeper insight into the mechanisms is urgently needed in order to define new meaningful therapeutic targets. Recently, the role of epigenetics in the development of various complex diseases has attracted much attention. In the present review, we provide a critical update on the available literature and ongoing research regarding the epigenetic modifications of genes involved in several pathways of elevated blood pressure, especially those linked to the vascular epithelium. This review also focuses on the role of microRNA (miRNA) in the regulation of gene expression associated with HT and of fetal programming mediating susceptibility to HT in adulthood.
Collapse
|
31
|
Tarry-Adkins JL, Ozanne SE. Nutrition in early life and age-associated diseases. Ageing Res Rev 2017; 39:96-105. [PMID: 27594376 DOI: 10.1016/j.arr.2016.08.003] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Revised: 03/24/2016] [Accepted: 08/05/2016] [Indexed: 02/06/2023]
Abstract
The prevalence of age-associated disease is increasing at a striking rate globally. It is known that a strong association exists between a suboptimal maternal and/or early-life environment and increased propensity of developing age-associated disease, including cardiovascular disease (CVD), type-2 diabetes (T2D) and obesity. The dissection of underlying molecular mechanisms to explain this phenomenon, which is known as 'developmental programming' is still emerging; however three common mechanisms have emerged in many models of developmental programming. These mechanisms are (a) changes in tissue structure, (b) epigenetic regulation and (c) accelerated cellular ageing. This review will examine the epidemiological evidence and the animal models of suboptimal maternal environments, focusing upon these molecular mechanisms and will discuss the progress being made in the development of safe and effective intervention strategies which ultimately could target those 'programmed' individuals who are known to be at-risk of age-associated disease.
Collapse
Affiliation(s)
- Jane L Tarry-Adkins
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Institute of Metabolic Science, Level 4, Box 289, Addenbrooke's Treatment Centre, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 OQQ, UK.
| | - Susan E Ozanne
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Institute of Metabolic Science, Level 4, Box 289, Addenbrooke's Treatment Centre, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 OQQ, UK.
| |
Collapse
|
32
|
El-Dahr S, Hilliard S, Saifudeen Z. Regulation of kidney development by the Mdm2/Mdm4-p53 axis. J Mol Cell Biol 2017; 9:26-33. [PMID: 28096292 PMCID: PMC5907835 DOI: 10.1093/jmcb/mjx005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 01/15/2017] [Indexed: 01/13/2023] Open
Abstract
While p53 activity is required for tumour suppression, unconstrained p53 activity on the other hand is detrimental to the organism, resulting in inappropriate cellular death or proliferation defects. Unimpeded p53 activity is lethal in the developing embryo, underlining the need for maintaining a tight control on p53 activity during this period. The critical role of the negative regulators of p53, Mdm2 and Mdm4, in vertebrate development came to light by fatal disruption of embryogenesis that was observed with Mdm2 and Mdm4 gene deletions in mice. Embryonic lethality was rescued only by superimposing p53 removal. Here we summarize the contribution of the Mdm2/Mdm4–p53 axis that occurs at multiple steps of kidney development. Conditional, cell type-specific deletions reveal distinct functions of these proteins in renal morphogenesis. The severe impact on the renal phenotype from targeted gene deletions underscores the critical role played by the Mdm2/Mdm4–p53 nexus on nephrogenesis, and emphasizes the need to monitor patients with aberrations in this pathway for kidney function defects and associated cardiovascular dysfunction.
Collapse
Affiliation(s)
- Samir El-Dahr
- Department of Pediatrics, Section of Pediatric Nephrology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Sylvia Hilliard
- Department of Pediatrics, Section of Pediatric Nephrology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Zubaida Saifudeen
- Department of Pediatrics, Section of Pediatric Nephrology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| |
Collapse
|
33
|
Ji C, Li Y, Cui L, Cai J, Shi J, Cheng FW, Li Y, Curhan GC, Wu S, Gao X. Prenatal Earthquake Exposure and Midlife Uric Acid Levels Among Chinese Adults. Arthritis Care Res (Hoboken) 2017; 69:703-708. [PMID: 27454342 DOI: 10.1002/acr.22973] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 05/26/2016] [Accepted: 06/21/2016] [Indexed: 01/21/2023]
Affiliation(s)
- Chunpeng Ji
- Hebei United University Kailuan General Hospital; Tangshan Hebei China
| | - Yanping Li
- Harvard School of Public Health; Boston Massachusetts
| | - Liufu Cui
- Hebei United University Kailuan General Hospital; Tangshan Hebei China
| | - Jianfang Cai
- Peking Union Medical College Hospital; Beijing China
| | - Jihong Shi
- Hebei United University Kailuan General Hospital; Tangshan Hebei China
| | - Feon W. Cheng
- Pennsylvania State University; State College Pennsylvania
| | - Yuqing Li
- Hebei United University Kailuan General Hospital; Tangshan Hebei China
| | - Gary C. Curhan
- Brigham and Women's Hospital and Harvard Medical School; Boston Massachusetts
| | - Shouling Wu
- Hebei United University Kailuan General Hospital; Tangshan Hebei China
| | - Xiang Gao
- Pennsylvania State University; State College Pennsylvania
| |
Collapse
|
34
|
Brophy P. Maternal determinants of renal mass and function in the fetus and neonate. Semin Fetal Neonatal Med 2017; 22:67-70. [PMID: 28347404 DOI: 10.1016/j.siny.2017.01.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The impact of adverse maternal and early gestational issues, ranging from maternal-fetal interactions all the way through to premature birth, are recognized as having influence on the subsequent development of chronic diseases later in life. The development of chronic kidney disease (CKD) as a direct result of early life renal injury or a sequela of diseases such as hypertension or diabetes is a good model example of the potential impact that early life events may have on renal development and lifelong function. The global monetary and human resource cost of CKD is exorbitant. Socio-economic factors, along with other factors (genetic and environmental) may significantly influence the timing and display of phenotypic expression in fetuses and neonates at risk for developing CKD, yet very few of these factors are studied or well understood. In general our focus has been directed at treatment once CKD is established. This strategy has been and remains short-sighted and costly. Earlier understanding of the intrauterine determinants of renal mass development (i.e. environmental "biomes", poor maternal-fetal health, socio-economic factors impacting early life events, diet, access to value based health care and educational opportunities on disease evolution) may allow us an opportunity for earlier intervention. This article aims to provide some foundation for improved understanding of the maternal determinants of renal mass and function in the fetus and neonate.
Collapse
Affiliation(s)
- Patrick Brophy
- University of Iowa Stead Family Children's Hospital, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
35
|
Wanner N, Bechtel-Walz W. Epigenetics of kidney disease. Cell Tissue Res 2017; 369:75-92. [PMID: 28286899 DOI: 10.1007/s00441-017-2588-x] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2016] [Accepted: 02/15/2017] [Indexed: 02/06/2023]
Abstract
DNA methylation and histone modifications determine renal programming and the development and progression of renal disease. The identification of the way in which the renal cell epigenome is altered by environmental modifiers driving the onset and progression of renal diseases has extended our understanding of the pathophysiology of kidney disease progression. In this review, we focus on current knowledge concerning the implications of epigenetic modifications during renal disease from early development to chronic kidney disease progression including renal fibrosis, diabetic nephropathy and the translational potential of identifying new biomarkers and treatments for the prevention and therapy of chronic kidney disease and end-stage kidney disease.
Collapse
Affiliation(s)
- Nicola Wanner
- Department of Medicine IV, Faculty of Medicine, University of Freiburg, Freiburg, Germany. .,Center for Systems Biology (ZBSA), Albert-Ludwigs-University, Freiburg, Germany. .,Renal Division, University Hospital Freiburg, Breisacher Strasse 66, 79106, Freiburg, Germany.
| | - Wibke Bechtel-Walz
- Department of Medicine IV, Faculty of Medicine, University of Freiburg, Freiburg, Germany. .,Renal Division, University Hospital Freiburg, Breisacher Strasse 66, 79106, Freiburg, Germany.
| |
Collapse
|
36
|
Murphy MO, Cohn DM, Loria AS. Developmental origins of cardiovascular disease: Impact of early life stress in humans and rodents. Neurosci Biobehav Rev 2017; 74:453-465. [PMID: 27450581 PMCID: PMC5250589 DOI: 10.1016/j.neubiorev.2016.07.018] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 07/06/2016] [Accepted: 07/19/2016] [Indexed: 02/07/2023]
Abstract
The Developmental Origins of Health and Disease (DOHaD) hypothesizes that environmental insults during childhood programs the individual to develop chronic disease in adulthood. Emerging epidemiological data strongly supports that early life stress (ELS) given by the exposure to adverse childhood experiences is regarded as an independent risk factor capable of predicting future risk of cardiovascular disease. Experimental animal models utilizing chronic behavioral stress during postnatal life, specifically maternal separation (MatSep) provides a suitable tool to elucidate molecular mechanisms by which ELS increases the risk to develop cardiovascular disease, including hypertension. The purpose of this review is to highlight current epidemiological studies linking ELS to the development of cardiovascular disease and to discuss the potential molecular mechanisms identified from animal studies. Overall, this review reveals the need for future investigations to further clarify the molecular mechanisms of ELS in order to develop more personalized therapeutics to mitigate the long-term consequences of chronic behavioral stress including cardiovascular and heart disease in adulthood.
Collapse
Affiliation(s)
- M O Murphy
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, United States
| | - D M Cohn
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, United States
| | - A S Loria
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, United States.
| |
Collapse
|
37
|
Tain YL, Hsu CN. Developmental Origins of Chronic Kidney Disease: Should We Focus on Early Life? Int J Mol Sci 2017; 18:ijms18020381. [PMID: 28208659 PMCID: PMC5343916 DOI: 10.3390/ijms18020381] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 01/24/2017] [Accepted: 02/03/2017] [Indexed: 12/20/2022] Open
Abstract
Chronic kidney disease (CKD) is becoming a global burden, despite recent advances in management. CKD can begin in early life by so-called "developmental programming" or "developmental origins of health and disease" (DOHaD). Early-life insults cause structural and functional changes in the developing kidney, which is called renal programming. Epidemiological and experimental evidence supports the proposition that early-life adverse events lead to renal programming and make subjects vulnerable to developing CKD and its comorbidities in later life. In addition to low nephron endowment, several mechanisms have been proposed for renal programming. The DOHaD concept opens a new window to offset the programming process in early life to prevent the development of adult kidney disease, namely reprogramming. Here, we review the key themes on the developmental origins of CKD. We have particularly focused on the following areas: evidence from human studies support fetal programming of kidney disease; insight from animal models of renal programming; hypothetical mechanisms of renal programming; alterations of renal transcriptome in response to early-life insults; and the application of reprogramming interventions to prevent the programming of kidney disease.
Collapse
Affiliation(s)
- You-Lin Tain
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan.
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan.
| | - Chien-Ning Hsu
- Department of Pharmacy, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan.
- School of Pharmacy, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| |
Collapse
|
38
|
N-acetylcysteine attenuates intrauterine growth retardation-induced hepatic damage in suckling piglets by improving glutathione synthesis and cellular homeostasis. Eur J Nutr 2016; 57:327-338. [DOI: 10.1007/s00394-016-1322-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 09/29/2016] [Indexed: 01/05/2023]
|
39
|
Wang YP, Chen X, Zhang ZK, Cui HY, Wang P, Wang Y. Increased renal apoptosis and reduced renin-angiotensin system in fetal growth restriction. J Renin Angiotensin Aldosterone Syst 2016; 17:17/3/1470320316654810. [PMID: 27534427 PMCID: PMC5843940 DOI: 10.1177/1470320316654810] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 04/05/2016] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVE The purpose of the study was to characterize changes in apoptosis and the renin-angiotensin system (RAS) in fetal growth restriction (FGR). MATERIALS AND METHOD Fetuses were collected from patients who visited our hospital to either terminate or abort their pregnancy. Kidneys of fetuses which suffered with FGR, (n=11) at gestational age of 33.4±0.5 weeks and those from non-FGR (n=12) at gestational age of 34.3±0.9 weeks were collected. TUNEL, Bax and Bcl-2 staining were examined. The number of nephrons was also counted. Both protein and mRNA levels of renin and angiotensinogen were analyzed. Ultrasound was applied to measure fetus parameters including biparietal diameter, head circumference, circumference of abdomen, and femur length. RESULTS The number of nephrons was positively correlated with fetal weight at termination. Kidneys in the FGR group presented more apoptotic cells than those in the non-FGR group. Renin and angiotensinogen both decreased in the FGR group. Ultrasound revealed that biparietal diameter, abdomen circumference, femur length, and birth weight were all reduced in the FGR group compared with the non-FGR group. Kidney size was also restricted in the FGR group as indicated by ultrasound. CONCLUSION Renal apoptosis might contribute to the reduction of nephrons, and ultrasound plays a vital role in early diagnosis of developmental origins of health and disease (DOHAD).
Collapse
Affiliation(s)
- Yan P Wang
- Department of Obstetrics, Tianjin Central Hospital of Gynecology and Obstetrics, China
| | - Xu Chen
- Department of Obstetrics, Tianjin Central Hospital of Gynecology and Obstetrics, China
| | - Zhi K Zhang
- Department of Obstetrics, Tianjin Central Hospital of Gynecology and Obstetrics, China
| | - Hong Y Cui
- Department of Obstetrics, Tianjin Central Hospital of Gynecology and Obstetrics, China
| | - Peng Wang
- Department of Obstetrics, Tianjin Central Hospital of Gynecology and Obstetrics, China
| | - Yue Wang
- Department of Obstetrics, Tianjin Central Hospital of Gynecology and Obstetrics, China
| |
Collapse
|
40
|
Fani S, Dehghan F, Karimian H, Mun Lo K, Ebrahimi Nigjeh S, Swee Keong Y, Soori R, May Chow K, Kamalidehghan B, Mohd Ali H, Mohd Hashim N. Monobenzyltin Complex C1 Induces Apoptosis in MCF-7 Breast Cancer Cells through the Intrinsic Signaling Pathway and through the Targeting of MCF-7-Derived Breast Cancer Stem Cells via the Wnt/β-Catenin Signaling Pathway. PLoS One 2016; 11:e0160836. [PMID: 27529753 PMCID: PMC4986984 DOI: 10.1371/journal.pone.0160836] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 07/26/2016] [Indexed: 01/06/2023] Open
Abstract
Monobenzyltin Schiff base complex, [N-(3,5-dichloro-2-oxidobenzylidene)-4-chlorobenzyhydrazidato](o-methylbenzyl)aquatin(IV) chloride, C1, is an organotin non-platinum metal-based agent. The present study was conducted to investigate its effects on MCF-7 cells with respect to the induction of apoptosis and its inhibitory effect against MCF-7 breast cancer stem cells. As determined in a previous study, compound C1 revealed strong antiproliferative activity on MCF-7 cells with an IC50 value of 2.5 μg/mL. Annexin V/propidium iodide staining coupled with flow cytometry indicated the induction of apoptosis in treated cells. Compound C1 induced apoptosis in MCF-7 cells and was mediated through the intrinsic pathway with a reduction in mitochondrial membrane potential and mitochondrial cytochrome c release to cytosol. Complex C1 activated caspase 9 as a result of cytochrome c release. Subsequently, western blot and real time PCR revealed a significant increase in Bax and Bad expression and a significant decrease in the expression levels of Bcl2 and HSP70. Furthermore, a flow cytometric analysis showed that treatment with compound C1 caused a significant arrest of MCF-7 cells in G0/G1 phase. The inhibitory analysis of compound C1 against derived MCF-7 stem cells showed a significant reduction in the aldehyde dehydrogenase-positive cell population and a significant reduction in the population of MCF-7 cancer stem cells in primary, secondary, and tertiary mammospheres. Moreover, treatment with C1 down-regulated the Wnt/β-catenin self-renewal pathway. These findings indicate that complex C1 is a suppressive agent of MCF-7 cells that functions through the induction of apoptosis, cell cycle arrest, and the targeting of MCF-7-derived cancer stem cells. This work may lead to a better treatment strategy for the reduction of breast cancer recurrence.
Collapse
Affiliation(s)
- Somayeh Fani
- Department of Pharmacy, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
- * E-mail: (SF); (NMH)
| | - Firouzeh Dehghan
- Department of Exercise Physiology, Faculty of Physical Education and Sport Sciences, University of Tehran, 14174 Tehran, Iran
- Department of Exercise Science, Sports Center, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Hamed Karimian
- Department of Pharmacy, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Kong Mun Lo
- Department of Chemistry, Faculty of Science, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | | | - Yeap Swee Keong
- Institute of Bioscience, University Putra Malaysia, 43400 Serdang, Malaysia
| | - Rahman Soori
- Department of Exercise Physiology, Faculty of Physical Education and Sport Sciences, University of Tehran, 14174 Tehran, Iran
| | - Kit May Chow
- Department of Chemistry, Faculty of Science, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Behnam Kamalidehghan
- Medical Genetics Department, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran-Karaj Highway, Tehran, Iran
| | - Hapipah Mohd Ali
- Department of Chemistry, Faculty of Science, University of Malaya, 50603 Kuala Lumpur, Malaysia
- Center for Natural Products and Drug Discovery (CENAR), Department of Chemistry, Faculty of Science, University of Malaya, Kuala Lumpur, Malaysia
| | - Najihah Mohd Hashim
- Department of Pharmacy, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
- Center for Natural Products and Drug Discovery (CENAR), Department of Chemistry, Faculty of Science, University of Malaya, Kuala Lumpur, Malaysia
- * E-mail: (SF); (NMH)
| |
Collapse
|
41
|
Wang Z, Li C, Yang Z, Zou Z, Ma J. Infant exposure to Chinese famine increased the risk of hypertension in adulthood: results from the China Health and Retirement Longitudinal Study. BMC Public Health 2016; 16:435. [PMID: 27225778 PMCID: PMC4880986 DOI: 10.1186/s12889-016-3122-x] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Accepted: 05/17/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Early-life developmental adaptations in response to severe malnutrition may play a crucial role in susceptibility to hypertension. This study aimed to explore the associations between exposure to the Chinese famine (1959-1961) at fetal, infant and preschool stages during fetal life or childhood and the risk of hypertension in adulthood. METHODS We used the data of 1,966 adults born between 1956 and 1964 in selected families from the China Health and Retirement Longitudinal Study (CHARLS) national survey. RESULTS Prevalence of hypertension among adults in non-exposed, fetal-exposed, infant-exposed, and preschool-exposed cohorts was 18.9, 20.7, 28.7, and 23.4 %, respectively. In severely affected famine areas, only infant-exposed cohort had a significant increased risk of hypertension compared with non-exposed cohort (OR 2.12; 95 % CI 1.19, 3.79; P = 0.011), and the significance remained after adjusted gender, smoking, and drinking (OR 2.11; 95 % CI 1.18, 3.77; P = 0.012). After stratification by BMI and economic status, the risk of hypertension was higher for subjects with BMI ≥ 24 kg/m(2)(OR 2.09; 95 % CI 1.09, 4.01; P = 0.026) or high economic status(OR 2.26; 95 % CI 1.19, 4.31; P = 0.013) than those with BMI < 24 kg/m(2)(OR 1.65; 95 % CI 0.71, 3.83; P = 0.246) or low economic status (OR 2.18; 95 % CI 1.14, 4.18; P = 0.019) in infant-exposed cohort of severely affected famine areas. However, there was no consistent association observed in less severely affected area or other exposed cohorts in severely affected areas. CONCLUSIONS Infanthood exposed to famine might increase the risk of hypertension in adulthood, and a postnatal 'rich' nutrient environment further increased the risk.
Collapse
Affiliation(s)
- Zhenghe Wang
- Institute of Child and Adolescent Health, School of Public Health, Peking University Health Science Center, No 38 Xue Yuan Road, Haidian District, Beijing, 100191, China
| | - Changwei Li
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA
| | - Zhongping Yang
- Institute of Child and Adolescent Health, School of Public Health, Peking University Health Science Center, No 38 Xue Yuan Road, Haidian District, Beijing, 100191, China
| | - Zhiyong Zou
- Institute of Child and Adolescent Health, School of Public Health, Peking University Health Science Center, No 38 Xue Yuan Road, Haidian District, Beijing, 100191, China.
| | - Jun Ma
- Institute of Child and Adolescent Health, School of Public Health, Peking University Health Science Center, No 38 Xue Yuan Road, Haidian District, Beijing, 100191, China.
| |
Collapse
|
42
|
Morton JS, Cooke CL, Davidge ST. In Utero Origins of Hypertension: Mechanisms and Targets for Therapy. Physiol Rev 2016; 96:549-603. [DOI: 10.1152/physrev.00015.2015] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The developmental origins of health and disease theory is based on evidence that a suboptimal environment during fetal and neonatal development can significantly impact the evolution of adult-onset disease. Abundant evidence exists that a compromised prenatal (and early postnatal) environment leads to an increased risk of hypertension later in life. Hypertension is a silent, chronic, and progressive disease defined by elevated blood pressure (>140/90 mmHg) and is strongly correlated with cardiovascular morbidity/mortality. The pathophysiological mechanisms, however, are complex and poorly understood, and hypertension continues to be one of the most resilient health problems in modern society. Research into the programming of hypertension has proposed pharmacological treatment strategies to reverse and/or prevent disease. In addition, modifications to the lifestyle of pregnant women might impart far-reaching benefits to the health of their children. As more information is discovered, more successful management of hypertension can be expected to follow; however, while pregnancy complications such as fetal growth restriction, preeclampsia, preterm birth, etc., continue to occur, their offspring will be at increased risk for hypertension. This article reviews the current knowledge surrounding the developmental origins of hypertension, with a focus on mechanistic pathways and targets for therapeutic and pharmacologic interventions.
Collapse
Affiliation(s)
- Jude S. Morton
- Departments of Obstetrics and Gynaecology and of Physiology, University of Alberta, Edmonton, Canada; Women and Children's Health Research Institute, Edmonton, Canada; and Cardiovascular Research Centre, Edmonton, Canada
| | - Christy-Lynn Cooke
- Departments of Obstetrics and Gynaecology and of Physiology, University of Alberta, Edmonton, Canada; Women and Children's Health Research Institute, Edmonton, Canada; and Cardiovascular Research Centre, Edmonton, Canada
| | - Sandra T. Davidge
- Departments of Obstetrics and Gynaecology and of Physiology, University of Alberta, Edmonton, Canada; Women and Children's Health Research Institute, Edmonton, Canada; and Cardiovascular Research Centre, Edmonton, Canada
| |
Collapse
|
43
|
Tang XS, Shen Q, Chen J, Zha XL, Xu H. Maternal protein restriction reduces perlecan at mid-metanephrogenesis in rats. Nephrology (Carlton) 2016; 21:200-8. [PMID: 26246161 DOI: 10.1111/nep.12583] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/04/2015] [Indexed: 12/19/2022]
Affiliation(s)
- Xiao-Shan Tang
- Department of Nephrology and Rheumatology; Children's Hospital of Fudan University; Shanghai China
| | - Qian Shen
- Department of Nephrology and Rheumatology; Children's Hospital of Fudan University; Shanghai China
| | - Jing Chen
- Department of Nephrology and Rheumatology; Children's Hospital of Fudan University; Shanghai China
| | - Xi-Liang Zha
- Department of Biochemistry and Molecular Biology; Shanghai Medical College; Fudan University; Shanghai China
| | - Hong Xu
- Department of Nephrology and Rheumatology; Children's Hospital of Fudan University; Shanghai China
| |
Collapse
|
44
|
Reynolds CM, Gray C, Li M, Segovia SA, Vickers MH. Early Life Nutrition and Energy Balance Disorders in Offspring in Later Life. Nutrients 2015; 7:8090-111. [PMID: 26402696 PMCID: PMC4586579 DOI: 10.3390/nu7095384] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Revised: 08/31/2015] [Accepted: 09/11/2015] [Indexed: 02/07/2023] Open
Abstract
The global pandemic of obesity and type 2 diabetes is often causally linked to changes in diet and lifestyle; namely increased intake of calorically dense foods and concomitant reductions in physical activity. Epidemiological studies in humans and controlled animal intervention studies have now shown that nutritional programming in early periods of life is a phenomenon that affects metabolic and physiological functions throughout life. This link is conceptualised as the developmental programming hypothesis whereby environmental influences during critical periods of developmental plasticity can elicit lifelong effects on the health and well-being of the offspring. The mechanisms by which early environmental insults can have long-term effects on offspring remain poorly defined. However there is evidence from intervention studies which indicate altered wiring of the hypothalamic circuits that regulate energy balance and epigenetic effects including altered DNA methylation of key adipokines including leptin. Studies that elucidate the mechanisms behind these associations will have a positive impact on the health of future populations and adopting a life course perspective will allow identification of phenotype and markers of risk earlier, with the possibility of nutritional and other lifestyle interventions that have obvious implications for prevention of non-communicable diseases.
Collapse
Affiliation(s)
- Clare M Reynolds
- Liggins Institute and Gravida: National Centre for Growth and Development, University of Auckland, Auckland 1142, New Zealand.
| | - Clint Gray
- Liggins Institute and Gravida: National Centre for Growth and Development, University of Auckland, Auckland 1142, New Zealand.
| | - Minglan Li
- Liggins Institute and Gravida: National Centre for Growth and Development, University of Auckland, Auckland 1142, New Zealand.
| | - Stephanie A Segovia
- Liggins Institute and Gravida: National Centre for Growth and Development, University of Auckland, Auckland 1142, New Zealand.
| | - Mark H Vickers
- Liggins Institute and Gravida: National Centre for Growth and Development, University of Auckland, Auckland 1142, New Zealand.
| |
Collapse
|
45
|
Early life obesity and chronic kidney disease in later life. Pediatr Nephrol 2015; 30:1255-63. [PMID: 25145270 DOI: 10.1007/s00467-014-2922-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Revised: 04/23/2014] [Accepted: 07/17/2014] [Indexed: 12/16/2022]
Abstract
The prevalence of chronic kidney disease (CKD) has increased considerably with a parallel rise in the prevalence of obesity. It is now recognized that early life nutrition has life-long effects on the susceptibility of an individual to develop obesity, diabetes, cardiovascular disease and CKD. The kidney can be programmed by a number of intrauterine and neonatal insults. Low birth weight (LBW) is one of the most identifiable markers of a suboptimal prenatal environment, and the important intrarenal factors sensitive to programming events include decreased nephron number and altered control of the renin-angiotensin system (RAS). LBW complicated by accelerated catch-up growth is associated with an increased risk of obesity, hypertension and CKD in later life. High birth weight and exposure to maternal diabetes or obesity can enhance the risk for developing CKD in later life. Rapid postnatal growth per se may also contribute to the subsequent development of obesity and CKD regardless of birth weight and prenatal nutrition. Although the mechanisms of renal risks due to early life nutritional programming remain largely unknown, experimental and clinical studies suggest the burdening role of early life obesity in longstanding cardiovascular and renal diseases.
Collapse
|
46
|
IUGR prevents IGF-1 upregulation in juvenile male mice by perturbing postnatal IGF-1 chromatin remodeling. Pediatr Res 2015; 78:14-23. [PMID: 25826117 DOI: 10.1038/pr.2015.70] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 01/09/2015] [Indexed: 12/11/2022]
Abstract
BACKGROUND Intrauterine growth restriction (IUGR) offspring with rapid catch-up growth are at increased risk for early obesity especially in males. Persistent insulin-like growth factor-1 (IGF-1) reduction is an important risk factor. Using a mouse model of maternal hypertension-induced IUGR, we examined IGF-1 levels, promoter DNA methylation, and histone H3 covalent modifications at birth (D1). We additionally investigated whether prenatal perturbations could reset at preadolescence (D21). METHODS IUGR was induced via maternal thromboxane A2-analog infusion in mice. RESULTS IUGR uniformly decreased D1 IGF-1 mRNA and protein levels with reduced promoter 1 (P1) transcription and increased P1 DNA methylation. IUGR males also had increased H3K4ac at exon 5 and 3' distal UTR. At D21, IUGR males continued to have decreased IGF-1 levels, originating from both P1 and P2 with reduced 1A variant. IUGR males also had decreased activation mark of H3K4me3 at P1 compared with sham males. In contrast, D21 IUGR females normalized their IGF-1 levels, in association with an increased activation mark of H3K4me3 at P1 compared with sham females. CONCLUSION IUGR uniformly affected D1 hepatic IGF-1 epigenetic modifications in both sexes. However, at preadolescence, IUGR males are unable to correct for the prenatal reduction possibly due to a more perturbed IGF-1 chromatin structure.
Collapse
|
47
|
Postnatal nutritional restriction affects growth and immune function of piglets with intra-uterine growth restriction. Br J Nutr 2015; 114:53-62. [DOI: 10.1017/s0007114515001579] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Postnatal rapid growth by excess intake of nutrients has been associated with an increased susceptibility to diseases in neonates with intra-uterine growth restricted (IUGR). The aim of the present study was to determine whether postnatal nutritional restriction could improve intestinal development and immune function of neonates with IUGR using piglets as model. A total of twelve pairs of normal-birth weight (NBW) and IUGR piglets (7 d old) were randomly assigned to receive adequate nutrient intake or restricted nutrient intake (RNI) by artificially liquid feeding for a period of 21 d. Blood samples and intestinal tissues were collected at necropsy and were analysed for morphology, digestive enzyme activities, immune cells and expression of innate immunity-related genes. The results indicated that both IUGR and postnatal nutritional restriction delayed the growth rate during the sucking period. Irrespective of nutrient intake, piglets with IUGR had a significantly lower villous height and crypt depth in the ileum than the NBW piglets. Moreover, IUGR decreased alkaline phosphatase activity while enhanced lactase activity in the jejunum and mRNA expressions of Toll-like receptor 9 (TLR-9) and DNA methyltransferase 1 (DNMT1) in the ileum of piglets. Irrespective of body weight, RNI significantly decreased the number and/or percentage of peripheral leucocytes, lymphocytes and monocytes of piglets, whereas the percentage of neutrophils and the ratio of CD4+ to CD8+ were increased. Furthermore, RNI markedly enhanced the mRNA expression of TLR-9 and DNMT1, but decreased the expression of NOD2 and TRAF-6 in the ileum of piglets. In summary, postnatal nutritional restriction led to abnormal cellular and innate immune response, as well as delayed the growth and intestinal development of IUGR piglets.
Collapse
|
48
|
Seferovic MD, Goodspeed DM, Chu DM, Krannich LA, Gonzalez-Rodriguez PJ, Cox JE, Aagaard KM. Heritable IUGR and adult metabolic syndrome are reversible and associated with alterations in the metabolome following dietary supplementation of 1-carbon intermediates. FASEB J 2015; 29:2640-52. [PMID: 25757570 PMCID: PMC4447228 DOI: 10.1096/fj.14-266387] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Accepted: 02/19/2015] [Indexed: 12/16/2022]
Abstract
Metabolic syndrome (MetS), following intrauterine growth restriction (IUGR), is epigenetically heritable. Recently, we abrogated the F2 adult phenotype with essential nutrient supplementation (ENS) of intermediates along the 1-carbon pathway. With the use of the same grandparental uterine artery ligation model, we profiled the F2 serum metabolome at weaning [postnatal day (d)21; n = 76] and adulthood (d160; n = 12) to test if MetS is preceded by alterations in the metabolome. Indicative of developmentally programmed MetS, adult F2, formerly IUGR rats, were obese (621 vs. 461 g; P < 0.0001), dyslipidemic (133 vs. 67 mg/dl; P < 0.001), and glucose intolerant (26 vs. 15 mg/kg/min; P < 0.01). Unbiased gas chromatography-mass spectrometry (GC-MS) profiling revealed 34 peaks corresponding to 12 nonredundant metabolites and 9 unknowns to be changing at weaning [false discovery rate (FDR) < 0.05]. Markers of later-in-life MetS included citric acid, glucosamine, myoinositol, and proline (P < 0.03). Hierarchical clustering revealed grouping by IUGR lineage and supplementation at d21 and d160. Weanlings grouped distinctly for ENS and IUGR by partial least-squares discriminate analysis (PLS-DA; P < 0.01), whereas paternal and maternal IUGR (IUGR(pat)/IUGR(mat), respectively) control-fed rats, destined for MetS, had a distinct metabolome at weaning (randomForest analysis; class error < 0.1) and adulthood (PLS-DA; P < 0.05). In sum, we have found that alterations in the metabolome accompany heritable IUGR, precede adult-onset MetS, and are partially amenable to dietary intervention.
Collapse
Affiliation(s)
- Maxim D Seferovic
- Departments of *Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, and Molecular and Cell Biology, Molecular and Human Genetics, and Molecular Physiology and Biophysics, and Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, Texas, USA; and Department of Biochemistry and Metabolomics Core, University of Utah, Salt Lake City, Utah, USA
| | - Danielle M Goodspeed
- Departments of *Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, and Molecular and Cell Biology, Molecular and Human Genetics, and Molecular Physiology and Biophysics, and Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, Texas, USA; and Department of Biochemistry and Metabolomics Core, University of Utah, Salt Lake City, Utah, USA
| | - Derrick M Chu
- Departments of *Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, and Molecular and Cell Biology, Molecular and Human Genetics, and Molecular Physiology and Biophysics, and Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, Texas, USA; and Department of Biochemistry and Metabolomics Core, University of Utah, Salt Lake City, Utah, USA
| | - Laura A Krannich
- Departments of *Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, and Molecular and Cell Biology, Molecular and Human Genetics, and Molecular Physiology and Biophysics, and Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, Texas, USA; and Department of Biochemistry and Metabolomics Core, University of Utah, Salt Lake City, Utah, USA
| | - Pablo J Gonzalez-Rodriguez
- Departments of *Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, and Molecular and Cell Biology, Molecular and Human Genetics, and Molecular Physiology and Biophysics, and Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, Texas, USA; and Department of Biochemistry and Metabolomics Core, University of Utah, Salt Lake City, Utah, USA
| | - James E Cox
- Departments of *Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, and Molecular and Cell Biology, Molecular and Human Genetics, and Molecular Physiology and Biophysics, and Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, Texas, USA; and Department of Biochemistry and Metabolomics Core, University of Utah, Salt Lake City, Utah, USA
| | - Kjersti M Aagaard
- Departments of *Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, and Molecular and Cell Biology, Molecular and Human Genetics, and Molecular Physiology and Biophysics, and Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, Texas, USA; and Department of Biochemistry and Metabolomics Core, University of Utah, Salt Lake City, Utah, USA
| |
Collapse
|
49
|
Awazu M, Hida M. Maternal nutrient restriction inhibits ureteric bud branching but does not affect the duration of nephrogenesis in rats. Pediatr Res 2015; 77:633-9. [PMID: 25675424 DOI: 10.1038/pr.2015.24] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Accepted: 10/29/2014] [Indexed: 12/17/2022]
Abstract
BACKGROUND Maternal nutrient restriction produces offspring with fewer nephrons. We studied whether the reduced nephron number is due to the inhibition of ureteric branching or early cessation of nephrogenesis in rats. Signaling pathways involved in kidney development were also examined. METHODS The offspring of dams given food ad libitum (control (CON)) and those subjected to 50% food restriction (nutrient restriceted (NR)) were examined. RESULTS At embryonic day 13 (E13), there was no difference between NR and CON in body weight or kidney size. Ureteric buds branched once in both NR and CON. At E14 and E15, body and kidney size were significantly reduced in NR. Ureteric bud tip numbers were also reduced to 50% of CON. On the other hand, the disappearance of nephrogenic zone and a nephron progenitor marker Cited1 was not different between CON and NR. The final glomerular number of NR was 80% of CON. Activated extracellular signal-regulated kinase (ERK), p38, PI3K, Akt, and mammallian target of rapamycin (mTOR), and protein expression of β-catenin were downregulated at E15. CONCLUSION Ureteric branching is inhibited and developmentally regulated signaling pathways are downregulated at an early stage by maternal nutrient restriction. These changes, not early cessation of nephrogenesis, may be a mechanism for the inhibited kidney growth and nephrogenesis.
Collapse
Affiliation(s)
- Midori Awazu
- Department of Pediatrics, Keio University School of Medicine, Tokyo, Japan
| | - Mariko Hida
- 1] Department of Pediatrics, Keio University School of Medicine, Tokyo, Japan [2] Perinatal Center and Department of Neonatology, Yokohama Rosai Hospital, Kanagawa, Japan
| |
Collapse
|
50
|
Abstract
Low birth weight serves as a crude proxy for impaired growth during fetal life and indicates a failure for the fetus to achieve its full growth potential. Low birth weight can occur in response to numerous etiologies that include complications during pregnancy, poor prenatal care, parental smoking, maternal alcohol consumption, or stress. Numerous epidemiological and experimental studies demonstrate that birth weight is inversely associated with blood pressure and coronary heart disease. Sex and age impact the developmental programming of hypertension. In addition, impaired growth during fetal life also programs enhanced vulnerability to a secondary insult. Macrosomia, which occurs in response to maternal obesity, diabetes, and excessive weight gain during gestation, is also associated with increased cardiovascular risk. Yet, the exact mechanisms that permanently change the structure, physiology, and endocrine health of an individual across their lifespan following altered growth during fetal life are not entirely clear. Transmission of increased risk from one generation to the next in the absence of an additional prenatal insult indicates an important role for epigenetic processes. Experimental studies also indicate that the sympathetic nervous system, the renin angiotensin system, increased production of oxidative stress, and increased endothelin play an important role in the developmental programming of blood pressure in later life. Thus, this review will highlight how adverse influences during fetal life and early development program an increased risk for cardiovascular disease including high blood pressure and provide an overview of the underlying mechanisms that contribute to the fetal origins of cardiovascular pathology.
Collapse
Affiliation(s)
- Barbara T Alexander
- Department of Physiology and Biophysics, Women's Health Research Center, Center for Cardiovascular-Renal Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | | | | |
Collapse
|