1
|
Travers CP, Ambalavanan N. More may be better: extended CPAP may help alveolar development. Eur Respir J 2025; 65:2500072. [PMID: 40180361 DOI: 10.1183/13993003.00072-2025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 01/17/2025] [Indexed: 04/05/2025]
Affiliation(s)
- Colm P Travers
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA
| | | |
Collapse
|
2
|
Fowden AL, Vaughan OR, Forhead AJ. Early-life programming of livestock metabolism by glucocorticoids. J Dev Orig Health Dis 2025; 16:e16. [PMID: 40104937 DOI: 10.1017/s2040174425000091] [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] [Indexed: 03/20/2025]
Abstract
Adverse environmental conditions during early life are known to determine adult metabolic phenotype in laboratory species and human populations. However, less is known about developmental programming of adult metabolic phenotype in livestock, given their size and longevity compared to laboratory animals. As maternal and/or fetal glucocorticoid (GC) concentrations rise in stressful conditions during pregnancy, GCs may act as a common mechanism linking early-life environmental conditions to the subsequent metabolic phenotype. This review examines prenatal and longer-term postnatal programming of metabolism by early-life GC overexposure in livestock species with a particular emphasis on sheep. It examines the effects of both cortisol, the natural glucocorticoid and more potent synthetic GCs used clinically to treat threatened pre-term delivery and other conditions during pregnancy. It considers the effects of early- life GC overexposure on the metabolism of specific feto-placental and adult tissues in relation to changes in the growth trajectory, other metabolic hormones and in the functioning of the hypothalamic-pituitary-adrenal axis itself. It highlights the role of GCs as maturational and environmental signals in programming development of a metabolic phenotype fit for survival at birth and future homeostatic challenges. However, the ensuing metabolic phenotype induced by early GC overexposure may become inappropriate for the prevailing postnatal conditions and lead to metabolic dysfunction as functional reserves decline with age. Further studies are needed in livestock to establish whether the metabolic outcomes of early-life GC overexposure are sex-linked, more pronounced in old age and inherited transgenerationally in these species.
Collapse
Affiliation(s)
- Abigail L Fowden
- Department of Physiology Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Owen R Vaughan
- Institute of Women's Health, University College London, London, UK
| | - Alison J Forhead
- Department of Physiology Development and Neuroscience, University of Cambridge, Cambridge, UK
- Department of Biological and Medical Sciences, Oxford Brookes University, Oxford, UK
| |
Collapse
|
3
|
Roff AJ, Davies AN, Clifton VL, Stark MJ, Tai A, Robinson JL, Hammond SJ, Darby JRT, Meakin AS, Lock MC, Wiese MD, Sharkey DJ, Bischof RJ, Morrison JL, Gatford KL. Pregnancy does not affect progression of mild experimental asthma in sheep. J Physiol 2025; 603:1241-1261. [PMID: 39915955 PMCID: PMC11870047 DOI: 10.1113/jp287583] [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: 08/31/2024] [Accepted: 01/15/2025] [Indexed: 03/01/2025] Open
Abstract
Asthma is the most common respiratory condition during pregnancy and increases the risks of adverse pregnancy and perinatal outcomes. Asthma symptoms change in ∼60% of pregnancies, but whether this is due to pregnancy itself is unclear. We tested the hypothesis that physiological changes during pregnancy worsen asthma severity in an ovine experimental model of allergic asthma. Three-year-old Merino ewes were randomly allocated to either control or allergic asthma groups. Asthma was induced through sensitisation and repeated airway challenge with house dust mite allergen. We compared airway function, circulating cytokine profile and airway immune response to allergen challenge throughout the study and airway structure between groups, in non-pregnant (control n = 8, asthma n = 9), singleton-pregnant (control n = 5, asthma n = 8) and twin-pregnant ewes (control n = 6, asthma n = 9). Within non-pregnant animals, transpulmonary pressure at Day 132 of the study was 37% higher in asthmatic than control ewes (P = 0.031), but not different between treatments in singleton-pregnant (P = 0.594) or twin-pregnant (P = 0.074) ewes. Between premating and Day 132, dynamic compliance decreased more in asthmatic than control ewes (P = 0.040), and this change did not differ between litter sizes (P = 0.096). Neither asthma nor pregnancy affected eosinophils in bronchoalveolar lavage or lung tissue. There was no evidence of lung airway remodelling in the cohort. The results of this study suggest that pregnancy does not increase asthma symptoms or severity of mild asthma. KEY POINTS: Asthma severity changes in ∼60% of pregnancies, but whether this is due to pregnancy itself is unclear. Using a sheep model of allergic asthma, we tested the hypothesis that physiological changes during pregnancy worsen asthma severity. Dynamic compliance decreased to a greater extent in asthmatic than control ewes over the course of the study, indicating the development of a mild asthma phenotype, and this decrease was similar in non-pregnant, singleton-pregnant and twin-pregnant ewes. Eosinophil proportions in bronchoalveolar lavage and lung tissue were not affected by either asthma or pregnancy, nor was there evidence of lung airway remodelling in this cohort. Our findings suggest that pregnancy does not increase asthma symptoms or severity of mild asthma.
Collapse
Affiliation(s)
- Andrea J. Roff
- Robinson Research InstituteUniversity of AdelaideAdelaideSAAustralia
- School of BiomedicineUniversity of AdelaideAdelaideSAAustralia
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, Clinical and Health SciencesUniversity of South AustraliaAdelaideAustralia
| | - Andrew N. Davies
- Biomedicine Discovery InstituteMonash UniversityFrankstonVICAustralia
| | - Vicki L. Clifton
- Mater Medical Research InstituteUniversity of QueenslandBrisbaneQLDAustralia
| | - Michael J. Stark
- Robinson Research InstituteUniversity of AdelaideAdelaideSAAustralia
- Adelaide Medical SchoolUniversity of AdelaideAdelaideSAAustralia
| | - Andrew Tai
- Robinson Research InstituteUniversity of AdelaideAdelaideSAAustralia
- Adelaide Medical SchoolUniversity of AdelaideAdelaideSAAustralia
- Women's & Children's HospitalNorth AdelaideSAAustralia
| | - Joshua L. Robinson
- Robinson Research InstituteUniversity of AdelaideAdelaideSAAustralia
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, Clinical and Health SciencesUniversity of South AustraliaAdelaideAustralia
- Adelaide Medical SchoolUniversity of AdelaideAdelaideSAAustralia
| | - Sarah J. Hammond
- Robinson Research InstituteUniversity of AdelaideAdelaideSAAustralia
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, Clinical and Health SciencesUniversity of South AustraliaAdelaideAustralia
| | - Jack R. T. Darby
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, Clinical and Health SciencesUniversity of South AustraliaAdelaideAustralia
| | - Ashley S. Meakin
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, Clinical and Health SciencesUniversity of South AustraliaAdelaideAustralia
| | - Mitchell C. Lock
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, Clinical and Health SciencesUniversity of South AustraliaAdelaideAustralia
| | - Michael D. Wiese
- Centre for Pharmaceutical Innovation, Clinical & Health SciencesUniversity of South AustraliaAdelaideSAAustralia
| | - David J. Sharkey
- Robinson Research InstituteUniversity of AdelaideAdelaideSAAustralia
- School of BiomedicineUniversity of AdelaideAdelaideSAAustralia
| | - Robert J. Bischof
- Institute of Innovation, Science and SustainabilityFederation University AustraliaBerwickVICAustralia
| | - Janna L. Morrison
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, Clinical and Health SciencesUniversity of South AustraliaAdelaideAustralia
| | - Kathryn L. Gatford
- Robinson Research InstituteUniversity of AdelaideAdelaideSAAustralia
- School of BiomedicineUniversity of AdelaideAdelaideSAAustralia
| |
Collapse
|
4
|
McGillick EV, Orgeig S, Allison BJ, Brain KL, Niu Y, Itani N, Skeffington KL, Kane AD, Herrera EA, Giussani DA, Morrison JL. Antenatal Vitamin C differentially affects lung development in normally grown and growth restricted sheep. Pediatr Res 2025:10.1038/s41390-025-03828-1. [PMID: 40000856 DOI: 10.1038/s41390-025-03828-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 12/07/2024] [Accepted: 12/31/2024] [Indexed: 02/27/2025]
Abstract
BACKGROUND Chronic hypoxemia is a common cause of fetal growth restriction and can have significant effects on the developing fetal lung. Maternal antioxidant treatment in hypoxic pregnancy protects against offspring cardiovascular dysfunction. The effects of antenatal antioxidants on lung development in the chronically hypoxic growth restricted fetus is unknown. METHODS We investigated the effect of maternal daily Vitamin C (200 mg/kg i.v. vs. Saline) for a month in late gestation on molecular markers regulating lung maturation between normoxic normally grown and hypoxic growth-restricted fetal sheep. Chronic fetal hypoxia and fetal growth restriction were induced by exposure to maternal chronic hypoxia (10% O2 vs. Normoxia=21% O2) from 105-138 d gestation (term=145 d). RESULTS The data show a differential effect of antenatal Vitamin C treatment on regulation of genes involved in surfactant maturation, sodium movement and hypoxia signaling. Limited responsiveness to antenatal Vitamin C exposure in the lung of the hypoxic fetus, compared to responsiveness to antenatal Vitamin C in the normoxic fetus, suggests a maximal upregulation of the molecular signaling pathways in response to the chronic hypoxic insult alone. CONCLUSION We provide molecular insight into the heterogeneity of antenatal Vitamin C treatment on development of the normoxic and growth restricted hypoxic fetal lung. IMPACT The effect of maternal Vitamin C on molecular markers of lung maturation between normoxic normally grown and hypoxic growth restricted fetal sheep was unknown. We show a differential effect of Vitamin C with a greater increase in molecular markers of lung maturation in normoxic compared with hypoxic fetuses. Limited responsiveness in the hypoxic fetal lung is likely due to maximal upregulation by the hypoxic insult alone, thus added exposure to Vitamin C is unable to upregulate the system further. The work highlights the need to understand differential effects of antenatal interventions in healthy and complicated pregnancy, prior to clinical translation.
Collapse
Affiliation(s)
- Erin V McGillick
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, Adelaide, Australia.
- UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, Australia.
| | - Sandra Orgeig
- UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, Australia
| | - Beth J Allison
- Department of Physiology, Development & Neuroscience, University of Cambridge, Cambridgeshire, Cambridge, United Kingdom
| | - Kirsty L Brain
- Department of Physiology, Development & Neuroscience, University of Cambridge, Cambridgeshire, Cambridge, United Kingdom
| | - Youguo Niu
- Department of Physiology, Development & Neuroscience, University of Cambridge, Cambridgeshire, Cambridge, United Kingdom
| | - Nozomi Itani
- Department of Physiology, Development & Neuroscience, University of Cambridge, Cambridgeshire, Cambridge, United Kingdom
| | - Katie L Skeffington
- Department of Physiology, Development & Neuroscience, University of Cambridge, Cambridgeshire, Cambridge, United Kingdom
| | - Andrew D Kane
- Department of Physiology, Development & Neuroscience, University of Cambridge, Cambridgeshire, Cambridge, United Kingdom
| | - Emilio A Herrera
- Programa de Fisiopatología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Dino A Giussani
- Department of Physiology, Development & Neuroscience, University of Cambridge, Cambridgeshire, Cambridge, United Kingdom
| | - Janna L Morrison
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, Adelaide, Australia.
- UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, Australia.
| |
Collapse
|
5
|
Tong W, Allison BJ, Brain KL, Patey OV, Niu Y, Botting KJ, Ford SG, Garrud TA, Wooding PFB, Lyu Q, Zhang L, Ma J, Sowton AP, O'Brien KA, Cindrova-Davies T, Yung HW, Burton GJ, Murray AJ, Giussani DA. Placental mitochondrial metabolic adaptation maintains cellular energy balance in pregnancy complicated by gestational hypoxia. J Physiol 2025. [PMID: 39868991 DOI: 10.1113/jp287897] [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: 10/17/2024] [Accepted: 12/18/2024] [Indexed: 01/28/2025] Open
Abstract
The mechanisms that drive placental dysfunction in pregnancies complicated by hypoxia and fetal growth restriction remain poorly understood. Changes to mitochondrial respiration contribute to cellular dysfunction in conditions of hypoxia and have been implicated in the pathoaetiology of pregnancy complications, such as pre-eclampsia. We used bespoke isobaric hypoxic chambers and a combination of functional, molecular and imaging techniques to study cellular metabolism and mitochondrial dynamics in sheep undergoing hypoxic pregnancy. We show that hypoxic pregnancy in sheep triggers a shift in capacity away from β-oxidation and complex I-mediated respiration, while maintaining total oxidative phosphorylation capacity. There are also complex-specific changes to electron transport chain composition and a switch in mitochondrial dynamics towards fission. Hypoxic placentas show increased activation of the non-canonical mitochondrial unfolded protein response pathway and enhanced insulin like growth factor 2 signalling. Combined, therefore, the data show that the hypoxic placenta undergoes significant metabolic and morphological adaptations to maintain cellular energy balance. Chronic hypoxia during pregnancy in sheep activated placental mitochondrial stress pathways, leading to alterations in mitochondrial respiration, mitochondrial energy metabolism and mitochondrial dynamics, as seen in the placenta of women with pre-eclampsia. KEY POINTS: Hypoxia shifts mitochondrial respiration away from β-oxidation and complex I. Complex-specific changes occur in the electron transport chain composition. Activation of the non-canonical mitochondrial unfolded protein response pathway is heightened in hypoxic placentas. Enhanced insulin like growth factor 2 signalling is observed in hypoxic placentas. Hypoxic placentas undergo significant functional adaptations for energy balance.
Collapse
Affiliation(s)
- Wen Tong
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
- Strategic Research Initiative in Reproduction, University of Cambridge, Cambridge, UK
| | - Beth J Allison
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Kirsty L Brain
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Olga V Patey
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Youguo Niu
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
- Strategic Research Initiative in Reproduction, University of Cambridge, Cambridge, UK
- Cardiovascular Strategic Research Initiative, University of Cambridge, Cambridge, UK
- Department of Aerospace Physiology, Fourth Military Medical University, Xi'an, China
| | - Kimberley J Botting
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
- Strategic Research Initiative in Reproduction, University of Cambridge, Cambridge, UK
- Cardiovascular Strategic Research Initiative, University of Cambridge, Cambridge, UK
| | - Sage G Ford
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Tess A Garrud
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
| | - Peter F B Wooding
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
| | - Qiang Lyu
- Department of Aerospace Physiology, Fourth Military Medical University, Xi'an, China
| | - Lin Zhang
- Department of Aerospace Physiology, Fourth Military Medical University, Xi'an, China
| | - Jin Ma
- Department of Aerospace Physiology, Fourth Military Medical University, Xi'an, China
| | - Alice P Sowton
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
| | - Katie A O'Brien
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
| | - Tereza Cindrova-Davies
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
- Strategic Research Initiative in Reproduction, University of Cambridge, Cambridge, UK
| | - Hong Wa Yung
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
- Strategic Research Initiative in Reproduction, University of Cambridge, Cambridge, UK
| | - Graham J Burton
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
- Strategic Research Initiative in Reproduction, University of Cambridge, Cambridge, UK
| | - Andrew J Murray
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
- Cardiovascular Strategic Research Initiative, University of Cambridge, Cambridge, UK
| | - Dino A Giussani
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
- Strategic Research Initiative in Reproduction, University of Cambridge, Cambridge, UK
- Cardiovascular Strategic Research Initiative, University of Cambridge, Cambridge, UK
- Department of Aerospace Physiology, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
6
|
Hammond SJ, Roff AJ, Robinson JL, Darby JRT, Meakin AS, Clifton VL, Bischof RJ, Stark MJ, Wallace MJ, Tai A, Morrison JL, Gatford KL. In utero exposure to experimental maternal asthma alters fetal airway development in sheep. Exp Physiol 2025. [PMID: 39869487 DOI: 10.1113/ep092502] [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: 12/10/2024] [Accepted: 12/17/2024] [Indexed: 01/29/2025]
Abstract
The mechanisms linking maternal asthma (MA) exposure in utero and subsequent risk of asthma in childhood are not fully understood. Pathological airway remodelling, including reticular basement membrane thickening, has been reported in infants and children who go on to develop asthma later in childhood. This suggests altered airway development before birth as a mechanism underlying increased risk of asthma in children exposed in utero to MA. We hypothesised that in utero MA exposure would reduce airway diameter and increase airway-associated smooth muscle area and reticular basement membrane thickness in neonatal offspring. Experimental MA was induced by maternal sensitisation followed by airway challenges with house dust mite before and during pregnancy. Lambs from control (n = 16) or MA (n = 26) ewes were delivered at ∼140 days gestation (term = 150 days), ventilated for 45 min, then humanely killed. Left lungs were inflation-fixed, and cross-sections of generation 2-5 airways were collected. Airway sections were stained with Haematoxylin and Eosin, Masson's Trichrome and Gordon and Sweet's histological stains for morphological analysis. Lamb body and lung weights were similar between groups (P > 0.5 and P > 0.7, respectively). Lambs that were exposed to MA had narrower airway diameters (P = 0.019) and thinner reticular basement membrane (P = 0.016) but similar airway-associated smooth muscle area (P = 0.152) compared with unexposed control lambs. Our results demonstrate a potential mechanism for increased risk of asthma in children of mothers with asthma, independent of genetic risk or behavioural changes during pregnancy.
Collapse
Affiliation(s)
- Sarah J Hammond
- Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
- School of Biomedicine, University of Adelaide, Adelaide, South Australia, Australia
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation and Health Sciences, University of South Australia, Adelaide, Australia
| | - Andrea J Roff
- Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
- School of Biomedicine, University of Adelaide, Adelaide, South Australia, Australia
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation and Health Sciences, University of South Australia, Adelaide, Australia
| | - Joshua L Robinson
- Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation and Health Sciences, University of South Australia, Adelaide, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Jack R T Darby
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation and Health Sciences, University of South Australia, Adelaide, Australia
| | - Ashley S Meakin
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation and Health Sciences, University of South Australia, Adelaide, Australia
| | - Vicki L Clifton
- Mater Medical Research Institute, University of Queensland, South Brisbane, Queensland, Australia
| | - Robert J Bischof
- Institute of Innovation, Science and Sustainability, Federation University Australia, Berwick, Victoria, Australia
| | - Michael J Stark
- Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
- Department of Neonatal Medicine, Women's & Children's Hospital, North Adelaide, South Australia, Australia
| | - Megan J Wallace
- Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, Australia
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Andrew Tai
- Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
- Respiratory and Sleep Medicine, Women's & Children's Hospital, North Adelaide, South Australia, Australia
| | - Janna L Morrison
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation and Health Sciences, University of South Australia, Adelaide, Australia
| | - Kathryn L Gatford
- Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
- School of Biomedicine, University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
7
|
Domingues RR, Wiltbank MC, Hernandez LL, Adcock SJJ. Prenatal treatment with the antidepressant fluoxetine on maternal and neonatal behavior in sheep. Pediatr Res 2025:10.1038/s41390-025-03799-3. [PMID: 39809853 DOI: 10.1038/s41390-025-03799-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 11/18/2024] [Accepted: 12/15/2024] [Indexed: 01/16/2025]
Abstract
BACKGROUND Fluoxetine is commonly prescribed to treat depression during pregnancy. We aimed to evaluate the effects of prenatal fluoxetine exposure on maternal-offspring behavior in a non-depressed sheep model. METHODS On day 119 ± 1 of a 151-day expected gestation, Hampshire ewes were randomly assigned to receive intravenous fluoxetine (10 mg/kg for the first 2 days and 5 mg/kg daily thereafter until parturition) or a control vehicle. Video was recorded of 8 fluoxetine-treated ewes and 10 control ewes for 2 h before and after parturition. RESULTS Fluoxetine did not alter dam behavior during the peripartum period, including time spent lying before the first birth, lying bout duration, probability of needing birth assistance, duration of birth assistance when provided, and time spent touching her lambs. However, in utero exposure impaired neonatal vigor as lambs spent less time standing and tended to spend less time nursing compared to unexposed lambs. CONCLUSION Neonatal behavioral impairments are consistent with those associated with fluoxetine exposure during human gestation. This effect appears to be independent of maternal behavior, which was unaffected by antidepressant use. IMPACT Lambs exposed to SSRI in utero spent less time standing and tended to spend less time nursing than control lambs, consistent with neonatal behavioral outcomes encountered in clinical practice. The reduced neonatal vigor was likely unrelated to maternal behavior, which was not altered by SSRI treatment. Non-depressed sheep models can help to elucidate the behavioral effects of antidepressant use during pregnancy to enhance health outcomes and patient care.
Collapse
Affiliation(s)
- Rafael R Domingues
- Department of Animal and Dairy Sciences, University of Wisconsin, Madison, WI, USA
- Endocrinology and Reproductive Physiology Program, University of Wisconsin, Madison, WI, USA
- Department of Animal Sciences, The Ohio State University, Columbus, OH, USA
| | - Milo C Wiltbank
- Department of Animal and Dairy Sciences, University of Wisconsin, Madison, WI, USA
- Endocrinology and Reproductive Physiology Program, University of Wisconsin, Madison, WI, USA
| | - Laura L Hernandez
- Department of Animal and Dairy Sciences, University of Wisconsin, Madison, WI, USA
- Endocrinology and Reproductive Physiology Program, University of Wisconsin, Madison, WI, USA
| | - Sarah J J Adcock
- Department of Animal and Dairy Sciences, University of Wisconsin, Madison, WI, USA.
| |
Collapse
|
8
|
Cai Y, Yang H, Xu H, Li S, Zhao B, Wang Z, Yao X, Wang F, Zhang Y. β-Nicotinamide Mononucleotide Reduces Oxidative Stress and Improves Steroidogenesis in Granulosa Cells Associated with Sheep Prolificacy via Activating AMPK Pathway. Antioxidants (Basel) 2024; 14:34. [PMID: 39857368 PMCID: PMC11762531 DOI: 10.3390/antiox14010034] [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: 11/10/2024] [Revised: 12/23/2024] [Accepted: 12/28/2024] [Indexed: 01/27/2025] Open
Abstract
Oxidative stress is a significant factor in the death of granulosa cells (GCs), leading to follicular atresia and consequently limiting the number of dominant follicles that can mature and ovulate within each follicular wave. Follicular fluid contains a diverse array of metabolites that play crucial roles in regulating GCs' proliferation and oocyte maturation, which are essential for follicle development and female fertility. However, the mechanisms behind metabolite heterogeneity and its effects on GCs' function remain poorly understood. Here, we identified elevated nicotinamide levels in the follicular fluid of high-prolificacy sheep, correlated with oxidative stress in GCs, by an integrated analysis. In vitro experiments demonstrated that supplementation with β-nicotinamide mononucleotide (NMN) significantly increased the levels of nicotinamide adenine dinucleotide (NAD+) and adenosine triphosphate (ATP) in GCs. NMN treatment effectively reduced Lipopolysaccharide (LPS)-induced apoptosis and mitigated mitochondrial dysfunction, while also decreasing the production of reactive oxygen species (ROS), thereby enhancing the activity of the antioxidant defense system. Importantly, NMN treatment improved the impairments in steroid hormone levels induced by LPS. Mechanistically, the protective effects of NMN against GCs function were mediated via the AMPK/mTOR pathway. Collectively, our findings elucidate the metabolic characteristics associated with sheep prolificacy and demonstrate that NMN effectively protects GCs from LPS-induced dysfunction and enhances ovarian responsiveness via the AMPK/mTOR pathway. These findings also position NMN as a potential novel metabolic biomarker in enhancing ovarian function.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Yanli Zhang
- Jiangsu Livestock Embryo Engineering Laboratory, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (Y.C.); (H.Y.); (H.X.); (S.L.); (B.Z.); (Z.W.); (X.Y.); (F.W.)
| |
Collapse
|
9
|
Cochrane ALK, Murphy MP, Ozanne SE, Giussani DA. Pregnancy in obese women and mechanisms of increased cardiovascular risk in offspring. Eur Heart J 2024; 45:5127-5145. [PMID: 39508438 DOI: 10.1093/eurheartj/ehae671] [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: 04/04/2024] [Revised: 07/12/2024] [Accepted: 09/19/2024] [Indexed: 11/15/2024] Open
Abstract
Pregnancy complicated by maternal obesity contributes to an increased cardiovascular risk in offspring, which is increasingly concerning as the rates of obesity and cardiovascular disease are higher than ever before and still growing. There has been much research in humans and preclinical animal models to understand the impact of maternal obesity on offspring health. This review summarizes what is known about the offspring cardiovascular phenotype, describing a mechanistic role for oxidative stress, metabolic inflexibility, and mitochondrial dysfunction in mediating these impairments. It also discusses the impact of secondary postnatal insults, which may reveal latent cardiovascular deficits that originated in utero. Finally, current interventional efforts and gaps of knowledge to limit the developmental origins of cardiovascular dysfunction in offspring of obese pregnancy are highlighted.
Collapse
Affiliation(s)
- Anna L K Cochrane
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3EG, UK
- Department of Medicine, University of Cambridge, Hills Road, Cambridge CB2 0QQ, UK
| | - Michael P Murphy
- Department of Medicine, University of Cambridge, Hills Road, Cambridge CB2 0QQ, UK
- MRC Mitochondrial Biology Unit, University of Cambridge, Hills Road, Cambridge CB2 0XY, UK
| | - Susan E Ozanne
- Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- Loke Centre for Trophoblast Research, University of Cambridge, Downing Street, Cambridge CB2 3EG, UK
- Cambridge Strategic Research Initiative in Reproduction, University of Cambridge, Cambridge, UK
- British Heart Foundation, Cambridge Cardiovascular Centre for Research Excellence, University of Cambridge, Cambridge, UK
| | - Dino A Giussani
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3EG, UK
- Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- Loke Centre for Trophoblast Research, University of Cambridge, Downing Street, Cambridge CB2 3EG, UK
- Cambridge Strategic Research Initiative in Reproduction, University of Cambridge, Cambridge, UK
- British Heart Foundation, Cambridge Cardiovascular Centre for Research Excellence, University of Cambridge, Cambridge, UK
| |
Collapse
|
10
|
Viola I, Accornero P, Manenti I, Miretti S, Baratta M, Toschi P. mTOR is an essential gate in adapting the functional response of ovine trophoblast cells under stress-inducing environments. Placenta 2024; 158:14-22. [PMID: 39341011 DOI: 10.1016/j.placenta.2024.09.011] [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: 11/14/2023] [Revised: 09/13/2024] [Accepted: 09/15/2024] [Indexed: 09/30/2024]
Abstract
INTRODUCTION During the early stage of pregnancy trophoblast cells adapt to adverse uterine environments characterized by oxygen and nutrient deprivation. Autophagy is an intracellular degradation process that aims to promote cell survival in response to stressful conditions. Autophagy activation passes through the mechanistic target of rapamycin (mTOR), also known as a placental nutrient sensor. Here, we tested the hypothesis that ovine trophoblast cells may adapt to a suboptimal environment through an mTOR dependent regulation of cell survival with relevant implications for key placental functionality. METHODS Primary ovine trophoblast cells subjected to mTOR inhibitor and low-nutrient conditions were used to explore how autophagy affects cellular functionality and expression of solute carriers' genes (SLCs). RESULTS Autophagy activation was confirmed both in rapamycin-treated and low-nutrient conditions, through the detection of specific autophagic markers. However, p-mTOR activation seems to be severely modified only following rapamycin treatment whereas 24h of starvation allowed p-mTOR reactivation. Starvation promoted migration compared to normal culture conditions whereas all trophoblast functional activities were decreased in rapamycin treatment. Interestingly in both conditions, the autophagy-activated environment did not affect the progesterone release. mRNA expression of amino acid transporters remains largely undisturbed except for SLC43A2 and SLC38A4 which are downregulated in starved and rapamycin-treated cells, respectively. DISCUSSION The study demonstrates that sheep trophoblast cells can adapt to adverse conditions in the early stage of placentation by balancing, in an mTOR dependent manner, nutrient recycling and transport with relevant effects for in vitro functional properties, which could potentially impact conceptus development and survival.
Collapse
Affiliation(s)
- Irene Viola
- Department of Veterinary Sciences, University of Turin, Largo Paolo Braccini 2, Grugliasco, 10095, Italy.
| | - Paolo Accornero
- Department of Veterinary Sciences, University of Turin, Largo Paolo Braccini 2, Grugliasco, 10095, Italy.
| | - Isabella Manenti
- Department of Veterinary Sciences, University of Turin, Largo Paolo Braccini 2, Grugliasco, 10095, Italy.
| | - Silvia Miretti
- Department of Veterinary Sciences, University of Turin, Largo Paolo Braccini 2, Grugliasco, 10095, Italy.
| | - Mario Baratta
- Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parco Area delle Scienze 11a, 43124, Parma, Italy.
| | - Paola Toschi
- Department of Veterinary Sciences, University of Turin, Largo Paolo Braccini 2, Grugliasco, 10095, Italy.
| |
Collapse
|
11
|
Collins B, Lemanski EA, Wright-Jin E. The Importance of Including Maternal Immune Activation in Animal Models of Hypoxic-Ischemic Encephalopathy. Biomedicines 2024; 12:2559. [PMID: 39595123 PMCID: PMC11591850 DOI: 10.3390/biomedicines12112559] [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: 09/10/2024] [Revised: 10/24/2024] [Accepted: 10/29/2024] [Indexed: 11/28/2024] Open
Abstract
Hypoxic-ischemic encephalopathy (HIE) is a perinatal brain injury that is the leading cause of cerebral palsy, developmental delay, and poor cognitive outcomes in children born at term, occurring in about 1.5 out of 1000 births. The only proven therapy for HIE is therapeutic hypothermia. However, despite this treatment, many children ultimately suffer disability, brain injury, and even death. Barriers to implementation including late diagnosis and lack of resources also lead to poorer outcomes. This demonstrates a critical need for additional treatments for HIE, and to facilitate this, we need translational models that accurately reflect risk factors and interactions present in HIE. Maternal or amniotic infection is a significant risk factor and possible cause of HIE in humans. Maternal immune activation (MIA) is a well-established model of maternal infection and inflammation that has significant developmental consequences largely characterized within the context of neurodevelopmental disorders such as autism spectrum disorder and schizophrenia. MIA can also lead to long-lasting changes within the neuroimmune system, which lead to compounding negative outcomes following a second insult. This supports the importance of understanding the interaction of maternal inflammation and hypoxic-ischemic outcomes. Animal models have been invaluable to understanding the pathophysiology of this injury and to the development of therapeutic hypothermia. However, each model system has its own limitations. Large animal models such as pigs may more accurately represent the brain and organ development and complexity in humans, while rodent models are more cost-effective and offer more possible molecular techniques. Recent studies have utilized MIA or direct inflammation prior to HIE insult. Investigators should thoughtfully consider the risk factors they wish to include in their HIE animal models. In the incorporation of MIA, investigators should consider the type, timing, and dose of the inflammatory stimulus, as well as the timing, severity, and type of hypoxic insult. Using a variety of animal models that incorporate the maternal-placental-fetal system of inflammation will most likely lead to a more robust understanding of the mechanisms of this injury that can guide future clinical decisions and therapies.
Collapse
Affiliation(s)
- Bailey Collins
- Division of Biomedical Research, Nemours Children’s Health, Wilmington, DE 19803, USA; (B.C.); (E.A.L.)
- Psychological and Brain Sciences, University of Delaware, Newark, DE 19716, USA
| | - Elise A. Lemanski
- Division of Biomedical Research, Nemours Children’s Health, Wilmington, DE 19803, USA; (B.C.); (E.A.L.)
- Psychological and Brain Sciences, University of Delaware, Newark, DE 19716, USA
| | - Elizabeth Wright-Jin
- Psychological and Brain Sciences, University of Delaware, Newark, DE 19716, USA
- Division of Neurology, Nemours Children’s Health, Wilmington, DE 19803, USA
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
12
|
Rezaei N, Dormiani K, Kiani-Esfahani A, Mirdamadian S, Rahmani M, Jafarpour F, Nasr-Esfahani MH. Characterization and functional evaluation of goat PDX1 regulatory modules through comparative analysis of conserved interspecies homologs. Sci Rep 2024; 14:26755. [PMID: 39500950 PMCID: PMC11538457 DOI: 10.1038/s41598-024-77614-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 10/23/2024] [Indexed: 11/08/2024] Open
Abstract
PDX1 is a crucial transcription factor in pancreas development and mature β-cell function. However, the regulation of PDX1 expression in larger animals mirroring human pancreas morphogenesis and endocrine maturation remains poorly understood. Therefore, we conducted a comparative analysis to characterize regulatory regions of goat PDX1 gene and assessed their transcriptional activity by transient transfection of several transgenic EGFP constructs in β- and non-β cell lines. We recognized several highly conserved regions encompassing the promoter and cis-regulatory elements (Area I-IV) at 5' flanking sequence of the genes. Within the promoter, we identified that a key E-box and nearby CAAT element synergistically drive transcription, constituting the basal promoter of goat PDX1 gene. Furthermore, each recognized regulatory area separately enhances this basal promoter activity in β-cells compared to non-β cells; however, cooperatively, they exhibit a bifunctional regulatory effect on transcription. Additionally, the intact ~ 3 kb upstream region (Area I-III) functions as the most efficient reporter transgene in vitro and shows islet-specific expression in native rat pancreas. Together, our findings suggest that the regulation of goat PDX1 gene is governed by conserved regions similar to other mammals, while both structurally and functionally, these regions exhibit a closer resemblance to those found in humans.
Collapse
Affiliation(s)
- Naeimeh Rezaei
- Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Kianoush Dormiani
- Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran.
| | - Abbas Kiani-Esfahani
- Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Somayeh Mirdamadian
- Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Mohsen Rahmani
- Department of Animal Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Farnoosh Jafarpour
- Department of Animal Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Mohammad Hossein Nasr-Esfahani
- Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran.
- Department of Animal Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran.
| |
Collapse
|
13
|
Robinson JL, Roff AJ, Hammond SJ, Darby JRT, Meakin AS, Holman SL, Tai A, Moss TJM, Dimasi CG, Jesse SM, Wiese MD, Davies AN, Muhlhausler BS, Bischof RJ, Wallace MJ, Clifton VL, Morrison JL, Stark MJ, Gatford KL. Betamethasone improved near-term neonatal lamb lung maturation in experimental maternal asthma. Exp Physiol 2024; 109:1967-1979. [PMID: 39436639 PMCID: PMC11522833 DOI: 10.1113/ep091997] [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: 04/30/2024] [Accepted: 08/09/2024] [Indexed: 10/23/2024]
Abstract
Maternal asthma is associated with increased rates of neonatal lung disease, and fetuses from asthmatic ewes have fewer surfactant-producing cells and lower surfactant-protein B gene (SFTPB) expression than controls. Antenatal betamethasone increases lung surfactant production in preterm babies, and we therefore tested this therapy in experimental maternal asthma. Ewes were sensitised to house dust mite allergen, and an asthmatic phenotype induced by fortnightly allergen lung challenges; controls received saline. Pregnant asthmatic ewes were randomised to receive antenatal saline (asthma) or 12 mg intramuscular betamethasone (asthma+beta) at 138 and 139 days of gestation (term = 150 days). Lambs were delivered by Caesarean section at 140 days of gestation and ventilated for 45 min before tissue collection. Lung function and structure were similar in control lambs (n = 16, 11 ewes) and lambs from asthma ewes (n = 14, 9 ewes). Dynamic lung compliance was higher in lambs from asthma+beta ewes (n = 12, 8 ewes) compared to those from controls (P = 0.003) or asthma ewes (P = 0.008). Lung expression of surfactant protein genes SFTPA (P = 0.048) and SFTPB (P < 0.001), but not SFTPC (P = 0.177) or SFTPD (P = 0.285), was higher in lambs from asthma+beta than those from asthma ewes. Female lambs had higher tidal volume (P = 0.007), dynamic lung compliance (P < 0.001), and SFTPA (P = 0.037) and SFTPB gene expression (P = 0.030) than males. These data suggest that betamethasone stimulates lung maturation and function of near-term neonates, even in the absence of impairment by maternal asthma.
Collapse
Affiliation(s)
- Joshua L. Robinson
- Robinson Research InstituteUniversity of AdelaideAdelaideSouth AustraliaAustralia
- Adelaide Medical SchoolUniversity of AdelaideAdelaideSouth AustraliaAustralia
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, Clinical and Health SciencesUniversity of South AustraliaAdelaideSouth AustraliaAustralia
| | - Andrea J. Roff
- Robinson Research InstituteUniversity of AdelaideAdelaideSouth AustraliaAustralia
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, Clinical and Health SciencesUniversity of South AustraliaAdelaideSouth AustraliaAustralia
- School of BiomedicineUniversity of AdelaideAdelaideSouth AustraliaAustralia
| | - Sarah J. Hammond
- Robinson Research InstituteUniversity of AdelaideAdelaideSouth AustraliaAustralia
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, Clinical and Health SciencesUniversity of South AustraliaAdelaideSouth AustraliaAustralia
| | - Jack R. T. Darby
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, Clinical and Health SciencesUniversity of South AustraliaAdelaideSouth AustraliaAustralia
| | - Ashley S. Meakin
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, Clinical and Health SciencesUniversity of South AustraliaAdelaideSouth AustraliaAustralia
| | - Stacey L. Holman
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, Clinical and Health SciencesUniversity of South AustraliaAdelaideSouth AustraliaAustralia
| | - Andrew Tai
- Robinson Research InstituteUniversity of AdelaideAdelaideSouth AustraliaAustralia
- Adelaide Medical SchoolUniversity of AdelaideAdelaideSouth AustraliaAustralia
- Respiratory and Sleep MedicineWomen's & Children's HospitalNorth AdelaideSouth AustraliaAustralia
| | - Tim J. M. Moss
- Department of Obstetrics and GynaecologyMonash UniversityClaytonVictoriaAustralia
| | - Catherine G. Dimasi
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, Clinical and Health SciencesUniversity of South AustraliaAdelaideSouth AustraliaAustralia
| | - Sarah M. Jesse
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, Clinical and Health SciencesUniversity of South AustraliaAdelaideSouth AustraliaAustralia
| | - Michael D. Wiese
- Centre for Pharmaceutical Innovation, Clinical & Health SciencesUniversity of South AustraliaAdelaideSouth AustraliaAustralia
| | - Andrew N. Davies
- Biomedicine Discovery InstituteMonash UniversityFrankstonVictoriaAustralia
| | - Beverly S. Muhlhausler
- Health and BiosecurityCommonwealth Scientific and Industrial Research OrganisationAdelaideSouth AustraliaAustralia
| | - Robert J. Bischof
- Institute of Innovation, Science, and SustainabilityFederation University AustraliaBerwickVictoriaAustralia
| | - Megan J. Wallace
- Department of Obstetrics and GynaecologyMonash UniversityClaytonVictoriaAustralia
- The Ritchie CentreHudson Institute of Medical ResearchClaytonVictoriaAustralia
| | - Vicki L. Clifton
- Mater Medical Research InstituteUniversity of QueenslandSouth BrisbaneQueenslandAustralia
| | - Janna L. Morrison
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, Clinical and Health SciencesUniversity of South AustraliaAdelaideSouth AustraliaAustralia
| | - Michael J. Stark
- Robinson Research InstituteUniversity of AdelaideAdelaideSouth AustraliaAustralia
- Adelaide Medical SchoolUniversity of AdelaideAdelaideSouth AustraliaAustralia
- Department of Neonatal MedicineWomen's & Children's HospitalNorth AdelaideSouth AustraliaAustralia
| | - Kathryn L. Gatford
- Robinson Research InstituteUniversity of AdelaideAdelaideSouth AustraliaAustralia
- School of BiomedicineUniversity of AdelaideAdelaideSouth AustraliaAustralia
| |
Collapse
|
14
|
Kleinová M, Varga I, Čeháková M, Valent M, Klein M. Exploring the black box of human reproduction: endometrial organoids and assembloids - generation, implantation modeling, and future clinical perspectives. Front Cell Dev Biol 2024; 12:1482054. [PMID: 39507423 PMCID: PMC11539068 DOI: 10.3389/fcell.2024.1482054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Accepted: 10/09/2024] [Indexed: 11/08/2024] Open
Abstract
One of the critical processes in human reproduction that is still poorly understood is implantation. The implantation of an early human embryo is considered a significant limitation of successful pregnancy. Therefore, researchers are trying to develop an ideal model of endometrium in vitro that can mimic the endometrial micro-environment in vivo as much as possible. The ultimate goal of endometrial modeling is to study the molecular interactions at the embryo-maternal interface and to use this model as an in vitro diagnostic tool for infertility. Significant progress has been made over the years in generating such models. The first experiments of endometrial modeling involved animal models, which are undoubtedly valuable, but at the same time, their dissimilarities with human tissue represent a significant obstacle to further research. This fact led researchers to develop basic monolayer coculture systems using uterine cells obtained from biopsies and, later on, complex and multilayer coculture models. With successful tissue engineering methods and various cultivation systems, it is possible to form endometrial two-dimensional (2D) models to three-dimensional (3D) organoids and novel assembloids that can recapitulate many aspects of endometrial tissue architecture and cell composition. These organoids have already helped to provide new insight into the embryo-endometrium interplay. The main aim of this paper is a comprehensive review of past and current approaches to endometrial model generation, their feasibility, and potential clinical application for infertility treatment.
Collapse
Affiliation(s)
- Mária Kleinová
- Institute of Histology and Embryology, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovakia
| | - Ivan Varga
- Institute of Histology and Embryology, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovakia
| | - Michaela Čeháková
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovakia
| | - Martin Valent
- Department of Gynecology and Obstetrics, University Hospital Bratislava – Kramáre Workplace, Bratislava, Slovakia
| | - Martin Klein
- Institute of Histology and Embryology, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovakia
| |
Collapse
|
15
|
Rousseau-Ralliard D, Bozec J, Ouidir M, Jovanovic N, Gayrard V, Mellouk N, Dieudonné MN, Picard-Hagen N, Flores-Sanabria MJ, Jammes H, Philippat C, Couturier-Tarrade A. Short-Half-Life Chemicals: Maternal Exposure and Offspring Health Consequences-The Case of Synthetic Phenols, Parabens, and Phthalates. TOXICS 2024; 12:710. [PMID: 39453131 PMCID: PMC11511413 DOI: 10.3390/toxics12100710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/12/2024] [Accepted: 09/19/2024] [Indexed: 10/26/2024]
Abstract
Phenols, parabens, and phthalates (PPPs) are suspected or known endocrine disruptors. They are used in consumer products that pregnant women and their progeny are exposed to daily through the placenta, which could affect offspring health. This review aims to compile data from cohort studies and in vitro and in vivo models to provide a summary regarding placental transfer, fetoplacental development, and the predisposition to adult diseases resulting from maternal exposure to PPPs during the gestational period. In humans, using the concentration of pollutants in maternal urine, and taking the offspring sex into account, positive or negative associations have been observed concerning placental or newborn weight, children's BMI, blood pressure, gonadal function, or age at puberty. In animal models, without taking sex into account, alterations of placental structure and gene expression linked to hormones or DNA methylation were related to phenol exposure. At the postnatal stage, pollutants affect the bodyweight, the carbohydrate metabolism, the cardiovascular system, gonadal development, the age of puberty, sex/thyroid hormones, and gamete quality, but these effects depend on the age and sex. Future challenges will be to explore the effects of pollutants in mixtures using models and to identify the early signatures of in utero exposure capable of predicting the health trajectory of the offspring.
Collapse
Affiliation(s)
- Delphine Rousseau-Ralliard
- Université Paris-Saclay, UVSQ, INRAE, BREED, 78350 Jouy-en-Josas, France
- Ecole Nationale Vétérinaire d’Alfort, BREED, 94700 Maisons-Alfort, France
| | - Jeanne Bozec
- Université Paris-Saclay, UVSQ, INRAE, BREED, 78350 Jouy-en-Josas, France
- Ecole Nationale Vétérinaire d’Alfort, BREED, 94700 Maisons-Alfort, France
| | - Marion Ouidir
- University Grenoble Alpes, Inserm U1209, CNRS UMR 5309, Team of Environmental Epidemiology Applied to Development and Respiratory Health, Institute for Advanced Biosciences, 38000 Grenoble, France
| | - Nicolas Jovanovic
- University Grenoble Alpes, Inserm U1209, CNRS UMR 5309, Team of Environmental Epidemiology Applied to Development and Respiratory Health, Institute for Advanced Biosciences, 38000 Grenoble, France
| | - Véronique Gayrard
- ToxAlim (Research Center in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, 31062 Toulouse, France
| | - Namya Mellouk
- Université Paris-Saclay, UVSQ, INRAE, BREED, 78350 Jouy-en-Josas, France
- Ecole Nationale Vétérinaire d’Alfort, BREED, 94700 Maisons-Alfort, France
| | - Marie-Noëlle Dieudonné
- Université Paris-Saclay, UVSQ, INRAE, BREED, 78350 Jouy-en-Josas, France
- Ecole Nationale Vétérinaire d’Alfort, BREED, 94700 Maisons-Alfort, France
| | - Nicole Picard-Hagen
- ToxAlim (Research Center in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, 31062 Toulouse, France
| | - Maria-José Flores-Sanabria
- Université Paris-Saclay, UVSQ, INRAE, BREED, 78350 Jouy-en-Josas, France
- Ecole Nationale Vétérinaire d’Alfort, BREED, 94700 Maisons-Alfort, France
| | - Hélène Jammes
- Université Paris-Saclay, UVSQ, INRAE, BREED, 78350 Jouy-en-Josas, France
- Ecole Nationale Vétérinaire d’Alfort, BREED, 94700 Maisons-Alfort, France
| | - Claire Philippat
- University Grenoble Alpes, Inserm U1209, CNRS UMR 5309, Team of Environmental Epidemiology Applied to Development and Respiratory Health, Institute for Advanced Biosciences, 38000 Grenoble, France
| | - Anne Couturier-Tarrade
- Université Paris-Saclay, UVSQ, INRAE, BREED, 78350 Jouy-en-Josas, France
- Ecole Nationale Vétérinaire d’Alfort, BREED, 94700 Maisons-Alfort, France
| |
Collapse
|
16
|
Dimasi CG, Darby JR, Holman SL, Quinn M, Meakin AS, Seed M, Wiese MD, Morrison JL. Cardiac growth patterns and metabolism before and after birth in swine: Role of miR in proliferation, hypertrophy and metabolism. JOURNAL OF MOLECULAR AND CELLULAR CARDIOLOGY PLUS 2024; 9:100084. [PMID: 39803591 PMCID: PMC11708124 DOI: 10.1016/j.jmccpl.2024.100084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 07/09/2024] [Accepted: 07/15/2024] [Indexed: 01/06/2025]
Abstract
The adult mammalian heart is unable to undergo cardiac repair, limiting potential treatment options after cardiac damage. However, the fetal heart is capable of cardiac repair. In preparation for birth, cardiomyocytes (CMs) undergo major maturational changes that include exit from the cell cycle, hypertrophic growth, and mitochondrial maturation. The timing and regulation of such events in large mammals is not fully understood. In the present study, we aimed to assess this critical CM transition period using pigs as a preclinically relevant model. Left ventricular myocardium from Large White cross Landrace gilts was collected at 91, 98, 106 and 111-113 days gestation (d GA; term = 115d GA) and in piglets at 0-1, 4-5, 14-18, 19-20 days after birth. We found that miR-133a, which has known roles in CM proliferation, was significantly downregulated before birth, before rising postnatally. Likewise, gene expression of PCNA and CDK1 was repressed until birth with a rise postnatally, suggesting a decline in proliferation during late gestation followed by the onset of multinucleation in postnatal life. The timing of the switch in myocardial metabolism was unclear; however, complexes within the electron transport chain and mitochondrial biogenesis followed a similar pattern of decreasing abundance during late gestation and then a rise postnatally. These data suggest that CM maturation events such as cell cycle arrest and mitochondrial maturation occur around birth. These results may prove important to consider for preclinical applications such as the development of new therapeutics for cardiac repair.
Collapse
Affiliation(s)
- Catherine G. Dimasi
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA 5001, Australia
| | - Jack R.T. Darby
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA 5001, Australia
| | - Stacey L. Holman
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA 5001, Australia
| | - Megan Quinn
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA 5001, Australia
| | - Ashley S. Meakin
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA 5001, Australia
| | - Mike Seed
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- The Hospital for Sick Children, Division of Cardiology, Toronto, ON, Canada
- Research Institute, The Hospital for Sick Children, 686 Bay Street, Toronto M5G0A4, Canada
| | - Michael D. Wiese
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA 5001, Australia
| | - Janna L. Morrison
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA 5001, Australia
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Research Institute, The Hospital for Sick Children, 686 Bay Street, Toronto M5G0A4, Canada
| |
Collapse
|
17
|
Mills A, Nassabeh S, Hurley A, Shouldis L, Chantler PD, Dakhlallah D, Olfert IM. Influence of gestational window on offspring vascular health in rodents with in utero exposure to electronic cigarettes. J Physiol 2024; 602:4271-4289. [PMID: 39106241 PMCID: PMC11376404 DOI: 10.1113/jp286493] [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: 03/05/2024] [Accepted: 07/04/2024] [Indexed: 08/09/2024] Open
Abstract
Studies have shown cerebrovascular dysfunction in offspring with full-gestational electronic cigarette (Ecig) exposure, but little is known about how individual trimester exposure impacts offspring health. This study aimed to determine if there is a critical window during gestation that contributes to vascular and anxiety-like behavioural changes seen with full-term exposure. To test this, rats were time-mated, and the pregnant dams were randomly assigned to Ecig exposure during first trimester (gestational day, GD2-7), second trimester (GD8-14), third trimester (GD15-21) or full-term gestation (GD2-21). We also assessed the effect of maternal preconception exposure. Both male and female offspring from all maternal exposure conditions were compared to offspring from dams under ambient air (control) conditions. Ecig exposure consisted of 60-puffs/day (5 days/week) using either 5 or 30 watts for each respective exposure group. We found that maternal exposure to Ecig in the second and third trimesters resulted in a decrease (23-38%) in vascular reactivity of the middle cerebral artery (MCA) reactivity in 3- and 6-month-old offspring compared to Air offspring. Further, the severity of impairment was comparable to the full-term exposure (31-46%). Offspring also displayed changes in body composition, body mass, anxiety-like behaviour and locomotor activity, indicating that Ecigs influence neurodevelopment and metabolism. Maternal preconception exposure showed no impact on offspring body mass, anxiety-like behaviour, or vascular function. Thus, the critical exposure window where Ecig affects vascular development in offspring occurs during mid- to late-gestation in pregnancy, and both 5 W and 30 W exposure produce significant vascular dysfunction compared to Air. KEY POINTS: Exposure to electronic cigarettes (Ecigs) is known to increase risk factors for cardiovascular disease in both animals and humans. Maternal Ecig use during pregnancy in rodents is found to impair the vascular health of adolescent and adult offspring, but the critical gestation window for Ecig-induced vascular impairment is not known. This study demonstrates Ecig exposure during mid- and late-gestation (i.e. second or third trimester) results in impaired endothelial cell-mediated dilatation (i.e. middle cerebral artery reactivity) and alters anxiety-like behaviour in offspring. Maternal exposure prior to conception did not impact offspring's vascular or anxiety-like behavioural outcomes. Rodent models have been a reliable and useful predictor of inhalation-induced harm to humans. These data indicate maternal use of Ecigs during pregnancy should not be considered safe, and begin to inform clinicians and women about potential long-term harm to their offspring.
Collapse
Affiliation(s)
- Amber Mills
- Dept. of Physiology, Pharmacology & Toxicology, West Virginia University School of Medicine, Morgantown, WV 26506
- Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, WV 26506
| | - Sydney Nassabeh
- Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, WV 26506
| | - Audra Hurley
- Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, WV 26506
| | - Lainey Shouldis
- Department of Immunology and Microbial Pathogenesis, West Virginia University School of Medicine, Morgantown, WV 26506
| | - Paul D. Chantler
- Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, WV 26506
- Dept. of Neuroscience, West Virginia University School of Medicine, Morgantown, WV 26506
| | - Duaa Dakhlallah
- Dept. of Medicine, West Virginia University School of Medicine, Morgantown, WV 26506
| | - I. Mark Olfert
- Dept. of Physiology, Pharmacology & Toxicology, West Virginia University School of Medicine, Morgantown, WV 26506
- Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, WV 26506
- Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, WV 26506
| |
Collapse
|
18
|
Davies BP, Crew RC, Cochrane ALK, Davies K, Figueiredo Baptista A, Jeckel S, McCrone IS, Niu Y, Strugnell BW, Waine K, Fowden AL, Bryant CE, Wills JW, Giussani DA, Hughes K. An ovine model for investigation of the microenvironment of the male mammary gland. J Anat 2024; 245:405-419. [PMID: 38735860 PMCID: PMC11306760 DOI: 10.1111/joa.14055] [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: 12/17/2023] [Revised: 04/19/2024] [Accepted: 04/22/2024] [Indexed: 05/14/2024] Open
Abstract
The specific biology of the male breast remains relatively unexplored in spite of the increasing global prevalence of male breast cancer. Delineation of the microenvironment of the male breast is restricted by the low availability of human samples and a lack of characterisation of appropriate animal models. Unlike the mouse, the male ovine gland persists postnatally. We suggest that the male ovine mammary gland constitutes a promising adjunctive model for the male breast. In this study, we evaluate the male ovine mammary gland microenvironment, comparing intact and neutered males. Assessment of the glandular histo-anatomy highlights the resemblance of the male gland to that of neonatal female sheep and confirms the presence of rudimentary terminal duct lobular units. Irrespective of neutered status, cell proliferation in epithelial and stromal compartments is similarly low in males, and cell proliferation in epithelial cells and in the intralobular stroma is significantly lower than in pubertal female sheep. Between 42% and 72% of the luminal mammary epithelial cells in the male gland express the androgen receptor and expression is significantly reduced by neutering. Luminal epithelial cells within the intact and neutered male gland also express oestrogen receptor alpha, but minimal progesterone receptor expression is observed. The distribution of leukocytes within the ducts and stroma is similar to the mammary gland of female sheep and females of other species. Both macrophages and T lymphocytes are intercalated in the epithelial bilayer and are more abundant in the intralobular stroma than the interlobular stroma, suggesting that they may have a protective immunological function within the vestigial glandular tissue of the male sheep. Mast cells are also observed within the stroma. These cells cluster near the glandular tissue and are frequently located adjacent to blood vessels. The abundance of mast cells is significantly higher in intact males compared to neutered males, suggesting that hormone signalling may impact mast cell recruitment. In this study, we demonstrate the utility of the male ovine mammary gland as a model for furthering our knowledge of postnatal male mammary biology.
Collapse
Affiliation(s)
| | - Rachael C. Crew
- Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
- Department of Obstetrics and GynaecologyUniversity of CambridgeCambridgeUK
- School of Human SciencesThe University of Western AustraliaPerthWestern AustraliaAustralia
| | - Anna L. K. Cochrane
- Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
| | - Katie Davies
- Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
| | | | - Sonja Jeckel
- Farm Animal Pathology and DiagnosticsThe Royal Veterinary CollegeHatfieldUK
| | - Ian S. McCrone
- Department of Veterinary MedicineUniversity of CambridgeCambridgeUK
| | - Youguo Niu
- Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
| | | | - Katie Waine
- Farm Post Mortems LtdDurhamUK
- Present address:
Faculty of Veterinary MedicineUniversity of CalgaryCalgaryAlbertaCanada.
| | - Abigail L. Fowden
- Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
| | - Clare E. Bryant
- Department of Veterinary MedicineUniversity of CambridgeCambridgeUK
| | - John W. Wills
- Department of Veterinary MedicineUniversity of CambridgeCambridgeUK
| | - Dino A. Giussani
- Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
| | - Katherine Hughes
- Department of Veterinary MedicineUniversity of CambridgeCambridgeUK
| |
Collapse
|
19
|
Martin S, Peiro JL, Oria M, Forde B. Comparison of Amnio-Exchange With a Novel Synthetic Amniotic Fluid Versus Commercially Used Fluids for Fetal Therapy: An In Vivo Rodent Model. Prenat Diagn 2024; 44:1242-1250. [PMID: 39123304 DOI: 10.1002/pd.6644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 07/23/2024] [Accepted: 07/28/2024] [Indexed: 08/12/2024]
Abstract
OBJECTIVE Normal Saline (NS) and Lactated Ringer's (LR) damage human amniotic epithelium in vitro when compared with a synthetic amniotic fluid (Amnio-well, AW). We sought to evaluate the effect of amnio-exchange with NS, LR, and AW in vivo. METHODS On day E17.5, pregnant rats underwent amnio-exchange with NS, LR, or AW. Fetuses in each pregnant rat that did not undergo amnio-exchange acted as controls. Amnions were harvested at E20.5 and ultrastructure evaluated via electron microscopy. Protein levels of cleaved matrix metalloproteinase 9 (MMP9) and collagen 1 (Col1a) were evaluated via Western Blot. Connexin-43 expression was evaluated via immunofluorescence (IF). RESULTS There was an increase in amnion microfractures and epithelial cellular shrinkage with NS and LR compared with control and AW. The cleaved MMP9/Col1 ratio was increased 3.9-fold in NS (p < 0.001) and 4.5-fold LR (p = 0.0201) relative to control, whereas AW expression was similar to control (p = 0.636). Connexin-43 was also increased on IF in NS and LR relative to AW (mean gray intensity 26.5 ± 4.5, 26.5 ± 6.7, 19.2 ± 3.4, p < 0.001). CONCLUSION Amnio-exchange with NS and LR led to increased amniotic microfractures and collagen degradation compared with synthetic amniotic fluid. Larger models are warranted to validate or refute these findings.
Collapse
Affiliation(s)
- Samuel Martin
- Center for Fetal and Placental Research, Cincinnati Children's Hospital Medical Center (CCHMC), Cincinnati, Ohio, USA
| | - Jose L Peiro
- Center for Fetal and Placental Research, Cincinnati Children's Hospital Medical Center (CCHMC), Cincinnati, Ohio, USA
- Division of General and Thoracic, Department of Pediatric Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Marc Oria
- Department of Radiation Oncology, University of Cincinnati Medical Center, Cincinnati, Ohio, USA
| | - Braxton Forde
- Center for Fetal and Placental Research, Cincinnati Children's Hospital Medical Center (CCHMC), Cincinnati, Ohio, USA
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, University of Cincinnati Medical Center, Cincinnati, Ohio, USA
| |
Collapse
|
20
|
Alkhatib B, Ciarelli J, Ghnenis A, Pallas B, Olivier N, Padmanabhan V, Vyas AK. Early- to mid-gestational testosterone excess leads to adverse cardiac outcomes in postpartum sheep. Am J Physiol Heart Circ Physiol 2024; 327:H315-H330. [PMID: 38819385 PMCID: PMC11687963 DOI: 10.1152/ajpheart.00763.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 05/13/2024] [Accepted: 05/22/2024] [Indexed: 06/01/2024]
Abstract
Cardiovascular dysfunctions complicate 10-20% of pregnancies, increasing the risk for postpartum mortality. Various gestational insults, including preeclampsia are reported to be associated with adverse maternal cardiovascular outcomes. One such insult, gestational hyperandrogenism increases the risk for preeclampsia and other gestational morbidities but its impact on postpartum maternal health is not well known. We hypothesize that gestational hyperandrogenism such as testosterone (T) excess will adversely impact the maternal heart in the postpartum period. Pregnant ewes were injected with T propionate from day 30 to day 90 of gestation (term 147 days). Three months postpartum, echocardiograms, plasma cytokine profiles, cardiac morphometric, and molecular analysis were conducted [control (C) n = 6, T-treated (T) n = 7 number of animals]. Data were analyzed by two-tailed Student's t test and Cohen's effect size (d) analysis. There was a nonsignificant large magnitude decrease in cardiac output (7.64 ± 1.27 L/min vs. 10.19 ± 1.40, P = 0.22, d = 0.81) and fractional shortening in the T ewes compared with C (35.83 ± 2.33% vs. 41.50 ± 2.84, P = 0.15, d = 0.89). T treatment significantly increased 1) left ventricle (LV) weight-to-body weight ratio (2.82 ± 0.14 g/kg vs. 2.46 ± 0.08) and LV thickness (14.56 ± 0.52 mm vs. 12.50 ± 0.75), 2) proinflammatory marker [tumor necrosis factor-alpha (TNF-α)] in LV (1.66 ± 0.35 vs. 1.06 ± 0.18), 3) LV collagen (Masson's Trichrome stain: 3.38 ± 0.35 vs. 1.49 ± 0.15 and Picrosirius red stain: 5.50 ± 0.32 vs. 3.01 ± 0.23), 4) markers of LV apoptosis, including TUNEL (8.3 ± 1.1 vs. 0.9 ± 0.18), bcl-2-associated X protein (Bax)+-to-b-cell lymphoma 2 (Bcl2)+ ratio (0.68 ± 0.30 vs. 0.13 ± 0.02), and cleaved caspase 3 (15.4 ± 1.7 vs. 4.4 ± 0.38). These findings suggest that gestational testosterone excess adversely programs the maternal LV, leading to adverse structural and functional consequences in the postpartum period.NEW & NOTEWORTHY Using a sheep model of human translational relevance, this study provides evidence that excess gestational testosterone exposure such as that seen in hyperandrogenic disorders adversely impacts postpartum maternal hearts.
Collapse
Affiliation(s)
- Bashar Alkhatib
- Department of Pediatrics, Washington University, St Louis, Missouri, United States
| | - Joseph Ciarelli
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan, United States
| | - Adel Ghnenis
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan, United States
| | - Brooke Pallas
- Unit for Laboratory Animal Medicine, University of Michigan, Ann Arbor, Michigan, United States
| | - Nicholas Olivier
- Department of Veterinary Medicine, Michigan State University, Lansing, Michigan, United States
| | - Vasantha Padmanabhan
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan, United States
| | - Arpita Kalla Vyas
- Department of Pediatrics, Washington University, St Louis, Missouri, United States
| |
Collapse
|
21
|
Zhang S, Lock MC, Tie M, McMillen IC, Botting KJ, Morrison JL. Cardiac programming in the placentally restricted sheep fetus in early gestation. J Physiol 2024; 602:3815-3832. [PMID: 38975864 DOI: 10.1113/jp286702] [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: 04/08/2024] [Accepted: 06/13/2024] [Indexed: 07/09/2024] Open
Abstract
Fetal growth restriction (FGR) occurs in 8% of human pregnancies, and the growth restricted newborn is at a greater risk of developing heart disease in later adult life. In sheep, experimental restriction of placental growth (PR) from conception results in FGR, a decrease in cardiomyocyte endowment and an upregulation of pathological hypertrophic signalling in the fetal heart in late gestation. However, there is no change in the expression of markers of cellular proliferation nor in the level of cardiomyocyte apoptosis in the heart of the PR fetus in late gestation. This suggests that FGR arises early in gestation and programs a decrease in cardiomyocyte endowment in early, rather than late, gestation. Here, control and PR fetal sheep were humanely killed at 55 days' gestation (term, 150 days). Fetal body and heart weight were lower in PR compared with control fetuses and there was evidence of sparing of fetal brain growth. While there was no change in the proportion of cardiomyocytes that were proliferating in the early gestation PR heart, there was an increase in measures of apoptosis, and markers of autophagy and pathological hypertrophy in the PR fetal heart. These changes in early gestation highlight that FGR is associated with evidence of early cell death and compensatory hypertrophic responses of cardiomyocytes in the fetal heart. The data suggest that early placental restriction results in a decrease in the pool of proliferative cardiomyocytes in early gestation, which would limit cardiomyocyte endowment in the heart of the PR fetus in late gestation. KEY POINTS: Placental restriction leading to fetal growth restriction (FGR) and chronic fetal hypoxaemia in sheep results in a decrease in cardiomyocyte endowment in late gestation. FGR did not change cardiomyocyte proliferation during early gestation but did result in increased apoptosis and markers of autophagy in the fetal heart, which may result in the decreased endowment of cardiomyocytes observed in late gestation. FGR in early gestation also results in increased hypoxia inducible factor signalling in the fetal heart, which in turn may result in the altered expression of epigenetic regulators, increased expression of insulin-like growth factor 2 and cardiomyocyte hypertrophy during late gestation and after birth.
Collapse
Affiliation(s)
- Song Zhang
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Science, University of South Australia, Adelaide, SA, Australia
| | - Mitchell C Lock
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Science, University of South Australia, Adelaide, SA, Australia
| | - Michelle Tie
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Science, University of South Australia, Adelaide, SA, Australia
| | - I Caroline McMillen
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Science, University of South Australia, Adelaide, SA, Australia
| | - Kimberley J Botting
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Science, University of South Australia, Adelaide, SA, Australia
| | - Janna L Morrison
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Science, University of South Australia, Adelaide, SA, Australia
| |
Collapse
|
22
|
Moses RM, Stenhouse C, Halloran KM, Sah N, Newton MG, Hoskins EC, Washburn SE, Johnson GA, Wu G, Bazer FW. Metabolic pathways of glucose and fructose: II Spatiotemporal expression of genes involved in synthesis and transport of lactate in ovine conceptuses†. Biol Reprod 2024; 111:159-173. [PMID: 38531778 DOI: 10.1093/biolre/ioae047] [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: 12/06/2023] [Revised: 02/14/2024] [Accepted: 03/22/2024] [Indexed: 03/28/2024] Open
Abstract
Lactate, an abundant molecule in fetal fluids and blood of mammalian species, is often overlooked as a metabolic waste product generated during pregnancy. Most of the glucose and fructose consumed by ovine conceptuses is converted to lactate, but proteins involved in lactate metabolism and transport have not been investigated. This study characterized total lactate produced by ovine conceptuses throughout gestation, as well as expression of mRNAs and proteins involved in lactate metabolism. Lactate increased in abundance in the uterine lumen during the preimplantation period and was more abundant than pyruvate. The abundance of lactate in allantoic and amniotic fluids increased with advancing days of gestation and most abundant on Day 125 of pregnancy (P < 0.05). Lactate dehydrogenase subunits A (converts pyruvate to lactate) and B (converts lactate to pyruvate) were expressed by conceptuses throughout gestation. Lactate is transported via monocarboxylic acid transporters SLC16A1 and SLC16A3, both of which were expressed by the conceptus throughout gestation. Additionally, the interplacentomal chorioallantois from Day 126 expressed SLC16A1 and SLC16A3 and transported lactate across the tissue. Hydrocarboxylic acid receptor 1 (HCAR1), a receptor for lactate, was localized to the uterine luminal and superficial glandular epithelia of pregnant ewes throughout gestation and conceptus trophectoderm during the peri-implantation period of gestation. These results provide novel insights into the spatiotemporal profiles of enzymes, transporters, and receptor for lactate by ovine conceptuses throughout pregnancy.
Collapse
Affiliation(s)
- Robyn M Moses
- Department of Animal Science, Texas A&M University, College Station, TX, USA
| | - Claire Stenhouse
- Department of Animal Science, Pennsylvania State University, University Park, PA, USA
| | - Katherine M Halloran
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor MI, USA
| | - Nirvay Sah
- Department of Pathology, University of California-San Diego, San Diego, CA, USA
| | - Makenzie G Newton
- Department of Animal Science, Texas A&M University, College Station, TX, USA
| | - Emily C Hoskins
- College of Biomedical and Diagnostic Sciences, University of Tennessee, Knoxville, TN, USA
| | - Shannon E Washburn
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX, USA
| | - Gregory A Johnson
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, USA
| | - Guoyao Wu
- Department of Animal Science, Texas A&M University, College Station, TX, USA
| | - Fuller W Bazer
- Department of Animal Science, Texas A&M University, College Station, TX, USA
| |
Collapse
|
23
|
Navarrete Á, Inostroza M, Utrera A, Bezmalinovic A, González-Candia A, Rivera E, Godoy-Guzmán C, Herrera EA, García-Herrera C. Biomechanical effects of hemin and sildenafil treatments on the aortic wall of chronic-hypoxic lambs. Front Bioeng Biotechnol 2024; 12:1406214. [PMID: 39021365 PMCID: PMC11252865 DOI: 10.3389/fbioe.2024.1406214] [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: 03/24/2024] [Accepted: 06/07/2024] [Indexed: 07/20/2024] Open
Abstract
Introduction: Gestation under chronic hypoxia causes pulmonary hypertension, cardiovascular remodeling, and increased aortic stiffness in the offspring. To mitigate the neonatal cardiovascular risk, pharmacological treatments (such as hemin and sildenafil) have been proposed to improve pulmonary vasodilation. However, little is known about the effects of these treatments on the aorta. Therefore, we studied the effect of hemin and sildenafil treatments in the aorta of lambs gestated and raised at highlands, thereby subjected to chronic hypoxia. Methods: Several biomechanical tests were conducted in the descending thoracic aorta (DTA) and the distal abdominal aorta (DAA), assessing 3 groups of study of hypoxic animals: non-treated (Control) and treated either with hemin or sildenafil. Based on them, the stiffness level has been quantified in both zones, along with the physiological strain in the unloaded aortic duct. Furthermore, a morphological study by histology was conducted in the DTA. Results: Biomechanical results indicate that treatments trigger an increment of axial pre-stress and circumferential residual stress levels in DTA and DAA of lambs exposed to high-altitude chronic hypoxia, which reveals a vasodilatation improvement along with an anti-hypertensive response under this characteristic environmental condition. In addition, histological findings do not reveal significant differences in either structure or microstructural content. Discussion: The biomechanics approach emerges as a valuable study perspective, providing insights to explain the physiological mechanisms of vascular function. According to established results, alterations in the function of the aortic wall may not necessarily be explained by morphostructural changes, but rather by the characteristic mechanical state of the microstructural components that are part of the studied tissue. In this sense, the reported biomechanical changes are beneficial in mitigating the adverse effects of hypobaric hypoxia exposure during gestation and early postnatal life.
Collapse
Affiliation(s)
- Álvaro Navarrete
- Laboratorio de Biomecánica y Biomateriales, Departamento de Ingeniería Mecánica, Universidad de Santiago de Chile, USACH, Santiago de Chile, Chile
| | - Matías Inostroza
- Laboratorio de Biomecánica y Biomateriales, Departamento de Ingeniería Mecánica, Universidad de Santiago de Chile, USACH, Santiago de Chile, Chile
| | - Andrés Utrera
- Laboratorio de Biomecánica y Biomateriales, Departamento de Ingeniería Mecánica, Universidad de Santiago de Chile, USACH, Santiago de Chile, Chile
| | - Alejandro Bezmalinovic
- Laboratorio de Biomecánica y Biomateriales, Departamento de Ingeniería Mecánica, Universidad de Santiago de Chile, USACH, Santiago de Chile, Chile
| | | | - Eugenio Rivera
- Laboratorio de Biomecánica y Biomateriales, Departamento de Ingeniería Mecánica, Universidad de Santiago de Chile, USACH, Santiago de Chile, Chile
| | - Carlos Godoy-Guzmán
- Laboratorio de Ingeniería de Tejidos, Centro de Investigación Biomédica y Aplicada (CIBAP), Escuela de Medicina, Universidad de Santiago de Chile, Santiago de Chile, Chile
| | - Emilio A. Herrera
- Pathophysiology Program, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago, Chile
- International Center for Andean Studies (INCAS), Universidad de Chile, Santiago, Chile
| | - Claudio García-Herrera
- Laboratorio de Biomecánica y Biomateriales, Departamento de Ingeniería Mecánica, Universidad de Santiago de Chile, USACH, Santiago de Chile, Chile
| |
Collapse
|
24
|
Darby JRT, Saini BS, Holman SL, Hammond SJ, Perumal SR, Macgowan CK, Seed M, Morrison JL. Acute-on-chronic: using magnetic resonance imaging to disentangle the haemodynamic responses to acute and chronic fetal hypoxaemia. Front Med (Lausanne) 2024; 11:1340012. [PMID: 38933113 PMCID: PMC11199546 DOI: 10.3389/fmed.2024.1340012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 06/04/2024] [Indexed: 06/28/2024] Open
Abstract
Introduction The fetal haemodynamic response to acute episodes of hypoxaemia are well characterised. However, how these responses change when the hypoxaemia becomes more chronic in nature such as that associated with fetal growth restriction (FGR), is less well understood. Herein, we utilised a combination of clinically relevant MRI techniques to comprehensively characterize and differentiate the haemodynamic responses occurring during acute and chronic periods of fetal hypoxaemia. Methods Prior to conception, carunclectomy surgery was performed on non-pregnant ewes to induce FGR. At 108-110 days (d) gestational age (GA), pregnant ewes bearing control (n = 12) and FGR (n = 9) fetuses underwent fetal catheterisation surgery. At 117-119 days GA, ewes underwent MRI sessions where phase-contrast (PC) and T2 oximetry were used to measure blood flow and oxygenation, respectively, throughout the fetal circulation during a normoxia and then an acute hypoxia state. Results Fetal oxygen delivery (DO2) was lower in FGR fetuses than controls during the normoxia state but cerebral DO2 remained similar between fetal groups. Acute hypoxia reduced both overall fetal and cerebral DO2. FGR increased ductus venosus (DV) and foramen ovale (FO) blood flow during both the normoxia and acute hypoxia states. Pulmonary blood flow (PBF) was lower in FGR fetuses during the normoxia state but similar to controls during the acute hypoxia state when PBF in controls was decreased. Conclusion Despite a prevailing level of chronic hypoxaemia, the FGR fetus upregulates the preferential streaming of oxygen-rich blood via the DV-FO pathway to maintain cerebral DO2. However, this upregulation is unable to maintain cerebral DO2 during further exposure to an acute episode of hypoxaemia. The haemodynamic alterations required at the level of the liver and lung to allow the DV-FO pathway to maintain cerebral DO2, may have lasting consequences on hepatic function and pulmonary vascular regulation after birth.
Collapse
Affiliation(s)
- Jack R. T. Darby
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA, Australia
| | - Brahmdeep S. Saini
- Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Research Institute, Toronto, ON, Canada
| | - Stacey L. Holman
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA, Australia
| | - Sarah J. Hammond
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA, Australia
| | - Sunthara Rajan Perumal
- Preclinical, Imaging & Research Laboratories, South Australian Health & Medical Research Institute, Adelaide, SA, Australia
| | - Christopher K. Macgowan
- Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Research Institute, Toronto, ON, Canada
| | - Mike Seed
- Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Research Institute, Toronto, ON, Canada
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Janna L. Morrison
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA, Australia
- Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Research Institute, Toronto, ON, Canada
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
25
|
Yan Z, Yang J, Wei WT, Zhou ML, Mo DX, Wan X, Ma R, Wu MM, Huang JH, Liu YJ, Lv FH, Li MH. A time-resolved multi-omics atlas of transcriptional regulation in response to high-altitude hypoxia across whole-body tissues. Nat Commun 2024; 15:3970. [PMID: 38730227 PMCID: PMC11087590 DOI: 10.1038/s41467-024-48261-w] [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: 08/23/2023] [Accepted: 04/23/2024] [Indexed: 05/12/2024] Open
Abstract
High-altitude hypoxia acclimatization requires whole-body physiological regulation in highland immigrants, but the underlying genetic mechanism has not been clarified. Here we use sheep as an animal model for low-to-high altitude translocation. We generate multi-omics data including whole-genome sequences, time-resolved bulk RNA-Seq, ATAC-Seq and single-cell RNA-Seq from multiple tissues as well as phenotypic data from 20 bio-indicators. We characterize transcriptional changes of all genes in each tissue, and examine multi-tissue temporal dynamics and transcriptional interactions among genes. Particularly, we identify critical functional genes regulating the short response to hypoxia in each tissue (e.g., PARG in the cerebellum and HMOX1 in the colon). We further identify TAD-constrained cis-regulatory elements, which suppress the transcriptional activity of most genes under hypoxia. Phenotypic and transcriptional evidence indicate that antenatal hypoxia could improve hypoxia tolerance in offspring. Furthermore, we provide time-series expression data of candidate genes associated with human mountain sickness (e.g., BMPR2) and high-altitude adaptation (e.g., HIF1A). Our study provides valuable resources and insights for future hypoxia-related studies in mammals.
Collapse
Affiliation(s)
- Ze Yan
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, 100193, China
- College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Ji Yang
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, 100193, China
- College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Wen-Tian Wei
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, 100193, China
- College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Ming-Liang Zhou
- Sichuan Academy of Grassland Science, Chengdu, 611743, China
| | - Dong-Xin Mo
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, 100193, China
- College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Xing Wan
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, 100193, China
- College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Rui Ma
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, 100193, China
- College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Mei-Ming Wu
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, 100193, China
- College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Jia-Hui Huang
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, 100193, China
- College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Ya-Jing Liu
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, 100193, China
- College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Feng-Hua Lv
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, 100193, China
- College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Meng-Hua Li
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, 100193, China.
- College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
26
|
Holmes H, Saini BS, Moir OJ, Darby JRT, Morrison JL, Sun L, Seed M. Pulmonary Vascular Regulation in the Fetal and Transitional Lung. Clin Perinatol 2024; 51:1-19. [PMID: 38325936 DOI: 10.1016/j.clp.2023.11.003] [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] [Indexed: 02/09/2024]
Abstract
Fetal lungs have fewer and smaller arteries with higher pulmonary vascular resistance (PVR) than a newborn. As gestation advances, the pulmonary circulation becomes more sensitive to changes in pulmonary arterial oxygen tension, which prepares them for the dramatic drop in PVR and increase in pulmonary blood flow (PBF) that occur when the baby takes its first few breaths of air, thus driving the transition from fetal to postnatal circulation. Dynamic and intricate regulatory mechanisms control PBF throughout development and are essential in supporting gas exchange after birth. Understanding these concepts is crucial given the role the pulmonary vasculature plays in the development of complications with transition, such as in the setting of persistent pulmonary hypertension of the newborn and congenital heart disease. An improved understanding of pulmonary vascular regulation may reveal opportunities for better clinical management.
Collapse
Affiliation(s)
- Hannah Holmes
- Division of Cardiology, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, M5G 1X8, Canada
| | - Brahmdeep S Saini
- Division of Cardiology, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, M5G 1X8, Canada
| | - Olivia J Moir
- Division of Cardiology, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, M5G 1X8, Canada
| | - Jack R T Darby
- Early Origins of Adult Health Research Group, University of South Australia, Adelaide, South Australia, 5001, Australia
| | - Janna L Morrison
- Early Origins of Adult Health Research Group, University of South Australia, Adelaide, South Australia, 5001, Australia; Department of Physiology, Faculty of Medicine, University of Toronto, 555 University Avenue, Toronto, Ontario, M5G 1X8 Canada; Translational Medicine Program, The Hospital for Sick Children, University of Toronto, 555 University Avenue, Toronto, Ontario, M5G 1X8 Canada
| | - Liqun Sun
- Division of Cardiology, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, M5G 1X8, Canada
| | - Mike Seed
- Division of Cardiology, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, M5G 1X8, Canada; Department of Physiology, Faculty of Medicine, University of Toronto, 555 University Avenue, Toronto, Ontario, M5G 1X8 Canada; Translational Medicine Program, The Hospital for Sick Children, University of Toronto, 555 University Avenue, Toronto, Ontario, M5G 1X8 Canada; Research Institute, The Hospital for Sick Children, University of Toronto, 555 University Avenue, Toronto, Ontario, M5G 1X8 Canada; Department of Diagnostic Imaging, The Hospital for Sick Children, University of Toronto, 555 University Avenue, Toronto, Ontario, M5G 1X8 Canada.
| |
Collapse
|
27
|
Robinson JL, Gatford KL, Bailey DN, Roff AJ, Clifton VL, Morrison JL, Stark MJ. Preclinical models of maternal asthma and progeny outcomes: a scoping review. Eur Respir Rev 2024; 33:230174. [PMID: 38417970 PMCID: PMC10900068 DOI: 10.1183/16000617.0174-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 12/09/2023] [Indexed: 03/01/2024] Open
Abstract
There is an increased risk of adverse perinatal outcomes in the ∼17% of women with asthma during pregnancy. The mechanisms linking maternal asthma and adverse outcomes are largely unknown, but reflect joint effects of genetics and prenatal exposure to maternal asthma. Animal models are essential to understand the underlying mechanisms independent of genetics and comorbidities, and enable safe testing of interventions. This scoping review aimed to explore the methodology, phenotype, characteristics, outcomes and quality of published studies using preclinical maternal asthma models. MEDLINE (PubMed), Embase (Elsevier) and Web of Science were systematically searched using previously validated search strings for maternal asthma and for animal models. Two reviewers independently screened titles and abstracts, full texts, and then extracted and assessed the quality of each study using the Animal Research: Reporting of In Vivo Experiments (ARRIVE) 2.0 guidelines. Out of 3618 studies identified, 39 were eligible for extraction. Most studies were in rodents (86%) and all were models of allergic asthma. Maternal and progeny outcomes included airway hyperresponsiveness, airway resistance, inflammation, lung immune cells, lung structure and serum immunoglobulins and cytokines. Experimental design (100%), procedural details (97%) and rationale (100%) were most often reported. Conversely, data exclusion (21%), blinding (18%) and adverse events (8%) were reported in a minority of studies. Species differences in physiology and timing of development, the use of allergens not relevant to humans and a lack of comparable outcome measures may impede clinical translation. Future studies exploring models of maternal asthma should adhere to the minimum core outcomes set presented in this review.
Collapse
Affiliation(s)
- Joshua L Robinson
- Robinson Research Institute, University of Adelaide, Adelaide, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, Australia
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, Australia
| | - Kathryn L Gatford
- Robinson Research Institute, University of Adelaide, Adelaide, Australia
- School of Biomedicine, University of Adelaide, Adelaide, Australia
| | - Danielle N Bailey
- Robinson Research Institute, University of Adelaide, Adelaide, Australia
| | - Andrea J Roff
- Robinson Research Institute, University of Adelaide, Adelaide, Australia
- School of Biomedicine, University of Adelaide, Adelaide, Australia
| | - Vicki L Clifton
- Mater Research Institute, University of Queensland, Brisbane, Australia
| | - Janna L Morrison
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, Australia
| | - Michael J Stark
- Robinson Research Institute, University of Adelaide, Adelaide, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, Australia
- Department of Neonatal Medicine, Women's & Children's Hospital, Adelaide, Australia
| |
Collapse
|
28
|
Cao M, Kuthiala S, Jean KJ, Liu HL, Courchesne M, Nygard K, Burns P, Desrochers A, Fecteau G, Faure C, Frasch MG. The Vagus Nerve Regulates Immunometabolic Homeostasis in the Ovine Fetus near Term: The Impact on Terminal Ileum. BIOLOGY 2024; 13:38. [PMID: 38248469 PMCID: PMC10812930 DOI: 10.3390/biology13010038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 01/04/2024] [Accepted: 01/05/2024] [Indexed: 01/23/2024]
Abstract
BACKGROUND Glucosensing elements are widely distributed throughout the body and relay information about circulating glucose levels to the brain via the vagus nerve. However, while anatomical wiring has been established, little is known about the physiological role of the vagus nerve in glucosensing. The contribution of the vagus nerve to inflammation in the fetus is poorly understood. Increased glucose levels and inflammation act synergistically when causing organ injury, but their interplay remains incompletely understood. We hypothesized that vagotomy (Vx) will trigger a rise in systemic glucose levels and this will be enhanced during systemic and organ-specific inflammation. Efferent vagus nerve stimulation (VNS) should reverse this phenotype. METHODS Near-term fetal sheep (n = 57) were surgically prepared using vascular catheters and ECG electrodes as the control and treatment groups (lipopolysaccharide (LPS), Vx + LPS, Vx + LPS + selective efferent VNS). The experiment was started 72 h postoperatively to allow for post-surgical recovery. Inflammation was induced with LPS bolus intravenously (LPS group, 400 ng/fetus/day for 2 days; n = 23). For the Vx + LPS group (n = 11), a bilateral cervical vagotomy was performed during surgery; of these n = 5 received double the LPS dose, LPS800. The Vx + LPS + efferent VNS group (n = 8) received cervical VNS probes bilaterally distal from Vx in eight animals. Efferent VNS was administered for 20 min on days 1 and 2 +/10 min around the LPS bolus. Fetal arterial blood samples were drawn on each postoperative day of recovery (-72 h, -48 h, and -24 h) as well as at the baseline and seven selected time points (3-54 h) to profile inflammation (ELISA IL-6, pg/mL), insulin (ELISA), blood gas, and metabolism (glucose). At 54 h post-LPS, a necropsy was performed, and the terminal ileum macrophages' CD11c (M1 phenotype) immunofluorescence was quantified to detect inflammation. The results are reported for p < 0.05 and for Spearman R2 > 0.1. The results are presented as the median (IQR). RESULTS Across the treatment groups, blood gas and cardiovascular changes indicated mild septicemia. At 3 h in the LPS group, IL-6 peaked. That peak was decreased in the Vx + LPS400 group and doubled in the Vx + LPS800 group. The efferent VNS sped up the reduction in the inflammatory response profile over 54 h. The M1 macrophage activity was increased in the LPS and Vx + LPS800 groups only. The glucose and insulin concentrations in the Vx + LPS group were, respectively, 1.3-fold (throughout the experiment) and 2.3-fold higher vs. control (at 3 h). The efferent VNS normalized the glucose concentrations. CONCLUSIONS The complete withdrawal of vagal innervation resulted in a 72-h delayed onset of a sustained increase in glucose for at least 54 h and intermittent hyperinsulinemia. Under the conditions of moderate fetal inflammation, this was related to higher levels of gut inflammation. The efferent VNS reduced the systemic inflammatory response as well as restored both the concentrations of glucose and the degree of terminal ileum inflammation, but not the insulin concentrations. Supporting our hypothesis, these findings revealed a novel regulatory, hormetic, role of the vagus nerve in the immunometabolic response to endotoxin in near-term fetuses.
Collapse
Affiliation(s)
- Mingju Cao
- Department of Obstetrics and Gynaecology and Department of Neurosciences, CHU Ste-Justine Research Centre, Université de Montréal, Montréal, QC H3T 1C5, Canada; (M.C.)
| | - Shikha Kuthiala
- Department of Obstetrics and Gynaecology and Department of Neurosciences, CHU Ste-Justine Research Centre, Université de Montréal, Montréal, QC H3T 1C5, Canada; (M.C.)
| | - Keven Jason Jean
- Department of Obstetrics and Gynaecology and Department of Neurosciences, CHU Ste-Justine Research Centre, Université de Montréal, Montréal, QC H3T 1C5, Canada; (M.C.)
| | - Hai Lun Liu
- Department of Obstetrics and Gynaecology and Department of Neurosciences, CHU Ste-Justine Research Centre, Université de Montréal, Montréal, QC H3T 1C5, Canada; (M.C.)
| | - Marc Courchesne
- Biotron Microscopy, Western University, London, ON N6A 3K7, Canada
| | - Karen Nygard
- Biotron Microscopy, Western University, London, ON N6A 3K7, Canada
| | - Patrick Burns
- Clinical Sciences, CHUV, Université de Montréal, St-Hyacinthe, QC J2S 2M2, Canada (A.D.)
| | - André Desrochers
- Clinical Sciences, CHUV, Université de Montréal, St-Hyacinthe, QC J2S 2M2, Canada (A.D.)
| | - Gilles Fecteau
- Clinical Sciences, CHUV, Université de Montréal, St-Hyacinthe, QC J2S 2M2, Canada (A.D.)
| | - Christophe Faure
- Department of Pediatrics, CHU Ste-Justine Research Centre, Université de Montréal, Montréal, QC H3T 1C5, Canada
| | - Martin G. Frasch
- Department of Obstetrics and Gynaecology and Department of Neurosciences, CHU Ste-Justine Research Centre, Université de Montréal, Montréal, QC H3T 1C5, Canada; (M.C.)
- Centre de Recherche en Reproduction Animale, l’Université de Montréal, St-Hyacinthe, QC H3T 1J4, Canada
- Department of Obstetrics and Gynecology and Institute on Human Development and Disability, School of Medicine, University of Washington, 1959 NE Pacific St Box 356460, Seattle, WA 98195, USA
| |
Collapse
|
29
|
Courchesne M, Wakefield C, Nygard K, Burns P, Fecteau G, Desrochers A, Cao M, Frasch MG. Vagus Nerve Manipulation and Microglial Plasticity in the Prenatal Brain. NEUROMETHODS 2024:69-94. [DOI: 10.1007/978-1-0716-3465-3_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
30
|
Lacan L, Garabedian C, De Jonckheere J, Ghesquiere L, Storme L, Sharma D, Nguyen The Tich S. Fetal brain response to worsening acidosis: an experimental study in a fetal sheep model of umbilical cord occlusions. Sci Rep 2023; 13:23050. [PMID: 38155199 PMCID: PMC10754920 DOI: 10.1038/s41598-023-49495-2] [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: 03/15/2023] [Accepted: 12/08/2023] [Indexed: 12/30/2023] Open
Abstract
Perinatal anoxia remains an important public health problem as it can lead to hypoxic-ischaemic encephalopathy (HIE) and cause significant neonatal mortality and morbidity. The mechanisms of the fetal brain's response to hypoxia are still unclear and current methods of in utero HIE prediction are not reliable. In this study, we directly analysed the brain response to hypoxia in fetal sheep using in utero EEG. Near-term fetal sheep were subjected to progressive hypoxia induced by repeated umbilical cord occlusions (UCO) at increasing frequency. EEG changes during and between UCO were analysed visually and quantitatively, and related with gasometric and haemodynamic data. EEG signal was suppressed during occlusions and progressively slowed between occlusions with the increasing severity of the occlusions. Per-occlusion EEG suppression correlated with per-occlusion bradycardia and increased blood pressure, whereas EEG slowing and amplitude decreases correlated with arterial hypotension and respiratory acidosis. The suppression of the EEG signal during cord occlusion, in parallel with cardiovascular adaptation could correspond to a rapid cerebral adaptation mechanism that may have a neuroprotective role. The progressive alteration of the signal with the severity of the occlusions would rather reflect the cerebral hypoperfusion due to the failure of the cardiovascular adaptation mechanisms.
Collapse
Affiliation(s)
- Laure Lacan
- CHU Lille, Univ. Lille, ULR 2694 - METRICS, 59000, Lille, France.
- Department of Pediatric Neurology, CHU Lille, 59000, Lille, France.
- Department of Pediatric Neurology, Hôpital Roger Salengro, CHU Lille, Avenue du Professeur Emile Laine, 59037, Lille Cedex, France.
| | - Charles Garabedian
- CHU Lille, Univ. Lille, ULR 2694 - METRICS, 59000, Lille, France
- Department of Obstetrics, CHU Lille, 59000, Lille, France
| | - Julien De Jonckheere
- CHU Lille, Univ. Lille, ULR 2694 - METRICS, 59000, Lille, France
- CHU Lille, CIC-IT 1403, 59000, Lille, France
| | - Louise Ghesquiere
- CHU Lille, Univ. Lille, ULR 2694 - METRICS, 59000, Lille, France
- Department of Obstetrics, CHU Lille, 59000, Lille, France
| | - Laurent Storme
- CHU Lille, Univ. Lille, ULR 2694 - METRICS, 59000, Lille, France
- Department of Neonatology, CHU Lille, 59000, Lille, France
| | - Dyuti Sharma
- CHU Lille, Univ. Lille, ULR 2694 - METRICS, 59000, Lille, France
- Department of Pediatric Surgery, CHU Lille, 59000, Lille, France
| | - Sylvie Nguyen The Tich
- CHU Lille, Univ. Lille, ULR 2694 - METRICS, 59000, Lille, France
- Department of Pediatric Neurology, CHU Lille, 59000, Lille, France
| |
Collapse
|
31
|
Chevalier G, Garabedian C, Pekar JD, Wojtanowski A, Le Hesran D, Galan LE, Sharma D, Storme L, Houfflin-Debarge V, De Jonckheere J, Ghesquière L. Early heart rate variability changes during acute fetal inflammatory response syndrome: An experimental study in a fetal sheep model. PLoS One 2023; 18:e0293926. [PMID: 38032884 PMCID: PMC10688759 DOI: 10.1371/journal.pone.0293926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 10/21/2023] [Indexed: 12/02/2023] Open
Abstract
INTRODUCTION Fetal infection during labor with fetal inflammatory response syndrome (FIRS) is associated with neurodevelopmental disabilities, cerebral palsy, neonatal sepsis, and mortality. Current methods to diagnose FIRS are inadequate. Thus, the study aim was to explore whether fetal heart rate variability (HRV) analysis can be used to detect FIRS. MATERIAL AND METHODS In chronically instrumented near-term fetal sheep, lipopolysaccharide (LPS) was injected intravenously to model FIRS. A control group received saline solution injection. Hemodynamic, blood gas analysis, interleukin-6 (IL-6), and 14 HRV indices were recorded for 6 h. In both groups, comparisons were made between the stability phase and the 6 h following injection (H1-H6, respectively) and between LPS and control groups. RESULTS Fifteen lambs were instrumented. In the LPS group (n = 8), IL-6 increased significantly after LPS injection (p < 0.001), confirming the FIRS model. Fetal heart rate increased significantly after H5 (p < 0.01). In our FIRS model without shock or cardiovascular decompensation, five HRV measures changed significantly after H2 until H4 in comparison to baseline. Moreover, significant differences between LPS and control groups were observed in HRV measures between H2 and H4. These changes appear to be mediated by an increase of global variability and a loss of signal complexity. CONCLUSION As significant HRV changes were detected before FHR increase, these indices may be valuable for early detection of acute FIRS.
Collapse
Affiliation(s)
- Geoffroy Chevalier
- ULR 2694—METRICS—Evaluation des Technologies de Santé et des Pratiques Médicales, University Lille, CHU Lille, France
- Department of Obstetrics, CHU Lille, France
| | - Charles Garabedian
- ULR 2694—METRICS—Evaluation des Technologies de Santé et des Pratiques Médicales, University Lille, CHU Lille, France
- Department of Obstetrics, CHU Lille, France
| | | | | | | | | | - Dyuti Sharma
- ULR 2694—METRICS—Evaluation des Technologies de Santé et des Pratiques Médicales, University Lille, CHU Lille, France
- Department of Pediatric Surgery, CHU Lille, France
| | - Laurent Storme
- ULR 2694—METRICS—Evaluation des Technologies de Santé et des Pratiques Médicales, University Lille, CHU Lille, France
- Department of Neonatology, CHU Lille, France
| | - Veronique Houfflin-Debarge
- ULR 2694—METRICS—Evaluation des Technologies de Santé et des Pratiques Médicales, University Lille, CHU Lille, France
- Department of Obstetrics, CHU Lille, France
| | - Julien De Jonckheere
- ULR 2694—METRICS—Evaluation des Technologies de Santé et des Pratiques Médicales, University Lille, CHU Lille, France
- CIC-IT 1403, CHU Lille, France
| | - Louise Ghesquière
- ULR 2694—METRICS—Evaluation des Technologies de Santé et des Pratiques Médicales, University Lille, CHU Lille, France
- Department of Obstetrics, CHU Lille, France
| |
Collapse
|
32
|
Lue PY, Oliver MH, Neeff M, Thorne PR, Suzuki-Kerr H. Sheep as a large animal model for hearing research: comparison to common laboratory animals and humans. Lab Anim Res 2023; 39:31. [PMID: 38012676 PMCID: PMC10680324 DOI: 10.1186/s42826-023-00182-3] [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: 07/25/2023] [Revised: 11/10/2023] [Accepted: 11/15/2023] [Indexed: 11/29/2023] Open
Abstract
Sensorineural hearing loss (SNHL), caused by pathology in the cochlea, is the most common type of hearing loss in humans. It is generally irreversible with very few effective pharmacological treatments available to prevent the degenerative changes or minimise the impact. Part of this has been attributed to difficulty of translating "proof-of-concept" for novel treatments established in small animal models to human therapies. There is an increasing interest in the use of sheep as a large animal model. In this article, we review the small and large animal models used in pre-clinical hearing research such as mice, rats, chinchilla, guinea pig, rabbit, cat, monkey, dog, pig, and sheep to humans, and compare the physiology, inner ear anatomy, and some of their use as model systems for SNHL, including cochlear implantation surgeries. Sheep have similar cochlear anatomy, auditory threshold, neonatal auditory system development, adult and infant body size, and number of birth as humans. Based on these comparisons, we suggest that sheep are well-suited as a potential translational animal model that bridges the gap between rodent model research to the clinical use in humans. This is especially in areas looking at changes across the life-course or in specific areas of experimental investigation such as cochlear implantation and other surgical procedures, biomedical device development and age-related sensorineural hearing loss research. Combined use of small animals for research that require higher throughput and genetic modification and large animals for medical translation could greatly accelerate the overall translation of basic research in the field of auditory neuroscience from bench to clinic.
Collapse
Affiliation(s)
- Po-Yi Lue
- Department of Physiology, The University of Auckland, Auckland, New Zealand
- Eisdell Moore Centre, The University of Auckland, Auckland, New Zealand
| | - Mark H Oliver
- Liggins Institute, The University of Auckland, Auckland, New Zealand
- Ngapouri Research Farm Laboratory, University of Auckland, Waiotapu, New Zealand
| | - Michel Neeff
- Department of Physiology, The University of Auckland, Auckland, New Zealand
- Department of Surgery, Auckland District Health Board, Auckland, New Zealand
| | - Peter R Thorne
- Department of Physiology, The University of Auckland, Auckland, New Zealand
- Eisdell Moore Centre, The University of Auckland, Auckland, New Zealand
- Section of Audiology, The University of Auckland, Auckland, New Zealand
| | - Haruna Suzuki-Kerr
- Department of Physiology, The University of Auckland, Auckland, New Zealand.
- Eisdell Moore Centre, The University of Auckland, Auckland, New Zealand.
| |
Collapse
|
33
|
Robinson JL, Gatford KL, Clifton VL, Morrison JL, Stark MJ. Preclinical models of maternal asthma and progeny outcomes: a scoping review protocol. JBI Evid Synth 2023; 21:2115-2126. [PMID: 37246955 DOI: 10.11124/jbies-23-00006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
OBJECTIVE This scoping review will describe the methodology, phenotype, and characteristics of maternal asthma models used in preclinical studies and the outcomes that have been measured in the mother and progeny. The review This will identify gaps in knowledge of maternal and progeny outcomes following maternal asthma in pregnancy. INTRODUCTION Maternal asthma affects up to 17% of pregnancies worldwide and is associated with adverse perinatal outcomes in mothers and babies, including pre-eclampsia, gestational diabetes, cesarean section, preterm birth, small for gestational age, nursery admission, and neonatal death. While the associations are well established, the mechanisms linking maternal asthma and adverse perinatal outcomes are largely unknown due to the difficulties of human mechanistic studies. The appropriate selection of animal models is vital to understanding the mechanisms underlying associations between human maternal asthma and adverse perinatal outcomes. INCLUSION CRITERIA This review will include primary studies published in English where outcomes have been studied in vivo in non-human mammalian species. METHODS This review will follow the JBI methodology for scoping reviews. We will search MEDLINE (PubMed), Embase, and Web of Science to identify papers published before the end of 2022. Initial keywords will include pregnancy, gestation, asthma , and wheeze , as well as validated search strings to identify papers that describe animal models. Extracted data will include information on methods used to induce maternal asthma; asthmatic phenotypes and characteristics; and maternal, pregnancy, placental, and progeny outcomes. The characteristics of each study will be presented in summary tables and a core outcome list to assist researchers in developing, reporting, and comparing future animal studies of maternal asthma. REVIEW REGISTRATION Open Science Framework osf.io/trwk5.
Collapse
Affiliation(s)
- Joshua L Robinson
- Robinson Research Institute, The University of Adelaide, Adelaide, SA, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA, Australia
| | - Kathy L Gatford
- Robinson Research Institute, The University of Adelaide, Adelaide, SA, Australia
- School of Biomedicine, The University of Adelaide, Adelaide, SA, Australia
| | - Vicki L Clifton
- Mater Research Institute, University of Queensland, Brisbane, QLD, Australia
| | - Janna L Morrison
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA, Australia
| | - Michael J Stark
- Robinson Research Institute, The University of Adelaide, Adelaide, SA, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
- Department of Neonatal Medicine, Women's and Children's Hospital, Adelaide, SA, Australia
| |
Collapse
|
34
|
Chappell J, Aughwane R, Clark AR, Ourselin S, David AL, Melbourne A. A review of feto-placental vasculature flow modelling. Placenta 2023; 142:56-63. [PMID: 37639951 PMCID: PMC10873207 DOI: 10.1016/j.placenta.2023.08.068] [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: 06/19/2023] [Revised: 08/18/2023] [Accepted: 08/20/2023] [Indexed: 08/31/2023]
Abstract
The placenta provides the vital nutrients and removal of waste products required for fetal growth and development. Understanding and quantifying the differences in structure and function between a normally functioning placenta compared to an abnormal placenta is vital to provide insights into the aetiology and treatment options for fetal growth restriction and other placental disorders. Computational modelling of blood flow in the placenta allows a new understanding of the placental circulation to be obtained. This structured review discusses multiple recent methods for placental vascular model development including analysis of the appearance of the placental vasculature and how placental haemodynamics may be simulated at multiple length scales.
Collapse
Affiliation(s)
- Joanna Chappell
- School of Biomedical Engineering and Imaging Sciences (BMEIS), King's College, London, UK.
| | - Rosalind Aughwane
- Elizabeth Garrett Anderson Institute for Women's Health, University College, London, UK
| | | | - Sebastien Ourselin
- School of Biomedical Engineering and Imaging Sciences (BMEIS), King's College, London, UK
| | - Anna L David
- Elizabeth Garrett Anderson Institute for Women's Health, University College, London, UK
| | - Andrew Melbourne
- School of Biomedical Engineering and Imaging Sciences (BMEIS), King's College, London, UK
| |
Collapse
|
35
|
Padhee M, Lock MC, McMillen IC, Zhang S, Botting KJ, Nyengaard JR, MacLaughlin SM, Kleemann DO, Walker SK, Kelly JM, Rudiger SR, Morrison JL. Sex-specific effects of in vitro culture and embryo transfer on cardiac growth in sheep offspring. JOURNAL OF MOLECULAR AND CELLULAR CARDIOLOGY PLUS 2023; 5:100039. [PMID: 39802173 PMCID: PMC11708372 DOI: 10.1016/j.jmccpl.2023.100039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/14/2023] [Accepted: 07/20/2023] [Indexed: 01/16/2025]
Abstract
Embryo culture with and without human serum supplementation, previously common practice in assisted reproductive technologies (ARTs), have been associated with increased heart weight in early and late gestation in the sheep fetus. The present study aimed to determine whether the effects of embryo culture and transfer on cardiac growth and associated signalling pathways persist after birth. Embryos were either transferred to an intermediate ewe (ET) or cultured in vitro in the absence (IVC) or presence of human serum (IVCHS) and with methionine supplementation (IVCHS+M) for 6 days after mating. Naturally mated (NM) ewes were used as controls. There was an increase in the number of cardiomyocytes in the left ventricle of IVC and IVCHS+M compared to IVCHS lambs, but only in males. There were no differences in birth weight, body weight, relative heart weight, left ventricular weight, signalling molecules involved in hypertrophy, apoptosis or fibrosis at 6 months of age between the treatment groups. However, there was increased protein abundance of signalling molecules involved in ribosomal biogenesis, in male offspring from the IVC and IVCHS+M groups compared to the IVCHS group. In conclusion, the composition of the culture media used for in vitro embryo culture altered the abundance of proteins involved in ribosomal biogenesis as well as cardiomyocyte endowment in a sex specific manner. Our data suggest that male embryos cultured in the presence of human serum leads to molecular and structural changes that may detrimentally impact cardiovascular health across the life-course.
Collapse
Affiliation(s)
- Monalisa Padhee
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Mitchell C. Lock
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - I. Caroline McMillen
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Song Zhang
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Kimberley J. Botting
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Jens R. Nyengaard
- Core Centre for Molecular Morphology, Section for Stereology and Microscopy, Department of Clinical Medicine, Aarhus University, Denmark
- Department of Pathology, Aarhus University Hospital, Aarhus, Denmark
| | - Severence M. MacLaughlin
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - David O. Kleemann
- South Australian Research and Development Institute, Turretfield, South Australia, Australia
| | - Simon K. Walker
- South Australian Research and Development Institute, Turretfield, South Australia, Australia
| | - Jennifer M. Kelly
- South Australian Research and Development Institute, Turretfield, South Australia, Australia
| | - Skye R. Rudiger
- South Australian Research and Development Institute, Turretfield, South Australia, Australia
| | - Janna L. Morrison
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| |
Collapse
|
36
|
Blomberg J, Luna Ramirez RI, Goyal D, Limesand SW, Goyal R. Sexual dimorphic gene expression profile of perirenal adipose tissue in ovine fetuses with growth restriction. Front Physiol 2023; 14:1179288. [PMID: 37601643 PMCID: PMC10437077 DOI: 10.3389/fphys.2023.1179288] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 07/06/2023] [Indexed: 08/22/2023] Open
Abstract
Worldwide, fetal growth restriction (FGR) affects 7%-10% of pregnancies, or roughly 20.5 million infants, each year. FGR increases not only neonatal mortality and morbidity but also the risk of obesity in later life. Currently, the molecular mechanisms by which FGR "programs" an obese phenotype are not well understood. Studies demonstrate that FGR females are more prone to obesity compared to males; however, the molecular mechanisms that lead to the sexually dimorphic programming of FGR are not known. Thus, we hypothesized that FGR leads to the sexually dimorphic programming of preadipocytes and reduces their ability to differentiate into mature adipocytes. To test the hypothesis, we utilized a maternal hyperthermia-induced placental insufficiency to restrict fetal growth in sheep. We collected perirenal adipose tissue from near-term (∼140 days gestation) male and female FGR and normal-weight fetal lambs (N = 4 to 5 in each group), examined the preadipocytes' differentiation potential, and identified differential mRNA transcript expression in perirenal adipose tissue. Male FGR fetuses have a lower cellular density (nuclei number/unit area) compared to control male fetuses. However, no difference was observed in female FGR fetuses compared to control female fetuses. In addition, the ability of preadipocytes to differentiate into mature adipocytes with fat accumulation was impaired in male FGR fetuses, but this was not observed in female FGR fetuses. Finally, we examined the genes and pathways involved in the sexually dimorphic programming of obesity by FGR. On enrichment of differentially expressed genes in males compared to females, the Thermogenesis KEGG Pathway was downregulated, and the Metabolic and Steroid Biosynthesis KEGG pathways were upregulated. On enrichment of differentially expressed genes in male FGR compared to male control, the Steroid Biosynthesis KEGG Pathway was downregulated, and the PPAR Signaling KEGG pathway was upregulated. No pathways were altered in females in response to growth restriction in perirenal adipose tissue. Thus, the present study demonstrates a sexually dimorphic program in response to growth restriction in sheep fetal perirenal adipose tissue.
Collapse
Affiliation(s)
| | | | | | | | - Ravi Goyal
- School of Animal and Comparative Biomedical Sciences, College of Agriculture and Life Sciences, University of Arizona, Tucson, AZ, United States
| |
Collapse
|
37
|
Lock MC, Botting KJ, Allison BJ, Niu Y, Ford SG, Murphy MP, Orgeig S, Giussani DA, Morrison JL. MitoQ as an antenatal antioxidant treatment improves markers of lung maturation in healthy and hypoxic pregnancy. J Physiol 2023; 601:3647-3665. [PMID: 37467062 PMCID: PMC10952154 DOI: 10.1113/jp284786] [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: 04/03/2023] [Accepted: 07/04/2023] [Indexed: 07/21/2023] Open
Abstract
Chronic fetal hypoxaemia is a common pregnancy complication that increases the risk of infants experiencing respiratory complications at birth. In turn, chronic fetal hypoxaemia promotes oxidative stress, and maternal antioxidant therapy in animal models of hypoxic pregnancy has proven to be protective with regards to fetal growth and cardiovascular development. However, whether antenatal antioxidant therapy confers any benefit on lung development in complicated pregnancies has not yet been investigated. Here, we tested the hypothesis that maternal antenatal treatment with MitoQ will protect the developing lung in hypoxic pregnancy in sheep, a species with similar fetal lung developmental milestones as humans. Maternal treatment with MitoQ during late gestation promoted fetal pulmonary surfactant maturation and an increase in the expression of lung mitochondrial complexes III and V independent of oxygenation. Maternal treatment with MitoQ in hypoxic pregnancy also increased the expression of genes regulating liquid reabsorption in the fetal lung. These data support the hypothesis tested and suggest that MitoQ as an antenatal targeted antioxidant treatment may improve lung maturation in the late gestation fetus. KEY POINTS: Chronic fetal hypoxaemia promotes oxidative stress, and maternal antioxidant therapy in hypoxic pregnancy has proven to be protective with regards to fetal growth and cardiovascular development. MitoQ is a targeted antioxidant that uses the cell and the mitochondrial membrane potential to accumulate within the mitochondria. Treatment of healthy or hypoxic pregnancy with MitoQ, increases the expression of key molecules involved in surfactant maturation, lung liquid reabsorption and in mitochondrial proteins driving ATP synthesis in the fetal sheep lung. There were no detrimental effects of MitoQ treatment alone on the molecular components measured in the present study, suggesting that maternal antioxidant treatment has no effect on other components of normal maturation of the surfactant system.
Collapse
Affiliation(s)
- Mitchell C. Lock
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health ScienceUniversity of South AustraliaAdelaideSouth AustraliaAustralia
| | - Kimberley J. Botting
- Department of Physiology, Development & NeuroscienceUniversity of CambridgeCambridgeUK
| | - Beth J. Allison
- Department of Physiology, Development & NeuroscienceUniversity of CambridgeCambridgeUK
| | - Youguo Niu
- Department of Physiology, Development & NeuroscienceUniversity of CambridgeCambridgeUK
| | - Sage G. Ford
- Department of Physiology, Development & NeuroscienceUniversity of CambridgeCambridgeUK
| | | | - Sandra Orgeig
- UniSA: Clinical and Health ScienceUniversity of South AustraliaAdelaideSouth AustraliaAustralia
| | - Dino A. Giussani
- Department of Physiology, Development & NeuroscienceUniversity of CambridgeCambridgeUK
| | - Janna L. Morrison
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health ScienceUniversity of South AustraliaAdelaideSouth AustraliaAustralia
| |
Collapse
|
38
|
Sun J, Chong J, Zhang J, Ge L. Preterm pigs for preterm birth research: reasonably feasible. Front Physiol 2023; 14:1189422. [PMID: 37520824 PMCID: PMC10374951 DOI: 10.3389/fphys.2023.1189422] [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: 03/19/2023] [Accepted: 07/07/2023] [Indexed: 08/01/2023] Open
Abstract
Preterm birth will disrupt the pattern and course of organ development, which may result in morbidity and mortality of newborn infants. Large animal models are crucial resources for developing novel, credible, and effective treatments for preterm infants. This review summarizes the classification, definition, and prevalence of preterm birth, and analyzes the relationship between the predicted animal days and one human year in the most widely used animal models (mice, rats, rabbits, sheep, and pigs) for preterm birth studies. After that, the physiological characteristics of preterm pig models at different gestational ages are described in more detail, including birth weight, body temperature, brain development, cardiovascular system development, respiratory, digestive, and immune system development, kidney development, and blood constituents. Studies on postnatal development and adaptation of preterm pig models of different gestational ages will help to determine the physiological basis for survival and development of very preterm, middle preterm, and late preterm newborns, and will also aid in the study and accurate optimization of feeding conditions, diet- or drug-related interventions for preterm neonates. Finally, this review summarizes several accepted pediatric applications of preterm pig models in nutritional fortification, necrotizing enterocolitis, neonatal encephalopathy and hypothermia intervention, mechanical ventilation, and oxygen therapy for preterm infants.
Collapse
Affiliation(s)
- Jing Sun
- Chongqing Academy of Animal Sciences, Chongqing, China
- National Center of Technology Innovation for Pigs, Chongqing, China
- Key Laboratory of Pig Industry Sciences, Ministry of Agriculture, Chongqing, China
| | - Jie Chong
- Chongqing Academy of Animal Sciences, Chongqing, China
- National Center of Technology Innovation for Pigs, Chongqing, China
| | - Jinwei Zhang
- Chongqing Academy of Animal Sciences, Chongqing, China
- National Center of Technology Innovation for Pigs, Chongqing, China
- Key Laboratory of Pig Industry Sciences, Ministry of Agriculture, Chongqing, China
| | - Liangpeng Ge
- Chongqing Academy of Animal Sciences, Chongqing, China
- National Center of Technology Innovation for Pigs, Chongqing, China
- Key Laboratory of Pig Industry Sciences, Ministry of Agriculture, Chongqing, China
| |
Collapse
|
39
|
Frasch MG. Heart Rate Variability Code: Does It Exist and Can We Hack It? Bioengineering (Basel) 2023; 10:822. [PMID: 37508849 PMCID: PMC10375964 DOI: 10.3390/bioengineering10070822] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 06/13/2023] [Accepted: 07/06/2023] [Indexed: 07/30/2023] Open
Abstract
A code is generally defined as a system of signals or symbols for communication. Experimental evidence is synthesized for the presence and utility of such communication in heart rate variability (HRV) with particular attention to fetal HRV: HRV contains signatures of information flow between the organs and of response to physiological or pathophysiological stimuli as signatures of states (or syndromes). HRV exhibits features of time structure, phase space structure, specificity with respect to (organ) target and pathophysiological syndromes, and universality with respect to species independence. Together, these features form a spatiotemporal structure, a phase space, that can be conceived of as a manifold of a yet-to-be-fully understood dynamic complexity. The objective of this article is to synthesize physiological evidence supporting the existence of HRV code: hereby, the process-specific subsets of HRV measures indirectly map the phase space traversal reflecting the specific information contained in the code required for the body to regulate the physiological responses to those processes. The following physiological examples of HRV code are reviewed, which are reflected in specific changes to HRV properties across the signal-analytical domains and across physiological states and conditions: the fetal systemic inflammatory response, organ-specific inflammatory responses (brain and gut), chronic hypoxia and intrinsic (heart) HRV (iHRV), allostatic load (physiological stress due to surgery), and vagotomy (bilateral cervical denervation). Future studies are proposed to test these observations in more depth, and the author refers the interested reader to the referenced publications for a detailed study of the HRV measures involved. While being exemplified mostly in the studies of fetal HRV, the presented framework promises more specific fetal, postnatal, and adult HRV biomarkers of health and disease, which can be obtained non-invasively and continuously.
Collapse
Affiliation(s)
- Martin Gerbert Frasch
- Department of Obstetrics and Gynecology and Institute on Human Development and Disability, University of Washington School of Medicine, Seattle, WA 98195, USA
| |
Collapse
|
40
|
Darby JR, Zhang S, Holman SL, Muhlhausler BS, McMillen IC, Morrison JL. Cardiac growth and metabolism of the fetal sheep are not vulnerable to a 10 day increase in fetal glucose and insulin concentrations during late gestation. Heliyon 2023; 9:e18292. [PMID: 37519661 PMCID: PMC10372399 DOI: 10.1016/j.heliyon.2023.e18292] [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: 10/09/2022] [Revised: 07/06/2023] [Accepted: 07/13/2023] [Indexed: 08/01/2023] Open
Abstract
Aims To evaluate the effects of fetal glucose infusion in late gestation on the mRNA expression and protein abundance of molecules involved in the regulation of cardiac growth and metabolism. Main methods Either saline or glucose was infused into fetal sheep from 130 to 140 days (d) gestation (term, 150 d). At 140 d gestation, left ventricle tissue samples were collected. Quantitative real-time RT-PCR and Western blot were used to determine the mRNA expression and protein abundance of key signalling molecules within the left ventricle of the fetal heart. Key findings Although intra-fetal glucose infusion increased fetal plasma glucose and insulin concentrations, there was no change in the expression of molecules within the signalling pathways that regulate proliferation, hypertrophy, apoptosis or fibrosis in the fetal heart. Cardiac Solute carrier family 2 member 1 (SLC2A1) mRNA expression was decreased by glucose infusion. Glucose infusion increased cardiac mRNA expression of both Peroxisome proliferator activated receptor alpha (PPARA) and peroxisome proliferator activated receptor gamma (PPARG). However, there was no change in the mRNA expression of PPAR cofactors or molecules with PPAR response elements. Furthermore, glucose infusion did not impact the protein abundance of the 5 oxidative phosphorylation complexes of the electron transport chain. Significance Despite a 10-day doubling of fetal plasma glucose and insulin concentrations, the present study suggests that within the fetal left ventricle, the mRNA and protein expression of the signalling molecules involved in cardiac growth, development and metabolism are relatively unaffected.
Collapse
Affiliation(s)
| | | | | | | | | | - Janna L. Morrison
- Corresponding author. Australian Research Council Future Fellow, Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, GPO Box 2471, Adelaide, SA, 5001, Australia,
| |
Collapse
|
41
|
Larson AC, Sridharan A, Moon JK, Agarwal D, Chang J, Wallace KD, Forsberg F, Didier RA. Contrast-enhanced subharmonic aided pressure estimation for assessment of intracranial pressure in vivo. Pediatr Radiol 2023; 53:1640-1647. [PMID: 37062765 DOI: 10.1007/s00247-023-05637-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 02/19/2023] [Accepted: 02/20/2023] [Indexed: 04/18/2023]
Abstract
BACKGROUND Intracranial pressure (ICP) monitoring in children currently requires invasive techniques. Subharmonic aided pressure estimation (SHAPE) uses contrast-enhanced ultrasound (CEUS) to measure intravascular and interstitial pressure, but utility in ICP measurements has yet to be explored. OBJECTIVE The objective of this study was to investigate SHAPE as a novel tool for noninvasive ICP measurements in fetal lambs. MATERIALS AND METHODS Eighteen fetal lambs at 107-139 days gestational age (term = 145 days) underwent subdural ICP catheter placement. The brain was imaged in the coronal plane in CEUS mode optimized for SHAPE, while infusing an US contrast agent into the fetal circulation. After SHAPE calibration, saline was infused via the subdural catheter to increase ICP. Five-second SHAPE cine clips were obtained at various ICPs. Subharmonic intensity values of the whole brain and thalami were correlated with ICP values using mixed effects linear regression analyses and the strength of the relationship was evaluated by Spearman's rank-order correlation. RESULTS Forty-nine experiments produced 723 datapoints, including SHAPE intensity values and mean ICP measurements. There was a statistically significant inverse relationship between SHAPE intensity values and ICP measurements in the whole brain and thalami (median rho value - 0.58 and - 0.56, respectively). CONCLUSION SHAPE intensity values of the brain demonstrate an inverse and statistically significant correlation with in vivo ICP measurements in an animal model.
Collapse
Affiliation(s)
- Abby C Larson
- Department of Surgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Anush Sridharan
- Department of Radiology, Children's Hospital of Philadelphia, 3401 Civic Center Blvd, Philadelphia, PA, 19104, USA
| | - James K Moon
- Department of Surgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Divyansh Agarwal
- Perelmen School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jonathan Chang
- Department of Surgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | | | - Flemming Forsberg
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Ryne A Didier
- Department of Surgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Department of Radiology, Children's Hospital of Philadelphia, 3401 Civic Center Blvd, Philadelphia, PA, 19104, USA.
- Perelmen School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
42
|
Tolladay J, Lear CA, Bennet L, Gunn AJ, Georgieva A. Prediction of Fetal Blood Pressure during Labour with Deep Learning Techniques. Bioengineering (Basel) 2023; 10:775. [PMID: 37508802 PMCID: PMC10376045 DOI: 10.3390/bioengineering10070775] [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: 05/18/2023] [Revised: 06/16/2023] [Accepted: 06/24/2023] [Indexed: 07/30/2023] Open
Abstract
Our objective is to develop a model for the prediction of minimum fetal blood pressure (FBP) during fetal heart rate (FHR) decelerations. Experimental data from umbilical occlusions in near-term fetal sheep (2698 occlusions from 57 near-term lambs) were used to train a convolutional neural network. This model was then used to estimate FBP for decelerations extracted from the final 90 min of 53,445 human FHR signals collected using cardiotocography. Minimum sheep FBP was predicted with a mean absolute error of 6.7 mmHg (25th, 50th, 75th percentiles of 2.3, 5.2, 9.7 mmHg), mean absolute percentage errors of 17.3% (5.5%, 12.5%, 23.9%) and a coefficient of determination R2=0.36. While the model was unable to clearly predict severe compromise at birth in humans, there is positive evidence that such a model could predict human FBP with further development. The neural network is capable of predicting FBP for many of the sheep decelerations accurately but performed far from satisfactory at identifying FHR segments that correspond to the highest or lowest minimum FBP. These results indicate that with further work and a larger, more variable training dataset, the model could achieve higher accuracy.
Collapse
Affiliation(s)
- John Tolladay
- Oxford Labour Monitoring Group, Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, OX1 2JD, UK
| | - Christopher A Lear
- The Fetal Physiology and Neuroscience Group, Department of Physiology, University of Auckland, Auckland 1010, New Zealand
| | - Laura Bennet
- The Fetal Physiology and Neuroscience Group, Department of Physiology, University of Auckland, Auckland 1010, New Zealand
| | - Alistair J Gunn
- The Fetal Physiology and Neuroscience Group, Department of Physiology, University of Auckland, Auckland 1010, New Zealand
| | - Antoniya Georgieva
- Oxford Labour Monitoring Group, Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, OX1 2JD, UK
- Big Data Institute, Old Road Campus, University of Oxford, Oxford, OX3 7LF, UK
| |
Collapse
|
43
|
Darby JRT, Williams GK, Cho SKS, Meakin AS, Holman SL, Quinn M, Wiese MD, Macgowan CK, Seed M, Morrison JL. Acute resveratrol exposure does not impact hemodynamics of the fetal sheep. Physiol Rep 2023; 11:e15749. [PMID: 37332034 PMCID: PMC10277215 DOI: 10.14814/phy2.15749] [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: 04/29/2023] [Revised: 05/29/2023] [Accepted: 05/29/2023] [Indexed: 06/20/2023] Open
Abstract
Babies born growth restricted are at an increased risk of both poor short-and long-term outcomes. Current interventions to improve fetal growth are ineffective and do not lower the lifetime risk of poor health status. Maternal resveratrol (RSV) treatment increases uterine artery blood flow, fetal oxygenation, and fetal weight. However, studies suggest that diets high in polyphenols such as RSV may impair fetal hemodynamics. We aimed to characterize the effect of RSV on fetal hemodynamics to further assess its safety as an intervention strategy. Pregnant ewes underwent magnetic resonance imaging (MRI) scans to measure blood flow and oxygenation within the fetal circulation using phase contrast-MRI and T2 oximetry. Blood flow and oxygenation measures were performed in a basal state and then repeated while the fetus was exposed to RSV. Fetal blood pressure and heart rate were not different between states. RSV did not impact fetal oxygen delivery (DO2 ) or consumption (VO2 ). Blood flow and oxygen delivery throughout the major vessels of the fetal circulation were not different between basal and RSV states. As such, acute exposure of the fetus to RSV does not directly impact fetal hemodynamics. This strengthens the rationale for the use of RSV as an intervention strategy against fetal growth restriction.
Collapse
Affiliation(s)
- Jack R. T. Darby
- Early Origins of Adult Health Research GroupUniSA: Clinical and Health SciencesUniversity of South AustraliaSouth AustraliaAdelaideAustralia
| | - Georgia K. Williams
- Preclinical, Imaging and Research LaboratoriesSouth Australian Health and Medical Research InstituteAdelaideSouth AustraliaAustralia
| | - Steven K. S. Cho
- Early Origins of Adult Health Research GroupUniSA: Clinical and Health SciencesUniversity of South AustraliaSouth AustraliaAdelaideAustralia
- Univeristy of Toronto and The Hospital for Sick ChildrenTorontoOntarioCanada
| | - Ashley S. Meakin
- Early Origins of Adult Health Research GroupUniSA: Clinical and Health SciencesUniversity of South AustraliaSouth AustraliaAdelaideAustralia
| | - Stacey L. Holman
- Early Origins of Adult Health Research GroupUniSA: Clinical and Health SciencesUniversity of South AustraliaSouth AustraliaAdelaideAustralia
| | - Megan Quinn
- Early Origins of Adult Health Research GroupUniSA: Clinical and Health SciencesUniversity of South AustraliaSouth AustraliaAdelaideAustralia
| | - Michael D. Wiese
- Centre for Pharmaceutical Innovation, UniSA: Clinical and Health SciencesUniversity of South AustraliaSouth AustraliaAdelaideAustralia
| | | | - Mike Seed
- Univeristy of Toronto and The Hospital for Sick ChildrenTorontoOntarioCanada
| | - Janna L. Morrison
- Early Origins of Adult Health Research GroupUniSA: Clinical and Health SciencesUniversity of South AustraliaSouth AustraliaAdelaideAustralia
| |
Collapse
|
44
|
Lear CA, Beacom MJ, Dhillon SK, Lear BA, Mills OJ, Gunning MI, Westgate JA, Bennet L, Gunn AJ. Dissecting the contributions of the peripheral chemoreflex and myocardial hypoxia to fetal heart rate decelerations in near-term fetal sheep. J Physiol 2023; 601:2017-2041. [PMID: 37017488 DOI: 10.1113/jp284286] [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: 12/19/2022] [Accepted: 03/30/2023] [Indexed: 04/06/2023] Open
Abstract
Brief repeated fetal hypoxaemia during labour can trigger intrapartum decelerations of the fetal heart rate (FHR) via the peripheral chemoreflex or the direct effects of myocardial hypoxia, but the relative contribution of these two mechanisms and how this balance changes with evolving fetal compromise remain unknown. In the present study, chronically instrumented near-term fetal sheep received surgical vagotomy (n = 8) or sham vagotomy (control, n = 11) to disable the peripheral chemoreflex and unmask myocardial hypoxia. One-minute complete umbilical cord occlusions (UCOs) were performed every 2.5 min for 4 h or until arterial pressure fell below 20 mmHg. Hypotension and severe acidaemia developed progressively after 65.7 ± 7.2 UCOs in control fetuses and 49.5 ± 7.8 UCOs after vagotomy. Vagotomy was associated with faster development of metabolic acidaemia and faster impairment of arterial pressure during UCOs without impairing centralization of blood flow or neurophysiological adaptation to UCOs. During the first half of the UCO series, before severe hypotension developed, vagotomy was associated with a marked increase in FHR during UCOs. After the onset of evolving severe hypotension, FHR fell faster in control fetuses during the first 20 s of UCOs, but FHR during the final 40 s of UCOs became progressively more similar between groups, with no difference in the nadir of decelerations. In conclusion, FHR decelerations were initiated and sustained by the peripheral chemoreflex at a time when fetuses were able to maintain arterial pressure. After the onset of evolving hypotension and acidaemia, the peripheral chemoreflex continued to initiate decelerations, but myocardial hypoxia became progressively more important in sustaining and deepening decelerations. KEY POINTS: Brief repeated hypoxaemia during labour can trigger fetal heart rate decelerations by either the peripheral chemoreflex or myocardial hypoxia, but how this balance changes with fetal compromise is unknown. Reflex control of fetal heart rate was disabled by vagotomy to unmask the effects of myocardial hypoxia in chronically instrumented fetal sheep. Fetuses were then subjected to repeated brief hypoxaemia consistent with the rates of uterine contractions during labour. We show that the peripheral chemoreflex controls brief decelerations in their entirety at a time when fetuses were able to maintain normal or increased arterial pressure. The peripheral chemoreflex still initiated decelerations even after the onset of evolving hypotension and acidaemia, but myocardial hypoxia made an increasing contribution to sustain and deepen decelerations.
Collapse
Affiliation(s)
- Christopher A Lear
- The Fetal Physiology and Neuroscience Group, Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Michael J Beacom
- The Fetal Physiology and Neuroscience Group, Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Simerdeep K Dhillon
- The Fetal Physiology and Neuroscience Group, Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Benjamin A Lear
- The Fetal Physiology and Neuroscience Group, Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Olivia J Mills
- The Fetal Physiology and Neuroscience Group, Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Mark I Gunning
- The Fetal Physiology and Neuroscience Group, Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Jenny A Westgate
- Department of Obstetrics and Gynaecology, The University of Auckland, Auckland, New Zealand
| | - Laura Bennet
- The Fetal Physiology and Neuroscience Group, Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Alistair J Gunn
- The Fetal Physiology and Neuroscience Group, Department of Physiology, The University of Auckland, Auckland, New Zealand
- Department of Paediatrics, Starship Children's Hospital, Auckland, New Zealand
| |
Collapse
|
45
|
Dimasi CG, Darby JRT, Morrison JL. A change of heart: understanding the mechanisms regulating cardiac proliferation and metabolism before and after birth. J Physiol 2023; 601:1319-1341. [PMID: 36872609 PMCID: PMC10952280 DOI: 10.1113/jp284137] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Accepted: 02/17/2023] [Indexed: 03/07/2023] Open
Abstract
Mammalian cardiomyocytes undergo major maturational changes in preparation for birth and postnatal life. Immature cardiomyocytes contribute to cardiac growth via proliferation and thus the heart has the capacity to regenerate. To prepare for postnatal life, structural and metabolic changes associated with increased cardiac output and function must occur. This includes exit from the cell cycle, hypertrophic growth, mitochondrial maturation and sarcomeric protein isoform switching. However, these changes come at a price: the loss of cardiac regenerative capacity such that damage to the heart in postnatal life is permanent. This is a significant barrier to the development of new treatments for cardiac repair and contributes to heart failure. The transitional period of cardiomyocyte growth is a complex and multifaceted event. In this review, we focus on studies that have investigated this critical transition period as well as novel factors that may regulate and drive this process. We also discuss the potential use of new biomarkers for the detection of myocardial infarction and, in the broader sense, cardiovascular disease.
Collapse
Affiliation(s)
- Catherine G. Dimasi
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health SciencesUniversity of South AustraliaAdelaideSAAustralia
| | - Jack R. T. Darby
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health SciencesUniversity of South AustraliaAdelaideSAAustralia
| | - Janna L. Morrison
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health SciencesUniversity of South AustraliaAdelaideSAAustralia
| |
Collapse
|
46
|
Thangaraj SV, Kachman M, Halloran KM, Sinclair KD, Lea R, Bellingham M, Evans NP, Padmanabhan V. Developmental programming: Preconceptional and gestational exposure of sheep to a real-life environmental chemical mixture alters maternal metabolome in a fetal sex-specific manner. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 864:161054. [PMID: 36565874 PMCID: PMC10322214 DOI: 10.1016/j.scitotenv.2022.161054] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 12/12/2022] [Accepted: 12/15/2022] [Indexed: 05/21/2023]
Abstract
BACKGROUND Everyday, humans are exposed to a mixture of environmental chemicals some of which have endocrine and/or metabolism disrupting actions which may contribute to non-communicable diseases. The adverse health impacts of real-world chemical exposure, characterized by chronic low doses of a mixture of chemicals, are only recently emerging. Biosolids derived from human waste represent the environmental chemical mixtures humans are exposed to in real life. Prior studies in sheep have shown aberrant reproductive and metabolic phenotypes in offspring after maternal biosolids exposure. OBJECTIVE To determine if exposure to biosolids perturbs the maternal metabolic milieu of pregnant ewes, in a fetal sex-specific manner. METHODS Ewes were grazed on inorganic fertilizer (Control) or biosolids-treated pastures (BTP) from before mating and throughout gestation. Plasma from pregnant ewes (Control n = 15, BTP n = 15) obtained mid-gestation were analyzed by untargeted metabolomics. Metabolites were identified using Agilent MassHunter. Multivariate analyses were done using MetaboAnalyst 5.0 and confirmed using SIMCA. RESULTS Univariate and multivariate analysis of 2301 annotated metabolites identified 193 differentially abundant metabolites (DM) between control and BTP sheep. The DM primarily belonged to the super-class of lipids and organic acids. 15-HeTrE, oleamide, methionine, CAR(3:0(OH)) and pyroglutamic acid were the top DM and have been implicated in the regulation of fetal growth and development. Fetal sex further exacerbated differences in metabolite profiles in the BTP group. The organic acids class of metabolites was abundant in animals with male fetuses. Prenol lipid, sphingolipid, glycerolipid, alkaloid, polyketide and benzenoid classes showed fetal sex-specific responses to biosolids. DISCUSSION Our study illustrates that exposure to biosolids significantly alters the maternal metabolome in a fetal sex-specific manner. The altered metabolite profile indicates perturbations to fatty acid, arginine, branched chain amino acid and one‑carbon metabolism. These factors are consistent with, and likely contribute to, the adverse phenotypic outcomes reported in the offspring.
Collapse
Affiliation(s)
- S V Thangaraj
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA
| | - M Kachman
- MM BRCF Metabolomics Core, University of Michigan, Ann Arbor, MI, USA
| | - K M Halloran
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA
| | - K D Sinclair
- University of Nottingham, Sutton Bonington Campus, Loughborough, UK
| | - R Lea
- University of Nottingham, Sutton Bonington Campus, Loughborough, UK
| | - M Bellingham
- School of Biodiversity One Health and Veterinary Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, UK
| | - N P Evans
- School of Biodiversity One Health and Veterinary Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, UK
| | - V Padmanabhan
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
47
|
Kyathanahalli C, Snedden M, Hirsch E. Is human labor at term an inflammatory condition?†. Biol Reprod 2023; 108:23-40. [PMID: 36173900 PMCID: PMC10060716 DOI: 10.1093/biolre/ioac182] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 09/16/2022] [Accepted: 09/22/2022] [Indexed: 01/20/2023] Open
Abstract
Parturition at term in normal pregnancy follows a predictable sequence of events. There is some evidence that a state of inflammation prevails in the reproductive tissues during labor at term, but it is uncertain whether this phenomenon is the initiating signal for parturition. The absence of a clear temporal sequence of inflammatory events prior to labor casts doubt on the concept that normal human labor at term is primarily the result of an inflammatory cascade. This review examines evidence linking parturition and inflammation in order to address whether inflammation is a cause of labor, a consequence of labor, or a separate but related phenomenon. Finally, we identify and suggest ways to reconcile inconsistencies regarding definitions of labor onset in published research, which may contribute to the variability in conclusions regarding the genesis and maintenance of parturition. A more thorough understanding of the processes underlying normal parturition at term may lead to novel insights regarding abnormal labor, including spontaneous preterm labor, preterm premature rupture of the fetal membranes, and dysfunctional labor, and the role of inflammation in each.
Collapse
Affiliation(s)
- Chandrashekara Kyathanahalli
- Department of Obstetrics and Gynecology, NorthShore University HealthSystem, Evanston, Illinois, USA
- Department of Obstetrics and Gynecology, Pritzker School of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Madeline Snedden
- Department of Obstetrics and Gynecology, NorthShore University HealthSystem, Evanston, Illinois, USA
| | - Emmet Hirsch
- Department of Obstetrics and Gynecology, NorthShore University HealthSystem, Evanston, Illinois, USA
- Department of Obstetrics and Gynecology, Pritzker School of Medicine, University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
48
|
Meakin AS, Amirmostofian M, Darby JRT, Holman SL, Morrison JL, Wiese MD. Characterisation of cytochrome P450 isoenzyme activity in sheep liver and placental microsomes. Placenta 2023; 131:82-89. [PMID: 36527743 DOI: 10.1016/j.placenta.2022.11.015] [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] [Received: 08/19/2022] [Revised: 10/17/2022] [Accepted: 11/30/2022] [Indexed: 12/02/2022]
Abstract
INTRODUCTION Drug metabolism during pregnancy is a complex process that involves maternal, placental and fetal sites of metabolism. Indeed, there is a lack of clarity provided from drug metabolism in human pregnancy due to ethical limitations. Large animal models of human pregnancy provide an opportunity to quantify activity of phase 1 drug metabolism mediated by cytochrome P450 (CYP) enzymes in the maternal, placental, and fetal compartments. Herein, we have validated a comprehensive assay to quantify maternal, placental, and fetal CYP activity. METHODS Isolated microsomes from sheep maternal liver, placenta, and fetal liver (140d gestation, term = 150d) were incubated with CYP-specific probe drugs to quantify the activity of CYP1A2, CYP2B6, CYP2C8, CYP2C9, CYP2C19, CYP2D6, CYP2E1 and CYP3A. Inhibition studies were performed to validate specificity of probe drugs. The validated assay was developed using liquid chromatography-tandem mass spectrometry (LC-MS/MS). RESULTS CYP1A2, CYP2B6, CYP2C8, CYP2C19, CYP2D6, CYP2E1 and CYP3A were active in maternal liver. In contrast, only CYP1A2, CYP2C8 and CYP2D6 were active in the placenta, whereas CYP2B6, CYP2C8 and CYP2D6 were active in the fetal liver. Of the placental-specific CYPs validated, CYP1A2 increased in type A compared with type D placentomes, whereas CYP2C8 activity increased in type B compared with type A and C. DISCUSSION This study has established conditions for compartment-specific CYP activity in the sheep maternal-placental-fetal unit using a validated and standardised experimental workflow. Compartment- and placentome type-specific CYP activity are important considerations when examining drug metabolism in the maternal-placental-fetal unit and in determining the impact of pregnancy complications.
Collapse
Affiliation(s)
- Ashley S Meakin
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Marzieh Amirmostofian
- Centre for Pharmaceutical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Jack RT Darby
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Stacey L Holman
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Janna L Morrison
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia.
| | - Michael D Wiese
- Centre for Pharmaceutical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia.
| |
Collapse
|
49
|
Alon T, Rosov A, Lifshitz L, Moallem U. Male fetuses negatively affect the vitality of the litter and the dam's metabolic and physiological state in multifetal pregnant ewe. PLoS One 2023; 18:e0285338. [PMID: 37159474 PMCID: PMC10168572 DOI: 10.1371/journal.pone.0285338] [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] [Accepted: 04/19/2023] [Indexed: 05/11/2023] Open
Abstract
In sheep, ~30% of fetuses do not survive till parturition, and 17.7% of the multifetal pregnancies experience partial litter loss (PLL). In humans, multifetal pregnancies are associated with a higher risk of perinatal mortality. Therefore, the objectives were to examine the association between partial litter loss, fetal sex, dam's metabolic and physiological state, and pregnancy outcome in multifetal pregnant ewes. The study includes two parts. The first was a retrospective study, in which we analyzed data of 675 lambings and examined the PLL incidence according to male ratio (MR) for all litter sizes (range 2-6). Lambings were categorized as having a low male ratio (LMR; <50% males) or a high male ratio (LMR; >50% males). In the second part, we monitored 24 ewes from 80 to 138 days in pregnancy every 10 days, and then daily until lambing, by ultrasound scanning for maternal heart rate (HR), and Doppler ultrasound for litter vitality. Blood samples were taken from dams on the days of scanning. Male ratio strongly affected PLL, where the general survival rate (for all lambings) was reduced from 90% in LMR lambings to 85% in HMR lambings. The odds ratio for PLL in HMR vs. LMR litters was 1.82. Birth body weight and the survival rate of female was higher in LMR than HMR lambings, with no differences for male lambs in both parameters. In the second part, dams' HR during the last trimester was 9.4% higher in LMR than in HMR pregnancies, with no differences in fetuses' HR. The plasma glucose and insulin concentrations were not significantly different between groups, but plasma β-hydroxybutyrate and nonesterified fatty acid concentrations were, respectively, 31% and 20% lower in HMR vs. LMR ewes. In conclusion, male fetuses negatively affect pregnancy outcomes and influence dams' metabolic and physiological state in sheep.
Collapse
Affiliation(s)
- Tamir Alon
- Department of Ruminant Science, Institute of Animal Sciences, The Volcani Center, Rishon LeZion, Israel
- Department of Animal Science, the Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Alexander Rosov
- Department of Ruminant Science, Institute of Animal Sciences, The Volcani Center, Rishon LeZion, Israel
| | - Lila Lifshitz
- Department of Ruminant Science, Institute of Animal Sciences, The Volcani Center, Rishon LeZion, Israel
| | - Uzi Moallem
- Department of Ruminant Science, Institute of Animal Sciences, The Volcani Center, Rishon LeZion, Israel
| |
Collapse
|
50
|
Spencer BL, Mychaliska GB. Milestones for clinical translation of the artificial placenta. Semin Fetal Neonatal Med 2022; 27:101408. [PMID: 36437184 DOI: 10.1016/j.siny.2022.101408] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Despite significant advances in the treatment of prematurity, premature birth results in significant mortality and morbidity. In particular, extremely low gestational age newborns (ELGANs) defined as <28 weeks estimated gestational age (EGA) suffer from disproportionate mortality and morbidity. A radical paradigm shift in the treatment of prematurity is to recreate fetal physiology using an extracorporeal VV-ECLS artificial placenta (AP) or an AV-ECLS artificial womb (AW). Over the past 15 years, tremendous advances have been made in the laboratory confirming long-term support and organ protection and ongoing development. The major milestones to clinical application are miniaturization, anticoagulation, clinical risk stratification, specialized critical care protocols, a regulatory path and a strategy and platform to translate technology to the bedside. Currently, several groups are addressing the remaining milestones for clinical translation.
Collapse
Affiliation(s)
- Brianna L Spencer
- Department of Surgery, University of Michigan, 2101 Taubman Center 1500 E Medical Center Dr, Ann Arbor, MI, 48109, USA.
| | - George B Mychaliska
- Section of Pediatric Surgery, Department of Surgery, Fetal Diagnosis and Treatment Center, C.S. Mott Children's Hospital, 1540 E Hospital Dr, Ann Arbor, MI, 48109, USA.
| |
Collapse
|