1
|
Mobedi E, Dehghan Harati HR, Allahyari I, Gharagozlou F, Vojgani M, Baghbanani RH, Akbarinejad A, Akbarinejad V. Developmental programming of production and reproduction in dairy cows: V. Association of the main and interactive effects of maternal level of milk production and milk fat to protein ratio with offspring's birth weight, survival, and productive and reproductive performance from birth to the first lactation period. Theriogenology 2024; 228:17-29. [PMID: 39084064 DOI: 10.1016/j.theriogenology.2024.07.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 07/23/2024] [Accepted: 07/23/2024] [Indexed: 08/02/2024]
Abstract
Level of dam milk production (DMP) and dam milk fat to protein ratio (DFPR), as an indicator of metabolic status in dairy cows, have been identified to be associated with productive and reproductive performance of the offspring. Yet whether the interaction of DMP by DFPR can be associated with performance of the offspring have not been studied to our knowledge. Therefore, the present study was conducted to investigate the association of the main and interactive effects of DMP and DFPR with offspring's birth weight, survival, milk yield and fertility. To this end, data of birth weight, culling rate, milk yield and reproductive variables of offspring born to lactating dams (n = 14,582) and data associated with DMP and DFPR during 305-day lactation were retrieved. Afterwards, offspring were classified in three categories of DMP, including DMP1 (dams with <10.00 × 103 kg of 305-day milk production), DMP2 (dams with ≥10.00 × 103 kg and <14.00 × 103 kg of 305-day milk production), DMP3 (dams with ≥14.00 × 103 kg of 305-day milk production), and three categories of DFPR, including DFPR1 (offspring born to dams with <1.00 FPR), DFPR2 (offspring born to dams with ≥1.00 and < 1.40 FPR) and DFPR3 (offspring born to dams with ≥1.40 FPR). Statistical analysis revealed no association of the interaction effect of DMP by DFPR with investigated variables in the offspring (P > 0.05). However, the main effect of DMP was positively associated with milk yield, but negatively associated with survival, age at first insemination and conception during nulliparity, and transgenerational improvement in milk yield in the offspring (P < 0.05). Moreover, the main effect of DFPR was positively associated with birth weight, survival and first service conception rate during nulliparity, but negatively associated with metabolic status and reproductive performance during primiparity in the offspring (P < 0.05). In conclusion, the present study did not find any interaction effect of DMP by DFPR on productive and reproductive variables in the offspring. This finding implicates the association of DMP with milk production in the offspring was regardless of DFPR. Moreover, this finding implies the association of DFPR with postpartum metabolic status and reproductive performance in the offspring was regardless of DMP.
Collapse
Affiliation(s)
- Emadeddin Mobedi
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | | | - Iman Allahyari
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Faramarz Gharagozlou
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Mehdi Vojgani
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Reza Hemmati Baghbanani
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | | | - Vahid Akbarinejad
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.
| |
Collapse
|
2
|
Hashim L, Vari D, Bhat AM, Tsuda T. Adaptive Growth of the Ductus Arteriosus and Aortic Isthmus in Various Ductus-Dependent Complex Congenital Heart Diseases. Pediatr Cardiol 2024; 45:1588-1595. [PMID: 37477699 DOI: 10.1007/s00246-023-03236-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 07/06/2023] [Indexed: 07/22/2023]
Abstract
BACKGROUND The ductus arteriosus (DA) is critical in maintaining postnatal circulation in neonates with obstructed systemic circulation (OSC) and pulmonary circulation (OPC). We hypothesized that the size of the DA and aortic isthmus (AoI) undergoes adaptive growth in utero to counteract the hemodynamic challenges in these congenital heart diseases (CHD). METHODS Postnatal echocardiograms of neonates diagnosed prenatally with ductal-dependent CHD who were started on prostaglandins within 24 h of birth were reviewed. We assessed the cross-sectional area of the aortic valve opening, pulmonary valve opening, AoI, and DA by calculating (diameter)2/body surface area. Neonates were classified into OSC or OPC then subgrouped depending upon the patency of semilunar valves: OSC with and without aortic atresia (OSC-AA and OSC-nAA, respectively) and OPC with and without pulmonary atresia (OPC-PA and OPC-nPA, respectively). RESULTS Ninety-four cases were studied. The DA in OSC was significantly larger than OPC, and the DA in OSC-AA was significantly larger than OSC-nAA. The size of the AoI was significantly larger in OPC than OSC and larger in OSC-AA than OSC-nAA. Within the OSC-nAA group, there was no significant difference in the size of the DA, AoI, or pulmonary valve opening between those with retrograde flow (RF) at the AoI and without (nRF) except the aortic valve opening was significantly larger in nRF. All groups had comparable cross-sectional areas of systemic output. CONCLUSIONS Our findings suggest that DA and AoI show compensatory growth to maintain critical blood flow to vital organs against primary anatomical abnormalities in ductus-dependent CHD. (249 words).
Collapse
Affiliation(s)
- Liza Hashim
- Nemours Cardiac Center, Nemours Children's Health, 1600 Rockland Rd, Wilmington, DE, 19803, USA
- Department of Pediatrics, Division of Pediatric Cardiology, University of Virginia Children's Hospital, 1204 W. Main St, Charlottesville, VA, 22903, USA
| | - Daniel Vari
- Nemours Cardiac Center, Nemours Children's Health, 1600 Rockland Rd, Wilmington, DE, 19803, USA
- Cincinnati Children's Hospital Medical Center, The Heart Institute, 3333 Burnet Ave, Cincinnati, OH, 45229, USA
| | - Abdul M Bhat
- Nemours Cardiac Center, Nemours Children's Health, 1600 Rockland Rd, Wilmington, DE, 19803, USA
- Department of Pediatrics, Sidney Kimmel Medical College at Thomas Jefferson University, 1025 Walnut St. #100, Philadelphia, PA, 19107, USA
| | - Takeshi Tsuda
- Nemours Cardiac Center, Nemours Children's Health, 1600 Rockland Rd, Wilmington, DE, 19803, USA.
- Department of Pediatrics, Sidney Kimmel Medical College at Thomas Jefferson University, 1025 Walnut St. #100, Philadelphia, PA, 19107, USA.
| |
Collapse
|
3
|
Krause BJ, Paz AA, Garrud TAC, Peñaloza E, Vega-Tapia F, Ford SG, Niu Y, Giussani DA. Epigenetic regulation by hypoxia, N-acetylcysteine and hydrogen sulphide of the fetal vasculature in growth restricted offspring: A study in humans and chicken embryos. J Physiol 2024; 602:3833-3852. [PMID: 38985827 DOI: 10.1113/jp286266] [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: 01/12/2024] [Accepted: 06/12/2024] [Indexed: 07/12/2024] Open
Abstract
Fetal growth restriction (FGR) is a common outcome in human suboptimal gestation and is related to prenatal origins of cardiovascular dysfunction in offspring. Despite this, therapy of human translational potential has not been identified. Using human umbilical and placental vessels and the chicken embryo model, we combined cellular, molecular, and functional studies to determine whether N-acetylcysteine (NAC) and hydrogen sulphide (H2S) protect cardiovascular function in growth-restricted unborn offspring. In human umbilical and placental arteries from control or FGR pregnancy and in vessels from near-term chicken embryos incubated under normoxic or hypoxic conditions, we determined the expression of the H2S gene CTH (i.e. cystathionine γ-lyase) (via quantitative PCR), the production of H2S (enzymatic activity), the DNA methylation profile (pyrosequencing) and vasodilator reactivity (wire myography) in the presence and absence of NAC treatment. The data show that FGR and hypoxia increased CTH expression in the embryonic/fetal vasculature in both species. NAC treatment increased aortic CTH expression and H2S production and enhanced third-order femoral artery dilator responses to the H2S donor sodium hydrosulphide in chicken embryos. NAC treatment also restored impaired endothelial relaxation in human third-to-fourth order chorionic arteries from FGR pregnancies and in third-order femoral arteries from hypoxic chicken embryos. This NAC-induced protection against endothelial dysfunction in hypoxic chicken embryos was mediated via nitric oxide independent mechanisms. Both developmental hypoxia and NAC promoted vascular changes in CTH DNA and NOS3 methylation patterns in chicken embryos. Combined, therefore, the data support that the effects of NAC and H2S offer a powerful mechanism of human translational potential against fetal cardiovascular dysfunction in complicated pregnancy. KEY POINTS: Gestation complicated by chronic fetal hypoxia and fetal growth restriction (FGR) increases a prenatal origin of cardiovascular disease in offspring, increasing interest in antenatal therapy to prevent against a fetal origin of cardiovascular dysfunction. We investigated the effects between N-acetylcysteine (NAC) and hydrogen sulphide (H2S) in the vasculature in FGR human pregnancy and in chronically hypoxic chicken embryos. Combining cellular, molecular, epigenetic and functional studies, we show that the vascular expression and synthesis of H2S is enhanced in hypoxic and FGR unborn offspring in both species and this acts to protect their vasculature. Therefore, the NAC/H2S pathway offers a powerful therapeutic mechanism of human translational potential against fetal cardiovascular dysfunction in complicated pregnancy.
Collapse
Affiliation(s)
- Bernardo J Krause
- Instituto de Ciencias de la Salud, Universidad O'Higgins, Santiago, Chile
| | - Adolfo A Paz
- Instituto de Ciencias de la Salud, Universidad O'Higgins, Santiago, Chile
| | - Tessa A C Garrud
- Department of Physiology, Development & Neuroscience, University of Cambridge, Cambridge, UK
| | - Estefanía Peñaloza
- Instituto de Ciencias de la Salud, Universidad O'Higgins, Santiago, Chile
| | - Fabian Vega-Tapia
- Instituto de Ciencias de la Salud, Universidad O'Higgins, Santiago, Chile
| | - Sage G Ford
- Department of Physiology, Development & Neuroscience, University of Cambridge, Cambridge, UK
| | - Youguo Niu
- Department of Physiology, Development & Neuroscience, University of Cambridge, Cambridge, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
| | - Dino A Giussani
- Department of Physiology, Development & Neuroscience, University of Cambridge, Cambridge, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
- BHF Cardiovascular Centre for Research Excellence, University of Cambridge, Cambridge, UK
- Strategic Research Initiative in Reproduction, University of Cambridge, Cambridge, UK
| |
Collapse
|
4
|
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)
- Jewish Heritage Fund for Excellence
- 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)
- 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
- 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
- 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
|
5
|
Graton ME, Spaans F, He R, Chatterjee P, Kirschenman R, Quon A, Phillips TJ, Case CP, Davidge ST. Sex-specific differences in the mechanisms for enhanced thromboxane A 2-mediated vasoconstriction in adult offspring exposed to prenatal hypoxia. Biol Sex Differ 2024; 15:52. [PMID: 38898532 PMCID: PMC11188502 DOI: 10.1186/s13293-024-00627-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 06/14/2024] [Indexed: 06/21/2024] Open
Abstract
BACKGROUND Prenatal hypoxia, a common pregnancy complication, leads to impaired cardiovascular outcomes in the adult offspring. It results in impaired vasodilation in coronary and mesenteric arteries of the adult offspring, due to reduced nitric oxide (NO). Thromboxane A2 (TxA2) is a potent vasoconstrictor increased in cardiovascular diseases, but its role in the impact of prenatal hypoxia is unknown. To prevent the risk of cardiovascular disease by prenatal hypoxia, we have tested a maternal treatment using a nanoparticle-encapsulated mitochondrial antioxidant (nMitoQ). We hypothesized that prenatal hypoxia enhances vascular TxA2 responses in the adult offspring, due to decreased NO modulation, and that this might be prevented by maternal nMitoQ treatment. METHODS Pregnant Sprague-Dawley rats received a single intravenous injection (100 µL) of vehicle (saline) or nMitoQ (125 µmol/L) on gestational day (GD)15 and were exposed to normoxia (21% O2) or hypoxia (11% O2) from GD15 to GD21 (term = 22 days). Coronary and mesenteric arteries were isolated from the 4-month-old female and male offspring, and vasoconstriction responses to U46619 (TxA2 analog) were evaluated using wire myography. In mesenteric arteries, L-NAME (pan-NO synthase (NOS) inhibitor) was used to assess NO modulation. Mesenteric artery endothelial (e)NOS, and TxA2 receptor expression, superoxide, and 3-nitrotyrosine levels were assessed by immunofluorescence. RESULTS Prenatal hypoxia resulted in increased U46619 responsiveness in coronary and mesenteric arteries of the female offspring, and to a lesser extent in the male offspring, which was prevented by nMitoQ. In females, there was a reduced impact of L-NAME in mesenteric arteries of the prenatal hypoxia saline-treated females, and reduced 3-nitrotyrosine levels. In males, L-NAME increased U46619 responses in mesenteric artery to a similar extent, but TxA2 receptor expression was increased by prenatal hypoxia. There were no changes in eNOS or superoxide levels. CONCLUSIONS Prenatal hypoxia increased TxA2 vasoconstrictor capacity in the adult offspring in a sex-specific manner, via reduced NO modulation in females and increased TP expression in males. Maternal placental antioxidant treatment prevented the impact of prenatal hypoxia. These findings increase our understanding of how complicated pregnancies can lead to a sex difference in the programming of cardiovascular disease in the adult offspring.
Collapse
Affiliation(s)
- Murilo E Graton
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, AB, T6G 2R3, Canada
- Women and Children's Health Research Institute, University of Alberta, Edmonton, AB, T6G 2R3, Canada
| | - Floor Spaans
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, AB, T6G 2R3, Canada
- Women and Children's Health Research Institute, University of Alberta, Edmonton, AB, T6G 2R3, Canada
| | - Rose He
- Women and Children's Health Research Institute, University of Alberta, Edmonton, AB, T6G 2R3, Canada
- Department of Physiology, University of Alberta, Edmonton, AB, T6G 2R3, Canada
| | - Paulami Chatterjee
- Women and Children's Health Research Institute, University of Alberta, Edmonton, AB, T6G 2R3, Canada
- Department of Physiology, University of Alberta, Edmonton, AB, T6G 2R3, Canada
| | - Raven Kirschenman
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, AB, T6G 2R3, Canada
- Women and Children's Health Research Institute, University of Alberta, Edmonton, AB, T6G 2R3, Canada
| | - Anita Quon
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, AB, T6G 2R3, Canada
- Women and Children's Health Research Institute, University of Alberta, Edmonton, AB, T6G 2R3, Canada
| | - Tom J Phillips
- UK Dementia Research Institute, Cardiff University, Cardiff, W1T 7NF, UK
| | - C Patrick Case
- Musculoskeletal Research Unit, University of Bristol, Bristol, BS8 1QU, UK
| | - Sandra T Davidge
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, AB, T6G 2R3, Canada.
- Women and Children's Health Research Institute, University of Alberta, Edmonton, AB, T6G 2R3, Canada.
| |
Collapse
|
6
|
Ghasemi Z, Alizadeh Mogadam Masouleh A, Rashki Ghaleno L, Akbarinejad V, Rezazadeh Valojerdi M, Shahverdi A. Maternal nutrition and fetal imprinting of the male progeny. Anim Reprod Sci 2024; 265:107470. [PMID: 38657462 DOI: 10.1016/j.anireprosci.2024.107470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 03/28/2024] [Accepted: 03/30/2024] [Indexed: 04/26/2024]
Abstract
The global population as well as the demand for human food is rapidly growing worldwide, which necessitates improvement of efficiency in livestock operations. In this context, environmental factors during fetal and/or neonatal life have been observed to influence normal physical and physiological function of an individual during adulthood, and this phenomenon is called fetal or developmental programming. While numerous studies have reported the impact of maternal factors on development of the female progeny, limited information is available on the potential effects of fetal programming on reproductive function of the male offspring. Therefore, the objective for this review article was to focus on available literature regarding the impact of maternal factors, particularly maternal nutrition, on reproductive system of the male offspring. To this end, we highlighted developmental programming of the male offspring in domestic species (i.e., pig, cow and sheep) as well as laboratory species (i.e., mice and rat) during pregnancy and lactation. In this sense, we pointed out the effects of maternal nutrition on various functions of the male offspring including hypothalamic-pituitary axis, hormonal levels, testicular tissue and semen parameters.
Collapse
Affiliation(s)
- Zahrasadat Ghasemi
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran; Animal Core Facility, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - AliReza Alizadeh Mogadam Masouleh
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran; Gyn-medicum, Center for Reproductive Medicine, Göttingen, Germany; Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Göttingen, Germany.
| | - Leila Rashki Ghaleno
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Vahid Akbarinejad
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Mojtaba Rezazadeh Valojerdi
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran; Department of Anatomy, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Abdolhossein Shahverdi
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| |
Collapse
|
7
|
Mobedi E, Harati HRD, Allahyari I, Gharagozlou F, Vojgani M, Baghbanani RH, Akbarinejad A, Akbarinejad V. Developmental programming of production and reproduction in dairy cows: IV. Association of maternal milk fat and protein percentage and milk fat to protein ratio with offspring's birth weight, survival, productive and reproductive performance and AMH concentration from birth to the first lactation period. Theriogenology 2024; 220:12-25. [PMID: 38457855 DOI: 10.1016/j.theriogenology.2024.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 02/22/2024] [Accepted: 03/01/2024] [Indexed: 03/10/2024]
Abstract
Although the association of maternal milk production with developmental programming of offspring has been investigated, there is limited information available on the relationship of maternal milk components with productive and reproductive performance of the offspring. Therefore, the present study was conducted to analyze the association of maternal milk fat and protein percentage and milk fat to protein ratio with birth weight, survival, productive and reproductive performance and AMH concentration in the offspring. In study I, data of birth weight, milk yield and reproductive variables of offspring born to lactating dams (n = 14,582) and data associated with average maternal milk fat percentage (MFP), protein percentage (MPP) and fat to protein ratio (MFPR) during 305-day lactation were retrieved. Afterwards, offspring were classified in various categories of MFP, MPP and MFPR. In study II, blood samples (n = 339) were collected from offspring in various categories of MFP, MPP and MFPR for measurement of serum AMH. Maternal milk fat percentage was positively associated with birth weight and average percentage of milk fat (APMF) and protein (APMP) and milk fat to protein ratio (FPR) during the first lactation, but negatively associated with culling rate during nulliparity in the offspring (P < 0.05). Maternal milk protein percentage was positively associated with birth weight, APMF, APMP, FPR and culling rate, but negatively associated with milk yield and fertility in the offspring (P < 0.05). Maternal FPR was positively associated with APMF and FPR, but negatively associated with culling rate, APMP and fertility in the offspring (P < 0.05). However, concentration of AMH in the offspring was not associated with MFP, MPP and MFPR (P > 0.05). In conclusion, the present study revealed that maternal milk fat and protein percentage and their ratio were associated with birth weight, survival, production and reproduction of the offspring. Yet it was a preliminary research and further studies are required to elucidate the mechanisms underlying these associations.
Collapse
Affiliation(s)
- Emadeddin Mobedi
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | | | - Iman Allahyari
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Faramarz Gharagozlou
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Mehdi Vojgani
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Reza Hemmati Baghbanani
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | | | - Vahid Akbarinejad
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.
| |
Collapse
|
8
|
Akbarinejad V, Cushman RA. Developmental programming of reproduction in the female animal. Anim Reprod Sci 2024; 263:107456. [PMID: 38503204 DOI: 10.1016/j.anireprosci.2024.107456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 03/09/2024] [Accepted: 03/12/2024] [Indexed: 03/21/2024]
Abstract
Successful reproduction is a cornerstone in food animal industry in order to sustain food production for human. Therefore, various methods focusing on genetics and postnatal environment have been identified and applied to improve fertility in livestock. Yet there is evidence indicating that environmental factors during prenatal and/or neonatal life can also impact the function of reproductive system and fertility in the animals during adulthood, which is called the developmental programming of reproduction. The current review summarizes data associated with the developmental origins of reproduction in the female animals. In this regard, this review focuses on the effect of plane of nutrition, maternal body condition, hypoxia, litter size, maternal age, parity, level of milk production and milk components, lactocrine signaling, stress, thermal stress, exposure to androgens, endocrine disrupting chemicals, mycotoxins and pollutants, affliction with infection and inflammation, and maternal gut microbiota during prenatal and neonatal periods on the neuroendocrine system, puberty, health of reproductive organs and fertility in the female offspring. It is noteworthy that these prenatal and neonatal factors do not always exert their effects on the reproductive performance of the female by compromising the development of organs directly related to reproductive function such as hypothalamus, pituitary, ovary, oviduct and uterus. Since they can impair the development of non-reproductive organs and systems modulating reproductive function as well (e.g., metabolic system and level of milk yield in dairy animals). Furthermore, when these factors affect the epigenetics of the offspring, their adverse effects will not be limited to one generation and can transfer transgenerationally. Hence, pinpointing the factors influencing developmental programming of reproduction and considering them in management of livestock operations could be a potential strategy to help improve fertility in food animals.
Collapse
Affiliation(s)
- Vahid Akbarinejad
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.
| | - Robert A Cushman
- USDA, Agricultural Research Service, US. Meat Animal Research Center, Clay Center, NE 68933-0166, United States
| |
Collapse
|
9
|
Schroeder M, Fuenzalida B, Yi N, Shahnawaz S, Gertsch J, Pellegata D, Ontsouka E, Leiva A, Gutiérrez J, Müller M, Brocco MA, Albrecht C. LAT1-dependent placental methionine uptake is a key player in fetal programming of metabolic disease. Metabolism 2024; 153:155793. [PMID: 38295946 DOI: 10.1016/j.metabol.2024.155793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 12/11/2023] [Accepted: 01/09/2024] [Indexed: 02/06/2024]
Abstract
The Developmental Origins of Health and Disease hypothesis sustains that exposure to different stressors during prenatal development prepares the offspring for the challenges to be encountered after birth. We studied the gestational period as a particularly vulnerable window where different stressors can have strong implications for fetal programming of the offspring's life-long metabolic status via alterations of specific placentally expressed nutrient transporters. To study this mechanism, we used a murine prenatal stress model, human preeclampsia, early miscarriage, and healthy placental tissue samples, in addition to in vitro models of placental cells. In stressed mice, placental overexpression of L-type amino acid transporter 1 (Lat1) and subsequent global placental DNA hypermethylation was accompanied by fetal and adult hypothalamic dysregulation in global DNA methylation and gene expression as well as long-term metabolic abnormalities exclusively in female offspring. In human preeclampsia, early miscarriage, and under hypoxic conditions, placental LAT1 was significantly upregulated, leading to increased methionine uptake and global DNA hypermethylation. Remarkably, subgroups of healthy term placentas with high expression of stress-related genes presented increased levels of placental LAT1 mRNA and protein, DNA and RNA hypermethylation, increased methionine uptake capacity, one-carbon metabolic pathway disruption, higher methionine concentration in the placenta and transport to the fetus specifically in females. Since LAT1 mediates the intracellular accumulation of methionine, global DNA methylation, and one-carbon metabolism in the placenta, our findings hint at a major sex-specific global response to a variety of prenatal stressors affecting placental function, epigenetic programming, and life-long metabolic disease and provide a much-needed insight into early-life factors predisposing females/women to metabolic disorders.
Collapse
Affiliation(s)
- Mariana Schroeder
- Faculty of Medicine, Institute of Biochemistry and Molecular Medicine, University of Bern, Switzerland; Swiss National Centre of Competence in Research, NCCR TransCure, University of Bern, Bern, Switzerland.
| | - Barbara Fuenzalida
- Faculty of Medicine, Institute of Biochemistry and Molecular Medicine, University of Bern, Switzerland
| | - Nan Yi
- Faculty of Medicine, Institute of Biochemistry and Molecular Medicine, University of Bern, Switzerland
| | - Saira Shahnawaz
- Department of Biochemistry, Sargodha Medical College, University of Sargodha, Sargodha, Pakistan; Department of Allied Health Sciences, Sargodha Medical College, University of Sargodha, Sargodha, Pakistan
| | - Jürg Gertsch
- Faculty of Medicine, Institute of Biochemistry and Molecular Medicine, University of Bern, Switzerland; Swiss National Centre of Competence in Research, NCCR TransCure, University of Bern, Bern, Switzerland
| | - Daniele Pellegata
- Faculty of Medicine, Institute of Biochemistry and Molecular Medicine, University of Bern, Switzerland
| | - Edgar Ontsouka
- Faculty of Medicine, Institute of Biochemistry and Molecular Medicine, University of Bern, Switzerland
| | - Andrea Leiva
- Faculty of Medicine and Science, Universidad of San Sebastian, Santiago, Chile
| | - Jaime Gutiérrez
- Faculty of Medicine and Science, Universidad of San Sebastian, Santiago, Chile
| | - Martin Müller
- Division of Gynecology and Obstetrics, Lindenhofgruppe, Bern, Switzerland
| | - Marcela A Brocco
- Institute of Biotechnological Research, University of San Martín, Buenos Aires, Argentina
| | - Christiane Albrecht
- Faculty of Medicine, Institute of Biochemistry and Molecular Medicine, University of Bern, Switzerland; Swiss National Centre of Competence in Research, NCCR TransCure, University of Bern, Bern, Switzerland
| |
Collapse
|
10
|
Wang B, Zhu Y, Wei B, Zeng H, Zhang P, Li L, Wang H, Wu X, Zheng Y, Sun M. miR-377-3p Regulates Hippocampal Neurogenesis via the Zfp462-Pbx1 Pathway and Mediates Anxiety-Like Behaviors in Prenatal Hypoxic Offspring. Mol Neurobiol 2024; 61:1920-1935. [PMID: 37817032 DOI: 10.1007/s12035-023-03683-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 09/29/2023] [Indexed: 10/12/2023]
Abstract
Prenatal hypoxia (PH) is one of the most common complications of obstetrics and is closely associated with many neurological disorders such as depression, anxiety, and cognitive impairment. Our previous study found that Zfp462 heterozygous (Het) mice exhibit significant anxiety-like behavior. Interestingly, offspring mice with PH also have anxiety-like behaviors in adulthood, accompanied by reduced expression of Zfp462 and increased expression of miR-377-3p; however, the exact regulatory mechanisms remain unclear. In this study, western blotting, gene knockdown, immunofluorescence, dual-luciferase reporter assay, immunoprecipitation, cell transfection with miR-377-3p mimics or inhibitors, quantitative real-time PCR, and rescue assay were used to detect changes in the miR-377-3p-Zfp462-Pbx1 (pre-B-cell leukemia homeobox1) pathway in the brains of prenatal hypoxic offspring to explain the pathogenesis of anxiety-like behaviors. We found that Zfp462 deficiency promoted Pbx1 protein degradation through ubiquitination and that Zfp462 Het mice showed downregulation of the protein kinase B (PKB, also called Akt)-glycogen synthase kinase-3β (GSK3β)-cAMP response element-binding protein (CREB) pathway and hippocampal neurogenesis with anxiety-like behavior. In addition, PH mice exhibited upregulation of miR-377-3p, downregulation of Zfp462/Pbx1-Akt-GSK3β-CREB pathway activity, reduced hippocampal neurogenesis, and an anxiety-like phenotype. Intriguingly, miR-377-3p directly targets the 3'UTR of Zfp462 mRNA to regulate Zfp462 expression. Importantly, microinjection of miR-377-3p antagomir into the hippocampal dentate gyrus of PH mice upregulated Zfp462/Pbx1-Akt-GSK3β-CREB pathway activity, increased hippocampal neurogenesis, and improved anxiety-like behaviors. Collectively, our findings demonstrated a crucial role for miR-377-3p in the regulation of hippocampal neurogenesis and anxiety-like behaviors via the Zfp462/Pbx1-Akt-GSK3β-CREB pathway. Therefore, miR-377-3p could be a potential therapeutic target for anxiety-like behavior in prenatal hypoxic offspring.
Collapse
Affiliation(s)
- Bin Wang
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou City, 215006, Jiangsu, China.
| | - Yichen Zhu
- Cambridge-Suda Genomic Resource Center, Jiangsu Key Laboratory of Neuropsychiatric Diseases Research, Suzhou Medical College of Soochow University, Jiangsu, 215123, China
| | - Bin Wei
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou City, 215006, Jiangsu, China
| | - Hongtao Zeng
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou City, 215006, Jiangsu, China
| | - Pengjie Zhang
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou City, 215006, Jiangsu, China
| | - Lingjun Li
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou City, 215006, Jiangsu, China
| | - Hongyan Wang
- Obstetrics and Gynecology Hospital Research Center, Institute of Reproduction and Development, Fudan University, Shanghai, 200433, China
- State Key Laboratory of Genetic Engineering, MOE Key Laboratory of Contemporary Anthropology, and Collaborative Innovation Center for Genetics & Development, School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Xiaohui Wu
- State Key Laboratory of Genetic Engineering, MOE Key Laboratory of Contemporary Anthropology, and Collaborative Innovation Center for Genetics & Development, School of Life Sciences, Fudan University, Shanghai, 200438, China
- Institute of Developmental Biology & Molecular Medicine, Fudan University, Shanghai, 200433, China
| | - Yufang Zheng
- Obstetrics and Gynecology Hospital Research Center, Institute of Reproduction and Development, Fudan University, Shanghai, 200433, China
- State Key Laboratory of Genetic Engineering, MOE Key Laboratory of Contemporary Anthropology, and Collaborative Innovation Center for Genetics & Development, School of Life Sciences, Fudan University, Shanghai, 200438, China
- Institute of Developmental Biology & Molecular Medicine, Fudan University, Shanghai, 200433, China
| | - Miao Sun
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou City, 215006, Jiangsu, China.
| |
Collapse
|
11
|
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
|
12
|
Kolesova H, Hrabalova P, Bohuslavova R, Abaffy P, Fabriciova V, Sedmera D, Pavlinkova G. Reprogramming of the developing heart by Hif1a-deficient sympathetic system and maternal diabetes exposure. Front Endocrinol (Lausanne) 2024; 15:1344074. [PMID: 38505753 PMCID: PMC10948485 DOI: 10.3389/fendo.2024.1344074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 02/14/2024] [Indexed: 03/21/2024] Open
Abstract
Introduction Maternal diabetes is a recognized risk factor for both short-term and long-term complications in offspring. Beyond the direct teratogenicity of maternal diabetes, the intrauterine environment can influence the offspring's cardiovascular health. Abnormalities in the cardiac sympathetic system are implicated in conditions such as sudden infant death syndrome, cardiac arrhythmic death, heart failure, and certain congenital heart defects in children from diabetic pregnancies. However, the mechanisms by which maternal diabetes affects the development of the cardiac sympathetic system and, consequently, heightens health risks and predisposes to cardiovascular disease remain poorly understood. Methods and results In the mouse model, we performed a comprehensive analysis of the combined impact of a Hif1a-deficient sympathetic system and the maternal diabetes environment on both heart development and the formation of the cardiac sympathetic system. The synergic negative effect of exposure to maternal diabetes and Hif1a deficiency resulted in the most pronounced deficit in cardiac sympathetic innervation and the development of the adrenal medulla. Abnormalities in the cardiac sympathetic system were accompanied by a smaller heart, reduced ventricular wall thickness, and dilated subepicardial veins and coronary arteries in the myocardium, along with anomalies in the branching and connections of the main coronary arteries. Transcriptional profiling by RNA sequencing (RNA-seq) revealed significant transcriptome changes in Hif1a-deficient sympathetic neurons, primarily associated with cell cycle regulation, proliferation, and mitosis, explaining the shrinkage of the sympathetic neuron population. Discussion Our data demonstrate that a failure to adequately activate the HIF-1α regulatory pathway, particularly in the context of maternal diabetes, may contribute to abnormalities in the cardiac sympathetic system. In conclusion, our findings indicate that the interplay between deficiencies in the cardiac sympathetic system and subtle structural alternations in the vasculature, microvasculature, and myocardium during heart development not only increases the risk of cardiovascular disease but also diminishes the adaptability to the stress associated with the transition to extrauterine life, thus increasing the risk of neonatal death.
Collapse
Affiliation(s)
- Hana Kolesova
- Institute of Anatomy, First Faculty of Medicine, Charles University, Prague, Czechia
- Department of Developmental Cardiology, Institute of Physiology Czech Academy of Sciences (CAS), Prague, Czechia
| | - Petra Hrabalova
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology Czech Academy of Sciences (CAS), BIOCEV, Vestec, Czechia
- Faculty of Science, Charles University, Prague, Czechia
| | - Romana Bohuslavova
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology Czech Academy of Sciences (CAS), BIOCEV, Vestec, Czechia
| | - Pavel Abaffy
- Laboratory of Gene Expression, Institute of Biotechnology Czech Academy of Sciences (CAS), BIOCEV, Vestec, Czechia
| | - Valeria Fabriciova
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology Czech Academy of Sciences (CAS), BIOCEV, Vestec, Czechia
| | - David Sedmera
- Institute of Anatomy, First Faculty of Medicine, Charles University, Prague, Czechia
- Department of Developmental Cardiology, Institute of Physiology Czech Academy of Sciences (CAS), Prague, Czechia
| | - Gabriela Pavlinkova
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology Czech Academy of Sciences (CAS), BIOCEV, Vestec, Czechia
| |
Collapse
|
13
|
Dehghan Harati HR, Mobedi E, Allahyari I, Gharagozlou F, Vojgani M, Hemmati Baghbanani R, Akbarinejad A, Akbarinejad V. Developmental programming of production and reproduction in dairy cows: III. Association of level of maternal milk production with offspring's birth weight, survival, productive and reproductive performance and AMH concentration from birth to the first lactation period. Theriogenology 2024; 216:155-167. [PMID: 38183932 DOI: 10.1016/j.theriogenology.2024.01.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 12/31/2023] [Accepted: 01/02/2024] [Indexed: 01/08/2024]
Abstract
Although some studies investigated the relationship of dam milk production (DMP) with offspring birth weight and productive performance, limited information is available on the association of level of DMP with reproductive performance in dairy cows. Therefore, the present study was conducted to understand whether dams with various levels of milk production produce offspring with different fertility. In study I, offspring were classified based on the level of DMP into five categories, including DMP1 (dams with <10.00 × 103 kg of 305-day milk production), DMP2 (dams with ≥10.00 × 103 kg and <12.00 × 103 kg of 305-day milk production), DMP3 (dams with ≥12.00 × 103 kg and <14.00 × 103 kg of 305-day milk production), DMP4 (dams with ≥14.00 × 103 kg and <16.00 × 103 kg of 305-day milk production) and DMP5 (dams with ≥16.00 × 103 kg of 305-day milk production). In study I, data of birth weight, milk yield and reproductive variables of 14,536 offspring born to lactating dams and corresponding data of DMP were retrieved. In study II, blood samples (n = 339) were collected from offspring in various categories of DMP for measurement of serum AMH. Offspring were heavier at birth in DMP4 and DMP5 categories than DMP1 and DMP2 categories (P < 0.05). Milk yield of offspring increased as DMP elevated (P < 0.05); however, offspring in DMP1 and DMP2 categories produced higher milk as compared with their dams during primiparity (P < 0.05) whereas offspring in DMP3, DMP4 and DMP5 categories produced less milk as compared with their dams during primiparity (P < 0.05). Milk fat to protein ratio during the first month of lactation was greater in DMP4 and DMP5 categories than DMP1 category (P < 0.05). Offspring of DMP4 and DMP5 categories were inseminated and conceived at younger ages than offspring of DMP1 category during nulliparity (P < 0.05). Calving to conception interval was longer in DMP5 than DMP1 category in primiparous offspring (P < 0.05), but concentration of AMH did not differ among various categories of DMP (P > 0.05). In conclusion, dams with greater level of milk production produced heavier offspring with higher milk yield but worse transgenerational improvement in milk production and diminished reproductive performance, which were seemingly under higher pressure of negative energy balance during the first month of lactation.
Collapse
Affiliation(s)
| | - Emadeddin Mobedi
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Iman Allahyari
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Faramarz Gharagozlou
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Mehdi Vojgani
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Reza Hemmati Baghbanani
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | | | - Vahid Akbarinejad
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.
| |
Collapse
|
14
|
Wang Z, Camm EJ, Nuzzo AM, Spiroski AM, Skeffington KL, Ashmore TJ, Rolfo A, Todros T, Logan A, Ma J, Murphy MP, Niu Y, Giussani DA. In vivo mitochondria-targeted protection against uterine artery vascular dysfunction and remodelling in rodent hypoxic pregnancy. J Physiol 2024; 602:1211-1225. [PMID: 38381050 DOI: 10.1113/jp286178] [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: 12/20/2023] [Accepted: 01/29/2024] [Indexed: 02/22/2024] Open
Abstract
Gestational hypoxia adversely affects uterine artery function, increasing complications. However, an effective therapy remains unidentified. Here, we show in rodent uterine arteries that hypoxic pregnancy promotes hypertrophic remodelling, increases constrictor reactivity via protein kinase C signalling, and triggers compensatory dilatation via nitric oxide-dependent mechanisms and stimulation of large conductance Ca2+ -activated K+ -channels. Maternal in vivo oral treatment with the mitochondria-targeted antioxidant MitoQ in hypoxic pregnancy normalises uterine artery reactivity and prevents vascular remodelling. From days 6-20 of gestation (term ∼22 days), female Wistar rats were randomly assigned to normoxic or hypoxic (13-14% O2 ) pregnancy ± daily maternal MitoQ treatment (500 µm in drinking water). At 20 days of gestation, maternal, placental and fetal tissue was frozen to determine MitoQ uptake. The uterine arteries were harvested and, in one segment, constrictor and dilator reactivity was determined by wire myography. Another segment was fixed for unbiased stereological analysis of vessel morphology. Maternal administration of MitoQ in both normoxic and hypoxic pregnancy crossed the placenta and was present in all tissues analysed. Hypoxia increased uterine artery constrictor responses to norepinephrine, angiotensin II and the protein kinase C activator, phorbol 12,13-dibutyrate. Hypoxia enhanced dilator reactivity to sodium nitroprusside, the large conductance Ca2+ -activated K+ -channel activator NS1619 and ACh via increased nitric oxide-dependent mechanisms. Uterine arteries from hypoxic pregnancy showed increased wall thickness and MitoQ treatment in hypoxic pregnancy prevented all effects on uterine artery reactivity and remodelling. The data support mitochondria-targeted therapy against adverse changes in uterine artery structure and function in high-risk pregnancy. KEY POINTS: Dysfunction and remodelling of the uterine artery are strongly implicated in many pregnancy complications, including advanced maternal age, maternal hypertension of pregnancy, maternal obesity, gestational diabetes and pregnancy at high altitude. Such complications not only have immediate adverse effects on the growth of the fetus, but also they can also increase the risk of cardiovascular disease in the mother and offspring. Despite this, there is a significant unmet clinical need for therapeutics that treat uterine artery vascular dysfunction in adverse pregnancy. Here, we show in a rodent model of gestational hypoxia that in vivo oral treatment of the mitochondria-targeted antioxidant MitoQ protects against uterine artery vascular dysfunction and remodelling, supporting the use of mitochondria-targeted therapy against adverse changes in uterine artery structure and function in high-risk pregnancy.
Collapse
Affiliation(s)
- Zhongchao Wang
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Department of Aerospace Physiology, Fourth Military Medical University, Xi'an, China
- Department of Congenital Heart Disease, General Hospital of Northern Theater Command, Shenyang, China
| | - Emily J Camm
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Anna Maria Nuzzo
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Department of Surgical Sciences, University of Turin, Turin, Italy
| | - Ana-Mishel Spiroski
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Cambridge Cardiovascular Strategic Research Initiative, University of Cambridge, Cambridge, UK
| | - Katie L Skeffington
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Thomas J Ashmore
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Alessandro Rolfo
- Department of Surgical Sciences, University of Turin, Turin, Italy
| | - Tullia Todros
- Department of Surgical Sciences, University of Turin, Turin, Italy
| | - Angela Logan
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK
| | - Jin Ma
- Department of Aerospace Physiology, Fourth Military Medical University, Xi'an, China
| | - Michael P Murphy
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK
| | - Youguo Niu
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Cambridge Cardiovascular Strategic Research Initiative, University of Cambridge, Cambridge, UK
| | - Dino A Giussani
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Cambridge Cardiovascular Strategic Research Initiative, University of Cambridge, Cambridge, UK
| |
Collapse
|
15
|
Fuenzalida B, Yañez MJ, Mueller M, Mistry HD, Leiva A, Albrecht C. Evidence for hypoxia-induced dysregulated cholesterol homeostasis in preeclampsia: Insights into the mechanisms from human placental cells and tissues. FASEB J 2024; 38:e23431. [PMID: 38265294 PMCID: PMC10953329 DOI: 10.1096/fj.202301708rr] [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: 08/24/2023] [Revised: 12/21/2023] [Accepted: 01/04/2024] [Indexed: 01/25/2024]
Abstract
Preeclampsia (PE) poses a considerable risk to the long-term cardiovascular health of both mothers and their offspring due to a hypoxic environment in the placenta leading to reduced fetal oxygen supply. Cholesterol is vital for fetal development by influencing placental function. Recent findings suggest an association between hypoxia, disturbed cholesterol homeostasis, and PE. This study investigates the influence of hypoxia on placental cholesterol homeostasis. Using primary human trophoblast cells and placentae from women with PE, various aspects of cholesterol homeostasis were examined under hypoxic and hypoxia/reoxygenation (H/R) conditions. Under hypoxia and H/R, intracellular total and non-esterified cholesterol levels were significantly increased. This coincided with an upregulation of HMG-CoA-reductase and HMG-CoA-synthase (key genes regulating cholesterol biosynthesis), and a decrease in acetyl-CoA-acetyltransferase-1 (ACAT1), which mediates cholesterol esterification. Hypoxia and H/R also increased the intracellular levels of reactive oxygen species and elevated the expression of hypoxia-inducible factor (HIF)-2α and sterol-regulatory-element-binding-protein (SREBP) transcription factors. Additionally, exposure of trophoblasts to hypoxia and H/R resulted in enhanced cholesterol efflux to maternal and fetal serum. This was accompanied by an increased expression of proteins involved in cholesterol transport such as the scavenger receptor class B type I (SR-BI) and the ATP-binding cassette transporter G1 (ABCG1). Despite these metabolic alterations, mitogen-activated-protein-kinase (MAPK) signaling, a key regulator of cholesterol homeostasis, was largely unaffected. Our findings indicate dysregulation of cholesterol homeostasis at multiple metabolic points in both the trophoblast hypoxia model and placentae from women with PE. The increased cholesterol efflux and intracellular accumulation of non-esterified cholesterol may have critical implications for both the mother and the fetus during pregnancy, potentially contributing to an elevated cardiovascular risk later in life.
Collapse
Affiliation(s)
- Barbara Fuenzalida
- Institute of Biochemistry and Molecular Medicine, Faculty of MedicineUniversity of BernBernSwitzerland
| | - Maria Jose Yañez
- School of Medical Technology, Faculty of Medicine and ScienceUniversidad San SebastiánSantiagoChile
| | - Martin Mueller
- Division of Gynecology and ObstetricsLindenhofgruppeBernSwitzerland
- Department for BioMedical ResearchUniversity of BernBernSwitzerland
| | - Hiten D. Mistry
- Department of Women and Children's HealthSchool of Life Course and Population Health Sciences, King's College LondonLondonUK
| | - Andrea Leiva
- School of Medical Technology, Faculty of Medicine and ScienceUniversidad San SebastiánSantiagoChile
| | - Christiane Albrecht
- Institute of Biochemistry and Molecular Medicine, Faculty of MedicineUniversity of BernBernSwitzerland
- Swiss National Center of Competence in Research, NCCR TransCureUniversity of BernBernSwitzerland
| |
Collapse
|
16
|
Mozaffari Makiabadi MJ, Bafandeh M, Gharagozlou F, Vojgani M, Mobedi E, Akbarinejad V. Developmental programming of production and reproduction in dairy cows: II. Association of gestational stage of maternal exposure to heat stress with offspring's birth weight, milk yield, reproductive performance and AMH concentration during the first lactation period. Theriogenology 2023; 212:41-49. [PMID: 37690376 DOI: 10.1016/j.theriogenology.2023.09.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 08/11/2023] [Accepted: 09/01/2023] [Indexed: 09/12/2023]
Abstract
Although the negative effect of maternal exposure to heat stress on production and reproduction of offspring has been reported, there are some discrepancies among various studies about which gestational stage is more critical in this regard. Therefore, the present research was conducted to identify during which stage(s) of pregnancy maternal exposure to heat stress would lead to more dramatic decrease in productive and reproductive performance of offspring. To this end, offspring were classified based on the gestational stage they were in utero exposed to heat stress into four categories, including heat stress exposure (HSE) during only the first trimester of gestation (HSE1), HSE during the first and second trimester of gestation (HSE2), HSE during the second and third trimester of gestation (HSE3) and HSE during only the third trimester of gestation (HSE4). In study I, data of birth weight, milk yield and reproductive variables of 11,788 offspring and data of the month they were conceived were retrieved. In study II, blood samples (n = 521) were collected from offspring in various categories of HSE for measurement of serum AMH. Offspring in HSE1 and HSE2 categories were heavier than offspring in HSE3 and HSE4 categories (P < 0.0001). Offspring in HSE1 and HSE3 categories had the lowest and highest milk production, respectively (P < 0.05). First service conception rate was the greatest and worst in HSE1 and HSE4 categories, respectively (P < 0.05). Service per conception and calving to conception interval were greater in HSE2 than HSE4 category (P < 0.05). Concentration of AMH was lower in HSE1 than HSE4 category (P < 0.05). In conclusion, the present study indicated that the early stage of gestation could be a more critical period for the negative impact of in utero heat stress on developmental programming of milk production and ovarian reserves. Yet an evident temporal pattern for the adverse effect of maternal heat stress on developmental programming of reproductive performance in offspring was not found.
Collapse
Affiliation(s)
| | - Mohammad Bafandeh
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Faramarz Gharagozlou
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Mehdi Vojgani
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Emadeddin Mobedi
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Vahid Akbarinejad
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.
| |
Collapse
|
17
|
Huang Y, Su S, Luo W, Zhong H, Wang X, Lyu G. Effects and mechanisms of intrauterine chronic hypoxia on ovarian reserve function of zygotic rats. Sci Rep 2023; 13:19771. [PMID: 37957219 PMCID: PMC10643435 DOI: 10.1038/s41598-023-47088-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Accepted: 11/08/2023] [Indexed: 11/15/2023] Open
Abstract
Chronic intrauterine hypoxia (ICH) may lead to permanent alterations in the offspring's body structure, function, and metabolism through the "developmental programming" pathway, resulting in lasting changes in physiology and metabolism, as well as the onset of adult-onset diseases. The aim was to investigate intrauterine growth restriction caused by ICH and its effect on ovarian reserve function in female offspring at different developmental stages after birth. Healthy female Sprague-Dawley rats (n = 20) were pregnant by normal mating, and the rats in the ICH group were treated with chronic intrauterine hypoxia twice a day for 04 h00 each time from day 4 to 21 of gestation. After the first hypoxic treatment, four pregnant rats were randomly selected from the ICH and natural control groups for arterial blood gas analysis. In the ICH group, birth weight and body weight on the 5th day after birth were less than in the control group, the total number of follicles and the number of primordial follicles in the offspring of the ICH group were significantly reduced on postnatal days 5, 20, and 40 (p < 0.05). ICH decreases ovarian reserve function in female offspring rats and programmatically regulates the differential expression of ovarian miRNAs in female offspring rats.
Collapse
Affiliation(s)
- Yanyan Huang
- Department of Reproductive Medicine, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, China
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, China
| | - Shanshan Su
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, China
| | - Weiwen Luo
- Department of Ultrasound, Zhangzhou Hospital, Zhangzhou, 363000, Fujian Province, China
| | - Huohu Zhong
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, China
| | - Xiali Wang
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, China
- Collaborative Innovation Center for Maternal and Infant Health Service Application Technology of Education Ministry, Quanzhou Medical College, Quanzhou, 362000, China
- Department of Clinical Medicine, Quanzhou Medical College, Quanzhou, 362000, China
| | - Guorong Lyu
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, China.
- Collaborative Innovation Center for Maternal and Infant Health Service Application Technology of Education Ministry, Quanzhou Medical College, Quanzhou, 362000, China.
- Department of Clinical Medicine, Quanzhou Medical College, Quanzhou, 362000, China.
| |
Collapse
|
18
|
Bafandeh M, Mozaffari Makiabadi MJ, Gharagozlou F, Vojgani M, Mobedi E, Akbarinejad V. Developmental programming of production and reproduction in dairy cows: I. Association of maternal parity with offspring's birth weight, milk yield, reproductive performance and AMH concentration during the first lactation period. Theriogenology 2023; 210:34-41. [PMID: 37473594 DOI: 10.1016/j.theriogenology.2023.07.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 07/05/2023] [Accepted: 07/12/2023] [Indexed: 07/22/2023]
Abstract
Multiparous dams have been reported to produce offspring with greater fertility and higher AMH concentration, as a marker of ovarian reserves, as compared with nulliparous and primiparous dams. Yet it has remained to be addressed whether this phenomenon can still be true for old multiparous cows which might experience some geriatric changes in their reproductive system. Therefore, the present study was conducted to evaluate the productive and reproductive performance of offspring with different maternal parity. To this end, offspring were classified based on their maternal parities into four categories, including offspring of nulliparous (no previous parity), primiparous (one previous parity), young multiparous (two to six previous parities) and old multiparous (seven or more previous parities) dams. In study I, data of birth weight, milk yield and reproductive variables of 11,788 offspring and data of their maternal parity were retrieved. In study II, blood samples (n = 521) were collected from offspring with various maternal parity for measurement of serum AMH. Birth weight was the lowest in the offspring of nulliparous dams (P < 0.0001) and it was lower in offspring of primiparous and old multiparous dams than offspring of young multiparous dams (P < 0.05). Milk production was the lowest in offspring of old multiparous dams (P < 0.01), and it was lower in offspring of young multiparous dams than offspring of nulliparous and primiparous dams (P < 0.0001). Offspring of old multiparous dams had greater first service conception rate, less services per conception and shorter calving to conception interval than offspring of nulliparous, primiparous and young multiparous dams (P < 0.05). Furthermore, AMH concentration was higher in offspring of old multiparous dams than offspring of nulliparous and primiparous dams (P < 0.05). In conclusion, the present study revealed greater milk production in offspring resulting from dams with lower parity, probably due to the genetic selection for improvement of milk production in dairy cows which imparts the younger generations greater genetic merits for milk production. Reproductive performance, however, was greater in offspring born to dams with higher parity, particularly those born to old multiparous dams, and this phenomenon might be related to their lower milk production and higher AMH concentration.
Collapse
Affiliation(s)
- Mohammad Bafandeh
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | | | - Faramarz Gharagozlou
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Mehdi Vojgani
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Emadeddin Mobedi
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Vahid Akbarinejad
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.
| |
Collapse
|
19
|
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
|
20
|
Galli GLJ, Lock MC, Smith KLM, Giussani DA, Crossley DA. Effects of Developmental Hypoxia on the Vertebrate Cardiovascular System. Physiology (Bethesda) 2023; 38:0. [PMID: 36317939 DOI: 10.1152/physiol.00022.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 10/24/2022] [Accepted: 10/26/2022] [Indexed: 01/04/2023] Open
Abstract
Developmental hypoxia has profound and persistent effects on the vertebrate cardiovascular system, but the nature, magnitude, and long-term outcome of the hypoxic consequences are species specific. Here we aim to identify common and novel cardiovascular responses among vertebrates that encounter developmental hypoxia, and we discuss the possible medical and ecological implications.
Collapse
Affiliation(s)
- Gina L J Galli
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Mitchell C Lock
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Kerri L M Smith
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Dino A Giussani
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Dane A Crossley
- Department of Biological Sciences, University of North Texas, Denton, Texas
| |
Collapse
|
21
|
Serum Adipocytokines Profile in Children Born Small and Appropriate for Gestational Age-A Comparative Study. Nutrients 2023; 15:nu15040868. [PMID: 36839226 PMCID: PMC9962615 DOI: 10.3390/nu15040868] [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: 12/09/2022] [Revised: 01/04/2023] [Accepted: 01/10/2023] [Indexed: 02/11/2023] Open
Abstract
BACKGROUND Adipose tissue is not only a storage place for fat, but also an endocrine organ, secreting bioactive molecules which influence body metabolism. Such molecules are known as adipocytokines. In the past years the coincidence between adipocytokines and fetal growth restriction disorders was found. The aim of the study was to estimate serum levels of adiponectin, leptin and resistin in children born small for gestational age, compared to children born at an appropriate size for gestational age. METHODS The study consisted of 35 children aged seven to nine years, born SGA (small for gestational age) on term and 25 healthy children (14 girls, 11 boys), born with proper birthweight (AGA-appropriate for gestational age)-control group. RESULTS Adiponectin and leptin levels were significantly higher in the SGA group compared to the AGA group (p = 0.023, p = 0.018 respectively). The resistin values were comparable in both groups of patients. There was a positive correlation between serum leptin concentration and current body weight in SGA group (r = 0.28; p = 0.108). In turn, adiponectin levels in this group of patients negatively correlated with actual body weight (r = -0.51; p = 0.002). The negative correlation between body mass index and plasma adiponectin levels was found only in children born SGA. SGA children had significantly higher values of diastolic blood pressure. There was negative correlation between serum adiponectin level and systolic blood pressure in SGA children. In the SGA group the phenomenon of catch-up growth was observed in 32 children. CONCLUSIONS Children born SGA have abnormal adipose tissue biomarkers profiles.
Collapse
|
22
|
Salinas-Salmon CE, Murillo-Jauregui C, Gonzales-Isidro M, Espinoza-Pinto V, Mendoza SV, Ruiz R, Vargas R, Perez Y, Montaño J, Toledo L, Badner A, Jimenez J, Peñaranda J, Romero C, Aguilar M, Riveros-Gonzales L, Arana I, Villamor E. Elevation of Pulmonary Artery Pressure in Newborns from High-Altitude Pregnancies Complicated by Preeclampsia. Antioxidants (Basel) 2023; 12:347. [PMID: 36829907 PMCID: PMC9952561 DOI: 10.3390/antiox12020347] [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: 11/30/2022] [Revised: 01/06/2023] [Accepted: 01/17/2023] [Indexed: 02/05/2023] Open
Abstract
We hypothesized that fetal exposure to the oxidative stress induced by the combined challenge of preeclampsia (PE) and high altitude would induce a significant impairment in the development of pulmonary circulation. We conducted a prospective study in La Paz (Bolivia, mean altitude 3625 m) in which newborns from singleton pregnancies with and without PE were compared (PE group n = 69, control n = 70). We conducted an echocardiographic study in these infants at the median age of two days. The percentage of cesarean deliveries and small for gestational age (SGA) infants was significantly higher in the PE group. Heart rate, respiratory rate, and oxygen saturation did not vary significantly between groups. Estimated pulmonary arterial pressure and pulmonary vascular resistance were 30% higher in newborns exposed to PE and high altitude compared with those exposed only to high altitude. We also detected signs of right ventricular hypertrophy in infants subjected to both exposures. In conclusion, this study provides evidence that the combination of PE and pregnancy at high altitude induces subclinical alterations in the pulmonary circulation of the newborn. Follow-up of this cohort may provide us with valuable information on the potential increased susceptibility to developing pulmonary hypertension or other pulmonary and cardiovascular disorders.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Jesús Jimenez
- Instituto Boliviano de Biología de Altura (IBBA), UMSA, La Paz, Bolivia
| | | | - Catherine Romero
- Instituto Boliviano de Biología de Altura (IBBA), UMSA, La Paz, Bolivia
| | - Martha Aguilar
- Instituto Boliviano de Biología de Altura (IBBA), UMSA, La Paz, Bolivia
| | | | | | - Eduardo Villamor
- Maastricht University Medical Center (MUMC+), School for Oncology and Reproduction (GROW), 6202AZ Maastricht, The Netherlands
| |
Collapse
|
23
|
Ramli I, Posadino AM, Giordo R, Fenu G, Fardoun M, Iratni R, Eid AH, Zayed H, Pintus G. Effect of Resveratrol on Pregnancy, Prenatal Complications and Pregnancy-Associated Structure Alterations. Antioxidants (Basel) 2023; 12:antiox12020341. [PMID: 36829900 PMCID: PMC9952837 DOI: 10.3390/antiox12020341] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/24/2023] [Accepted: 01/28/2023] [Indexed: 02/04/2023] Open
Abstract
Adverse pregnancy outcomes are considered significant health risks for pregnant women and their offspring during pregnancy and throughout their lifespan. These outcomes lead to a perturbated in-utero environment that impacts critical phases of the fetus's life and correlates to an increased risk of chronic pathological conditions, such as diabetes, obesity, and cardiovascular diseases, in both the mother's and adult offspring's life. The dietary intake of naturally occurring antioxidants promotes health benefits and disease prevention. In this regard, maternal dietary intake of polyphenolic antioxidants is linked to a reduced risk of maternal obesity and cardio-metabolic disorders, positively affecting both the fetus and offspring. In this work, we will gather and critically appraise the current literature highlighting the effect/s of the naturally occurring polyphenol antioxidant resveratrol on oxidative stress, inflammation, and other molecular and physiological phenomena associated with pregnancy and pregnancy conditions, such as gestational diabetes, preeclampsia, and preterm labor. The resveratrol impact on prenatal complications and pregnancy-associated structures, such as the fetus and placenta, will also be discussed. Finally, we will draw conclusions from the current knowledge and provide future perspectives on potentially exploiting resveratrol as a therapeutic tool in pregnancy-associated conditions.
Collapse
Affiliation(s)
- Iman Ramli
- Departement de Biologie Animale, Université des Frères Mentouri Constantine 1, Constantine 25000, Algeria
| | - Anna Maria Posadino
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy
| | - Roberta Giordo
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai 505055, United Arab Emirates
| | - Grazia Fenu
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy
| | - Manal Fardoun
- Department of Pharmacology and Toxicology, American University of Beirut, Beirut 11-0236, Lebanon
| | - Rabah Iratni
- Department of Biology, United Arab Emirates University, Al-Ain 15551, United Arab Emirates
| | - Ali H. Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha 2713, Qatar
| | - Hatem Zayed
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha 2713, Qatar
| | - Gianfranco Pintus
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy
- Correspondence:
| |
Collapse
|
24
|
The Long-Term Effects of Prenatal Hypoxia on Coronary Artery Function of the Male and Female Offspring. Biomedicines 2022; 10:biomedicines10123019. [PMID: 36551775 PMCID: PMC9776081 DOI: 10.3390/biomedicines10123019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/18/2022] [Accepted: 11/19/2022] [Indexed: 11/25/2022] Open
Abstract
Prenatal hypoxia predisposes the offspring to the development of cardiovascular (CV) dysfunction in adult life. Using a rat model, we assessed the effect of prenatal hypoxia on vasoconstrictive and vasodilative mechanisms in left anterior descending coronary arteries of 4- and 9.5-month-old offspring. Endothelium-dependent relaxation to methylcholine and vasoconstriction responses to endothelin-1 (ET-1) were assessed by wire myography. Prenatal hypoxia impaired endothelium-dependent vasodilation in 4- and 9.5-month-old offspring. Inhibition of nitric oxide (NO) synthase prevented coronary artery relaxation in all groups. Inhibition of prostaglandin H synthase (PGHS) improved relaxation in prenatally hypoxic males and tended to improve vasorelaxation in females, suggesting that impaired vasodilation was mediated via increased PGHS-dependent vasoconstriction. An enhanced contribution of endothelium-dependent hyperpolarization to coronary artery vasodilation was observed in prenatally hypoxic males and females. No changes in endothelial NO synthase (eNOS) and PGHS-1 expressions were observed, while PGHS-2 expression was decreased in only prenatally hypoxic males. At 4 months, ET-1 responses were similar between groups, while ETB inhibition (with BQ788) tended to decrease ET-1-mediated responses in only prenatally hypoxic females. At 9.5 months, ET-1-mediated responses were decreased in only prenatally hypoxic females. Our data suggest that prenatal hypoxia has long-term similar effects on the mechanisms of impaired endothelium-dependent vasodilation in coronary arteries from adult male and female offspring; however, coronary artery contractile capacity is impaired only in prenatally hypoxic females. Understanding the mechanistic pathways involved in the programming of CV disease may allow for the development of therapeutic interventions.
Collapse
|
25
|
Hif-1α regulates Tet1-c-Myc binding involved in depression-like behavior in prenatal hypoxia offspring. Neuroscience 2022; 502:41-51. [PMID: 36041588 DOI: 10.1016/j.neuroscience.2022.08.014] [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: 06/21/2022] [Revised: 08/03/2022] [Accepted: 08/12/2022] [Indexed: 11/21/2022]
Abstract
Prenatal hypoxia (PH) is one of the most common adverse stimulation during pregnancy. The brain is fragile in the fetal period and sensitive to hypoxia. The offspring who have experienced PH may be at increased risk of developing neurodevelopmental disorders after birth and various neuropsychiatric diseases after adulthood. In this study, pregnant mice used to generate PH offspring were treated with hypoxia (10.5% oxygen) from gestational day 12.5 to 17.5. Compared with control mice, the birth weight of offspring in the PH group was significantly lower and the male adult offspring exhibited significant depression-like behavior. The expression of the oxygen-sensitive subunit of hypoxia-inducible factor (Hif-1α) was significantly elevated, whereas Ten-eleven translocated methylcytosine dioxygenase 1 (Tet1) and c-Myc, which is closely related to cell proliferation, was significantly decreased in the hippocampus of the male offspring in the PH group. In addition, the PH group showed increased binding of Hif-1α to Tet1, and decreased binding of Tet1 to c-Myc, resulting in increased ubiquitinated degradation of c-Myc and decreased neurogenesis in the hippocampus of the male offspring. These findings suggest that Hif-1α regulates Tet1-c-Myc binding involved in depression-like behavior in PH offspring and Hif-1α can be used as a detection index of stress-related diseases.
Collapse
|
26
|
Effects of Maternal Nutrient Restriction and Melatonin Supplementation on Cardiomyocyte Cell Development Parameters Using Machine Learning Techniques. Animals (Basel) 2022; 12:ani12141818. [PMID: 35883365 PMCID: PMC9311781 DOI: 10.3390/ani12141818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 06/07/2022] [Accepted: 07/02/2022] [Indexed: 11/17/2022] Open
Abstract
The objective of the current study was to examine the effects of maternal feed restriction and melatonin supplementation on fetal cardiomyocyte cell development parameters and predict binucleation and hypertrophy using machine learning techniques using pregnant beef heifers. Brangus heifers (n = 29) were assigned to one of four treatment groups in a 2 × 2 factorial design at day 160 of gestation: (1) 100% of nutrient requirements (adequately fed; ADQ) with no dietary melatonin (CON); (2) 100% of nutrient requirements (ADQ) with 20 mg/d of dietary melatonin (MEL); (3) 60% of nutrient requirements (nutrient-restricted; RES) with no dietary melatonin (CON); (4) 60% of nutrient requirements (RES) with 20 mg/d of dietary melatonin (MEL). On day 240 of gestation, fetuses were removed, and fetal heart weight and thickness were determined. The large blood vessel perimeter was increased in fetuses from RES compared with ADQ (p = 0.05). The total number of capillaries per tissue area exhibited a nutrition by treatment interaction (p = 0.01) where RES-MEL increased capillary number compared (p = 0.03) with ADQ-MEL. The binucleated cell number per tissue area showed a nutrition by treatment interaction (p = 0.010), where it was decreased in RES-CON vs. ADQ-CON fetuses. Hypertrophy was estimated by dividing ventricle thickness by heart weight. Based on machine learning results, for the binucleation and hypertrophy target variables, the Bagging model with 5 Decision Tree estimators and 3 Decision Tree estimators produced the best results without overfitting. In the prediction of binucleation, left heart ventricular thickness feature had the highest Gin importance weight followed by fetal body weight. In the case of hypertrophy, heart weight was the most important feature. This study provides evidence that restricted maternal nutrition leads to a reduction in the number of cardiomyocytes while melatonin treatment can mitigate some of these disturbances.
Collapse
|
27
|
Intergenerational effects of prenatal hypoxia exposure on uterine artery adaptations to pregnancies in the female offspring. J Dev Orig Health Dis 2022; 13:794-799. [PMID: 35616050 DOI: 10.1017/s2040174422000216] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Prenatal hypoxia is a common complication of pregnancy and is associated with detrimental health outcomes, such as impaired cardiac and vascular function, in adult offspring. Exposure to prenatal hypoxia reportedly impacts the reproductive system of female offspring. Whether exposure to prenatal hypoxia influences pregnancy adaptations and outcomes in these female offspring is unknown. We hypothesised that prenatal hypoxia impairs uterine artery adaptations in pregnancies of the adult offspring. Pregnancy outcomes and uterine artery function were assessed in 14-16 weeks old non-pregnant and late pregnant (gestational day 20; term = 22 days) adult female offspring born to rats exposed to prenatal normoxia (21% oxygen) or hypoxia (11% oxygen, between days 15-21 of gestation). Compared with normoxia controls, prenatal hypoxia was associated with pregnant adult offspring having reduced placental weights in their litters, and uterine artery circumferential stress that increased with pregnancy. Overall, prenatal hypoxia adversely, albeit mildly, compromised pregnancies of adult offspring.
Collapse
|
28
|
Yao C, Lu L, Ji Y, Zhang Y, Li W, Shi Y, Liu J, Sun M, Xia F. Hypo-Hydroxymethylation of Nobox is Associated with Ovarian Dysfunction in Rat Offspring Exposed to Prenatal Hypoxia. Reprod Sci 2022; 29:1424-1436. [PMID: 35257353 PMCID: PMC9005429 DOI: 10.1007/s43032-022-00866-6] [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: 04/27/2021] [Accepted: 01/23/2022] [Indexed: 11/23/2022]
Abstract
Prenatal hypoxia (PH) is a common feature of a suboptimal intrauterine environment affecting the development of fetuses. Whether PH leads to abnormal ovary development is not yet clear. This study investigated ovarian function in offspring exposed to PH and the potential underlying molecular mechanisms. SD female rats (n = 12 per group) at 9 weeks of age were housed in individual cages (21% O2). After the pregnant rats were exposed to hypoxia (10.5% oxygen) from embryonic day (E) 5 to E21, PH offspring were generated. All animals maintained normoxia during lactation. The number of follicles was counted in female offspring at 3 months under an optical microscope. The expression of Nobox, Gdf9, and Tets was detected by quantitative real-time polymerase chain reaction (PCR) and Western blot. Global DNA hydroxymethylation was measured by dot blot. The hydroxymethylation level of the Nobox gene was evaluated with an NGS-based multiple targeted CpG hydroxymethylation analysis method. Body weight and ovary weight were significantly decreased in the PH group compared with the control group. PH offspring have abnormal estrous cycle, decreased serum anti-Mullerian hormone (AMH), and increased serum follicle-stimulating hormone (FSH), and follicular atresia, which are consistent with the clinical manifestations in patients with ovarian dysfunction. In terms of mechanism, the expression of Nobox was significantly decreased in the PH group. Subsequent high-throughput sequencing results showed that the level of hydroxymethylation in the candidate region of the Nobox gene was reduced. Cultured cells treated with hypoxia exhibited lower levels of both 5hmC and Nobox, while vitamin C, a coactivator of Tets, rescued hypo-hydroxymethylation and increased the expression level of Nobox. This study indicated that PH could cause hypo-hydroxymethylation of Nobox through epigenetic regulation and may consequently contribute to ovarian dysfunction in adult rat offspring.
Collapse
Affiliation(s)
- Changfang Yao
- Reproductive Medicine Center of the First Affiliated Hospital of Soochow University, Suzhou, 215006 Jiangsu China
- Obstetrics of the Third Affiliated Hospital of Soochow University, Changzhou, 213000 Jiangsu China
| | - Likui Lu
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu China
| | - Yiting Ji
- Reproductive Medicine Center of the First Affiliated Hospital of Soochow University, Suzhou, 215006 Jiangsu China
| | - Yingying Zhang
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu China
| | - Weisheng Li
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu China
| | - Yajun Shi
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu China
| | - Jinliu Liu
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu China
| | - Miao Sun
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu China
| | - Fei Xia
- Reproductive Medicine Center of the First Affiliated Hospital of Soochow University, Suzhou, 215006 Jiangsu China
| |
Collapse
|
29
|
Grant ID, Giussani DA, Aiken CE. Fetal growth and spontaneous preterm birth in high-altitude pregnancy: A systematic review, meta-analysis, and meta-regression. Int J Gynaecol Obstet 2022; 157:221-229. [PMID: 34101174 DOI: 10.1002/ijgo.13779] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 05/17/2021] [Accepted: 06/07/2021] [Indexed: 01/12/2023]
Abstract
OBJECTIVE To understand the relationship between birth weight and altitude to improve health outcomes in high-altitude populations, to systematically assess the impact of altitude on the likelihood of low birth weight (LBW), small for gestational age (SGA), and spontaneous preterm birth (sPTB), and to estimate the magnitude of reduced birth weight associated with altitude. METHODS PubMed, OvidEMBASE, Cochrane Library, Medline, Web of Science, and clinicaltrials.gov were searched (from inception to November 11, 2020). Observational, cohort, or case-control studies were included if they reported a high altitude (>2500 m) and appropriate control population. RESULTS Of 2524 studies identified, 59 were included (n = 1 604 770 pregnancies). Data were abstracted according to PRISMA guidelines, and were pooled using random-effects models. There are greater odds of LBW (odds ratio [OR] 1.47, 95% confidence interval [CI] 1.33-1.62, P < 0.001), SGA (OR 1.88, 95% CI 1.08-3.28, P = 0.026), and sPTB (OR 1.23, 95% CI 1.04-1.47, P = 0.016) in high- versus low-altitude pregnancies. Birth weight decreases by 54.7 g (±13.0 g, P < 0.0001) per 1000 m increase in altitude. Average gestational age at delivery was not significantly different. CONCLUSION Globally, the likelihood of adverse perinatal outcomes, including LBW, SGA, and sPTB, increases in high-altitude pregnancies. There is an inverse relationship between birth weight and altitude. These findings have important implications for the increasing global population living at altitudes above 2500 m.
Collapse
Affiliation(s)
- Imogen D Grant
- University Department of Obstetrics and Gynaecology, University of Cambridge, NIHR Cambridge Biomedical Research Centre, Cambridge, UK
| | - Dino A Giussani
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Catherine E Aiken
- University Department of Obstetrics and Gynaecology, University of Cambridge, NIHR Cambridge Biomedical Research Centre, Cambridge, UK
| |
Collapse
|
30
|
Starr VJ, Dzialowski EM. Developing chicken cardiac muscle mitochondria are resistant to variations in incubation oxygen levels. Curr Res Physiol 2022; 5:151-157. [PMID: 35345510 PMCID: PMC8956876 DOI: 10.1016/j.crphys.2022.03.001] [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: 12/22/2021] [Revised: 03/05/2022] [Accepted: 03/14/2022] [Indexed: 11/25/2022] Open
Abstract
Background Chronic exposure to hypoxia during vertebrate development can produce abnormal cardiovascular morphology and function. The aim of this study was to examine cardiac mitochondria function in an avian model, the chicken, in response to embryonic development under hypoxic (15% O2), normoxic (21% O2), or hyperoxic (40% O2) incubation conditions. Methods Chicken embryos were incubated in hypoxia, normoxia, or hyperoxia beginning on day 5 of incubation through hatching. Cardiac mitochondria oxygen flux and reactive oxygen species production were measured in permeabilized cardiac fibers from externally pipped and 1-day post hatchlings. Results Altering oxygen during development had a large effect on body and heart masses of externally pipped embryos and 1-day old hatchlings. Hypoxic animals had smaller body masses and absolute heart masses, but proportionally similar sized hearts compared to normoxic animals during external pipping. Hyperoxic animals were larger with larger hearts than normoxic animals during external pipping. Mitochondrial oxygen flux in permeabilized cardiac muscle fibers revealed limited effects of developing under altered oxygen conditions, with only oxygen flux through cytochrome oxidase being lower in hypoxic hearts compared with hyperoxic hearts. Oxygen flux in leak and oxidative phosphorylation states were not affected by developmental oxygen levels. Mitochondrial reactive oxygen species production under leak and oxidative phosphorylation states studied did not differ between any developmental oxygen treatment. Conclusions These results suggest that cardiac mitochondria function of the developing chicken is not altered by developing in ovo under different oxygen levels. Chicken heart mass is influenced by oxygen availability during development. Cardiac mitochondria respiration was unchanged by developing under hypoxic or hyperoxic oxygen stress. Cardiac mitochondria ROS production was not altered by developmental oxygen stress.
Collapse
Key Words
- AA, Antimycin A
- ADP, adenosine diphosphate
- COX, cytochrome oxidase
- Cardiac mitochondria
- Chicken
- EP, external pipping
- GMP, glutamate, malate, and pyruvate
- Hyperoxia
- Hypoxia
- IP, internal pipping
- LEAK, mitochondrial leak respiration
- OMY, oligomycin
- OXPHOS, mitochondrial oxidative phosphorylation
- ROS, reactive oxygen species
- ROT, rotenone
- Reactive oxygen species
- S, succinate
- TMPD, N,N,N’,N’-tetramethyl-p-phenylenediamine
- dph, days post hatching
Collapse
Affiliation(s)
- Vanessa J Starr
- Developmental Integrative Biology, Department of Biological Sciences, 1155 Union Circle #305220, University of North Texas, Denton, TX, 76203, USA
| | - Edward M Dzialowski
- Developmental Integrative Biology, Department of Biological Sciences, 1155 Union Circle #305220, University of North Texas, Denton, TX, 76203, USA
| |
Collapse
|
31
|
Liu Y, Luo Z, Liao Z, Wang M, Zhou Y, Luo S, Ding Y, Liu T, Cao C, Yue S. Effects of Excessive Activation of N-methyl-D-aspartic Acid Receptors in Neonatal Cardiac Mitochondrial Dysfunction Induced by Intrauterine Hypoxia. Front Cardiovasc Med 2022; 9:837142. [PMID: 35498024 PMCID: PMC9039344 DOI: 10.3389/fcvm.2022.837142] [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: 12/16/2021] [Accepted: 03/04/2022] [Indexed: 11/13/2022] Open
Abstract
Intrauterine hypoxia is a common complication during pregnancy and could increase the risk of cardiovascular disease in offspring. However, the underlying mechanism is controversial. Memantine, an NMDA receptor antagonist, is reported to be a potential cardio-protective agent. We hypothesized that antenatal memantine treatment could prevent heart injury in neonatal offspring exposed to intrauterine hypoxia. Pregnant rats were exposed to gestational hypoxia or antenatal memantine treatment during late pregnancy. Newborns were then sacrificed to assess multiple parameters. The results revealed that Intrauterine hypoxia resulted in declining birth weight, heart weight, and an abnormally high heart weight/birth weight ratio. Furthermore, intrauterine hypoxia caused mitochondrial structural, functional abnormalities and decreased expression of DRP1, and upregulation of NMDAR1 in vivo. Antenatal memantine treatment,an NMDARs antagonist, improved these changes. In vitro, hypoxia increased the glutamate concentration and expression of NMDAR1. NMDAR activation may lead to similar changes in mitochondrial function, structure, and downregulation of DRP1 in vitro. Pharmacological blockade of NMDARs by the non-competitive NMDA antagonist MK-801 or knockdown of the glutamate receptor NR1 significantly attenuated the increased mitochondrial reactive oxygen species and calcium overload-induced by hypoxia exposure. These facts suggest that memantine could provide a novel and promising treatment for clinical use in intrauterine hypoxia during pregnancy to protect the cardiac mitochondrial function in the offspring. To our best knowledge, our research is the first study that shows intrauterine hypoxia can excessively activate cardiac NMDARs and thus cause mitochondrial dysfunction.
Collapse
Affiliation(s)
- Yang Liu
- Department of Neonatology, Xiangya Hospital, Central South University, Changsha, China
| | - Ziqiang Luo
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, China
| | - Zhengchang Liao
- Department of Neonatology, Xiangya Hospital, Central South University, Changsha, China
| | - Mingjie Wang
- Department of Neonatology, Xiangya Hospital, Central South University, Changsha, China
| | - Yan Zhou
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, China
| | - Siwei Luo
- Departments of Pediatrics and Neonatology, Children's Hospital of Fudan University, Shanghai, China
- Laboratory of Neonatal Diseases, National Children's Medical Center, National Commission of Health, Shanghai, China
| | - Ying Ding
- Department of Neonatology, Xiangya Hospital, Central South University, Changsha, China
| | - Teng Liu
- Department of Neonatology, Xiangya Hospital, Central South University, Changsha, China
| | - Chuangding Cao
- Department of Neonatology, Xiangya Hospital, Central South University, Changsha, China
| | - Shaojie Yue
- Department of Neonatology, Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Shaojie Yue
| |
Collapse
|
32
|
Sutovska H, Babarikova K, Zeman M, Molcan L. Prenatal Hypoxia Affects Foetal Cardiovascular Regulatory Mechanisms in a Sex- and Circadian-Dependent Manner: A Review. Int J Mol Sci 2022; 23:2885. [PMID: 35270026 PMCID: PMC8910900 DOI: 10.3390/ijms23052885] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 02/28/2022] [Accepted: 03/05/2022] [Indexed: 11/17/2022] Open
Abstract
Prenatal hypoxia during the prenatal period can interfere with the developmental trajectory and lead to developing hypertension in adulthood. Prenatal hypoxia is often associated with intrauterine growth restriction that interferes with metabolism and can lead to multilevel changes. Therefore, we analysed the effects of prenatal hypoxia predominantly not associated with intrauterine growth restriction using publications up to September 2021. We focused on: (1) The response of cardiovascular regulatory mechanisms, such as the chemoreflex, adenosine, nitric oxide, and angiotensin II on prenatal hypoxia. (2) The role of the placenta in causing and attenuating the effects of hypoxia. (3) Environmental conditions and the mother's health contribution to the development of prenatal hypoxia. (4) The sex-dependent effects of prenatal hypoxia on cardiovascular regulatory mechanisms and the connection between hypoxia-inducible factors and circadian variability. We identified that the possible relationship between the effects of prenatal hypoxia on the cardiovascular regulatory mechanism may vary depending on circadian variability and phase of the days. In summary, even short-term prenatal hypoxia significantly affects cardiovascular regulatory mechanisms and programs hypertension in adulthood, while prenatal programming effects are not only dependent on the critical period, and sensitivity can change within circadian oscillations.
Collapse
Affiliation(s)
| | | | - Michal Zeman
- Department of Animal Physiology and Ethology, Faculty of Natural Sciences, Comenius University, 842 15 Bratislava, Slovakia; (H.S.); (K.B.); (L.M.)
| | | |
Collapse
|
33
|
Maternal metabolizable protein restriction during gestation affects the vascular function of maternal and fetal placental arteries in sheep. Theriogenology 2022; 185:24-33. [DOI: 10.1016/j.theriogenology.2022.03.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 02/10/2022] [Accepted: 03/15/2022] [Indexed: 11/19/2022]
|
34
|
Graf AV, Maslova MV, Artiukhov AV, Ksenofontov AL, Aleshin VA, Bunik VI. Acute Prenatal Hypoxia in Rats Affects Physiology and Brain Metabolism in the Offspring, Dependent on Sex and Gestational Age. Int J Mol Sci 2022; 23:2579. [PMID: 35269722 PMCID: PMC8910449 DOI: 10.3390/ijms23052579] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Revised: 02/22/2022] [Accepted: 02/23/2022] [Indexed: 01/27/2023] Open
Abstract
Hypoxia is damaging to the fetus, but the developmental impact may vary, with underlying molecular mechanisms unclear. We demonstrate the dependence of physiological and biochemical effects of acute prenatal hypoxia (APH) on sex and gestational age. Compared to control rats, APH on the 10th day of pregnancy (APH-10) increases locomotion in both the male and female offspring, additionally increasing exploratory activity and decreasing anxiety in the males. Compared to APH-10, APH on the 20th day of pregnancy (APH-20) induces less behavioral perturbations. ECG is changed similarly in all offspring only by APH-10. Sexual dimorphism in the APH outcome on behavior is also observed in the brain acetylation system and 2-oxoglutarate dehydrogenase reaction, essential for neurotransmitter metabolism. In view of the perturbed behavior, more biochemical parameters in the brains are assessed after APH-20. Of the six enzymes, APH-20 significantly decreases the malic enzyme activity in both sexes. Among 24 amino acids and dipeptides, APH-20 increases the levels of only three amino acids (Phe, Thr, and Trp) in male offspring, and of seven amino acids (Glu, Gly, Phe, Trp, Ser, Thr, Asn) and carnosine in the female offspring. Thus, a higher reactivity of the brain metabolism to APH stabilizes the behavior. The behavior and brain biochemistry demonstrate sexually dimorphic responses to APH at both gestational stages, whereas the APH effects on ECG depend on gestational age rather than sex.
Collapse
Affiliation(s)
- Anastasia V. Graf
- Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia; (A.V.G.); (M.V.M.)
- Department of Biokinetics, A. N. Belozersky Institute of Physicochemical Biology, Lomonosov Moscow State University, 119234 Moscow, Russia; (A.V.A.); (A.L.K.); (V.A.A.)
| | - Maria V. Maslova
- Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia; (A.V.G.); (M.V.M.)
| | - Artem V. Artiukhov
- Department of Biokinetics, A. N. Belozersky Institute of Physicochemical Biology, Lomonosov Moscow State University, 119234 Moscow, Russia; (A.V.A.); (A.L.K.); (V.A.A.)
- Department of Biochemistry, Sechenov University, 119048 Moscow, Russia
| | - Alexander L. Ksenofontov
- Department of Biokinetics, A. N. Belozersky Institute of Physicochemical Biology, Lomonosov Moscow State University, 119234 Moscow, Russia; (A.V.A.); (A.L.K.); (V.A.A.)
| | - Vasily A. Aleshin
- Department of Biokinetics, A. N. Belozersky Institute of Physicochemical Biology, Lomonosov Moscow State University, 119234 Moscow, Russia; (A.V.A.); (A.L.K.); (V.A.A.)
- Department of Biochemistry, Sechenov University, 119048 Moscow, Russia
| | - Victoria I. Bunik
- Department of Biokinetics, A. N. Belozersky Institute of Physicochemical Biology, Lomonosov Moscow State University, 119234 Moscow, Russia; (A.V.A.); (A.L.K.); (V.A.A.)
- Department of Biochemistry, Sechenov University, 119048 Moscow, Russia
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119234 Moscow, Russia
| |
Collapse
|
35
|
Jones AK, Wang D, Goldstrohm DA, Brown LD, Rozance PJ, Limesand SW, Wesolowski SR. Tissue-specific responses that constrain glucose oxidation and increase lactate production with the severity of hypoxemia in fetal sheep. Am J Physiol Endocrinol Metab 2022; 322:E181-E196. [PMID: 34957858 PMCID: PMC8816623 DOI: 10.1152/ajpendo.00382.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Fetal hypoxemia decreases insulin and increases cortisol and norepinephrine concentrations and may restrict growth by decreasing glucose utilization and altering substrate oxidation. Specifically, we hypothesized that hypoxemia would decrease fetal glucose oxidation and increase lactate and pyruvate production. We tested this by measuring whole body glucose oxidation and lactate production, and molecular pathways in liver, muscle, adipose, and pancreas tissues of fetuses exposed to maternal hypoxemia for 9 days (HOX) compared with control fetal sheep (CON) in late gestation. Fetuses with more severe hypoxemia had lower whole body glucose oxidation rates, and HOX fetuses had increased lactate production from glucose. In muscle and adipose tissue, expression of the glucose transporter GLUT4 was decreased. In muscle, pyruvate kinase (PKM) and lactate dehydrogenase B (LDHB) expression was decreased. In adipose tissue, LDHA and lactate transporter (MCT1) expression was increased. In liver, there was decreased gene expression of PKLR and MPC2 and phosphorylation of PDH, and increased LDHA gene and LDH protein abundance. LDH activity, however, was decreased only in HOX skeletal muscle. There were no differences in basal insulin signaling across tissues, nor differences in pancreatic tissue insulin content, β-cell area, or genes regulating β-cell function. Collectively, these results demonstrate coordinated metabolic responses across tissues in the hypoxemic fetus that limit glucose oxidation and increase lactate and pyruvate production. These responses may be mediated by hypoxemia-induced endocrine responses including increased norepinephrine and cortisol, which inhibit pancreatic insulin secretion resulting in lower insulin concentrations and decreased stimulation of glucose utilization.NEW & NOTEWORTHY Hypoxemia lowered fetal glucose oxidation rates, based on severity of hypoxemia, and increased lactate production. This was supported by tissue-specific metabolic responses that may result from increased norepinephrine and cortisol concentrations, which decrease pancreatic insulin secretion and insulin concentrations and decrease glucose utilization. This highlights the vulnerability of metabolic pathways in the fetus and demonstrates that constrained glucose oxidation may represent an early event in response to sustained hypoxemia and fetal growth restriction.
Collapse
Affiliation(s)
- Amanda K Jones
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado
| | - Dong Wang
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado
| | - David A Goldstrohm
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado
| | - Laura D Brown
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado
| | - Paul J Rozance
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado
| | - Sean W Limesand
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, Arizona
| | | |
Collapse
|
36
|
Hansell JA, Richter HG, Camm EJ, Herrera EA, Blanco CE, Villamor E, Patey OV, Lock MC, Trafford AW, Galli GLJ, Giussani DA. Maternal melatonin: Effective intervention against developmental programming of cardiovascular dysfunction in adult offspring of complicated pregnancy. J Pineal Res 2022; 72:e12766. [PMID: 34634151 DOI: 10.1111/jpi.12766] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 08/16/2021] [Accepted: 08/26/2021] [Indexed: 12/01/2022]
Abstract
Adopting an integrative approach, by combining studies of cardiovascular function with those at cellular and molecular levels, this study investigated whether maternal treatment with melatonin protects against programmed cardiovascular dysfunction in the offspring using an established rodent model of hypoxic pregnancy. Wistar rats were divided into normoxic (N) or hypoxic (H, 10% O2 ) pregnancy ± melatonin (M) treatment (5 μg·ml-1 .day-1 ) in the maternal drinking water. Hypoxia ± melatonin treatment was from day 15-20 of gestation (term is ca. 22 days). To control for possible effects of maternal hypoxia-induced reductions in maternal food intake, additional dams underwent pregnancy under normoxic conditions but were pair-fed (PF) to the daily amount consumed by hypoxic dams from day 15 of gestation. In one cohort of animals from each experimental group (N, NM, H, HM, PF, PFM), measurements were made at the end of gestation. In another, following delivery of the offspring, investigations were made at adulthood. In both fetal and adult offspring, fixed aorta and hearts were studied stereologically and frozen hearts were processed for molecular studies. In adult offspring, mesenteric vessels were isolated and vascular reactivity determined by in-vitro wire myography. Melatonin treatment during normoxic, hypoxic or pair-fed pregnancy elevated circulating plasma melatonin in the pregnant dam and fetus. Relative to normoxic pregnancy, hypoxic pregnancy increased fetal haematocrit, promoted asymmetric fetal growth restriction and resulted in accelerated postnatal catch-up growth. Whilst fetal offspring of hypoxic pregnancy showed aortic wall thickening, adult offspring of hypoxic pregnancy showed dilated cardiomyopathy. Similarly, whilst cardiac protein expression of eNOS was downregulated in the fetal heart, eNOS protein expression was elevated in the heart of adult offspring of hypoxic pregnancy. Adult offspring of hypoxic pregnancy further showed enhanced mesenteric vasoconstrictor reactivity to phenylephrine and the thromboxane mimetic U46619. The effects of hypoxic pregnancy on cardiovascular remodelling and function in the fetal and adult offspring were independent of hypoxia-induced reductions in maternal food intake. Conversely, the effects of hypoxic pregnancy on fetal and postanal growth were similar in pair-fed pregnancies. Whilst maternal treatment of normoxic or pair-fed pregnancies with melatonin on the offspring cardiovascular system was unremarkable, treatment of hypoxic pregnancies with melatonin in doses lower than those recommended for overcoming jet lag in humans enhanced fetal cardiac eNOS expression and prevented all alterations in cardiovascular structure and function in fetal and adult offspring. Therefore, the data support that melatonin is a potential therapeutic target for clinical intervention against developmental origins of cardiovascular dysfunction in pregnancy complicated by chronic fetal hypoxia.
Collapse
Affiliation(s)
- Jeremy A Hansell
- Department of Physiology Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Hans G Richter
- Facultad de Medicina, Instituto de Anatomía, Histología y Patología, Universidad Austral de Chile, Valdivia, Chile
| | - Emily J Camm
- Department of Physiology Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Emilio A Herrera
- Programa de Fisiopatología, Facultad de Medicina, Instituto de Ciencias Biomédicas (ICBM), Universidad de Chile, Santiago, Chile
| | - Carlos E Blanco
- National Children's Research Centre, Our Lady's Children's Hospital, Dublin, Ireland
| | - Eduardo Villamor
- Department of Pediatrics, School for Oncology and Developmental Biology (GROW), Maastricht University Medical Center (MUMC+), Maastricht, The Netherlands
| | - Olga V Patey
- Department of Physiology Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Mitchell C Lock
- Division of Cardiovascular Sciences, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Andrew W Trafford
- Division of Cardiovascular Sciences, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Gina L J Galli
- Division of Cardiovascular Sciences, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Dino A Giussani
- Department of Physiology Development and Neuroscience, University of Cambridge, Cambridge, UK
- Cambridge BHF Centre for Research Excellence, Cambridge, UK
- Cambridge Strategic Research Initiative in Reproduction, Cambridge, UK
| |
Collapse
|
37
|
Lakshman R, Spiroski AM, McIver LB, Murphy MP, Giussani DA. Noninvasive Biomarkers for Cardiovascular Dysfunction Programmed in Male Offspring of Adverse Pregnancy. Hypertension 2021; 78:1818-1828. [PMID: 34757774 PMCID: PMC8577293 DOI: 10.1161/hypertensionaha.121.17926] [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] [Indexed: 11/18/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Rama Lakshman
- Department of Physiology, Development and Neuroscience (R.L., A.-M.S., L.B.M., D.A.G.), University of Cambridge, United Kingdom
| | - Ana-Mishel Spiroski
- Department of Physiology, Development and Neuroscience (R.L., A.-M.S., L.B.M., D.A.G.), University of Cambridge, United Kingdom.,Cambridge BHF Centre of Research Excellence (A.-M.S., M.P.M., D.A.G.), University of Cambridge, United Kingdom
| | - Lauren B McIver
- Department of Physiology, Development and Neuroscience (R.L., A.-M.S., L.B.M., D.A.G.), University of Cambridge, United Kingdom
| | - Michael P Murphy
- MRC Mitochondria Biology Unit (M.P.M.), University of Cambridge, United Kingdom.,Cambridge BHF Centre of Research Excellence (A.-M.S., M.P.M., D.A.G.), University of Cambridge, United Kingdom.,Department of Medicine (M.P.M., D.A.G.), University of Cambridge, United Kingdom
| | - Dino A Giussani
- Department of Physiology, Development and Neuroscience (R.L., A.-M.S., L.B.M., D.A.G.), University of Cambridge, United Kingdom.,Cambridge BHF Centre of Research Excellence (A.-M.S., M.P.M., D.A.G.), University of Cambridge, United Kingdom.,Department of Medicine (M.P.M., D.A.G.), University of Cambridge, United Kingdom.,Cambridge Strategic Research Initiative in Reproduction, United Kingdom (D.A.G.)
| |
Collapse
|
38
|
Song H, Polster BM, Thompson LP. Chronic hypoxia alters cardiac mitochondrial complex protein expression and activity in fetal guinea pigs in a sex-selective manner. Am J Physiol Regul Integr Comp Physiol 2021; 321:R912-R924. [PMID: 34730023 PMCID: PMC8714812 DOI: 10.1152/ajpregu.00004.2021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 10/22/2021] [Accepted: 10/22/2021] [Indexed: 11/22/2022]
Abstract
We hypothesize that intrauterine hypoxia (HPX) alters the mitochondrial phenotype in fetal hearts contributing to developmental programming. Pregnant guinea pigs were exposed to normoxia (NMX) or hypoxia (HPX, 10.5% O2), starting at early [25 days (25d), 39d duration] or late gestation (50d, 14d duration). Near-term (64d) male and female fetuses were delivered by hysterotomy from anesthetized sows, and body/organ weights were measured. Left ventricles of fetal hearts were excised and frozen for measurement of expression of complex (I-V) subunits, fusion (Mfn2/OPA1) and fission (DRP1/Fis1) proteins, and enzymatic rates of I and IV from isolated mitochondrial proteins. Chronic HPX decreased fetal body weight and increased relative placenta weight regardless of timing. Early-onset HPX increased I, III, and V subunit levels, increased complex I but decreased IV activities in males but not females (all P < 0.05). Late-onset HPX decreased (P < 0.05) I, III, and V levels in both sexes but increased I and decreased IV activities in males only. Both HPX conditions decreased cardiac mitochondrial DNA content in males only. Neither early- nor late-onset HPX had any effect on Mfn2 levels but increased OPA1 in both sexes. Both HPX treatments increased DRP1/Fis1 levels in males. In females, early-onset HPX increased DRP1 with no effect on Fis1, whereas late-onset HPX increased Fis1 with no effect on DRP1. We conclude that both early- and late-onset HPX disrupts the expression/activities of select complexes that could reduce respiratory efficiency and shifts dynamics toward fission in fetal hearts. Thus, intrauterine HPX disrupts the mitochondrial phenotype predominantly in male fetal hearts, potentially altering cardiac metabolism and predisposing the offspring to heart dysfunction.
Collapse
Affiliation(s)
- Hong Song
- Department of Obstetrics, Gynecology and Reproductive Sciences, School of Medicine, University of Maryland, Baltimore, Maryland
| | - Brian M Polster
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research, School of Medicine, University of Maryland, Baltimore, Maryland
| | - Loren P Thompson
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research, School of Medicine, University of Maryland, Baltimore, Maryland
| |
Collapse
|
39
|
Abstract
Heart disease remains one of the greatest killers. In addition to genetics and traditional lifestyle risk factors, we now understand that adverse conditions during pregnancy can also increase susceptibility to cardiovascular disease in the offspring. Therefore, the mechanisms by which this occurs and possible preventative therapies are of significant contemporary interest to the cardiovascular community. A common suboptimal pregnancy condition is a sustained reduction in fetal oxygenation. Chronic fetal hypoxia results from any pregnancy with increased placental vascular resistance, such as in preeclampsia, placental infection, or maternal obesity. Chronic fetal hypoxia may also arise during pregnancy at high altitude or because of maternal respiratory disease. This article reviews the short- and long-term effects of hypoxia on the fetal cardiovascular system, and the importance of chronic fetal hypoxia in triggering a developmental origin of future heart disease in the adult progeny. The work summarizes evidence derived from human studies as well as from rodent, avian, and ovine models. There is a focus on the discovery of the molecular link between prenatal hypoxia, oxidative stress, and increased cardiovascular risk in adult offspring. Discussion of mitochondria-targeted antioxidant therapy offers potential targets for clinical intervention in human pregnancy complicated by chronic fetal hypoxia.
Collapse
Affiliation(s)
- Dino A Giussani
- Department of Physiology, Development, and Neuroscience; The Barcroft Centre; Cambridge Cardiovascular British Heart Foundation Centre for Research Excellence; and Cambridge Strategic Research Initiative in Reproduction, University of Cambridge, UK
| |
Collapse
|
40
|
Knutson AK, Williams AL, Boisvert WA, Shohet RV. HIF in the heart: development, metabolism, ischemia, and atherosclerosis. J Clin Invest 2021; 131:137557. [PMID: 34623330 DOI: 10.1172/jci137557] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The heart forms early in development and delivers oxygenated blood to the rest of the embryo. After birth, the heart requires kilograms of ATP each day to support contractility for the circulation. Cardiac metabolism is omnivorous, utilizing multiple substrates and metabolic pathways to produce this energy. Cardiac development, metabolic tuning, and the response to ischemia are all regulated in part by the hypoxia-inducible factors (HIFs), central components of essential signaling pathways that respond to hypoxia. Here we review the actions of HIF1, HIF2, and HIF3 in the heart, from their roles in development and metabolism to their activity in regeneration and preconditioning strategies. We also discuss recent work on the role of HIFs in atherosclerosis, the precipitating cause of myocardial ischemia and the leading cause of death in the developed world.
Collapse
|
41
|
Sutovska H, Molcan L, Koprdova R, Piesova M, Mach M, Zeman M. Prenatal hypoxia increases blood pressure in male rat offspring and affects their response to artificial light at night. J Dev Orig Health Dis 2021; 12:587-594. [PMID: 33109302 DOI: 10.1017/s2040174420000963] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Prenatal hypoxia (PH) has negative consequences on the cardiovascular system in adulthood and can affect the responses to additional insults later in life. We explored the effects of PH imposed during embryonic day 20 (10.5% O2 for 12 h) on circadian rhythms of systolic blood pressure (BP) and heart rate (HR) in mature male rat offspring measured by telemetry. We evaluated: (1) stability of BP and HR changes after PH; (2) circadian variability of BP and HR after 2 and 5 weeks of exposure to artificial light at night (ALAN; 1-2 lx); and (3) response of BP and HR to norepinephrine. PH increased BP in the dark (134 ± 2 mmHg vs. control 127 ± 2 mmHg; p = 0.05) and marginally in the light (125 ± 1 mmHg vs. control 120 ± 2 mmHg) phase of the day but not HR. The effect of PH was highly repeatable between 21- and 27-week-old PH male offspring. Two weeks of ALAN decreased the circadian variability of HR (p < 0.05) and BP more in control than PH rats. After 5 weeks of ALAN, the circadian variability of HR and BP were damped compared to LD and did not differ between control and PH rats (p < 0.05). Responses of BP and HR to norepinephrine did not differ between control and PH rats. Hypoxia at the end of the embryonic period increases BP and affects the functioning of the cardiovascular system in mature male offspring. ALAN in adulthood decreased the circadian variability of cardiovascular parameters, more in control than PH rats.
Collapse
Affiliation(s)
- Hana Sutovska
- Department of Animal Physiology and Ethology, Faculty of Natural Sciences, Comenius University, Bratislava, Slovakia
| | - Lubos Molcan
- Department of Animal Physiology and Ethology, Faculty of Natural Sciences, Comenius University, Bratislava, Slovakia
| | - Romana Koprdova
- Centre of Experimental Medicine SAS, Institute of Experimental Pharmacology and Toxicology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Michaela Piesova
- Centre of Experimental Medicine SAS, Institute of Experimental Pharmacology and Toxicology, Slovak Academy of Sciences, Bratislava, Slovakia
- Department of Pharmacology, Jessenius Faculty of Medicine, Comenius University, Martin, Slovakia
| | - Mojmír Mach
- Centre of Experimental Medicine SAS, Institute of Experimental Pharmacology and Toxicology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Michal Zeman
- Department of Animal Physiology and Ethology, Faculty of Natural Sciences, Comenius University, Bratislava, Slovakia
| |
Collapse
|
42
|
The newborn sheep translational model for pulmonary arterial hypertension of the neonate at high altitude. J Dev Orig Health Dis 2021; 11:452-463. [PMID: 32705972 DOI: 10.1017/s2040174420000616] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Chronic hypoxia during gestation induces greater occurrence of perinatal complications such as intrauterine growth restriction, fetal hypoxia, newborn asphyxia, and respiratory distress, among others. This condition may also cause a failure in the transition of the fetal to neonatal circulation, inducing pulmonary arterial hypertension of the neonate (PAHN), a syndrome that involves pulmonary vascular dysfunction, increased vasoconstrictor tone and pathological remodeling. As this syndrome has a relatively low prevalence in lowlands (~7 per 1000 live births) and very little is known about its prevalence and clinical evolution in highlands (above 2500 meters), our understanding is very limited. Therefore, studies on appropriate animal models have been crucial to comprehend the mechanisms underlying this pathology. Considering the strengths and weaknesses of any animal model of human disease is fundamental to achieve an effective and meaningful translation to clinical practice. The sheep model has been used to study the normal and abnormal cardiovascular development of the fetus and the neonate for almost a century. The aim of this review is to highlight the advances in our knowledge on the programming of cardiopulmonary function with the use of high-altitude newborn sheep as a translational model of PAHN.
Collapse
|
43
|
Prenatal hypoxia affects scaling of blood pressure and arterial wall mechanics in the common snapping turtle, Chelydra serpentina. Comp Biochem Physiol A Mol Integr Physiol 2021; 260:111023. [PMID: 34224856 DOI: 10.1016/j.cbpa.2021.111023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 06/30/2021] [Accepted: 06/30/2021] [Indexed: 11/24/2022]
Abstract
In reptiles, exposure to hypoxia during embryonic development affects several cardiovascular parameters. These modifications may impose different mechanical stress to the arterial system, and we speculated that the arterial wall of major outflow vessels would be modified accordingly. Since non-crocodilian reptiles possess a partially divided ventricle, ensuing similar systemic and pulmonary systolic pressures, we investigated how morphological and mechanical properties of segments from the left aortic arch (LAo) and the proximal and distal segments of the left pulmonary artery (LPAp and LPAd, respectively) change as body mass (Mb) increases. Eggs from common snapping turtles, Chelydra serpentina, were incubated under normoxia (21% O2; N21) or hypoxia (10% O2; H10), hatched and maintained in normoxia thereafter. Turtles (0.11-6.85 kg) were cannulated to measure arterial pressures, and an injection of adrenaline was used to increase pressures. Portions of the LAo, LPAp and LPAd were fixed under physiological hydrostatic pressures for histology and mechanical assessment. Arterial pressures increased with Mb for N21 but not for H10. Although mechanical and functional characteristics from the LPAp and LPAd were similar between N21 and H10, wall thickness from LAo did not change with Mb in the H10 group, thus wall stress increased in larger turtles. This indicates that larger H10 turtles probably experience an elevated probability of arterial wall rupture without concomitant changes in the cardiovascular system to prevent it. Finally, collagen content of the LPAp and LAo was smaller than in LPAd, suggesting a more distensible arterial wall could attenuate higher pressures from larger turtles.
Collapse
|
44
|
Camm EJ, Cross CM, Kane AD, Tarry-Adkins JL, Ozanne SE, Giussani DA. Maternal antioxidant treatment protects adult offspring against memory loss and hippocampal atrophy in a rodent model of developmental hypoxia. FASEB J 2021; 35:e21477. [PMID: 33891326 DOI: 10.1096/fj.202002557rr] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 02/11/2021] [Accepted: 02/11/2021] [Indexed: 02/02/2023]
Abstract
Chronic fetal hypoxia is one of the most common outcomes in complicated pregnancy in humans. Despite this, its effects on the long-term health of the brain in offspring are largely unknown. Here, we investigated in rats whether hypoxic pregnancy affects brain structure and function in the adult offspring and explored underlying mechanisms with maternal antioxidant intervention. Pregnant rats were randomly chosen for normoxic or hypoxic (13% oxygen) pregnancy with or without maternal supplementation with vitamin C in their drinking water. In one cohort, the placenta and fetal tissues were collected at the end of gestation. In another, dams were allowed to deliver naturally, and offspring were reared under normoxic conditions until 4 months of age (young adult). Between 3.5 and 4 months, the behavior, cognition and brains of the adult offspring were studied. We demonstrated that prenatal hypoxia reduced neuronal number, as well as vascular and synaptic density, in the hippocampus, significantly impairing memory function in the adult offspring. These adverse effects of prenatal hypoxia were independent of the hypoxic pregnancy inducing fetal growth restriction or elevations in maternal or fetal plasma glucocorticoid levels. Maternal vitamin C supplementation during hypoxic pregnancy protected against oxidative stress in the placenta and prevented the adverse effects of prenatal hypoxia on hippocampal atrophy and memory loss in the adult offspring. Therefore, these data provide a link between prenatal hypoxia, placental oxidative stress, and offspring brain health in later life, providing insight into mechanism and identifying a therapeutic strategy.
Collapse
Affiliation(s)
- Emily J Camm
- Department of Physiology, Development & Neuroscience, University of Cambridge, Cambridge, UK
| | - Christine M Cross
- Department of Physiology, Development & Neuroscience, University of Cambridge, Cambridge, UK
| | - Andrew D Kane
- Department of Physiology, Development & Neuroscience, University of Cambridge, Cambridge, UK
| | - Jane L Tarry-Adkins
- University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK
| | - Susan E Ozanne
- University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK.,Cambridge Strategic Initiative in Reproduction, Cambridge, UK
| | - Dino A Giussani
- Department of Physiology, Development & Neuroscience, University of Cambridge, Cambridge, UK.,Cambridge Strategic Initiative in Reproduction, Cambridge, UK
| |
Collapse
|
45
|
Spiroski AM, Niu Y, Nicholas LM, Austin-Williams S, Camm EJ, Sutherland MR, Ashmore TJ, Skeffington KL, Logan A, Ozanne SE, Murphy MP, Giussani DA. Mitochondria antioxidant protection against cardiovascular dysfunction programmed by early-onset gestational hypoxia. FASEB J 2021; 35:e21446. [PMID: 33788974 PMCID: PMC7612077 DOI: 10.1096/fj.202002705r] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 01/19/2021] [Accepted: 02/01/2021] [Indexed: 02/02/2023]
Abstract
Mitochondria-derived oxidative stress during fetal development increases cardiovascular risk in adult offspring of pregnancies complicated by chronic fetal hypoxia. We investigated the efficacy of the mitochondria-targeted antioxidant MitoQ in preventing cardiovascular dysfunction in adult rat offspring exposed to gestational hypoxia, integrating functional experiments in vivo, with those at the isolated organ and molecular levels. Rats were randomized to normoxic or hypoxic (13%-14% O2 ) pregnancy ± MitoQ (500 μM day-1 ) in the maternal drinking water. At 4 months of age, one cohort of male offspring was chronically instrumented with vascular catheters and flow probes to test in vivo cardiovascular function. In a second cohort, the heart was isolated and mounted onto a Langendorff preparation. To establish mechanisms linking gestational hypoxia with cardiovascular dysfunction and protection by MitoQ, we quantified the expression of antioxidant system, β-adrenergic signaling, and calcium handling genes in the fetus and adult, in frozen tissues from a third cohort. Maternal MitoQ in hypoxic pregnancy protected offspring against increased α1 -adrenergic reactivity of the cardiovascular system, enhanced reactive hyperemia in peripheral vascular beds, and sympathetic dominance, hypercontractility and diastolic dysfunction in the heart. Inhibition of Nfe2l2-mediated oxidative stress in the fetal heart and preservation of calcium regulatory responses in the hearts of fetal and adult offspring link molecular mechanisms to the protective actions of MitoQ treatment of hypoxic pregnancy. Therefore, these data show the efficacy of MitoQ in buffering mitochondrial stress through NADPH-induced oxidative damage and the prevention of programmed cardiovascular disease in adult offspring of hypoxic pregnancy.
Collapse
Affiliation(s)
- Ana-Mishel Spiroski
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK.,Cambridge Cardiovascular Strategic Research Initiative, Cambridge, UK
| | - Youguo Niu
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK.,Cambridge Cardiovascular Strategic Research Initiative, Cambridge, UK
| | - Lisa M Nicholas
- Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Shani Austin-Williams
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Emily J Camm
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Megan R Sutherland
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Thomas J Ashmore
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Katie L Skeffington
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Angela Logan
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK
| | - Susan E Ozanne
- Cambridge Cardiovascular Strategic Research Initiative, Cambridge, UK.,Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK.,Strategic Research Initiative in Reproduction, Cambridge, UK
| | - Michael P Murphy
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK.,Department of Medicine, University of Cambridge, Cambridge, UK
| | - Dino A Giussani
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK.,Cambridge Cardiovascular Strategic Research Initiative, Cambridge, UK.,Strategic Research Initiative in Reproduction, Cambridge, UK
| |
Collapse
|
46
|
Romanowicz J, Guerrelli D, Dhari Z, Mulvany C, Reilly M, Swift L, Vasandani N, Ramadan M, Leatherbury L, Ishibashi N, Posnack NG. Chronic perinatal hypoxia delays cardiac maturation in a mouse model for cyanotic congenital heart disease. Am J Physiol Heart Circ Physiol 2021; 320:H1873-H1886. [PMID: 33739154 DOI: 10.1152/ajpheart.00870.2020] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Compared with acyanotic congenital heart disease (CHD), cyanotic CHD has an increased risk of lifelong mortality and morbidity. These adverse outcomes may be attributed to delayed cardiomyocyte maturation, since the transition from a hypoxic fetal milieu to oxygen-rich postnatal environment is disrupted. We established a rodent model to replicate hypoxic myocardial conditions spanning perinatal development, and tested the hypothesis that chronic hypoxia impairs cardiac development. Pregnant mice were housed in hypoxia beginning at embryonic day 16. Pups stayed in hypoxia until postnatal day (P)8 when cardiac development is nearly complete. Global gene expression was quantified at P8 and at P30, after recovering in normoxia. Phenotypic testing included electrocardiogram, echocardiogram, and ex vivo electrophysiology study. Hypoxic P8 animals were 47% smaller than controls with preserved heart size. Gene expression was grossly altered by hypoxia at P8 (1,427 genes affected), but normalized after recovery (P30). Electrocardiograms revealed bradycardia and slowed conduction velocity in hypoxic animals at P8, with noticeable resolution after recovery (P30). Notable differences that persisted after recovery (P30) included a 65% prolongation in ventricular effective refractory period, sinus node dysfunction, 23% reduction in ejection fraction, and 16% reduction in fractional shortening in animals exposed to hypoxia. We investigated the impact of chronic hypoxia on the developing heart. Perinatal hypoxia was associated with changes in gene expression and cardiac function. Persistent changes to the electrophysiological substrate and contractile function warrant further investigation and may contribute to adverse outcomes observed in the cyanotic CHD population.NEW & NOTEWORTHY We utilized a new mouse model of chronic perinatal hypoxia to simulate the hypoxic myocardial conditions present in cyanotic congenital heart disease. Hypoxia caused numerous abnormalities in cardiomyocyte gene expression, the electrophysiologic substrate of the heart, and contractile function. Taken together, alterations observed in the neonatal period suggest delayed cardiac development immediately following hypoxia.
Collapse
Affiliation(s)
- Jennifer Romanowicz
- Children's National Heart Institute, Children's National Hospital, Washington, District of Columbia
| | - Devon Guerrelli
- Children's National Heart Institute, Children's National Hospital, Washington, District of Columbia.,Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's National Research Institute, Washington, District of Columbia.,Department of Biomedical Engineering, George Washington University, Washington, District of Columbia
| | - Zaenab Dhari
- Center for Neuroscience Research, Children's National Research Institute, Washington, District of Columbia
| | - Colm Mulvany
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's National Research Institute, Washington, District of Columbia
| | - Marissa Reilly
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's National Research Institute, Washington, District of Columbia
| | - Luther Swift
- Children's National Heart Institute, Children's National Hospital, Washington, District of Columbia.,Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's National Research Institute, Washington, District of Columbia
| | - Nimisha Vasandani
- Center for Neuroscience Research, Children's National Research Institute, Washington, District of Columbia
| | - Manelle Ramadan
- Children's National Heart Institute, Children's National Hospital, Washington, District of Columbia.,Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's National Research Institute, Washington, District of Columbia
| | - Linda Leatherbury
- Children's National Heart Institute, Children's National Hospital, Washington, District of Columbia
| | - Nobuyuki Ishibashi
- Children's National Heart Institute, Children's National Hospital, Washington, District of Columbia.,Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's National Research Institute, Washington, District of Columbia.,Center for Neuroscience Research, Children's National Research Institute, Washington, District of Columbia.,Department of Pediatrics, George Washington University, Washington, District of Columbia.,Department of Pharmacology & Physiology, George Washington University, Washington, District of Columbia
| | - Nikki Gillum Posnack
- Children's National Heart Institute, Children's National Hospital, Washington, District of Columbia.,Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's National Research Institute, Washington, District of Columbia.,Department of Pediatrics, George Washington University, Washington, District of Columbia.,Department of Pharmacology & Physiology, George Washington University, Washington, District of Columbia
| |
Collapse
|
47
|
Working towards precision medicine in developmental programming. Pediatr Res 2021; 89:1606-1607. [PMID: 33753895 PMCID: PMC8249231 DOI: 10.1038/s41390-021-01466-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 02/22/2021] [Indexed: 11/08/2022]
|
48
|
Hellgren KT, Premanandhan H, Quinn CJ, Trafford AW, Galli GLJ. Sex-dependent effects of developmental hypoxia on cardiac mitochondria from adult murine offspring. Free Radic Biol Med 2021; 162:490-499. [PMID: 33186741 DOI: 10.1016/j.freeradbiomed.2020.11.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 10/29/2020] [Accepted: 11/04/2020] [Indexed: 12/12/2022]
Abstract
Insufficient oxygen supply (hypoxia) during fetal and embryonic development can lead to latent phenotypical changes in the adult cardiovascular system, including altered cardiac function and increased susceptibility to ischemia reperfusion injury. While the cellular mechanisms underlying this phenomenon are largely unknown, several studies have pointed towards metabolic disturbances in the heart of offspring from hypoxic pregnancies. To this end, we investigated mitochondrial function in the offspring of a mouse model of prenatal hypoxia. Pregnant C57 mice were subjected to either normoxia (21%) or hypoxia (14%) during gestational days 6-18. Offspring were reared in normoxia for up to 8 months and mitochondrial biology was assessed with electron microscopy (ultrastructure), spectrophotometry (enzymatic activity of electron transport chain complexes), microrespirometry (oxidative phosphorylation and H202 production) and Western Blot (protein expression). Our data showed that male adult offspring from hypoxic pregnancies possessed mitochondria with increased H202 production and lower respiratory capacity that was associated with reduced protein expression of complex I, II and IV. In contrast, females from hypoxic pregnancies had a higher respiratory capacity and lower H202 production that was associated with increased enzymatic activity of complex IV. From these results, we speculate that early exposure to hypoxia has long term, sex-dependent effects on cardiac metabolic function, which may have implications for cardiovascular health and disease in adulthood.
Collapse
Affiliation(s)
- Kim T Hellgren
- Division of Cardiovascular Sciences, School of Medical Sciences, University of Manchester, Manchester M13 9NT, UK
| | - Hajani Premanandhan
- Division of Cardiovascular Sciences, School of Medical Sciences, University of Manchester, Manchester M13 9NT, UK
| | - Callum J Quinn
- Division of Cardiovascular Sciences, School of Medical Sciences, University of Manchester, Manchester M13 9NT, UK
| | - Andrew W Trafford
- Division of Cardiovascular Sciences, School of Medical Sciences, University of Manchester, Manchester M13 9NT, UK
| | - Gina L J Galli
- Division of Cardiovascular Sciences, School of Medical Sciences, University of Manchester, Manchester M13 9NT, UK.
| |
Collapse
|
49
|
Martín-Estal I, Castilla-Cortázar I, Castorena-Torres F. The Placenta as a Target for Alcohol During Pregnancy: The Close Relation with IGFs Signaling Pathway. Rev Physiol Biochem Pharmacol 2021; 180:119-153. [PMID: 34159446 DOI: 10.1007/112_2021_58] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Alcohol is one of the most consumed drugs in the world, even during pregnancy. Its use is a risk factor for developing adverse outcomes, e.g. fetal death, miscarriage, fetal growth restriction, and premature birth, also resulting in fetal alcohol spectrum disorders. Ethanol metabolism induces an oxidative environment that promotes the oxidation of lipids and proteins, triggers DNA damage, and advocates mitochondrial dysfunction, all of them leading to apoptosis and cellular injury. Several organs are altered due to this harmful behavior, the brain being one of the most affected. Throughout pregnancy, the human placenta is one of the most important organs for women's health and fetal development, as it secretes numerous hormones necessary for a suitable intrauterine environment. However, our understanding of the human placenta is very limited and even more restricted is the knowledge of the impact of toxic substances in its development and fetal growth. So, could ethanol consumption during this period have wounding effects in the placenta, compromising proper fetal organ development? Several studies have demonstrated that alcohol impairs various signaling cascades within G protein-coupled receptors and tyrosine kinase receptors, mainly through its action on insulin and insulin-like growth factor 1 (IGF-1) signaling pathway. This last cascade is involved in cell proliferation, migration, and differentiation and in placentation. This review tries to examine the current knowledge and gaps in our existing understanding of the ethanol effects in insulin/IGFs signaling pathway, which can explain the mechanism to elucidate the adverse actions of ethanol in the maternal-fetal interface of mammals.
Collapse
Affiliation(s)
- Irene Martín-Estal
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey, NL, Mexico
| | | | | |
Collapse
|
50
|
The path ahead: From global pandemic to health promotion. Prev Med Rep 2020; 21:101271. [PMID: 33364151 PMCID: PMC7750161 DOI: 10.1016/j.pmedr.2020.101271] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 11/17/2020] [Accepted: 11/19/2020] [Indexed: 02/06/2023] Open
Abstract
The current COVID-19 pandemic represents an acute threat to the health of adults and children across the globe. In addition, it has the potential to worsen the health of future generations through intergenerational health effects. Examples from history, including the Dutch famine (Hongerwinter), suggest that in utero and early life environments may have significant implications for health outcomes throughout the lifespan and are important in determining risk of chronic disease in adulthood. Parental health status, stress, and nutrition appear to affect offspring health and are all affected by the COVID-19 pandemic. Thus, it is critical that we consider the potential impacts of the current pandemic on pregnant women, infants and children and take public health and medical actions to mitigate risk and promote health in future generations.
Collapse
|