1
|
Unger HW, Dadi A, Brown K, Simon D, Guthridge S. Intergenerational risk of preterm birth in First Nations Australians: a population-based cohort study from the Northern Territory, Australia. BMC Pregnancy Childbirth 2024; 24:855. [PMID: 39716120 DOI: 10.1186/s12884-024-07053-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 12/09/2024] [Indexed: 12/25/2024] Open
Abstract
BACKGROUND PTB increases the risk of health problems such as chronic renal disease and diabetes in later life and adverse impacts are inversely correlated with gestational age at birth. Rates of PTB in the Northern Territory (NT) of Australia are amongst the highest nationally and globally, with First Nations babies most affected. This study assessed the magnitude and potential drivers of intergenerational PTB recurrence in the NT. METHODS A retrospective intergenerational cohort study (1986-2017) was conducted amongst 5,366 mothers born singleton who had 9,571 singleton live births (7,673 First Nations, and 1,898 non-First Nations babies). Maternal and offspring PTB was categorised as early (< 34 weeks) and late (34-36 gestational weeks). Modified Poisson regression was used to estimate the relative risk (RR) of PTB associated with maternal PTB, adjusting for moderators such as receipt of antenatal care prior to the offspring PTB. Secondary analyses assessed the impact of additional adjustment for conditions with a familial component, or that PTB predisposes to, on the risk estimate. Mediation analysis assessed the degree of mediation of maternal-offspring PTB relationships by these conditions. RESULTS Overall, First Nations women born preterm (< 37 weeks) had an increased risk of delivering before 37 gestational weeks (aRR 1.28; 95%CI 1.08, 1.51). Women born preterm had a higher risk of delivering early (< 34 gestational weeks) but not late preterm (34-36 weeks): the risk of early offspring PTB was increased amongst women themselves born early preterm (aRR 1.95, 95%CI 1.17, 3.24) or late preterm (aRR 1.41, 95%CI 1.01, 1.97). Adjustment for pre-eclampsia, intrauterine growth restriction, and hypertensive renal disease attenuated the observed intergenerational PTB associations. Mediation analysis suggested these conditions may mediate up to 26% of the observed intergenerational PTB recurrence. Similar trends were observed when first-time mothers were considered only. Maternal PTB status was not associated with PTB amongst non-First Nations women. CONCLUSIONS First Nations women born preterm have an increased risk of early PTB. This association is in part driven by pre-eclampsia and hypertensive renal disease. Routine inquiry of maternal birth status may be a useful tool to identify NT First Nations women who may benefit from preventative measures.
Collapse
Affiliation(s)
- Holger W Unger
- Menzies School of Health Research, Charles Darwin University, Darwin, NT, Australia.
- Department of Obstetrics and Gynaecology, Royal Darwin Hospital, Darwin, NT, Australia.
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK.
- Department of Infectious Diseases, University of Melbourne, Melbourne, Australia.
- Menzies School of Health Research, Northern Territory, Charles Darwin University, PO Box 41096, Casuarina, 0810, Australia.
| | - Abel Dadi
- Menzies School of Health Research, Charles Darwin University, Darwin, NT, Australia
- Addis Continental Institute of Public Health, Addis Ababa, Ethiopia
| | - Kiarna Brown
- Menzies School of Health Research, Charles Darwin University, Darwin, NT, Australia
- Department of Obstetrics and Gynaecology, Royal Darwin Hospital, Darwin, NT, Australia
| | - David Simon
- Katherine District Hospital, Katherine, NT, Australia
| | - Steven Guthridge
- Menzies School of Health Research, Charles Darwin University, Darwin, NT, Australia
| |
Collapse
|
2
|
Brennan S, Rudd D, Watson D, Kandasamy Y. The relationship between maternal health during pregnancy and infant kidney development: a prospective cohort study. J Nephrol 2024; 37:2531-2539. [PMID: 39638985 DOI: 10.1007/s40620-024-02141-0] [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: 04/18/2024] [Accepted: 10/19/2024] [Indexed: 12/07/2024]
Abstract
BACKGROUND The significance of intergenerational impacts on fetal and infant kidney development and function remains to be fully understood. This is particularly relevant for certain populations, for example the Indigenous Australians since their risk of developing chronic kidney disease (CKD) is twice that of non-Indigenous Australians. The aim of this study was to assess the impact of maternal health and kidney size and function on infant kidney development. METHODS This study was open to all pregnant women receiving antenatal care at Townsville University Hospital, Australia. It presents data from a larger, ongoing prospective, longitudinal cohort study which commenced August 2019, involving mother-infant dyads. This manuscript reports on term mother-infants' dyads from singleton pregnancies. Ultrasound was used to measure renal parenchymal thickness, a surrogate for nephron number, of the mother and their newborn. Kidney function was assessed using serum cystatin C and creatinine. RESULTS Analysis was conducted on 80 mother-infant dyads, 17 Indigenous and 63 non-Indigenous. Multivariate regression modeling showed maternal renal parenchymal thickness (ß = 0.31, p = 0.004), smoking (ß = - 0.70, p = 0.022) and maternal serum cystatin C (ß = - 0.34, p = 0.014) significantly predicted newborn renal parenchymal thickness. No significant differences were found between the maternal and newborn renal parenchymal thickness and function between Indigenous and non-Indigenous participants. CONCLUSIONS Our study suggests that maternal kidney size and function has a significant intergenerational effect on kidney development of their infants. Newborn renal parenchymal thickness was positively associated with maternal renal parenchymal thickness and negatively associated with smoking and maternal serum cystatin C.
Collapse
Affiliation(s)
- Sonja Brennan
- Maternal Fetal Medicine and Ultrasound, Townsville University Hospital, Douglas, Townsville, Australia.
- Division of Tropical Health and Medicine, James Cook University, Townsville, Australia.
| | - Donna Rudd
- Division of Tropical Health and Medicine, James Cook University, Townsville, Australia
| | - David Watson
- Division of Tropical Health and Medicine, James Cook University, Townsville, Australia
- Department of Maternal Fetal Medicine, Townsville University Hospital, Townsville, Australia
| | - Yogavijayan Kandasamy
- Division of Tropical Health and Medicine, James Cook University, Townsville, Australia
- Department of Neonatology, Townsville University Hospital, Townsville, Australia
- Mothers and Babies Research Centre, Hunter Medical Research Institute, John Hunter Hospital, The University of Newcastle, Newcastle, Australia
| |
Collapse
|
3
|
Doan TNA, Cowley JM, Phillips AL, Briffa JF, Leemaqz SY, Burton RA, Romano T, Wlodek ME, Bianco-Miotto T. Imprinted gene alterations in the kidneys of growth restricted offspring may be mediated by a long non-coding RNA. Epigenetics 2024; 19:2294516. [PMID: 38126131 PMCID: PMC10761017 DOI: 10.1080/15592294.2023.2294516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 12/06/2023] [Indexed: 12/23/2023] Open
Abstract
Altered epigenetic mechanisms have been previously reported in growth restricted offspring whose mothers experienced environmental insults during pregnancy in both human and rodent studies. We previously reported changes in the expression of the DNA methyltransferase Dnmt3a and the imprinted genes Cdkn1c (Cyclin-dependent kinase inhibitor 1C) and Kcnq1 (Potassium voltage-gated channel subfamily Q member 1) in the kidney tissue of growth restricted rats whose mothers had uteroplacental insufficiency induced on day 18 of gestation, at both embryonic day 20 (E20) and postnatal day 1 (PN1). To determine the mechanisms responsible for changes in the expression of these imprinted genes, we investigated DNA methylation of KvDMR1, an imprinting control region (ICR) that includes the promoter of the antisense long non-coding RNA Kcnq1ot1 (Kcnq1 opposite strand/antisense transcript 1). Kcnq1ot1 expression decreased by 51% in growth restricted offspring compared to sham at PN1. Interestingly, there was a negative correlation between Kcnq1ot1 and Kcnq1 in the E20 growth restricted group (Spearman's ρ = 0.014). No correlation was observed between Kcnq1ot1 and Cdkn1c expression in either group at any time point. Additionally, there was a 11.25% decrease in the methylation level at one CpG site within KvDMR1 ICR. This study, together with others in the literature, supports that long non-coding RNAs may mediate changes seen in tissues of growth restricted offspring.
Collapse
Affiliation(s)
- Thu N. A. Doan
- School of Agriculture, Food and Wine, & Waite Research Institute, University of Adelaide, Adelaide, South Australia, Australia
- Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - James M. Cowley
- School of Agriculture, Food and Wine, & Waite Research Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - Aaron L. Phillips
- School of Agriculture, Food and Wine, & Waite Research Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - Jessica F. Briffa
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, Victoria, Australia
| | - Shalem Y. Leemaqz
- Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
- SAHMRI Women and Kids, South Australian Health & Medical Research Institute, Adelaide, South Australia, Australia
- College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia
| | - Rachel A. Burton
- School of Agriculture, Food and Wine, & Waite Research Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - Tania Romano
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, Victoria, Australia
| | - Mary E. Wlodek
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, Victoria, Australia
- Department of Obstetrics and Gynaecology, The University of Melbourne, Parkville, Victoria, Australia
| | - Tina Bianco-Miotto
- School of Agriculture, Food and Wine, & Waite Research Institute, University of Adelaide, Adelaide, South Australia, Australia
- Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
4
|
Zhang B, Ban M, Chen X, Zhang Y, Wang Z, Feng W, Zhao H, Li J, Zhang T, Hu J, Hu K, Cui L, Chen ZJ. Associations Between Paternal Obesity and Cardiometabolic Alterations in Offspring via Assisted Reproductive Technology. J Clin Endocrinol Metab 2024; 109:e2309-e2316. [PMID: 38375892 PMCID: PMC11570360 DOI: 10.1210/clinem/dgae096] [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: 08/12/2023] [Revised: 02/01/2024] [Accepted: 02/16/2024] [Indexed: 02/21/2024]
Abstract
CONTEXT Both assisted reproductive technology (ART) and obesity are associated with adverse cardiometabolic alterations in offspring. However, the combined effects of paternal obesity and ART on offspring cardiometabolic health are still unclear. OBJECTIVE To clarify cardiometabolic changes in offspring of obese fathers conceived using ART. This was a retrospective cohort study conducted between June 2014 and October 2019 at a center for reproductive medicine. A total of 2890 singleton visits aged 4-10 years were followed. Age-and sex-specific z-score of body mass index (BMI), blood pressure, insulin resistance, and lipid profile were examined. RESULTS We observed a strong association between paternal BMI categories and offspring BMI, blood pressure, and insulin resistance. Compared with offspring of fathers with normal weight, multivariable-adjusted mean differences for BMI z-score were 0.53 (95% CI 0.37-0.68) for obese fathers, 0.17 (95% CI 0.05-0.30) for overweight fathers, and -0.55 (95% CI -0.95-0.15) for underweight fathers; corresponding values for systolic blood pressure z-score were 0.21(95% CI 0.07-0.35), 0.10 (95% CI -0.01-0.21), and -0.24 (95% CI -0.59-0.11), and corresponding values for homeostatic model assessment for insulin resistance z-score were 0.31 (95% CI 0.16-0.46), 0.09 (95% CI -0.02-0.21), and -0.11 (95% CI -0.48-0.28), respectively. The mediation analyses suggested that 57.48% to 94.75% of the associations among paternal obesity and offspring cardiometabolic alterations might be mediated by offspring BMI. CONCLUSION Paternal obesity was associated with an unfavorable cardiometabolic profile in ART-conceived offspring. Mediation analyses indicated that offspring BMI was a possible mediator of the association between paternal obesity and the offspring impaired metabolic changes.
Collapse
Affiliation(s)
- Bingqian Zhang
- Children and Reproductive Health, Institute of Women, The Second Hospital, Shandong University, Shandong, 250012, China
- School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, Shandong, 250012, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, 250012, China
- Key laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, Shandong, 250012, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong, 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong, 250012, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250012, China
- Chinese Academy of Medical Sciences (No. 2021RU001), Research Unit of Gametogenesis and Health of ART-Offspring, Jinan, Shandong, 250012, China
| | - Miaomiao Ban
- Children and Reproductive Health, Institute of Women, The Second Hospital, Shandong University, Shandong, 250012, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, Shandong, 250012, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, 250012, China
- Key laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, Shandong, 250012, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong, 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong, 250012, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250012, China
- Chinese Academy of Medical Sciences (No. 2021RU001), Research Unit of Gametogenesis and Health of ART-Offspring, Jinan, Shandong, 250012, China
| | - Xiaojing Chen
- Children and Reproductive Health, Institute of Women, The Second Hospital, Shandong University, Shandong, 250012, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, Shandong, 250012, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, 250012, China
- Key laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, Shandong, 250012, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong, 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong, 250012, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250012, China
- Chinese Academy of Medical Sciences (No. 2021RU001), Research Unit of Gametogenesis and Health of ART-Offspring, Jinan, Shandong, 250012, China
| | - Yiyuan Zhang
- Children and Reproductive Health, Institute of Women, The Second Hospital, Shandong University, Shandong, 250012, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, Shandong, 250012, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, 250012, China
- Key laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, Shandong, 250012, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong, 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong, 250012, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250012, China
- Chinese Academy of Medical Sciences (No. 2021RU001), Research Unit of Gametogenesis and Health of ART-Offspring, Jinan, Shandong, 250012, China
| | - Zijing Wang
- Children and Reproductive Health, Institute of Women, The Second Hospital, Shandong University, Shandong, 250012, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, Shandong, 250012, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, 250012, China
- Key laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, Shandong, 250012, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong, 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong, 250012, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250012, China
- Chinese Academy of Medical Sciences (No. 2021RU001), Research Unit of Gametogenesis and Health of ART-Offspring, Jinan, Shandong, 250012, China
| | - Wanbing Feng
- Children and Reproductive Health, Institute of Women, The Second Hospital, Shandong University, Shandong, 250012, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, Shandong, 250012, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, 250012, China
- Key laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, Shandong, 250012, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong, 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong, 250012, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250012, China
- Chinese Academy of Medical Sciences (No. 2021RU001), Research Unit of Gametogenesis and Health of ART-Offspring, Jinan, Shandong, 250012, China
| | - Han Zhao
- Children and Reproductive Health, Institute of Women, The Second Hospital, Shandong University, Shandong, 250012, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, Shandong, 250012, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, 250012, China
- Key laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, Shandong, 250012, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong, 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong, 250012, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250012, China
- Chinese Academy of Medical Sciences (No. 2021RU001), Research Unit of Gametogenesis and Health of ART-Offspring, Jinan, Shandong, 250012, China
| | - Jingyu Li
- Children and Reproductive Health, Institute of Women, The Second Hospital, Shandong University, Shandong, 250012, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, Shandong, 250012, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, 250012, China
- Key laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, Shandong, 250012, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong, 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong, 250012, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250012, China
- Chinese Academy of Medical Sciences (No. 2021RU001), Research Unit of Gametogenesis and Health of ART-Offspring, Jinan, Shandong, 250012, China
| | - Tao Zhang
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Jingmei Hu
- Children and Reproductive Health, Institute of Women, The Second Hospital, Shandong University, Shandong, 250012, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, Shandong, 250012, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, 250012, China
- Key laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, Shandong, 250012, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong, 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong, 250012, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250012, China
- Chinese Academy of Medical Sciences (No. 2021RU001), Research Unit of Gametogenesis and Health of ART-Offspring, Jinan, Shandong, 250012, China
| | - Kuona Hu
- Children and Reproductive Health, Institute of Women, The Second Hospital, Shandong University, Shandong, 250012, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, Shandong, 250012, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, 250012, China
- Key laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, Shandong, 250012, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong, 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong, 250012, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250012, China
- Chinese Academy of Medical Sciences (No. 2021RU001), Research Unit of Gametogenesis and Health of ART-Offspring, Jinan, Shandong, 250012, China
| | - Linlin Cui
- Children and Reproductive Health, Institute of Women, The Second Hospital, Shandong University, Shandong, 250012, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, Shandong, 250012, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, 250012, China
- Key laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, Shandong, 250012, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong, 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong, 250012, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250012, China
- Chinese Academy of Medical Sciences (No. 2021RU001), Research Unit of Gametogenesis and Health of ART-Offspring, Jinan, Shandong, 250012, China
| | - Zi-Jiang Chen
- Children and Reproductive Health, Institute of Women, The Second Hospital, Shandong University, Shandong, 250012, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, Shandong, 250012, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, 250012, China
- Key laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, Shandong, 250012, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong, 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong, 250012, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250012, China
- Chinese Academy of Medical Sciences (No. 2021RU001), Research Unit of Gametogenesis and Health of ART-Offspring, Jinan, Shandong, 250012, China
- Department of Reproductive Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200135, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, 200135, China
| |
Collapse
|
5
|
Michael N, Sadananthan SA, Yuan WL, Ong YY, Loy SL, Huang JY, Tint MT, Padmapriya N, Choo J, Ling LH, Kramer MS, Godfrey KM, Gluckman PD, Tan KH, Eriksson JG, Chong YS, Lee YS, Karnani N, Yap F, Shek LPC, Fortier MV, Moritz KM, Chan SY, Velan SS, Wlodek ME. Associations of maternal and foetoplacental factors with prehypertension/hypertension in early childhood. J Hypertens 2022; 40:2171-2179. [PMID: 36205012 DOI: 10.1097/hjh.0000000000003241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
OBJECTIVE To evaluate whether characterization of maternal and foetoplacental factors beyond birthweight can enable early identification of children at risk of developing prehypertension/hypertension. METHODS We recruited 693 mother-offspring dyads from the GUSTO prospective mother-offspring cohort. Prehypertension/hypertension at age 6 years was identified using the simplified paediatric threshold of 110/70 mmHg. We evaluated the associations of pregnancy complications (gestational diabetes, excessive/inadequate gestational weight gain, hypertensive disorders of pregnancy), foetal growth deceleration (decline in foetal abdominal circumference at least 0.67 standard deviations between second and third trimesters), high foetoplacental vascular resistance (third trimester umbilical artery systolic-to-diastolic ratio ≥90th centile), preterm birth, small-for-gestational age and neonatal kidney volumes with risk of prehypertension/hypertension at age 6 years, after adjusting for sex, ethnicity, maternal education and prepregnancy BMI. RESULTS Pregnancy complications, small-for-gestational age, preterm birth, and low neonatal kidney volume were not associated with an increased risk of prehypertension/hypertension at age 6 years. In contrast, foetal growth deceleration was associated with a 72% higher risk [risk ratio (RR) = 1.72, 95% confidence interval (CI) 1.18-2.52]. High foetoplacental vascular resistance was associated with a 58% higher risk (RR = 1.58, 95% CI 0.96-2.62). Having both these characteristics, relative to having neither, was associated with over two-fold higher risk (RR = 2.55, 95% CI 1.26-5.16). Over 85% of the foetuses with either of these characteristics were born appropriate or large for gestational age. CONCLUSION Foetal growth deceleration and high foetoplacental vascular resistance may be helpful in prioritizing high-risk children for regular blood pressure monitoring and preventive interventions, across the birthweight spectrum.
Collapse
Affiliation(s)
- Navin Michael
- Singapore Institute for Clinical Sciences, Agency for Science, Technology, and Research, Singapore
| | - Suresh Anand Sadananthan
- Singapore Institute for Clinical Sciences, Agency for Science, Technology, and Research, Singapore
| | - Wen Lun Yuan
- Université de Paris, CRESS, Inserm, INRAE, Paris, France
| | | | - See Ling Loy
- Department of Reproductive Medicine, KK Women's and Children's Hospital, Singapore
- Duke-National University of Singapore Medical School
| | - Jonathan Y Huang
- Singapore Institute for Clinical Sciences, Agency for Science, Technology, and Research, Singapore
| | - Mya-Thway Tint
- Singapore Institute for Clinical Sciences, Agency for Science, Technology, and Research, Singapore
| | - Natarajan Padmapriya
- Department of Obstetrics & Gynaecology and Human Potential Translational Research Programme, Yong Loo Lin School of Medicine
- Saw Swee Hock School of Public Health
| | | | - Lieng Hsi Ling
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore
- Department of Cardiology, National University Heart Centre
| | - Michael S Kramer
- Department of Obstetrics & Gynaecology and Human Potential Translational Research Programme, Yong Loo Lin School of Medicine
| | - Keith M Godfrey
- Medical Research Council Lifecourse Epidemiology Unit and National Institute for Health Research Southampton Biomedical Research Centre, University of Southampton and University Hospital, Southampton National Health Service Foundation Trust, Southampton, United Kingdom
| | - Peter D Gluckman
- Singapore Institute for Clinical Sciences, Agency for Science, Technology, and Research, Singapore
- Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Kok Hian Tan
- Department of Maternal Fetal Medicine
- Duke-National University of Singapore Medical School
| | - Johan G Eriksson
- Singapore Institute for Clinical Sciences, Agency for Science, Technology, and Research, Singapore
- Department of Obstetrics & Gynaecology and Human Potential Translational Research Programme, Yong Loo Lin School of Medicine
- Department of General Practice and Primary Healthcare, University of Helsinki and Helsinki University Hospital
- Folkhälsan Research Center, Helsinki, Finland
| | - Yap-Seng Chong
- Singapore Institute for Clinical Sciences, Agency for Science, Technology, and Research, Singapore
- Department of Obstetrics & Gynaecology and Human Potential Translational Research Programme, Yong Loo Lin School of Medicine
| | - Yung Seng Lee
- Singapore Institute for Clinical Sciences, Agency for Science, Technology, and Research, Singapore
- Department of Paediatrics
- Khoo Teck Puat-National University Children's Medical Institute, National University Health System, Singapore
| | - Neerja Karnani
- Singapore Institute for Clinical Sciences, Agency for Science, Technology, and Research, Singapore
| | - Fabian Yap
- Department of Pediatric Endocrinology
- Duke-National University of Singapore Medical School
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Lynette Pei-Chi Shek
- Singapore Institute for Clinical Sciences, Agency for Science, Technology, and Research, Singapore
| | - Marielle V Fortier
- Singapore Institute for Clinical Sciences, Agency for Science, Technology, and Research, Singapore
- Department of Diagnostic and Interventional Imaging
| | | | - Shiao-Yng Chan
- Singapore Institute for Clinical Sciences, Agency for Science, Technology, and Research, Singapore
- Department of Obstetrics & Gynaecology and Human Potential Translational Research Programme, Yong Loo Lin School of Medicine
| | - S Sendhil Velan
- Singapore Institute for Clinical Sciences, Agency for Science, Technology, and Research, Singapore
| | - Mary E Wlodek
- Singapore Institute for Clinical Sciences, Agency for Science, Technology, and Research, Singapore
- Department of Obstetrics & Gynaecology and Human Potential Translational Research Programme, Yong Loo Lin School of Medicine
- University of Melbourne, Parkville, Australia
| |
Collapse
|
6
|
Perl AJ, Schuh MP, Kopan R. Regulation of nephron progenitor cell lifespan and nephron endowment. Nat Rev Nephrol 2022; 18:683-695. [PMID: 36104510 PMCID: PMC11078284 DOI: 10.1038/s41581-022-00620-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/27/2022] [Indexed: 11/08/2022]
Abstract
Low nephron number - resulting, for example, from prematurity or developmental anomalies - is a risk factor for the development of hypertension, chronic kidney disease and kidney failure. Considerable interest therefore exists in the mechanisms that regulate nephron endowment and contribute to the premature cessation of nephrogenesis following preterm birth. The cessation of nephrogenesis in utero or shortly after birth is synchronized across multiple niches in all mammals, and is coupled with the exhaustion of nephron progenitor cells. Consequently, no nephrons are formed after the cessation of developmental nephrogenesis, and lifelong renal function therefore depends on the complement of nephrons generated during gestation. In humans, a tenfold variation in nephron endowment between individuals contributes to differences in susceptibility to kidney disease; however, the mechanisms underlying this variation are not yet clear. Salient advances in our understanding of environmental inputs, and of intrinsic molecular mechanisms that contribute to the regulation of cessation timing or nephron progenitor cell exhaustion, have the potential to inform interventions to enhance nephron endowment and improve lifelong kidney health for susceptible individuals.
Collapse
Affiliation(s)
- Alison J Perl
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Meredith P Schuh
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Raphael Kopan
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| |
Collapse
|
7
|
Bajinka O, Barrow A, Mendy S, Jallow BJJ, Jallow J, Barrow S, Bah O, Camara S, Colley ML, Nyabally S, Joof AN, Qi M, Tan Y. The Influence of Parental Environmental Exposure and Nutrient Restriction on the Early Life of Offspring Growth in Gambia-A Pilot Study. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:13045. [PMID: 36293620 PMCID: PMC9603272 DOI: 10.3390/ijerph192013045] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 09/21/2022] [Accepted: 09/23/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND The role of the germline in epigenetic transgenerational inheritance starts with environmental factors, acting on the first generation of a gestating mother. These factors influence the developing second-generation fetus by altering gonadal development, thereby reprogramming the primordial germ cell DNA methylation and leading to consequences that might be seen along generations. OBJECTIVE Despite these epigenetic factors now surfacing, the few available studies are on animal-based experiments, and conducting a follow-up on human intergenerational trials might take decades. To this response, this study aimed to determine the influence of parental energy, toxicant exposure, age, and nutrient restriction on the early life of offspring growth in Gambia. METHOD This pilot study was based on population observation and combined both maternal and paternal factors across the country between August and October 2021. It captures the lifestyle and health detailed account of 339 reproductive parents and their last born (child under 5 years) using a structured interview questionnaire performed by nurses and public health officers. RESULTS This study showed that parents who worked in industrial areas were more likely to have offspring with poor psychosocial skills. In addition, mothers who are exposed to oxidative stress and high temperatures are more likely to have offspring with poor psychosocial skills. Mothers who consume a high-protein diet were almost three times more likely to have infants with good psychosocial skills in their offspring. Furthermore, there was a negative correlation between maternal stress during pregnancy and the psychosocial skills of offspring. CONCLUSION This study was able to ascertain if the maternal diet during gestation, toxicant exposure, maternal stress, and parental smoking habits have an influence on the early life of offspring. While the study recommends a large sample size study to eliminate selection bias, there should be an increased level of awareness of mothers of their offspring's health and their husbands' lifestyles that might influence the adulthood health of their offspring.
Collapse
Affiliation(s)
- Ousman Bajinka
- Department of Medical Microbiology, Central South University, Changsha 410078, China
- China-Africa Research Center of Infectious Diseases, School of Basic and Medical Sciences, Central South University, Changsha 410078, China
- School of Medicine and Allied Health Sciences, University of The Gambia, Kanifing 3530, The Gambia
| | - Amadou Barrow
- School of Medicine and Allied Health Sciences, University of The Gambia, Kanifing 3530, The Gambia
| | - Sang Mendy
- Ministry of Health, Banjul P.O. Box 273, The Gambia
| | - Binta J. J. Jallow
- Department of Medical Microbiology, Central South University, Changsha 410078, China
| | - Jarry Jallow
- School of Medicine and Allied Health Sciences, University of The Gambia, Kanifing 3530, The Gambia
| | - Sulayman Barrow
- School of Medicine and Allied Health Sciences, University of The Gambia, Kanifing 3530, The Gambia
| | - Ousman Bah
- Ministry of Health, Banjul P.O. Box 273, The Gambia
| | - Saikou Camara
- School of Medicine and Allied Health Sciences, University of The Gambia, Kanifing 3530, The Gambia
| | - Modou Lamin Colley
- School of Medicine and Allied Health Sciences, University of The Gambia, Kanifing 3530, The Gambia
| | - Sankung Nyabally
- School of Medicine and Allied Health Sciences, University of The Gambia, Kanifing 3530, The Gambia
| | - Amie N. Joof
- Department of Medical Microbiology, Central South University, Changsha 410078, China
| | - Mingming Qi
- Department of Obstetrics, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Changsha 410017, China
| | - Yurong Tan
- Department of Medical Microbiology, Central South University, Changsha 410078, China
- China-Africa Research Center of Infectious Diseases, School of Basic and Medical Sciences, Central South University, Changsha 410078, China
| |
Collapse
|
8
|
Liu C, Ma K, Zhang Y, He X, Song L, Chi M, Han Z, Li G, Zhang Q, Liu C. Kidney diseases and long non-coding RNAs in the limelight. Front Physiol 2022; 13:932693. [PMID: 36299256 PMCID: PMC9589442 DOI: 10.3389/fphys.2022.932693] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 09/21/2022] [Indexed: 11/13/2022] Open
Abstract
The most extensively and well-investigated sequences in the human genome are protein-coding genes, while large numbers of non-coding sequences exist in the human body and are even more diverse with more potential roles than coding sequences. With the unveiling of non-coding RNA research, long-stranded non-coding RNAs (lncRNAs), a class of transcripts >200 nucleotides in length primarily expressed in the nucleus and rarely in the cytoplasm, have drawn our attention. LncRNAs are involved in various levels of gene regulatory processes, including but not limited to promoter activity, epigenetics, translation and transcription efficiency, and intracellular transport. They are also dysregulated in various pathophysiological processes, especially in diseases and cancers involving genomic imprinting. In recent years, numerous studies have linked lncRNAs to the pathophysiology of various kidney diseases. This review summarizes the molecular mechanisms involved in lncRNAs, their impact on kidney diseases, and associated complications, as well as the value of lncRNAs as emerging biomarkers for the prevention and prognosis of kidney diseases, suggesting their potential as new therapeutic tools.
Collapse
Affiliation(s)
- Chenxin Liu
- Reproductive and Women-Children Hospital, School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Kuai Ma
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yunchao Zhang
- Reproductive and Women-Children Hospital, School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xing He
- School of Clinical Medicine, Chengdu Medical College, Chengdu, China
| | - Linjiang Song
- Reproductive and Women-Children Hospital, School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Mingxuan Chi
- Department of Nephrology, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, Sichuan Renal Disease Clinical Research Center, University of Electronic Science and Technology of China, Chengdu, China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| | - Zhongyu Han
- Reproductive and Women-Children Hospital, School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Guanhua Li
- Department of Cardiovascular Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- *Correspondence: Guanhua Li, ; Qinxiu Zhang, ; Chi Liu,
| | - Qinxiu Zhang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- *Correspondence: Guanhua Li, ; Qinxiu Zhang, ; Chi Liu,
| | - Chi Liu
- Department of Nephrology, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, Sichuan Renal Disease Clinical Research Center, University of Electronic Science and Technology of China, Chengdu, China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
- *Correspondence: Guanhua Li, ; Qinxiu Zhang, ; Chi Liu,
| |
Collapse
|
9
|
Associations of maternal periconceptional alcohol consumption with offspring prehypertension/hypertension at age 6 years: the Growing Up in Singapore Towards healthy Outcomes prospective mother-offspring cohort study. J Hypertens 2022; 40:1212-1222. [PMID: 35703883 DOI: 10.1097/hjh.0000000000003134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE To evaluate the relationship of the levels of maternal alcohol consumption during the 1 year before pregnancy recognition with childhood cardiorenal, metabolic, and neurocognitive health. METHODS In 1106 women and their children from the Growing Up in Singapore Towards healthy Outcomes mother-offspring cohort, quantity of maternal alcohol consumption in the 12 months prior to pregnancy recognition was categorized as high (≥75th percentile: 1.9 g/day), low (<1.9 g/day), and none, and frequency of alcohol consumption was categorized as high (≥2-3 times/week), low (<2-3 times/week), and none. Offspring MRI-based abdominal fat depot, kidney, and brain volumes, blood pressure, metabolic syndrome score, and cognitive intelligence scores were assessed. Child prehypertension/hypertension at age 6 years was defined using a simplified pediatric threshold of 110/70 mmHg. RESULTS The average maternal alcohol consumption in the year prior to pregnancy recognition was 2.5 g/day, which is lower than the daily maximal limit of one standard drink (10 g) recommended for women by Singapore's Ministry of Health. After adjusting for participant characteristics, alcohol consumption at least 1.9 g/day was associated with over two-fold higher risk (risk ratio = 2.18, P = 0.013) of child prehypertension and 15% greater kidney growth between early infancy and age 6 years (P = 0.040) compared with abstinence. Alcohol consumption was not associated with metabolic and neurocognitive health at age 6-7 years. The associations with high frequency of alcohol consumption were concordant with those obtained for quantity of alcohol consumption. CONCLUSION Maternal self-reported alcohol consumption at least 1.9 g/day prior to pregnancy recognition was associated with increased risk of child prehypertension and rapid kidney growth. Our findings highlight the potential detrimental effects of low periconceptional alcohol consumption, below national guidelines on offspring cardiorenal health.
Collapse
|
10
|
Zhao X, Li B, Xiong Y, Xia Z, Hu S, Sun Z, Wang H, Ao Y. Prenatal caffeine exposure induced renal developmental toxicity and transgenerational effect in rat offspring. Food Chem Toxicol 2022; 165:113082. [PMID: 35537649 DOI: 10.1016/j.fct.2022.113082] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 03/24/2022] [Accepted: 04/23/2022] [Indexed: 10/18/2022]
Abstract
Epidemiological studies revealed that prenatal caffeine exposure (PCE) is associated with adverse gestational outcomes and susceptibility to chronic diseases in offspring, yet the effects of PCE on glomerulosclerosis susceptibility in adult female offspring and its intergenerational transmission remain to be further investigated. Here, we found that PCE caused fetal kidney dysplasia and glomerulosclerosis of the female offspring. Besides, the kidney of F1 offspring in PCE group exhibited the "low expressional programming of AT2R" and "GC-IGF1 programming" alteration. Intergenerational genetic studies revealed that the renal defect and GC-IGF1 programming alteration was inherited to F2 adult female offspring derived from the female germ line, but Low expression of AT2R did not extend to the F2 female offspring. Taken together, PCE caused renal dysplasia and adult glomerulosclerosis in the F1 female offspring, which might be mediated by renal AT2R low expressional programming and GC-IGF1 axis alteration. Furthermore, PCE induced transgenerational toxicity on kidney, and GC-IGF1 programming alteration might be the potential molecular mechanism. This study provided experimental evidence for the mechanism study of the intergenerational inheritance of kidney developmental toxicity caused by PCE.
Collapse
Affiliation(s)
- Xiaoqi Zhao
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan, 430071, China
| | - Bin Li
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan, 430071, China; Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Ying Xiong
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan, 430071, China
| | - Zhiping Xia
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan, 430071, China
| | - Shuangshuang Hu
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan, 430071, China
| | - Zhaoxia Sun
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan, 430071, China
| | - Hui Wang
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan, 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disorder, Wuhan, 430071, China
| | - Ying Ao
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan, 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disorder, Wuhan, 430071, China.
| |
Collapse
|
11
|
Iyengar A, Bonilla-Félix M. Effects of Prematurity and Growth Restriction on Adult Blood Pressure and Kidney Volume. Adv Chronic Kidney Dis 2022; 29:243-250. [PMID: 36084971 DOI: 10.1053/j.ackd.2022.02.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 01/11/2022] [Accepted: 02/15/2022] [Indexed: 11/11/2022]
Abstract
Gaining insight into the complex cycle of renal programming and its early-life clinical associations is essential to understand the origins of kidney disease. Prematurity and intrauterine growth restriction are associated with low nephron endowment. This increases the risk of developing hypertension and chronic kidney disease later in life. There is appreciable evidence to support mechanistic links between low nephron endowment secondary to intrauterine events and kidney size, kidney function, and blood pressure in postnatal life. A clear understanding of the cycle of developmental programming and consequences of fetal insults on the kidney is critical. In addition, the impact of events in the early postnatal period (accelerated postnatal growth, development of obesity, exposure to nephrotoxins) on the cardiovascular system and blood pressure of individuals born prematurely or with low birth weight is discussed. In summary, this review draws attention to the concepts of renal programming and nephron endowment and underscores the associations between intrauterine growth restriction, prematurity, and its clinical consequences in adult life.
Collapse
Affiliation(s)
- Arpana Iyengar
- Department of Pediatric Nephrology, St John's Medical College Hospital, Bangalore, India
| | | |
Collapse
|
12
|
Costa TJ, De Oliveira JC, Giachini FR, Lima VV, Tostes RC, Bomfim GF. Programming of Vascular Dysfunction by Maternal Stress: Immune System Implications. Front Physiol 2022; 13:787617. [PMID: 35360231 PMCID: PMC8961444 DOI: 10.3389/fphys.2022.787617] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 01/13/2022] [Indexed: 11/13/2022] Open
Abstract
A growing body of evidence highlights that several insults during pregnancy impact the vascular function and immune response of the male and female offspring. Overactivation of the immune system negatively influences cardiovascular function and contributes to cardiovascular disease. In this review, we propose that modulation of the immune system is a potential link between prenatal stress and offspring vascular dysfunction. Glucocorticoids are key mediators of stress and modulate the inflammatory response. The potential mechanisms whereby prenatal stress negatively impacts vascular function in the offspring, including poor hypothalamic–pituitary–adrenal axis regulation of inflammatory response, activation of Th17 cells, renin–angiotensin–aldosterone system hyperactivation, reactive oxygen species imbalance, generation of neoantigens and TLR4 activation, are discussed. Alterations in the immune system by maternal stress during pregnancy have broad relevance for vascular dysfunction and immune-mediated diseases, such as cardiovascular disease.
Collapse
Affiliation(s)
- Tiago J. Costa
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Júlio Cezar De Oliveira
- Health Education Research Center (NUPADS), Institute of Health Sciences, Federal University of Mato Grosso, Sinop, Brazil
| | - Fernanda Regina Giachini
- Institute of Biological Sciences and Health, Federal University of Mato Grosso, Barra do Garças, Brazil
| | - Victor Vitorino Lima
- Institute of Biological Sciences and Health, Federal University of Mato Grosso, Barra do Garças, Brazil
| | - Rita C. Tostes
- Health Education Research Center (NUPADS), Institute of Health Sciences, Federal University of Mato Grosso, Sinop, Brazil
| | - Gisele Facholi Bomfim
- Health Education Research Center (NUPADS), Institute of Health Sciences, Federal University of Mato Grosso, Sinop, Brazil
- *Correspondence: Gisele Facholi Bomfim,
| |
Collapse
|
13
|
Ojeda ML, Carreras O, Nogales F. The Role of Selenoprotein Tissue Homeostasis in MetS Programming: Energy Balance and Cardiometabolic Implications. Antioxidants (Basel) 2022; 11:antiox11020394. [PMID: 35204276 PMCID: PMC8869711 DOI: 10.3390/antiox11020394] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 02/11/2022] [Accepted: 02/12/2022] [Indexed: 11/16/2022] Open
Abstract
Selenium (Se) is an essential trace element mainly known for its antioxidant, anti-inflammatory, and anti-apoptotic properties, as it is part of the catalytic center of 25 different selenoproteins. Some of them are related to insulin resistance (IR) and metabolic syndrome (MetS) generation, modulating reactive oxygen species (ROS), and the energetic sensor AMP-activated protein kinase (AMPK); they can also regulate the nuclear transcription factor kappa-B (NF-kB), leading to changes in inflammation production. Selenoproteins are also necessary for the correct synthesis of insulin and thyroid hormones. They are also involved in endocrine central regulation of appetite and energy homeostasis, affecting growth and development. MetS, a complex metabolic disorder, can appear during gestation and lactation in mothers, leading to energetic and metabolic changes in their offspring that, according to the metabolic programming theory, will produce cardiovascular and metabolic diseases later in life. However, there is a gap concerning Se tissue levels and selenoproteins’ implications in MetS generation, which is even greater during MetS programming. This narrative review also provides an overview of the existing evidence, based on experimental research from our laboratory, which strengthens the fact that maternal MetS leads to changes in Se tissue deposits and antioxidant selenoproteins’ expression in their offspring. These changes contribute to alterations in tissues’ oxidative damage, inflammation, energy balance, and tissue function, mainly in the heart. Se imbalance also could modulate appetite and endocrine energy balance, affecting pups’ growth and development. MetS pups present a profile similar to that of diabetes type 1, which also appeared when dams were exposed to low-Se dietary supply. Maternal Se supplementation should be taken into account if, during gestation and/or lactation periods, there are suspicions of endocrine energy imbalance in the offspring, such as MetS. It could be an interesting therapy to induce heart reprogramming. However, more studies are necessary.
Collapse
|
14
|
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
|
15
|
Hu C, Tao Y, Deng Y, Cai Q, Ren H, Yu C, Zheng S, Yang J, Zeng C. Paternal long-term PM2.5 exposure causes hypertension via increased renal AT1R expression and function in male offspring. Clin Sci (Lond) 2021; 135:2575-2588. [PMID: 34779863 PMCID: PMC8628185 DOI: 10.1042/cs20210802] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 11/10/2021] [Accepted: 11/15/2021] [Indexed: 01/15/2023]
Abstract
Maternal exposure to fine particulate matter (PM2.5) causes hypertension in offspring. However, paternal contribution of PM2.5 exposure to hypertension in offspring remains unknown. In the present study, male Sprague-Dawley rats were treated with PM2.5 suspension (10 mg/ml) for 12 weeks and/or fed with tap water containing an antioxidant tempol (1 mM/L) for 16 weeks. The blood pressure, 24 h-urine volume and sodium excretion were determined in male offspring. The offspring were also administrated with losartan (20 mg/kg/d) for 4 weeks. The expressions of angiotensin II type 1 receptor (AT1R) and G-protein-coupled receptor kinase type 4 (GRK4) were determined by qRT-PCR and immunoblotting. We found that long-term PM2.5 exposure to paternal rats caused hypertension and impaired urine volume and sodium excretion in male offspring. Both the mRNA and protein expression of GRK4 and its downstream target AT1R were increased in offspring of PM2.5-exposed paternal rats, which was reflected in its function because treatment with losartan, an AT1R antagonist, decreased the blood pressure and increased urine volume and sodium excretion. In addition, the oxidative stress level was increased in PM2.5-treated paternal rats. Administration with tempol in paternal rats restored the increased blood pressure and decreased urine volume and sodium excretion in the offspring of PM2.5-exposed paternal rats. Treatment with tempol in paternal rats also reversed the increased expressions of AT1R and GRK4 in the kidney of their offspring. We suggest that paternal PM2.5 exposure causes hypertension in offspring. The mechanism may be involved that paternal PM2.5 exposure-associated oxidative stress induces the elevated renal GRK4 level, leading to the enhanced AT1R expression and its-mediated sodium retention, consequently causes hypertension in male offspring.
Collapse
Affiliation(s)
- Cuimei Hu
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, P. R. China
| | - Yu Tao
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, P. R. China
| | - Yi Deng
- Department of General Practice Medicine, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Qi Cai
- Department of Cardiology, Fujian Heart Center, Provincial Institute of Coronary Disease, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Hongmei Ren
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, P. R. China
| | - Cheng Yu
- Department of Cardiology, Fujian Heart Center, Provincial Institute of Coronary Disease, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Shuo Zheng
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, P. R. China
| | - Jian Yang
- Department of Clinical Nutrition, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chunyu Zeng
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, P. R. China
- State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, The Third Military Medical University, Chongqing, China
- Cardiovascular Research Center of Chongqing College, Department of Cardiology of Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, China
| |
Collapse
|
16
|
A Scoping Review of Life-Course Psychosocial Stress and Kidney Function. CHILDREN-BASEL 2021; 8:children8090810. [PMID: 34572242 PMCID: PMC8467128 DOI: 10.3390/children8090810] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 09/06/2021] [Accepted: 09/08/2021] [Indexed: 11/16/2022]
Abstract
Increased exposure to maternal psychosocial stress during gestation and adverse neonatal environments has been linked to alterations in developmental programming and health consequences in offspring. A programmed low nephron endowment, among other altered pathways of susceptibility, likely increases the vulnerability to develop chronic kidney disease in later life. Our aim in this scoping review was to identify gaps in the literature by focusing on understanding the association between life-course exposure to psychosocial stress, and the risk of reduced kidney function. A systematic search in four databases (PubMed, ProQuest, Wed of Science, and Scopus) was performed, yielding 609 articles. Following abstract and full-text review, we identified 19 articles meeting our inclusion criteria, reporting associations between different psychosocial stressors and an increase in the prevalence of kidney disease or decline in kidney function, mainly in adulthood. There are a lack of studies that specifically evaluated the association between gestational exposure to psychosocial stress and measures of kidney function or disease in early life, despite the overall evidence consistent with the independent effects of prenatal stress on other perinatal and postnatal outcomes. Further research will establish epidemiological studies with clear and more comparable psychosocial stressors to solve this critical research gap.
Collapse
|
17
|
Jorge BC, Reis ACC, Stein J, Balin PDS, Sterde ÉT, Barbosa MG, de Aquino AM, Kassuya CAL, Arena AC. Parental exposure to benzo(a)pyrene in the peripubertal period impacts reproductive aspects of the F1 generation in rats. Reprod Toxicol 2021; 100:126-136. [PMID: 33513405 DOI: 10.1016/j.reprotox.2021.01.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 12/21/2020] [Accepted: 01/22/2021] [Indexed: 10/22/2022]
Abstract
Benzo(a)pyrene (BaP) is an ubiquitous environmental pollutant which can lead to adverse effects on male reproduction. However, the persistence of these changes on a multigenerational scale has not been sufficiently explored. This study evaluated if peripubertal exposure to BaP in male rats can induce reproductive impairment in offspring. Male rats received BaP at environmentally relevant doses (0, 0.1, 1, or 10 μg/kg/day) orally from post-natal (PND) 23-53. On PND 90, treated males were mated with non-treated females for obtaining the next generation (F1). The paternal exposure to BaP decreased the body weight of offspring on PND 1, 13 and 22, as well as it provoked a reduction in the relative anogenital distance of the males. This exposure also brought forward the onset of puberty, evidenced by an earlier vaginal opening and first estrous in females of the lowest dose group and by a delay in the testicular descent and preputial separation ages in males. The males presented a decrease in the daily sperm production and a disrupted sperm morphology. Furthermore, the testicular histology was altered, evidenced by a reduction in the Leydig cell numbers and in the seminiferous tubules diameter, as well as a disrupted seminiferous tubules staging. The estrous cyclicity and some fertility parameters were changed in the females, as well as alterations in the ovary and uterus histology were observed. BaP compromised several reproductive parameters of the F1 generation, suggesting that peripubertal exposure to this compound provokes permanent modifications in male germ line of F0 generation.
Collapse
Affiliation(s)
- Bárbara Campos Jorge
- Department of Structural and Functional Biology, Institute of Biosciences of Botucatu, Univ. Estadual Paulista - Botucatu (UNESP), São Paulo, Brazil
| | - Ana Carolina Casali Reis
- Department of Structural and Functional Biology, Institute of Biosciences of Botucatu, Univ. Estadual Paulista - Botucatu (UNESP), São Paulo, Brazil
| | - Julia Stein
- Department of Structural and Functional Biology, Institute of Biosciences of Botucatu, Univ. Estadual Paulista - Botucatu (UNESP), São Paulo, Brazil
| | - Paola da Silva Balin
- Department of Structural and Functional Biology, Institute of Biosciences of Botucatu, Univ. Estadual Paulista - Botucatu (UNESP), São Paulo, Brazil
| | - Érika Tissiana Sterde
- Department of Structural and Functional Biology, Institute of Biosciences of Botucatu, Univ. Estadual Paulista - Botucatu (UNESP), São Paulo, Brazil
| | - Mariana Gazoli Barbosa
- Department of Structural and Functional Biology, Institute of Biosciences of Botucatu, Univ. Estadual Paulista - Botucatu (UNESP), São Paulo, Brazil
| | - Ariana Musa de Aquino
- Department of Structural and Functional Biology, Institute of Biosciences of Botucatu, Univ. Estadual Paulista - Botucatu (UNESP), São Paulo, Brazil
| | | | - Arielle Cristina Arena
- Department of Structural and Functional Biology, Institute of Biosciences of Botucatu, Univ. Estadual Paulista - Botucatu (UNESP), São Paulo, Brazil; Center of Toxicological Assistance (CEATOX), Institute of Biosciences of Botucatu, Univ. Estadual Paulista - Botucatu (UNESP), São Paulo State, Brazil.
| |
Collapse
|
18
|
Lurbe E, Ingelfinger J. Developmental and Early Life Origins of Cardiometabolic Risk Factors: Novel Findings and Implications. Hypertension 2021; 77:308-318. [PMID: 33390043 DOI: 10.1161/hypertensionaha.120.14592] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The intent of this review is to critically consider the data that support the concept of programming and its implications. Birth weight and growth trajectories during childhood are associated with cardiometabolic disease in adult life. Both extremes, low and high birth weight coupled with postnatal growth increase the early presence of cardiometabolic risk factors and vascular imprinting, crucial elements of this framework. Data coming from epigenetics, proteomics, metabolomics, and microbiota added relevant information and contribute to better understanding of mechanisms as well as development of biomarkers helping to move forward to take actions. Research has reached a stage in which sufficiently robust data calls for new initiatives focused on early life. Prevention starting early in life is likely to have a very large impact on reducing disease incidence and its associated effects at the personal, economic, and social levels.
Collapse
Affiliation(s)
- Empar Lurbe
- From the Pediatric Department, Consorcio Hospital General, University of Valencia (E.L.)
- CIBER Fisiopatología Obesidad y Nutrición (CB06/03), Instituto de Salud Carlos III, Spain (E.L.)
| | - Julie Ingelfinger
- Department of Pediatrics, Harvard Medical School, Mass General Hospital for Children, Massachusetts General Hospital, Boston (J.I.)
| |
Collapse
|
19
|
Argeri R, Thomazini F, Lichtenecker DCK, Thieme K, do Carmo Franco M, Gomes GN. Programmed Adult Kidney Disease: Importance of Fetal Environment. Front Physiol 2020; 11:586290. [PMID: 33101064 PMCID: PMC7546361 DOI: 10.3389/fphys.2020.586290] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 09/07/2020] [Indexed: 12/29/2022] Open
Abstract
The Barker hypothesis strongly supported the influence of fetal environment on the development of chronic diseases in later life. Multiple experimental and human studies have identified that the deleterious effect of fetal programming commonly leads to alterations in renal development. The interplay between environmental insults and fetal genome can induce epigenetic changes and lead to alterations in the expression of renal phenotype. In this review, we have explored the renal development and its functions, while focusing on the epigenetic findings and functional aspects of the renin-angiotensin system and its components.
Collapse
Affiliation(s)
- Rogério Argeri
- Department of Physiology, School of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - Fernanda Thomazini
- Department of Physiology, School of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | | | - Karina Thieme
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, Universidade de Sao Paulo, São Paulo, Brazil
| | - Maria do Carmo Franco
- Department of Physiology, School of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - Guiomar Nascimento Gomes
- Department of Physiology, School of Medicine, Federal University of São Paulo, São Paulo, Brazil
| |
Collapse
|
20
|
Barbosa MG, Jorge BC, Stein J, Santos Ferreira DA, Barreto ACDS, Reis ACC, Moreira SDS, Inocencio LCDL, Macorini LFB, Arena AC. Pre-pubertal exposure to ibuprofen impairs sperm parameters in male adult rats and compromises the next generation. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2020; 83:559-572. [PMID: 32615883 DOI: 10.1080/15287394.2020.1786483] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Ibuprofen is one of the most commonly prescribed anti-inflammatory drugs in pediatric practice. This drug inhibits the cyclooxygenase enzyme, reducing the production of prostaglandin, an important mediator on male reproductive function. We examined if pre-pubertal treatment with ibuprofen in male rats can affect the reproductive parameters of these animals in adult life and on their descendants. Male rats (23 days old) received ibuprofen (0; 2.4; 7.2 or 14.3 mg/kg/day), per gavage, from postnatal day (PND) 23 to 53. At sexual maturity, treated males were placed with untreated females for obtaining the next generation (F1). The highest dose of ibuprofen interfered in sexual behavior and reduced the fertility potential of these animals in adulthood. Additionally, the ibuprofen treatment altered the sperm quantity and quality, as evidenced by a decrease in sperm motility and in the daily sperm production in the testis. Testosterone levels were also reduced by pre-pubertal treatment. The paternal treatment with this drug also influenced the reproductive outcomes of progeny. The male offspring from males treated exhibited acceleration in sperm transit time in the epididymis and the number and volume of Leydig cell nuclei were decreased, while the estrous cyclicity was displayed and the fertility potential reduced in the female offspring. The pre-pubertal ibuprofen-treatment caused negative reproductive impacts in adulthood, compromising sperm quality and quantity, as well as interfered in the reproductive outcomes of the next generation.
Collapse
Affiliation(s)
- Mariana Gazoli Barbosa
- Department of Structural and Functional Biology, Institute of Biosciences of Botucatu, Univ. Estadual Paulista - Botucatu (UNESP) , São Paulo, Brazil
| | - Bárbara Campos Jorge
- Department of Structural and Functional Biology, Institute of Biosciences of Botucatu, Univ. Estadual Paulista - Botucatu (UNESP) , São Paulo, Brazil
| | - Julia Stein
- Department of Structural and Functional Biology, Institute of Biosciences of Botucatu, Univ. Estadual Paulista - Botucatu (UNESP) , São Paulo, Brazil
| | - Dayana Agnes Santos Ferreira
- Department of Structural and Functional Biology, Institute of Biosciences of Botucatu, Univ. Estadual Paulista - Botucatu (UNESP) , São Paulo, Brazil
| | - Ana Carolina da Silva Barreto
- Department of Structural and Functional Biology, Institute of Biosciences of Botucatu, Univ. Estadual Paulista - Botucatu (UNESP) , São Paulo, Brazil
| | - Ana Carolina Casali Reis
- Department of Structural and Functional Biology, Institute of Biosciences of Botucatu, Univ. Estadual Paulista - Botucatu (UNESP) , São Paulo, Brazil
| | - Suyane Da Silva Moreira
- Department of Structural and Functional Biology, Institute of Biosciences of Botucatu, Univ. Estadual Paulista - Botucatu (UNESP) , São Paulo, Brazil
| | - Leonardo Cesar De Lima Inocencio
- Department of Structural and Functional Biology, Institute of Biosciences of Botucatu, Univ. Estadual Paulista - Botucatu (UNESP) , São Paulo, Brazil
| | | | - Arielle Cristina Arena
- Department of Structural and Functional Biology, Institute of Biosciences of Botucatu, Univ. Estadual Paulista - Botucatu (UNESP) , São Paulo, Brazil
- Department of Structural and Functional Biology, Institute of Biosciences of Botucatu, São Paulo State University (UNESP) , Botucatu, Brazil
| |
Collapse
|
21
|
Torreggiani M, Fois A, D’Alessandro C, Colucci M, Orozco Guillén AO, Cupisti A, Piccoli GB. Of Mice and Men: The Effect of Maternal Protein Restriction on Offspring's Kidney Health. Are Studies on Rodents Applicable to Chronic Kidney Disease Patients? A Narrative Review. Nutrients 2020; 12:E1614. [PMID: 32486266 PMCID: PMC7352514 DOI: 10.3390/nu12061614] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 05/19/2020] [Accepted: 05/28/2020] [Indexed: 12/20/2022] Open
Abstract
In the almost 30 years that have passed since the postulation of the "Developmental Origins of Health and Disease" theory, it has been clearly demonstrated that a mother's dietary habits during pregnancy have potential consequences for her offspring that go far beyond in utero development. Protein malnutrition during pregnancy, for instance, can cause severe alterations ranging from intrauterine growth retardation to organ damage and increased susceptibility to hypertension, diabetes mellitus, cardiovascular diseases and chronic kidney disease (CKD) later in life both in experimental animals and humans. Conversely, a balanced mild protein restriction in patients affected by CKD has been shown to mitigate the biochemical derangements associated with kidney disease and even slow its progression. The first reports on the management of pregnant CKD women with a moderately protein-restricted plant-based diet appeared in the literature a few years ago. Today, this approach is still being debated, as is the optimal source of protein during gestation in CKD. The aim of this report is to critically review the available literature on the topic, focusing on the similarities and differences between animal and clinical studies.
Collapse
Affiliation(s)
- Massimo Torreggiani
- Nephrology and Dialysis, Centre Hospitalier Le Mans, Avenue Roubillard 194, 72000 Le Mans, France; (A.F.); (G.B.P.)
| | - Antioco Fois
- Nephrology and Dialysis, Centre Hospitalier Le Mans, Avenue Roubillard 194, 72000 Le Mans, France; (A.F.); (G.B.P.)
| | - Claudia D’Alessandro
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (C.D.); (A.C.)
| | - Marco Colucci
- Unit of Nephrology and Dialysis, ICS Maugeri S.p.A. SB, Via S. Maugeri 10, 27100 Pavia, Italy;
| | | | - Adamasco Cupisti
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (C.D.); (A.C.)
| | - Giorgina Barbara Piccoli
- Nephrology and Dialysis, Centre Hospitalier Le Mans, Avenue Roubillard 194, 72000 Le Mans, France; (A.F.); (G.B.P.)
- Dipartimento di Scienze Cliniche e Biologiche, Università di Torino, 10100 Torino, Italy
| |
Collapse
|
22
|
Akison LK, Probyn ME, Gray SP, Cullen-McEwen LA, Tep K, Steane SE, Gobe GC, Wlodek ME, Bertram JF, Moritz KM. Moderate prenatal ethanol exposure in the rat promotes kidney cell apoptosis, nephron deficits, and sex-specific kidney dysfunction in adult offspring. Anat Rec (Hoboken) 2020; 303:2632-2645. [PMID: 31984647 DOI: 10.1002/ar.24370] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 10/31/2019] [Accepted: 12/07/2019] [Indexed: 12/12/2022]
Abstract
Alcohol during pregnancy can impair fetal development and result in offspring with neurodevelopmental deficits. Less is known about how low to moderate alcohol exposure can affect other organs, such as the kidney. Here, the effects of moderate ethanol exposure throughout pregnancy on kidney development were examined using a rat model. Rats were fed a liquid diet containing 6% ethanol (vol/vol) or control (0% ethanol) throughout pregnancy. Kidneys were collected at embryonic day (E) 20 or postnatal day (PN) 30 and total glomerular (nephron) number determined using unbiased stereology. Kidney function was examined in offspring at 8 and 19 months. At E20, fetuses exposed to ethanol had fewer nephrons with increased apoptosis. Alcohol exposure caused kidney dysregulation of pro- (Bax) and anti- (Bcl-2) apoptotic factors, and reduced expression of the cell proliferation marker, Ki67. Prenatal alcohol decreased expression of Gdnf and Tgfb1, important regulators of branching morphogenesis, in male fetuses. At PN30, kidney volume and nephron number were lower in offspring exposed to prenatal alcohol. Urine flow and osmolality were normal in offspring exposed to alcohol however sodium excretion tended to be lower in females prenatally exposed to alcohol. Findings suggest exposure to moderate levels of alcohol during pregnancy results in impaired kidney development and leads to a permanent nephron deficit. Although the impact on adult kidney function was relatively minor, these data highlight that even at moderate levels, alcohol consumption during pregnancy can have deleterious long-term outcomes and should be avoided.
Collapse
Affiliation(s)
- Lisa K Akison
- School of Biomedical Sciences, The University of Queensland, Brisbane, Australia.,Child Health Research Centre, The University of Queensland, Brisbane, Australia
| | - Megan E Probyn
- School of Biomedical Sciences, The University of Queensland, Brisbane, Australia
| | - Stephen P Gray
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Clayton, Australia.,Department of Anatomy and Developmental Biology, Monash University, Clayton, Australia
| | - Louise A Cullen-McEwen
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Clayton, Australia.,Department of Anatomy and Developmental Biology, Monash University, Clayton, Australia
| | - Karrona Tep
- School of Biomedical Sciences, The University of Queensland, Brisbane, Australia
| | - Sarah E Steane
- School of Biomedical Sciences, The University of Queensland, Brisbane, Australia
| | - Glenda C Gobe
- School of Biomedical Sciences, The University of Queensland, Brisbane, Australia
| | - Mary E Wlodek
- Department of Physiology, The University of Melbourne, Parkville, Australia
| | - John F Bertram
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Clayton, Australia.,Department of Anatomy and Developmental Biology, Monash University, Clayton, Australia
| | - Karen M Moritz
- School of Biomedical Sciences, The University of Queensland, Brisbane, Australia.,Child Health Research Centre, The University of Queensland, Brisbane, Australia
| |
Collapse
|
23
|
Zhu Z, Cao F, Li X. Epigenetic Programming and Fetal Metabolic Programming. Front Endocrinol (Lausanne) 2019; 10:764. [PMID: 31849831 PMCID: PMC6901800 DOI: 10.3389/fendo.2019.00764] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 10/21/2019] [Indexed: 12/30/2022] Open
Abstract
Fetal metabolic programming caused by the adverse intrauterine environment can induce metabolic syndrome in adult offspring. Adverse intrauterine environment introduces fetal long-term relatively irreversible changes in organs and metabolism, and thus causes fetal metabolic programming leading metabolic syndrome in adult offspring. Fetal metabolic programming of obesity and insulin resistance plays a key role in this process. The mechanism of fetal metabolic programming is still not very clear. It is suggested that epigenetic programming, also induced by the adverse intrauterine environment, is a critical underlying mechanism of fetal metabolic programming. Fetal epigenetic programming affects gene expression changes and cellular function through epigenetic modifications without DNA nucleotide sequence changes. Epigenetic modifications can be relatively stably retained and transmitted through mitosis and generations, and thereby induce the development of metabolic syndrome in adult offspring. This manuscript provides an overview of the critical role of epigenetic programming in fetal metabolic programming.
Collapse
Affiliation(s)
- Ziqiang Zhu
- Children's Hospital of Soochow University, Suzhou, China
- Changzhou Maternity and Child Health Care Hospital affiliated to Nanjing Medical University, Changzhou, China
| | - Fang Cao
- Changzhou Maternity and Child Health Care Hospital affiliated to Nanjing Medical University, Changzhou, China
| | - Xiaozhong Li
- Children's Hospital of Soochow University, Suzhou, China
| |
Collapse
|
24
|
Hofstee P, Bartho LA, McKeating DR, Radenkovic F, McEnroe G, Fisher JJ, Holland OJ, Vanderlelie JJ, Perkins AV, Cuffe JSM. Maternal selenium deficiency during pregnancy in mice increases thyroid hormone concentrations, alters placental function and reduces fetal growth. J Physiol 2019; 597:5597-5617. [PMID: 31562642 DOI: 10.1113/jp278473] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 09/26/2019] [Indexed: 12/25/2022] Open
Abstract
KEY POINTS Inappropriate intake of key micronutrients in pregnancy is known to alter maternal endocrine status, impair placental development and induce fetal growth restriction. Selenium is an essential micronutrient required for the function of approximately 25 important proteins. However, the specific effects of selenium deficiency during pregnancy on maternal, placental and fetal outcomes are poorly understood. The present study demonstrates that maternal selenium deficiency increases maternal triiodothyronine and tetraiodothyronine concentrations, reduces fetal blood glucose concentrations, and induces fetal growth restriction. Placental expression of key selenium-dependent thyroid hormone converting enzymes were reduced, whereas the expression of key placental nutrient transporters was dysregulated. Selenium deficiency had minimal impact on selenium-dependent anti-oxidants but increased placental copper concentrations and expression of superoxide dismutase 1. These results highlight the idea that selenium deficiency during pregnancy may contribute to thyroid dysfunction, causing reduced fetal growth, that may precede programmed disease outcomes in offspring. ABSTRACT Selenium is a trace element fundamental to diverse homeostatic processes, including anti-oxidant regulation and thyroid hormone metabolism. Selenium deficiency in pregnancy is common and increases the risk of pregnancy complications including fetal growth restriction. Although altered placental formation may contribute to these poor outcomes, the mechanism by which selenium deficiency contributes to complications in pregnancy is poorly understood. Female C57BL/6 mice were randomly allocated to control (>190 µg kg-1 , n = 8) or low selenium (<50 µg kg-1 , n = 8) diets 4 weeks prior to mating and throughout gestation. Pregnant mice were killed at embryonic day 18.5 followed by collection of maternal and fetal tissue. Maternal and fetal plasma thyroid hormone concentrations were analysed, as was placental expression of key selenoproteins involved in thyroid metabolism and anti-oxidant defences. Selenium deficiency increased plasma tetraiodothyronine and triiodothyronine concentrations. This was associated with a reduction in placental expression of key selenodependent deiodinases, DIO2 and DIO3. Placental expression of selenium-dependent anti-oxidants was unaffected by selenium deficiency. Selenium deficiency reduced fetal glucose concentrations, leading to reduced fetal weight. Placental glycogen content was increased within the placenta, as was Slc2a3 mRNA expression. This is the first study to demonstrate that selenium deficiency may reduce fetal weight through increased maternal thyroid hormone concentrations, impaired placental thyroid hormone metabolism and dysregulated placental nutrient transporter expression. The study suggests that the magnitude of selenium deficiency commonly reported in pregnant women may be sufficient to impair thyroid metabolism but not placental anti-oxidant concentrations.
Collapse
Affiliation(s)
- Pierre Hofstee
- School of Medical Science, Menzies Health Institute Queensland, Griffith University Gold Coast Campus, Southport, QLD, Australia
| | - Lucy A Bartho
- School of Medical Science, Menzies Health Institute Queensland, Griffith University Gold Coast Campus, Southport, QLD, Australia
| | - Daniel R McKeating
- School of Medical Science, Menzies Health Institute Queensland, Griffith University Gold Coast Campus, Southport, QLD, Australia
| | - Filip Radenkovic
- School of Medical Science, Menzies Health Institute Queensland, Griffith University Gold Coast Campus, Southport, QLD, Australia
| | - Georgia McEnroe
- School of Medical Science, Menzies Health Institute Queensland, Griffith University Gold Coast Campus, Southport, QLD, Australia
| | - Joshua J Fisher
- School of Medical Science, Menzies Health Institute Queensland, Griffith University Gold Coast Campus, Southport, QLD, Australia
| | - Olivia J Holland
- School of Medical Science, Menzies Health Institute Queensland, Griffith University Gold Coast Campus, Southport, QLD, Australia
| | | | - Anthony V Perkins
- School of Medical Science, Menzies Health Institute Queensland, Griffith University Gold Coast Campus, Southport, QLD, Australia
| | - James S M Cuffe
- School of Medical Science, Menzies Health Institute Queensland, Griffith University Gold Coast Campus, Southport, QLD, Australia
- The School of Biomedical Sciences, The University of Queensland, St Lucia, QLD, Australia
| |
Collapse
|
25
|
From Genotype to Phenotype: Through Chromatin. Genes (Basel) 2019; 10:genes10020076. [PMID: 30678090 PMCID: PMC6410296 DOI: 10.3390/genes10020076] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 01/16/2019] [Accepted: 01/21/2019] [Indexed: 02/07/2023] Open
Abstract
Advances in sequencing technologies have enabled the exploration of the genetic basis for several clinical disorders by allowing identification of causal mutations in rare genetic diseases. Sequencing technology has also facilitated genome-wide association studies to gather single nucleotide polymorphisms in common diseases including cancer and diabetes. Sequencing has therefore become common in the clinic for both prognostics and diagnostics. The success in follow-up steps, i.e., mapping mutations to causal genes and therapeutic targets to further the development of novel therapies, has nevertheless been very limited. This is because most mutations associated with diseases lie in inter-genic regions including the so-called regulatory genome. Additionally, no genetic causes are apparent for many diseases including neurodegenerative disorders. A complementary approach is therefore gaining interest, namely to focus on epigenetic control of the disease to generate more complete functional genomic maps. To this end, several recent studies have generated large-scale epigenetic datasets in a disease context to form a link between genotype and phenotype. We focus DNA methylation and important histone marks, where recent advances have been made thanks to technology improvements, cost effectiveness, and large meta-scale epigenome consortia efforts. We summarize recent studies unravelling the mechanistic understanding of epigenetic processes in disease development and progression. Moreover, we show how methodology advancements enable causal relationships to be established, and we pinpoint the most important issues to be addressed by future research.
Collapse
|