1
|
Paz AA, Jiménez TA, Ibarra-Gonzalez J, Astudillo-Maya C, Beñaldo FA, Figueroa EG, Llanos AJ, Gonzalez-Candia A, Herrera EA. Gestational hypoxia elicits long-term cardiovascular dysfunction in female guinea pigs. Life Sci 2025; 361:123282. [PMID: 39615619 DOI: 10.1016/j.lfs.2024.123282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 11/21/2024] [Accepted: 11/26/2024] [Indexed: 01/06/2025]
Abstract
BACKGROUND Gestational hypoxia (GH) has been implicated in the developmental programming of cardiovascular diseases (CVDs) in the offspring, with most studies focusing on males, conversely, the effects on female cardiovascular health remain understudied. We aimed to investigate the impact of GH on the cardiovascular system of female guinea pig offspring from the early postnatal period to adulthood. METHODS Pregnant guinea pigs were subjected to normoxic or hypoxic conditions from gestational day 30 until delivery (∼70 days). Female offspring were monitored with biometric parameters and peripheral vascular function (ultrasound) from birth to one year old. In addition, we assessed cardiovascular structure, oxidative stress, inflammatory state (IHC, qPCR, and immunoblot assays), and thoracic aorta reactivity (wire-myography) at one year of age. KEY FINDINGS GH increased heart rate and peripheral pulsatility index. At one year old, GH-exposed females exhibited cardiac remodeling, characterized by increased left ventricular luminal area and coronary artery muscle occupation. Furthermore, GH increased aortic vascular wall, intima-media thickness and contractile capacity. This was accompanied by reduced endothelium-dependent vasodilation and enhanced oxidative stress. Additionally, GH increased collagen deposition and oxidative stress in the right ventricle, accompanied by reduced antioxidant enzymes expression and reduced inflammatory mediator levels. SIGNIFICANCE GH exerts long-lasting effects on the cardiovascular health of female guinea pig offspring, contributing to cardiac remodeling, vascular dysfunction, oxidative stress, and inflammatory changes. These findings highlight the importance of GH as a risk factor for developing CVDs in female offspring and emphasize the need for sex-specific interventions to mitigate adverse long-term gestational effects.
Collapse
Affiliation(s)
- Adolfo A Paz
- Laboratorio de Función y Reactividad Vascular, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Tamara A Jiménez
- Laboratorio de Función y Reactividad Vascular, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Julieta Ibarra-Gonzalez
- Laboratorio de Función y Reactividad Vascular, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Cristian Astudillo-Maya
- Laboratorio de Función y Reactividad Vascular, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Felipe A Beñaldo
- Laboratorio de Función y Reactividad Vascular, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Esteban G Figueroa
- Instituto de Ciencias de la Salud, Universidad de O'Higgins, Rancagua, Chile
| | - Aníbal J Llanos
- Laboratorio de Función y Reactividad Vascular, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile; International Center for Andean Studies (INCAS), Universidad de Chile, Putre, Chile
| | | | - Emilio A Herrera
- Laboratorio de Función y Reactividad Vascular, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile; International Center for Andean Studies (INCAS), Universidad de Chile, Putre, Chile.
| |
Collapse
|
2
|
Wilson RL, Davenport BN, Jones HN. Mid-Pregnancy Placental Transcriptome in a Model of Placental Insufficiency with and without Novel Intervention. Reprod Sci 2024:10.1007/s43032-024-01769-4. [PMID: 39707140 DOI: 10.1007/s43032-024-01769-4] [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: 06/06/2024] [Accepted: 12/09/2024] [Indexed: 12/23/2024]
Abstract
Fetal growth restriction (FGR) affects between 5-10% of all live births. Placental insufficiency is a leading cause of FGR, resulting in reduced nutrient and oxygen delivery to the fetus. Currently, there are no effective in utero treatment options for FGR, or placental insufficiency. We have developed a gene therapy to deliver, via a non-viral nanoparticle, human insulin-like 1 growth factor (hIGF1) to the placenta as a potential treatment for placenta insufficiency and FGR. Using a guinea pig maternal nutrient restriction (MNR) model of FGR, we aimed to understand the transcriptional changes within the placenta associated with placental insufficiency that occur prior to/at initiation of FGR, and the impact of short-term hIGF1 nanoparticle treatment. Using RNAsequencing, we analyzed protein coding genes of three experimental groups: Control and MNR dams receiving a sham treatment, and MNR dams receiving hIGF1 nanoparticle treatment. Pathway enrichment analysis comparing differentially expressed genelists in sham-treated MNR placentas to sham-treated Control revealed upregulation of pathways associated with degradation and repair of genetic information and downregulation of pathways associated with transmembrane transport. When compared to sham-treated MNR placentas, MNR + hIGF1 placentas demonstrated changes to genelists associated with transmembrane transporter activity including ion, vitamin and solute carrier transport. Overall, this study identifies the key signaling and metabolic changes occurring in the placenta contributing to placental insufficiency prior to/at initiation of FGR, and increases our understanding of the pathways that our nanoparticle-mediated gene therapy intervention regulates.
Collapse
Affiliation(s)
- Rebecca L Wilson
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL, USA.
- Center for Research in Perinatal Outcomes, College of Medicine, University of Florida, Gainesville, FL, USA.
| | - Baylea N Davenport
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL, USA
- Center for Research in Perinatal Outcomes, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Helen N Jones
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL, USA
- Center for Research in Perinatal Outcomes, College of Medicine, University of Florida, Gainesville, FL, USA
| |
Collapse
|
3
|
Davenport BN, Wilson RL, Williams AA, Jones HN. Placental nanoparticle-mediated IGF1 gene therapy corrects fetal growth restriction in a guinea pig model. Gene Ther 2024:10.1038/s41434-024-00508-3. [PMID: 39627510 DOI: 10.1038/s41434-024-00508-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 11/13/2024] [Accepted: 11/20/2024] [Indexed: 12/11/2024]
Abstract
Fetal growth restriction (FGR) caused by placental insufficiency is a major contributor to neonatal morbidity and mortality. There is currently no in utero treatment for placental insufficiency or FGR. The placenta serves as the vital communication, supply, exchange, and defense organ for the developing fetus and offers an excellent opportunity for therapeutic interventions. Here we show efficacy of repeated treatments of trophoblast-specific human insulin-like 1 growth factor (IGF1) gene therapy delivered in a non-viral, polymer nanoparticle to the placenta for the treatment of FGR. Using a guinea pig maternal nutrient restriction model (70% food intake) of FGR, nanoparticle-mediated IGF1 treatment was delivered to the placenta via ultrasound guidance across the second half of pregnancy, after establishment of FGR. This treatment resulted in correction of fetal weight in MNR + IGF1 animals compared to sham treated controls on an ad libitum diet, increased fetal blood glucose and decreased fetal blood cortisol levels compared to sham treated MNR, and showed no negative maternal side-effects. Overall, we show a therapy capable of positively impacting the entire pregnancy environment: maternal, placental, and fetal. This combined with our previous studies using this therapy at mid pregnancy in the guinea pig and in two different mouse model and three different human in vitro/ex vivo models, demonstrate the plausibility of this therapy for future human translation. Our overall goal is to improve health outcomes of neonates and decrease numerous morbidities associated with the developmental origins of disease.
Collapse
Affiliation(s)
- Baylea N Davenport
- Center for Research in Perinatal Outcomes, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Rebecca L Wilson
- Center for Research in Perinatal Outcomes, College of Medicine, University of Florida, Gainesville, FL, USA.
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL, USA.
| | - Alyssa A Williams
- Center for Research in Perinatal Outcomes, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Helen N Jones
- Center for Research in Perinatal Outcomes, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL, USA
| |
Collapse
|
4
|
Sethi S, Friesen-Waldner LJ, Regnault TRH, McKenzie CA. Quantifying Brain Myelin Water Fraction in a Guinea Pig Model of Spontaneous Intrauterine Growth Restriction. J Magn Reson Imaging 2024; 60:2615-2625. [PMID: 38445838 DOI: 10.1002/jmri.29332] [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: 07/26/2023] [Revised: 02/17/2024] [Accepted: 02/20/2024] [Indexed: 03/07/2024] Open
Abstract
BACKGROUND Intrauterine growth restriction (IUGR) is an obstetrical condition where a fetus has not achieved its genetic potential. A consequence of IUGR is a decrease in brain myelin content. Myelin water imaging (MWI) has been used to assess fetal myelin water fraction (MWF) and might potentially assess myelination changes associated with IUGR. PURPOSE To quantify and compare the MWF of non-IUGR and IUGR fetal guinea pigs (GPs) in late gestation. STUDY TYPE Prospective animal model. ANIMAL MODEL Dunkin-Hartley GP model of spontaneous IUGR (mean ± SD: 60 ± 1.2 days gestation; 19 IUGR, 52 control). FIELD STRENGTH/SEQUENCE Eight spoiled gradient-recalled (SPGR) gradient echo volumes (flip angles [α]: 2°-16°), and two sets of eight balanced steady-state free precession (bSSFP) gradient echo volumes (α: 8° - 64°), at 0° and 180° phase increments, at 3.0 T. ASSESSMENT MWF maps were generated for each fetal GP brain using multicomponent driven equilibrium single pulse observation of T1/T2 (mcDESPOT). MWF was quantified in the fetal corpus callosum (CC), fornix (FOR), parasagittal white matter (PSW), and whole fetal brain. STATISTICAL TESTS Linear regression was performed between five fetal IUGR markers (body volume, body-to-pregnancy volume ratio, brain-to-liver volume ratio, brain-to-placenta volume ratio, and brain-to-body volume ratio) and MWF (coefficient of determination, R2). A t-test with a linear mixed model compared the MWF of non-IUGR and IUGR fetal GPs (significance was determined at α < 0.05). RESULTS The MWF of the control fetuses are (mean ± SD): 0.23 ± 0.02 (CC), 0.31 ± 0.02 (FOR), 0.28 ± 0.02 (PSW), and 0.20 ± 0.01 (whole brain). The MWF of the IUGR fetuses are (mean ± SD): 0.19 ± 0.02 (CC), 0.27 ± 0.01 (FOR), 0.24 ± 0.03 (PSW), and 0.16 ± 0.01 (whole brain). Significant differences in MWF were found between the non-IUGR and IUGR fetuses in every comparison. DATA CONCLUSION The mean MWF of IUGR fetal GPs is significantly lower than non-IUGR fetal GPs. EVIDENCE LEVEL 2 TECHNICAL EFFICACY: Stage 1.
Collapse
Affiliation(s)
- Simran Sethi
- Department of Medical Biophysics, Western University, London, Ontario, Canada
| | | | - Timothy R H Regnault
- Department of Obstetrics & Gynaecology, Western University, London, Ontario, Canada
- Department of Physiology & Pharmacology, Western University, London, Ontario, Canada
- Division of Maternal, Fetal and Newborn Health, Children's Health Research Institute, Lawson Health Research Institute, London, Ontario, Canada
| | - Charles A McKenzie
- Department of Medical Biophysics, Western University, London, Ontario, Canada
- Division of Maternal, Fetal and Newborn Health, Children's Health Research Institute, Lawson Health Research Institute, London, Ontario, Canada
- Robarts Research Institute, Western University, London, Ontario, Canada
| |
Collapse
|
5
|
Rock CR, Miller SL, Allison BJ. The Use of Antioxidants for Cardiovascular Protection in Fetal Growth Restriction: A Systematic Review. Antioxidants (Basel) 2024; 13:1400. [PMID: 39594542 PMCID: PMC11591491 DOI: 10.3390/antiox13111400] [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: 09/30/2024] [Revised: 11/09/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
Fetal growth restriction (FGR) increases the risk of cardiovascular disease. There are currently no treatment options available; however, antioxidants have shown potential to improve cardiovascular deficits associated with FGR. This systematic review aimed to determine whether antenatal antioxidant intervention can effectively protect the developing cardiovascular system in FGR. We searched for interventional studies that used an antenatal antioxidant intervention to improve cardiac and/or vascular outcomes in FGR published between 01/1946 and 09/2024 using MEDLINE and Embase (PROSPERO: CRD42024503756). The risk of bias was assessed with SYRCLE. The studies were assessed for cardiovascular protection based on the percentage of cardiac and/or vascular deficits that were restored with the antioxidant treatment. Studies were characterised as showing strong cardiovascular protection (≥50% restoration), mild cardiovascular protection (>0% but <50% restoration), an antioxidant-only effect (this did not include control group which showed a change with antioxidant intervention compared to FGR) or no cardiovascular protection (0% restoration). Thirty-eight publications met the inclusion criteria, encompassing 43 studies and investigating 15 antioxidant interventions. Moreover, 29/43 studies (71%) reported the restoration of at least one cardiac or vascular deficit with antioxidant intervention, and 21/43 studies (51%) were classified as strong cardiovascular protection. An ex vivo analysis of the arterial function in seven studies revealed endothelial dysfunction in growth-restricted offspring and antioxidant interventions restored the endothelial function in all cases. Additionally, four studies demonstrated that antioxidants reduced peroxynitrite-mediated oxidative stress. Notably, only 13/43 studies (32%) delayed antioxidant administration until after the induction of FGR. Antenatal antioxidant interventions show promise for providing cardiovascular protection in FGR. Melatonin was the most frequently studied intervention followed by nMitoQ, vitamin C and N-acetylcysteine, all of which demonstrated a strong capacity to reduce oxidative stress and improve nitric oxide bioavailability in the cardiovascular system of growth-restricted offspring; however, this systematic review highlights critical knowledge gaps and inconsistencies in preclinical research, which hinder our ability to determine which antioxidant treatments are currently suitable for clinical translation.
Collapse
Affiliation(s)
- Charmaine R. Rock
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright Street, Clayton 3168, Australia;
- Department of Obstetrics and Gynaecology, Monash University, Clayton 3168, Australia
| | - Suzanne L. Miller
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright Street, Clayton 3168, Australia;
- Department of Obstetrics and Gynaecology, Monash University, Clayton 3168, Australia
| | - Beth J. Allison
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright Street, Clayton 3168, Australia;
- Department of Obstetrics and Gynaecology, Monash University, Clayton 3168, Australia
| |
Collapse
|
6
|
Wilson RL, Schmidt JK, Davenport BN, Ren E, Keding LT, Shaw SA, Schotzko ML, Antony KM, Simmons HA, Golos TG, Jones HN. Placental gene therapy in nonhuman primates: a pilot study of maternal, placental, and fetal response to non-viral, polymeric nanoparticle delivery of IGF1. Mol Hum Reprod 2024; 30:gaae038. [PMID: 39499161 PMCID: PMC11562130 DOI: 10.1093/molehr/gaae038] [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: 07/09/2024] [Revised: 10/14/2024] [Indexed: 11/07/2024] Open
Abstract
Currently, there are no placenta-targeted treatments to alter the in utero environment for administration to pregnant women who receive a diagnosis of fetal growth restriction (FGR). Water-soluble polymers have a distinguished record of clinical relevance outside of pregnancy. We have demonstrated the effective delivery of polymer-based nanoparticles containing a non-viral human insulin-like growth factor 1 (IGF1) transgene to correct placental insufficiency in small animal models of FGR. Our goals were to extend these studies to a proof-of-concept study in the pregnant macaque, establish feasibility of nanoparticle-mediated gene therapy delivery to trophoblasts, and investigate the acute maternal, placental, and fetal responses to treatment. Pregnant macaques underwent ultrasound-guided intraplacental injections of nanoparticles (GFP- or IGF1-expressing plasmid under the control of the trophoblast-specific PLAC1 promoter complexed with a HPMA-DMEAMA co-polymer) at approximately gestational day 100 (term = 165 days). Fetectomy was performed 24 h (GFP; n = 1), 48 h (IGF1; n = 3) or 10 days (IGF1; n = 3) after nanoparticle delivery. Routine pathological assessment was performed on biopsied maternal tissues and placental and fetal tissues. Maternal blood was analyzed for complete blood count (CBC), immunomodulatory proteins and growth factors, progesterone (P4), and estradiol (E2). Placental ERK/AKT/mTOR signaling was assessed using Western blot and qPCR. Fluorescent microscopy and in situ hybridization confirmed placental uptake and transient transgene expression in villous syncytiotrophoblast. No off-target expression was observed in either maternal or fetal tissues. Histopathological assessment of the placenta recorded observations not necessarily related to the IGF1 nanoparticle treatment. In maternal blood, CBCs, P4, and E2 remained within the normal range for pregnant macaques across the treatment period. Changes to placental ERK and AKT signaling at 48 h and 10 days after IGF1 nanoparticle treatment indicated an upregulation in placental homeostatic mechanisms to prevent overactivity in the normal pregnancy environment. The lack of adverse maternal reaction to nanoparticle-mediated IGF1 treatment, combined with changes in placental signaling to maintain homeostasis, indicates no deleterious impact of treatment during the acute phase of study.
Collapse
Affiliation(s)
- Rebecca L Wilson
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL, USA
- Center for Research in Perinatal Outcomes, University of Florida, Gainesville, FL, USA
| | - Jenna Kropp Schmidt
- Wisconsin National Primate Research Center, University of Wisconsin–Madison, Madison, WI, USA
| | - Baylea N Davenport
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL, USA
- Center for Research in Perinatal Outcomes, University of Florida, Gainesville, FL, USA
| | - Emily Ren
- Wisconsin National Primate Research Center, University of Wisconsin–Madison, Madison, WI, USA
| | - Logan T Keding
- Wisconsin National Primate Research Center, University of Wisconsin–Madison, Madison, WI, USA
| | - Sarah A Shaw
- Wisconsin National Primate Research Center, University of Wisconsin–Madison, Madison, WI, USA
| | - Michele L Schotzko
- Wisconsin National Primate Research Center, University of Wisconsin–Madison, Madison, WI, USA
| | - Kathleen M Antony
- Department of Obstetrics and Gynecology, University of Wisconsin–Madison, Madison, WI, USA
| | - Heather A Simmons
- Wisconsin National Primate Research Center, University of Wisconsin–Madison, Madison, WI, USA
| | - Thaddeus G Golos
- Wisconsin National Primate Research Center, University of Wisconsin–Madison, Madison, WI, USA
- Department of Obstetrics and Gynecology, University of Wisconsin–Madison, Madison, WI, USA
- Department of Comparative Biosciences, University of Wisconsin–Madison, Madison, WI, USA
| | - Helen N Jones
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL, USA
- Center for Research in Perinatal Outcomes, University of Florida, Gainesville, FL, USA
| |
Collapse
|
7
|
Rollman TB, Berkebile ZW, Hicks DM, Hatfield JS, Chauhan P, Pravetoni M, Schleiss MR, Milligan GN, Morgan TK, Bierle CJ. CD4+ but not CD8+ T cells are required for protection against severe guinea pig cytomegalovirus infections. PLoS Pathog 2024; 20:e1012515. [PMID: 39495799 PMCID: PMC11563410 DOI: 10.1371/journal.ppat.1012515] [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: 08/19/2024] [Revised: 11/14/2024] [Accepted: 09/27/2024] [Indexed: 11/06/2024] Open
Abstract
Human cytomegalovirus (HCMV) is a ubiquitous herpesvirus and the leading cause of infectious disease related birth defects worldwide. How the immune response modulates the risk of intrauterine transmission of HCMV after maternal infection remains poorly understood. Maternal T cells likely play a critical role in preventing infection at the maternal-fetal interface and limiting spread across the placenta, but concerns exist that immune responses to infection may also cause placental dysfunction and adverse pregnancy outcomes. This study investigated the role of CD4+ and CD8+ T cells in a guinea pig model of primary cytomegalovirus infection. Monoclonal antibodies specific to guinea pig CD4 and CD8 were used to deplete T cells in non-pregnant and in pregnant guinea pigs after mid-gestation. CD4+ T cell depletion increased the severity of illness, caused significantly elevated viral loads, and increased the rate of congenital guinea pig cytomegalovirus (GPCMV) infection relative to animals treated with control antibody. CD8+ T cell depletion was comparably well tolerated and did not significantly affect the weight of infected guinea pigs or viral loads in their blood or tissue. However, significantly more viral genomes and transcripts were detected in the placenta and decidua of CD8+ T cell depleted dams post-infection. This study corroborates earlier findings made in nonhuman primates that maternal CD4+ T cells play a critical role in limiting the severity of primary CMV infection during pregnancy while also revealing that other innate and adaptive immune responses can compensate for an absent CD8+ T cell response in α-CD8-treated guinea pigs.
Collapse
Affiliation(s)
- Tyler B. Rollman
- Division of Pediatric Infectious Diseases, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Zachary W. Berkebile
- Division of Pediatric Infectious Diseases, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Dustin M. Hicks
- Department of Pharmacology, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Jason S. Hatfield
- Division of Pediatric Infectious Diseases, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Priyanka Chauhan
- Division of Pediatric Infectious Diseases, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Marco Pravetoni
- Center for Medication Development for Substance Use Disorders and Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Mark R. Schleiss
- Division of Pediatric Infectious Diseases, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Gregg N. Milligan
- Division of Vaccinology, Department of Pediatrics, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Terry K. Morgan
- Department of Pathology, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Craig J. Bierle
- Division of Pediatric Infectious Diseases, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, United States of America
| |
Collapse
|
8
|
Sixtus RP, Gray C, Barnes H, Paterson ESJ, Berry MJ, Dyson RM. Cardiovascular responses to heat and cold exposure are altered by preterm birth in guinea pigs. Physiol Rep 2024; 12:e70098. [PMID: 39435736 PMCID: PMC11494451 DOI: 10.14814/phy2.70098] [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: 08/16/2024] [Revised: 10/10/2024] [Accepted: 10/10/2024] [Indexed: 10/23/2024] Open
Abstract
Adversity early in life can modify the trajectory for disease risk extending decades beyond the event. Preterm birth produces persistent cardiovascular alterations that may appear maladaptive in adulthood. We have previously hypothesized that those born preterm may exhibit cardiovascular vulnerability in the climate change context. Further, this vulnerability may be present as early as childhood. We aimed to identify the early signs of cardiovascular dysfunction at childhood-equivalent age using our animal model of preterm birth. Using a whole-body thermal stress test, guinea pigs aged 35-d and 38-d (equivalent to 8-10-year-old children) and born at term or preterm gestations were exposed to progressive hyper- (TC = 41.5°C) and hypo-thermia (TC = 34°C; normothermia TC = 39°C). Comprehensive cardiovascular monitoring included ECG, blood pressure, microvascular perfusion, blood gas, and catecholamine profile, as well as skin and core body temperature. Preterm-born animals exhibited attenuated vascular responses to hyperthermic stress, and a significant elevation in systolic blood pressure in response to hypothermic stress. Such responses are similar to those observed in elderly populations and indicate the presence of cardiovascular dysfunction. This is the first study to demonstrate the impact of preterm birth on the cardiovascular response to both heat and cold stress. Further, this dysfunction has been observed at an earlier age than that achievable using traditional stress testing techniques. The present findings warrant further investigation.
Collapse
Affiliation(s)
- Ryan Phillip Sixtus
- Department of Paediatrics and Child HealthUniversity of OtagoWellingtonNew Zealand
- Present address:
Department of Biological and Life SciencesCardiff UniversityWalesUK
| | - Clint Gray
- Department of Paediatrics and Child HealthUniversity of OtagoWellingtonNew Zealand
| | - Heather Barnes
- Department of Paediatrics and Child HealthUniversity of OtagoWellingtonNew Zealand
| | | | - Mary Judith Berry
- Department of Paediatrics and Child HealthUniversity of OtagoWellingtonNew Zealand
| | - Rebecca Maree Dyson
- Department of Paediatrics and Child HealthUniversity of OtagoWellingtonNew Zealand
| |
Collapse
|
9
|
Guan T, Guo J, Lin R, Liu J, Luo R, Zhang Z, Pei D, Liu J. Single-cell analysis of preimplantation embryonic development in guinea pigs. BMC Genomics 2024; 25:911. [PMID: 39350018 PMCID: PMC11440810 DOI: 10.1186/s12864-024-10815-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 09/19/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND Guinea pigs exhibit numerous physiological similarities to humans, yet the details of their preimplantation embryonic development remain largely unexplored. RESULTS To address this, we conducted single-cell sequencing on the transcriptomes of cells isolated from the zygote stage through preimplantation stages in guinea pigs. This study identified seven distinct cell types within guinea pig preimplantation embryos and pinpointed the timing of zygotic gene activation (ZGA). Trajectory analysis revealed a bifurcation into two lineage-specific branches, accompanied by alterations in specific pathways, including oxidative phosphorylation and vascular endothelial growth factor (VEGF). Additionally, co-expressed gene network analysis highlighted the most enriched functional modules for the epiblast (EPI), primitive endoderm (PrE), and inner cell mass (ICM). Finally, we compared the similarities and differences between human and guinea pig epiblasts (EPIs). CONCLUSION This study systematically constructs a cell atlas of guinea pig preimplantation embryonic development, offering fresh insights into mammalian embryonic development and providing alternative experimental models for studying human embryonic development.
Collapse
Affiliation(s)
- Tongxing Guan
- School of Life Sciences, University of Science and Technology of China, Hefei, 230026, China
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
- Center for Development and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Jing Guo
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
- Center for Development and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Runxia Lin
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
- Center for Development and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Jinpeng Liu
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
- University of Chinese Academy of Science, Beijing, 100049, People's Republic of China
- Center for Development and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Rongping Luo
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
- Center for Development and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Zhen Zhang
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.
- Center for Development and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.
| | - Duanqing Pei
- School of Life Sciences, University of Science and Technology of China, Hefei, 230026, China.
| | - Jing Liu
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.
- Joint School of Life Sciences,Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, 510530, China.
- Center for Development and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.
| |
Collapse
|
10
|
Davenport B, Wilson R, Williams A, Jones H. Placental Nanoparticle-mediated IGF1 Gene Therapy Corrects Fetal Growth Restriction in a Guinea Pig Model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.05.587765. [PMID: 38645174 PMCID: PMC11030242 DOI: 10.1101/2024.04.05.587765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Fetal growth restriction (FGR) caused by placental insufficiency is a major contributor to neonatal morbidity and mortality. There is currently no in utero treatment for placental insufficiency or FGR. The placenta serves as the vital communication, supply, exchange, and defense organ for the developing fetus and offers an excellent opportunity for therapeutic interventions. Here we show efficacy of repeated treatments of trophoblast-specific human insulin-like 1 growth factor (IGF1) gene therapy delivered in a non-viral, polymer nanoparticle to the placenta for the treatment of FGR. Using a guinea pig maternal nutrient restriction model (70% food intake) of FGR, nanoparticle-mediated IGF1 treatment was delivered to the placenta via ultrasound guidance across the second half of pregnancy, after establishment of FGR. This treatment resulted in correction of fetal weight in MNR + IGF1 animals compared to sham treated controls on an ad libitum diet, increased fetal blood glucose and decreased fetal blood cortisol levels compared to sham treated MNR, and showed no negative maternal side-effects. Overall, we show a therapy capable of positively impacting the entire pregnancy environment: maternal, placental, and fetal. This combined with our previous studies using this therapy at mid pregnancy in the guinea pig and in two different mouse model and three different human in vitro/ex vivo models, demonstrate the plausibility of this therapy for future human translation. Our overall goal is to improve health outcomes of neonates and decrease numerous morbidities associated with the developmental origins of disease.
Collapse
|
11
|
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
- Department of Physiology, Pharmacology & Toxicology, West Virginia University School of Medicine, Morgantown, WV, USA
- Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Sydney Nassabeh
- Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Audra Hurley
- Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Lainey Shouldis
- Department of Immunology and Microbial Pathogenesis, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Paul D Chantler
- Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, WV, USA
- Department of Neuroscience, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Duaa Dakhlallah
- Department of Medicine, West Virginia University School of Medicine, Morgantown, WV, USA
| | - I Mark Olfert
- Department of Physiology, Pharmacology & Toxicology, West Virginia University School of Medicine, Morgantown, WV, USA
- Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, WV, USA
- Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, WV, USA
| |
Collapse
|
12
|
Haq KT, McLean K, Salameh S, Swift LM, Posnack NG. Electroanatomical adaptations in the guinea pig heart from neonatal to adulthood. Europace 2024; 26:euae158. [PMID: 38864516 PMCID: PMC11218563 DOI: 10.1093/europace/euae158] [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: 02/21/2024] [Accepted: 06/06/2024] [Indexed: 06/13/2024] Open
Abstract
AIMS Electroanatomical adaptations during the neonatal to adult phase have not been comprehensively studied in preclinical animal models. To explore the impact of age as a biological variable on cardiac electrophysiology, we employed neonatal and adult guinea pigs, which are a recognized animal model for developmental research. METHODS AND RESULTS Electrocardiogram recordings were collected in vivo from anaesthetized animals. A Langendorff-perfusion system was employed for the optical assessment of action potentials and calcium transients. Optical data sets were analysed using Kairosight 3.0 software. The allometric relationship between heart weight and body weight diminishes with age, it is strongest at the neonatal stage (R2 = 0.84) and abolished in older adults (R2 = 1E-06). Neonatal hearts exhibit circular activation, while adults show prototypical elliptical shapes. Neonatal conduction velocity (40.6 ± 4.0 cm/s) is slower than adults (younger: 61.6 ± 9.3 cm/s; older: 53.6 ± 9.2 cm/s). Neonatal hearts have a longer action potential duration (APD) and exhibit regional heterogeneity (left apex; APD30: 68.6 ± 5.6 ms, left basal; APD30: 62.8 ± 3.6), which was absent in adults. With dynamic pacing, neonatal hearts exhibit a flatter APD restitution slope (APD70: 0.29 ± 0.04) compared with older adults (0.49 ± 0.04). Similar restitution characteristics are observed with extrasystolic pacing, with a flatter slope in neonates (APD70: 0.54 ± 0.1) compared with adults (younger: 0.85 ± 0.4; older: 0.95 ± 0.7). Neonatal hearts display unidirectional excitation-contraction coupling, while adults exhibit bidirectionality. CONCLUSION Postnatal development is characterized by transient changes in electroanatomical properties. Age-specific patterns can influence cardiac physiology, pathology, and therapies for cardiovascular diseases. Understanding heart development is crucial to evaluating therapeutic eligibility, safety, and efficacy.
Collapse
Affiliation(s)
- Kazi T Haq
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children’s National Hospital, 111 Michigan Avenue, NW, Washington, DC 20010, USA
- Children’s National Heart Institute, Children’s National Hospital, Washington, DC, USA
| | - Kate McLean
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children’s National Hospital, 111 Michigan Avenue, NW, Washington, DC 20010, USA
| | - Shatha Salameh
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children’s National Hospital, 111 Michigan Avenue, NW, Washington, DC 20010, USA
- Children’s National Heart Institute, Children’s National Hospital, Washington, DC, USA
- Department of Pharmacology and Physiology, The George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Luther M Swift
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children’s National Hospital, 111 Michigan Avenue, NW, Washington, DC 20010, USA
- Children’s National Heart Institute, Children’s National Hospital, Washington, DC, USA
| | - Nikki Gillum Posnack
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children’s National Hospital, 111 Michigan Avenue, NW, Washington, DC 20010, USA
- Children’s National Heart Institute, Children’s National Hospital, Washington, DC, USA
- Department of Pharmacology and Physiology, The George Washington University School of Medicine and Health Sciences, Washington, DC, USA
- Department of Pediatrics, The George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| |
Collapse
|
13
|
Collins HE, Alexander BT, Care AS, Davenport MH, Davidge ST, Eghbali M, Giussani DA, Hoes MF, Julian CG, LaVoie HA, Olfert IM, Ozanne SE, Bytautiene Prewit E, Warrington JP, Zhang L, Goulopoulou S. Guidelines for assessing maternal cardiovascular physiology during pregnancy and postpartum. Am J Physiol Heart Circ Physiol 2024; 327:H191-H220. [PMID: 38758127 PMCID: PMC11380979 DOI: 10.1152/ajpheart.00055.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 04/22/2024] [Accepted: 05/08/2024] [Indexed: 05/18/2024]
Abstract
Maternal mortality rates are at an all-time high across the world and are set to increase in subsequent years. Cardiovascular disease is the leading cause of death during pregnancy and postpartum, especially in the United States. Therefore, understanding the physiological changes in the cardiovascular system during normal pregnancy is necessary to understand disease-related pathology. Significant systemic and cardiovascular physiological changes occur during pregnancy that are essential for supporting the maternal-fetal dyad. The physiological impact of pregnancy on the cardiovascular system has been examined in both experimental animal models and in humans. However, there is a continued need in this field of study to provide increased rigor and reproducibility. Therefore, these guidelines aim to provide information regarding best practices and recommendations to accurately and rigorously measure cardiovascular physiology during normal and cardiovascular disease-complicated pregnancies in human and animal models.
Collapse
Grants
- HL169157 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01 HD088590 NICHD NIH HHS
- HD083132 HHS | NIH | Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD)
- The Biotechnology and Biological Sciences Research Council
- P20GM103499 HHS | NIH | National Institute of General Medical Sciences (NIGMS)
- British Heart Foundation (BHF)
- R21 HD111908 NICHD NIH HHS
- Distinguished University Professor
- The Lister Insititute
- ES032920 HHS | NIH | National Institute of Environmental Health Sciences (NIEHS)
- Canadian Insitute's of Health Research Foundation Grant
- HL149608 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- Royal Society (The Royal Society)
- U.S. Department of Defense (DOD)
- HL138181 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- MC_00014/4 UKRI | Medical Research Council (MRC)
- RG/17/8/32924 British Heart Foundation
- Jewish Heritage Fund for Excellence
- HD111908 HHS | NIH | Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD)
- HL163003 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- APP2002129 NHMRC Ideas Grant
- HL159865 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- HL131182 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- HL163818 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- NS103017 HHS | NIH | National Institute of Neurological Disorders and Stroke (NINDS)
- HL143459 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- HL146562 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01 HL138181 NHLBI NIH HHS
- 20CSA35320107 American Heart Association (AHA)
- RG/17/12/33167 British Heart Foundation (BHF)
- National Heart Foundation Future Leader Fellowship
- P20GM121334 HHS | NIH | National Institute of General Medical Sciences (NIGMS)
- HL146562-04S1 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- HL155295 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- HD088590-06 HHS | NIH | Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD)
- HL147844 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- WVU SOM Synergy Grant
- R01 HL146562 NHLBI NIH HHS
- R01 HL159865 NHLBI NIH HHS
- HL159447 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- ES034646-01 HHS | NIH | National Institute of Environmental Health Sciences (NIEHS)
- HL150472 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- 2021T017 Dutch Heart Foundation Dekker Grant
- MC_UU_00014/4 Medical Research Council
- R01 HL163003 NHLBI NIH HHS
- Christenson professor In Active Healthy Living
- National Heart Foundation
- Dutch Heart Foundation Dekker
- WVU SOM Synergy
- Jewish Heritage
- Department of Health | National Health and Medical Research Council (NHMRC)
- Gouvernement du Canada | Canadian Institutes of Health Research (Instituts de recherche en santé du Canada)
Collapse
Affiliation(s)
- Helen E Collins
- University of Louisville, Louisville, Kentucky, United States
| | - Barbara T Alexander
- University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Alison S Care
- University of Adelaide, Adelaide, South Australia, Australia
| | | | | | - Mansoureh Eghbali
- University of California Los Angeles, Los Angeles, California, United States
| | | | | | - Colleen G Julian
- University of Colorado School of Medicine, Aurora, Colorado, United States
| | - Holly A LaVoie
- University of South Carolina School of Medicine, Columbia, South Carolina, United States
| | - I Mark Olfert
- West Virginia University School of Medicine, Morgantown, West Virginia, United States
| | | | | | - Junie P Warrington
- University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Lubo Zhang
- Loma Linda University School of Medicine, Loma Linda, California, United States
| | | |
Collapse
|
14
|
Wilson RL, Davenport BN, Jones HN. Mid-pregnancy placental transcriptome in a model of placental insufficiency with and without novel intervention. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.05.597621. [PMID: 38895312 PMCID: PMC11185618 DOI: 10.1101/2024.06.05.597621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Fetal growth restriction (FGR) affects between 5-10% of all live births. Placental insufficiency is a leading cause of FGR, resulting in reduced nutrient and oxygen delivery to the fetus. Currently, there are no effective in utero treatment options for FGR, or placental insufficiency. We have developed a gene therapy to deliver, via a non-viral nanoparticle, human insulin-like 1 growth factor ( hIGF1 ) to the placenta as potential treatment of placenta insufficiency and FGR. Using a guinea pig maternal nutrient restriction (MNR) model of FGR, we aimed to understand the transcriptional changes within the placenta associated with placental insufficiency that occur prior to/at initiation of FGR, and the impact of short-term hIGF1 nanoparticle treatment. Using RNAsequencing, we analyzed protein coding genes of three experimental groups: Control and MNR dams receiving a sham treatment, and MNR dams receiving hIGF1 nanoparticle treatment. Pathway enrichment analysis comparing differentially expressed genelists in sham-treated MNR placentas to Control revealed upregulation of pathways associated with degradation and repair of genetic information and downregulation of pathways associated with transmembrane transport. When compared to sham-treated MNR placentas, MNR + hIGF1 placentas demonstrated changes to genelists associated with transmembrane transporter activity including ion, vitamin and solute carrier transport. Overall, this study identifies the key signaling and metabolic changes occurring in the placenta contributing to placental insufficiency prior to/at initiation of FGR, and increases our understanding of the pathways that our nanoparticle-mediated gene therapy intervention regulates. Statements and Declarations Competing Interests: Authors declare no conflicts of interest.
Collapse
|
15
|
Hamada H, Casciaro C, Moisiadis VG, Constantinof A, Kostaki A, Matthews SG. Prenatal maternal glucocorticoid exposure modifies sperm miRNA profiles across multiple generations in the guinea-pig. J Physiol 2024; 602:2127-2139. [PMID: 38285002 DOI: 10.1113/jp284942] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 01/11/2024] [Indexed: 01/30/2024] Open
Abstract
Maternal stress and glucocorticoid exposure during pregnancy have multigenerational effects on neuroendocrine function and behaviours in offspring. Importantly, effects are transmitted through the paternal lineage. Altered phenotypes are associated with profound differences in transcription and DNA methylation in the brain. In the present study, we hypothesized that maternal prenatal synthetic glucocorticoid (sGC) exposure in the F0 pregnancy will result in differences in miRNA levels in testes germ cells and sperm across multiple generations, and that these changes will associate with modified microRNA (miRNA) profiles and gene expression in the prefrontal cortex (PFC) of subsequent generations. Pregnant guinea-pigs (F0) were treated with multiple courses of the sGC betamethasone (Beta) (1 mg kg-1; gestational days 40, 41, 50, 51, 60 and 61) in late gestation. miRNA levels were assessed in testes germ cells and in F2 PFC using the GeneChip miRNA 4.0 Array and candidate miRNA measured in epididymal sperm by quantitative real-time PCR. Maternal Beta exposure did not alter miRNA levels in germ cells derived from the testes of adult male offspring. However, there were significant differences in the levels of four candidate miRNAs in the sperm of F1 and F2 adult males. There were no changes in miRNA levels in the PFC of juvenile F2 female offspring. The present study has identified that maternal Beta exposure leads to altered miRNA levels in sperm that are apparent for at least two generations. The fact that differences were confined to epididymal sperm suggests that the intergenerational effects of Beta may target the epididymis. KEY POINTS: Paternal glucocorticoid exposure prior to conception leads to profound epigenetic changes in the brain and somatic tissues in offspring, and microRNAs (miRNAs) in sperm may mediate these changes. We show that there were significant differences in the miRNA profile of epididymal sperm in two generations following prenatal glucocorticoid exposure that were not observed in germ cells derived from the testes. The epididymis is a probable target for intergenerational programming. The effects of prenatal glucocorticoid treatment may span multiple generations.
Collapse
Affiliation(s)
- Hirotaka Hamada
- Departments of Physiology, Obstetrics and Gynaecology and Medicine, University of Toronto, Toronto, ON, Canada
- Department of Gynecology and Obstetrics, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Christopher Casciaro
- Departments of Physiology, Obstetrics and Gynaecology and Medicine, University of Toronto, Toronto, ON, Canada
| | - Vasilis G Moisiadis
- Departments of Physiology, Obstetrics and Gynaecology and Medicine, University of Toronto, Toronto, ON, Canada
| | - Andrea Constantinof
- Departments of Physiology, Obstetrics and Gynaecology and Medicine, University of Toronto, Toronto, ON, Canada
| | - Alisa Kostaki
- Departments of Physiology, Obstetrics and Gynaecology and Medicine, University of Toronto, Toronto, ON, Canada
| | - Stephen G Matthews
- Departments of Physiology, Obstetrics and Gynaecology and Medicine, University of Toronto, Toronto, ON, Canada
- Lunenfeld-Tanenbaum Research Institute, Sinai Health Systems, Toronto, ON, Canada
| |
Collapse
|
16
|
Moloney RA, Palliser HK, Dyson RM, Pavy CL, Berry M, Hirst JJ, Shaw JC. Ongoing effects of preterm birth on the dopaminergic and noradrenergic pathways in the frontal cortex and hippocampus of guinea pigs. Dev Neurobiol 2024; 84:93-110. [PMID: 38526217 DOI: 10.1002/dneu.22937] [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: 08/10/2023] [Revised: 03/04/2024] [Accepted: 03/07/2024] [Indexed: 03/26/2024]
Abstract
Children born preterm have an increased likelihood of developing neurobehavioral disorders such as attention-deficit hyperactivity disorder (ADHD) and anxiety. These disorders have a sex bias, with males having a higher incidence of ADHD, whereas anxiety disorder tends to be more prevalent in females. Both disorders are underpinned by imbalances to key neurotransmitter systems, with dopamine and noradrenaline in particular having major roles in attention regulation and stress modulation. Preterm birth disturbances to neurodevelopment may affect this neurotransmission in a sexually dimorphic manner. Time-mated guinea pig dams were allocated to deliver by preterm induction of labor (gestational age 62 [GA62]) or spontaneously at term (GA69). The resultant offspring were randomized to endpoints as neonates (24 h after term-equivalence age) or juveniles (corrected postnatal day 40, childhood equivalence). Relative mRNA expressions of key dopamine and noradrenaline pathway genes were examined in the frontal cortex and hippocampus and quantified with real-time PCR. Myelin basic protein and neuronal nuclei immunostaining were performed to characterize the impact of preterm birth. Within the frontal cortex, there were persisting reductions in the expression of dopaminergic pathway components that occurred in preterm males only. Conversely, preterm-born females had increased expression of key noradrenergic receptors and a reduction of the noradrenergic transporter within the hippocampus. This study demonstrated that preterm birth results in major changes in dopaminergic and noradrenergic receptor, transporter, and synthesis enzyme gene expression in a sex- and region-based manner that may contribute to the sex differences in susceptibility to neurobehavioral disorders. These findings highlight the need for the development of sex-based treatments for improving these conditions.
Collapse
Affiliation(s)
- Roisin A Moloney
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, Australia
- Hunter Medical Research Institute, Mothers and Babies Research Centre, Newcastle, Australia
| | - Hannah K Palliser
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, Australia
- Hunter Medical Research Institute, Mothers and Babies Research Centre, Newcastle, Australia
| | - Rebecca M Dyson
- Department of Paediatrics and Child Health, University of Otago, Wellington, New Zealand
- Biomedical Research Unit, University of Otago, Wellington, New Zealand
| | - Carlton L Pavy
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, Australia
- Hunter Medical Research Institute, Mothers and Babies Research Centre, Newcastle, Australia
| | - Max Berry
- Department of Paediatrics and Child Health, University of Otago, Wellington, New Zealand
- Biomedical Research Unit, University of Otago, Wellington, New Zealand
| | - Jonathon J Hirst
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, Australia
- Hunter Medical Research Institute, Mothers and Babies Research Centre, Newcastle, Australia
| | - Julia C Shaw
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, Australia
- Hunter Medical Research Institute, Mothers and Babies Research Centre, Newcastle, Australia
| |
Collapse
|
17
|
Hargett SE, Leslie EF, Chapa HO, Gaharwar AK. Animal models of postpartum hemorrhage. Lab Anim (NY) 2024; 53:93-106. [PMID: 38528231 DOI: 10.1038/s41684-024-01349-8] [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: 06/16/2023] [Accepted: 02/26/2024] [Indexed: 03/27/2024]
Abstract
Postpartum hemorrhage (PPH)-heavy bleeding following childbirth-is a leading cause of morbidity and mortality worldwide. PPH can affect individuals regardless of risks factors and its incidence has been increasing in high-income countries including the United States. The high incidence and severity of this childbirth complication has propelled research into advanced treatments and alternative solutions for patients facing PPH; however, the development of novel treatments is limited by the absence of a common, well-established and well-validated animal model of PPH. A variety of animals have been used for in vivo studies of novel therapeutic materials; however, each of these animals differs considerably from the anatomy and physiology of a postpartum woman, and the methods used for achieving a postpartum hemorrhagic condition vary widely. Here we critically evaluate the various animal models of PPH presented in the literature and propose additional and alternative methods for modeling PPH in in vivo studies. We highlight how current animal models successfully or unsuccessfully mimic the anatomy and physiology of a postpartum woman and how this may impact treatment development. We aim to equip researchers with the necessary background information to select appropriate animal models for their research related to PPH solutions, while supporting the goals of refinement, reduction and replacement (3Rs) in preclinical animal studies.
Collapse
Affiliation(s)
- Sarah E Hargett
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX, USA
| | - Elaine F Leslie
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX, USA
| | - Hector O Chapa
- Medical Education, College of Medicine, Texas A&M University, Bryan, TX, USA
| | - Akhilesh K Gaharwar
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX, USA.
- Interdisciplinary Program in Genetics and Genomics, Texas A&M University, College Station, TX, USA.
- Department of Material Science and Engineering, College of Engineering, Texas A&M University, College Station, TX, USA.
- Center for Remote Health Technologies and Systems, Texas A&M University, College Station, TX, USA.
| |
Collapse
|
18
|
Lopes B, Coelho A, Alvites R, Sousa AC, Sousa P, Moreira A, Atayde L, Salgado A, Geuna S, Maurício AC. Animal models in peripheral nerve transection studies: a systematic review on study design and outcomes assessment. Regen Med 2024; 19:189-203. [PMID: 37855207 DOI: 10.2217/rme-2023-0102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2023] Open
Abstract
Aim: Peripheral nerve injury regeneration studies using animal models are crucial to different pre-clinical therapeutic approaches efficacy evaluation whatever the surgical technique explored. Materials & methods: A 944 articles systematic review on 'peripheral nerve injury in animal models' over the last 9 years was carried out. Results: It was found that 91% used rodents, and only 9% employed large animals. Different nerves are studied, with generated gaps (10,78 mm) and methods applied for regeneration evaluation uniformed. Sciatic nerve was the most used (88%), followed by median and facial nerves (2.6%), significantly different. Conclusion: There has not been a significant scale-up of the in vivo testing to large animal models (anatomically/physiologically closer to humans), allowing an improvement in translational medicine for clinical cases.
Collapse
Affiliation(s)
- Bruna Lopes
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, Porto, 4051-401, Portugal
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, No. 228, Porto, 4050-313, Portugal
- Associate Laboratory for Animal & Veterinary Science (AL4AnimalS), Lisboa, 1300-477, Portugal
| | - André Coelho
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, Porto, 4051-401, Portugal
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, No. 228, Porto, 4050-313, Portugal
- Associate Laboratory for Animal & Veterinary Science (AL4AnimalS), Lisboa, 1300-477, Portugal
| | - Rui Alvites
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, Porto, 4051-401, Portugal
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, No. 228, Porto, 4050-313, Portugal
- Associate Laboratory for Animal & Veterinary Science (AL4AnimalS), Lisboa, 1300-477, Portugal
- Instituto Universitário de Ciências da Saúde (CESPU), Avenida Central de Gandra 1317, Gandra, Paredes, 4585-116, Portugal
| | - Ana Catarina Sousa
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, Porto, 4051-401, Portugal
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, No. 228, Porto, 4050-313, Portugal
- Associate Laboratory for Animal & Veterinary Science (AL4AnimalS), Lisboa, 1300-477, Portugal
| | - Patrícia Sousa
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, Porto, 4051-401, Portugal
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, No. 228, Porto, 4050-313, Portugal
- Associate Laboratory for Animal & Veterinary Science (AL4AnimalS), Lisboa, 1300-477, Portugal
| | - Alícia Moreira
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, Porto, 4051-401, Portugal
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, No. 228, Porto, 4050-313, Portugal
- Associate Laboratory for Animal & Veterinary Science (AL4AnimalS), Lisboa, 1300-477, Portugal
| | - Luís Atayde
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, Porto, 4051-401, Portugal
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, No. 228, Porto, 4050-313, Portugal
- Associate Laboratory for Animal & Veterinary Science (AL4AnimalS), Lisboa, 1300-477, Portugal
| | - António Salgado
- Life & Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, Braga, Portugal
- ICVS/3B's e PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Stefano Geuna
- Department of Clinical & Biological Sciences, & Cavalieri Ottolenghi Neuroscience Institute, University of Turin, Ospedale San Luigi, Orbassano, Turin, 10043, Italy
| | - Ana Colette Maurício
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, Porto, 4051-401, Portugal
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, No. 228, Porto, 4050-313, Portugal
- Associate Laboratory for Animal & Veterinary Science (AL4AnimalS), Lisboa, 1300-477, Portugal
| |
Collapse
|
19
|
Thompson LP, Song H, Hartnett J. Nicotinamide Riboside, an NAD + Precursor, Protects Against Cardiac Mitochondrial Dysfunction in Fetal Guinea Pigs Exposed to Gestational Hypoxia. Reprod Sci 2024; 31:975-986. [PMID: 37957471 PMCID: PMC10959782 DOI: 10.1007/s43032-023-01387-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 10/18/2023] [Indexed: 11/15/2023]
Abstract
Gestational hypoxia inhibits mitochondrial function in the fetal heart and placenta contributing to fetal growth restriction and organ dysfunction. NAD + deficiency may contribute to a metabolic deficit by inhibiting oxidative phosphorylation and ATP synthesis. We tested the effects of nicotinamide riboside (NR), an NAD + precursor, as a treatment for reversing known mitochondrial dysfunction in hypoxic fetal hearts. Pregnant guinea pigs were housed in room air (normoxia) or placed in a hypoxic chamber (10.5%O2) for the last 14 days of gestation (term = 65 days) and administered either water or NR (1.6 mg/ml) in the drinking bottle. Fetuses were excised at term, and NAD + levels of maternal liver, placenta, and fetal heart ventricles were measured. Indices of mitochondrial function (complex IV activity, sirtuin 3 activity, protein acetylation) and ATP synthesis were measured in fetal heart ventricles of NR-treated/untreated normoxic and hypoxic animals. Hypoxia reduced fetal body weight in both sexes (p = 0.01), which was prevented by NR. Hypoxia had no effect on maternal liver NAD + levels but decreased (p = 0.04) placenta NAD + levels, the latter normalized with NR treatment. Hypoxia had no effect on fetal heart NAD + but decreased (p < 0.05) mitochondrial complex IV and sirtuin 3 activities, ATP content, and increased mitochondrial acetylation, which were all normalized with maternal NR. Hypoxia increased (p < 0.05) mitochondrial acetylation in female fetal hearts but had no effect on other mitochondrial indices. We conclude that maternal NR is an effective treatment for normalizing mitochondrial dysfunction and ATP synthesis in the hypoxic fetal heart.
Collapse
Affiliation(s)
- Loren P Thompson
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland, Baltimore, School of Medicine, 655 W. Baltimore St., Baltimore, MD, 21201, USA.
| | - Hong Song
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland, Baltimore, School of Medicine, 655 W. Baltimore St., Baltimore, MD, 21201, USA
| | - Jamie Hartnett
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland, Baltimore, School of Medicine, 655 W. Baltimore St., Baltimore, MD, 21201, USA
| |
Collapse
|
20
|
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
|
21
|
Haq KT, McLean K, Salameh S, Swift L, Posnack NG. Electroanatomical Adaptations in the Guinea Pig Heart from Neonatal to Adulthood. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.26.577234. [PMID: 38352347 PMCID: PMC10862765 DOI: 10.1101/2024.01.26.577234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2024]
Abstract
Background Electroanatomical adaptations during the neonatal to adult phase have not been comprehensively studied in preclinical animal models. To explore the impact of age as a biological variable on cardiac electrophysiology, we employed neonatal and adult guinea pigs, which are a recognized animal model for developmental research. Methods Healthy guinea pigs were categorized into three age groups (neonates, n=10; younger adults, n=13; and older adults, n=26). Electrocardiogram (ECG) recordings were collected in vivo from anesthetized animals (2-3% isoflurane). A Langendorff-perfusion system was employed for optical assessment of epicardial action potentials and calcium transients, using intact excised heart preparations. Optical data sets were analyzed and metric maps were constructed using Kairosight 3.0. Results The allometric relationship between heart weight and body weight diminishes with age, as it is strongest at the neonatal stage (R 2 = 0.84) and completely abolished in older adults (R 2 = 1E-06). Neonatal hearts exhibit circular activation waveforms, while adults show prototypical elliptical shapes. Neonatal conduction velocity (40.6±4.0 cm/s) is slower than adults (younger adults: 61.6±9.3 cm/s; older adults: 53.6±9.2 cm/s). Neonatal hearts have a longer action potential duration (APD) and exhibit regional heterogeneity (left apex; APD30: 68.6±5.6 ms, left basal; APD30: 62.8±3.6), which was absent in adult epicardium. With dynamic pacing, neonatal hearts exhibit a flatter APD restitution slope (APD70: 0.29±0.04) compared to older adults (0.49±0.04). Similar restitution characteristics are observed with extrasystolic pacing, with a flatter slope in neonatal hearts (APD70: 0.54±0.1) compared to adults (Younger adults: 0.85±0.4; Older adults: 0.95±0.7). Finally, neonatal hearts display unidirectional excitation-contraction coupling, while adults exhibit bidirectionality. Conclusion The transition from neonatal to adulthood in guinea pig hearts is characterized by transient changes in electroanatomic properties. Age-specific patterns can influence cardiac physiology, pathology, and therapies for cardiovascular diseases. Understanding postnatal heart development is crucial to evaluating therapeutic eligibility, safety, and efficacy. What is Known Age-specific cardiac electroanatomical characteristics have been documented in humans and some preclinical animal models. These age-specific patterns can influence cardiac physiology, pathology, and therapies for cardiovascular diseases. What the Study Adds Cardiac electroanatomical characteristics are age-specific in guinea pigs, a well-known preclinical model for developmental studies. Age-dependent adaptations in cardiac electrophysiology are readily observed in the electrocardiogram recordings and via optical mapping of epicardial action potentials and calcium transients. Our findings reveal unique activation and repolarization characteristics between neonatal and adult animals.
Collapse
|
22
|
Coker SJ, Berry MJ, Vissers MCM, Dyson RM. Maternal Vitamin C Intake during Pregnancy Influences Long-Term Offspring Growth with Timing- and Sex-Specific Effects in Guinea Pigs. Nutrients 2024; 16:369. [PMID: 38337653 PMCID: PMC10857109 DOI: 10.3390/nu16030369] [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: 12/18/2023] [Revised: 01/18/2024] [Accepted: 01/25/2024] [Indexed: 02/12/2024] Open
Abstract
Our previous work in guinea pigs revealed that low vitamin C intake during preconception and pregnancy adversely affects fertility, pregnancy outcomes, and foetal and neonatal growth in a sex-dependent manner. To investigate the long-term impact on offspring, we monitored their growth from birth to adolescence (four months), recorded organ weights at childhood equivalence (28 days) and adolescence, and assessed physiological parameters like oral glucose tolerance and basal cortisol concentrations. We also investigated the effects of the timing of maternal vitamin C restriction (early vs. late gestation) on pregnancy outcomes and the health consequences for offspring. Dunkin Hartley guinea pigs were fed an optimal (900 mg/kg feed) or low (100 mg/kg feed) vitamin C diet ad libitum during preconception. Pregnant dams were then randomised into four feeding regimens: consistently optimal, consistently low, low during early pregnancy, or low during late pregnancy. We found that low maternal vitamin C intake during early pregnancy accelerated foetal and neonatal growth in female offspring and altered glucose homeostasis in the offspring of both sexes at an age equivalent to early childhood. Conversely, low maternal vitamin C intake during late pregnancy resulted in foetal growth restriction and reduced weight gain in male offspring throughout their lifespan. We conclude that altered vitamin C during development has long-lasting, sex-specific consequences for offspring and that the timing of vitamin C depletion is also critical, with low levels during early development being associated with the development of a metabolic syndrome-related phenotype, while later deprivation appears to be linked to a growth-faltering phenotype.
Collapse
Affiliation(s)
- Sharna J. Coker
- Perinatal and Developmental Physiology Group, Department of Paediatrics and Child Health, University of Otago, Wellington 6242, New Zealand; (M.J.B.); (R.M.D.)
| | - Mary J. Berry
- Perinatal and Developmental Physiology Group, Department of Paediatrics and Child Health, University of Otago, Wellington 6242, New Zealand; (M.J.B.); (R.M.D.)
| | - Margreet C. M. Vissers
- Mātai Hāora-Centre for Redox Biology and Medicine, Department of Pathology and Biomedical Science, University of Otago, Christchurch 8140, New Zealand;
| | - Rebecca M. Dyson
- Perinatal and Developmental Physiology Group, Department of Paediatrics and Child Health, University of Otago, Wellington 6242, New Zealand; (M.J.B.); (R.M.D.)
| |
Collapse
|
23
|
Canizo J, Biondic S, Lenghan KV, Petropoulos S. Guinea Pig Preimplantation Embryos: Generation, Collection, and Immunofluorescence. Methods Mol Biol 2024; 2767:275-292. [PMID: 37284942 DOI: 10.1007/7651_2023_488] [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: 06/08/2023]
Abstract
Studying various animal models is important for comparative biology and to better understand evolutionary development. Furthermore, when aiming to translate findings to human development, it is crucial to select an appropriate animal model that closely resembles the specific aspect of development under study. The guinea pig is highlighted as a useful platform for reproductive studies due to similarities in in utero development and general physiology with the human. This chapter outlines the methods required for guinea pig mating and collection of embryos for in vitro culture and molecular characterization. Specifically, this chapter provides detailed guidance on monitoring the estrus cycle to determine the mating time, performing a vaginal flush and smear to confirm successful mating, performing euthanasia of the guinea pig, and flushing in vivo embryos. Once collected, the embryos can be utilized for numerous downstream applications. Here we will cover embryo culturing and processing embryos for immunofluorescence.
Collapse
Affiliation(s)
- Jesica Canizo
- Molecular Biology Program, Université de Montréal, Montreal, QC, Canada
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montreal, QC, Canada
| | - Savana Biondic
- Molecular Biology Program, Université de Montréal, Montreal, QC, Canada
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montreal, QC, Canada
| | - Katherine Vandal Lenghan
- Molecular Biology Program, Université de Montréal, Montreal, QC, Canada
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montreal, QC, Canada
| | - Sophie Petropoulos
- Molecular Biology Program, Université de Montréal, Montreal, QC, Canada
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montreal, QC, Canada
- Département de Médecine, Université de Montréal, Montreal, QC, Canada
- Clinical Science, Investigation and Technology, Karolinska Institutet, Huddinge, Sweden
| |
Collapse
|
24
|
Wilson RL, Kropp Schmidt J, Davenport BN, Ren E, Keding LT, Shaw SA, Schotzko ML, Antony KM, Simmons HA, Golos TG, Jones HN. Maternal, placental and fetal response to a non-viral, polymeric nanoparticle gene therapy in nonhuman primates. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.16.545278. [PMID: 38168281 PMCID: PMC10760006 DOI: 10.1101/2023.06.16.545278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Background Currently, there are no placenta-targeted treatments to alter the in utero environment. Water-soluble polymers have a distinguished record of clinical relevance outside of pregnancy. We have demonstrated the effective delivery of polymer-based nanoparticles containing a non-viral human insulin-like 1 growth factor ( IGF1 ) transgene to correct placental insufficiency in small animal models of fetal growth restriction (FGR). Our goal was to extend these studies to the pregnant nonhuman primate (NHP) and assess maternal, placental and fetal responses to nanoparticle-mediated IGF1 treatment. Methods Pregnant macaques underwent ultrasound-guided intraplacental injections of nanoparticles ( GFP- or IGF1- expressing plasmid under the control of the trophoblast-specific PLAC1 promoter complexed with a HPMA-DMEAMA co-polymer) at approximately gestational day 100 (term = 165 days). Fetectomy was performed 24 h ( GFP ; n =1), 48 h ( IGF1 ; n = 3) or 10 days ( IGF1 ; n = 3) after nanoparticle delivery. Routine pathological assessment was performed on biopsied maternal tissues, and placental and fetal tissues. Maternal blood was analyzed for complete blood count (CBC), immunomodulatory proteins and growth factors, progesterone (P4) and estradiol (E2). Placental ERK/AKT/mTOR signaling was assessed using western blot and qPCR. Findings Fluorescent microscopy and in situ hybridization confirmed placental uptake and transgene expression in villous syncytiotrophoblast. No off-target expression was observed in maternal and fetal tissues. Histopathological assessment of the placenta recorded observations not necessarily related to the IGF1 nanoparticle treatment. In maternal blood, CBCs, P4 and E2 remained within the normal range for pregnant macaques across the treatment period. Changes to placental ERK and AKT signaling at 48 h and 10 d after IGF1 nanoparticle treatment indicated an upregulation in placental homeostatic mechanisms to prevent over activity in the normal pregnancy environment. Interpretation Maternal toxicity profile analysis and lack of adverse reaction to nanoparticle-mediated IGF1 treatment, combined with changes in placental signaling to maintain homeostasis indicates no deleterious impact of treatment. Funding National Institutes of Health, and Wisconsin National Primate Research Center.
Collapse
|
25
|
Le HH, Hagen MW, Louey S, Tavori H, Thornburg KL, Giraud GD, Hinds MT, Barnes AP. Development of a novel Guinea Pig model producing transgenerational endothelial transcriptional changes driven by maternal food restriction and a second metabolic insult of high fat diet. Front Physiol 2023; 14:1266444. [PMID: 37942229 PMCID: PMC10628814 DOI: 10.3389/fphys.2023.1266444] [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: 07/24/2023] [Accepted: 09/26/2023] [Indexed: 11/10/2023] Open
Abstract
Developmental programming of chronic adverse cardiovascular health outcomes has been studied both using numerous human populations and an array of animal models. However, the mechanisms that produce transgenerational effects have been difficult to study due to a lack of developmentally relevant models. As such, how increased disease risk is carried to the second generation has been poorly studied. We hypothesized that the endothelium which mediates many acute and chronic vascular inflammatory responses is a key player in these effects, and epidemiological studies implicate transgenerational nutritional effects on endothelial health. To study the mutigenerational effects of maternal undernutrition on offspring endothelial health, we developed a model of transgenerational nutritional stress in guinea pigs, a translationally relevant precocial species with a relatively short lifespan. First- and second-generation offspring were subjected to a high fat diet in adolescence to exacerbate negative cardiovascular health. To assess transcriptional changes, we performed bulk RNA-sequencing in carotid artery endothelial cells, with groups stratified as prenatal control or food restricted, and postnatal control or high fat diet. We detected statistically significant gene alterations for each dietary permutation, some of which were unique to treatments and other transcriptional signatures shared by multiple or all conditions. These findings highlight a core group of genes altered by high fat diet that is shared by all cohorts and a divergence of transgenerational effects between the prenatal ad libitum and dietary restriction groups. This study establishes the groundwork for this model to be used to better understand the interplay of prenatal stress and genetic reprogramming.
Collapse
Affiliation(s)
- Hillary H. Le
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR, United States
| | - Matthew W. Hagen
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR, United States
| | - Samantha Louey
- Center for Developmental Health, Portland, OR, United States
- Knight Cardiovascular Institute, Portland, OR, United States
| | - Hagai Tavori
- Knight Cardiovascular Institute, Portland, OR, United States
| | - Kent L. Thornburg
- Center for Developmental Health, Portland, OR, United States
- Knight Cardiovascular Institute, Portland, OR, United States
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, OR, United States
| | - George D. Giraud
- Center for Developmental Health, Portland, OR, United States
- Knight Cardiovascular Institute, Portland, OR, United States
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, OR, United States
- VA Portland Health Care System, Portland, OR, United States
| | - Monica T. Hinds
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR, United States
- Center for Developmental Health, Portland, OR, United States
| | | |
Collapse
|
26
|
Song H, Thompson LP. Effects of Gestational Hypoxia on PGC1α and Mitochondrial Acetylation in Fetal Guinea Pig Hearts. Reprod Sci 2023; 30:2996-3009. [PMID: 37138147 PMCID: PMC10556133 DOI: 10.1007/s43032-023-01245-5] [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: 02/06/2023] [Accepted: 04/20/2023] [Indexed: 05/05/2023]
Abstract
Chronic intrauterine hypoxia is a significant pregnancy complication impacting fetal heart growth, metabolism, and mitochondrial function, contributing to cardiovascular programming of the offspring. PGC1α (peroxisome proliferator-activated receptor γ co-activator 1α) is the master regulator of mitochondrial biogenesis. We investigated the effects of hypoxia on PGC1α expression following exposure at different gestational ages. Time-mated pregnant guinea pigs were exposed to normoxia (NMX, 21% O2) or hypoxia (HPX, 10.5% O2) at either 25-day (early-onset) or 50-day (late-onset) gestation, and all fetuses were extracted at term (term = ~65-day gestation). Expression of nuclear PGC1α, sirtuin 1 (SIRT1), AMP-activated protein kinase (AMPK), and mitochondrial sirtuin 3 (SIRT3) was measured, along with SIRT3 activity and mitochondrial acetylation of heart ventricles of male and female fetuses. Early-onset hypoxia increased (P<0.05) fetal cardiac nuclear PGC1α and had no effect on mitochondrial acetylation of either growth-restricted males or females. Late-onset hypoxia had either no effect or decreased (P<0.05) PCC1α expression in males and females, respectively, but increased (P<0.05) mitochondrial acetylation in both sexes. Hypoxia had variable effects on expression of SIRT1, AMPK, SIRT3, and SIRT3 activity depending on the sex. The capacity of the fetal heart to respond to hypoxia differs depending on the gestational age of exposure and sex of the fetus. Further, the effects of late-onset hypoxia on fetal heart function impose a greater risk to male than female fetuses, which has implications toward cardiovascular programming effects of the offspring.
Collapse
Affiliation(s)
- Hong Song
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland, Baltimore, School of Medicine, 655 W. Baltimore St., Baltimore, MD, 21201, USA
| | - Loren P Thompson
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland, Baltimore, School of Medicine, 655 W. Baltimore St., Baltimore, MD, 21201, USA.
| |
Collapse
|
27
|
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
|
28
|
Coker SJ, Dyson RM, Smith-Díaz CC, Vissers MCM, Berry MJ. Effects of Low Vitamin C Intake on Fertility Parameters and Pregnancy Outcomes in Guinea Pigs. Nutrients 2023; 15:4107. [PMID: 37836389 PMCID: PMC10574174 DOI: 10.3390/nu15194107] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 09/21/2023] [Accepted: 09/21/2023] [Indexed: 10/15/2023] Open
Abstract
Identifying how specific nutrients can impact fertility, pregnancy, and neonatal outcomes will yield important insights into the biological mechanisms linking diet and reproductive health. Our study investigates how dietary vitamin C intake affects various fertility parameters and pregnancy and neonatal outcomes in the guinea pig, a natural model of vitamin C dependency. Dunkin Hartley guinea pigs were fed an optimal (900 mg/kg feed) or low (100 mg/kg feed) vitamin C diet ad libitum for at least three weeks prior to mating and throughout pregnancy. We found that animals receiving the low vitamin C diet had an increased number of unsuccessful matings, a higher incidence of foetal reabsorption, and, among pregnancies resulting in delivery at term, produced fewer offspring. Neonates from mothers on the low vitamin C diet had significantly decreased plasma vitamin C concentrations at birth and exhibited mild growth impairments in a sex-dependent manner. We conclude that a diet low of vitamin C induces a state of subfertility, reduces overall fecundity, and adversely impacts both pregnancy outcomes and growth in the offspring. Our study provides an essential foundation for future investigations to determine whether these findings translate to humans. If so, they could have important clinical implications for assisted reproductive technologies and nutritional recommendations for couples trying to conceive, pregnant women, and breastfeeding mothers.
Collapse
Affiliation(s)
- Sharna J. Coker
- Perinatal and Developmental Physiology Group, Department of Paediatrics and Child Health, University of Otago, Wellington 6242, New Zealand; (S.J.C.); (R.M.D.)
| | - Rebecca M. Dyson
- Perinatal and Developmental Physiology Group, Department of Paediatrics and Child Health, University of Otago, Wellington 6242, New Zealand; (S.J.C.); (R.M.D.)
| | - Carlos C. Smith-Díaz
- Mātai Hāora—Centre for Redox Biology and Medicine, Department of Pathology and Biomedical Science, University of Otago, Christchurch 8140, New Zealand;
| | - Margreet C. M. Vissers
- Mātai Hāora—Centre for Redox Biology and Medicine, Department of Pathology and Biomedical Science, University of Otago, Christchurch 8140, New Zealand;
| | - Mary J. Berry
- Perinatal and Developmental Physiology Group, Department of Paediatrics and Child Health, University of Otago, Wellington 6242, New Zealand; (S.J.C.); (R.M.D.)
| |
Collapse
|
29
|
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
|
30
|
Casciaro C, Hamada H, Kostaki A, Matthews SG. Glucocorticoid exposure modifies the miRNA profile of sperm in the guinea pig: Implications for intergenerational transmission. FASEB J 2023; 37:e22879. [PMID: 36928999 DOI: 10.1096/fj.202201784r] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 02/27/2023] [Accepted: 03/02/2023] [Indexed: 03/18/2023]
Abstract
Approximately 1%-3% of the adult population are treated with synthetic glucocorticoids (sGCs) for a variety of conditions. Studies have demonstrated that adversities experienced by males prior to conception may lead to abnormal neuroendocrine function and behaviors in offspring and that epigenetic factors including microRNA (miRNA) within sperm may be responsible for driving these effects. However, it remains unclear where in the epididymis sperm miRNA changes are occurring. Here, we hypothesized that sGC exposure will alter the miRNA profile of sperm in the epididymis in a region-specific manner. Adult male guinea pigs were exposed to regular drinking water (Ctrl) or water with the sGC dexamethasone (Dex; 3mg/kg) (n = 6/group) every other day for 48 days. Sperms were collected from epididymal seminal fluid in the caput and cauda regions of the epididymis and total RNA was extracted. miRNAs were assessed by miRNA 4.0 microarray; data were processed by TAC 4.0.1 and R. miRNA analysis revealed one miRNA in the caput that was significantly decreased by Dex in sperm. In the cauda, 31 miRNAs were reduced in sperm following Dex-exposure. The findings of this study demonstrate that Dex-exposure influences miRNA profile of sperm in the cauda but not the caput of the epididymis. This suggests that glucocorticoids target the epididymis to modify sperm miRNA and do not modify the miRNA content during spermiation in the testes.
Collapse
Affiliation(s)
- Christopher Casciaro
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Hirotaka Hamada
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Department of Gynecology and Obstetrics, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Alisa Kostaki
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Stephen G Matthews
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Department of Obstetrics and Gynecology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
| |
Collapse
|
31
|
Crombie GK, Palliser HK, Shaw JC, Hanley BA, Moloney RA, Hirst JJ. Prenatal Stress Induces Translational Disruption Associated with Myelination Deficits. Dev Neurosci 2023; 45:290-308. [PMID: 37004512 DOI: 10.1159/000530282] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Accepted: 03/03/2023] [Indexed: 04/03/2023] Open
Abstract
Disruptions to neurodevelopment are known to be linked to behavioral disorders in childhood and into adulthood. The fetal brain is extremely vulnerable to stimuli that alter inhibitory GABAergic pathways and critical myelination processes, programing long-term neurobehavioral disruption. The maturation of the GABAergic system into the major inhibitory pathway in the brain and the development of oligodendrocytes into mature cells capable of producing myelin are integral components of optimal neurodevelopment. The current study aimed to elucidate prenatal stress-induced mechanisms that disrupt these processes and to delineate the role of placental pathways in these adverse outcomes. Pregnant guinea pig dams were exposed to prenatal stress with strobe light exposure for 2 h/day on gestational age (GA) 35, 40, 45, 50, 55, 60, and 65, and groups of fetuses and placentae were collected after the stress exposure on GA40, GA50, GA60, and GA69 (term). Fetal plasma, placental, and brain tissue were collected for allopregnanolone and cortisol quantification with ELISA. Relative mRNA expression of genes of specific pathways of interest was examined with real-time PCR in placental and hippocampal tissue, and myelin basic protein (MBP) was quantified immunohistochemically in the hippocampus and surrounding regions for assessment of mature myelin. Prenatal stress in mid-late gestation resulted in disruptions to the translational machinery responsible for the production of myelin and decreased myelin coverage in the hippocampus and surrounding regions. The male placenta showed an initial protective increase in allopregnanolone concentrations in response to maternal psychosocial stress. The male and female placentae had a sex-dependent increase in neurosteroidogenic enzymes at term following prenatal stress. Independent from exposure to prenatal stress, at gestational day 60 - a critical period for myelin development, the placentae of female fetuses had increased capability of preventing cortisol transfer to the fetus through expression of 11-beta-hydroxysteroid dehydrogenase types 1 and 2. The deficits early in the process of maturation of myelination indicate that the reduced myelination observed at childhood equivalence in previous studies begins in fetal life. This negative programing persists into childhood, potentially due to dysregulation of MBP translation processes. Expression patterns of neurosteroidogenic enzymes in the placenta at term following stress may identify at-risk fetuses that have been exposed to a stressful in utero environment.
Collapse
Affiliation(s)
- Gabrielle K Crombie
- Mothers and Babies Research Centre, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, New South Wales, Australia
| | - Hannah K Palliser
- Mothers and Babies Research Centre, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, New South Wales, Australia
| | - Julia C Shaw
- Mothers and Babies Research Centre, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, New South Wales, Australia
| | - Bethany A Hanley
- Mothers and Babies Research Centre, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, New South Wales, Australia
| | - Roisin A Moloney
- Mothers and Babies Research Centre, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, New South Wales, Australia
| | - Jonathan J Hirst
- Mothers and Babies Research Centre, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, New South Wales, Australia
| |
Collapse
|
32
|
Kang M, Blenkiron C, Chamley L. The biodistribution of placental and fetal extracellular vesicles during pregnancy following placentation. Clin Sci (Lond) 2023; 137:385-399. [PMID: 36920079 PMCID: PMC10017278 DOI: 10.1042/cs20220301] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 02/15/2023] [Accepted: 02/20/2023] [Indexed: 03/16/2023]
Abstract
Human pregnancy is a highly orchestrated process requiring extensive cross-talk between the mother and the fetus. Extracellular vesicles released by the fetal tissue, particularly the placenta, are recognized as important mediators of this process. More recently, the importance of placental extracellular vesicle biodistribution studies in animal models has received increasing attention as identifying the organs to which extracellular vesicles are targeted to helps us understand more about this communication system. Placental extracellular vesicles are categorized based on their size into macro-, large-, and small-extracellular vesicles, and their biodistribution is dependent on the extracellular vesicle's particle size, the direction of blood flow, the recirculation of blood, as well as the retention capacity in organs. Macro-extracellular vesicles are exclusively localized to the lungs, while large- and small-extracellular vesicles show high levels of distribution to the lungs and liver, while there is inconsistency in the reporting of distribution to the spleen and kidneys. This inconsistency may be due to the differences in the methodologies employed between studies and their limitations. Future studies should incorporate analysis of placental extracellular vesicle biodistribution at the macroscopic level on whole animals and organs/tissues, as well as the microscopic cellular level.
Collapse
Affiliation(s)
- Matthew Kang
- Department of Obstetrics and Gynaecology, University of Auckland, 1023, Auckland, New Zealand
- Correspondence: Matt Kang ()
| | - Cherie Blenkiron
- Department of Obstetrics and Gynaecology, University of Auckland, 1023, Auckland, New Zealand
- Hub for Extracellular Vesicle Investigations (HEVI), University of Auckland, 1023, Auckland, New Zealand
- Auckland Cancer Society Research Center (ACSRC), University of Auckland, 1023, Auckland, New Zealand
- Molecular Medicine and Pathology, University of Auckland, 1023, Auckland, New Zealand
| | - Lawrence W. Chamley
- Department of Obstetrics and Gynaecology, University of Auckland, 1023, Auckland, New Zealand
- Hub for Extracellular Vesicle Investigations (HEVI), University of Auckland, 1023, Auckland, New Zealand
| |
Collapse
|
33
|
Genzer SC, Flietstra T, Coleman-McCray JD, Tansey C, Welch SR, Spengler JR. Effect of Parental Age, Parity, and Pairing Approach on Reproduction in Strain 13/N Guinea Pigs ( Cavia porcellus). Animals (Basel) 2023; 13:ani13050895. [PMID: 36899751 PMCID: PMC10000240 DOI: 10.3390/ani13050895] [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: 01/26/2023] [Revised: 02/27/2023] [Accepted: 02/27/2023] [Indexed: 03/06/2023] Open
Abstract
Guinea pigs are important animal models for human disease, and both outbred and inbred lines are utilized in biomedical research. The optimal maintenance of guinea pig colonies, commercially and in research settings, relies on robust informed breeding programs, however, breeding data on specialized inbred strains are limited. Here, we investigated the effects of parental age, parity, and pairing approaches on mean total fetus count, percentage of female pups in the litter, and pup survival rate after 10 days in strain 13/N guinea pigs. Our analysis of colony breeding data indicates that the average litter size is 3.3 pups, with a 25.2% stillbirth rate, a failure-to-thrive outcome in 5.1% of pups, and a 10 day survival rate of 69.7%. The only variable to significantly affect the reproductive outcomes examined was parental age (p < 0.05). In comparison to adults, both juvenile and geriatric sows had lower total fetus counts; juvenile boars had a higher percentage of females in litters, and geriatric boars had a lower 10 day survival rate of pups. These studies provide valuable information regarding the reproductive characteristics of strain 13/N guinea pigs, and support a variety of breeding approaches without significant effects on breeding success.
Collapse
Affiliation(s)
- Sarah C. Genzer
- Comparative Medicine Branch, Division of Scientific Resources, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA
| | - Timothy Flietstra
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA
| | - JoAnn D. Coleman-McCray
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA
| | - Cassandra Tansey
- Comparative Medicine Branch, Division of Scientific Resources, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA
| | - Stephen R. Welch
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA
| | - Jessica R. Spengler
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA
- Correspondence:
| |
Collapse
|
34
|
Sixtus RP, Berry MJ, Gray CL, Dyson RM. A novel whole-body thermal stress test for monitoring cardiovascular responses in Guinea pigs. J Therm Biol 2023; 113:103500. [PMID: 37055107 DOI: 10.1016/j.jtherbio.2023.103500] [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] [Received: 05/09/2022] [Revised: 01/27/2023] [Accepted: 02/04/2023] [Indexed: 03/12/2023]
Abstract
Cardiovascular disease is a leading cause of morbidity and mortality worldwide. Stress tests are frequently employed to expose early signs of cardiovascular dysfunction or disease and can be employed, for example, in the context of preterm birth. We aimed to establish a safe and effective thermal stress test to examine cardiovascular function. Guinea pigs were anaesthetized using a 0.8% isoflurane, 70% N2O mix. ECG, non-invasive blood pressure, laser Doppler flowmetry, respiratory rate, and an array of skin and rectal thermistors were applied. A physiologically relevant heating and a cooling thermal stress test was developed. Upper and lower thermal limits for core body temperature were set at 41.5 OC and 34 OC, for the safe recovery of animals. This protocol therefore presents a viable thermal stress test for use in guinea pig models of health and disease that facilitates exploration of whole-system cardiovascular function.
Collapse
Affiliation(s)
- Ryan P Sixtus
- Department of Paediatrics and Child Health, University of Otago, Wellington, New Zealand.
| | - Mary J Berry
- Department of Paediatrics and Child Health, University of Otago, Wellington, New Zealand
| | - Clint L Gray
- Department of Paediatrics and Child Health, University of Otago, Wellington, New Zealand
| | - Rebecca M Dyson
- Department of Paediatrics and Child Health, University of Otago, Wellington, New Zealand
| |
Collapse
|
35
|
Candia AA, Jiménez T, Navarrete Á, Beñaldo F, Silva P, García-Herrera C, Sferruzzi-Perri AN, Krause BJ, González-Candia A, Herrera EA. Developmental Ultrasound Characteristics in Guinea Pigs: Similarities with Human Pregnancy. Vet Sci 2023; 10:144. [PMID: 36851448 PMCID: PMC9963037 DOI: 10.3390/vetsci10020144] [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: 12/02/2022] [Revised: 01/27/2023] [Accepted: 02/07/2023] [Indexed: 02/12/2023] Open
Abstract
BACKGROUND Biometrical and blood flow examinations are fundamental for assessing fetoplacental development during pregnancy. Guinea pigs have been proposed as a good model to study fetal development and related gestational complications; however, longitudinal growth and blood flow changes in utero have not been properly described. This study aimed to describe fetal and placental growth and blood flow of the main intrauterine vascular beds across normal guinea pig pregnancy and to discuss the relevance of this data for human pregnancy. METHODS Pregnant guinea pigs were studied from day 25 of pregnancy until term (day ~70) by ultrasound and Doppler assessment. The results were compared to human data from the literature. RESULTS Measurements of biparietal diameter (BPD), cranial circumference (CC), abdominal circumference, and placental biometry, as well as pulsatility index determination of umbilical artery, middle cerebral artery (MCA), and cerebroplacental ratio (CPR), were feasible to determine across pregnancy, and they could be adjusted to linear or nonlinear functions. In addition, several of these parameters showed a high correlation coefficient and could be used to assess gestational age in guinea pigs. We further compared these data to ultrasound variables from human pregnancy with high similarities. CONCLUSIONS BPD and CC are the most reliable measurements to assess fetal growth in guinea pigs. Furthermore, this is the first report in which the MCA pulsatility index and CPR are described across guinea pig gestation. The guinea pig is a valuable model to assess fetal growth and blood flow distribution, variables that are comparable with human pregnancy.
Collapse
Affiliation(s)
- Alejandro A. Candia
- Laboratorio de Función y Reactividad Vascular, Programa de Fisiopatología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago 7500922, Chile
- Institute of Health Sciences, University of O’Higgins, Rancagua 2841959, Chile
| | - Tamara Jiménez
- Laboratorio de Función y Reactividad Vascular, Programa de Fisiopatología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago 7500922, Chile
| | - Álvaro Navarrete
- Departamento de Ingeniería Mecánica, Facultad de Ingeniería, Universidad de Santiago de Chile, Santiago 9170022, Chile
| | - Felipe Beñaldo
- Laboratorio de Función y Reactividad Vascular, Programa de Fisiopatología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago 7500922, Chile
| | - Pablo Silva
- Laboratorio de Función y Reactividad Vascular, Programa de Fisiopatología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago 7500922, Chile
| | - Claudio García-Herrera
- Departamento de Ingeniería Mecánica, Facultad de Ingeniería, Universidad de Santiago de Chile, Santiago 9170022, Chile
| | - Amanda N. Sferruzzi-Perri
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK
| | - Bernardo J. Krause
- Institute of Health Sciences, University of O’Higgins, Rancagua 2841959, Chile
| | - Alejandro González-Candia
- Laboratorio de Función y Reactividad Vascular, Programa de Fisiopatología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago 7500922, Chile
- Institute of Health Sciences, University of O’Higgins, Rancagua 2841959, Chile
| | - Emilio A. Herrera
- Laboratorio de Función y Reactividad Vascular, Programa de Fisiopatología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago 7500922, Chile
- International Center for Andean Studies (INCAS), University of Chile, Putre 1070000, Chile
| |
Collapse
|
36
|
Davenport BN, Jones HN, Wilson RL. Placental treatment with insulin-like growth factor 1 via nanoparticle differentially impacts vascular remodeling factors in guinea pig sub-placenta/decidua. Front Physiol 2023; 13:1055234. [PMID: 36685211 PMCID: PMC9845775 DOI: 10.3389/fphys.2022.1055234] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 12/13/2022] [Indexed: 01/06/2023] Open
Abstract
Clinically, fetal growth restriction (FGR) is only detectable in later gestation, despite pathophysiological establishment likely earlier in pregnancy. Additionally, there are no effective in utero treatment options for FGR. We have developed a nanoparticle to deliver human insulin-like 1 growth factor (hIGF-1) in a trophoblast-specific manner which results in increased expression of hIGF-1. IGF-1 signaling in the placenta regulates multiple developmental processes including trophoblast invasion and maternal vascular remodeling, both of which can be diminished in the FGR placenta. We aimed to determine the effects of short-term hIGF-1 nanoparticle treatment on sub-placenta/decidua trophoblast signaling mechanisms in FGR and under normal growth conditions. Using the guinea pig maternal nutrient restriction (MNR) model of FGR, ultrasound-guided, intra-placenta injections of hIGF-1 nanoparticle were performed at gestational day 30-33, and dams sacrificed 5 days later. Sub-placenta/decidua tissue was separated from placenta for further analyses. Western blot was used to analyze protein expression of ERK/AKT/mTOR signaling proteins (phospho-Erk (pERK), phospho-Akt (pAKT), raptor, rictor and deptor). qPCR was used to analyze gene expression of vascular/remodeling factors [vascular endothelial growth factor (Vegf), placenta growth factor (Pgf), platelet-derived growth factor (Pdgf)) and tight junction/adhesion proteins (claudin 5 (Cldn5), p-glycoprotein (Abcb1), occludin (Ocln) and tight junction protein 1 (Zo1)]. MNR reduced expression of pERK, PdgfB and Cldn5, and increased expression of Ocln and Zo1 in the sub-placenta/decidua. In MNR + hIGF1 nanoparticle sub-placenta/decidua, expression of PdgfB, Ocln and Zo1 was normalized, whilst pAkt, VegfB, Vegf receptor 1 and PdgfB receptor were increased compared to MNR. In contrast, hIGF-1 nanoparticle treatment of normal placentas reduced expression of pERK, raptor and increased expression of the mTOR inhibitor deptor. This was associated with reduced expression of VegfA, Plgf, and PdgfB. Here we have shown that the impact of hIGF-1 nanoparticle treatment is dependent on pregnancy environment. Under MNR/FGR, hIGF-1 nanoparticle treatment triggers increased expression of growth factors and normalization of EMT factors. However, under normal conditions, the response of the placenta is to decrease AKT/mTOR signaling and growth factor expression to achieve homeostasis.
Collapse
Affiliation(s)
- Baylea N. Davenport
- Center for Research in Perinatal Outcomes, University of Florida College of Medicine, Gainesville, FL, United States
- Department of Physiology and Aging, University of Florida College of Medicine, Gainesville, FL, United States
| | - Helen N. Jones
- Center for Research in Perinatal Outcomes, University of Florida College of Medicine, Gainesville, FL, United States
- Department of Physiology and Aging, University of Florida College of Medicine, Gainesville, FL, United States
| | - Rebecca L. Wilson
- Center for Research in Perinatal Outcomes, University of Florida College of Medicine, Gainesville, FL, United States
- Department of Physiology and Aging, University of Florida College of Medicine, Gainesville, FL, United States
| |
Collapse
|
37
|
Cumberland A, Hale N, Azhan A, Gilchrist CP, Chincarini G, Tolcos M. Excitatory and inhibitory neuron imbalance in the intrauterine growth restricted fetal guinea pig brain: Relevance to the developmental origins of schizophrenia and autism. Dev Neurobiol 2023; 83:40-53. [PMID: 36373424 PMCID: PMC10953391 DOI: 10.1002/dneu.22907] [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: 04/25/2022] [Revised: 10/15/2022] [Accepted: 10/27/2022] [Indexed: 11/16/2022]
Abstract
Neurodevelopmental disorders such as schizophrenia and autism are thought to involve an imbalance of excitatory and inhibitory signaling in the brain. Intrauterine growth restriction (IUGR) is a risk factor for these disorders, with IUGR onset occurring during critical periods of neurodevelopment. The aim of this study was to determine the impact of IUGR on excitatory and inhibitory neurons of the fetal neocortex and hippocampus. Fetal brains (n = 2) were first collected from an unoperated pregnant guinea pig at mid-gestation (32 days of gestation [dg]; term ∼67 dg) to visualize excitatory (Ctip2) and inhibitory (calretinin [CR] and somatostatin [SST]) neurons via immunohistochemistry. Chronic placental insufficiency (CPI) was then induced via radial artery ablation at 30 dg in another cohort of pregnant guinea pigs (n = 8) to generate IUGR fetuses (52 dg; n = 8); control fetuses (52 dg; n = 7) were from sham surgeries with no radial artery ablation. At 32 dg, Ctip2- and CR-immunoreactive (IR) cells had populated the cerebral cortex, whereas SST-IR cells had not, suggesting these neurons were yet to complete migration. At 52 dg, in IUGR versus control fetuses, there was a reduction in SST-IR cell density in the cerebral cortex (p = .0175) and hilus of the dentate gyrus (p = .0035) but not the striatum (p > .05). There was no difference between groups in the density of Ctip2-IR (cortex) or CR-IR (cortex, hippocampus) neurons (p > 0.05). Thus, we propose that an imbalance in inhibitory (SST-IR) and excitatory (Ctip2-IR) neurons in the IUGR fetal guinea pig brain could lead to excitatory/inhibitory dysfunction commonly seen in neurodevelopmental disorders such as autism and schizophrenia.
Collapse
Affiliation(s)
- Angela Cumberland
- School of Health and Biomedical SciencesRMIT UniversityBundooraVictoriaAustralia
| | - Nadia Hale
- The Ritchie Centre, Hudson Institute of Medical ResearchMonash UniversityMelbourneVictoriaAustralia
| | - Aminath Azhan
- The Ritchie Centre, Hudson Institute of Medical ResearchMonash UniversityMelbourneVictoriaAustralia
| | - Courtney P. Gilchrist
- School of Health and Biomedical SciencesRMIT UniversityBundooraVictoriaAustralia
- Victorian Infant Brain StudiesMurdoch Children's Research InstituteParkvilleVictoriaAustralia
| | - Ginevra Chincarini
- School of Health and Biomedical SciencesRMIT UniversityBundooraVictoriaAustralia
| | - Mary Tolcos
- School of Health and Biomedical SciencesRMIT UniversityBundooraVictoriaAustralia
| |
Collapse
|
38
|
Wilson RL, Lampe K, Gupta MK, Duvall CL, Jones HN. Nanoparticle-mediated transgene expression of insulin-like growth factor 1 in the growth restricted guinea pig placenta increases placenta nutrient transporter expression and fetal glucose concentrations. Mol Reprod Dev 2022; 89:540-553. [PMID: 36094907 PMCID: PMC10947605 DOI: 10.1002/mrd.23644] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/23/2022] [Accepted: 08/28/2022] [Indexed: 12/25/2022]
Abstract
Fetal growth restriction (FGR) significantly contributes to neonatal and perinatal morbidity and mortality. Currently, there are no effective treatment options for FGR during pregnancy. We have developed a nanoparticle gene therapy targeting the placenta to increase expression of human insulin-like growth factor 1 (hIGF1) to correct fetal growth trajectories. Using the maternal nutrient restriction guinea pig model of FGR, an ultrasound-guided, intraplacental injection of nonviral, polymer-based hIGF1 nanoparticle containing plasmid with the hIGF1 gene and placenta-specific Cyp19a1 promotor was administered at mid-pregnancy. Sustained hIGF1 expression was confirmed in the placenta 5 days after treatment. Whilst increased hIGF1 did not change fetal weight, circulating fetal glucose concentration were 33%-67% higher. This was associated with increased expression of glucose and amino acid transporters in the placenta. Additionally, hIGF1 nanoparticle treatment increased the fetal capillary volume density in the placenta, and reduced interhaemal distance between maternal and fetal circulation. Overall, our findings, that trophoblast-specific increased expression of hIGF1 results in changes to glucose transporter expression and increases fetal glucose concentrations within a short time period, highlights the translational potential this treatment could have in correcting impaired placental nutrient transport in human pregnancies complicated by FGR.
Collapse
Affiliation(s)
- Rebecca L. Wilson
- Center for Research in Perinatal Outcomes, University of Florida College of Medicine, Gainesville, Florida, USA
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Kristin Lampe
- Center for Fetal and Placental Research, Cincinnati Children’s Hospital and Medical Center, Cincinnati, Ohio, USA
| | - Mukesh K. Gupta
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
| | - Craig L. Duvall
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
| | - Helen N. Jones
- Center for Research in Perinatal Outcomes, University of Florida College of Medicine, Gainesville, Florida, USA
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida, USA
| |
Collapse
|
39
|
Sethi S, Friesen-Waldner LJ, Wade TP, Courchesne M, Nygard K, Sarr O, Sutherland B, Regnault TRH, McKenzie CA. Feasibility of MRI Quantification of Myelin Water Fraction in the Fetal Guinea Pig Brain. J Magn Reson Imaging 2022; 57:1856-1864. [PMID: 36239714 DOI: 10.1002/jmri.28482] [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] [Received: 07/25/2022] [Revised: 09/29/2022] [Accepted: 09/30/2022] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Fetal myelination assessment is important for understanding neurodevelopment and neurodegeneration. Myelin water imaging (MWI) quantifies myelin water fraction (MWF), a validated marker for myelin content, and has been used to assess brain myelin in children and neonates. PURPOSE To demonstrate that MWI can quantify MWF in fetal guinea pigs (GPs). STUDY TYPE Animal model. ANIMAL MODEL Nine pregnant, Dunkin-Hartley GPs with 31 fetuses (mean ± standard deviation = 60 ± 1.5 days gestation). FIELD STRENGTH/SEQUENCE 3D spoiled gradient echo and balanced steady-state free precession sequences at 3.0 T. ASSESSMENT MWF maps were reconstructed for maternal and fetal GP brains using the multicomponent driven equilibrium single pulse observation of T1 and T2 (mcDESPOT) approach. Myelin basic protein (MBP) stain provided histological validation of the MWF. Regions of interest were placed in the maternal corpus callosum (CC), maternal fornix (FOR), fetal CC, and fetal FOR in MWF maps and MBP stains. STATISTICAL TESTS Linear regression between MWF and MBP stain intensity (SI) of all four regions (coefficient of determination, R2 ). A paired t-test compared the MWF of maternal and mean fetal CC, MBP SI of maternal and mean fetal CC, MWF of maternal and mean fetal FOR, MBP SI of maternal and mean fetal FOR. A paired t-test with a linear mixed model compared the MWF of fetal CC and fetal FOR, and MBP SI of fetal CC and fetal FOR. A P value < 0.0083 was considered statistically significant. RESULTS The mean MWF of the analyzed regions are as follows (mean ± standard deviation): 0.338 + 0.016 (maternal CC), 0.340 ± 0.017 (maternal FOR), 0.214 ± 0.016 (fetal CC), and 0.305 ± 0.025 (fetal FOR). MWF correlated with MBP SI in all regions (R2 = 0.81). Significant differences were found between MWF and MBP SI of maternal and fetal CC, and MWF and MBP SI of fetal CC and fetal FOR. DATA CONCLUSION This study demonstrated the feasibility of MWI in assessing fetal brain myelin content. EVIDENCE LEVEL 2 Technical Efficacy: Stage 1.
Collapse
Affiliation(s)
- Simran Sethi
- Department of Medical Biophysics, Western University, London, Ontario, Canada
| | | | - Trevor P Wade
- Department of Medical Biophysics, Western University, London, Ontario, Canada.,Robarts Research Institute, Western University, London, Ontario, Canada
| | - Marc Courchesne
- Biotron Experimental Climate Change Research Centre, Western University, London, Ontario, Canada
| | - Karen Nygard
- Biotron Experimental Climate Change Research Centre, Western University, London, Ontario, Canada
| | - Ousseynou Sarr
- Department of Physiology & Pharmacology, Western University, London, Ontario, Canada
| | - Brian Sutherland
- Department of Physiology & Pharmacology, Western University, London, Ontario, Canada
| | - Timothy R H Regnault
- Department of Physiology & Pharmacology, Western University, London, Ontario, Canada.,Department of Obstetrics & Gynaecology, Western University, London, Ontario, Canada.,Division of Maternal, Fetal, and Newborn Health, Children's Health Research Institute, Lawson Health Research Institute, Western University, London, Ontario, Canada
| | - Charles A McKenzie
- Department of Medical Biophysics, Western University, London, Ontario, Canada.,Robarts Research Institute, Western University, London, Ontario, Canada.,Division of Maternal, Fetal, and Newborn Health, Children's Health Research Institute, Lawson Health Research Institute, Western University, London, Ontario, Canada
| |
Collapse
|
40
|
Navarrete Á, Varela P, López M, García-Herrera CM, Celentano DJ, Krause B. Characterization of the active response of a guinea pig carotid artery. Front Bioeng Biotechnol 2022; 10:924019. [PMID: 36091433 PMCID: PMC9458959 DOI: 10.3389/fbioe.2022.924019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 07/15/2022] [Indexed: 11/13/2022] Open
Abstract
This work presents a characterization of the active response of the carotid artery of guinea pig fetuses through a methodology that encompasses experiments, modeling and numerical simulation. To this end, the isometric contraction test is carried out in ring samples subjected to different levels of KCl concentrations and pre-stretching. Then, a coupled mechanochemical model, aimed at describing the smooth cell behavior and its influence on the passive and active mechanical response of the vascular tissue, is calibrated from the experimental measurements. Due to the complex stress and strain fields developed in the artery, a finite element numerical simulation of the test is performed to fit the model parameters, where those related to the phosphorylation and dephosphorylation activity along with the load-bearing capacity of the myosin cross-bridges are found to be the most predominant when sensitizing the active response. The main strengths of the model are associated with the prediction of the stationary state of the active mechanical response of the tissue through a realistic description of the mechanochemical process carried out at its cellular level.
Collapse
Affiliation(s)
- Álvaro Navarrete
- Departamento de Ingeniería Mecánica, Universidad de Santiago de Chile, Santiago, Chile
| | - Pablo Varela
- Departamento de Ingeniería Mecánica, Universidad de Santiago de Chile, Santiago, Chile
| | - Miguel López
- Departamento de Ingeniería Mecánica, Universidad de Santiago de Chile, Santiago, Chile
| | - Claudio M. García-Herrera
- Departamento de Ingeniería Mecánica, Universidad de Santiago de Chile, Santiago, Chile
- *Correspondence: Claudio M. García-Herrera,
| | - Diego J. Celentano
- Departamento de Ingeniería Mecánica y Metalúrgica, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Bernardo Krause
- Instituto de Ciencias de la Salud, Universidad de O’Higgins, Rancagua, Chile
| |
Collapse
|
41
|
Sze Y, Brunton PJ. Neurosteroids and early-life programming: An updated perspective. CURRENT OPINION IN ENDOCRINE AND METABOLIC RESEARCH 2022; 25:100367. [PMID: 36561280 PMCID: PMC7613978 DOI: 10.1016/j.coemr.2022.100367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Early-life stress can lead to detrimental offspring outcomes, including an increased risk for mood disorders and hypothalamic-pituitary-adrenal axis dysregulation. Neurosteroids bind to ligand-gated neurotransmitter receptors, rapidly modulating neuronal excitability and promoting termination of stress responses. Reduced neurosteroidogenesis underlies some of the aberrant neuroendocrine and behavioural phenotypes observed in adult prenatally stressed rodents. During development, disruptions in neurosteroid generation and action also lead to long-term programming effects on the off-spring's brain and behaviour. Here, we review recent advances in the field, focusing on the interaction between neurosteroids and early-life stress outcomes in adulthood and in the perinatal period. We also discuss the direction of future research, with emphasis on quantification methods, sex differences, and neurosteroids as targets for therapeutic intervention.
Collapse
Affiliation(s)
- Ying Sze
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, EH8 9XD, Scotland, UK
| | - Paula J Brunton
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, EH8 9XD, Scotland, UK
| |
Collapse
|
42
|
Davenport BN, Wilson RL, Jones HN. Interventions for placental insufficiency and fetal growth restriction. Placenta 2022; 125:4-9. [PMID: 35414477 PMCID: PMC10947607 DOI: 10.1016/j.placenta.2022.03.127] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 03/09/2022] [Accepted: 03/28/2022] [Indexed: 01/16/2023]
Abstract
Pregnancy complications adversely impact both mother and/or fetus throughout the lifespan. Fetal growth restriction (FGR) occurs when a fetus fails to reach their intrauterine potential for growth, it is the second highest leading cause of infant mortality, and leads to increased risk of developing non-communicable diseases in later life due 'fetal programming'. Abnormal placental development, growth and/or function underlies approximately 75% of FGR cases and there is currently no treatment save delivery, often prematurely. We previously demonstrated in a murine model of FGR that nanoparticle mediated, intra-placental human IGF-1 gene therapy maintains normal fetal growth. Multiple models of FGR currently exist reflecting the etiologies of human FGR and have been used by us and others to investigate the development of in utero therapeutics as discussed here. In addition to the in vivo models discussed herein, utilizing human models including in vitro (Choriocarcinoma cell lines and primary trophoblasts) and ex vivo (term villous fragments and placenta cotyledon perfusion) we have demonstrated robust nanoparticle uptake, transgene expression, nutrient transporter regulation without transfer to the fetus. For translational gene therapy application in the human placenta, there are multiple avenues that require investigation including syncytial uptake from the maternal circulation, transgene expression, functionality and longevity of treatment, impact of treatment on the mother and developing fetus. The potential impact of treating the placenta during gestation is high, wide-ranging across pregnancy complications, and may offer reduced risk of developing associated cardio-metabolic diseases in later life impacting at both an individual and societal level.
Collapse
Affiliation(s)
- Baylea N Davenport
- Center for Research in Perinatal Outcomes, University of Florida College of Medicine, United States
| | - Rebecca L Wilson
- Center for Research in Perinatal Outcomes, University of Florida College of Medicine, United States
| | - Helen N Jones
- Center for Research in Perinatal Outcomes, University of Florida College of Medicine, United States.
| |
Collapse
|
43
|
S. Aljaser F. Cryopreservation Methods and Frontiers in the Art of Freezing Life in Animal Models. Vet Med Sci 2022. [DOI: 10.5772/intechopen.101750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
The development in cryobiology in animal breeding had revolutionized the field of reproductive medicine. The main objective to preserve animal germplasm stems from variety of reasons such as conservation of endangered animal species, animal diversity, and an increased demand of animal models and/or genetically modified animals for research involving animal and human diseases. Cryopreservation has emerged as promising technique for fertility preservation and assisted reproduction techniques (ART) for production of animal breeds and genetically engineered animal species for research. Slow rate freezing and rapid freezing/vitrification are the two main methods of cryopreservation. Slow freezing is characterized by the phase transition (liquid turning into solid) when reducing the temperature below freezing point. Vitrification, on the other hand, is a phenomenon in which liquid solidifies without the formation of ice crystals, thus the process is referred to as a glass transition or ice-free cryopreservation. The vitrification protocol applies high concentrations of cryoprotective agents (CPA) used to avoid cryoinjury. This chapter provides a brief overview of fundamentals of cryopreservation and established methods adopted in cryopreservation. Strategies involved in cryopreserving germ cells (sperm and egg freezing) are included in this chapter. Last section describes the frontiers and advancement of cryopreservation in some of the important animal models like rodents (mouse and rats) and in few large animals (sheep, cow etc).
Collapse
|
44
|
Adaptations in the Hippocampus during the Fetal to Neonatal Transition in Guinea Pigs. REPRODUCTIVE MEDICINE 2022. [DOI: 10.3390/reprodmed3020008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
(Background) The transition from in utero to ex utero life is associated with rapid changes in the brain that are both protective and required for newborn functional activities, allowing adaption to the changing environment. The current study aimed to reveal new insights into adaptations required for normal ongoing brain development and function after birth. (Methods) Time-mated dams were randomly allocated to fetal collection at gestational age 68 or spontaneous term delivery followed by neonatal collection within 24 h of birth. Immunohistochemistry was performed to examine mature myelin formation and neuronal nuclei coverage. RT-PCR was used to quantify the mRNA expression of key markers of the oligodendrocyte lineage, neuronal development, and GABAergic/glutamatergic pathway maturation. (Results) Mature myelin was reduced in the subcortical white matter of the neonate, whilst neuronal nuclei coverage was increased in both the hippocampus and the overlying cortical region. Increased mRNA expression in neonates was observed for oligodendrocyte and neuronal markers. There were also widespread mRNA changes across the inhibitory GABAergic and excitatory glutamatergic pathways in neonates. (Conclusions) This study has identified important adaptations in the expression of key neurodevelopmental structures, including oligodendrocytes and neurons, that may be essential for appropriate transition in neurodevelopment to the postnatal period.
Collapse
|
45
|
Tain YL, Hsu CN. Hypertension of Developmental Origins: Consideration of Gut Microbiome in Animal Models. Biomedicines 2022; 10:biomedicines10040875. [PMID: 35453625 PMCID: PMC9030804 DOI: 10.3390/biomedicines10040875] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 03/29/2022] [Accepted: 04/08/2022] [Indexed: 11/29/2022] Open
Abstract
Hypertension is the leading cause of global disease burden. Hypertension can arise from early life. Animal models are valuable for giving cogent evidence of a causal relationship between various environmental insults in early life and the hypertension of developmental origins in later life. These insults consist of maternal malnutrition, maternal medical conditions, medication use, and exposure to environmental chemicals/toxins. There is a burgeoning body of evidence on maternal insults can shift gut microbiota, resulting in adverse offspring outcomes later in life. Emerging evidence suggests that gut microbiota dysbiosis is involved in hypertension of developmental origins, while gut microbiota-targeted therapy, if applied early, is able to help prevent hypertension in later life. This review discusses the innovative use of animal models in addressing the mechanisms behind hypertension of developmental origins. We will also highlight the application of animal models to elucidate how the gut microbiota connects with other core mechanisms, and the potential of gut microbiota-targeted therapy as a novel preventive strategy to prevent hypertension of developmental origins. These animal models have certainly enhanced our understanding of hypertension of developmental origins, closing the knowledge gap between animal models and future clinical translation.
Collapse
Affiliation(s)
- You-Lin Tain
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan;
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan
| | - Chien-Ning Hsu
- Department of Pharmacy, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
- School of Pharmacy, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Correspondence: ; Tel.: +886-975-368-975; Fax: +886-7733-8009
| |
Collapse
|
46
|
Sex-specific alterations in hepatic cholesterol metabolism in low birth weight adult guinea pigs. Pediatr Res 2022; 91:1078-1089. [PMID: 34230622 DOI: 10.1038/s41390-021-01491-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 02/19/2021] [Accepted: 02/26/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND Intrauterine growth restriction and low birth weight (LBW) have been widely reported as an independent risk factor for adult hypercholesterolaemia and increased hepatic cholesterol in a sex-specific manner. However, the specific impact of uteroplacental insufficiency (UPI), a leading cause of LBW in developed world, on hepatic cholesterol metabolism in later life, is ill defined and is clinically relevant in understanding later life liver metabolic health trajectories. METHODS Hepatic cholesterol, transcriptome, cholesterol homoeostasis regulatory proteins, and antioxidant markers were studied in UPI-induced LBW and normal birth weight (NBW) male and female guinea pigs at 150 days. RESULTS Hepatic free and total cholesterol were increased in LBW versus NBW males. Transcriptome analysis of LBW versus NBW livers revealed that "cholesterol metabolism" was an enriched pathway in LBW males but not in females. Microsomal triglyceride transfer protein and cytochrome P450 7A1 protein, involved in hepatic cholesterol efflux and catabolism, respectively, and catalase activity were decreased in LBW male livers. Superoxide dismutase activity was reduced in LBW males but increased in LBW females. CONCLUSIONS UPI environment is associated with a later life programed hepatic cholesterol accumulation via impaired cholesterol elimination in a sex-specific manner. These programmed alterations could underlie later life cholesterol-induced hepatic lipotoxicity in LBW male offspring. IMPACT Low birth weight (LBW) is a risk factor for increased hepatic cholesterol. Uteroplacental insufficiency (UPI) resulting in LBW increased hepatic cholesterol content, altered hepatic expression of cholesterol metabolism-related genes in young adult guinea pigs. UPI-induced LBW was also associated with markers of a compromised hepatic cholesterol elimination process and failing antioxidant system in young adult guinea pigs. These changes, at the current age studied, were sex-specific, only being observed in LBW males and not in LBW females. These programmed alterations could lead to further hepatic damage and greater predisposition to liver diseases in UPI-induced LBW male offspring as they age.
Collapse
|
47
|
Eng ME, Bloise E, Matthews SG. Fetal glucocorticoid exposure leads to sex-specific changes in drug-transporter function at the blood-brain barrier in juvenile guinea pigs. FASEB J 2022; 36:e22245. [PMID: 35262963 PMCID: PMC9311705 DOI: 10.1096/fj.202101552rr] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 02/17/2022] [Accepted: 02/22/2022] [Indexed: 11/18/2022]
Abstract
Antenatal synthetic glucocorticoids (sGCs) are a life‐saving treatment in managing pre‐term birth. However, off‐target effects of sGCs can impact blood‐brain barrier (BBB) drug transporters essential for fetal brain protection, including P‐glycoprotein (P‐gp/Abcb1) and breast cancer resistance protein (BCRP/Abcg2). We hypothesized that maternal antenatal sGC treatment modifies BBB function in juvenile offspring in a sex‐dependent manner. Thus, the objective of this study was to determine the long‐term impact of a single or multiple courses of betamethasone on P‐gp/Abcb1 and BCRP/Abcg2 expression and function at the BBB. Pregnant guinea pigs (N = 42) received 3 courses (gestation days (GDs) 40, 50, and 60) or a single course (GD50) of betamethasone (1 mg/kg) or vehicle (saline). Cerebral microvessels and brain endothelial cells (BEC) were collected from the post‐natal day (PND) 14 offspring to measure protein, gene expression, and function of the drug transporters P‐gp/Abcb1 and BCRP/Abcg2. P‐gp protein expression was decreased (p < .05) in microvessels from male offspring that had been exposed to multiple courses and a single course of sGC, in utero. Multiple courses of sGC resulted in a significant decrease in P‐gp function in BECs from males (p < .05), but not females. There was a very strong trend for increased P‐gp function in males compared to females (p = .055). Reduced P‐gp expression and function at the BBB of young male offspring following multiple prenatal sGC exposures, is clinically relevant as many drugs administered postnatally are P‐gp substrates. These novel sex differences in drug transporter function may underlie potential sexual dimorphism in drug sensitivity and toxicity in the newborn and juvenile brain.
Collapse
Affiliation(s)
- Margaret Elizabeth Eng
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Enrrico Bloise
- Department of Morphology, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Stephen G Matthews
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.,Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada.,Department of Obstetrics and Gynecology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.,Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
48
|
Coker SJ, Smith-Díaz CC, Dyson RM, Vissers MCM, Berry MJ. The Epigenetic Role of Vitamin C in Neurodevelopment. Int J Mol Sci 2022; 23:ijms23031208. [PMID: 35163133 PMCID: PMC8836017 DOI: 10.3390/ijms23031208] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/16/2022] [Accepted: 01/17/2022] [Indexed: 02/07/2023] Open
Abstract
The maternal diet during pregnancy is a key determinant of offspring health. Early studies have linked poor maternal nutrition during gestation with a propensity for the development of chronic conditions in offspring. These conditions include cardiovascular disease, type 2 diabetes and even compromised mental health. While multiple factors may contribute to these outcomes, disturbed epigenetic programming during early development is one potential biological mechanism. The epigenome is programmed primarily in utero, and during this time, the developing fetus is highly susceptible to environmental factors such as nutritional insults. During neurodevelopment, epigenetic programming coordinates the formation of primitive central nervous system structures, neurogenesis, and neuroplasticity. Dysregulated epigenetic programming has been implicated in the aetiology of several neurodevelopmental disorders such as Tatton-Brown-Rahman syndrome. Accordingly, there is great interest in determining how maternal nutrient availability in pregnancy might affect the epigenetic status of offspring, and how such influences may present phenotypically. In recent years, a number of epigenetic enzymes that are active during embryonic development have been found to require vitamin C as a cofactor. These enzymes include the ten-eleven translocation methylcytosine dioxygenases (TETs) and the Jumonji C domain-containing histone lysine demethylases that catalyse the oxidative removal of methyl groups on cytosines and histone lysine residues, respectively. These enzymes are integral to epigenetic regulation and have fundamental roles in cellular differentiation, the maintenance of pluripotency and development. The dependence of these enzymes on vitamin C for optimal catalytic activity illustrates a potentially critical contribution of the nutrient during mammalian development. These insights also highlight a potential risk associated with vitamin C insufficiency during pregnancy. The link between vitamin C insufficiency and development is particularly apparent in the context of neurodevelopment and high vitamin C concentrations in the brain are indicative of important functional requirements in this organ. Accordingly, this review considers the evidence for the potential impact of maternal vitamin C status on neurodevelopmental epigenetics.
Collapse
Affiliation(s)
- Sharna J. Coker
- Perinatal & Developmental Physiology Group, Department of Paediatrics & Child Health, University of Otago, Wellington 6242, New Zealand; (S.J.C.); (R.M.D.)
| | - Carlos C. Smith-Díaz
- Centre for Free Radical Research, Department of Pathology and Biomedical Science, University of Otago, Christchurch 8140, New Zealand;
| | - Rebecca M. Dyson
- Perinatal & Developmental Physiology Group, Department of Paediatrics & Child Health, University of Otago, Wellington 6242, New Zealand; (S.J.C.); (R.M.D.)
| | - Margreet C. M. Vissers
- Centre for Free Radical Research, Department of Pathology and Biomedical Science, University of Otago, Christchurch 8140, New Zealand;
- Correspondence: (M.C.M.V.); (M.J.B.)
| | - Mary J. Berry
- Perinatal & Developmental Physiology Group, Department of Paediatrics & Child Health, University of Otago, Wellington 6242, New Zealand; (S.J.C.); (R.M.D.)
- Correspondence: (M.C.M.V.); (M.J.B.)
| |
Collapse
|
49
|
Sinclair AJ, Guo XF, Abedin L. Dietary Alpha-Linolenic Acid Supports High Retinal DHA Levels. Nutrients 2022; 14:nu14020301. [PMID: 35057481 PMCID: PMC8779487 DOI: 10.3390/nu14020301] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/06/2022] [Accepted: 01/10/2022] [Indexed: 02/01/2023] Open
Abstract
The retina requires docosahexaenoic acid (DHA) for optimal function. Alpha-linolenic acid (ALA) and DHA are dietary sources of retinal DHA. This research investigated optimizing retinal DHA using dietary ALA. Previous research identified 19% DHA in retinal phospholipids was associated with optimal retinal function in guinea pigs. Pregnant guinea pigs were fed dietary ALA from 2.8% to 17.3% of diet fatty acids, at a constant level of linoleic acid (LA) of 18% for the last one third of gestation and retinal DHA levels were assessed in 3-week-old offspring maintained on the same diets as their mothers. Retinal DHA increased in a linear fashion with the maximum on the diet with LA:ALA of 1:1. Feeding diets with LA:ALA of 1:1 during pregnancy and assessing retinal DHA in 3-week-old offspring was associated with optimized retinal DHA levels. We speculate that the current intakes of ALA in human diets, especially in relation to LA intakes, are inadequate to support high DHA levels in the retina.
Collapse
Affiliation(s)
- Andrew J. Sinclair
- Department of Nutrition, Dietetics and Food, School of Clinical Sciences, Monash University, Melbourne, VIC 3168, Australia
- Correspondence: ; Tel.: +61-(0)414-906-341
| | - Xiao-Fei Guo
- Institute of Nutrition & Health, College of Public Health, Qingdao University, Qingdao 266071, China;
| | - Lavinia Abedin
- Department of Food Science and Technology, School of Science, RMIT University, Melbourne, VIC 3001, Australia;
| |
Collapse
|
50
|
Quebedeaux TM, Song H, Giwa-Otusajo J, Thompson LP. Chronic Hypoxia Inhibits Respiratory Complex IV Activity and Disrupts Mitochondrial Dynamics in the Fetal Guinea Pig Forebrain. Reprod Sci 2022; 29:184-192. [PMID: 34750769 DOI: 10.1007/s43032-021-00779-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: 04/09/2021] [Accepted: 10/19/2021] [Indexed: 11/25/2022]
Abstract
Mitochondrial dysfunction is an underlying cause of childhood neurological disease secondary to the crucial role of mitochondria in proper neurodevelopment. We hypothesized that chronic intrauterine hypoxia (HPX) induces mitochondrial deficits by altering mitochondrial biogenesis and dynamics in the fetal brain. Pregnant guinea pigs were exposed to either normoxia (NMX, 21%O2) or HPX (10.5%O2) starting at 28-day (early onset, EO-HPX) or 50-day (late onset, LO-HPX) gestation until term (65 days). Near-term male and female fetuses were extracted from anesthetized sows, and mitochondria were isolated from excised fetal forebrains (n = 6/group). Expression of mitochondrial complex subunits I-V (CI-CV), fission (Drp-1), and fusion (Mfn-2) proteins was measured by Western blot. CI and CIV enzyme activities were measured by colorimetric assays. Chronic HPX reduced fetal body wts and increased (P < 0.05) brain/body wt ratios of both sexes. CV subunit levels were increased in EO-HPX males only and CII levels increased in LO-HPX females only compared to NMX. Both EO- and LO-HPX decreased CIV activity in both sexes but had no effect on CI activity. EO-HPX increased Drp1 and decreased Mfn2 levels in males, while LO-HPX had no effect on either protein levels. In females, both EO-HPX and LO-HPX increased Drp1 but had no effect on Mfn2 levels. Chronic HPX alters abundance and activity of select complex subunits and shifts mitochondrial dynamics toward fission in a sex-dependent manner in the fetal guinea pig brain. This may be an underlying mechanism of reduced respiratory efficiency leading to disrupted metabolism and increased vulnerability to a second neurological injury at the time of birth in HPX fetal brains.
Collapse
Affiliation(s)
- Tabitha M Quebedeaux
- Department of Obstetrics, Gynecology, & Reproductive Sciences, University of Maryland, Baltimore, School of Medicine, 655 W. Baltimore St., Baltimore, MD, 21201, USA
| | - Hong Song
- Department of Obstetrics, Gynecology, & Reproductive Sciences, University of Maryland, Baltimore, School of Medicine, 655 W. Baltimore St., Baltimore, MD, 21201, USA
| | - Jamiu Giwa-Otusajo
- Department of Obstetrics, Gynecology, & Reproductive Sciences, University of Maryland, Baltimore, School of Medicine, 655 W. Baltimore St., Baltimore, MD, 21201, USA
| | - Loren P Thompson
- Department of Obstetrics, Gynecology, & Reproductive Sciences, University of Maryland, Baltimore, School of Medicine, 655 W. Baltimore St., Baltimore, MD, 21201, USA.
| |
Collapse
|