1
|
Cortese R, Cataldo K, Hummel J, Smith G, Ortega M, Richey M, Winn H, Gozal D, Goodman JR. Fetal growth disorders detection during first trimester gestation through comprehensive maternal circulating DNA profiling. Hum Mol Genet 2025:ddaf055. [PMID: 40263746 DOI: 10.1093/hmg/ddaf055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 03/18/2025] [Accepted: 04/04/2025] [Indexed: 04/24/2025] Open
Abstract
BACKGROUND Early diagnosis, close follow-up and timely delivery constitute the main elements for appropriate detection and management of Fetal Growth Disorders (FGD). We hypothesized that fetoplacental FGD-associated alterations can be detected in circulating DNA (cirDNA) samples isolated from maternal blood, as early as the first gestational trimester. METHODS Plasma cirDNA was isolated from samples prospectively collected during first trimester gestation (n = 56). Small, Large and Appropriate for Gestational Age (SGA n = 11, LGA n = 18, and AGA n = 29, respectively) status was determined at birth according to weight and gestational age. cirDNA amount, fragmentation, mitochondrial/nuclear ratio and cirDNA methylation profiles were quantified using qPCR-based assays. Machine learning approaches were applied to build a molecular signature for prediction of LGA and SGA. Prediction accuracy was assessed by Receiver-Operating Curve (ROC) analysis, and Positive and Negative Predictive values (PPV and NPV, respectively) were calculated. RESULTS Total concentration of plasma cirDNA, cirDNA fragmentation and ratio of mitochondrial/nuclear cirDNA were increased in SGA and LGA compared to AGA pregnancies. Out of the 10 selected loci, we detected 5 genes (HSD2, RASSF1, CYP19A1, IL10, and LEP) showing significant differential methylation differences (p < 0.05) across the SGA, AGA and LGA samples at first trimester. cirDNA marker signature discriminated between FGD and AGA pregnancies with high accuracy (AUC > 0.95), achieving 88.8% PPV and 85.7% NPV. CONCLUSIONS Maternal blood cirDNA profiles accurately detects early gestation FGD. The proposed novel marker panel hold great potential for implementation of low invasive approaches for reliable prediction of FGDs, enabling a disruptive path toward precision medicine in FGD.
Collapse
Affiliation(s)
- Rene Cortese
- Department of Gynecology, Obstetrics and Women's Health, University of Missouri, 1 Hospital Dr. Columbia, MO 65212, United States
- Department of Pediatrics, School of Medicine, University of Missouri, 1 Hospital Dr. Columbia, MO 65212, United States
- Institute for Data Science and Informatics, University of Missouri, Columbia, 22 Heinkel Building, MO 65211 United States
| | - Kylie Cataldo
- Department of Gynecology, Obstetrics and Women's Health, University of Missouri, 1 Hospital Dr. Columbia, MO 65212, United States
- Department of Pediatrics, School of Medicine, University of Missouri, 1 Hospital Dr. Columbia, MO 65212, United States
| | - Justin Hummel
- Institute for Data Science and Informatics, University of Missouri, Columbia, 22 Heinkel Building, MO 65211 United States
| | - Gracie Smith
- Department of Gynecology, Obstetrics and Women's Health, University of Missouri, 1 Hospital Dr. Columbia, MO 65212, United States
| | - Madison Ortega
- Department of Gynecology, Obstetrics and Women's Health, University of Missouri, 1 Hospital Dr. Columbia, MO 65212, United States
| | - Madison Richey
- Department of Gynecology, Obstetrics and Women's Health, University of Missouri, 1 Hospital Dr. Columbia, MO 65212, United States
| | - Hung Winn
- Department of Gynecology, Obstetrics and Women's Health, University of Missouri, 1 Hospital Dr. Columbia, MO 65212, United States
- Saint Luke's Hospital Maternal Fetal Medicine Specialists, Kansas City, MO, 64111, United States
| | - David Gozal
- Department of Pediatrics, School of Medicine, University of Missouri, 1 Hospital Dr. Columbia, MO 65212, United States
| | - Jean Ricci Goodman
- Department of Gynecology, Obstetrics and Women's Health, University of Missouri, 1 Hospital Dr. Columbia, MO 65212, United States
| |
Collapse
|
2
|
Woestenberg PJ, Maas VYF, Vissers LCM, Oliveri NMB, Kant AC, de Feijter M. The association between coronavirus disease 2019 vaccination during pregnancy and neonatal health outcomes. Pediatr Investig 2025; 9:41-51. [PMID: 40241888 PMCID: PMC11998170 DOI: 10.1002/ped4.12456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 09/12/2024] [Indexed: 04/18/2025] Open
Abstract
Importance Pregnant women have a higher risk of severe illness or complications due to a severe acute respiratory syndrome coronavirus 2 infection. To reduce these risks, pregnant women are advised to coronavirus disease 2019 (COVID-19) vaccination. Continued monitoring of the safety of maternal COVID-19 vaccination remains important. Objective To evaluate the association between maternal COVID-19 vaccination and neonatal health. Methods Data from the Dutch Pregnancy Drug Register were used. In this prospective cohort study, pregnant women self-reported COVID-19 vaccination and neonatal health outcomes. We included women with a due date between January 15, 2021, and May 15, 2022, and a singleton live birth after at least 24 weeks gestation. Using log-binomial regression analysis we studied the association between COVID-19 vaccination during pregnancy and the health outcomes; small for gestational age (SGA), large for gestational age (LGA), and neonatal health problems. We corrected for potential confounders using inverse probability of treatment weighting. Results In total, 3655 participants were included (92.1% COVID-19 vaccinated during pregnancy). Of all participants, 8.9% reported SGA, 11.1% reported LGA, and 16.4% reported neonatal health problems. Maternal COVID-19 vaccination was not statistically significantly associated with SGA (adjusted prevalence ratio [aPR]: 0.90; 95% confidence interval [CI]: 0.59-1.36), LGA (aPR: 1.07; 95% CI: 0.70-1.63), or neonatal health problems (aPR: 0.84; 95% CI: 0.63-1.11). Interpretation This study indicates that COVID-19 vaccination during pregnancy is not associated with self-reported adverse neonatal health outcomes. These findings contribute to the growing body of evidence on the safety of COVID-19 vaccination during pregnancy.
Collapse
Affiliation(s)
| | | | | | | | - Agnes C. Kant
- Netherlands Pharmacovigilance Centre Lareb’s‐Hertogenboschthe Netherlands
- Department of Clinical Pharmacology and ToxicologyLeiden University Medical CentreLeidenthe Netherlands
| | - Maud de Feijter
- Netherlands Pharmacovigilance Centre Lareb’s‐Hertogenboschthe Netherlands
| |
Collapse
|
3
|
Walsh CJ, Meyers ML, Chandnani N, Barker AJ, Fujiwara T, Mirsky DM, Englund EK. Quantitative evaluation of placental microvascular blood flow and microstructure in fetal growth restriction with IVIM MRI. Pediatr Radiol 2025; 55:546-555. [PMID: 39853392 DOI: 10.1007/s00247-024-06151-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 12/17/2024] [Accepted: 12/23/2024] [Indexed: 01/26/2025]
Abstract
BACKGROUND Intravoxel incoherent motion (IVIM) MRI uses diffusion-weighted (DW) MRI acquisitions to evaluate the microvascular and cellular environments of tissue. Due to these properties, IVIM has been increasingly utilized to evaluate abnormal placentation. OBJECTIVE Our primary objective was to compare IVIM parameters in the placenta of patients with fetal growth restriction and appropriate for gestational age controls across gestational ages. Our secondary aim was to quantify IVIM parameters in fetal versus maternal placental aspects to evaluate microvascular and parenchymal differences. MATERIALS AND METHODS With IRB approval, pregnant individuals with a diagnosis of fetal growth restriction (n=24) and controls (n=22) were retrospectively analyzed. DW-MRI data were collected at 1.5 T with nine b-values (range, 0 s/mm2 to 1,000 s/mm2). Data were processed by one non-blinded reader to obtain mean perfusion fraction (f), pseudo-diffusion coefficient (D*), their product fD*, and diffusion coefficient (D) in maternal and fetal aspects of the placenta, defined by bisecting the thickness of the placenta. Repeated measures of two-way ANOVAs were used to compare IVIM parameters in fetal and maternal placental aspects between participant groups. Correlations between IVIM parameters and gestational age were also evaluated in both groups. RESULTS The average gestational age at MRI was 27±4 weeks for both groups. The percentile estimated fetal weight was 5.4±5.9% for the fetal growth restriction group and 42.8±28.5% for controls (P<0.001). The parameters D* and fD*, related to microvascular blood flow and tissue perfusion, were significantly lower in participants with fetal growth restriction compared to controls (D*, 40.5 vs. 52.4×10-3 mm2/s, P=0.043; fD*, 14.7 vs. 21.0×10-3 mm2/s, P=0.019). No other differences were observed, nor associations between gestational age and IVIM parameters in either group. CONCLUSION Our results suggest reduced microvascular flow in placentas of patients with fetal growth restriction compared to controls.
Collapse
Affiliation(s)
- Caroline J Walsh
- University of Colorado Anschutz Medical Campus, 13001 E 17th Pl, Aurora, CO, 80045, USA.
| | - Mariana L Meyers
- University of Colorado Anschutz Medical Campus, 13001 E 17th Pl, Aurora, CO, 80045, USA
- Children's Hospital Colorado, Aurora, USA
| | - Neal Chandnani
- University of Colorado Anschutz Medical Campus, 13001 E 17th Pl, Aurora, CO, 80045, USA
| | - Alex J Barker
- University of Colorado Anschutz Medical Campus, 13001 E 17th Pl, Aurora, CO, 80045, USA
- Children's Hospital Colorado, Aurora, USA
| | - Takashi Fujiwara
- University of Colorado Anschutz Medical Campus, 13001 E 17th Pl, Aurora, CO, 80045, USA
- Children's Hospital Colorado, Aurora, USA
| | - David M Mirsky
- University of Colorado Anschutz Medical Campus, 13001 E 17th Pl, Aurora, CO, 80045, USA
- Children's Hospital Colorado, Aurora, USA
| | - Erin K Englund
- University of Colorado Anschutz Medical Campus, 13001 E 17th Pl, Aurora, CO, 80045, USA.
- Children's Hospital Colorado, Aurora, USA.
| |
Collapse
|
4
|
Benítez-Marín MJ, Blasco-Alonso M, de Rodríguez de Fonseca F, Jiménez JS, Rivera P, González-Mesa E. Evaluating neuronal damage biomarkers at birth for predicting neurodevelopmental risks in foetal growth restriction. Acta Paediatr 2025; 114:272-284. [PMID: 39601356 DOI: 10.1111/apa.17521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 10/22/2024] [Accepted: 11/19/2024] [Indexed: 11/29/2024]
Abstract
AIM This study was based on the need to predict neurodevelopmental outcomes of children with foetal growth restriction. The aim was to systematically review the correlation between biomarkers of neural injury in children with foetal growth restriction and their neurodevelopment. METHOD Following Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines, the review included studies on growth-restricted foetuses that measured biomarkers of postpartum brain injury and assessed neurodevelopment in childhood. Studies published between 1 January 2014 and 31 March 2024 were identified through PubMed and Embase, with the study protocol registered in PROSPERO (CRD42024520254). RESULTS Only five met the inclusion criteria. Results showed that urinary S100B levels were significantly elevated in foetal growth restriction, negatively correlating with neurological development at 7 days of life. Neuron-specific enolase negatively correlated with cognitive, motor and socio-emotional development. Urinary nerve growth factor levels were significantly lower in neonates with foetal growth restriction, correlating with poor neurodevelopment. No alterations in BDNF levels were observed. Tau protein levels were lower in children with foetal growth restriction and adverse outcomes. CONCLUSION The study emphasised the need for further research on biomarkers and predictive models of neurodevelopment in children with foetal growth restriction.
Collapse
Affiliation(s)
- Mª José Benítez-Marín
- Research Group in Maternal-Fetal Medicine, Epigenetics, Women's Diseases and Reproductive Health, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma Bionand, Málaga, Spain
- Obstetrics and Gynecology Service, Regional University Hospital of Malaga, Málaga, Spain
- Obstetrics and Gynecology Service, Virgen de la Victoria University Hospital, Málaga, Spain
| | - Marta Blasco-Alonso
- Research Group in Maternal-Fetal Medicine, Epigenetics, Women's Diseases and Reproductive Health, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma Bionand, Málaga, Spain
- Obstetrics and Gynecology Service, Regional University Hospital of Malaga, Málaga, Spain
- Surgical Specialties, Biochemistry and Immunology Department, Málaga University, Málaga, Spain
| | - Fernando de Rodríguez de Fonseca
- Instituto de Investigación Biomédica de Málaga (IBIMA)-Plataforma BIONAND, Málaga, Spain
- Servicio Neurologia, Hospital Regional Universitario de Málaga, Málaga, Spain
| | - Jesús S Jiménez
- Research Group in Maternal-Fetal Medicine, Epigenetics, Women's Diseases and Reproductive Health, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma Bionand, Málaga, Spain
- Obstetrics and Gynecology Service, Regional University Hospital of Malaga, Málaga, Spain
- Surgical Specialties, Biochemistry and Immunology Department, Málaga University, Málaga, Spain
| | - Patricia Rivera
- Instituto de Investigación Biomédica de Málaga (IBIMA)-Plataforma BIONAND, Málaga, Spain
- UGC Salud Mental, Hospital Regional Universitario de Málaga, Málaga, Spain
| | - Ernesto González-Mesa
- Research Group in Maternal-Fetal Medicine, Epigenetics, Women's Diseases and Reproductive Health, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma Bionand, Málaga, Spain
- Obstetrics and Gynecology Service, Regional University Hospital of Malaga, Málaga, Spain
- Surgical Specialties, Biochemistry and Immunology Department, Málaga University, Málaga, Spain
| |
Collapse
|
5
|
Saeed B, ALbalawi A, Bintalib M, Alturki A, De Vol EB, ALzayed B, Mohty D, Veldtman G, AlMugbel M, Latta N, Joueidi F, Kurdi W. Pregnancy Outcomes in Women with Low and Ultra-Low Ejection Fraction: A Retrospective Study in a Tertiary Care Center. J Clin Med 2025; 14:745. [PMID: 39941415 PMCID: PMC11818610 DOI: 10.3390/jcm14030745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 11/06/2024] [Accepted: 11/13/2024] [Indexed: 02/16/2025] Open
Abstract
The data about pregnancy while having a low ejection fraction are scarce, since pregnancy is not recommended for women with an ejection fraction of less than 30%. There is an increased risk of obstetrical complications and adverse maternal-fetal outcomes. Pregnancy is a rough journey for this group of patients. However, a successful pregnancy can be achieved when cardiac complications are managed during pregnancy. The early recognition of women at risk of cardiovascular events and early referral can optimize the maternal and neonatal outcomes with close collaboration between the maternal-fetal medicine specialist and the cardiologist. The study's aim was to assess the experience of our tertiary center with regard to the adverse maternal outcome for women with an ejection fraction ≤ 30% compared to those with an EF > 30% in our tertiary center. The fetal and obstetric outcome for pregnancies with an EF ≤ 30% was compared to that for pregnancies with an EF > 30%. Methodology: After receiving the approval of the local Ethical Board Review, a retrospective study was conducted at King Faisal Specialist Hospital and Research Center (KFSHRC) in the city of Riyadh, Kingdom of Saudi Arabia. Our study population included women with cardiomyopathy (acquired or congenital) who were followed up or delivered in KFSHRC from the period of January 2004 till March 2020. Cases were identified by reviewing the database from the Cardiac Center Echocardiograph and maternal fetal medicine unit. The data on the maternal and fetal outcome were gathered from the hospital medical records. An adverse maternal outcome included: death, hospitalization due to decompensated heart failure, and worsening cardiovascular status during pregnancy. Adverse fetal outcomes included: miscarriages, termination of pregnancy, FGR, and placental insufficiency. Obstetrics complications included: complications related to the mode of delivery, antepartum hemorrhage, postpartum hemorrhage, or preeclampsia. Results: Our study included 44 subjects, examining the differences between those with an ejection fraction greater than 30 (n = 21 subjects) and those with an ejection fraction less than or equal to 30 (n = 23) with respect to demographics, co-morbidities, and outcomes (maternal, pregnancy, fetal, ultrasound, and baby). There was no evidence of any differences in the demographics. From among the co-morbidities, there was a statistically higher rate of dilated cardiomyopathy and lower rate of rheumatic heart disease in those with a lower ejection fraction. Also, women with a lower ejection fraction tended to deliver through a means other than simple vaginal delivery. There was a significant association (p = 0.0296) indicating that individuals with a lower ejection fraction tended to have a lower gestational age at delivery. The information on whether the pregnancy resulted in a live birth was available for all but one of the mothers. Across all the mothers, 32 (74%) resulted in a live birth and 11 did not. The percentage of pregnancies resulting in a live birth in the group for which the ejection fraction was greater than 30 was 90% and that in the group for which the ejection fraction was less than or equal to 30 was 59% (p = 0.0339). From among the ultrasound and baby outcomes, only the rate of the babies being discharged alive differed statistically between the two ejection fraction groups, with those mothers having a lower ejection fraction experiencing fewer babies being discharged alive (p = 0.0310). Conclusions: In conclusion, women with a low ejection fraction are at an increased risk of maternal-fetal complications. In our study, the lower the EF (≤30) the worse were the fetal and neonatal outcomes; however, in terms of the maternal outcomes, it was the same whether the EF was low or ultra-low. Yet, these groups of patients need to be counseled about the facts of poor obstetrical outcomes with an emphasis on preconceptual counseling.
Collapse
Affiliation(s)
- Bashayer Saeed
- Department of Obstetrics & Gynecology, King Faisal Specialist Hospital & Research Centre, Riyadh 11211, Saudi Arabia; (B.S.); (A.A.); (M.B.); (A.A.); (M.A.)
| | - Amani ALbalawi
- Department of Obstetrics & Gynecology, King Faisal Specialist Hospital & Research Centre, Riyadh 11211, Saudi Arabia; (B.S.); (A.A.); (M.B.); (A.A.); (M.A.)
| | - Marwah Bintalib
- Department of Obstetrics & Gynecology, King Faisal Specialist Hospital & Research Centre, Riyadh 11211, Saudi Arabia; (B.S.); (A.A.); (M.B.); (A.A.); (M.A.)
| | - Amjad Alturki
- Department of Obstetrics & Gynecology, King Faisal Specialist Hospital & Research Centre, Riyadh 11211, Saudi Arabia; (B.S.); (A.A.); (M.B.); (A.A.); (M.A.)
| | - Edward B. De Vol
- Biostatistics, Epidemiology & Scientific Computing, King Faisal Specialist Hospital & Research Centre, Riyadh 11211, Saudi Arabia; (E.B.D.V.); (B.A.)
| | - Balqees ALzayed
- Biostatistics, Epidemiology & Scientific Computing, King Faisal Specialist Hospital & Research Centre, Riyadh 11211, Saudi Arabia; (E.B.D.V.); (B.A.)
| | - Dania Mohty
- Cardiovascular Unit, King Faisal Specialist Hospital & Research Centre, Riyadh 11211, Saudi Arabia; (D.M.); (G.V.)
| | - Gruschen Veldtman
- Cardiovascular Unit, King Faisal Specialist Hospital & Research Centre, Riyadh 11211, Saudi Arabia; (D.M.); (G.V.)
| | - Maisoon AlMugbel
- Department of Obstetrics & Gynecology, King Faisal Specialist Hospital & Research Centre, Riyadh 11211, Saudi Arabia; (B.S.); (A.A.); (M.B.); (A.A.); (M.A.)
| | - Nayef Latta
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (N.L.); (F.J.)
| | - Faisal Joueidi
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (N.L.); (F.J.)
| | - Wesam Kurdi
- Department of Obstetrics & Gynecology, King Faisal Specialist Hospital & Research Centre, Riyadh 11211, Saudi Arabia; (B.S.); (A.A.); (M.B.); (A.A.); (M.A.)
| |
Collapse
|
6
|
Aziz Ali S, Genkinger J, Kahe K, Valeri L, Khowaja N, Krebs NF, Kuhn L. Role of preconception nutrition supplements in maternal anemia and intrauterine growth: a systematic review and meta-analysis of randomized controlled trials. Syst Rev 2025; 14:11. [PMID: 39806515 PMCID: PMC11727609 DOI: 10.1186/s13643-024-02726-7] [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: 09/20/2024] [Accepted: 11/28/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Impaired intrauterine growth, a significant global health problem, contributes to a higher burden of infant morbidity and mortality, mainly in resource-poor settings. Maternal anemia and undernutrition, two important causes of impaired intrauterine growth, are prioritized by global nutrition targets of 2030. We synthesized the evidence on the role of preconception nutrition supplements in reducing maternal anemia and improving intrauterine growth. METHODS We undertook a review of the randomized controlled trials (RCTs) assessing the effect of preconception nutrition supplements on maternal hemoglobin, an indicator to estimate maternal anemia, and markers of intrauterine growth including birth weight, length, head circumference, and small for gestational age. Additionally, we examined preterm birth as an important perinatal outcome. We searched PubMed, CINAHL, Web of Science, Cochrane Central, and Embase. We computed summary mean differences and risk ratios (RR) with 95% confidence intervals (CIs) using random-effect models. We employed I2 and Cochran's Q test statistics to assess heterogeneity. We used a revised Cochrane risk-of-bias (RoB version 2.0) and GRADE (grading of recommendations, assessment, development, and evaluation) tools to assess the risk of bias and quality of evidence of eligible RCTs, respectively. RESULTS We identified 20 eligible RCTs (n = 27,659 women). Preconception nutrition supplements (iron and folic acid, multiple micronutrients, and a lipid-based nutrient supplement) overall increased maternal hemoglobin by 0.30 g/dL ((0.03, 0.57); I2 = 79%; n=9). However, we did not find a significant effect of the supplements on birth weight (12.25 gm ((- 22.66, 47.16); I2 = 55%; n=10)), length (0.15 cm (- 0.26, 0.56); I2 = 68%; n = 5), head circumference (- 0.23 cm (- 0.88, 0.43); I2 = 84%; n=4), small for gestational age (RR 0.91 (0.80, 1.04); I2 = 31%; n=8), or preterm birth (RR 0.93 (0.69,1.25); I2 = 57%; n=12). In general, the quality of evidence was assessed as very low to moderate. CONCLUSION Preconception nutrition supplements studied to date appear to reduce maternal anemia. However, it is uncertain whether there are beneficial effects of the supplements on intrauterine growth. Low quality of evidence warrants future well-designed RCTs to produce solid scientific data, particularly of a more comprehensive package of preconception nutrition supplements that include both macro- and micronutrients. SYSTEMATIC REVIEW REGISTRATION PROSPERO CRD42023464966.
Collapse
Affiliation(s)
- Sumera Aziz Ali
- Department of Epidemiology, Mailman School of Public Health, Columbia University Irving Medical Center, New York, NY, USA.
| | - Jeanine Genkinger
- Department of Epidemiology, Mailman School of Public Health, Columbia University Irving Medical Center, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Ka Kahe
- Department of Epidemiology, Mailman School of Public Health, Columbia University Irving Medical Center, New York, NY, USA
- Department of Obstetrics and Gynecology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Linda Valeri
- Department of Biostatistics, Mailman School of Public Health, Columbia University Irving Medical Center, New York, NY, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Nayab Khowaja
- Liaquat University of Medical and Health Sciences, Jamshoro, Pakistan
| | - Nancy F Krebs
- Department of Paediatrics, Section of Nutrition, University of Colorado School of Medicine, Aurora, CO, USA
| | - Louise Kuhn
- Department of Epidemiology, Mailman School of Public Health, Columbia University Irving Medical Center, New York, NY, USA
- Gertrude H. Sergievsky Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
7
|
Puglisi CJ, McDonough J, Bianco-Miotto T, Grieger JA. General Practitioners perspectives on infant telomere length screening after a pregnancy complication: a qualitative analysis. Fam Pract 2024; 41:1025-1031. [PMID: 37262296 PMCID: PMC11636557 DOI: 10.1093/fampra/cmad064] [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] [Indexed: 06/03/2023] Open
Abstract
BACKGROUND Pregnancy complications can impact the mother and child's health in the short and longterm resulting in an increased risk of chronic disease later in life. Telomere length is a biomarker of future cardiometabolic diseases and may offer a novel way of identifying offspring most at risk for future chronic diseases. OBJECTIVE(S) To qualitatively explore General Practitioners' (GPs) perspectives on the feasibility and uptake for recommending a telomere screening test in children who were born after a pregnancy complication. METHODS Twelve semi-structured interviews were conducted with GPs within metropolitan Adelaide, South Australia. Interviews were audio recorded, transcribed verbatim, and analysed for codes and themes. RESULTS Two themes were generated: ethical considerations and practical considerations. Ethically, the GP participants discussed barriers including consenting on behalf of a child, parental guilt, and the impact of health insurance, whereas viewing it for health promotion was a facilitator. For practical considerations, barriers included the difficulty in identifying people eligible for screening, maintaining medical communication between service providers, and time and financial constraints, whereas linking screening for telomere length with existing screening would facilitate uptake. CONCLUSIONS GPs were generally supportive of potential telomere screening in infants, particularly via a saliva test that could be embedded in current antenatal care. However, several challenges, such as lack of knowledge, ethical considerations, and time and financial constraints, need to be overcome before such a test could be implemented into practice.
Collapse
Affiliation(s)
- Carolyn J Puglisi
- School of Agriculture, Food and Wine, University of Adelaide, Adelaide 5005, Australia
| | - Joshua McDonough
- School of Public Health, University of Adelaide, Adelaide 5005, Australia
| | - Tina Bianco-Miotto
- School of Agriculture, Food and Wine, University of Adelaide, Adelaide 5005, Australia
- Robinson Research Institute, University of Adelaide, Adelaide 5005, Australia
| | - Jessica A Grieger
- Robinson Research Institute, University of Adelaide, Adelaide 5005, Australia
- Adelaide Medical School, University of Adelaide, Adelaide 5005, Australia
| |
Collapse
|
8
|
Santos BM, de Souza JPA, Goulart LRDP, Petrine JCP, Alves FHF, Del Bianco-Borges B. Impacts of Anabolic-androgenic steroid supplementation on female health and offspring: Mechanisms, side effects, and medical perspectives. Saudi Pharm J 2024; 32:102205. [PMID: 39697477 PMCID: PMC11653648 DOI: 10.1016/j.jsps.2024.102205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 11/19/2024] [Indexed: 12/20/2024] Open
Abstract
The increasing prevalence of Anabolic-androgenic steroids (AAS) among women, driven by the pursuit of improved body aesthetics, characterized by higher lean mass and reduced adipose tissue, raises significant health concerns, particularly due to the limited knowledge regarding their effects on the female organism. Prolonged use and/or high doses of AAS are linked to various harmful side effects, including mood changes, psychiatric disorders, voice deepening, clitoromegaly, menstrual irregularities, and cardiovascular complications, prompting medical societies to discourage their widespread use due to insufficient evidence supporting their safety and efficacy. Studies in female rodents have shown that AAS can lead to increased aggression, inflammation, reduced neuronal density, and negative impacts on the myocardium and blood vessels. Additionally, maternal administration of androgens during pregnancy can adversely affect offspring's reproductive, neuronal, and metabolic health, resulting in long-term impairments. The complexity of the mechanisms underlying AAS effects, and their potential genotoxicity remains poorly understood. This review aims to elucidate the various ways in which AAS can impact female physiology and that of their offspring, highlight commonly used anabolic substances, and discuss the positions of medical societies regarding AAS use.
Collapse
Affiliation(s)
- Beatriz Menegate Santos
- Postgraduate Program in Health Science, Lavras Federal University - UFLA; University Campus, CP: 3037, Lavras 37203-202, Brazil
| | - Jessica Peres Alves de Souza
- Postgraduate Program in Health Science, Lavras Federal University - UFLA; University Campus, CP: 3037, Lavras 37203-202, Brazil
| | - Luísa Rodrigues de Paula Goulart
- Medicine Department, Health Science Faculty, Lavras Federal University - UFLA; University Campus, CP: 3037, Lavras 37203-202, Brazil
| | - Jéssica Castro Pereira Petrine
- Postgraduate Program in Health Science, Lavras Federal University - UFLA; University Campus, CP: 3037, Lavras 37203-202, Brazil
| | - Fernando Henrique Ferrari Alves
- Institute of Science, Technology and Innovation – Federal University of Lavras, Jardim Califórnia Garden 37950-000, São Sebastião do Paraíso, Minas Gerais, Brazil
| | - Bruno Del Bianco-Borges
- Postgraduate Program in Health Science, Lavras Federal University - UFLA; University Campus, CP: 3037, Lavras 37203-202, Brazil
- Medicine Department, Health Science Faculty, Lavras Federal University - UFLA; University Campus, CP: 3037, Lavras 37203-202, Brazil
| |
Collapse
|
9
|
Wu P, Wang J, Ji X, Chai J, Chen L, Zhang T, Long X, Tu Z, Chen S, Zhang L, Wang K, Zhang L, Guo Z, Wang J. Maternal Hypermethylated Genes Contribute to Intrauterine Growth Retardation of Piglets in Rongchang Pigs. Int J Mol Sci 2024; 25:6462. [PMID: 38928167 PMCID: PMC11203632 DOI: 10.3390/ijms25126462] [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: 04/16/2024] [Revised: 06/04/2024] [Accepted: 06/08/2024] [Indexed: 06/28/2024] Open
Abstract
The placenta is a crucial determinant of fetal survival, growth, and development. Deficiency in placental development directly causes intrauterine growth retardation (IUGR). IUGR can lead to fetal growth restriction and an increase in the mortality rate. The genetic mechanisms underlying IUGR development, however, remain unclear. In the present study, we integrated whole-genome DNA methylation and transcriptomic analyses to determine distinct gene expression patterns in various placental tissues to identify pivotal genes that are implicated with IUGR development. By performing RNA-sequencing analysis, 1487 differentially expressed genes (DEGs), with 737 upregulated and 750 downregulated genes, were identified in IUGR pigs (H_IUGR) compared with that in normal birth weight pigs (N_IUGR) (p < 0.05); furthermore, 77 miRNAs, 1331 lncRNAs, and 61 circRNAs were differentially expressed. The protein-protein interaction network analysis revealed that among these DEGs, the genes GNGT1, ANXA1, and CDC20 related to cellular developmental processes and blood vessel development were the key genes associated with the development of IUGR. A total of 495,870 differentially methylated regions were identified between the N_IUGR and H_IUGR groups, which included 25,053 differentially methylated genes (DMEs); moreover, the overall methylation level was higher in the H_IUGR group than in the N_IUGR group. Combined analysis showed an inverse correlation between methylation levels and gene expression. A total of 1375 genes involved in developmental processes, tissue development, and immune system regulation exhibited methylation differences in gene expression levels in the promoter regions and gene ontology regions. Five genes, namely, ANXA1, ADM, NRP2, SHH, and SMAD1, with high methylation levels were identified as potential contributors to IUGR development. These findings provide valuable insights that DNA methylation plays a crucial role in the epigenetic regulation of gene expression and mammalian development and that DNA-hypermethylated genes contribute to IUGR development in Rongchang pigs.
Collapse
Affiliation(s)
- Pingxian Wu
- Chongqing Academy of Animal Sciences, Rongchang, Chongqing 402460, China (S.C.)
- National Center of Technology Innovation for Pigs, Rongchang, Chongqing 402460, China
- Chongqing Modern Agricultural Industry Technology System, Chongqing 401120, China
| | - Junge Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Xiang Ji
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Jie Chai
- Chongqing Academy of Animal Sciences, Rongchang, Chongqing 402460, China (S.C.)
- National Center of Technology Innovation for Pigs, Rongchang, Chongqing 402460, China
- Chongqing Modern Agricultural Industry Technology System, Chongqing 401120, China
| | - Li Chen
- Chongqing Academy of Animal Sciences, Rongchang, Chongqing 402460, China (S.C.)
- National Center of Technology Innovation for Pigs, Rongchang, Chongqing 402460, China
- Chongqing Modern Agricultural Industry Technology System, Chongqing 401120, China
| | - Tinghuan Zhang
- Chongqing Academy of Animal Sciences, Rongchang, Chongqing 402460, China (S.C.)
- National Center of Technology Innovation for Pigs, Rongchang, Chongqing 402460, China
| | - Xi Long
- Chongqing Academy of Animal Sciences, Rongchang, Chongqing 402460, China (S.C.)
- National Center of Technology Innovation for Pigs, Rongchang, Chongqing 402460, China
- Chongqing Modern Agricultural Industry Technology System, Chongqing 401120, China
| | - Zhi Tu
- Chongqing Academy of Animal Sciences, Rongchang, Chongqing 402460, China (S.C.)
- National Center of Technology Innovation for Pigs, Rongchang, Chongqing 402460, China
- Chongqing Modern Agricultural Industry Technology System, Chongqing 401120, China
| | - Siqing Chen
- Chongqing Academy of Animal Sciences, Rongchang, Chongqing 402460, China (S.C.)
- National Center of Technology Innovation for Pigs, Rongchang, Chongqing 402460, China
| | - Lijuan Zhang
- Chongqing Academy of Animal Sciences, Rongchang, Chongqing 402460, China (S.C.)
- National Center of Technology Innovation for Pigs, Rongchang, Chongqing 402460, China
| | - Ketian Wang
- Chongqing Academy of Animal Sciences, Rongchang, Chongqing 402460, China (S.C.)
- National Center of Technology Innovation for Pigs, Rongchang, Chongqing 402460, China
| | - Liang Zhang
- Chongqing Academy of Animal Sciences, Rongchang, Chongqing 402460, China (S.C.)
- National Center of Technology Innovation for Pigs, Rongchang, Chongqing 402460, China
| | - Zongyi Guo
- Chongqing Academy of Animal Sciences, Rongchang, Chongqing 402460, China (S.C.)
- National Center of Technology Innovation for Pigs, Rongchang, Chongqing 402460, China
- Chongqing Modern Agricultural Industry Technology System, Chongqing 401120, China
| | - Jinyong Wang
- Chongqing Academy of Animal Sciences, Rongchang, Chongqing 402460, China (S.C.)
- National Center of Technology Innovation for Pigs, Rongchang, Chongqing 402460, China
- Chongqing Modern Agricultural Industry Technology System, Chongqing 401120, China
| |
Collapse
|
10
|
Han W, Song Z, Shan D, Shi Q. Fetal origins of obesity: a novel pathway of regulating appetite neurons in the hypothalamus of growth-restricted rat offspring. Arch Gynecol Obstet 2024; 309:2411-2419. [PMID: 37378669 PMCID: PMC11147910 DOI: 10.1007/s00404-023-07108-3] [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: 04/20/2023] [Accepted: 06/11/2023] [Indexed: 06/29/2023]
Abstract
PURPOSE Fetal growth restriction causes a series of sequelae, some of which, such as hyperphagia, reduced satiety and postnatal obesity, are believed to be associated with embryonic hypothalamic neurons impairment. The mechanisms underlying the linkage of fetal brain injuries to break the energy homeostasis have not been elucidated completely. Here, we aim to investigate the effect of intrauterine energy restriction on remodeling appetite neurons in the hypothalamus of fetal and postnatal infant rats. METHODS Low-protein (8%) diet combined with 75% energy restriction was used to establish an animal model. Rats offspring brain tissues, harvested from embryo day 18 and postnatal infant day 1, were sampled for dependent regulator analyses and master neuron assessment. RESULTS Growth-restricted rats showed the increased expression of Bsx and NPY in the hypothalamus as well as remodeling hypothalamic neurons differentiation compared to controls. Intriguingly, in cells cultured in vitro test, we found that activated effects of Bsx and NPY could be exacerbated by DNMT1 inhibitor. CONCLUSIONS In embryonic and early postnatal stage of FGR rats, we detected high concentrations of orexigenic neurons in the hypothalamus. DNMT1 activity is correlated with early embryonic neurogenesis by mediating the expression of Bsx and NPY. It may be one of the reasons for the abnormal development of the appetite regulation pathway and higher susceptibility to obesity in FGR offspring.
Collapse
Affiliation(s)
- Weiling Han
- Department of Obstetrics, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, No. 251 Yao Jia Yuan Road, Chao Yang District, Beijing, 100026, China
| | - Zhaoyi Song
- STI-Zhilian Research Institute for Innovation and Digital Health, Beijing, China
| | - Dan Shan
- Department of Obstetrics, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, No. 251 Yao Jia Yuan Road, Chao Yang District, Beijing, 100026, China
- Department of Obstetrics and Gynecology, The People's Hospital of Yongcheng, Dongcheng District, Yongcheng City, Henan Province, China
| | - Qingyun Shi
- Department of Obstetrics, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, No. 251 Yao Jia Yuan Road, Chao Yang District, Beijing, 100026, China.
| |
Collapse
|
11
|
Yadav MK, Khan ZA, Wang JH, Ansari A. Impact of Gut–Brain Axis on Hepatobiliary Diseases in Fetal Programming. JOURNAL OF MOLECULAR PATHOLOGY 2024; 5:215-227. [DOI: 10.3390/jmp5020014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2025] Open
Abstract
The hepatobiliary system is vital for the biotransformation and disposition of endogenous molecules. Any impairment in the normal functioning of the hepatobiliary system leads to a spectrum of hepatobiliary diseases (HBDs), such as liver cirrhosis, fatty liver, biliary dyskinesia, gallbladder cancer, etc. Especially in pregnancy, HBD may result in increased maternal and fetal morbidity and mortality. Maternal HBD is a burden to the fetus’s growth, complicates fetal development, and risks the mother’s life. In fetal programming, the maternal mechanism is significantly disturbed by multiple factors (especially diet) that influence the development of the fetus and increase the frequency of metabolic diseases later in life. Additionally, maternal under-nutrition or over-nutrition (especially in high-fat, high-carbohydrate, or protein-rich diets) lead to dysregulation in gut hormones (CCK, GLP-1, etc.), microbiota metabolite production (SCFA, LPS, TMA, etc.), neurotransmitters (POMC, NPY, etc.), and hepatobiliary signaling (insulin resistance, TNF-a, SREBPs, etc.), which significantly impact fetal programming. Recently, biotherapeutics have provided a new horizon for treating HBD during fetal programming to save the lives of the mother and fetus. This review focuses on how maternal impaired hepatobiliary metabolic signaling leads to disease transmission to the fetus mediated through the gut–brain axis.
Collapse
Affiliation(s)
- Mukesh Kumar Yadav
- Department of Microbiology, Central University of Punjab, Bathinda 151401, India
| | - Zeeshan Ahmad Khan
- Department of Physical Therapy, College of Health Medical Science and Engineering, Inje University, Gimhae 50834, Republic of Korea
| | - Jing-Hua Wang
- Department of Korean Medicine, Daejeon University, Daejeon 35235, Republic of Korea
| | - AbuZar Ansari
- Department of Obstetrics and Gynecology, Ewha Womans University Mokdong Hospital, Seoul 07984, Republic of Korea
| |
Collapse
|
12
|
Barbian ME, Owens JA, Naudin CR, Denning P, Patel RM, Jones RM. A high fiber diet or supplementation with Lactococcus lactis subspecies cremoris to pregnant mice confers protection against intestinal injury in adult offspring. Gut Microbes 2024; 16:2337317. [PMID: 38619316 PMCID: PMC11020553 DOI: 10.1080/19490976.2024.2337317] [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: 07/07/2023] [Accepted: 03/27/2024] [Indexed: 04/16/2024] Open
Abstract
The diet during pregnancy, or antenatal diet, influences the offspring's intestinal health. We previously showed that antenatal butyrate supplementation reduces injury in adult murine offspring with dextran sulfate sodium (DSS)-induced colitis. Potential modulators of butyrate levels in the intestine include a high fiber diet or dietary supplementation with probiotics. To test this, we supplemented the diet of pregnant mice with high fiber, or with the probiotic bacteria Lactococcus lactis subspecies cremoris or Lactobacillus rhamnosus GG. We then induced chronic colitis with DSS in their adult offspring. We demonstrate that a high fiber antenatal diet, or supplementation with Lactococcus lactis subspecies cremoris during pregnancy diminished the injury from DSS-induced colitis in offspring. These data are evidence that antenatal dietary interventions impact offspring gut health and define the antenatal diet as a therapeutic modality to enhance offspring intestinal health.
Collapse
Affiliation(s)
- Maria E. Barbian
- Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, Atlanta, USA
| | | | - Crystal R. Naudin
- Department of Medicine, Emory University School of Medicine, Atlanta, USA
| | - Patricia Denning
- Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, Atlanta, USA
| | - Ravi M. Patel
- Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, Atlanta, USA
| | - Rheinallt M. Jones
- Department of Pediatrics, Emory University School of Medicine, Atlanta, USA
| |
Collapse
|
13
|
Oh CM, Lee S, Kwon H, Hwangbo B, Cho H. Prevalence of pre-existing lung diseases and their association with income level among patients with lung cancer: a nationwide population-based case-control study in South Korea. BMJ Open Respir Res 2023; 10:e001772. [PMID: 37940354 PMCID: PMC10632895 DOI: 10.1136/bmjresp-2023-001772] [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: 04/17/2023] [Accepted: 10/13/2023] [Indexed: 11/10/2023] Open
Abstract
BACKGROUND This study aimed to estimate the prevalence of pre-existing lung diseases in patients with lung cancer compared to people without lung cancer and examine the association between income levels and pre-existing lung diseases. METHODS Data on patients with lung cancer (case) and the general population without lung cancer (non-cancer controls) matched by age, sex and region were obtained from the Korea National Health Insurance Service-National Health Information Database (n=51 586). Insurance premiums were divided into quintiles and medicaid patients. Conditional logistic regression models were used to examine the association between pre-existing lung diseases and the risk of lung cancer. The relationship between income level and the prevalence of pre-existing lung disease among patients with lung cancer was analysed using logistic regression models. RESULTS The prevalence of asthma (17.3%), chronic obstructive lung disease (COPD) (9.3%), pneumonia (9.1%) and pulmonary tuberculosis (1.6%) in patients with lung cancer were approximately 1.6-3.2 times higher compared with the general population without lung cancer. A significantly higher risk for lung cancer was observed in individuals with pre-existing lung diseases (asthma: OR=1.36, 95% CI 1.29 to 1.44; COPD: 2.11, 95% CI 1.94 to 2.31; pneumonia: 1.49, 95% CI 1.38 to 1.61; pulmonary tuberculosis: 2.16, 95% CI 1.75 to 2.66). Patients with lung cancer enrolled in medicaid exhibited higher odds of having pre-existing lung diseases compared with those in the top 20% income level (asthma: OR=1.75, 95% CI 1.56 to 1.96; COPD: 1.91, 95% CI 1.65 to 2.21; pneumonia: 1.73, 95% CI 1.50 to 2.01; pulmonary tuberculosis: 2.45, 95% CI 1.78 to 3.36). CONCLUSIONS Pre-existing lung diseases were substantially higher in patients with lung cancer than in the general population. The high prevalence odds of pre-existing lung diseases in medicaid patients suggests the health disparity arising from the lowest income group, underscoring a need for specialised lung cancer surveillance.
Collapse
Affiliation(s)
- Chang-Mo Oh
- Departments of Preventive Medicine, School of Medicine, Kyung Hee University, Seoul, South Korea
| | - Sanghee Lee
- Department of Cancer Control and Population Health, National Cancer Center, Goyang, Gyeonggi-do, South Korea
- Health Insurance Research Institute, National Health Insurance Service, Wonju, Gangwon-do, South Korea
| | - Hoejun Kwon
- Department of Cancer Control and Population Health, National Cancer Center, Goyang, Gyeonggi-do, South Korea
| | - Bin Hwangbo
- Division of Pulmonology, Center for Lung Cancer, National Cancer Center, Goyang, Gyeonggi-do, South Korea
| | - Hyunsoon Cho
- Department of Cancer AI and Digital Health, National Cancer Center, Goyang, Gyeonggi-do, South Korea
- Integrated Biostatistics Branch, Division of Cancer Data Science, National Cancer Center, Goyang, Gyeonggi-do, South Korea
| |
Collapse
|
14
|
Miranda J, Paules C, Noell G, Youssef L, Paternina-Caicedo A, Crovetto F, Cañellas N, Garcia-Martín ML, Amigó N, Eixarch E, Faner R, Figueras F, Simões RV, Crispi F, Gratacós E. Similarity network fusion to identify phenotypes of small-for-gestational-age fetuses. iScience 2023; 26:107620. [PMID: 37694157 PMCID: PMC10485038 DOI: 10.1016/j.isci.2023.107620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 04/19/2023] [Accepted: 08/09/2023] [Indexed: 09/12/2023] Open
Abstract
Fetal growth restriction (FGR) affects 5-10% of pregnancies, is the largest contributor to fetal death, and can have long-term consequences for the child. Implementation of a standard clinical classification system is hampered by the multiphenotypic spectrum of small fetuses with substantial differences in perinatal risks. Machine learning and multiomics data can potentially revolutionize clinical decision-making in FGR by identifying new phenotypes. Herein, we describe a cluster analysis of FGR based on an unbiased machine-learning method. Our results confirm the existence of two subtypes of human FGR with distinct molecular and clinical features based on multiomic analysis. In addition, we demonstrated that clusters generated by machine learning significantly outperform single data subtype analysis and biologically support the current clinical classification in predicting adverse maternal and neonatal outcomes. Our approach can aid in the refinement of clinical classification systems for FGR supported by molecular and clinical signatures.
Collapse
Affiliation(s)
- Jezid Miranda
- BCNatal – Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), IDIBAPS, University of Barcelona, and Centre for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain
- Department of Obstetrics and Gynecology, Faculty of Medicine, Universidad de Cartagena, Cartagena de Indias, Colombia
| | - Cristina Paules
- BCNatal – Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), IDIBAPS, University of Barcelona, and Centre for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain
- Aragon Institute of Health Research (IIS Aragon), Obstetrics Department, Hospital Clínico Universitario Lozano Blesa, Zaragoza, Spain
| | - Guillaume Noell
- University of Barcelona, Biomedicine Department, IDIBAPS, Centre for Biomedical Research on Respiratory Diseases (CIBERES), Barcelona, Spain
| | - Lina Youssef
- BCNatal – Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), IDIBAPS, University of Barcelona, and Centre for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain
| | | | - Francesca Crovetto
- BCNatal – Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), IDIBAPS, University of Barcelona, and Centre for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain
| | - Nicolau Cañellas
- Metabolomics Platform, IISPV, DEEiA, Universidad Rovira i Virgili, Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Tarragona, Spain
| | - María L. Garcia-Martín
- BIONAND, Andalusian Centre for Nanomedicine and Biotechnology, Junta de Andalucía, Universidad de Málaga, Málaga, Spain
| | | | - Elisenda Eixarch
- BCNatal – Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), IDIBAPS, University of Barcelona, and Centre for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain
| | - Rosa Faner
- University of Barcelona, Biomedicine Department, IDIBAPS, Centre for Biomedical Research on Respiratory Diseases (CIBERES), Barcelona, Spain
| | - Francesc Figueras
- BCNatal – Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), IDIBAPS, University of Barcelona, and Centre for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain
| | - Rui V. Simões
- BCNatal – Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), IDIBAPS, University of Barcelona, and Centre for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain
- Institute for Research & Innovation in Health (i3S), University of Porto, Porto, Portugal
| | - Fàtima Crispi
- BCNatal – Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), IDIBAPS, University of Barcelona, and Centre for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain
| | - Eduard Gratacós
- BCNatal – Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), IDIBAPS, University of Barcelona, and Centre for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain
| |
Collapse
|
15
|
Kent NL, Atluri SC, Moritz KM, Cuffe JSM. Maternal hypothyroidism in rats impairs placental nutrient transporter expression, increases labyrinth zone size, and impairs fetal growth. Placenta 2023; 139:148-158. [PMID: 37406552 DOI: 10.1016/j.placenta.2023.06.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 06/05/2023] [Accepted: 06/14/2023] [Indexed: 07/07/2023]
Abstract
INTRODUCTION Hypothyroidism during pregnancy is associated with fetal growth restriction (FGR). FGR is commonly caused by placental insufficiency and yet the role of hypothyroidism in placental regulation of fetal growth is unknown. This study aimed to investigate the effects of maternal hypothyroidism on placental nutrient transporter expression, placental morphology, and placental metabolism. METHODS Hypothyroidism was induced in female Sprague-Dawley rats by adding methimazole (MMI) to drinking water at moderate (MOD, MMI at 0.005% w/v) and severe (SEV, MMI at 0.02% w/v) doses from one week prior to pregnancy and throughout gestation. Maternal and fetal tissues were collected on embryonic day 20 (E20). RESULTS Hypothyroidism reduced fetal weight (PTrt<0.001) despite causing fetal hyperglycaemia (PTrt = 0.016). Placental weight was not affected by hypothyroidism however placental efficiency was reduced (PTrt<0.001), as was the junctional zone (JZ):labyrinth zone (LZ) weight ratio (PTrt = 0.005). LZ glycogen content was increased (PTrt = 0.029) and while mRNA expression of glucose transporters was reduced by hypothyroidism, only GLUT1 protein expression was reduced in male LZs. Maternal hypothyroidism reduced mitochondrial content (PTrt = 0.031), particularly in SEV males relative to CON males (P = 0.004). Protein expression of Complex V (P < 0.001) and Complex III (P = 0.002) of the electron transport chain were also reduced in males. Maternal hypothyroidism reduced LZ (PTrt<0.001) and fetal plasma triglycerides (P = 0.019) while fetal free fatty acids and the expression of LZ lipid transporters was not affected. DISCUSSION Overall, maternal hypothyroidism may lead to FGR through reduced maternal T4 availability, changes to placental morphology, altered nutrient transporter expression and sex-specific effects on placental metabolism. Changes to LZ glycogen and triglyceride stores as well as mitochondrial content suggest a metabolic shift from oxidative phosphorylation to anaerobic glycolysis in males. These changes also likely impact fetal substrate availability and therefore fetal growth.
Collapse
Affiliation(s)
- Nykola L Kent
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia
| | - Sharat C Atluri
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia
| | - Karen M Moritz
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia
| | - James S M Cuffe
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
16
|
Whitlock AE, Moskowitzova K, Kycia I, Zurakowski D, Fauza DO. Morphometric, Developmental, and Anti-Inflammatory Effects of Transamniotic Stem Cell Therapy (TRASCET) on the Fetal Heart and Lungs in a Model of Intrauterine Growth Restriction. Stem Cells Dev 2023; 32:484-490. [PMID: 37358376 DOI: 10.1089/scd.2023.0040] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/27/2023] Open
Abstract
Transamniotic stem cell therapy (TRASCET) with mesenchymal stem cells (MSCs) can attenuate placental inflammation and minimize intrauterine growth restriction (IUGR). We sought to determine whether MSC-based TRASCET could mitigate fetal cardiopulmonary effects of IUGR. Pregnant Sprague-Dawley dams were exposed to alternating 12-h hypoxia (10.5% O2) cycles in the last fourth of gestation. Their fetuses (n = 155) were divided into 4 groups. One group remained untreated (n = 42), while three groups received volume-matched intra-amniotic injections of either saline (sham; n = 34), or of syngeneic amniotic fluid-derived MSCs, either in their native state (TRASCET; n = 36) or "primed" by exposure to interferon-gamma and interleukin-1beta before administration in vivo (TRASCET-primed; n = 43). Normal fetuses served as additional controls (n = 30). Multiple morphometric and biochemical analyses were performed at term for select markers of cardiopulmonary development and inflammation previously shown to be affected by IUGR. Among survivors (75%; 117/155), fetal heart-to-body weight ratio was increased in both the sham and untreated groups (P < 0.001 for both) but normalized in the TRASCET and TRASCET-primed groups (P = 0.275, 0.069, respectively). Cardiac b-type natriuretic peptide levels were increased in all hypoxia groups compared with normal (P < 0.001), but significantly decreased from sham and untreated in both TRASCET groups (P < 0.0001-0.005). Heart tumor necrosis factor-alpha levels were significantly elevated in sham and TRASCET groups (P = 0.009, 0.002), but normalized in the untreated and TRASCET-primed groups (P = 0.256, 0.456). Lung transforming growth factor-beta levels were significantly increased in both sham and untreated groups (P < 0.001, 0.003), but normalized in both TRASCET groups (P = 0.567, 0.303). Similarly, lung endothelin-1 levels were elevated in sham and untreated groups (P < 0.001 for both), but normalized in both TRASCET groups (P = 0.367, 0.928). We conclude that TRASCET with MSCs decreases markers of fetal cardiac strain, insufficiency, and inflammation, as well as of pulmonary fibrosis and hypertension in the rodent model of IUGR.
Collapse
Affiliation(s)
- Ashlyn E Whitlock
- Department of Surgery, Boston Children's Hospital/Harvard Medical School, Boston, Massachusetts, USA
| | - Kamila Moskowitzova
- Department of Surgery, Boston Children's Hospital/Harvard Medical School, Boston, Massachusetts, USA
| | - Ina Kycia
- Department of Surgery, Boston Children's Hospital/Harvard Medical School, Boston, Massachusetts, USA
| | - David Zurakowski
- Department of Surgery, Boston Children's Hospital/Harvard Medical School, Boston, Massachusetts, USA
| | - Dario O Fauza
- Department of Surgery, Boston Children's Hospital/Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
17
|
Foteva V, Fisher JJ, Qiao Y, Smith R. Does the Micronutrient Molybdenum Have a Role in Gestational Complications and Placental Health? Nutrients 2023; 15:3348. [PMID: 37571285 PMCID: PMC10421405 DOI: 10.3390/nu15153348] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 07/24/2023] [Accepted: 07/25/2023] [Indexed: 08/13/2023] Open
Abstract
Molybdenum is an essential trace element for human health and survival, with molybdenum-containing enzymes catalysing multiple reactions in the metabolism of purines, aldehydes, and sulfur-containing amino acids. Recommended daily intakes vary globally, with molybdenum primarily sourced through the diet, and supplementation is not common. Although the benefits of molybdenum as an anti-diabetic and antioxidant inducer have been reported in the literature, there are conflicting data on the benefits of molybdenum for chronic diseases. Overexposure and deficiency can result in adverse health outcomes and mortality, although physiological doses remain largely unexplored in relation to human health. The lack of knowledge surrounding molybdenum intake and the role it plays in physiology is compounded during pregnancy. As pregnancy progresses, micronutrient demand increases, and diet is an established factor in programming gestational outcomes and maternal health. This review summarises the current literature concerning varied recommendations on molybdenum intake, the role of molybdenum and molybdoenzymes in physiology, and the contribution these play in gestational outcomes.
Collapse
Affiliation(s)
- Vladimira Foteva
- Mothers and Babies Research Program, Hunter Medical Research Institute, Newcastle, NSW 2305, Australia; (J.J.F.); (R.S.)
- School of Medicine and Public Health, University of Newcastle, Newcastle, NSW 2308, Australia
| | - Joshua J. Fisher
- Mothers and Babies Research Program, Hunter Medical Research Institute, Newcastle, NSW 2305, Australia; (J.J.F.); (R.S.)
- School of Medicine and Public Health, University of Newcastle, Newcastle, NSW 2308, Australia
| | - Yixue Qiao
- Academy of Pharmacy, Xi’an Jiaotong Liverpool University, Suzhou 215000, China;
| | - Roger Smith
- Mothers and Babies Research Program, Hunter Medical Research Institute, Newcastle, NSW 2305, Australia; (J.J.F.); (R.S.)
- School of Medicine and Public Health, University of Newcastle, Newcastle, NSW 2308, Australia
| |
Collapse
|
18
|
Opoku R, DeCata J, Phillips CL, Schulz LC. Effect of Genetically Reduced Maternal Myostatin on Late Gestation Maternal, Fetal, and Placental Metabolomes in Mice. Metabolites 2023; 13:719. [PMID: 37367877 PMCID: PMC10302353 DOI: 10.3390/metabo13060719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 05/26/2023] [Accepted: 05/28/2023] [Indexed: 06/28/2023] Open
Abstract
Myostatin (gene symbol: Mstn) is an autocrine and paracrine inhibitor of muscle growth. Pregnant mice with genetically reduced levels of myostatin give birth to offspring with greater adult muscle mass and bone biomechanical strength. However, maternal myostatin is not detectable in fetal circulations. Fetal growth is dependent on the maternal environment, and the provisioning of nutrients and growth factors by the placenta. Thus, this study examined the effect of reduced maternal myostatin on maternal and fetal serum metabolomes, as well as the placental metabolome. Fetal and maternal serum metabolomes were highly distinct, which is consistent with the role of the placenta in creating a specific fetal nutrient environment. There was no effect from myostatin on maternal glucose tolerance or fasting insulin. In comparisons between pregnant control and Mstn+/- mice, there were more significantly different metabolite concentrations in fetal serum, at 50, than in the mother's serum at 33, confirming the effect of maternal myostatin reduction on the fetal metabolic milieu. Polyamines, lysophospholipids, fatty acid oxidation, and vitamin C, in fetal serum, were all affected by maternal myostatin reduction.
Collapse
Affiliation(s)
- Ruth Opoku
- Division of Biological Sciences, University of Missouri, Columbia, MO 65211, USA; (R.O.); (J.D.)
| | - Jenna DeCata
- Division of Biological Sciences, University of Missouri, Columbia, MO 65211, USA; (R.O.); (J.D.)
| | | | - Laura C. Schulz
- Department of Obstetrics, Gynecology and Women’s Health, University of Missouri, Columbia, MO 65212, USA
| |
Collapse
|
19
|
Pierce M, Di Prinzio P, Dalman C, Abel KM, Morgan VA. Hospital inpatient admissions of children of mothers with severe mental illness: A Western Australian cohort study. Aust N Z J Psychiatry 2023; 57:528-536. [PMID: 35642532 DOI: 10.1177/00048674221100161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND Children of parents with mental illness face a number of adversities, potentially contributing to poor health. AIM The aim of this study was to quantify the association between maternal severe mental illness and children's hospital admissions. METHOD Record linkage cohort study of 467,945 children born in Western Australia between 1 January 1980 and 31 December 2001. Follow-up was from age 28 days until fifth birthday. Linked registers captured information on potential confounders. Rate ratios and adjusted rate ratios measured relative change in the numbers of admissions and total days of stay, while rate differences measured absolute change in outcomes. Cause-specific increases were calculated for ICD-9 chapters and for 'potentially preventable' conditions. RESULTS After adjusting for potential confounders, children of mothers with severe mental illness had a 46% relative increased rate in hospital admissions (95% confidence interval = [38%, 54%]) and an absolute increase in 0.69 extra days in hospital per child, per year (95% confidence interval = [0.67, 0.70]). The relative increase in admissions was greatest in the child's first year of life (adjusted rate ratio = 1.76, 95% confidence interval = [1.64, 1.88]; rate difference = 0.32, 95% confidence interval = [0.30, 0.34]). Rates of admissions were increased for a range of causes, particularly injuries, infections and respiratory disease, and for conditions classified as 'potentially preventable'. CONCLUSION Children of mothers with severe mental illness have a substantial excess in hospital use compared to children of well mothers. This vulnerable group should be targeted with interventions to avert preventable morbidity and premature mortality in later life.
Collapse
Affiliation(s)
- Matthias Pierce
- Centre for Women's Mental Health, Division of Psychology and Mental Health, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Patsy Di Prinzio
- Neuropsychiatric Epidemiology Research Unit, School of Population and Global Health, University of Western Australia, Crawley, WA, Australia
| | - Christina Dalman
- Department of Global Public Health, Karolinska Institutet, Stockholm, Sweden
- Department of Global Public Health, Karolinska Institute, Stockholm, Sweden
| | - Kathryn M Abel
- Centre for Women's Mental Health, Division of Psychology and Mental Health, Faculty of Biology, Medicine and Health, University of Manchester, Greater Manchester Mental Health Trust, Manchester, UK
| | - Vera A Morgan
- Neuropsychiatric Epidemiology Research Unit, School of Population and Global Health, University of Western Australia, Crawley, WA, Australia
| |
Collapse
|
20
|
Thermoregulatory, metabolic and stress responses to spring shearing of aged ewes born to undernourished mothers. J Therm Biol 2023; 113:103503. [PMID: 37055122 DOI: 10.1016/j.jtherbio.2023.103503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 01/20/2023] [Accepted: 02/05/2023] [Indexed: 02/12/2023]
Abstract
Maternal undernutrition during gestation affects the behaviour, metabolism, and sensitivity to stressors of the offspring. Shearing is a stressor that triggers physiological and behavioural changes and augments the thermoregulatory demands in sheep. The aim of this study was to compare the thermoregulatory, metabolic, and behavioural responses to spring shearing of aged ewes born to mothers who grazed different pasture allowances during gestation. Nineteen non-gestating six-year-old Corriedale ewes born to mothers who grazed two pasture allowances from 23 days before conception until 122 days of gestation were used. The pasture allowance offered to the mothers was high [HPA group; n = 11; 10-12 kg of dry matter (DM)/100 kg of body weight (BW)/day] or low [LPA group: n = 8; 5-8 kg of DM/100 kg of BW/day]. The adult offspring of both experimental groups were sheared during spring (Day 0), and remained outdoors, grazing natural grassland, and the behaviour, the surface temperature and the rectal temperature were recorded. Blood concentrations of albumin, total protein, glucose, and insulin were also determined. Data were compared with a mixed model. The LPA ewes had lower ear and nose maximum and minimum surface temperatures before shearing (P < 0.05). On Day 15, the average surface temperature of the vulva was lower in LPA than in HPA ewes (P < 0.05). After shearing, rumination frequency was greater in HPA than in LPA ewes (P = 0.01), and LPA ewes were observed more time standing up than HPA ewes (P < 0.0001). Insulin concentration tended to be greater in LPA than HPA ewes (P = 0.06). Maternal undernutrition during gestation modified the thermoregulatory responses and the acute behavioural changes after shearing in aged female offspring, whilst the metabolism was affected to a lesser degree. The long-term effects noticed in this study highlight the importance of providing proper nutrition to pregnant ewes.
Collapse
|
21
|
Trevisi P, Negrini C, Correa F, Virdis S, Laghi L, Marcello M, Conte G, Mazzoni M, Luise D. Insight into the long-term impact of birth weight on intestinal development, microbial settlement, and the metabolism of weaned piglets. J Anim Sci 2023; 101:skad395. [PMID: 38064718 PMCID: PMC10963063 DOI: 10.1093/jas/skad395] [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: 09/13/2023] [Accepted: 11/28/2023] [Indexed: 12/23/2023] Open
Abstract
Infant mortality of low birth body weight (LBBW) piglets can reach 10% and is mainly due to gut and immune system immaturity which can lead to a higher risk in the long term. This study aimed to assess the impact of birth body weight (BBW) on piglet metabolism, gut status, and microbial profile from weaning to 21 d postweaning. At birth, 32 piglets were selected for their BBW and inserted into the normal BBW (NBBW:1.38 ± 0.09 g) or the LBBW (0.92 ± 0.07 g) group. The piglets were weighed weekly from weaning (d0) to d21. At d9 and d21, 8 piglets/group were slaughtered to obtain the distal jejunum for morphology, immunohistochemistry, and gene expression analysis, colon content for microbiota and short-chain fatty acid (SCFA) analysis, and intestinal content for pH measurement. Blood was collected for metabolomic, haptoglobin (Hp), and reactive oxygen metabolite (ROM) analysis. The LBBW group had a lower body weight (BW) throughout the study (P < 0.01), a lower average daily gain from d9-d21 (P = 0.002), and lower feed intake (P = 0.02). The LBBW piglets had lower Hp at d9 (P = 0.03), higher ROMs at d21 (P = 0.06), and a net alteration of the amino acid (AA) metabolism at d9 and d21. A higher expression of NFKB2 was observed in the LBBW piglets at d9 (P = 0.003) and d21 (P < 0.001). MYD88 expression was enhanced in NBBW piglets at d9 (P < 0.001). The LBBW piglets had a lower villus height, absorptive mucosal surface (P = 0.01), and villus height:crypt depth ratio (P = 0.02), and a greater number of T-lymphocytes in both the epithelium and the crypts (P < 0.001) at d21. At d21, the LBBW piglets had higher lactic acid, acetate, butyrate, and valerate, and also higher SCFA in the colon (P < 0.05). The LBBW piglets had a higher Shannon index (P = 0.01) at d9 and a higher abundance of SCFA-fermenting bacteria. In conclusion, the present study confirmed that LBBW could impact the gut mucosal structure, immunity, and inflammatory and oxidative status, leading to an altered AA metabolism, and delaying the recovery from weaning.
Collapse
Affiliation(s)
- Paolo Trevisi
- Department of Agricultural and Food Sciences, University of Bologna, Bologna, Italy
| | - Clara Negrini
- Department of Agricultural and Food Sciences, University of Bologna, Bologna, Italy
| | - Federico Correa
- Department of Agricultural and Food Sciences, University of Bologna, Bologna, Italy
| | - Sara Virdis
- Department of Agricultural and Food Sciences, University of Bologna, Bologna, Italy
| | - Luca Laghi
- Department of Agricultural and Food Sciences, University of Bologna, Cesena, Italy
| | - Mele Marcello
- Department of Agriculture, Food and Environment, University of Pisa, Pisa, Italy
| | - Giuseppe Conte
- Department of Agriculture, Food and Environment, University of Pisa, Pisa, Italy
| | - Maurizio Mazzoni
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano Emilia, Italy
| | - Diana Luise
- Department of Agricultural and Food Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|
22
|
Demirdjian SP, Meller CH, Berruet MC, Dosdoglirian G, Etchegaray A. Perinatal outcomes of two consecutive strategies for the management of fetal growth restriction: a before-after study. Arch Gynecol Obstet 2023; 307:319-326. [PMID: 35688941 DOI: 10.1007/s00404-022-06641-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 05/18/2022] [Indexed: 02/02/2023]
Abstract
PURPOSE We aim to compare the perinatal outcomes of two consecutive management strategies for fetal growth restriction (FGR), with or without the inclusion of additional Doppler parameters. METHODS A quasi-experimental before/after study was conducted in which we compared a composite perinatal outcome, prematurity rate, and neonatal complications between two management strategies in small fetuses. In the strategy 1 (S1), the management was based on fetal biometry and umbilical artery Doppler. The second strategy (S2) added the assessment of uterine and middle cerebral artery Doppler. We also compared outcomes between strategies according to early (≤ 32 weeks) and late (> 32 weeks) diagnosis subgroups. RESULTS We included 396 patients, 163 in S1 and 233 in S2. There were no significant differences in the perinatal composite outcome (p 0.98), prematurity (p 0.19), or in the subgroup analysis. We found a significant reduction in respiratory distress syndrome (RDS) rate with S2 both globally (OR 0.50, p 0.02), and in the early diagnosis subgroup (OR 0.45, p 0.01). In addition, we observed a significant reduction in the incidence of sepsis with S2 both globally (OR 0.30, p 0.04) and in the early diagnosis subgroup (OR 0.25, p 0.02). We did not observe significant differences in necrotizing enterocolitis (p 0.41) and intraventricular hemorrhage (p 1.00). CONCLUSION The expanded strategy for the management of FGR did not show significant differences in the primary composite outcome or prematurity. However, it was associated with a lower incidence of RDS and neonatal sepsis.
Collapse
Affiliation(s)
- Sabrina Paola Demirdjian
- Fetal Medicine Unit, Department of Obstetrics and Gynecology, Hospital Universitario Austral, Buenos Aires, Argentina.
| | - Cesar Hernan Meller
- Fetal Medicine Unit, Department of Obstetrics and Gynecology, Hospital Italiano de Buenos Aires, Instituto Universitario Hospital Italiano, Buenos Aires, Argentina
| | - Maria Celeste Berruet
- Fetal Medicine Unit, Department of Obstetrics and Gynecology, Hospital Universitario Austral, Buenos Aires, Argentina
| | - Gonzalo Dosdoglirian
- Fetal Medicine Unit, Department of Obstetrics and Gynecology, Hospital Universitario Austral, Buenos Aires, Argentina
| | - Adolfo Etchegaray
- Fetal Medicine Unit, Department of Obstetrics and Gynecology, Hospital Universitario Austral, Buenos Aires, Argentina
| |
Collapse
|
23
|
Balbona JV, Kim Y, Keller MC. The estimation of environmental and genetic parental influences. Dev Psychopathol 2022; 34:1876-1886. [PMID: 36524242 PMCID: PMC10272284 DOI: 10.1017/s0954579422000761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Parents share half of their genes with their children, but they also share background social factors and actively help shape their child's environment - making it difficult to disentangle genetic and environmental causes of parent-offspring similarity. While adoption and extended twin family designs have been extremely useful for distinguishing genetic and nongenetic parental influences, these designs entail stringent assumptions about phenotypic similarity between relatives and require samples that are difficult to collect and therefore are typically small and not publicly shared. Here, we describe these traditional designs, as well as modern approaches that use large, publicly available genome-wide data sets to estimate parental effects. We focus in particular on an approach we recently developed, structural equation modeling (SEM)-polygenic score (PGS), that instantiates the logic of modern PGS-based methods within the flexible SEM framework used in traditional designs. Genetically informative designs such as SEM-PGS rely on different and, in some cases, less rigid assumptions than traditional approaches; thus, they allow researchers to capitalize on new data sources and answer questions that could not previously be investigated. We believe that SEM-PGS and similar approaches can lead to improved insight into how nature and nurture combine to create the incredible diversity underlying human behavior.
Collapse
Affiliation(s)
- Jared V. Balbona
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO 80303, USA
- Department of Psychology & Neuroscience, University of Colorado at Boulder, Boulder, CO 80303, USA
| | - Yongkang Kim
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO 80303, USA
| | - Matthew C. Keller
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO 80303, USA
- Department of Psychology & Neuroscience, University of Colorado at Boulder, Boulder, CO 80303, USA
| |
Collapse
|
24
|
Korhonen P, Tihtonen K, Isojärvi J, Ojala R, Ashorn U, Ashorn P, Tammela O. Calcium supplementation during pregnancy and long‐term offspring outcome: a systematic literature review and meta‐analysis. Ann N Y Acad Sci 2022; 1510:36-51. [PMID: 34978718 PMCID: PMC9304138 DOI: 10.1111/nyas.14729] [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: 08/04/2021] [Revised: 10/04/2021] [Accepted: 11/04/2021] [Indexed: 11/29/2022]
Abstract
The World Health Organization currently recommends calcium supplementation for pregnant women, especially those with low calcium intakes, to reduce the risk of hypertension and preeclampsia. We aimed to evaluate the effect of this intervention on selected offspring outcomes. A systematic search was conducted in 11 databases for published randomized controlled trials (RCTs) on the effect of maternal calcium supplementation with or without vitamin D during pregnancy on selected offspring cardiovascular, growth, and metabolic and neurodevelopmental outcomes. Screening of titles and abstracts of 3555 records and full texts of 31 records yielded six RCTs (nine reports, n = 1616). Forest plot analyses were performed if at least two studies presented comparable data on the same outcome. In one study (n = 591), high‐dose calcium supplementation during pregnancy was associated with a decreased risk of offspring high systolic blood pressure at 5–7 years of age (risk ratio = 0.59; 95% confidence interval: 0.39–0.90). The effects of the intervention on offspring growth, metabolic, and neurodevelopmental outcomes remain unknown because of conflicting or insufficient data. High risk of attrition bias decreased the quality of the evidence. Limited available data from RCTs do not provide sufficient evidence to conclude that prenatal calcium supplementation influences offspring health outcomes beyond the newborn period.
Collapse
Affiliation(s)
- Päivi Korhonen
- Department of Pediatrics Tampere University Hospital Tampere Finland
- Faculty of Medicine and Health Technology, Center for Child, Adolescent, and Maternal Health Research Tampere University Tampere Finland
| | - Kati Tihtonen
- Faculty of Medicine and Health Technology, Center for Child, Adolescent, and Maternal Health Research Tampere University Tampere Finland
- Department of Obstetrics and Gynecology Tampere University Hospital Tampere Finland
| | | | - Riitta Ojala
- Department of Pediatrics Tampere University Hospital Tampere Finland
- Faculty of Medicine and Health Technology, Center for Child, Adolescent, and Maternal Health Research Tampere University Tampere Finland
| | - Ulla Ashorn
- Faculty of Medicine and Health Technology, Center for Child, Adolescent, and Maternal Health Research Tampere University Tampere Finland
| | - Per Ashorn
- Department of Pediatrics Tampere University Hospital Tampere Finland
- Faculty of Medicine and Health Technology, Center for Child, Adolescent, and Maternal Health Research Tampere University Tampere Finland
| | - Outi Tammela
- Department of Pediatrics Tampere University Hospital Tampere Finland
- Faculty of Medicine and Health Technology, Center for Child, Adolescent, and Maternal Health Research Tampere University Tampere Finland
| |
Collapse
|
25
|
Stevenson NJ, Lai MM, Starkman HE, Colditz PB, Wixey JA. Electroencephalographic studies in growth-restricted and small-for-gestational-age neonates. Pediatr Res 2022; 92:1527-1534. [PMID: 35197567 PMCID: PMC9771813 DOI: 10.1038/s41390-022-01992-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 01/25/2022] [Accepted: 01/31/2022] [Indexed: 12/30/2022]
Abstract
Foetal growth restriction (FGR) and being born small for gestational age (SGA) are associated with neurodevelopmental delay. Early diagnosis of neurological damage is difficult in FGR and SGA neonates. Electroencephalography (EEG) has the potential as a tool for the assessment of brain development in FGR/SGA neonates. In this review, we analyse the evidence base on the use of EEG for the assessment of neonates with FGR or SGA. We found consistent findings that FGR/SGA is associated with measurable changes in the EEG that present immediately after birth and persist into childhood. Early manifestations of FGR/SGA in the EEG include changes in spectral power, symmetry/synchrony, sleep-wake cycling, and the continuity of EEG amplitude. Later manifestations of FGR/SGA into infancy and early childhood include changes in spectral power, sleep architecture, and EEG amplitude. FGR/SGA infants had poorer neurodevelopmental outcomes than appropriate for gestational age controls. The EEG has the potential to identify FGR/SGA infants and assess the functional correlates of neurological damage. IMPACT: FGR/SGA neonates have significantly different EEG activity compared to AGA neonates. EEG differences persist into childhood and are associated with adverse neurodevelopmental outcomes. EEG has the potential for early identification of brain impairment in FGR/SGA neonates.
Collapse
Affiliation(s)
- Nathan J. Stevenson
- grid.1049.c0000 0001 2294 1395Brain Modelling Group, QIMR Berghofer Medical Research Institute, Brisbane, QLD Australia
| | - Melissa M. Lai
- grid.1003.20000 0000 9320 7537UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Herston, QLD 4029 Australia ,grid.416100.20000 0001 0688 4634Perinatal Research Centre, Royal Brisbane and Women’s Hospital, Herston, QLD 4029 Australia
| | - Hava E. Starkman
- grid.1003.20000 0000 9320 7537UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Herston, QLD 4029 Australia ,grid.17063.330000 0001 2157 2938Department of Obstetrics and Gynaecology, University of Toronto, King’s College Circle, Toronto, ON M5S Canada
| | - Paul B. Colditz
- grid.1003.20000 0000 9320 7537UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Herston, QLD 4029 Australia ,grid.416100.20000 0001 0688 4634Perinatal Research Centre, Royal Brisbane and Women’s Hospital, Herston, QLD 4029 Australia
| | - Julie A. Wixey
- grid.1003.20000 0000 9320 7537UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Herston, QLD 4029 Australia
| |
Collapse
|
26
|
Mutamba AK, He X, Wang T. Therapeutic advances in overcoming intrauterine growth restriction induced metabolic syndrome. Front Pediatr 2022; 10:1040742. [PMID: 36714657 PMCID: PMC9875160 DOI: 10.3389/fped.2022.1040742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 12/21/2022] [Indexed: 01/13/2023] Open
Abstract
Intrauterine growth restriction (IUGR) remains a great public health challenge as it affects neonatal survival and influences their normal biological development and metabolism. Several clinical researches have revealed the occurrence of metabolic syndrome, such as insulin resistance, obesity, type 2 diabetes mellitus, oxidative stress, dyslipidemia, as direct results of IUGR. Therefore, it is essential to understand its underlying mechanism, impact and develop effective therapies. The purpose of this work is to review the current knowledge on IUGR induced metabolic syndrome and relevant therapies. Here in, we elaborate on the characteristics and causes of IUGR by pointing out recent research findings. Furthermore, we discuss the impact of IUGR on different organs of the body, followed by preclinical studies on IUGR using suitable animal models. Additionally, various metabolic disorders with their genetic implications, such as insulin resistance, type 2 diabetes mellitus, dyslipidemia, obesity are detailed. Finally, the current therapeutic options used in the treatment of IUGR are summarized with some prospective therapies highlighted.
Collapse
Affiliation(s)
- Alpha Kalonda Mutamba
- Department of Pediatrics, Neonatology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xiaori He
- Department of Pediatrics, Neonatology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Tao Wang
- Laboratory of Neonatal Disease, Institute of Pediatrics, Central South University, Changsha, China
| |
Collapse
|
27
|
Dalle Molle R, de Mendonça Filho EJ, Minuzzi L, Machado TD, Reis RS, Rodrigues DM, Mucellini AB, Franco AR, Buchweitz A, Toazza R, Bortoluzzi A, Salum GA, Boscenco S, Meaney MJ, Levitan RD, Manfro GG, Silveira PP. Thrifty-Eating Behavior Phenotype at the Food Court - Programming Goes Beyond Food Preferences. Front Endocrinol (Lausanne) 2022; 13:882532. [PMID: 35677721 PMCID: PMC9168906 DOI: 10.3389/fendo.2022.882532] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 04/14/2022] [Indexed: 11/18/2022] Open
Abstract
INTRODUCTION Prenatal growth impairment leads to higher preference for palatable foods in comparison to normal prenatal growth subjects, which can contribute to increased body fat mass and a higher risk for developing chronic diseases in small-for-gestational-age (SGA) individuals throughout life. This study aimed to investigate the effect of SGA on feeding behavior in children and adolescents, as well as resting-state connectivity between areas related to reward, self-control, and value determination, such as orbitofrontal cortex (OFC), dorsolateral prefrontal cortex (DL-PFC), amygdala and dorsal striatum (DS). METHODS Caregivers and their offspring were recruited from two independent cohorts in Brazil (PROTAIA) and Canada (MAVAN). Both cohorts included anthropometric measurements, food choice tasks, and resting-state functional magnetic resonance imaging (fMRI) data. RESULTS In the Brazilian sample (17 ± 0.28 years, n=70), 21.4% of adolescents were classified as SGA. They exhibited lower monetary-related expenditure to buy a snack compared to controls in the food choice test. Decreased functional connectivity (n=40) between left OFC and left DL-PFC; and between right OFC and: left amygdala, right DS, and left DS were observed in the Brazilian SGA participants. Canadian SGA participants (14.9%) had non-significant differences in comparison with controls in a food choice task at 4 years old ( ± 0.01, n=315). At a follow-up brain scan visit (10.21 ± 0.140 years, n=49), SGA participants (28.6%) exhibited higher connectivity between the left OFC and left DL-PFC, also higher connectivity between the left OFC and right DL-PFC. We did not observe significant anthropometric neither nutrients' intake differences between groups in both samples. CONCLUSIONS Resting-state fMRI results showed that SGA individuals had altered connectivity between areas involved in encoding the subjective value for available goods and decision-making in both samples, which can pose them in disadvantage when facing food options daily. Over the years, the cumulative exposure to particular food cues together with the altered behavior towards food, such as food purchasing, as seen in the adolescent cohort, can play a role in the long-term risk for developing chronic non-communicable diseases.
Collapse
Affiliation(s)
- Roberta Dalle Molle
- Programa de Pós-Graduação em Ciências da Nutrição, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
- Ludmer Centre for Neuroinformatics and Mental Health, Douglas Hospital Research Center, Montreal, QC, Canada
- Programa de Pós-Graduação em Saúde da Criança e do Adolescente, Faculdade de Medicina, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Euclides José de Mendonça Filho
- Ludmer Centre for Neuroinformatics and Mental Health, Douglas Hospital Research Center, Montreal, QC, Canada
- Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Luciano Minuzzi
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, ON, Canada
| | - Tania Diniz Machado
- Programa de Pós-Graduação em Saúde da Criança e do Adolescente, Faculdade de Medicina, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Roberta Sena Reis
- Programa de Pós-Graduação em Saúde da Criança e do Adolescente, Faculdade de Medicina, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Faculdade de Nutrição, Universidade Federal de Goiás, Goiânia, Brazil
| | - Danitsa Marcos Rodrigues
- Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Amanda Brondani Mucellini
- Programa de Pós-Graduação em Ciências Médicas: Psiquiatria, Faculdade de Medicina, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Alexandre Rosa Franco
- Instituto do Cérebro (InsCer), Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
- Faculdade de Medicina, Programa de Pós-Graduação em Medicina e Ciências da Saúde, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
- Faculdade de Engenharia, Programa de Pós-Graduação em Engenharia Elétrica, PUCRS, Porto Alegre, Brazil
| | - Augusto Buchweitz
- Instituto do Cérebro (InsCer), Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
- Faculdade de Medicina, Programa de Pós-Graduação em Medicina e Ciências da Saúde, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
- Faculdade de Letras, Programa de Pós-Graduação em Letras, Linguística, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Rudineia Toazza
- Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Andressa Bortoluzzi
- Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Giovanni Abrahão Salum
- Programa de Pós-Graduação em Ciências Médicas: Psiquiatria, Faculdade de Medicina, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Sonia Boscenco
- Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Michael J. Meaney
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | - Robert D. Levitan
- Department of Psychiatry, University of Toronto and Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Gisele Gus Manfro
- Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Programa de Pós-Graduação em Ciências Médicas: Psiquiatria, Faculdade de Medicina, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Patricia Pelufo Silveira
- Ludmer Centre for Neuroinformatics and Mental Health, Douglas Hospital Research Center, Montreal, QC, Canada
- Department of Psychiatry, McGill University, Montreal, QC, Canada
- *Correspondence: Patricia Pelufo Silveira,
| |
Collapse
|
28
|
Zhang H, Zheng P, Chen D, Yu B, He J, Mao X, Yu J, Luo Y, Luo J, Huang Z, Yan H. Dietary Arginine Supplementation Improves Intestinal Mitochondrial Functions in Low-Birth-Weight Piglets but Not in Normal-Birth-Weight Piglets. Antioxidants (Basel) 2021; 10:antiox10121995. [PMID: 34943098 PMCID: PMC8698761 DOI: 10.3390/antiox10121995] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/09/2021] [Accepted: 12/10/2021] [Indexed: 12/16/2022] Open
Abstract
Our previous studies revealed that L-arginine supplementation had beneficial effects on intestinal barrier functions of low-birth-weight (LBW) piglets, which were associated with the enhanced antioxidant capacity. Moreover, mitochondrial functions are closely related to the redox state. This study was to explore potential mechanisms of L-arginine-induced beneficial effects against intestinal dysfunction by regulating mitochondrial function of LBW piglets. Twenty 4-day-old normal birth weight (NBW) piglets (BW: 2.08 ± 0.09 kg) and 20 LBW siblings (BW: 1.16 ± 0.07 kg) were artificially fed either a basal diet or a basal diet supplemented with 1.0% L-arginine for 21 d, respectively. Growth performance, intestinal morphology, redox status, mitochondrial morphology, and mitochondrial functions were examined. Data were subjected to two-way analysis of variance. LBW piglets presented lower (p < 0.05) ADG, shorter (p < 0.05) intestinal villus height, lower (p < 0.05) jejunal adenosine triphosphate (ATP) content and higher (p < 0.05) concentrations of Ca2+ and 8-OH-dG in jejunal mitochondria, compared with NBW piglets. Supplementation with 1.0% L-arginine significantly increased (p < 0.05) ADG, the activities of CAT, SOD, and GPx, intestinal villus height and mRNA abundances of ZO-1 (2-fold) in the jejunum of LBW piglets, but not in NBW piglets. Furthermore, the concentrations of ATP and the transcription of COX IV, COX V genes were up-regulated (p < 0.05) and the concentration of Ca2+ and 8-OH-dG were decreased (p < 0.05) in arginine-treated LBW piglets. The results suggest that mitochondrial morphology is affected, and mitochondrial functions are impaired in the jejunum of LBW piglets. While supplementation with 1.0% L-arginine relieved intestinal dysfunction through enhancing antioxidant capacity and improving mitochondrial functions via repairing mitochondrial morphology, normalizing mitochondrial calcium, and increasing ATP concentration in the jejunum of LBW piglets. However, supplementation with L-arginine has no significant beneficial effects on intestinal health in NBW piglets.
Collapse
Affiliation(s)
- Hao Zhang
- Animal Nutrition Insititute, Sichuan Agricultural University, Chengdu 611130, China; (H.Z.); (D.C.); (B.Y.); (J.H.); (X.M.); (J.Y.); (Y.L.); (J.L.); (Z.H.); (H.Y.)
| | - Ping Zheng
- Animal Nutrition Insititute, Sichuan Agricultural University, Chengdu 611130, China; (H.Z.); (D.C.); (B.Y.); (J.H.); (X.M.); (J.Y.); (Y.L.); (J.L.); (Z.H.); (H.Y.)
- Key Laboratory for Animal Disease-Resistance Nutrition, China Ministry of Education, Chengdu 611130, China
- Correspondence: ; Tel.: +86-028-86290922
| | - Daiwen Chen
- Animal Nutrition Insititute, Sichuan Agricultural University, Chengdu 611130, China; (H.Z.); (D.C.); (B.Y.); (J.H.); (X.M.); (J.Y.); (Y.L.); (J.L.); (Z.H.); (H.Y.)
- Key Laboratory for Animal Disease-Resistance Nutrition, China Ministry of Education, Chengdu 611130, China
| | - Bing Yu
- Animal Nutrition Insititute, Sichuan Agricultural University, Chengdu 611130, China; (H.Z.); (D.C.); (B.Y.); (J.H.); (X.M.); (J.Y.); (Y.L.); (J.L.); (Z.H.); (H.Y.)
- Key Laboratory for Animal Disease-Resistance Nutrition, China Ministry of Education, Chengdu 611130, China
| | - Jun He
- Animal Nutrition Insititute, Sichuan Agricultural University, Chengdu 611130, China; (H.Z.); (D.C.); (B.Y.); (J.H.); (X.M.); (J.Y.); (Y.L.); (J.L.); (Z.H.); (H.Y.)
- Key Laboratory for Animal Disease-Resistance Nutrition, China Ministry of Education, Chengdu 611130, China
| | - Xiangbing Mao
- Animal Nutrition Insititute, Sichuan Agricultural University, Chengdu 611130, China; (H.Z.); (D.C.); (B.Y.); (J.H.); (X.M.); (J.Y.); (Y.L.); (J.L.); (Z.H.); (H.Y.)
- Key Laboratory for Animal Disease-Resistance Nutrition, China Ministry of Education, Chengdu 611130, China
| | - Jie Yu
- Animal Nutrition Insititute, Sichuan Agricultural University, Chengdu 611130, China; (H.Z.); (D.C.); (B.Y.); (J.H.); (X.M.); (J.Y.); (Y.L.); (J.L.); (Z.H.); (H.Y.)
- Key Laboratory for Animal Disease-Resistance Nutrition, China Ministry of Education, Chengdu 611130, China
| | - Yuheng Luo
- Animal Nutrition Insititute, Sichuan Agricultural University, Chengdu 611130, China; (H.Z.); (D.C.); (B.Y.); (J.H.); (X.M.); (J.Y.); (Y.L.); (J.L.); (Z.H.); (H.Y.)
- Key Laboratory for Animal Disease-Resistance Nutrition, China Ministry of Education, Chengdu 611130, China
| | - Junqiu Luo
- Animal Nutrition Insititute, Sichuan Agricultural University, Chengdu 611130, China; (H.Z.); (D.C.); (B.Y.); (J.H.); (X.M.); (J.Y.); (Y.L.); (J.L.); (Z.H.); (H.Y.)
- Key Laboratory for Animal Disease-Resistance Nutrition, China Ministry of Education, Chengdu 611130, China
| | - Zhiqing Huang
- Animal Nutrition Insititute, Sichuan Agricultural University, Chengdu 611130, China; (H.Z.); (D.C.); (B.Y.); (J.H.); (X.M.); (J.Y.); (Y.L.); (J.L.); (Z.H.); (H.Y.)
| | - Hui Yan
- Animal Nutrition Insititute, Sichuan Agricultural University, Chengdu 611130, China; (H.Z.); (D.C.); (B.Y.); (J.H.); (X.M.); (J.Y.); (Y.L.); (J.L.); (Z.H.); (H.Y.)
| |
Collapse
|
29
|
Wu T, Gong X, Zhao Y, Zhang L, You Y, Wei H, Zuo X, Zhou Y, Xing X, Meng Z, Lv Q, Liu Z, Zhang J, Hu L, Li J, Li L, Chen C, Liu C, Sun G, Liu A, Chen J, Lv Y, Wang X, Wei Y. Fetal growth velocity references from a Chinese population-based fetal growth study. BMC Pregnancy Childbirth 2021; 21:688. [PMID: 34627184 PMCID: PMC8501691 DOI: 10.1186/s12884-021-04149-x] [Citation(s) in RCA: 4] [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: 05/10/2021] [Accepted: 09/15/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Fetal growth velocity standards have yet to be established for the Chinese population. This study aimed to establish such standards suitable for the Chinese population. METHODS We performed a multicenter, population-based longitudinal cohort study including 9075 low-risk singleton pregnant women. Data were collected from the clinical records of 24 hospitals in 18 provinces of China. Demographic characteristics, reproductive history, fetal ultrasound measurements, and perinatal outcome data were collected. The fetal ultrasound measurements included biparietal diameter (BPD), abdominal circumference (AC), head circumference (HC), and femur diaphysis length (FDL). We used linear mixed models with cubic splines to model the trajectory of four ultrasound parameters and estimate fetal weight. Fetal growth velocity was determined by calculating the first derivative of fetal size curves. We also used logistic regression to estimate the association between fetal growth velocities in the bottom 10th percentile and adverse perinatal outcomes. RESULTS Fetal growth velocity was not consistent over time or among individuals. The estimated fetal weight (EFW) steadily increased beginning at 12 gestational weeks and peaked at 35 gestational weeks. The maximum velocity was 211.71 g/week, and there was a steady decrease in velocity from 35 to 40 gestational weeks. The four ultrasound measurements increased in the early second trimester; BPD and HC peaked at 13 gestational weeks, AC at 14 gestational weeks, and FDL at 15 gestational weeks. BPD and HC also increased from 19 to 24 and 19 to 21 gestational weeks, respectively. EFW velocity in the bottom 10th percentile indicated higher risks of neonatal complications (odds ratio [OR] = 2.23, 95% confidence interval [CI]: 1.79-2.78) and preterm birth < 37 weeks (OR = 3.68, 95% CI: 2.64-5.14). Sensitivity analyses showed that EFW velocity in the bottom 10th percentile was significantly associated with more adverse pregnancy outcomes for appropriate-for-gestational age neonates. CONCLUSIONS We established fetal growth velocity curves for the Chinese population based on real-world clinical data. Our findings demonstrated that Chinese fetal growth patterns are somewhat different from those of other populations. Fetal growth velocity could provide more information to understand the risk of adverse perinatal outcomes, especially for appropriate-for-gestational age neonates.
Collapse
Affiliation(s)
- Tianchen Wu
- Department of Maternal and Child Health, School of Public Health, Peking University, Beijing, 100191, China
| | - Xiaoli Gong
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China
| | - Yangyu Zhao
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China
| | - Lizhen Zhang
- Department of Obstetrics and Gynecology, Maternal and Child Health Hospital of Qinhuangdao, Qinhuangdao, 066000, China
| | - Yiping You
- Department of Obstetrics, Maternal and Child Health Hospital of Hunan, Changsha, 410007, China
| | - Hongwei Wei
- Department of Obstetrics, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530000, China
| | - Xifang Zuo
- Department of Obstetrics, Tongzhou Maternal and Child Health Hospital of Beijing, Beijing, 101100, China
| | - Ying Zhou
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Xiamen University, Xiamen, 361003, China
| | - Xinli Xing
- Maternal and Child Health Hospital of Dongchangfu District, Liaocheng, 252004, China
| | - Zhaoyan Meng
- Department of Obstetrics, Gansu Maternal and Child Health Hospital, Lanzhou, 730050, China
| | - Qi Lv
- Department of Obstetrics and Gynecology, Changchun Obstetrics-Gynecology Hospital, Changchun, 130042, China
| | - Zhaodong Liu
- Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fu Zhou, 350122, China
| | - Jian Zhang
- Department of Function, Maternal and Child Health Hospital of Shijiazhuang, Shijiazhuang, 050051, China
| | - Liyan Hu
- Children's Hospital of Shanxi, Women Health Center of Shanxi, Taiyuan, 030013, China
| | - Junnan Li
- Department of Obstetrics, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Li Li
- Department of Obstetrics, Zhengzhou Central Hospital, Zhengzhou, 450007, China
| | - Chulin Chen
- Department of Obstetrics, Maternal and Child Health Hospital of Changzhi, Changzhi, 046011, China
| | - Chunyan Liu
- Department of Obstetrics, Maternity and Infant Hospital of Shenyang, Shenyang, 110014, China
| | - Guoqiang Sun
- Department of Obstetrics, Hubei Maternal and Child Health Hospital, Wuhan, 430070, China
| | - Aiju Liu
- Department of Obstetrics, Inner Mongolia Maternal and Child Health Hospital, Hohhot, 010060, China
| | - Jingsi Chen
- Department of Prenatal Diagnosis, the Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Yuan Lv
- Shengjing Hospital Affiliated to China Medical University, Shenyang, 117004, China
| | - Xiaoli Wang
- Department of Maternal and Child Health, School of Public Health, Peking University, Beijing, 100191, China.
| | - Yuan Wei
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China.
| |
Collapse
|
30
|
Tsantekidou I, Evangelinakis N, Bargiota A, Vrachnis N, Kalantaridou S, Valsamakis G. Macrosomia and fetal growth restriction: evidence for similar extrauterine metabolic risks but with differences in pathophysiology. J Matern Fetal Neonatal Med 2021; 35:8450-8455. [PMID: 34555989 DOI: 10.1080/14767058.2021.1980531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
PURPOSE To investigate and compare the causes of macrosomia and FGR fetuses, their ultrasound characteristics and the importance of their similar metabolic profile in intrauterine and extrauterine life. MATERIALS AND METHODS We searched Pubmed/Google Scholar database up until 15 December 2020 using keywords. Out of the 70 matching results we selected 50 most representative and matching papers. RESULTS We found similar causes and metabolic profiles and in both conditions offspring are at increased risk of developing metabolic and cardiovascular diseases in the extrauterine life. CONCLUSION Despite similarities of the maternal factors and fetal metabolic profile it is still unknown which of them has worse metabolic status during intrauterine and extrauterine life.
Collapse
Affiliation(s)
- Inga Tsantekidou
- School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Nikolaos Evangelinakis
- Reproductive Endocrinology Unit, 3rd Department of Obstetrics and Gynecology, University General Hospital "Attikon", Medical School, National and Kapodistrian University of Athens Greece, Athens, Greece
| | - Alexandra Bargiota
- Department of Endocrinology and Metabolic Disorders, University Hospital of Larissa, Medical School of Larissa, University of Thessaly, Larissa, Greece
| | - Nikolaos Vrachnis
- Reproductive Endocrinology Unit, 3rd Department of Obstetrics and Gynecology, University General Hospital "Attikon", Medical School, National and Kapodistrian University of Athens Greece, Athens, Greece
| | - Sophia Kalantaridou
- Reproductive Endocrinology Unit, 3rd Department of Obstetrics and Gynecology, University General Hospital "Attikon", Medical School, National and Kapodistrian University of Athens Greece, Athens, Greece
| | - Georgios Valsamakis
- Department of Endocrinology and Metabolic Disorders, University Hospital of Larissa, Medical School of Larissa, University of Thessaly, Larissa, Greece.,2nd Department of Obstetrics and Gynecology, University Hospital "Aretaieion", Medical School, National and Kapodistrian University of Athens Greece, Athens, Greece
| |
Collapse
|
31
|
Areola ED, Adewuyi IJ, Usman TO, Tamunoibuomi G, Arogundade LK, Olaoye B, Matt-Ojo DD, Jeje AO, Oyabambi AO, Afolayan EA, Olatunji LA. Sildenafil augments fetal weight and placental adiponectin in gestational testosterone-induced glucose intolerant rats. Toxicol Rep 2021; 8:1358-1368. [PMID: 34277360 PMCID: PMC8271103 DOI: 10.1016/j.toxrep.2021.06.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 06/04/2021] [Accepted: 06/14/2021] [Indexed: 02/07/2023] Open
Abstract
Testosterone induces intra-uterine growth restriction (IUGR) with maternal glucose dysregulation and oxidant release in various tissues. Adiponectin, which modulates the antioxidant nuclear factor erythroid 2-related factor 2 (Nrf2) signaling is expressed in the placenta and affects fetal growth. Sildenafil, a phosphodiesterase type 5 inhibitor (PDE5i), used mainly in erectile dysfunction has been widely studied as a plausible pharmacologic candidate in IUGR. Therefore, the present study sought to determine the effect of PDE5i on placental adiponectin/Nrf2 pathway in gestational testosterone-induced impaired glucose tolerance and fetal growth. Fifteen pregnant Wistar rats were allotted into three groups (n = 5/group) receiving vehicles (Ctr; distilled water and olive oil), testosterone propionate (Tes; 3.0 mg/kg; sc) or combination of testosterone propionate (3.0 mg/kg; sc) and sildenafil (50.0 mg/kg; po) from gestational day 14-19. On gestational day 20, plasma and placenta homogenates were obtained for biochemical analysis as well as fetal biometry. Pregnant rats exposed to testosterone had 4-fold increase in circulating testosterone compared with control (20.9 ± 2.8 vs 5.1 ± 1.7 ng/mL; p < 0.05) whereas placenta testosterone levels were similar in testosterone- and vehicle-treated rats. Exposure to gestational testosterone caused reduction in fetal and placental weights, placental Nrf2 and adiponectin. Moreover, impaired glucose tolerance, elevated plasma triglyceride-glucose (TyG) index, placental triglyceride, total cholesterol, lactate, malondialdehyde and alanine aminotransferase were observed in testosterone-exposed rats. Treatment with sildenafil improved glucose tolerance, plasma TyG index, fetal and placental weights and reversed placental adiponectin in testosterone-exposed pregnant rats without any effect on placental Nrf2. Therefore, in testosterone-exposed rats, sildenafil improves impaired glucose tolerance, poor fetal outcome which is accompanied by augmented placental adiponectin regardless of depressed Nrf2.
Collapse
Affiliation(s)
- Emmanuel Damilare Areola
- HOPE Cardiometabolic Research Team and Department of Physiology, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Ifeoluwa Jesufemi Adewuyi
- HOPE Cardiometabolic Research Team and Department of Physiology, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Taofeek Olumayowa Usman
- HOPE Cardiometabolic Research Team and Department of Physiology, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
- Cardiovascular Unit, Department of Physiology, College of Health Sciences, Osun State University, Osogbo, Nigeria
| | - God’sgift Tamunoibuomi
- HOPE Cardiometabolic Research Team and Department of Physiology, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Lucy Kemi Arogundade
- HOPE Cardiometabolic Research Team and Department of Physiology, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Barakat Olaoye
- HOPE Cardiometabolic Research Team and Department of Physiology, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Deborah Damilayo Matt-Ojo
- HOPE Cardiometabolic Research Team and Department of Physiology, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Abdulrazaq Olatunji Jeje
- HOPE Cardiometabolic Research Team and Department of Physiology, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Adewumi Oluwafemi Oyabambi
- HOPE Cardiometabolic Research Team and Department of Physiology, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Enoch Abiodun Afolayan
- Department of Pathology, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Lawrence Aderemi Olatunji
- HOPE Cardiometabolic Research Team and Department of Physiology, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
- Corresponding author at: Department of Physiology, University of Ilorin, P.M.B. 1515, Ilorin, 240003, Nigeria.
| |
Collapse
|
32
|
Pendleton AL, Wesolowski SR, Regnault TRH, Lynch RM, Limesand SW. Dimming the Powerhouse: Mitochondrial Dysfunction in the Liver and Skeletal Muscle of Intrauterine Growth Restricted Fetuses. Front Endocrinol (Lausanne) 2021; 12:612888. [PMID: 34079518 PMCID: PMC8165279 DOI: 10.3389/fendo.2021.612888] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 04/22/2021] [Indexed: 11/14/2022] Open
Abstract
Intrauterine growth restriction (IUGR) of the fetus, resulting from placental insufficiency (PI), is characterized by low fetal oxygen and nutrient concentrations that stunt growth rates of metabolic organs. Numerous animal models of IUGR recapitulate pathophysiological conditions found in human fetuses with IUGR. These models provide insight into metabolic dysfunction in skeletal muscle and liver. For example, cellular energy production and metabolic rate are decreased in the skeletal muscle and liver of IUGR fetuses. These metabolic adaptations demonstrate that fundamental processes in mitochondria, such as substrate utilization and oxidative phosphorylation, are tempered in response to low oxygen and nutrient availability. As a central metabolic organelle, mitochondria coordinate cellular metabolism by coupling oxygen consumption to substrate utilization in concert with tissue energy demand and accretion. In IUGR fetuses, reducing mitochondrial metabolic capacity in response to nutrient restriction is advantageous to ensure fetal survival. If permanent, however, these adaptations may predispose IUGR fetuses toward metabolic diseases throughout life. Furthermore, these mitochondrial defects may underscore developmental programming that results in the sequela of metabolic pathologies. In this review, we examine how reduced nutrient availability in IUGR fetuses impacts skeletal muscle and liver substrate catabolism, and discuss how enzymatic processes governing mitochondrial function, such as the tricarboxylic acid cycle and electron transport chain, are regulated. Understanding how deficiencies in oxygen and substrate metabolism in response to placental restriction regulate skeletal muscle and liver metabolism is essential given the importance of these tissues in the development of later lifer metabolic dysfunction.
Collapse
Affiliation(s)
- Alexander L. Pendleton
- School of Animal and Comparative Biomedical Sciences, The University of Arizona, Tucson, AZ, United States
| | - Stephanie R. Wesolowski
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, United States
| | | | - Ronald M. Lynch
- School of Animal and Comparative Biomedical Sciences, The University of Arizona, Tucson, AZ, United States
| | - Sean W. Limesand
- School of Animal and Comparative Biomedical Sciences, The University of Arizona, Tucson, AZ, United States
| |
Collapse
|
33
|
Rouhani S, Griffin NW, Yori PP, Gehrig JL, Olortegui MP, Salas MS, Trigoso DR, Moulton LH, Houpt ER, Barratt MJ, Kosek MN, Gordon JI. Diarrhea as a Potential Cause and Consequence of Reduced Gut Microbial Diversity Among Undernourished Children in Peru. Clin Infect Dis 2021; 71:989-999. [PMID: 31773127 PMCID: PMC7053391 DOI: 10.1093/cid/ciz905] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Accepted: 09/13/2019] [Indexed: 12/18/2022] Open
Abstract
Background Detrimental effects of diarrhea on child growth and survival are well documented, but details of the underlying mechanisms remain poorly understood. Recent evidence demonstrates that perturbations to normal development of the gut microbiota in early life may contribute to growth faltering and susceptibility to related childhood diseases. We assessed associations between diarrhea, gut microbiota configuration, and childhood growth in the Peruvian Amazon. Methods Growth, diarrhea incidence, illness, pathogen infection, and antibiotic exposure were assessed monthly in a birth cohort of 271 children aged 0–24 months. Gut bacterial diversity and abundances of specific bacterial taxa were quantified by sequencing 16S rRNA genes in fecal samples collected at 6, 12, 18, and 24 months. Linear and generalized linear models were used to determine whether diarrhea was associated with altered microbiota and, in turn, if features of the microbiota were associated with the subsequent risk of diarrhea. Results Diarrheal frequency, duration, and severity were negatively associated with bacterial diversity and richness (P < .05). Children born stunted (length-for-age z-score [LAZ] ≤ −2) who were also severely stunted (LAZ ≤ −3) at the time of sampling exhibited the greatest degree of diarrhea-associated reductions in bacterial diversity and the slowest recovery of bacterial diversity after episodes of diarrhea. Increased bacterial diversity was predictive of reduced subsequent diarrhea from age 6 to 18 months. Conclusions Persistent, severe growth faltering may reduce the gut microbiota's resistance and resilience to diarrhea, leading to greater losses of diversity and longer recovery times. This phenotype, in turn, denotes an increased risk of future diarrheal disease and growth faltering.
Collapse
Affiliation(s)
- Saba Rouhani
- Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Nicholas W Griffin
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, Washington.,Center for Gut Microbiome and Nutrition Research, St. Louis, Missouri
| | - Pablo Peñataro Yori
- Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland.,Asociación Benéfica Preferred Reporting Items for Systematic Reviews and Meta-analyses, Iquitos, Peru
| | - Jeanette L Gehrig
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, Washington.,Center for Gut Microbiome and Nutrition Research, St. Louis, Missouri
| | - Maribel Paredes Olortegui
- Asociación Benéfica Preferred Reporting Items for Systematic Reviews and Meta-analyses, Iquitos, Peru
| | - Mery Siguas Salas
- Asociación Benéfica Preferred Reporting Items for Systematic Reviews and Meta-analyses, Iquitos, Peru
| | - Dixner Rengifo Trigoso
- Asociación Benéfica Preferred Reporting Items for Systematic Reviews and Meta-analyses, Iquitos, Peru
| | | | - Eric R Houpt
- University of Virginia, Charlottesville, Virginia
| | - Michael J Barratt
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, Washington.,Center for Gut Microbiome and Nutrition Research, St. Louis, Missouri
| | - Margaret N Kosek
- Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland.,University of Virginia, Charlottesville, Virginia
| | - Jeffrey I Gordon
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, Washington.,Center for Gut Microbiome and Nutrition Research, St. Louis, Missouri
| |
Collapse
|
34
|
Cerón NA, Gutiérrez OO, Cerón OM, Ortiz RA. Complicaciones cardiovasculares en relación con la programación fetal. REPERTORIO DE MEDICINA Y CIRUGÍA 2021. [DOI: 10.31260/repertmedcir.01217273.943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Introducción: la programación fetal ofrece nuevas perspectivas sobre el origen de las enfermedades cardiovasculares, relacionando su aparición con factores perinatales. Objetivo: exponer evidencia que vincule las alteraciones gestacionales con las enfermedades cardiovasculares en la vida adulta del feto. Metodología: búsqueda en las bases de datos EBSCO, COCHRANE, MEDLINE, PROQUEST y SciELO de los artículos de revisión e investigaciones originales en inglés publicados en los últimos diez años. Se utilizaron términos MeSH para búsqueda controlada y se evaluaron los estudios con STROBE y PRISMA según correspondía. Resultados: los hallazgos sugieren que nacer con menos de 2600 k guarda relación con diabetes mellitus (OR de 1.607 IC 95% 1.324-1.951), hipertensión arterial (OR de 1.15 IC 95% 1.043-1.288) y menor función endotelial (1.94+0.37 vs 2.68+0.41, p: 0.0001) en la adultez. La prematuridad se asocia con mayores presiones arteriales sistólicas (4.2 mmHg IC 95%; 2.8 - 5.7 p 0.001) y diastólicas (2.6 mmHg IC 95%; 1.2-4.0; p 0.001). Las alteraciones nutricionales maternas y la diabetes gestacional aumentan el riesgo de síndrome metabólico (OR 1.2 IC 95% 0.9-1.7) y sobrepeso en la edad escolar (OR 1.81 IC 95% 1.18 - 2.86). Conclusión: los resultados adversos en la gestación están relacionados con el desarrollo de enfermedades cardiovasculares en la vida adulta del feto expuesto.
Collapse
|
35
|
Ding S, Yan W, Ma Y, Fang J. The impact of probiotics on gut health via alternation of immune status of monogastric animals. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2021; 7:24-30. [PMID: 33997328 PMCID: PMC8110871 DOI: 10.1016/j.aninu.2020.11.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 10/25/2020] [Accepted: 11/04/2020] [Indexed: 12/29/2022]
Abstract
The intestinal immune system is affected by various factors during its development, such as maternal antibodies, host genes, intestinal microbial composition and activity, and various stresses (such as weaning stress). Intestinal microbes may have an important impact on the development of the host immune system. Appropriate interventions such as probiotics may have a positive effect on intestinal immunity by regulating the composition and activity of intestinal microbes. Moreover, probiotics participate in the regulation of host health in many ways; for instance, by improving digestion and the absorption of nutrients, immune response, increasing the content of intestinal-beneficial microorganisms, and inhibiting intestinal-pathogenic bacteria, and they participate in regulating intestinal diseases in various ways. Probiotics are widely used as additives in livestock and the poultry industry and bring health benefits to hosts by improving intestinal microbes and growth performance, which provides more choices for promoting strong and efficient productivity.
Collapse
Affiliation(s)
- Sujuan Ding
- College of Bioscience and Biotechnology, Hunan Agricultural University, Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, Changsha, 410128, China
| | - Wenxin Yan
- College of Bioscience and Biotechnology, Hunan Agricultural University, Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, Changsha, 410128, China
| | - Yong Ma
- College of Bioscience and Biotechnology, Hunan Agricultural University, Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, Changsha, 410128, China
| | - Jun Fang
- College of Bioscience and Biotechnology, Hunan Agricultural University, Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, Changsha, 410128, China
| |
Collapse
|
36
|
Rahmawati W, van der Pligt P, Worsley A, Willcox JC. Indonesian antenatal nutrition education: A qualitative study of healthcare professional views. WOMEN'S HEALTH (LONDON, ENGLAND) 2021; 17:17455065211066077. [PMID: 34892998 PMCID: PMC8669879 DOI: 10.1177/17455065211066077] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 11/11/2021] [Accepted: 11/22/2021] [Indexed: 11/15/2022]
Abstract
BACKGROUND Early lifestyle intervention, including antenatal nutrition education, is required to reduce the triple burden of malnutrition. Understanding healthcare professionals' views and experiences is essential for improving future nutrition education programmes for Indonesian pregnant women. This study aimed to investigate the views of Indonesian antenatal healthcare professionals regarding nutrition education for pregnant women and the improvements required to provide more effective antenatal nutrition education. METHODS A descriptive qualitative study involved semi-structured interviews was conducted with 24 healthcare professionals, including nutritionists (n = 10), midwives (n = 9) and obstetricians (n = 5) in Malang, Indonesia, between December 2018 and January 2019. Data were analysed using thematic analysis. RESULTS The study identified four main themes. First, healthcare professionals were aware of the importance of providing antenatal nutrition education, which included supporting its targeted delivery. Second, there were differing views on who should provide nutrition education. Most midwives and obstetricians viewed nutritionists as the prime nutrition education provider. Nutritionists were confident in their capability to provide nutrition education. However, some nutritionists reported that only a few women visited primary health centres and received nutrition counselling via this pathway. Third, healthcare professionals revealed some barriers in providing education for women. These barriers included a limited number of nutritionists, lack of consistent guidelines, lack of healthcare professionals' nutrition knowledge and lack of time during antenatal care services. Fourth, participants expressed the need to strengthen some system elements, including reinforcing collaboration, developing guidelines, and enhancing capacity building to improve future antenatal nutrition education. CONCLUSIONS Healthcare professionals play a central role in the provision of antenatal nutrition education. This study highlighted the importance of educational models that incorporate various antenatal nutrition education delivery strategies. These methods include maximizing referral systems and optimizing education through multiple delivery methods, from digital modes to traditional face-to-face nutrition education in pregnancy classes and community-based health services.
Collapse
Affiliation(s)
- Widya Rahmawati
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, VIC, Australia
- Department of Nutrition Science, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
| | - Paige van der Pligt
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, VIC, Australia
| | - Anthony Worsley
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, VIC, Australia
| | - Jane C Willcox
- School of Allied Health, College of Science, Health and Engineering, La Trobe University, Bundoora, VIC, Australia
- Centre for Quality and Patient Safety Research–Epworth HealthCare Partnership, Institute of Health Transformation, School of Nursing & Midwifery, Deakin University, Burwood, VIC, Australia
- School of Human Movement and Nutrition Sciences, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
37
|
Jayalekshmi VS, Ramachandran S. Maternal cholesterol levels during gestation: boon or bane for the offspring? Mol Cell Biochem 2021; 476:401-416. [PMID: 32964393 DOI: 10.1007/s11010-020-03916-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 09/15/2020] [Indexed: 02/08/2023]
Abstract
An increase in cholesterol levels is perceived during pregnancy and is considered as a normal adaptive response to the development of the fetus. In some pregnancies, excessive increase in total cholesterol with high levels of Low-Density Lipoprotein leads to maladaptation by the fetus to cholesterol demands, resulting in a pathological condition termed as maternal hypercholesterolemia (MH). MH is considered clinically irrelevant and therefore cholesterol levels are not routinely checked during pregnancy, as a consequence of which there is scarce information on its global prevalence in pregnant women. Studies have reported that MH during pregnancy can cause atherogenesis in adults emphasizing the concept of in utero programming of fetus. Moreover, Gestational Diabetes Mellitus, obesity and Polycystic Ovary Syndrome are potential risk factors which strengthen combined pathologies in placenta and fetuses of mothers with MH. However, lack of conclusive evidence on cholesterol transport and underlying programming demand substantial research to develop population-based life style strategies for women in their childbearing years. The current review focuses on the mechanisms and outcomes of MH from existing epidemiological as well as experimental data and presents a detailed insight on this novel risk factor of cardiovascular diseases.
Collapse
Affiliation(s)
- V S Jayalekshmi
- Cardiovascular Diseases and Diabetes Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
- PhD Program in Biotechnology, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Surya Ramachandran
- Cardiovascular Diseases and Diabetes Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India.
| |
Collapse
|
38
|
Intrauterine growth restriction: Clinical consequences on health and disease at adulthood. Reprod Toxicol 2021; 99:168-176. [DOI: 10.1016/j.reprotox.2020.10.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 10/01/2020] [Accepted: 10/04/2020] [Indexed: 02/06/2023]
|
39
|
Mendelian randomization and experimental IUGR reveal the adverse effect of low birth weight on lung structure and function. Sci Rep 2020; 10:22395. [PMID: 33372189 PMCID: PMC7769986 DOI: 10.1038/s41598-020-79245-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 12/03/2020] [Indexed: 11/17/2022] Open
Abstract
Intrauterine growth restriction (IUGR) and low birth weigth (LBW) are risk factors for neonatal chronic lung disease. However, maternal and fetal genetic factors and the molecular mechanisms remain unclear. We investigated the relationship between LBW and lung function with Mendelian randomisation analyses and studied angiogenesis in a low protein diet rat model of IUGR. Our data indicate a possible association between LBW and reduced FEV1 (p = 5.69E−18, MR-PRESSO) and FVC (6.02E-22, MR-PRESSO). Complimentary, we demonstrated two-phased perinatal programming after IUGR. The intrauterine phase (embryonic day 21) is earmarked by a reduction of endothelial cell markers (e.g. CD31) as well as mRNA expression of angiogenic factors (e.g., Vegfa, Flt1, Klf4). Protein analysis identified an activation of anti-angiogenic mTOR effectors. In the postnatal phase, lung capillaries (< 20 µm) were significantly reduced, expression of CD31 and VE-Cadherin were unaffected, whereas SMAD1/5/8 signaling and Klf4 protein were increased (p < 0.01). Moreover, elevated proteolytic activity of MMP2 and MMP9 was linked to a 50% reduction of lung elastic fibres. In conclusion, we show a possible link of LBW in humans and reduced lung function in adulthood. Experimental IUGR identifies an intrauterine phase with inhibition of angiogenic signaling, and a postnatal phase with proteolytic activity and reduced elastic fibres.
Collapse
|
40
|
Moreno-Fernandez J, Ochoa JJ, Lopez-Frias M, Diaz-Castro J. Impact of Early Nutrition, Physical Activity and Sleep on the Fetal Programming of Disease in the Pregnancy: A Narrative Review. Nutrients 2020; 12:nu12123900. [PMID: 33419354 PMCID: PMC7766505 DOI: 10.3390/nu12123900] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 12/17/2020] [Accepted: 12/18/2020] [Indexed: 12/13/2022] Open
Abstract
Early programming is the adaptation process by which nutrition and environmental factors alter development pathways during prenatal growth, inducing changes in postnatal metabolism and diseases. The aim of this narrative review, is evaluating the current knowledge in the scientific literature on the effects of nutrition, environmental factors, physical activity and sleep on development pathways. If in utero adaptations were incorrect, this would cause a mismatch between prenatal programming and adulthood. Adequate caloric intake, protein, mineral, vitamin, and long-chain fatty acids, have been noted for their relevance in the offspring brain functions and behavior. Fetus undernutrition/malnutrition causes a delay in growth and have detrimental effects on the development and subsequent functioning of the organs. Pregnancy is a particularly vulnerable period for the development of food preferences and for modifications in the emotional response. Maternal obesity increases the risk of developing perinatal complications and delivery by cesarean section and has long-term implications in the development of metabolic diseases. Physical exercise during pregnancy contributes to overall improved health post-partum. It is also interesting to highlight the relevance of sleep problems during pregnancy, which influence adequate growth and fetal development. Taking into account these considerations, we conclude that nutrition and metabolic factors during early life play a key role of health promotion and public health nutrition programs worldwide to improve the health of the offspring and the health costs of hospitalization.
Collapse
Affiliation(s)
- Jorge Moreno-Fernandez
- Department of Physiology, Faculty of Pharmacy, Campus Universitario de Cartuja, E-18071 Granada, Spain; (J.M.-F.); (M.L.-F.); (J.D.-C.)
- Institute of Nutrition and Food Technology “José Mataix Verdú”, University of Granada, E-18071 Granada, Spain
| | - Julio J. Ochoa
- Department of Physiology, Faculty of Pharmacy, Campus Universitario de Cartuja, E-18071 Granada, Spain; (J.M.-F.); (M.L.-F.); (J.D.-C.)
- Institute of Nutrition and Food Technology “José Mataix Verdú”, University of Granada, E-18071 Granada, Spain
- Correspondence: ; Tel.: +34-958-241-000 (ext. 20317)
| | - Magdalena Lopez-Frias
- Department of Physiology, Faculty of Pharmacy, Campus Universitario de Cartuja, E-18071 Granada, Spain; (J.M.-F.); (M.L.-F.); (J.D.-C.)
- Institute of Nutrition and Food Technology “José Mataix Verdú”, University of Granada, E-18071 Granada, Spain
| | - Javier Diaz-Castro
- Department of Physiology, Faculty of Pharmacy, Campus Universitario de Cartuja, E-18071 Granada, Spain; (J.M.-F.); (M.L.-F.); (J.D.-C.)
- Institute of Nutrition and Food Technology “José Mataix Verdú”, University of Granada, E-18071 Granada, Spain
| |
Collapse
|
41
|
Wu Y, Yin G, Wang P, Huang Z, Lin S. Effects of different diet-induced postnatal catch-up growth on glycolipid metabolism in intrauterine growth retardation male rats. Exp Ther Med 2020; 20:134. [PMID: 33082866 PMCID: PMC7560533 DOI: 10.3892/etm.2020.9263] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Accepted: 06/24/2020] [Indexed: 02/05/2023] Open
Abstract
A number of studies have reported the occurrence of long-term metabolic disorders in mammals following intrauterine growth retardation (IUGR). However, the effects of dietary patterns during IUGR have not been fully elucidated. The present study aimed to evaluate the effects of different dietary patterns during critical growth windows on metabolic outcomes in the offspring of rats with IUGR. Male offspring rats from mothers fed either a normal or low-protein diet were randomly assigned to one of the following groups: Normal diet throughout pregnancy, lactation and after weaning (CON); normal diet throughout pregnancy and high-fat diet throughout lactation and after weaning (N + H + H); low-protein diet throughout pregnancy and high-fat diet throughout lactation and after weaning (IUGR + H + H); low-protein diet throughout pregnancy and lactation and high-fat diet after weaning (IUGR + L + H); and low-protein diet throughout pregnancy and normal diet throughout lactation and after weaning. During lactation, the male offspring in the N + H + H group exhibited the fastest growth rate, whereas the slowest rate was in the IUGR + L + H group. Following weaning, all IUGR groups demonstrated significant catch-up growth. Abnormal insulin tolerance were observed in the N + H + H, IUGR + H + H and IUGR + L + H groups and insulin sensitivity was decreased in IUGR + L + H group. The triglycerides/high-density lipoprotein ratio in the IUGR + L + H group was significantly higher compared with in the other groups. The abdominal circumference, Lee's index and adipocyte diameter of IUGR groups were significantly increased compared with the CON group. High levels of leptin and interleukin-6 in adipose tissues, and low adiponectin were observed in the IUGR + L + H group. Different dietary patterns during specific growth windows showed numerous impacts on glycolipid metabolism in IUGR offspring. The present study elucidated the mechanisms and potential options for IUGR treatment and prevention.
Collapse
Affiliation(s)
- Yixi Wu
- Department of Endocrinology, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515000, P.R. China
| | - Guoshu Yin
- Department of Endocrinology, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515000, P.R. China
| | - Ping Wang
- Center of Reproductive Medicine, Shantou University Medical College, Shantou, Guangdong 515000, P.R. China
| | - Zhihua Huang
- Center of Reproductive Medicine, Shantou University Medical College, Shantou, Guangdong 515000, P.R. China
| | - Shaoda Lin
- Department of Endocrinology, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515000, P.R. China
- Correspondence to: Dr Shaoda Lin, Department of Endocrinology, The First Affiliated Hospital of Shantou University Medical College, 57 Changping Road, Shantou, Guangdong 515000, P.R. China
| |
Collapse
|
42
|
Hendrix MLE, van Kuijk SMJ, El Bahaey SE, Gerver WJM, Feron FJM, Kuin ME, Spaanderman MEA, Bons JAP, Al-Nasiry S. 'Postnatal growth during the first five years of life in SGA and AGA neonates with reduced fetal growth'. Early Hum Dev 2020; 151:105199. [PMID: 33032049 DOI: 10.1016/j.earlhumdev.2020.105199] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 09/17/2020] [Indexed: 10/23/2022]
Abstract
BACKGROUND Even though a lot of research has been done on postnatal growth and the occurrence of catch-up growth in small-for-gestational age (SGA) neonates, this phenomenon has not been studied well in appropriate-for-gestational age (AGA) neonates. Postnatal catch-up growth may also occur in AGA neonates indicating a compensatory mechanism for undiagnosed intrauterine growth restriction, especially in AGA neonates with reduced fetal growth velocity. AIMS To describe postnatal growth during the first 5 years of life in SGA and AGA neonates and evaluating the role of fetal growth velocity in catch-up growth. STUDY DESIGN Retrospective study in a Dutch tertiary hospital. SUBJECTS 740 singleton neonates, without congenital anomalies, with ultrasound fetal growth data from 20 weeks and 32 weeks of pregnancy. OUTCOME MEASURES Postnatal growth measurements of height (cm) and weight (kg) from birth until five years of age. Postnatal catch-up growth defined as difference (delta) in both height and weight between 4 weeks and 3 years of age. RESULTS AND CONCLUSIONS SGA neonates had a significantly lower height and weight compared to the AGA group for all available measurement moments till 3 years. The catch-up growth between the SGA and AGA groups from 4 weeks up to 3 years after birth was not different between the two groups. However, neonates with reduced fetal growth velocity had a significantly higher risk for catch-up growth in height during the first 3 years after birth. This suggests a role for fetal growth velocity measurement in predicting fetal and subsequent postnatal growth potential.
Collapse
Affiliation(s)
- M L E Hendrix
- Department of Obstetrics & Gynaecology, GROW School of Oncology and Developmental Biology, Maastricht University Medical Centre (MUMC+), Maastricht, the Netherlands.
| | - S M J van Kuijk
- Department of Clinical Epidemiology and Medical Technology Assessment (KEMTA), Maastricht University Medical Centre, Maastricht, the Netherlands
| | - S E El Bahaey
- Department of Obstetrics & Gynaecology, GROW School of Oncology and Developmental Biology, Maastricht University Medical Centre (MUMC+), Maastricht, the Netherlands
| | - W J M Gerver
- Department of Paediatrics Endocrinology, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - F J M Feron
- Department of Social Medicine, CAPHRI, Care and Public Health Research Institute, Maastricht University, Maastricht, the Netherlands
| | - M E Kuin
- Department of Obstetrics & Gynaecology, GROW School of Oncology and Developmental Biology, Maastricht University Medical Centre (MUMC+), Maastricht, the Netherlands
| | - M E A Spaanderman
- Department of Obstetrics & Gynaecology, GROW School of Oncology and Developmental Biology, Maastricht University Medical Centre (MUMC+), Maastricht, the Netherlands
| | - J A P Bons
- Central Diagnostic Laboratory, Maastricht University Medical Centre (MUMC+), Maastricht, the Netherlands
| | - S Al-Nasiry
- Department of Obstetrics & Gynaecology, GROW School of Oncology and Developmental Biology, Maastricht University Medical Centre (MUMC+), Maastricht, the Netherlands
| |
Collapse
|
43
|
Early life overnutrition impairs plasticity of non-neuronal brainstem cells and drives obesity in offspring across development in rats. Int J Obes (Lond) 2020; 44:2405-2418. [PMID: 32999409 DOI: 10.1038/s41366-020-00658-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 07/17/2020] [Accepted: 08/15/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND The prevalence of adolescent obesity has increased dramatically, becoming a serious public health concern. While previous evidence suggests that in utero- and early postnatal overnutrition increases adult-onset obesity risk, the neurobiological mechanisms underlying this outcome are not well understood. Non-neuronal cells play an underestimated role in the physiological responses to metabolic/nutrient signals. Hypothalamic glial-mediated inflammation is now considered a contributing factor in the development and perpetuation of obesity; however, attention on the role of gliosis and microglia activation in other nuclei is still needed. METHODS/RESULTS Here, we demonstrate that early life consumption of high-fat/sucrose diet (HFSD) is sufficient to increase offspring body weight, hyperleptinemia and potentially maladaptive cytoarchitectural changes in the brainstem dorsal-vagal-complex (DVC), an essential energy balance processing hub, across postnatal development. Our data demonstrate that pre- and postnatal consumption of HFSD result in increased body weight, hyperleptinemia and dramatically affects the non-neuronal landscape, and therefore the plasticity of the DVC in the developing offspring. CONCLUSIONS Current findings are very provocative, considering the importance of the DVC in appetite regulation, suggesting that HFSD-consumption during early life may contribute to subsequent obesity risk via DVC cytoarchitectural changes.
Collapse
|
44
|
Lager S, Sovio U, Eddershaw E, van der Linden MW, Yazar C, Cook E, Happerfield L, Jessop FA, Sebire NJ, Charnock-Jones DS, Smith GCS. Abnormal placental CD8 + T-cell infiltration is a feature of fetal growth restriction and pre-eclampsia. J Physiol 2020; 598:5555-5571. [PMID: 32886802 DOI: 10.1113/jp279532] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 08/28/2020] [Indexed: 12/13/2022] Open
Abstract
KEY POINTS Placental pathological abnormalities are more frequently observed in complicated pregnancies than in healthy pregnancies. Infiltration of CD8+ T-cells into the placental villous tissue occurred in both fetal growth restriction and pre-eclampsia, whereas CD79α+ B-cell infiltration was only apparent with reduced fetal growth. Vascularization, fibrin depositions, macrophage and neutrophil infiltration in the placenta did not differ between healthy and complicated pregnancies. ABSTRACT Fetal growth restriction (FGR) and pre-eclampsia are severe, adverse pregnancy outcomes. Alterations in placental histology are frequently reported in these pregnancy complications and are often based upon scoring by pathologists. However, many alterations are also observed in placenta from uncomplicated pregnancies. Moreover, knowledge of disease state may bias assessment. We sought to perform an objective comparison of placental microscopic appearance in normal and complicated pregnancies. Placental villous tissue (n = 823) and edge biopsies (n = 488) from 871 individual, singleton pregnancies were collected after delivery. Cases of small-for-gestational age (SGA) or pre-eclampsia were matched with healthy controls. A subset of the SGA cases displayed signs of FGR. Cases of preterm delivery were also included. Tissue sections were stained with haematoxylin and eosin or antibodies for CD8, CD14, CD31, CD79α and elastase. Images were scored by two experienced pathologists for pathological features or analysed by image analysis and stereology. Analyses were performed blind to case-control status and gestational age. Volume fraction of T-cells increased in placentas from pregnancies complicated by pre-eclampsia (adjusted odds ratio (aOR) 1.46, 95% CI: 1.12-1.90) and FGR (aOR 1.64, 95% CI: 1.11-2.43), whereas B-cells only increased in FGR (aOR 1.65, 95% CI: 1.05-2.60). Pathological abnormalities in villous tissue were reported in 21.4% (88/411) of complicated pregnancies and 14.3% (52/363) of controls (OR 1.62, 95% CI: 1.12-2.37). There were no differences in the fractions of endothelial cells, fibrin deposition, macrophages and neutrophils when comparing normal and complicated pregnancies. In conclusion, FGR and pre-eclampsia are associated with T-cell infiltration of the placenta and placental pathological abnormalities.
Collapse
Affiliation(s)
- Susanne Lager
- Department of Obstetrics & Gynaecology, University of Cambridge and NIHR Cambridge Biomedical Research Centre, Cambridge, UK.,Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, UK.,Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden
| | - Ulla Sovio
- Department of Obstetrics & Gynaecology, University of Cambridge and NIHR Cambridge Biomedical Research Centre, Cambridge, UK.,Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, UK
| | - Elizabeth Eddershaw
- Department of Obstetrics & Gynaecology, University of Cambridge and NIHR Cambridge Biomedical Research Centre, Cambridge, UK
| | - Margaretha W van der Linden
- Department of Obstetrics & Gynaecology, University of Cambridge and NIHR Cambridge Biomedical Research Centre, Cambridge, UK
| | - Cansu Yazar
- Department of Obstetrics & Gynaecology, University of Cambridge and NIHR Cambridge Biomedical Research Centre, Cambridge, UK
| | - Emma Cook
- Department of Obstetrics & Gynaecology, University of Cambridge and NIHR Cambridge Biomedical Research Centre, Cambridge, UK
| | - Lisa Happerfield
- Department of Paediatric Pathology, Addenbrooke's Hospital, Cambridge, UK
| | - Flora A Jessop
- Department of Paediatric Pathology, Addenbrooke's Hospital, Cambridge, UK
| | - Neil J Sebire
- Departments of Paediatric Pathology, Camelia Botnar Laboratories, Great Ormond Street Hospital and Institute of Child Health, London, UK
| | - D Stephen Charnock-Jones
- Department of Obstetrics & Gynaecology, University of Cambridge and NIHR Cambridge Biomedical Research Centre, Cambridge, UK.,Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, UK
| | - Gordon C S Smith
- Department of Obstetrics & Gynaecology, University of Cambridge and NIHR Cambridge Biomedical Research Centre, Cambridge, UK.,Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, UK
| |
Collapse
|
45
|
Habibi N, Grieger JA, Bianco-Miotto T. A Review of the Potential Interaction of Selenium and Iodine on Placental and Child Health. Nutrients 2020; 12:nu12092678. [PMID: 32887377 PMCID: PMC7551633 DOI: 10.3390/nu12092678] [Citation(s) in RCA: 9] [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: 07/10/2020] [Revised: 08/28/2020] [Accepted: 08/28/2020] [Indexed: 12/12/2022] Open
Abstract
A healthy pregnancy is important for the growth and development of a baby. An adverse pregnancy outcome is associated with increased chronic disease risk for the mother and offspring. An optimal diet both before and during pregnancy is essential to support the health of the mother and offspring. A key mediator of the effect of maternal nutrition factors on pregnancy outcomes is the placenta. Complicated pregnancies are characterized by increased oxidative stress in the placenta. Selenium and iodine are micronutrients that are involved in oxidative stress in placental cells. To date, there has been no comprehensive review investigating the potential synergistic effect of iodine and selenium in the placenta and how maternal deficiencies may be associated with increased oxidative stress and hence adverse pregnancy outcomes. We undertook a hypothesis-generating review on selenium and iodine, to look at how they may relate to pregnancy complications through oxidative stress. We propose how they may work together to impact pregnancy and placental health and explore how deficiencies in these micronutrients during pregnancy may impact the future health of offspring.
Collapse
Affiliation(s)
- Nahal Habibi
- School of Agriculture, Food and Wine, Waite Research Institute, and Robinson Research Institute, University of Adelaide, Adelaide 5005, Australia;
| | - Jessica A. Grieger
- Adelaide Medical School, and Robinson Research Institute, University of Adelaide, Adelaide 5005, Australia
- Correspondence: (J.A.G.); (T.B.-M.)
| | - Tina Bianco-Miotto
- School of Agriculture, Food and Wine, Waite Research Institute, and Robinson Research Institute, University of Adelaide, Adelaide 5005, Australia;
- Correspondence: (J.A.G.); (T.B.-M.)
| |
Collapse
|
46
|
Ma L, Tian MX, Sun QY, Liu NN, Dong JF, Feng K, Wu YK, Wang YX, Wang GY, Chen W, Xi JJ, Kang JH. Fetal growth restriction mice are more likely to exhibit depression-like behaviors due to stress-induced loss of dopaminergic neurons in the VTA. FASEB J 2020; 34:13257-13271. [PMID: 32860269 DOI: 10.1096/fj.202000534r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 07/05/2020] [Accepted: 07/16/2020] [Indexed: 11/11/2022]
Abstract
Fetal growth restriction (FGR) is a severe perinatal complication that can increase risk for mental illness. To investigate the mechanism by which FGR mice develop mental illness in adulthood, we established the FGR mouse model and the FGR mice did not display obvious depression-like behaviors, but after environmental stress exposure, FGR mice were more likely to exhibit depression-like behaviors than control mice. Moreover, FGR mice had significantly fewer dopaminergic neurons in the ventral tegmental area but no difference in serotoninergic neurons in the dorsal raphe. RNA-seq analysis showed that the downregulated genes in the midbrain of FGR mice were associated with many mental diseases and were especially involved in the regulation of NMDA-selective glutamate receptor (NMDAR) activity. Furthermore, the NMDAR antagonist memantine can relieve the stress-induced depression-like behaviors of FGR mice. In summary, our findings provide a theoretical basis for future research and treatment of FGR-related depression.
Collapse
Affiliation(s)
- Li Ma
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Collaborative Innovation Center for Brain Science, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Meng-Xue Tian
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Collaborative Innovation Center for Brain Science, School of Life Sciences and Technology, Tongji University, Shanghai, China.,Institute of Translational Research, Tongji Hospital, School of Life Sciences and Technology, Shanghai Key Laboratory of Signaling and Disease Research, Collaborative Innovation Center for Brain Science, Tongji University, Shanghai, China
| | - Qiao-Yi Sun
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Collaborative Innovation Center for Brain Science, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Na-Na Liu
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Collaborative Innovation Center for Brain Science, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Jian-Feng Dong
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Collaborative Innovation Center for Brain Science, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Ke Feng
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Collaborative Innovation Center for Brain Science, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Yu-Kang Wu
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Collaborative Innovation Center for Brain Science, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Yu-Xi Wang
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Collaborative Innovation Center for Brain Science, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Gui-Ying Wang
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Collaborative Innovation Center for Brain Science, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Wen Chen
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Collaborative Innovation Center for Brain Science, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Jia-Jie Xi
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Collaborative Innovation Center for Brain Science, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Jiu-Hong Kang
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Collaborative Innovation Center for Brain Science, School of Life Sciences and Technology, Tongji University, Shanghai, China
| |
Collapse
|
47
|
Kadayifci FZ, Haggard S, Jeon S, Ranard K, Tao D, Pan YX. Early-life Programming of Type 2 Diabetes Mellitus: Understanding the Association between Epigenetics/Genetics and Environmental Factors. Curr Genomics 2020; 20:453-463. [PMID: 32477001 PMCID: PMC7235385 DOI: 10.2174/1389202920666191009110724] [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: 04/15/2019] [Revised: 07/03/2019] [Accepted: 09/06/2019] [Indexed: 11/22/2022] Open
Abstract
Type 2 Diabetes Mellitus is an increasing public health problem that poses a severe social and economic burden affecting both developed and developing countries. Defects in insulin signaling itself are among the earliest indications that an individual is predisposed to the development of insulin resistance and subsequently Type 2 Diabetes Mellitus. To date, however, the underlying molecular mechanisms which result in resistance to the actions of insulin are poorly understood. Furthermore, it has been shown that maternal obesity is associated with an increased risk of obesity and insulin resistance in the offspring. However, the genetic and/or epigenetic modifications within insulin-sensitive tissues such as the liver and skeletal muscle, which contribute to the insulin-resistant phenotype, still remain unknown. More importantly, a lack of in-depth understanding of how the early life environment can have long-lasting effects on health and increased risk of Type 2 Diabetes Mellitus in adulthood poses a major limitation to such efforts. The focus of the current review is thus to discuss recent experimental and human evidence of an epigenetic component associated with components of nutritional programming of Type 2 Diabetes Mellitus, including altered feeding behavior, adipose tissue, and pancreatic beta-cell dysfunction, and transgenerational risk transmission.
Collapse
Affiliation(s)
- Fatma Z Kadayifci
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Sage Haggard
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Sookyoung Jeon
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Katie Ranard
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Dandan Tao
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Yuan-Xiang Pan
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, IL, USA.,Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA.,Illinois Informatics Institute, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| |
Collapse
|
48
|
Manangama G, Audignon-Durand S, Migault L, Gramond C, Zaros C, Teysseire R, Sentilhes L, Brochard P, Lacourt A, Delva F. Maternal occupational exposure to carbonaceous nanoscale particles and small for gestational age and the evolution of head circumference in the French Longitudinal Study of Children - Elfe study. ENVIRONMENTAL RESEARCH 2020; 185:109394. [PMID: 32247149 DOI: 10.1016/j.envres.2020.109394] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 03/03/2020] [Accepted: 03/13/2020] [Indexed: 06/11/2023]
Abstract
OBJECTIVES To investigate the association between exposure to unintentionally emitted carbonaceous nanoscale particles (NPs) and small for gestational age (SGA), as well as head circumference (HC) at birth and at two years of age. METHODS Mothers from the French Longitudinal Study of Children (Elfe cohort) who worked during pregnancy were selected for our study. Data collected at birth and during follow-up (up to two years) were used. The probability and frequency of maternal occupational exposure to unintentionally emitted carbonaceous NPs was estimated using a job exposure matrix (MatPUF). Multivariate logistic regression, linear regression, and mixed models were applied to estimate any associations. Analyses were carried out depending on whether mothers stopped working during the first, second, or third trimester of pregnancy. RESULTS Maternal occupational exposure to unintentionally emitted carbonaceous NPs was associated with SGA in the overall study population by multivariate analysis (ORa = 1.80, 95% CI: 1.29, 2.46), as well as in sub-groups of mothers who stopped working during the second (ORa = 1.84, 95% CI: 1.13, 3.02) or third (ORa = 1.80, 95% CI: 1.10, 2.95) trimesters. There were no significant associations with HC at birth or two years of age. CONCLUSIONS We found a significant association between occupational exposure to carbonaceous NPs and SGA, with the effect depending on the period of exposure during pregnancy. These results should encourage further studies concerning the adverse effects of exposure to carbonaceous NPs on the development of offspring.
Collapse
Affiliation(s)
- Guyguy Manangama
- Bordeaux Teaching Hospital, Artemis Center, Bordeaux, France; University of Bordeaux, Inserm UMR1219-EPICENE, Bordeaux Population Health Research Center, Bordeaux, France.
| | - Sabyne Audignon-Durand
- University of Bordeaux, Inserm UMR1219-EPICENE, Bordeaux Population Health Research Center, Bordeaux, France
| | - Lucile Migault
- French Agency for Food, Environmental and Occupational Health & Safety, Paris, France
| | - Céline Gramond
- University of Bordeaux, Inserm UMR1219-EPICENE, Bordeaux Population Health Research Center, Bordeaux, France
| | - Cécile Zaros
- Joint Research Unit Elfe, Ined-Inserm-EFS, France
| | - Raphaëlle Teysseire
- Bordeaux Teaching Hospital, Artemis Center, Bordeaux, France; University of Bordeaux, Inserm UMR1219-EPICENE, Bordeaux Population Health Research Center, Bordeaux, France
| | - Loïc Sentilhes
- Bordeaux Teaching Hospital, Artemis Center, Bordeaux, France; Department of Obstetrics and Gynecology, Bordeaux University Hospital, Bordeaux, France
| | - Patrick Brochard
- Bordeaux Teaching Hospital, Artemis Center, Bordeaux, France; University of Bordeaux, Inserm UMR1219-EPICENE, Bordeaux Population Health Research Center, Bordeaux, France
| | - Aude Lacourt
- University of Bordeaux, Inserm UMR1219-EPICENE, Bordeaux Population Health Research Center, Bordeaux, France
| | - Fleur Delva
- Bordeaux Teaching Hospital, Artemis Center, Bordeaux, France; Clinical and Epidemiological Research Unit, INSERM CIC1401, F-33000, Bordeaux, France; University of Bordeaux, Inserm UMR1219-EPICENE, Bordeaux Population Health Research Center, Bordeaux, France
| |
Collapse
|
49
|
Felicioni F, Santos TG, Paula TDMDE, Chiarini-Garcia H, de Almeida FRCL. Intrauterine growth restriction: screening and diagnosis using animal models. Anim Reprod 2020; 16:66-71. [PMID: 33299479 PMCID: PMC7720938 DOI: 10.21451/1984-3143-ar2018-127] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Intrauterine growth restriction (IUGR) is a serious condition of multifactorial origin, mainly caused by maternal malnutrition, multiple gestation associated with nutrient competition, abuse of nocive substances and infections. The diagnosis of such syndrome is complex, as its own manifestations can mask its occurrence, requiring a thorough assessment of body weight and size. Moreover, it is not responsive to any kind of treatment. There is evidence that IUGR may predispose the individual to several pathologies, such as diabetes, hypertension and metabolic syndrome in adulthood, and it has also been linked to thrifty phenotype hypothesis. Thus, a healthy lifestyle is needed to better prevent those pathologies. Given the world high prevalence and importance of IUGR, mainly in developing countries, this review is focused on discussing how different animal models contribute to the biological screening and diagnosis of this condition.
Collapse
Affiliation(s)
- Fernando Felicioni
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Thaís Garcia Santos
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Hélio Chiarini-Garcia
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | | |
Collapse
|
50
|
Krishna RG, Vishnu Bhat B, Bobby Z, Papa D, Badhe B, Kalidoss VK, Karli S. Identification of differentially methylated candidate genes and their biological significance in IUGR neonates by methylation EPIC array. J Matern Fetal Neonatal Med 2020; 35:525-533. [PMID: 32091279 DOI: 10.1080/14767058.2020.1727881] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Background: Intrauterine growth restriction (IUGR) is a pregnancy-associated disease manifested by decreased growth rate of fetus than the normal genetic growth potential. It is associated with increased susceptibility to metabolic diseases later in life. Although the mechanisms underlying the origin of metabolic diseases are poorly understood, DNA methylation is a crucial investigation for the identification of epigenetic changes.Objectives: To assess the degree of change of DNA methylation in IUGR neonates and compare with that of appropriate for gestational age (AGA) neonates and to explore the differentially methylated candidate genes and their biological significance.Methods: This cohort study was conducted in the Neonatology Department of JIPMER during the period of November 2017 to December 2018. Forty each of IUGR and gestation matched AGA neonates were recruited. Umbilical cord blood samples were collected at birth. DNA was separated from the blood samples; and, using 5-mC DNA ELISA method, the percentage of genomic DNA methylated in these neonates was established. Data were expressed as mean ± standard deviation. Methylation EPIC array was performed to identify the differentially methylated candidate genes. David analysis was used to find out the functional annotation chart by KEGG pathway.Results: Genomic DNA methylation varied significantly between IUGR and AGA neonates (IUGR: 3.12 ± 1.24; AGA: 4.40 ± 2.03; p value: <.01). A global shift toward hypomethylation was seen in IUGR compared with AGA, targeted to regulatory regions of the genome, and specifically promoters. Pathway analysis identified deregulation of pathways involved in metabolic diseases. Altered methylation of PTPRN2 & HLADQB1 genes leads to dysregulation of T-cells and reactive oxygen species (ROS). These changes may lead to complications later among these neonates subjected to IUGR.Conclusion: Our findings show significant changes in the methylation pattern of genes among IUGR and AGA babies. Steps for correcting the changes may help in reducing later complications among IUGR babies.
Collapse
Affiliation(s)
- Rao Gurugubelli Krishna
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS), Mangalagiri, Andhra Pradesh, India.,Department of Neonatology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Puducherry, India
| | - Ballambattu Vishnu Bhat
- Department of Neonatology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Puducherry, India.,Department of Pediatrics, AVMC, Puducherry, India
| | | | - Dasari Papa
- Department of Obstetrics & Gynaecology, JIPMER, Puducherry, India
| | | | | | | |
Collapse
|