1
|
Sedmera D, Drobna Krejci E, Nanka O, Eckhardt A. Proteomic analysis of chick embryonic heart in experimental hypoxia. Dev Biol 2025; 521:28-36. [PMID: 39933632 DOI: 10.1016/j.ydbio.2025.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 01/08/2025] [Accepted: 02/08/2025] [Indexed: 02/13/2025]
Abstract
Investigating prenatal hypoxia is difficult in mammals, as there are confounding factors stemming from maternal adaptations and compensatory mechanisms. We have thus established an avian model of hypoxic incubation (starting after 2 days of normoxia, 15% O2, normobaric, until the time of sampling at embryonic day 8) to study embryonic reactions to low oxygen concentration. Our previous studies have shown increased vascularization, oedema, and ventricular wall thinning preceding the lethality at mid-gestation. Analysis of the cardiac proteome after 6 days of hypoxic incubation showed strong upregulation of enzymes involved in anaerobic glycolysis as well as an increase in apoptosis-related proteins, cell adhesion proteins, and secretory activity.
Collapse
Affiliation(s)
- David Sedmera
- Institute of Anatomy, First Faculty of Medicine, Charles University, U Nemocnice 3, 128 00, Prague 2, Czech Republic; Institute of Physiology, The Czech Academy of Sciences, Videnska 1024, 142 00, Prague 4, Czech Republic.
| | - Eliska Drobna Krejci
- Institute of Anatomy, First Faculty of Medicine, Charles University, U Nemocnice 3, 128 00, Prague 2, Czech Republic
| | - Ondrej Nanka
- Institute of Anatomy, First Faculty of Medicine, Charles University, U Nemocnice 3, 128 00, Prague 2, Czech Republic
| | - Adam Eckhardt
- Institute of Physiology, The Czech Academy of Sciences, Videnska 1024, 142 00, Prague 4, Czech Republic
| |
Collapse
|
2
|
Sierla JR, Pagerols Raluy L, Trochimiuk M, Trah J, Petrosyan M, Velasquez LN, Schumacher U, Singer D, Heiter J. Disparity Between Functional and Structural Recovery of Placental Mitochondria After Exposure to Hypoxia. Int J Mol Sci 2025; 26:2956. [PMID: 40243595 PMCID: PMC11988352 DOI: 10.3390/ijms26072956] [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: 02/05/2025] [Revised: 03/07/2025] [Accepted: 03/09/2025] [Indexed: 04/18/2025] Open
Abstract
Intrauterine growth restriction (IUGR) affects 5-10% of pregnancies with placental hypoxia, playing a key role as a common pathophysiological pathway of different etiologies. Despite the high metabolic rate of the placenta and its "gatekeeper" role in protecting the fetus from hypoxia, the response of placental mitochondria to hypoxic stress is not well understood. This study tested the hypothesis that transient exposure to hypoxia leads to a loss of placental mitochondria and affects their function. Human villous trophoblastic (JEG-3) cells were cultured under normoxic and hypoxic conditions for 24 h. Mitochondrial content was determined by flow cytometry before and after hypoxic exposure and after 24 h of normoxic recovery. Parameters of oxidative phosphorylation were assessed using a respirometric analyzer before hypoxic exposure and after normoxic recovery. Mitochondrial content decreased significantly from 88.5% to 26.7% during hypoxic incubation. Although it had increased to 84.2% after 24 h of normoxic recovery, oxidative phosphorylation parameters were still significantly suppressed to 1/2 to 1/3 of the pre-incubation levels. The results underscore the ability of placental cells to adapt mitochondrial content to O2 supply. Despite rapid recovery under normoxia, respiratory function remains suppressed, which may result in persistent impairment of adenosine triphosphate (ATP)-dependent synthetic and transport functions.
Collapse
Affiliation(s)
- Jonathan R. Sierla
- Division of Neonatology and Pediatric Critical Care Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (J.R.S.)
| | - Laia Pagerols Raluy
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Magdalena Trochimiuk
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Julian Trah
- Division of Neonatology and Pediatric Critical Care Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (J.R.S.)
| | - Mariam Petrosyan
- Institute of Anatomy and Experimental Morphology, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Lis N. Velasquez
- Division of Molecular Immunology und Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Udo Schumacher
- Institute of Anatomy and Experimental Morphology, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Dominique Singer
- Division of Neonatology and Pediatric Critical Care Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (J.R.S.)
| | - Julia Heiter
- Division of Neonatology and Pediatric Critical Care Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (J.R.S.)
| |
Collapse
|
3
|
Carver AJ, Dunnwald M, Stevens HE. A head start: The relationship of placental factors to craniofacial and brain development. Dev Dyn 2025. [PMID: 40105397 DOI: 10.1002/dvdy.70018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 02/17/2025] [Accepted: 03/02/2025] [Indexed: 03/20/2025] Open
Abstract
In recent years, the importance of placental function for fetal neurodevelopment has become increasingly studied. This field, known as neuroplacentology, has greatly expanded possible etiologies of neurodevelopmental disorders by exploring the influence of placental function on brain development. It is also well-established that brain development is influenced by craniofacial morphogenesis. However, there is less focus on the impact of the placenta on craniofacial development. Recent research suggests the functional influence of placental nutrients and hormones on craniofacial skeletal growth, such as prolactin, growth hormone, insulin-like growth factor 1, vitamin D, sulfate, and calcium, impacting both craniofacial and brain development. Therefore, interactions between the placenta and both fetal neurodevelopment and craniofacial development likely influence the growth and morphology of the head as a whole. This review discusses the role of placental hormone production and nutrient delivery in the development of the fetal head-defined as craniofacial and brain tissue together-expanding on the more established focus on brain development to also include the skull (or cranium) and face.
Collapse
Affiliation(s)
- Annemarie Jenna Carver
- Interdisciplinary Graduate Program in Genetics, University of Iowa, Iowa City, Iowa, USA
- Psychiatry Department, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Martine Dunnwald
- Interdisciplinary Graduate Program in Genetics, University of Iowa, Iowa City, Iowa, USA
- Department of Anatomy and Cell Biology, Carver College of Medicine, Iowa City, Iowa, USA
| | - Hanna Elizabeth Stevens
- Interdisciplinary Graduate Program in Genetics, University of Iowa, Iowa City, Iowa, USA
- Psychiatry Department, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
- Hawk Intellectual and Developmental Disabilities Research Center, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
4
|
Ahrens S, Singer D. Placental Adaptation to Hypoxia: The Case of High-Altitude Pregnancies. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2025; 22:214. [PMID: 40003440 PMCID: PMC11855801 DOI: 10.3390/ijerph22020214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 01/22/2025] [Accepted: 01/28/2025] [Indexed: 02/27/2025]
Abstract
Even in the highest inhabited regions of the world, well above 2500 m altitude, women become pregnant and give birth to healthy children. The underlying adaptation to hypobaric hypoxia provides interesting insights into the physio(patho)logy of the human placenta. Although increasing altitude is regularly associated with fetal growth restriction (FGR), oxygen deficiency does not appear to be a direct cause. Rather, placental oxygen consumption is reduced to maintain the oxygen supply to the fetus. This comes at the expense of placental synthesis and transport functions, resulting in inappropriate nutrient supply. The hypoxia-inducible factor (HIF-1α), which modulates the mitochondrial electron transport chain to protect placental tissue from reactive oxygen species, plays a key role here. Reduced oxygen consumption also reflects decreased placental vascularization and perfusion, which is accompanied by an increased risk of maternal pre-eclampsia at high altitude. In native highlanders, the latter seems to be attenuated, partly due to a lower release of HIF-1α. In addition, metabolic peculiarities have been described in indigenous people that enhance glucose availability and thus reduce the extent of FGR. This review attempts to revisit the (albeit incomplete) knowledge in this area to draw the clinical reader's attention to the crucial role of the placenta in defending the fetus against hypoxia.
Collapse
Affiliation(s)
- Sofia Ahrens
- Department of Pediatric Surgery, Altona Children’s Hospital, University Medical Center Eppendorf (UKE), 20251 Hamburg, Germany;
- Division of Neonatology and Pediatric Critical Care Medicine, University Medical Center Eppendorf (UKE), 20251 Hamburg, Germany
| | - Dominique Singer
- Division of Neonatology and Pediatric Critical Care Medicine, University Medical Center Eppendorf (UKE), 20251 Hamburg, Germany
| |
Collapse
|
5
|
Tong W, Allison BJ, Brain KL, Patey OV, Niu Y, Botting KJ, Ford SG, Garrud TA, Wooding PFB, Lyu Q, Zhang L, Ma J, Sowton AP, O'Brien KA, Cindrova-Davies T, Yung HW, Burton GJ, Murray AJ, Giussani DA. Placental mitochondrial metabolic adaptation maintains cellular energy balance in pregnancy complicated by gestational hypoxia. J Physiol 2025. [PMID: 39868991 DOI: 10.1113/jp287897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 12/18/2024] [Indexed: 01/28/2025] Open
Abstract
The mechanisms that drive placental dysfunction in pregnancies complicated by hypoxia and fetal growth restriction remain poorly understood. Changes to mitochondrial respiration contribute to cellular dysfunction in conditions of hypoxia and have been implicated in the pathoaetiology of pregnancy complications, such as pre-eclampsia. We used bespoke isobaric hypoxic chambers and a combination of functional, molecular and imaging techniques to study cellular metabolism and mitochondrial dynamics in sheep undergoing hypoxic pregnancy. We show that hypoxic pregnancy in sheep triggers a shift in capacity away from β-oxidation and complex I-mediated respiration, while maintaining total oxidative phosphorylation capacity. There are also complex-specific changes to electron transport chain composition and a switch in mitochondrial dynamics towards fission. Hypoxic placentas show increased activation of the non-canonical mitochondrial unfolded protein response pathway and enhanced insulin like growth factor 2 signalling. Combined, therefore, the data show that the hypoxic placenta undergoes significant metabolic and morphological adaptations to maintain cellular energy balance. Chronic hypoxia during pregnancy in sheep activated placental mitochondrial stress pathways, leading to alterations in mitochondrial respiration, mitochondrial energy metabolism and mitochondrial dynamics, as seen in the placenta of women with pre-eclampsia. KEY POINTS: Hypoxia shifts mitochondrial respiration away from β-oxidation and complex I. Complex-specific changes occur in the electron transport chain composition. Activation of the non-canonical mitochondrial unfolded protein response pathway is heightened in hypoxic placentas. Enhanced insulin like growth factor 2 signalling is observed in hypoxic placentas. Hypoxic placentas undergo significant functional adaptations for energy balance.
Collapse
Affiliation(s)
- Wen Tong
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
- Strategic Research Initiative in Reproduction, University of Cambridge, Cambridge, UK
| | - Beth J Allison
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Kirsty L Brain
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Olga V Patey
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Youguo Niu
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
- Strategic Research Initiative in Reproduction, University of Cambridge, Cambridge, UK
- Cardiovascular Strategic Research Initiative, University of Cambridge, Cambridge, UK
- Department of Aerospace Physiology, Fourth Military Medical University, Xi'an, China
| | - Kimberley J Botting
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
- Strategic Research Initiative in Reproduction, University of Cambridge, Cambridge, UK
- Cardiovascular Strategic Research Initiative, University of Cambridge, Cambridge, UK
| | - Sage G Ford
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Tess A Garrud
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
| | - Peter F B Wooding
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
| | - Qiang Lyu
- Department of Aerospace Physiology, Fourth Military Medical University, Xi'an, China
| | - Lin Zhang
- Department of Aerospace Physiology, Fourth Military Medical University, Xi'an, China
| | - Jin Ma
- Department of Aerospace Physiology, Fourth Military Medical University, Xi'an, China
| | - Alice P Sowton
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
| | - Katie A O'Brien
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
| | - Tereza Cindrova-Davies
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
- Strategic Research Initiative in Reproduction, University of Cambridge, Cambridge, UK
| | - Hong Wa Yung
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
- Strategic Research Initiative in Reproduction, University of Cambridge, Cambridge, UK
| | - Graham J Burton
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
- Strategic Research Initiative in Reproduction, University of Cambridge, Cambridge, UK
| | - Andrew J Murray
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
- Cardiovascular Strategic Research Initiative, University of Cambridge, Cambridge, UK
| | - Dino A Giussani
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
- Strategic Research Initiative in Reproduction, University of Cambridge, Cambridge, UK
- Cardiovascular Strategic Research Initiative, University of Cambridge, Cambridge, UK
- Department of Aerospace Physiology, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
6
|
Eiríksdóttir DH, Hvass GG, Zimmermann H, Jan Struijk J, Schmidt SE. Noise reduction in abdominal acoustic recordings of maternal placental murmurs. Biomed Phys Eng Express 2025; 11:025005. [PMID: 39773991 DOI: 10.1088/2057-1976/ada6bb] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 01/07/2025] [Indexed: 01/11/2025]
Abstract
Fetal phonocardiography is a well-known auscultation technique for evaluation of fetal health. However, murmurs that are synchronous with the maternal heartbeat can often be heard while listening to fetal heart sounds. Maternal placental murmurs (MPM) could be used to detect maternal cardiovascular and placental abnormalities, but the recorded MPMs are often contaminated by ambient interference and noise.Objective:The aim of this study was to compare noise reduction methods to reduce noise in the recorded MPMs. Approach:1) Bandpass filtering (BPF), 2) a multichannel noise reduction (MCh) using either Wiener filter (WF), Least-mean-square or Independent component analysis, 3) a combination of BPF with wavelet transient reduction (WTR) and 4) a combination of MCh and WTR. The methods were tested on signals recorded with two microphone units placed on the abdomen of pregnant women with an electrocardiogram (ECG) recorded simultaneously. The performance was evaluated using coherence and heart cycle duration error (HCDError) as compared with the ECG. Results: The mean of the absolute HCDErrorwas 32.7 ms for the BPF with all methods significantly lower (p < 0.05) than BPF. The lowest errors were obtained for WTR-WF where the HCDErrorranged 16.68-17.72 ms for seven different filter orders. All methods had significantly different coherence measure compared with BPF (p < 0.05). The lowest coherence was reached with WTR-WF (filter order 640) where the mean value decreased from 0.50 for BPF to 0.03.Significance:These results show how noise reduction techniques such as WF combined with wavelet denoising can greatly enhance the quality of MPM recordings.
Collapse
Affiliation(s)
| | - Gry Grønborg Hvass
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Henrik Zimmermann
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Johannes Jan Struijk
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Samuel Emil Schmidt
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| |
Collapse
|
7
|
Hu Y, Lin Y, Yang J, Wang S, Gao L, Bi Y, Wang Y. Mitochondrial dysfunction and oxidative stress in selective fetal growth restriction. Placenta 2024; 156:46-54. [PMID: 39265375 DOI: 10.1016/j.placenta.2024.09.005] [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: 05/29/2024] [Revised: 09/02/2024] [Accepted: 09/09/2024] [Indexed: 09/14/2024]
Abstract
INTRODUCTION Placental dysfunction is the primary cause of selective fetal growth restriction (sFGR), and the specific role of mitochondria remains unclear. This study aims to elucidate mitochondrial functional defects in sFGR placentas and explore the roles of mitochondrial genomic and epigenetic alterations in its pathogenesis. METHODS The placental villi of MCDA twins with sFGR were collected and the morphology and number of mitochondria were observed by transmission electron microscopy. Meanwhile, the levels of reactive oxygen species (ROS), ATP and oxidative damage markers were assessed. Mitochondrial DNA (mtDNA) copy number detection, targeted sequencing and methylation sequencing were performed. The expression of placental cytochrome c oxidase subunit I (COX I) and mitochondrial long non-coding RNAs (lncRNAs) were evaluated by Western blotting and qPCR. RESULTS Compared with placentae from normal fetuses, pronounced mitochondrial damage within cytotrophoblast was revealed in sFGR placentae, alongside augmented mitochondrial number in syncytiotrophoblast. Enhanced oxidative stress in these placentae was evidenced by elevated markers of oxidative damage, accompanied by increased ROS production and diminished ATP generation. In sFGR placentae, a notable rise in mitochondrial copy number and one heterozygous mutation in the MT-RNR2 gene were observed, along with decreased COX Ⅰ levels, increased lncND5, lncND6, lncCyt b, and MDL1 synthesis, and decreased RMRP synthesis. DISCUSSION Findings collectively confirmed an exacerbation of oxidative stress within sFGR placentae, coinciding with mitochondrial dysfunction, compromised energy production, and ultimately the failure of compensatory mechanisms to restore energy balance, which may result from mutations in the mitochondrial genome and abnormal expression of epigenetic regulatory genes.
Collapse
Affiliation(s)
- Yucheng Hu
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China; Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, 200030, China
| | - Yuhong Lin
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China; Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, 200030, China
| | - Jiawen Yang
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China; Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, 200030, China
| | - Shan Wang
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China; Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, 200030, China
| | - Li Gao
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China; Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, 200030, China
| | - Yan Bi
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China; Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, 200030, China.
| | - Yanlin Wang
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China; Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, 200030, China.
| |
Collapse
|
8
|
Bezemer RE, Faas MM, van Goor H, Gordijn SJ, Prins JR. Decidual macrophages and Hofbauer cells in fetal growth restriction. Front Immunol 2024; 15:1379537. [PMID: 39007150 PMCID: PMC11239338 DOI: 10.3389/fimmu.2024.1379537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 06/14/2024] [Indexed: 07/16/2024] Open
Abstract
Placental macrophages, which include maternal decidual macrophages and fetal Hofbauer cells, display a high degree of phenotypical and functional plasticity. This provides these macrophages with a key role in immunologically driven events in pregnancy like host defense, establishing and maintaining maternal-fetal tolerance. Moreover, placental macrophages have an important role in placental development, including implantation of the conceptus and remodeling of the intrauterine vasculature. To facilitate these processes, it is crucial that placental macrophages adapt accordingly to the needs of each phase of pregnancy. Dysregulated functionalities of placental macrophages are related to placental malfunctioning and have been associated with several adverse pregnancy outcomes. Although fetal growth restriction is specifically associated with placental insufficiency, knowledge on the role of macrophages in fetal growth restriction remains limited. This review provides an overview of the distinct functionalities of decidual macrophages and Hofbauer cells in each trimester of a healthy pregnancy and aims to elucidate the mechanisms by which placental macrophages could be involved in the pathogenesis of fetal growth restriction. Additionally, potential immune targeted therapies for fetal growth restriction are discussed.
Collapse
Affiliation(s)
- Romy Elisa Bezemer
- Department of Obstetrics and Gynecology, University Medical Center Groningen, Groningen, Netherlands
- Department of Pathology and Medical Biology, University Medical Center Groningen, Groningen, Netherlands
| | - Marijke M Faas
- Department of Obstetrics and Gynecology, University Medical Center Groningen, Groningen, Netherlands
- Department of Pathology and Medical Biology, University Medical Center Groningen, Groningen, Netherlands
| | - Harry van Goor
- Department of Pathology and Medical Biology, University Medical Center Groningen, Groningen, Netherlands
| | - Sanne Jehanne Gordijn
- Department of Obstetrics and Gynecology, University Medical Center Groningen, Groningen, Netherlands
| | - Jelmer R Prins
- Department of Obstetrics and Gynecology, University Medical Center Groningen, Groningen, Netherlands
| |
Collapse
|
9
|
Shabanov P, Samorodov A, Urakova N, Fisher E, Shchemeleva A. Low Fetal Resistance to Hypoxia as a Cause of Stillbirth and Neonatal Encephalopathy. CLIN EXP OBSTET GYN 2024; 51. [DOI: 10.31083/j.ceog5102033] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2025]
Abstract
Objective: Low fetal resistance to hypoxia is a factor in stillbirth and neonatal encephalopathy. This review examines fetal movement patterns in response to hypoxia as a predictor of the likelihood of stillbirth. Monitoring the dynamics of fetal movements during maternal apnea could allow the assessment of fetal resistance to hypoxia. The goal of this study is to describe the practical application of this method by doctors and pregnant women. Mechanism: We searched relevant keywords in the international scientific literature databases Scopus and Web of Science, as well as databases for patents granted in China, India, USA, Japan, Germany, Russia and other countries. Devices, drugs and medical technologies that provide diagnosis, modeling, prevention and treatment of intrauterine fetal hypoxia, stillbirth and neonatal encephalopathy were considered. Findings in Brief: During apnea by a pregnant woman in the second half of normal pregnancy, if the maximum duration of fetal immobility exceeds 30 seconds from the onset of breath-holding, then the fetus is considered to show good resistance to hypoxia, thus preserving its health and life during vaginal delivery. On the other hand, excessive fetal movements <10 seconds after the onset of apnea in a pregnant woman indicates low fetal resistance to hypoxia. When fetal resistance to hypoxia is low, there is no alternative to immediate cesarean section for the preservation of fetal life and health. Conclusions: The monitoring of fetal movements during apnea in pregnant women allows real-time assessment of fetal resistance to intrauterine hypoxia. Obtaining timely information on fetal resistance to hypoxia is critical for determining the optimal timing and type of delivery in order to prevent encephalopathy and stillbirth.
Collapse
Affiliation(s)
- Petr Shabanov
- Department of Psychopharmacology, Institute of Experimental Medicine, 197376 Saint Petersburg, Russia
| | - Aleksandr Samorodov
- Department of Pharmacology with a Course of Сlinical Pharmacology, Bashkir State Medical University, 450008 Ufa, Bashkortostan, Russia
| | - Natalya Urakova
- Department of Obstetrics and Gynecology, Izhevsk State Medical Academy, 426034 Izhevsk, Udmurt Republic, Russia
- Department of Experimental and Clinical Research and Inventive, Institute of Thermology, 426054 Izhevsk, Udmurt Republic, Russia
| | - Evgeny Fisher
- Department of Experimental and Clinical Research and Inventive, Institute of Thermology, 426054 Izhevsk, Udmurt Republic, Russia
| | - Albina Shchemeleva
- Department of Experimental and Clinical Research and Inventive, Institute of Thermology, 426054 Izhevsk, Udmurt Republic, Russia
| |
Collapse
|
10
|
O'Brien KA, Gu W, Houck JA, Holzner LMW, Yung HW, Armstrong JL, Sowton AP, Baxter R, Darwin PM, Toledo-Jaldin L, Lazo-Vega L, Moreno-Aramayo AE, Miranda-Garrido V, Shortt JA, Matarazzo CJ, Yasini H, Burton GJ, Moore LG, Simonson TS, Murray AJ, Julian CG. Genomic Selection Signals in Andean Highlanders Reveal Adaptive Placental Metabolic Phenotypes That Are Disrupted in Preeclampsia. Hypertension 2024; 81:319-329. [PMID: 38018457 PMCID: PMC10841680 DOI: 10.1161/hypertensionaha.123.21748] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 10/24/2023] [Indexed: 11/30/2023]
Abstract
BACKGROUND The chronic hypoxia of high-altitude residence poses challenges for tissue oxygen supply and metabolism. Exposure to high altitude during pregnancy increases the incidence of hypertensive disorders of pregnancy and fetal growth restriction and alters placental metabolism. High-altitude ancestry protects against altitude-associated fetal growth restriction, indicating hypoxia tolerance that is genetic in nature. Yet, not all babies are protected and placental pathologies associated with fetal growth restriction occur in some Andean highlanders. METHODS We examined placental metabolic function in 79 Andeans (18-45 years; 39 preeclamptic and 40 normotensive) living in La Paz, Bolivia (3600-4100 m) delivered by unlabored Cesarean section. Using a selection-nominated approach, we examined links between putatively adaptive genetic variation and phenotypes related to oxygen delivery or placental metabolism. RESULTS Mitochondrial oxidative capacity was associated with fetal oxygen delivery in normotensive but not preeclamptic placenta and was also suppressed in term preeclamptic pregnancy. Maternal haplotypes in or within 200 kb of selection-nominated genes were associated with lower placental mitochondrial respiratory capacity (PTPRD [protein tyrosine phosphatase receptor-δ]), lower maternal plasma erythropoietin (CPT2 [carnitine palmitoyl transferase 2], proopiomelanocortin, and DNMT3 [DNA methyltransferase 3]), and lower VEGF (vascular endothelial growth factor) in umbilical venous plasma (TBX5 [T-box transcription factor 5]). A fetal haplotype within 200 kb of CPT2 was associated with increased placental mitochondrial complex II capacity, placental nitrotyrosine, and GLUT4 (glucose transporter type 4) protein expression. CONCLUSIONS Our findings reveal novel associations between putatively adaptive gene regions and phenotypes linked to oxygen delivery and placental metabolic function in highland Andeans, suggesting that such effects may be of genetic origin. Our findings also demonstrate maladaptive metabolic mechanisms in the context of preeclampsia, including dysregulation of placental oxygen consumption.
Collapse
Affiliation(s)
- Katie A O'Brien
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom (K.A.O., L.M.W.H., H.W.Y., J.L.A., A.P.S., R.B., P.M.D., G.J.B., A.J.M.)
- Department of Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine (K.A.O., W.G., T.S.S.), University of California San Diego, La Jolla, CA
- Department of Biomedical Informatics (K.A.O., J.A.H., J.A.S., C.J.M., H.Y., C.G.J.), University of Colorado School of Medicine, Aurora, CO
| | - Wanjun Gu
- Department of Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine (K.A.O., W.G., T.S.S.), University of California San Diego, La Jolla, CA
- Herbert Wertheim School of Public Health and Longevity Sciences (W.G.), University of California San Diego, La Jolla, CA
| | - Julie A Houck
- Department of Biomedical Informatics (K.A.O., J.A.H., J.A.S., C.J.M., H.Y., C.G.J.), University of Colorado School of Medicine, Aurora, CO
- Department of Obstetrics and Gynecology, Division of Reproductive Sciences (J.A.H., L.G.M.), University of Colorado School of Medicine, Aurora, CO
| | - Lorenz M W Holzner
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom (K.A.O., L.M.W.H., H.W.Y., J.L.A., A.P.S., R.B., P.M.D., G.J.B., A.J.M.)
| | - Hong Wa Yung
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom (K.A.O., L.M.W.H., H.W.Y., J.L.A., A.P.S., R.B., P.M.D., G.J.B., A.J.M.)
| | - Jenna L Armstrong
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom (K.A.O., L.M.W.H., H.W.Y., J.L.A., A.P.S., R.B., P.M.D., G.J.B., A.J.M.)
| | - Alice P Sowton
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom (K.A.O., L.M.W.H., H.W.Y., J.L.A., A.P.S., R.B., P.M.D., G.J.B., A.J.M.)
| | - Ruby Baxter
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom (K.A.O., L.M.W.H., H.W.Y., J.L.A., A.P.S., R.B., P.M.D., G.J.B., A.J.M.)
| | - Paula M Darwin
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom (K.A.O., L.M.W.H., H.W.Y., J.L.A., A.P.S., R.B., P.M.D., G.J.B., A.J.M.)
| | - Lilian Toledo-Jaldin
- Department of Obstetrics, Hospital Materno-Infantil, La Paz, Bolivia (L.T.-J., L.L.-V., A.E.M.-M., V.M.-G.)
| | - Litzi Lazo-Vega
- Department of Obstetrics, Hospital Materno-Infantil, La Paz, Bolivia (L.T.-J., L.L.-V., A.E.M.-M., V.M.-G.)
| | - Any Elena Moreno-Aramayo
- Department of Obstetrics, Hospital Materno-Infantil, La Paz, Bolivia (L.T.-J., L.L.-V., A.E.M.-M., V.M.-G.)
| | - Valquiria Miranda-Garrido
- Department of Obstetrics, Hospital Materno-Infantil, La Paz, Bolivia (L.T.-J., L.L.-V., A.E.M.-M., V.M.-G.)
| | - Jonathan A Shortt
- Department of Biomedical Informatics (K.A.O., J.A.H., J.A.S., C.J.M., H.Y., C.G.J.), University of Colorado School of Medicine, Aurora, CO
| | - Christopher J Matarazzo
- Department of Biomedical Informatics (K.A.O., J.A.H., J.A.S., C.J.M., H.Y., C.G.J.), University of Colorado School of Medicine, Aurora, CO
| | - Hussna Yasini
- Department of Biomedical Informatics (K.A.O., J.A.H., J.A.S., C.J.M., H.Y., C.G.J.), University of Colorado School of Medicine, Aurora, CO
| | - Graham J Burton
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom (K.A.O., L.M.W.H., H.W.Y., J.L.A., A.P.S., R.B., P.M.D., G.J.B., A.J.M.)
| | - Lorna G Moore
- Department of Obstetrics and Gynecology, Division of Reproductive Sciences (J.A.H., L.G.M.), University of Colorado School of Medicine, Aurora, CO
| | - Tatum S Simonson
- Department of Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine (K.A.O., W.G., T.S.S.), University of California San Diego, La Jolla, CA
| | - Andrew J Murray
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom (K.A.O., L.M.W.H., H.W.Y., J.L.A., A.P.S., R.B., P.M.D., G.J.B., A.J.M.)
| | - Colleen G Julian
- Department of Biomedical Informatics (K.A.O., J.A.H., J.A.S., C.J.M., H.Y., C.G.J.), University of Colorado School of Medicine, Aurora, CO
| |
Collapse
|
11
|
Gou R, Zhang X. Glycolysis: A fork in the path of normal and pathological pregnancy. FASEB J 2023; 37:e23263. [PMID: 37889786 DOI: 10.1096/fj.202301230r] [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: 06/19/2023] [Revised: 09/17/2023] [Accepted: 10/04/2023] [Indexed: 10/29/2023]
Abstract
Glucose metabolism is vital to the survival of living organisms. Since the discovery of the Warburg effect in the 1920s, glycolysis has become a major research area in the field of metabolism. Glycolysis has been extensively studied in the field of cancer and is considered as a promising therapeutic target. However, research on the role of glycolysis in pregnancy is limited. Recent evidence suggests that blastocysts, trophoblasts, decidua, and tumors all acquire metabolic energy at specific stages in a highly similar manner. Glycolysis, carefully controlled throughout pregnancy, maintains a dynamic and coordinated state, so as to maintain the homeostasis of the maternal-fetal interface and ensure normal gestation. In the present review, we investigate metabolic remodeling and the selective propensity of the embryo and placenta for glycolysis. We then address dysregulated glycolysis that occurs in the cellular interactive network at the maternal-fetal interface in miscarriage, preeclampsia, fetal growth restriction, and gestational diabetes mellitus. We provide new insights into the field of maternal-fetal medicine from a metabolic perspective, thus revealing the mystery of human pregnancy.
Collapse
Affiliation(s)
- Rui Gou
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing, P.R. China
| | - Xiaohong Zhang
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing, P.R. China
| |
Collapse
|
12
|
Karner E, Muin DA, Klebermass-Schrehof K, Waldhoer T, Yang L. Altitude Modifies the Effect of Parity on Birth Weight/Length Ratio: A Study Comprising 2,057,702 Newborns between 1984 and 2020 in Austria. Life (Basel) 2023; 13:1718. [PMID: 37629575 PMCID: PMC10455113 DOI: 10.3390/life13081718] [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: 07/19/2023] [Revised: 08/07/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
(1) Background: Lower birth weight among newborns in higher altitudes has been well documented in previous literature. Several possible causes for this phenomenon have been investigated, including biophysiological adaptation, epigenetic or genetic mechanisms or lifestyle changes. This is the first study to show the effect modification of altitude and parity on the birth weight length ratio (BWLR) in women resident in moderate altitudes compared to a low sea level.; (2) Methods: This population-based study obtained data on altitude (0-300, 300-500, 500-700,700-900, >900 m), parity (1, 2, …, 7, 8/9), birth weight and length on all births in Austria between 1984 and 2020 from birth certificates provided by Statistics Austria. The BWLR was calculated, and the effect of moderate altitude and parity was estimated using multivariable linear mixed models adjusting for predefined variables. Sub-group regression analyses were conducted by altitude group. (3) Results: Data on 2,057,702 newborns from 1,280,272 mothers were analyzed. The effect of parity on BWLR, as indicated by the difference of BWLR between the first- and second-born infants, ranged between 1.87 to 2.09 g per centimeter across all altitude groups. Our analyses found that the effect of parity on BWLR diminished from parity three onwards at altitude 0-300, whilst the effect of parity on BWLR continued to increase at higher than 300 m and was most notable in the highest altitude group >900 m. (4) Conclusions: Findings from our study indicated that the negative effect of increasing altitude on BWLR was deprived for newborns of higher parity. It shows that the residential altitude can modify the effect of parity on BWLR.
Collapse
Affiliation(s)
- Eva Karner
- Division of Feto-Maternal Medicine, Department of Obstetrics and Gynecology, Medical University of Vienna, 1090 Vienna, Austria; (E.K.); (D.A.M.)
| | - Dana A. Muin
- Division of Feto-Maternal Medicine, Department of Obstetrics and Gynecology, Medical University of Vienna, 1090 Vienna, Austria; (E.K.); (D.A.M.)
| | - Katrin Klebermass-Schrehof
- Division of Neonatology, Pediatric Intensive Care and Neuropediatrics, Department of Pediatrics, Medical University of Vienna, 1090 Vienna, Austria;
| | - Thomas Waldhoer
- Department of Epidemiology, Center for Public Health, Medical University of Vienna, 1090 Vienna, Austria
| | - Lin Yang
- Department of Cancer Epidemiology and Prevention Research, Alberta Health Services, Calgary, AB T2S 3C3, Canada;
- Departments of Oncology and Community Health Sciences, University of Calgary, Calgary, AB T2S 3C3, Canada
| |
Collapse
|
13
|
Kyllo HM, Wang D, Lorca RA, Julian CG, Moore LG, Wilkening RB, Rozance PJ, Brown LD, Wesolowski SR. Adaptive responses in uteroplacental metabolism and fetoplacental nutrient shuttling and sensing during placental insufficiency. Am J Physiol Endocrinol Metab 2023; 324:E556-E568. [PMID: 37126847 PMCID: PMC10259853 DOI: 10.1152/ajpendo.00046.2023] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 04/12/2023] [Accepted: 04/25/2023] [Indexed: 05/03/2023]
Abstract
Glucose, lactate, and amino acids are major fetal nutrients. During placental insufficiency-induced intrauterine growth restriction (PI-IUGR), uteroplacental weight-specific oxygen consumption rates are maintained, yet fetal glucose and amino acid supply is decreased and fetal lactate concentrations are increased. We hypothesized that uteroplacental metabolism adapts to PI-IUGR by altering nutrient allocation to maintain oxidative metabolism. Here, we measured nutrient flux rates, with a focus on nutrients shuttled between the placenta and fetus (lactate-pyruvate, glutamine-glutamate, and glycine-serine) in a sheep model of PI-IUGR. PI-IUGR fetuses weighed 40% less and had decreased oxygen, glucose, and amino acid concentrations and increased lactate and pyruvate versus control (CON) fetuses. Uteroplacental weight-specific rates of oxygen, glucose, lactate, and pyruvate uptake were similar. In PI-IUGR, fetal glucose uptake was decreased and pyruvate output was increased. In PI-IUGR placental tissue, pyruvate dehydrogenase (PDH) phosphorylation was decreased and PDH activity was increased. Uteroplacental glutamine output to the fetus and expression of genes regulating glutamine-glutamate metabolism were lower in PI-IUGR. Fetal glycine uptake was lower in PI-IUGR, with no differences in uteroplacental glycine or serine flux. These results suggest increased placental utilization of pyruvate from the fetus, without higher maternal glucose utilization, and lower fetoplacental amino acid shuttling during PI-IUGR. Mechanistically, AMP-activated protein kinase (AMPK) activation was higher and associated with thiobarbituric acid-reactive substances (TBARS) content, a marker of oxidative stress, and PDH activity in the PI-IUGR placenta, supporting a potential link between oxidative stress, AMPK, and pyruvate utilization. These differences in fetoplacental nutrient sensing and shuttling may represent adaptive strategies enabling the placenta to maintain oxidative metabolism.NEW & NOTEWORTHY These results suggest increased placental utilization of pyruvate from the fetus, without higher maternal glucose uptake, and lower amino acid shuttling in the placental insufficiency-induced intrauterine growth restriction (PI-IUGR) placenta. AMPK activation was associated with oxidative stress and PDH activity, supporting a putative link between oxidative stress, AMPK, and pyruvate utilization. These differences in fetoplacental nutrient sensing and shuttling may represent adaptive strategies enabling the placenta to maintain oxidative metabolism at the expense of fetal growth.
Collapse
Affiliation(s)
- Hannah M Kyllo
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado, United States
| | - Dong Wang
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado, United States
| | - Ramón A Lorca
- Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, Colorado, United States
| | - Colleen G Julian
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, Colorado, United States
| | - Lorna G Moore
- Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, Colorado, United States
| | - Randall B Wilkening
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado, United States
| | - Paul J Rozance
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado, United States
| | - Laura D Brown
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado, United States
| | - Stephanie R Wesolowski
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado, United States
| |
Collapse
|
14
|
Salazar-Petres E, Pereira-Carvalho D, Lopez-Tello J, Sferruzzi-Perri AN. Maternal and Intrauterine Influences on Feto-Placental Growth Are Accompanied by Sexually Dimorphic Changes in Placental Mitochondrial Respiration, and Metabolic Signalling Pathways. Cells 2023; 12:797. [PMID: 36899933 PMCID: PMC10000946 DOI: 10.3390/cells12050797] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 02/26/2023] [Accepted: 03/01/2023] [Indexed: 03/08/2023] Open
Abstract
Adverse maternal environments such as small size, malnutrition, and metabolic conditions are known to influence fetal growth outcomes. Similarly, fetal growth and metabolic alterations may alter the intrauterine environment and affect all fetuses in multiple gestation/litter-bearing species. The placenta is the site of convergence between signals derived from the mother and the developing fetus/es. Its functions are fuelled by energy generated by mitochondrial oxidative phosphorylation (OXPHOS). The aim of this study was to delineate the role of an altered maternal and/or fetal/intrauterine environment in feto-placental growth and placental mitochondrial energetic capacity. To address this, in mice, we used disruptions of the gene encoding phosphoinositol 3-kinase (PI3K) p110α, a growth and metabolic regulator to perturb the maternal and/or fetal/intrauterine environment and study the impact on wildtype conceptuses. We found that feto-placental growth was modified by a perturbed maternal and intrauterine environment, and effects were most evident for wildtype males compared to females. However, placental mitochondrial complex I+II OXPHOS and total electron transport system (ETS) capacity were similarly reduced for both fetal sexes, yet reserve capacity was additionally decreased in males in response to the maternal and intrauterine perturbations. These were also sex-dependent differences in the placental abundance of mitochondrial-related proteins (e.g., citrate synthase and ETS complexes), and activity of growth/metabolic signalling pathways (AKT and MAPK) with maternal and intrauterine alterations. Our findings thus identify that the mother and the intrauterine environment provided by littermates modulate feto-placental growth, placental bioenergetics, and metabolic signalling in a manner dependent on fetal sex. This may have relevance for understanding the pathways leading to reduced fetal growth, particularly in the context of suboptimal maternal environments and multiple gestation/litter-bearing species.
Collapse
Affiliation(s)
- Esteban Salazar-Petres
- Departamento de Ciencias Básicas, Facultad de Ciencias, Universidad Santo Tomás, Valdivia 5090000, Chile
| | - Daniela Pereira-Carvalho
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK
| | - Jorge Lopez-Tello
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK
| | - Amanda N. Sferruzzi-Perri
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK
| |
Collapse
|
15
|
Abstract
AbstractThe idea of putting astronauts into a hibernation-like state during interplanetary spaceflights has sparked new interest in the evolutionary roots of hibernation and torpor. In this context, it should be noted that mammalian fetuses and neonates respond to the environmental challenges in the perinatal period with a number of physiological mechanisms that bear striking similarity to hibernation and torpor. These include three main points: first, prenatal deviation from the overall metabolic size relationship, which adapts the fetus to the low-oxygen conditions in the womb and corresponds to the metabolic reduction during hibernation and estivation; second, intranatal diving bradycardia in response to shortened O2 supply during birth, comparable to the decrease in heart rate preceding the drop in body temperature upon entry into torpor; and third, postnatal onset of nonshivering thermogenesis in the brown adipose tissue, along with the increase in basal metabolic rate up to the level expected from body size, such as during arousal from hibernation. The appearance of hibernation-like adaptations in the perinatal period suggests that, conversely, hibernation and torpor may be composed of mechanisms shared by all mammals around birth. This hypothesis sheds new light on the origins of hibernation and supports its potential accessibility to nonhibernating species, including humans.
Collapse
|
16
|
Zhou J, Jiang Z, Fu L, Qu F, Dai M, Xie N, Zhang S, Wang F. Contribution of labor related gene subtype classification on heterogeneity of polycystic ovary syndrome. PLoS One 2023; 18:e0282292. [PMID: 36857354 PMCID: PMC9977056 DOI: 10.1371/journal.pone.0282292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 02/11/2023] [Indexed: 03/02/2023] Open
Abstract
OBJECTIVE As one of the most common endocrine disorders in women of reproductive age, polycystic ovary syndrome (PCOS) is highly heterogeneous with varied clinical features and diverse gestational complications among individuals. The patients with PCOS have 2-fold higher risk of preterm labor which is associated with substantial infant morbidity and mortality and great socioeconomic cost. The study was designated to identify molecular subtypes and the related hub genes to facilitate the susceptibility assessment of preterm labor in women with PCOS. METHODS Four mRNA datasets (GSE84958, GSE5090, GSE43264 and GSE98421) were obtained from Gene Expression Omnibus database. Twenty-eight candidate genes related to preterm labor or labor were yielded from the researches and our unpublished data. Then, we utilized unsupervised clustering to identify molecular subtypes in PCOS based on the expression of above candidate genes. Key modules were generated with weighted gene co-expression network analysis R package, and their hub genes were generated with CytoHubba. The probable biological function and mechanism were explored through Gene Ontology analysis and Kyoto Encyclopedia of Genes and Genomes pathway analysis. In addition, STRING and Cytoscape software were used to identify the protein-protein interaction (PPI) network, and the molecular complex detection (MCODE) was used to identify the hub genes. Then the overlapping hub genes were predicted. RESULTS Two molecular subtypes were found in women with PCOS based on the expression similarity of preterm labor or labor-related genes, in which two modules were highlighted. The key modules and PPI network have five overlapping five hub genes, two of which, GTF2F2 and MYO6 gene, were further confirmed by the comparison between clustering subgroups according to the expression of hub genes. CONCLUSIONS Distinct PCOS molecular subtypes were identified with preterm labor or labor-related genes, which might uncover the potential mechanism underlying heterogeneity of clinical pregnancy complications in women with PCOS.
Collapse
Affiliation(s)
- Jue Zhou
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, China
| | - Zhou Jiang
- Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Leyi Fu
- Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Fan Qu
- Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Minchen Dai
- Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Ningning Xie
- Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Songying Zhang
- Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- * E-mail: (FW); (SZ)
| | - Fangfang Wang
- Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- * E-mail: (FW); (SZ)
| |
Collapse
|
17
|
Filippi L, Scaramuzzo RT, Pascarella F, Pini A, Morganti R, Cammalleri M, Bagnoli P, Ciantelli M. Fetal oxygenation in the last weeks of pregnancy evaluated through the umbilical cord blood gas analysis. Front Pediatr 2023; 11:1140021. [PMID: 37152310 PMCID: PMC10160648 DOI: 10.3389/fped.2023.1140021] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Accepted: 04/04/2023] [Indexed: 05/09/2023] Open
Abstract
Introduction Embryo and fetus grow and mature over the first trimester of pregnancy in a dynamic hypoxic environment, where placenta development assures an increased oxygen availability. However, it is unclear whether and how oxygenation changes in the later trimesters and, more specifically, in the last weeks of pregnancy. Methods Observational study that evaluated the gas analysis of the umbilical cord blood collected from a cohort of healthy newborns with gestational age ≥37 weeks. Umbilical venous and arterial oxygen levels as well as fetal oxygen extraction were calculated to establish whether oxygenation level changes over the last weeks of pregnancy. In addition, fetal lactate, and carbon dioxide production were analyzed to establish whether oxygen oscillations may induce metabolic effects in utero. Results This study demonstrates a progressive increase in fetal oxygenation levels from the 37th to the 41st weeks of gestation (mean venous PaO2 approximately from 20 to 25 mmHg; p < 0.001). This increase is largely attributable to growing umbilical venous PaO2, regardless of delivery modalities. In neonates born by vaginal delivery, the increased oxygen availability is associated with a modest increase in oxygen extraction, while in neonates born by cesarean section, it is associated with reduced lactate production. Independently from the type of delivery, carbon dioxide production moderately increased. These findings suggest a progressive shift from a prevalent anaerobic metabolism (Warburg effect) towards a growing aerobic metabolism. Conclusion This study confirms that fetuses grow in a hypoxic environment that becomes progressively less hypoxic in the last weeks of gestation. The increased oxygen availability seems to favor aerobic metabolic shift during the last weeks of intrauterine life; we hypothesize that this environmental change may have implications for fetal maturation during intrauterine life.
Collapse
Affiliation(s)
- Luca Filippi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
- Neonatology Unit, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
- Correspondence: Luca Filippi
| | | | | | - Alessandro Pini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Riccardo Morganti
- Section of Statistics, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Maurizio Cammalleri
- Unit of General Physiology, Department of Biology, University of Pisa, Pisa, Italy
| | - Paola Bagnoli
- Unit of General Physiology, Department of Biology, University of Pisa, Pisa, Italy
| | | |
Collapse
|
18
|
Prioreschi A, Aronoff DM, Koethe J, Norris SA, Goldstein JA. Interactions between maternal health and placental morphology on neonate body composition. GLOBAL PEDIATRICS 2022; 3. [PMID: 37073307 PMCID: PMC10107011 DOI: 10.1016/j.gpeds.2022.100030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Background We aimed to examine maternal metabolic correlates of neonate body composition, and the potential mediating effect of the placenta. Methods Data were collected throughout pregnancy and at delivery. An oral glucose tolerance test (OGTT) was conducted in order to diagnose or rule out gestational diabetes mellitus (GDM). Maternal weight and blood pressure were taken and hypertension and gestational weight gain (GWG) were defined. Gestational age, birth weight (BW) and weight to length ration (WLR) were recorded. Photographs were taken of the placenta, and the widths and lengths were measured digitally. Body composition was analysed via air displacement plethysmography or dual-energy x-ray absorptiometry. Mediation models were conducted to determine the mediation effect of the placental variables on the relationships between maternal health variables and neonate outcomes. Next, interaction terms were added to models to determine how maternal and placental variables interacted in their effect on neonate outcomes. Results A total of n = 280 women were included in the analysis. Majority were overweight or obese. Fourteen percent of women developed GDM during pregnancy, 5% had hypertension during pregnancy, 32% were HIV positive, and 32% had anemia. For the birth weight outcome, coefficients of BMI were attenuated by the addition of placental variables (Model 1 β=18.66 vs Model 2 β=16.40). Similar patterns were evident for GWG and hypertension, and for the WLR outcome. In all cases the addition of the placental variables attenuated associations between maternal exposures and neonatal outcomes, yet the level of significance did not change. Inclusion of interaction terms reversed the direction of the relationships between hypertension and BW and WLR, and between GWG and WLR. Conclusion The placenta buffers some harmful effects of obesity, GWG, and hypertension on neonate size, and placental efficiency interacted with most maternal risk factors to either counteract, or attenuate relationships with neonate size at birth. However the placenta was unable to completely counteract the negative effect of excess nutrient supply on in utero growth.
Collapse
Affiliation(s)
- Alessandra Prioreschi
- SAMRC/Wits Developmental Pathways for Health Research Unit, Department of Paediatrics, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Corresponding author at: Chris Hani Baragwanath Academic Hospital, Corner College and Clinic Roads, Soweto, South Africa. (A. Prioreschi)
| | - David M Aronoff
- SAMRC/Wits Developmental Pathways for Health Research Unit, Department of Paediatrics, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232
- Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, Nashville, TN 37232
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232
| | - John Koethe
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232
- Vanderbilt Institute for Global Health, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Shane A Norris
- SAMRC/Wits Developmental Pathways for Health Research Unit, Department of Paediatrics, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Jeffrey A Goldstein
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| |
Collapse
|
19
|
Bahabayi A, Yang N, Xu T, Xue Y, Ma L, Gu X, Wang Y, Jia K. Expression of Matrix Metalloproteinase-2,-7,-9 in Serum during Pregnancy in Patients with Pre-Eclampsia: A Prospective Study. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:14500. [PMID: 36361378 PMCID: PMC9654649 DOI: 10.3390/ijerph192114500] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/26/2022] [Accepted: 10/28/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND Matrix Metalloproteinases (MMPs) have been found to have important roles in vascular pathology and may be involved in the occurrence of pre-eclampsia. In this study, the serum levels of MMP-2, -7, -9 in normal pregnant women and pre-eclampsia patients were analyzed to assess their predictive value. METHODS A total of 1563 pregnant women from Peking University Third Hospital, from February 2021 to October 2021, were enrolled. Serum samples were collected from patients one to three times, during the different trimesters. Among the 102 singleton pre-eclampsia patients, we collected samples from 33 patients in the first trimester (6-13 GW), 33 in the second trimester (14-28 GW), 41 in the third trimester (29-41 GW) and 28 after onset of pre-eclampsia. Samples from each trimester were collected before the onset of pre-eclampsia. Then we selected 35, 37, 43 and 25 samples from 124 healthy pregnant women by matching their age, BMI and gestational weeks, using these as the control groups. Serum levels of MMP-2, -7, -9 were detected by ELISA. The receiver operating characteristic (ROC) curve was used to evaluate their predictive value. RESULTS Except for the first trimester, MMP-2 and MMP-7 were significantly higher in the pre-eclampsia group (p < 0.5). Additionally, in the pre-eclampsia group, MMP-9 increased significantly in the first trimester and after the onset of pre-eclampsia but decreased significantly in the second and third trimesters (p < 0.5). The ROC curve indicated that MMP-9, MMP-2 and MMP-7 were the best indicators for predicting pre-eclampsia in the first, second and third trimesters, respectively. CONCLUSION Increased MMP-2 and MMP-7 levels and a decreased MMP-9 level seem to be related to the pathogenesis of pre-eclampsia and are expected to be potential predictors of pre-eclampsia.
Collapse
Affiliation(s)
- Ayibaota Bahabayi
- Department of Clinical Laboratory, Peking University Third Hospital, Beijing 100191, China
- Department of Clinical Laboratory, Peking University People’s Hospital, Beijing 100033, China
| | - Nan Yang
- Department of Blood Transfusion, Peking University Third Hospital, Beijing 100191, China
| | - Tong Xu
- Department of Clinical Laboratory, Peking University Third Hospital, Beijing 100191, China
| | - Yuting Xue
- Department of Clinical Laboratory, Peking University Third Hospital, Beijing 100191, China
| | - Lijuan Ma
- Department of Clinical Laboratory, Peking University Third Hospital, Beijing 100191, China
| | - Xunke Gu
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China
| | - Yongqing Wang
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China
| | - Keke Jia
- Department of Clinical Laboratory, Peking University Third Hospital, Beijing 100191, China
| |
Collapse
|
20
|
Moore LG, Wesolowski SR, Lorca RA, Murray AJ, Julian CG. Why is human uterine artery blood flow during pregnancy so high? Am J Physiol Regul Integr Comp Physiol 2022; 323:R694-R699. [PMID: 36094446 PMCID: PMC9602899 DOI: 10.1152/ajpregu.00167.2022] [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: 06/29/2022] [Revised: 08/12/2022] [Accepted: 09/07/2022] [Indexed: 11/22/2022]
Abstract
In healthy near-term women, blood flow to the uteroplacental circulation is estimated as 841 mL/min, which is greater than in other mammalian species. We argue that as uterine venous Po2 sets the upper limit for O2 diffusion to the fetus, high uterine artery blood flow serves to narrow the maternal arterial-to-uterine venous Po2 gradient and thereby raise uterine vein Po2. In support, we show that the reported levels for uterine artery blood flow agree with what is required to maintain normal fetal growth. Although residence at high altitudes (>2,500 m) depresses fetal growth, not all populations are equally affected; Tibetans and Andeans have higher levels of uterine artery blood flow than newcomers and exhibit normal fetal growth. Estimates of uterine venous Po2 from the umbilical blood-gas data available from healthy Andean pregnancies indicate that their high levels of uterine artery blood flow are consistent with their reported, normal birth weights. Unknown, however, are the effects on placental gas exchange of the lower levels of uterine artery blood flow seen in high-altitude newcomers or hypoxia-associated pregnancy complications. We speculate that, by widening the maternal artery to uterine vein Po2 gradient, lower levels of uterine artery blood flow prompt metabolic changes that slow fetal growth to match O2 supply.
Collapse
Affiliation(s)
- Lorna G Moore
- Department of Obstetrics and Gynecology, University of Colorado Denver, Aurora, Colorado
| | | | - Ramón A Lorca
- Department of Obstetrics and Gynecology, University of Colorado Denver, Aurora, Colorado
| | - Andrew J Murray
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Colleen G Julian
- Department of Biomedical Informatics, University of Colorado Denver, Aurora, Colorado
| |
Collapse
|
21
|
Ortega MA, Fraile-Martínez O, García-Montero C, Sáez MA, Álvarez-Mon MA, Torres-Carranza D, Álvarez-Mon M, Bujan J, García-Honduvilla N, Bravo C, Guijarro LG, De León-Luis JA. The Pivotal Role of the Placenta in Normal and Pathological Pregnancies: A Focus on Preeclampsia, Fetal Growth Restriction, and Maternal Chronic Venous Disease. Cells 2022; 11:cells11030568. [PMID: 35159377 PMCID: PMC8833914 DOI: 10.3390/cells11030568] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 02/02/2022] [Accepted: 02/05/2022] [Indexed: 12/01/2022] Open
Abstract
The placenta is a central structure in pregnancy and has pleiotropic functions. This organ grows incredibly rapidly during this period, acting as a mastermind behind different fetal and maternal processes. The relevance of the placenta extends far beyond the pregnancy, being crucial for fetal programming before birth. Having integrative knowledge of this maternofetal structure helps significantly in understanding the development of pregnancy either in a proper or pathophysiological context. Thus, the aim of this review is to summarize the main features of the placenta, with a special focus on its early development, cytoarchitecture, immunology, and functions in non-pathological conditions. In contraposition, the role of the placenta is examined in preeclampsia, a worrisome hypertensive disorder of pregnancy, in order to describe the pathophysiological implications of the placenta in this disease. Likewise, dysfunction of the placenta in fetal growth restriction, a major consequence of preeclampsia, is also discussed, emphasizing the potential clinical strategies derived. Finally, the emerging role of the placenta in maternal chronic venous disease either as a causative agent or as a consequence of the disease is equally treated.
Collapse
Affiliation(s)
- Miguel A. Ortega
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Madrid, Spain; (O.F.-M.); (C.G.-M.); (M.A.S.); (M.A.Á.-M.); (D.T.-C.); (M.Á.-M.); (J.B.); (N.G.-H.)
- Ramón y Cajal Institute of Healthcare Research (IRYCIS), 28034 Madrid, Spain;
- Cancer Registry and Pathology Department, Hospital Universitario Principe de Asturias, 28801 Alcalá de Henares, Madrid, Spain
- Correspondence: ; Tel.: +34-91-885-4540; Fax: +34-91-885-4885
| | - Oscar Fraile-Martínez
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Madrid, Spain; (O.F.-M.); (C.G.-M.); (M.A.S.); (M.A.Á.-M.); (D.T.-C.); (M.Á.-M.); (J.B.); (N.G.-H.)
- Ramón y Cajal Institute of Healthcare Research (IRYCIS), 28034 Madrid, Spain;
| | - Cielo García-Montero
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Madrid, Spain; (O.F.-M.); (C.G.-M.); (M.A.S.); (M.A.Á.-M.); (D.T.-C.); (M.Á.-M.); (J.B.); (N.G.-H.)
- Ramón y Cajal Institute of Healthcare Research (IRYCIS), 28034 Madrid, Spain;
| | - Miguel A. Sáez
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Madrid, Spain; (O.F.-M.); (C.G.-M.); (M.A.S.); (M.A.Á.-M.); (D.T.-C.); (M.Á.-M.); (J.B.); (N.G.-H.)
- Ramón y Cajal Institute of Healthcare Research (IRYCIS), 28034 Madrid, Spain;
- Pathological Anatomy Service, Central University Hospital of Defence-UAH, 28047 Madrid, Spain
| | - Miguel Angel Álvarez-Mon
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Madrid, Spain; (O.F.-M.); (C.G.-M.); (M.A.S.); (M.A.Á.-M.); (D.T.-C.); (M.Á.-M.); (J.B.); (N.G.-H.)
- Ramón y Cajal Institute of Healthcare Research (IRYCIS), 28034 Madrid, Spain;
| | - Diego Torres-Carranza
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Madrid, Spain; (O.F.-M.); (C.G.-M.); (M.A.S.); (M.A.Á.-M.); (D.T.-C.); (M.Á.-M.); (J.B.); (N.G.-H.)
| | - Melchor Álvarez-Mon
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Madrid, Spain; (O.F.-M.); (C.G.-M.); (M.A.S.); (M.A.Á.-M.); (D.T.-C.); (M.Á.-M.); (J.B.); (N.G.-H.)
- Ramón y Cajal Institute of Healthcare Research (IRYCIS), 28034 Madrid, Spain;
- Immune System Diseases-Rheumatology and Oncology Service, University Hospital Príncipe de Asturias, CIBEREHD, 28801 Alcalá de Henares, Madrid, Spain
| | - Julia Bujan
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Madrid, Spain; (O.F.-M.); (C.G.-M.); (M.A.S.); (M.A.Á.-M.); (D.T.-C.); (M.Á.-M.); (J.B.); (N.G.-H.)
- Ramón y Cajal Institute of Healthcare Research (IRYCIS), 28034 Madrid, Spain;
| | - Natalio García-Honduvilla
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Madrid, Spain; (O.F.-M.); (C.G.-M.); (M.A.S.); (M.A.Á.-M.); (D.T.-C.); (M.Á.-M.); (J.B.); (N.G.-H.)
- Ramón y Cajal Institute of Healthcare Research (IRYCIS), 28034 Madrid, Spain;
| | - Coral Bravo
- Department of Public and Maternal and Child Health, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain; (C.B.); (J.A.D.L.-L.)
- Department of Obstetrics and Gynecology, University Hospital Gregorio Marañón, 28009 Madrid, Spain
- Health Research Institute Gregorio Marañón, 28009 Madrid, Spain
| | - Luis G. Guijarro
- Ramón y Cajal Institute of Healthcare Research (IRYCIS), 28034 Madrid, Spain;
- Unit of Biochemistry and Molecular Biology (CIBEREHD), Department of System Biology, University of Alcalá, 28801 Alcalá de Henares, Madrid, Spain
| | - Juan A. De León-Luis
- Department of Public and Maternal and Child Health, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain; (C.B.); (J.A.D.L.-L.)
- Department of Obstetrics and Gynecology, University Hospital Gregorio Marañón, 28009 Madrid, Spain
- Health Research Institute Gregorio Marañón, 28009 Madrid, Spain
| |
Collapse
|
22
|
SARS-CoV-2 and pregnancy: placental lesions and therapeutic conduct. Case presentation. GINECOLOGIA.RO 2022. [DOI: 10.26416/gine.36.2.2022.6550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
|
23
|
Ortega MA, Sáez MA, Fraile-Martínez O, Álvarez-Mon MA, García-Montero C, Guijarro LG, Asúnsolo Á, Álvarez-Mon M, Bujan J, García-Honduvilla N, De León-Luis JA, Bravo C. Overexpression of glycolysis markers in placental tissue of pregnant women with chronic venous disease: a histological study. Int J Med Sci 2022; 19:186-194. [PMID: 34975312 PMCID: PMC8692115 DOI: 10.7150/ijms.65419] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 11/24/2021] [Indexed: 12/15/2022] Open
Abstract
Chronic Venous Disease (CVD) refers to a wide variety of venous disorders being the varicose veins its most common manifestation. It is well-established the link between pregnancy and the risk of suffering CVD, due to hormonal or haematological factors, especially during the third trimester. In the same manner, previous studies have demonstrated the detrimental effect of this condition in the placental tissue of pregnant women, including in the normal physiology and the metabolomic profile of this organ. In this context, the aim of this study was to evaluate the glucose homeostasis in the placental tissue of women presenting CVD. Through immunohistochemistry, we studied the protein expression of the glucose transporter 1 (GLUT-1), Phosphoglycerate kinase 1 (PGK1), aldolase (ALD), Glyceraldehyde-3-phosphate dehydrogenase (GA3PDH) and lactate dehydrogenase (LDH). Our results have reported a significative increase in the expression of GLUT-1, PGK1, ALD, GA3PDH and the isoenzyme LDHA in placentas of women with CVD. This work has proven for the first-time an altered glucose metabolism in the placental tissue of women affected by CVD, what may aid to understand the pathophysiological mechanisms of this condition in more distant organs such as placenta. Furthermore, our research also supports the basis for further studies in the metabolic phenotyping of the human placenta due to CVD, which may be considered during the late pregnancy in these women.
Collapse
Affiliation(s)
- Miguel A Ortega
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Madrid, Spain
- Ramón y Cajal Institute of Healthcare Research (IRYCIS), Madrid, Spain
- Cancer Registry and Pathology Department, Hospital Universitario Principe de Asturias, Alcalá de Henares, Spain
| | - Miguel A Sáez
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Madrid, Spain
- Ramón y Cajal Institute of Healthcare Research (IRYCIS), Madrid, Spain
- Pathological Anatomy Service, Central University Hospital of Defence-UAH Madrid, Spain
| | - Oscar Fraile-Martínez
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Madrid, Spain
| | - Miguel A Álvarez-Mon
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Madrid, Spain
- Ramón y Cajal Institute of Healthcare Research (IRYCIS), Madrid, Spain
| | - Cielo García-Montero
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Madrid, Spain
| | - Luis G Guijarro
- Ramón y Cajal Institute of Healthcare Research (IRYCIS), Madrid, Spain
- Unit of Biochemistry and Molecular Biology (CIBEREHD), Department of System Biology, University of Alcalá, 28801 Alcalá de Henares, Spain
| | - Ángel Asúnsolo
- Ramón y Cajal Institute of Healthcare Research (IRYCIS), Madrid, Spain
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Madrid, Spain
| | - Melchor Álvarez-Mon
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Madrid, Spain
- Ramón y Cajal Institute of Healthcare Research (IRYCIS), Madrid, Spain
- Immune System Diseases-Rheumatology and Oncology Service, University Hospital Príncipe de Asturias, CIBEREHD, Alcalá de Henares, Madrid, Spain
| | - Julia Bujan
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Madrid, Spain
- Ramón y Cajal Institute of Healthcare Research (IRYCIS), Madrid, Spain
| | - Natalio García-Honduvilla
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Madrid, Spain
- Ramón y Cajal Institute of Healthcare Research (IRYCIS), Madrid, Spain
| | - Juan A De León-Luis
- Department of Public and Maternal and Child Health, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain
- Department of Obstetrics and Gynecology, University Hospital Gregorio Marañón, Madrid 28009, Spain
- Health Research Institute Gregorio Marañón, 28009 Madrid, Spain
| | - Coral Bravo
- Department of Public and Maternal and Child Health, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain
- Department of Obstetrics and Gynecology, University Hospital Gregorio Marañón, Madrid 28009, Spain
- Health Research Institute Gregorio Marañón, 28009 Madrid, Spain
| |
Collapse
|
24
|
Zhang Y, Zhou W, Feng W, Hu J, Hu K, Cui L, Chen ZJ. Assisted Reproductive Technology Treatment, the Catalyst to Amplify the Effect of Maternal Infertility on Preterm Birth. Front Endocrinol (Lausanne) 2022; 13:791229. [PMID: 35721737 PMCID: PMC9200983 DOI: 10.3389/fendo.2022.791229] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 04/27/2022] [Indexed: 11/21/2022] Open
Abstract
OBJECTIVE To identify the influence of different infertility causes and assisted reproductive technology (ART) treatment on perinatal outcomes and clarify the relationship between the maternal pathophysiological changes and artificial interventions. METHODS A total of 1,629 fertile women and 27,112 infertile women with sole infertility causes were prospectively recruited from July 2014 to December 2017, and 9,894 singletons were finally enrolled into the study. Pregnancies with more than one cause of infertility and/or multiple births were excluded. According to the causes of infertility and the exposure of ART treatment, the participants were divided into four groups, namely, fertile naturally conceived (NC) group, infertile NC group, female factor ART group, and male factor ART group. Perinatal outcomes, including gestational age of delivery (GA), birth weight (BW), preterm birth (PTB), low birth weight (LBW), small for gestational age (SGA), and large for gestational age (LGA), were compared among groups. Logistic regression was performed for the adjustment of several covariates. RESULTS The birth outcomes of the infertile NC group and fertile NC group, female factor ART group, and infertile NC group were comparable. Compared to the fertile NC group, the female factor ART group had a shorter GA (39.0 ± 1.6 vs. 39.3 ± 1.5 weeks, BW: P < 0.05). An interaction test showed that ART treatment had an interaction on the effect of female infertility on GA (P = 0.023). The female factor ART group also had a higher risk of PTB (OR 1.56, 95% CI 1.18-2.07) and LGA (OR 1.27, 95% CI 1.10-1.47) compared to the fertile NC group. The risk of PTB was increased for tubal factor ART (OR 1.49, 95% CI 1.12-2.00), ovulatory dysfunction ART (OR 1.87, 95% CI 1.29-2.72), and unexplained infertility ART (OR 1.88, 95% CI 1.11-3.17). The risk of LGA was increased for tubal factor ART (OR 1.28, 95% CI 1.11-1.48) and ovulatory dysfunction ART (OR 1.27, 95% CI 1.03-1.57). CONCLUSIONS Our findings indicated that ART treatment could amplify the adverse effect of female infertility on neonates. Women with tubal factor infertility, ovulatory dysfunction, and unexplained infertility have a higher risk of PTB after ART treatment. Thus, clinicians should be vigilant in such patients and provide corresponding prevention strategies before and during pregnancy.
Collapse
Affiliation(s)
- Youzhen Zhang
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, China
| | - Wei Zhou
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, China
| | - Wanbing Feng
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, China
| | - Jingmei Hu
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, China
| | - Kuona Hu
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, China
| | - Linlin Cui
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, China
- *Correspondence: Linlin Cui,
| | - Zi-Jiang Chen
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
25
|
Placental mtDNA copy number and methylation in association with macrosomia in healthy pregnancy. Placenta 2021; 118:1-9. [PMID: 34972066 DOI: 10.1016/j.placenta.2021.12.021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 12/21/2021] [Accepted: 12/23/2021] [Indexed: 11/20/2022]
Abstract
INTRODUCTION Fetal growth and development depend on metabolic energy from placental mitochondria. However, the impact of placental mitochondria on the occurrence of macrosomia remains unclear. We aimed to explore the association between macrosomia without gestational diabetes mellitus (non-GDM) and changes in placental mitochondrial DNA (mtDNA) copy number and methylation. METHODS Fifty-four newborns with macrosomia and 54 normal birthweight controls were enrolled in this study. Placental mtDNA copy number and mRNA expression of nuclear genes related to mitochondrial replication or ATP synthesis-related genes were measured by real-time quantitative polymerase chain reaction (qPCR). Methylation levels of the non-coding regulatory region D-loop and ATP synthesis-related genes were detected by targeted bisulfite sequencing. RESULTS Newborns with macrosomia had lower placental mtDNA copy number and higher methylation rates of the CpG15 site in the D-loop region (D-CpG15) and CpG6 site in the cytochrome C oxidase III (COX3) gene (COX3-CpG6) than normal birth weight newborns. After adjusting for potential covariates (gestational age, prepregnancy BMI, and infant sex), decreased placental mtDNA copy number (adjusted odds ratio [aOR] = 2.09, 95% confidence interval [CI] 1.03-4.25), elevated methylation rate of D-CpG15 (aOR = 2.06, 95% CI 1.03-4.09) and COX3-CpG6 (aOR = 2.13, 95% CI 1.08-4.20) remained significantly associated with a higher risk of macrosomia. DISCUSSION Reduced mtDNA copy number and increased methylation levels of specific loci at mtDNA would increase the risk of macrosomia. However, the detailed molecular mechanism needs further identification.
Collapse
|
26
|
Lee SSY, Mackey DA, Sanfilippo PG, Hewitt AW, Craig JE, Yazar S. In Utero Exposure to Smoking and Alcohol, and Passive Smoking during Childhood: Effect on the Retinal Nerve Fibre Layer in Young Adulthood. Ophthalmic Epidemiol 2021; 29:507-514. [PMID: 34486472 DOI: 10.1080/09286586.2021.1968005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
PURPOSE In utero exposure to cigarette smoke has been suggested to result in thinner retinal nerve fibre layer (RNFL). However, the potential cofounding effects of in utero alcohol exposure and passive smoking during childhood had not been considered. We explored RNFL thickness in young adults in relation to these early life factors. METHODS In 1989-1991, pregnant women completed questionnaires on their current smoking and alcohol drinking patterns. Following the birth of their offspring, information on household smokers was obtained between the 1- and 13-year follow-ups. At the 20-year follow-up, these offspring underwent an eye examination including optical coherence tomography imaging of the RNFL. RESULTS Participants (n = 1,287) were 19-22 years old at time of eye examination. Most participants (77%) had no in utero exposure to cigarette smoke; 1.3% were initially exposed but not after 18 weeks' gestation, while 21% had continual in utero smoking exposure. Half of the mothers never consumed alcohol or only consumed alcohol once during their pregnancies. After correcting for potential confounders, including in utero alcohel exposure and childhood passive smoking, participants who had continued in utero exposure to >10 cigarettes/day and ≤10 cigarettes/day had thinner RNFLs by 6.6 (95% confidence interval [CI] = 4.4-8.7) and 3.7 µm (95%[CI] = 2.3-5.5), respectively, than those with no exposure (p < .001). In utero alcohol exposure and childhood passive smoking were not significantly associated with RNFL thickness after accounting for in utero exposure to smoking. CONCLUSIONS In utero exposure to cigarette smoke is associated with thinner RFNL in young adulthood, independent of other early life environmental factors.
Collapse
Affiliation(s)
- Samantha Sze-Yee Lee
- Centre for Ophthalmology and Visual Science (Incorporating Lions Eye Institute), the University of Western Australia, Perth, Western Australia, Australia
| | - David A Mackey
- Centre for Ophthalmology and Visual Science (Incorporating Lions Eye Institute), the University of Western Australia, Perth, Western Australia, Australia.,School Of Medicine, Menzies Research Institute Tasmania, University Of Tasmania, Hobart, TAS, Australia.,Centre For Eye Research Australia, University Of Melbourne, Royal Victorian Eye And Ear Hospital, East Melbourne, VIC, Australia
| | - Paul G Sanfilippo
- Centre For Eye Research Australia, University Of Melbourne, Royal Victorian Eye And Ear Hospital, East Melbourne, VIC, Australia
| | - Alex W Hewitt
- Centre for Ophthalmology and Visual Science (Incorporating Lions Eye Institute), the University of Western Australia, Perth, Western Australia, Australia.,School Of Medicine, Menzies Research Institute Tasmania, University Of Tasmania, Hobart, TAS, Australia.,Centre For Eye Research Australia, University Of Melbourne, Royal Victorian Eye And Ear Hospital, East Melbourne, VIC, Australia
| | - Jamie E Craig
- Eye And Vision, Flinders Health And Medical Institute, Flinders University, Adelaide, South Australia, Australia
| | - Seyhan Yazar
- Centre for Ophthalmology and Visual Science (Incorporating Lions Eye Institute), the University of Western Australia, Perth, Western Australia, Australia.,Single Cell and Computational Genomics Laboratory, Garvan-Weizmann Centre For Cellular Genomics, Garvan Institute Of Medical Research, Sydney, NSW, Australia
| |
Collapse
|
27
|
Wilsterman K, Cheviron ZA. Fetal growth, high altitude, and evolutionary adaptation: a new perspective. Am J Physiol Regul Integr Comp Physiol 2021; 321:R279-R294. [PMID: 34259046 PMCID: PMC8530763 DOI: 10.1152/ajpregu.00067.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 07/07/2021] [Accepted: 07/09/2021] [Indexed: 11/22/2022]
Abstract
Residence at high altitude is consistently associated with low birthweight among placental mammals. This reduction in birthweight influences long-term health trajectories for both the offspring and mother. However, the physiological processes that contribute to fetal growth restriction at altitude are still poorly understood, and thus our ability to safely intervene remains limited. One approach to identify the factors that mitigate altitude-dependent fetal growth restriction is to study populations that are protected from fetal growth restriction through evolutionary adaptations (e.g., high altitude-adapted populations). Here, we examine human gestational physiology at high altitude from a novel evolutionary perspective that focuses on patterns of physiological plasticity, allowing us to identify 1) the contribution of specific physiological systems to fetal growth restriction and 2) the mechanisms that confer protection in highland-adapted populations. Using this perspective, our review highlights two general findings: first, that the beneficial value of plasticity in maternal physiology is often dependent on factors more proximate to the fetus; and second, that our ability to understand the contributions of these proximate factors is currently limited by thin data from altitude-adapted populations. Expanding the comparative scope of studies on gestational physiology at high altitude and integrating studies of both maternal and fetal physiology are needed to clarify the mechanisms by which physiological responses to altitude contribute to fetal growth outcomes. The relevance of these questions to clinical, agricultural, and basic research combined with the breadth of the unknown highlight gestational physiology at high altitude as an exciting niche for continued work.
Collapse
Affiliation(s)
- Kathryn Wilsterman
- Division of Biological Sciences, University of Montana, Missoula, Montana
| | - Zachary A Cheviron
- Division of Biological Sciences, University of Montana, Missoula, Montana
| |
Collapse
|
28
|
Grant ID, Giussani DA, Aiken CE. Blood pressure and hypertensive disorders of pregnancy at high altitude: a systematic review and meta-analysis. Am J Obstet Gynecol MFM 2021; 3:100400. [PMID: 34023533 DOI: 10.1016/j.ajogmf.2021.100400] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/06/2021] [Accepted: 05/15/2021] [Indexed: 01/17/2023]
Abstract
OBJECTIVE Exposure to high altitude (≥2500 m) is associated with increased arterial blood pressure. During pregnancy, even a mild elevation of maternal blood pressure is associated with reduced birthweight and increased prevalence of pregnancy complications. This study aimed to systematically assess the impact of altitude on maternal blood pressure at term and on the prevalence of hypertensive disorders of pregnancy. DATA SOURCES PubMed, Ovid Embase, Cochrane Library, Medline, Web of Science, and ClinicalTrials.gov were searched (inception to November 11, 2020). STUDY APPRAISAL AND SYNTHESIS METHODS Observational, cohort, or case-control studies were included if they reported a high-altitude and appropriate control pregnant population. Studies published >50 years ago were excluded; 2 reviewers independently assessed articles for eligibility and risk of bias. RESULTS At high altitude, maternal systolic and diastolic blood pressure at term was higher than at low altitude (4.8±1.6 mm Hg; P<.001; 4.0±0.8 mm Hg; P<.001, respectively). Hypertensive disorders of pregnancy were more common at high altitude (odds ratio, 1.31 [1.03-1.65]; P<.05). The prevalence of gestational hypertension was nearly twice as high at high altitude (odds ratio, 1.92 [1.15-3.22]; P<.05) but the prevalence of preeclampsia was half as high (odds ratio, 0.57 [0.46-0.70]; P<.001). The likelihood of stillbirth was increased by 63% in pregnancies at high altitude compared with low altitude (odds ratio, 1.63 [1.12-2.35]; P<.01). CONCLUSION Maternal blood pressure is higher at term in pregnancies at high altitude than low altitude, accompanied with an increased risk of gestational hypertension but not preeclampsia. Risk of stillbirth at high altitude is also increased. With a growing population residing at high altitude worldwide, it is essential to clearly define the associated risk of adverse pregnancy outcomes.
Collapse
Affiliation(s)
- Imogen D Grant
- Department of Obstetrics and Gynaecology (Ms Grant and Dr Aiken); NIHR Cambridge Biomedical Research Centre, Cambridge, United Kingdom (Ms Grant and Dr Aiken).
| | - Dino A Giussani
- Department of Physiology, Development and Neuroscience (Dr Giussani), University of Cambridge, Cambridge, United Kingdom
| | - Catherine E Aiken
- Department of Obstetrics and Gynaecology (Ms Grant and Dr Aiken); NIHR Cambridge Biomedical Research Centre, Cambridge, United Kingdom (Ms Grant and Dr Aiken)
| |
Collapse
|
29
|
Hypoxia and the integrated stress response promote pulmonary hypertension and preeclampsia: Implications in drug development. Drug Discov Today 2021; 26:2754-2773. [PMID: 34302972 DOI: 10.1016/j.drudis.2021.07.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 03/31/2021] [Accepted: 07/14/2021] [Indexed: 11/21/2022]
Abstract
Chronic hypoxia is a common cause of pulmonary hypertension, preeclampsia, and intrauterine growth restriction (IUGR). The molecular mechanisms underlying these diseases are not completely understood. Chronic hypoxia may induce the generation of reactive oxygen species (ROS) in mitochondria, promote endoplasmic reticulum (ER) stress, and result in the integrated stress response (ISR) in the pulmonary artery and uteroplacental tissues. Numerous studies have implicated hypoxia-inducible factors (HIFs), oxidative stress, and ER stress/unfolded protein response (UPR) in the development of pulmonary hypertension, preeclampsia and IUGR. This review highlights the roles of HIFs, mitochondria-derived ROS and UPR, as well as their interplay, in the pathogenesis of pulmonary hypertension and preeclampsia, and their implications in drug development.
Collapse
|
30
|
Alhassen S, Chen S, Alhassen L, Phan A, Khoudari M, De Silva A, Barhoosh H, Wang Z, Parrocha C, Shapiro E, Henrich C, Wang Z, Mutesa L, Baldi P, Abbott GW, Alachkar A. Intergenerational trauma transmission is associated with brain metabotranscriptome remodeling and mitochondrial dysfunction. Commun Biol 2021; 4:783. [PMID: 34168265 PMCID: PMC8225861 DOI: 10.1038/s42003-021-02255-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 05/18/2021] [Indexed: 12/21/2022] Open
Abstract
Intergenerational trauma increases lifetime susceptibility to depression and other psychiatric disorders. Whether intergenerational trauma transmission is a consequence of in-utero neurodevelopmental disruptions versus early-life mother–infant interaction is unknown. Here, we demonstrate that trauma exposure during pregnancy induces in mouse offspring social deficits and depressive-like behavior. Normal pups raised by traumatized mothers exhibited similar behavioral deficits to those induced in pups raised by their biological traumatized mothers. Good caregiving by normal mothers did not reverse prenatal trauma-induced behaviors, indicating a two-hit stress mechanism comprising both in-utero abnormalities and early-life poor parenting. The behavioral deficits were associated with profound changes in the brain metabotranscriptome. Striking increases in the mitochondrial hypoxia marker and epigenetic modifier 2-hydroxyglutaric acid in the brains of neonates and adults exposed prenatally to trauma indicated mitochondrial dysfunction and epigenetic mechanisms. Bioinformatic analyses revealed stress- and hypoxia-response metabolic pathways in the neonates, which produced long-lasting alterations in mitochondrial energy metabolism and epigenetic processes (DNA and chromatin modifications). Most strikingly, early pharmacological interventions with acetyl-L-carnitine (ALCAR) supplementation produced long-lasting protection against intergenerational trauma-induced depression. Sammy Alhassen, Siwei Chen, et al. use mouse models to examine the effects of prenatal and postnatal stress on metabolomic and transcriptomic pathways in the brain. Their results suggest that altered mitochondrial metabolism may underlie trauma-induced behavioral deficits, and that correcting metabolism with ALCAR supplementation may protect against intergenerational transmission of traumatic stress.
Collapse
Affiliation(s)
- Sammy Alhassen
- Department of Pharmaceutical Sciences, University of California, Irvine, CA, USA
| | - Siwei Chen
- Department of Computer Science, School of Information and Computer Sciences, University of California, Irvine, CA, USA.,Institute for Genomics and Bioinformatics, School of Information and Computer Sciences, University of California, Irvine, CA, USA
| | - Lamees Alhassen
- Department of Pharmaceutical Sciences, University of California, Irvine, CA, USA
| | - Alvin Phan
- Department of Pharmaceutical Sciences, University of California, Irvine, CA, USA
| | - Mohammad Khoudari
- Department of Pharmaceutical Sciences, University of California, Irvine, CA, USA
| | - Angele De Silva
- Bioelectricity Laboratory, Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, USA
| | - Huda Barhoosh
- Department of Pharmaceutical Sciences, University of California, Irvine, CA, USA
| | - Zitong Wang
- Department of Pharmaceutical Sciences, University of California, Irvine, CA, USA
| | - Chelsea Parrocha
- Department of Pharmaceutical Sciences, University of California, Irvine, CA, USA
| | - Emily Shapiro
- Department of Pharmaceutical Sciences, University of California, Irvine, CA, USA
| | - Charity Henrich
- Department of Pharmaceutical Sciences, University of California, Irvine, CA, USA
| | - Zicheng Wang
- Department of Pharmaceutical Sciences, University of California, Irvine, CA, USA
| | - Leon Mutesa
- Center for Human Genetics, College of Medicine and Health Sciences, University of Rwanda, Kigali, Rwanda
| | - Pierre Baldi
- Department of Computer Science, School of Information and Computer Sciences, University of California, Irvine, CA, USA.,Institute for Genomics and Bioinformatics, School of Information and Computer Sciences, University of California, Irvine, CA, USA
| | - Geoffrey W Abbott
- Bioelectricity Laboratory, Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, USA
| | - Amal Alachkar
- Department of Pharmaceutical Sciences, University of California, Irvine, CA, USA. .,Institute for Genomics and Bioinformatics, School of Information and Computer Sciences, University of California, Irvine, CA, USA.
| |
Collapse
|
31
|
Hu XQ, Zhang L. Hypoxia and Mitochondrial Dysfunction in Pregnancy Complications. Antioxidants (Basel) 2021; 10:antiox10030405. [PMID: 33800426 PMCID: PMC7999178 DOI: 10.3390/antiox10030405] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/02/2021] [Accepted: 03/03/2021] [Indexed: 02/07/2023] Open
Abstract
Hypoxia is a common and severe stress to an organism's homeostatic mechanisms, and hypoxia during gestation is associated with significantly increased incidence of maternal complications of preeclampsia, adversely impacting on the fetal development and subsequent risk for cardiovascular and metabolic disease. Human and animal studies have revealed a causative role of increased uterine vascular resistance and placental hypoxia in preeclampsia and fetal/intrauterine growth restriction (FGR/IUGR) associated with gestational hypoxia. Gestational hypoxia has a major effect on mitochondria of uteroplacental cells to overproduce reactive oxygen species (ROS), leading to oxidative stress. Excess mitochondrial ROS in turn cause uteroplacental dysfunction by damaging cellular macromolecules, which underlies the pathogenesis of preeclampsia and FGR. In this article, we review the current understanding of hypoxia-induced mitochondrial ROS and their role in placental dysfunction and the pathogenesis of pregnancy complications. In addition, therapeutic approaches selectively targeting mitochondrial ROS in the placental cells are discussed.
Collapse
|
32
|
Gragasin FS, Ospina MB, Serrano-Lomelin J, Kim SH, Kokotilo M, Woodman AG, Renaud SJ, Bourque SL. Maternal and Cord Blood Hemoglobin as Determinants of Placental Weight: A Cross-Sectional Study. J Clin Med 2021; 10:jcm10050997. [PMID: 33801215 PMCID: PMC7957881 DOI: 10.3390/jcm10050997] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 02/16/2021] [Accepted: 02/20/2021] [Indexed: 12/12/2022] Open
Abstract
Background: Both high and low placental weights are associated with adverse pregnancy outcomes. Maternal hemoglobin levels can influence placental weight, but the evidence is conflicting. Since maternal hemoglobin does not invariably correlate with fetal/neonatal blood hemoglobin levels, we sought to determine whether cord blood hemoglobin or maternal hemoglobin status more closely associates with placental weight in women undergoing elective cesarean section at term. Methods: This was a cross-sectional study conducted at the Royal Alexandra Hospital, Edmonton, Canada, involving 202 women with term singleton pregnancies undergoing elective cesarean section. Maternal blood and mixed cord blood hemoglobin levels were analyzed using a HemoCue Hb201+ system. Birth weight, placental weight, one- and five-minute APGAR scores, American Society of Anesthesiologists physical state classification, maternal age, and maternal height were also recorded. Relationships between maternal and cord blood hemoglobin levels with placental weight, birth weight, and birth weight to placental weight ratio were the main outcome measures. Results: A total of 182 subjects were included in the analysis. Regression analysis showed that cord blood hemoglobin, but not maternal hemoglobin, was inversely related with placental weight (β = −2.4, p = 0.001) and positively related with the birth weight to placental weight ratio (β = 0.015, p = 0.001 and p = 0.63, respectively). Conclusions: Our findings suggest that measuring cord blood hemoglobin levels, rather than maternal hemoglobin levels, may provide important diagnostic information about in utero fetal adaptation to suboptimal placental function and neonatal health.
Collapse
Affiliation(s)
- Ferrante S. Gragasin
- Department of Anesthesiology & Pain Medicine, University of Alberta, Edmonton, AB T6G 2G3, Canada; (F.S.G.); (M.K.)
- Women and Children’s Health Research Institute, University of Alberta, Edmonton, AB T6G 1C9, Canada; (M.B.O.); (J.S.-L.); (A.G.W.)
| | - Maria B. Ospina
- Women and Children’s Health Research Institute, University of Alberta, Edmonton, AB T6G 1C9, Canada; (M.B.O.); (J.S.-L.); (A.G.W.)
- Department of Obstetrics & Gynecology, University of Alberta, Edmonton, AB T6G 2R7, Canada
| | - Jesus Serrano-Lomelin
- Women and Children’s Health Research Institute, University of Alberta, Edmonton, AB T6G 1C9, Canada; (M.B.O.); (J.S.-L.); (A.G.W.)
- Department of Obstetrics & Gynecology, University of Alberta, Edmonton, AB T6G 2R7, Canada
| | - Su Hwan Kim
- Department of Mathematical & Statistical Sciences, University of Alberta, Edmonton, AB T6G 2G1, Canada;
| | - Matthew Kokotilo
- Department of Anesthesiology & Pain Medicine, University of Alberta, Edmonton, AB T6G 2G3, Canada; (F.S.G.); (M.K.)
| | - Andrew G. Woodman
- Women and Children’s Health Research Institute, University of Alberta, Edmonton, AB T6G 1C9, Canada; (M.B.O.); (J.S.-L.); (A.G.W.)
- Department of Pharmacology, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Stephen J. Renaud
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON N6A 5C1, Canada;
| | - Stephane L. Bourque
- Department of Anesthesiology & Pain Medicine, University of Alberta, Edmonton, AB T6G 2G3, Canada; (F.S.G.); (M.K.)
- Women and Children’s Health Research Institute, University of Alberta, Edmonton, AB T6G 1C9, Canada; (M.B.O.); (J.S.-L.); (A.G.W.)
- Department of Pharmacology, University of Alberta, Edmonton, AB T6G 2H7, Canada
- Correspondence: ; Tel.: +1-780-492-6000
| |
Collapse
|
33
|
Sharma K, Gupta S, Sharma C, Chhabra A, Jeengar L, Sharma N. Pathophysiologic and anaesthetic considerations in iron deficiency anaemia and pregnancy; An update. JOURNAL OF OBSTETRIC ANAESTHESIA AND CRITICAL CARE 2021. [DOI: 10.4103/joacc.joacc_46_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
34
|
Colson A, Sonveaux P, Debiève F, Sferruzzi-Perri AN. Adaptations of the human placenta to hypoxia: opportunities for interventions in fetal growth restriction. Hum Reprod Update 2020; 27:531-569. [PMID: 33377492 DOI: 10.1093/humupd/dmaa053] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 10/15/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The placenta is the functional interface between the mother and the fetus during pregnancy, and a critical determinant of fetal growth and life-long health. In the first trimester, it develops under a low-oxygen environment, which is essential for the conceptus who has little defense against reactive oxygen species produced during oxidative metabolism. However, failure of invasive trophoblasts to sufficiently remodel uterine arteries toward dilated vessels by the end of the first trimester can lead to reduced/intermittent blood flow, persistent hypoxia and oxidative stress in the placenta with consequences for fetal growth. Fetal growth restriction (FGR) is observed in ∼10% of pregnancies and is frequently seen in association with other pregnancy complications, such as preeclampsia (PE). FGR is one of the main challenges for obstetricians and pediatricians, as smaller fetuses have greater perinatal risks of morbidity and mortality and postnatal risks of neurodevelopmental and cardio-metabolic disorders. OBJECTIVE AND RATIONALE The aim of this review was to examine the importance of placental responses to changing oxygen environments during abnormal pregnancy in terms of cellular, molecular and functional changes in order to highlight new therapeutic pathways, and to pinpoint approaches aimed at enhancing oxygen supply and/or mitigating oxidative stress in the placenta as a mean of optimizing fetal growth. SEARCH METHODS An extensive online search of peer-reviewed articles using PubMed was performed with combinations of search terms including pregnancy, placenta, trophoblast, oxygen, hypoxia, high altitude, FGR and PE (last updated in May 2020). OUTCOMES Trophoblast differentiation and placental establishment are governed by oxygen availability/hypoxia in early pregnancy. The placental response to late gestational hypoxia includes changes in syncytialization, mitochondrial functions, endoplasmic reticulum stress, hormone production, nutrient handling and angiogenic factor secretion. The nature of these changes depends on the extent of hypoxia, with some responses appearing adaptive and others appearing detrimental to the placental support of fetal growth. Emerging approaches that aim to increase placental oxygen supply and/or reduce the impacts of excessive oxidative stress are promising for their potential to prevent/treat FGR. WIDER IMPLICATIONS There are many risks and challenges of intervening during pregnancy that must be considered. The establishment of human trophoblast stem cell lines and organoids will allow further mechanistic studies of the effects of hypoxia and may lead to advanced screening of drugs for use in pregnancies complicated by placental insufficiency/hypoxia. Since no treatments are currently available, a better understanding of placental adaptations to hypoxia would help to develop therapies or repurpose drugs to optimize placental function and fetal growth, with life-long benefits to human health.
Collapse
Affiliation(s)
- Arthur Colson
- Pole of Obstetrics, Institute of Experimental and Clinical Research (IREC), Université catholique de Louvain, Brussels, Belgium.,Pole of Pharmacology & Therapeutics, Institute of Experimental and Clinical Research (IREC), Université catholique de Louvain, Brussels, Belgium.,Department of Obstetrics, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Pierre Sonveaux
- Pole of Pharmacology & Therapeutics, Institute of Experimental and Clinical Research (IREC), Université catholique de Louvain, Brussels, Belgium
| | - Frédéric Debiève
- Pole of Obstetrics, Institute of Experimental and Clinical Research (IREC), Université catholique de Louvain, Brussels, Belgium.,Department of Obstetrics, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Amanda N Sferruzzi-Perri
- Department of Physiology, Development and Neuroscience, Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
| |
Collapse
|
35
|
Singer D. [Surviving the Lack: Natural Adaptations in Newborns]. Z Geburtshilfe Neonatol 2020; 225:203-215. [PMID: 33285584 DOI: 10.1055/a-1019-6007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Newborns are equipped with a number of natural adaptation mechanisms preventing them from impaired energy supply, despite their elevated (size-related) metabolic rate. These include the diving response known from aquatic mammals, which - being composed of apnea, bradycardia, and vasoconstriction - ensures an economical use of O2 reserves and results in a subsequent influx of lactate out of peripheral tissues. From a metabolic point of view, mammalian fetuses behave "like an organ of the mother" and thus exhibit a hibernation-like deviation from the overall metabolic size relationship that adapts them to the limited intrauterine O2/substrate availability. In case of lacking supply, they can reduce their energy demands even further by foregoing growth, with the placenta acting as a gatekeeper. Postnatal hypoxia does not only result in the suppression of non-shivering thermogenesis, but also in a hypoxic hypometabolism that otherwise has only been known from poikilothermic animals. After prolonged apnea, gasps do occur that maintain a rudimentary heart action through short elevations in pO2 (autoresuscitation). Overall, these mechanisms postpone a critical O2 deficit and thereby provide a "resistance" rather than a "tolerance" to hypoxia. As they are based on an (active) reduction in energy demand, they are not easy to distinguish from the (passive) breakdown of metabolism resulting from hypoxia.
Collapse
|
36
|
Khatami M. Deceptology in cancer and vaccine sciences: Seeds of immune destruction-mini electric shocks in mitochondria: Neuroplasticity-electrobiology of response profiles and increased induced diseases in four generations - A hypothesis. Clin Transl Med 2020; 10:e215. [PMID: 33377661 PMCID: PMC7749544 DOI: 10.1002/ctm2.215] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 10/08/2020] [Accepted: 10/13/2020] [Indexed: 12/16/2022] Open
Abstract
From Rockefeller's support of patent medicine to Gates' patent vaccines, medical establishment invested a great deal in intellectual ignorance. Through the control over medical education and research it has created a public illusion to prop up corporate profit and encouraged the lust for money and power. An overview of data on cancer and vaccine sciences, the status of Americans' health, a survey of repeated failed projects, economic toxicity, and heavy drug consumption or addiction among young and old provide compelling evidence that in the twentieth century nearly all classic disease categories (congenital, inheritance, neonatal, or induced) shifted to increase induced diseases. Examples of this deceptology in ignoring or minimizing, and mocking fundamental discoveries and theories in cancer and vaccine sciences are attacks on research showing that (a), effective immunity is responsible for defending and killing pathogens and defective cancerous cells, correcting and repairing genetic mutations; (b) viruses cause cancer; and (c), abnormal gene mutations are often the consequences of (and secondary to) disturbances in effective immunity. The outcomes of cancer reductionist approaches to therapies reveal failure rates of 90% (+/-5) for solid tumors; loss of over 50 million lives and waste of $30-50 trillions on too many worthless, out-of-focus, and irresponsible projects. Current emphasis on vaccination of public with pathogen-specific vaccines and ingredients seems new terms for drugging young and old. Cumulative exposures to low level carcinogens and environmental hazards or high energy electronic devices (EMF; 5G) are additional triggers to vaccine toxicities (antigen-mitochondrial overload) or "seeds of immune destruction" that create mini electrical shocks (molecular sinks holes) in highly synchronized and regulated immune network that retard time-energy-dependent biorhythms in organs resulting in causes, exacerbations or consequences of mild, moderate or severe immune disorders. Four generations of drug-dependent Americans strongly suggest that medical establishment has practiced decades of intellectual deception through its claims on "war on cancer"; that cancer is 100, 200, or 1000 diseases; identification of "individual" genetic mutations to cure diseases; "vaccines are safe". Such immoral and unethical practices, along with intellectual harassment and bullying, censoring or silencing of independent and competent professionals ("Intellectual Me Too") present grave concerns, far greater compared with the sexual harassment of 'Me Too' movement that was recently spearheaded by NIH. The principal driving forces behind conducting deceptive and illogical medical/cancer and vaccine projects seem to be; (a) huge return of investment and corporate profit for selling drugs and vaccines; (b) maintenance of abusive power over public health; (c) global control of population growth via increased induction of diseases, infertility, decline in life-span, and death. An overview of accidental discoveries that we established and extended since 1980s, on models of acute and chronic ocular inflammatory diseases, provides series of the first evidence for a direct link between inflammation and multistep immune dysfunction in tumorigenesis and angiogenesis. Results are relevant to demonstrate that current emphasis on vaccinating the unborn, newborn, or infant would induce immediate or long-term immune disorders (eg, low birth weight, preterm birth, fatigue, autism, epilepsy/seizures, BBB leakage, autoimmune, neurodegenerative or digestive diseases, obesity, diabetes, cardiovascular problems, or cancers). Vaccination of the unborn is likely to disturb trophoblast-embryo-fetus-placenta biology and orderly growth of embryo-fetus, alter epithelial-mesenchymal transition or constituent-inducible receptors, damage mitochondria, and diverse function of histamine-histidine pathways. Significant increased in childhood illnesses are likely due to toxicities of vaccine and incipient (eg, metals [Al, Hg], detergents, fetal tissue, DNA/RNA) that retard bioenergetics of mitochondria, alter polarization-depolarization balance of tumoricidal (Yin) and tumorigenic (Yang) properties of immunity. Captivated by complex electobiology of immunity, this multidisciplinary perspective is an attempt to initiate identifying bases for increased induction of immune disorders in three to four generations in America. We hypothesize that (a) gene-environment-immune biorhythms parallel neuronal function (brain neuroplasticity) with super-packages of inducible (adaptive or horizontal) electronic signals and (b) autonomic sympathetic and parasympathetic circuitry that shape immunity (Yin-Yang) cannot be explained by limited genomics (innate, perpendicular) that conventionally explain certain inherited diseases (eg, sickle cell anemia, progeria). Future studies should focus on deep learning of complex electrobiology of immunity that requires differential bioenergetics from mitochondria and cytoplasm. Approaches to limit or control excessive activation of gene-environment-immunity are keys to assess accurate disease risk formulations, prevent inducible diseases, and develop universal safe vaccines that promote health, the most basic human right.
Collapse
Affiliation(s)
- Mahin Khatami
- Inflammation, Aging and Cancer, National Cancer Institute (NCI)the National Institutes of Health (NIH) (Retired)BethesdaMarylandUSA
| |
Collapse
|
37
|
Lorca RA, Matarazzo CJ, Bales ES, Houck JA, Orlicky DJ, Euser AG, Julian CG, Moore LG. AMPK activation in pregnant human myometrial arteries from high-altitude and intrauterine growth-restricted pregnancies. Am J Physiol Heart Circ Physiol 2020; 319:H203-H212. [PMID: 32502374 DOI: 10.1152/ajpheart.00644.2019] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
High-altitude (>2,500 m) residence increases the incidence of intrauterine growth restriction (IUGR) due, in part, to reduced uterine artery blood flow and impaired myometrial artery (MA) vasodilator response. A role for the AMP-activated protein kinase (AMPK) pathway in protecting against hypoxia-associated IUGR is suggested by genomic and transcriptomic studies in humans and functional studies in mice. AMPK is a hypoxia-sensitive metabolic sensor with vasodilatory properties. Here we hypothesized that AMPK-dependent vasodilation was increased in MAs from high versus low-altitude (<1,700 m) Colorado women with appropriate for gestational age (AGA) pregnancies and reduced in IUGR pregnancies regardless of altitude. Vasoreactivity studies showed that, in AGA pregnancies, MAs from high-altitude women were more sensitive to vasodilation by activation of AMPK with A769662 due chiefly to increased endothelial nitric oxide production, whereas MA responses to AMPK activation in the low-altitude women were endothelium independent. MAs from IUGR compared with AGA pregnancies had blunted vasodilator responses to acetylcholine at high altitude. We concluded that 1) blunted vasodilator responses in IUGR pregnancies confirm the importance of MA vasodilation for normal fetal growth and 2) the increased sensitivity to AMPK activation in AGA pregnancies at high altitude suggests that AMPK activation helped maintain MA vasodilation and fetal growth. These results highlight a novel mechanism for vasodilation of MAs under conditions of chronic hypoxia and suggest that AMPK activation could provide a therapy for increasing uteroplacental blood flow and improving fetal growth in IUGR pregnancies.NEW & NOTEWORTHY Intrauterine growth restriction (IUGR) impairs infant well- being and increases susceptibility to later-in-life diseases for mother and child. Our study reveals a novel role for AMPK in vasodilating the myometrial artery (MA) from women residing at high altitude (>2,500 m) with appropriate for gestational age pregnancies but not in IUGR pregnancies at any altitude.
Collapse
Affiliation(s)
- Ramón A Lorca
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Denver-Anschutz Medical Campus, Aurora, Colorado
| | - Christopher J Matarazzo
- Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, University of Colorado Denver-Anschutz Medical Campus, Aurora, Colorado
| | - Elise S Bales
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Denver-Anschutz Medical Campus, Aurora, Colorado
| | - Julie A Houck
- Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, University of Colorado Denver-Anschutz Medical Campus, Aurora, Colorado
| | - David J Orlicky
- Department of Pathology, University of Colorado Denver-Anschutz Medical Campus, Aurora, Colorado
| | - Anna G Euser
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, University of Colorado Denver-Anschutz Medical Campus, Aurora, Colorado
| | - Colleen G Julian
- Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, University of Colorado Denver-Anschutz Medical Campus, Aurora, Colorado
| | - Lorna G Moore
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Denver-Anschutz Medical Campus, Aurora, Colorado
| |
Collapse
|
38
|
Sun C, Groom KM, Oyston C, Chamley LW, Clark AR, James JL. The placenta in fetal growth restriction: What is going wrong? Placenta 2020; 96:10-18. [PMID: 32421528 DOI: 10.1016/j.placenta.2020.05.003] [Citation(s) in RCA: 127] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 04/17/2020] [Accepted: 05/07/2020] [Indexed: 02/06/2023]
Abstract
The placenta is essential for the efficient delivery of nutrients and oxygen from mother to fetus to maintain normal fetal growth. Dysfunctional placental development underpins many pregnancy complications, including fetal growth restriction (FGR) a condition in which the fetus does not reach its growth potential. The FGR placenta is smaller than normal placentae throughout gestation and displays maldevelopment of both the placental villi and the fetal vasculature within these villi. Specialized epithelial cells called trophoblasts exhibit abnormal function and development in FGR placentae. This includes an altered balance between proliferation and apoptotic death, premature cellular senescence, and reduced colonisation of the maternal decidual tissue. Thus, the placenta undergoes aberrant changes at the macroscopic to cellular level in FGR, which can limit exchange capacity and downstream fetal growth. This review aims to compile stereological, in vitro, and imaging data to create a holistic overview of the FGR placenta and its pathophysiology, with a focus on the contribution of trophoblasts.
Collapse
Affiliation(s)
- Cherry Sun
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, The University of Auckland, 85 Park Road, Grafton, Auckland, 1023, New Zealand.
| | - Katie M Groom
- Liggins Institute, The University of Auckland, 85 Park Road, Grafton, Auckland, 1023, New Zealand
| | - Charlotte Oyston
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, The University of Auckland, 85 Park Road, Grafton, Auckland, 1023, New Zealand
| | - Lawrence W Chamley
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, The University of Auckland, 85 Park Road, Grafton, Auckland, 1023, New Zealand
| | - Alys R Clark
- Auckland Bioengineering Institute, The University of Auckland, Auckland Bioengineering, House, Level 6/70 Symonds Street, Grafton, Auckland, 1010, New Zealand
| | - Joanna L James
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, The University of Auckland, 85 Park Road, Grafton, Auckland, 1023, New Zealand
| |
Collapse
|
39
|
Läsche M, Emons G, Gründker C. Shedding New Light on Cancer Metabolism: A Metabolic Tightrope Between Life and Death. Front Oncol 2020; 10:409. [PMID: 32300553 PMCID: PMC7145406 DOI: 10.3389/fonc.2020.00409] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 03/09/2020] [Indexed: 12/13/2022] Open
Abstract
Since the earliest findings of Otto Warburg, who discovered the first metabolic differences between lactate production of cancer cells and non-malignant tissues in the 1920s, much time has passed. He explained the increased lactate levels with dysfunctional mitochondria and aerobic glycolysis despite adequate oxygenation. Meanwhile, we came to know that mitochondria remain instead functional in cancer cells; hence, metabolic drift, rather than being linked to dysfunctional mitochondria, was found to be an active act of direct response of cancer cells to cell proliferation and survival signals. This metabolic drift begins with the use of sugars and the full oxidative phosphorylation via the mitochondrial respiratory chain to form CO2, and it then leads to the formation of lactic acid via partial oxidation. In addition to oncogene-driven metabolic reprogramming, the oncometabolites themselves alter cell signaling and are responsible for differentiation and metastasis of cancer cells. The aberrant metabolism is now considered a major characteristic of cancer within the past 15 years. However, the proliferating anabolic growth of a tumor and its spread to distal sites of the body is not explainable by altered glucose metabolism alone. Since a tumor consists of malignant cells and its tumor microenvironment, it was important for us to understand the bilateral interactions between the primary tumor and its microenvironment and the processes underlying its successful metastasis. We here describe the main metabolic pathways and their implications in tumor progression and metastasis. We also portray that metabolic flexibility determines the fate of the cancer cell and ultimately the patient. This flexibility must be taken into account when deciding on a therapy, since singular cancer therapies only shift the metabolism to a different alternative path and create resistance to the medication used. As with Otto Warburg in his days, we primarily focused on the metabolism of mitochondria when dealing with this scientific question.
Collapse
Affiliation(s)
- Matthias Läsche
- Department of Gynecology and Obstetrics, University Medicine Göttingen, Göttingen, Germany
| | - Günter Emons
- Department of Gynecology and Obstetrics, University Medicine Göttingen, Göttingen, Germany
| | - Carsten Gründker
- Department of Gynecology and Obstetrics, University Medicine Göttingen, Göttingen, Germany
| |
Collapse
|
40
|
Elkin ER, Harris SM, Su AL, Lash LH, Loch-Caruso R. Placenta as a target of trichloroethylene toxicity. ENVIRONMENTAL SCIENCE. PROCESSES & IMPACTS 2020; 22:472-486. [PMID: 32022077 PMCID: PMC7103546 DOI: 10.1039/c9em00537d] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Trichloroethylene (TCE) is an industrial solvent and a common environmental contaminant detected in thousands of hazardous waste sites. Risk of exposure is a concern for workers in occupations that use TCE as well as for residents who live near industries that use TCE or who live near TCE-contaminated sites. Although renal, hepatic and carcinogenic effects of TCE have been documented, less is known about TCE impacts on reproductive functions despite epidemiology reports associating maternal TCE exposure with adverse pregnancy outcomes. Toxicological evidence suggests that the placenta mediates at least some of the adverse pregnancy outcomes associated with TCE exposure. Toxicology studies show that the TCE metabolite, S-(1,2-dichlorovinyl)-l-cysteine (DCVC) generates toxic effects such as mitochondrial dysfunction, apoptosis, oxidative stress, and release of prostaglandins and pro-inflammatory cytokines in placental cell lines. Each of these mechanisms of toxicity have significant implications for placental functions and, thus, ultimately the health of mother and developing child. Despite these findings there remain significant gaps in our knowledge about effects of TCE on the placenta, including effects on specific placental cell types and functions as well as sex differences in response to TCE exposure. Due to the critical role that the placenta plays in pregnancy, future research addressing some of these knowledge gaps could lead to significant gains in public health.
Collapse
Affiliation(s)
- Elana R Elkin
- Department of Environmental Health Sciences, University of Michigan, Ann Arbor, MI 48109-2029, USA.
| | | | | | | | | |
Collapse
|
41
|
Preeclampsia: The Interplay Between Oxygen-Sensitive miRNAs and Erythropoietin. J Clin Med 2020; 9:jcm9020574. [PMID: 32093169 PMCID: PMC7073952 DOI: 10.3390/jcm9020574] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 02/14/2020] [Accepted: 02/18/2020] [Indexed: 12/21/2022] Open
Abstract
Changes in the oxygen partial pressure caused by a violation of uteroplacental perfusion are considered a powerful inducer of a cascade of reactions leading to the clinical manifestation of preeclampsia (PE). At the same time, the induction of oxygen-dependent molecule expression, in particular, miRNA and erythropoietin, is modulated. Therefore, the focus of our study was aimed at estimating the miRNA expression profile of placental tissue and blood plasma in pregnant women with preeclampsia using deep sequencing and quantitative RT-PCR, as well as determining the concentration of erythropoietin. The expression of miR-27b-3p, miR-92b-3p, miR-125b-5p, miR-181a-5p, and miR-186-5p, as regulated by hypoxia/reoxygenation, was significantly increased in blood plasma during early-onset preeclampsia. The possibility of detecting early PE according to the logistic regression model (miR-92b-3p, miR-125b-5p, and miR-181a-5p (AUC = 0.91)) was evaluated. Furthermore, the erythropoietin level, which is regulated by miR-125b-5p, was significantly increased. According to PANTHER14.1, the participation of these miRNAs in the regulation of pathways, such as the hypoxia’s response via HIF activation, oxidative stress response, angiogenesis, and the VEGF signaling pathway, were determined.
Collapse
|
42
|
Elkin ER, Bridges D, Harris SM, Loch-Caruso RK. Exposure to Trichloroethylene Metabolite S-(1,2-Dichlorovinyl)-L-cysteine Causes Compensatory Changes to Macronutrient Utilization and Energy Metabolism in Placental HTR-8/SVneo Cells. Chem Res Toxicol 2020; 33:1339-1355. [PMID: 31951115 PMCID: PMC7299793 DOI: 10.1021/acs.chemrestox.9b00356] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
![]()
Trichloroethylene
(TCE) is a widespread environmental contaminant
following decades of use as an industrial solvent, improper disposal,
and remediation challenges. Consequently, TCE exposure continues to
constitute a risk to human health. Despite epidemiological evidence
associating exposure with adverse birth outcomes, the effects of TCE
and its metabolite S-(1, 2-dichlorovinyl)-L-cysteine
(DCVC) on the placenta remain undetermined. Flexible and efficient
macronutrient and energy metabolism pathway utilization is essential
for placental cell physiological adaptability. Because DCVC is known
to compromise cellular energy status and disrupt energy metabolism
in renal proximal tubular cells, this study investigated the effects
of DCVC on cellular energy status and energy metabolism pathways in
placental cells. Human extravillous trophoblast cells, HTR-8/SVneo,
were exposed to 5–20 μM DCVC for 6 or 12 h. After establishing
concentration and exposure duration thresholds for DCVC-induced cytotoxicity,
targeted metabolomics was used to evaluate overall energy status and
metabolite concentrations from energy metabolism pathways. The data
revealed glucose metabolism perturbations including a time-dependent
accumulation of glucose-6-phosphate+frutose-6-phosphate (G6P+F6P)
as well as independent shunting of glucose intermediates that diminished
with time, with modest energy status decline but in the absence of
significant changes in ATP concentrations. Furthermore, metabolic
profiling suggested that DCVC stimulated compensatory utilization
of glycerol, lipid, and amino acid metabolism to provide intermediate
substrates entering downstream in the glycolytic pathway or the tricarboxylic
acid cycle. Lastly, amino acid deprivation increased susceptibility
to DCVC-induced cytotoxicity. Taken together, these results suggest
that DCVC caused metabolic perturbations necessitating adaptations
in macronutrient and energy metabolism pathway utilization to maintain
adequate ATP levels.
Collapse
Affiliation(s)
- Elana R Elkin
- Department of Environmental Health Sciences, University of Michigan, Ann Arbor, Michigan 48109-2029, United States
| | - Dave Bridges
- Department of Nutritional Sciences, University of Michigan, Ann Arbor, Michigan 48109-2029, United States
| | - Sean M Harris
- Department of Environmental Health Sciences, University of Michigan, Ann Arbor, Michigan 48109-2029, United States
| | - Rita Karen Loch-Caruso
- Department of Environmental Health Sciences, University of Michigan, Ann Arbor, Michigan 48109-2029, United States
| |
Collapse
|
43
|
Llurba Olive E, Xiao E, Natale DR, Fisher SA. Oxygen and lack of oxygen in fetal and placental development, feto-placental coupling, and congenital heart defects. Birth Defects Res 2019; 110:1517-1530. [PMID: 30576091 DOI: 10.1002/bdr2.1430] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 11/12/2018] [Indexed: 12/19/2022]
Abstract
Low oxygen concentration (hypoxia) is part of normal embryonic development, yet the situation is complex. Oxygen (O2 ) is a janus gas with low levels signaling through hypoxia-inducible transcription factor (HIF) that are required for development of fetal and placental vasculature and fetal red blood cells. This results in coupling of fetus and mother around midgestation as a functional feto-placental unit (FPU) for O2 transport, which is required for continued growth and development of the fetus. Defects in these processes may leave the developing fetus vulnerable to O2 deprivation or other stressors during this critical midgestational transition when common septal and conotruncal heart defects (CHDs) are likely to arise. Recent human epidemiological and case-control studies support an association between placental dysfunction, manifest as early onset pre-eclampsia (PE) and increased serum bio-markers, and CHD. Animal studies support this association, in particular those using gene inactivation in the mouse. Sophisticated methods for gene inactivation, cell fate mapping, and a quantitative bio-reporter of O2 concentration support the premise that hypoxic stress at critical stages of development leads to CHD. The secondary heart field contributing to the cardiac outlet is a key target, with activation of the un-folded protein response and abrogation of FGF signaling or precocious activation of a cardiomyocyte transcriptional program for differentiation, suggested as mechanisms. These studies provide a strong foundation for further study of feto-placental coupling and hypoxic stress in the genesis of human CHD.
Collapse
Affiliation(s)
- Elisa Llurba Olive
- Director of the Obstetrics and Gynecology Department, Sant Pau University Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain.,Maternal and Child Health and Development Network II (SAMID II) RD16/0022, Institute of Health Carlos III, Madrid, Spain
| | - Emily Xiao
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland.,Department of Pediatrics, University of Maryland School of Medicine, Baltimore, Maryland
| | - David R Natale
- Department of Obstetrics and Gynecology and Reproductive Sciences, University of California San Diego, San Diego, California
| | - Steven A Fisher
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland.,Department of Physiology and Biophysics, University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
44
|
Hart B, Morgan E, Alejandro EU. Nutrient sensor signaling pathways and cellular stress in fetal growth restriction. J Mol Endocrinol 2019; 62:R155-R165. [PMID: 30400060 PMCID: PMC6443503 DOI: 10.1530/jme-18-0059] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 10/09/2018] [Indexed: 12/24/2022]
Abstract
Fetal growth restriction is one of the most common obstetrical complications resulting in significant perinatal morbidity and mortality. The most frequent etiology of human singleton fetal growth restriction is placental insufficiency, which occurs secondary to reduced utero-placental perfusion, abnormal placentation, impaired trophoblast invasion and spiral artery remodeling, resulting in altered nutrient and oxygen transport. Two nutrient-sensing proteins involved in placental development and glucose and amino acid transport are mechanistic target of rapamycin (mTOR) and O-linked N-acetylglucosamine transferase (OGT), which are both regulated by availability of oxygen. Impairment in either of these pathways is associated with fetal growth restriction and accompanied by cellular stress in the forms of hypoxia, oxidative and endoplasmic reticulum (ER) stress, metabolic dysfunction and nutrient starvation in the placenta. Recent evidence has emerged regarding the potential impact of nutrient sensors on fetal stress response, which occurs in a sexual dysmorphic manner, indicating a potential element of genetic gender susceptibility to fetal growth restriction. In this mini review, we focus on the known role of mTOR and OGT in placental development, nutrient regulation and response to cellular stress in human fetal growth restriction with supporting evidence from rodent models.
Collapse
Affiliation(s)
- Bethany Hart
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Women's Health, University of Minnesota, Minneapolis, Minnesota, USA
| | - Elizabeth Morgan
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Women's Health, University of Minnesota, Minneapolis, Minnesota, USA
| | - Emilyn U Alejandro
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
45
|
Placental mitochondria adapt developmentally and in response to hypoxia to support fetal growth. Proc Natl Acad Sci U S A 2019; 116:1621-1626. [PMID: 30655345 DOI: 10.1073/pnas.1816056116] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Mitochondria respond to a range of stimuli and function in energy production and redox homeostasis. However, little is known about the developmental and environmental control of mitochondria in the placenta, an organ vital for fetal growth and pregnancy maintenance in eutherian mammals. Using respirometry and molecular analyses, the present study examined mitochondrial function in the distinct transport and endocrine zones of the mouse placenta during normal pregnancy and maternal inhalation hypoxia. The data show that mitochondria of the two zones adopt different strategies in modulating their respiration, substrate use, biogenesis, density, and efficiency to best support the growth and energy demands of fetoplacental tissues during late gestation in both normal and hypoxic conditions. The findings have important implications for environmentally induced adaptations in mitochondrial function in other tissues and for compromised human pregnancy in which hypoxia and alterations in placental mitochondrial function are associated with poor outcomes like fetal growth restriction.
Collapse
|
46
|
Song H, Telugu BP, Thompson LP. Sexual dimorphism of mitochondrial function in the hypoxic guinea pig placenta. Biol Reprod 2019; 100:208-216. [PMID: 30085007 PMCID: PMC6335207 DOI: 10.1093/biolre/ioy167] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 04/16/2018] [Accepted: 07/30/2018] [Indexed: 12/13/2022] Open
Abstract
Placental hypoxia can stimulate oxidative stress and mitochondrial dysfunction reducing placental efficiency and inducing fetal growth restriction (FGR). We hypothesized that chronic hypoxia inhibits mitochondrial function in the placenta as an underlying cause of cellular mechanisms contributing to FGR. Pregnant guinea pigs were exposed to either normoxia (NMX) or hypoxia (HPX; 10.5% O2) at 25 day gestation until term (65 day). Guinea pigs were anesthetized, and fetuses and placentas were excised at either mid (40 day) or late gestation (64 day), weighed, and placental tissue stored at -80°C until assayed. Mitochondrial DNA content, protein expression of respiratory Complexes I-V, and nitration and activity rates of Complexes I and IV were measured in NMX and HPX male (N = 6 in each treatment) and female (N = 6 in each treatment) placentas. Mitochondrial density was not altered by HPX in either mid- or late-term placentas. In mid gestation, HPX slightly increased expression of Complexes I-III and V in male placentas only, but had no effect on either Complex I or IV activity rates or nitrotyrosine expression. In late gestation, HPX significantly decreased CI/CIV activity rates and increased CI/CIV nitration in male but not female placentas exhibiting a sexual dimorphism. Complex I-V expression was reduced from mid to late gestation in both male and female placentas regardless of treatment. We conclude that chronic HPX decreases mitochondrial function by inhibiting Complex I/IV activity via increased peroxynitrite in a sex-related manner. Further, there may be a progressive decrease in energy metabolism of placental cell types with gestation that increases the vulnerability of placental function to intrauterine stress.
Collapse
Affiliation(s)
- Hong Song
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Bhanu P Telugu
- Animal Biosciences and Biotechnology Laboratory, USDA-ARS, Beltsville, Maryland, USA
- Animal and Avian Science, University of Maryland, College Park, Maryland, USA
| | - Loren P Thompson
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
47
|
Guo L, Liu Y, Guo Y, Yang Y, Chen B. MicroRNA-423-5p inhibits the progression of trophoblast cells via targeting IGF2BP1. Placenta 2018; 74:1-8. [PMID: 30587375 DOI: 10.1016/j.placenta.2018.12.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 11/14/2018] [Accepted: 12/06/2018] [Indexed: 01/25/2023]
Abstract
INTRODUCTION Preeclampsia (PE) is one of the leading causes of maternal and fetal mortality globally. The imbalance of trophoblast homeostasis is closely linked with the pathogenesis of PE. MicroRNA-423-5p (miR-423-5p) has been reported to be abnormally expressed in placenta and blood plasma of pregnant women with PE. In the present study, miR-423-5p expression in blood plasma of pregnant women with PE and healthy pregnant women was detected. Also, the roles and molecular mechanisms of miR-423-5p in the development of trophoblast cells were further investigated. METHODS Expression of miR-423-5p and insulin like growth factor 2 mRNA binding protein 1 (IGF2BP1) mRNA was detected by RT-qPCR assay. Protein expression of IGF2BP1, Bcl-2 and Bax was determined using western blot assay. Cell migratory and invasive capacities were assessed by transwell migration and invasion assay. Cell apoptotic rate was determined using flow cytometry via the double-staining of Annexin V-FITC/Propidium Iodide. The interaction between miR-423-5p and IGF2BP1 was demonstrated by bioinformatics analysis and luciferase reporter assay. RESULTS MiR-423-5p was highly expressed in blood plasma of pregnant women with PE. MiR-423-5p inhibited migration, invasion and proliferation as well as induced apoptosis in HTR-8/SVneo cells. Further investigation revealed that IGF2BP1 was a target of miR-423-5p. Moreover, IGF2BP1 overexpression promoted migration, invasion and proliferation, suppressed apoptosis, and weakened miR-423-5p function in HTR-8/SVneo cells. DISCUSSION MiR-423-5p inhibited migration, invasion and proliferation as well as induced apoptosis by targeting IGF2BP1 in HTR-8/SVneo cells, presenting a novel molecular basis implicated in PE pathogenesis.
Collapse
Affiliation(s)
- Li Guo
- Department of Obstetrics and Gynecology, Xijing Hospital,the Military Medical University of PLA Airforce (Fourth Military Medical University), China; Department of Obstetrics, 215 Hospital of Shaanxi Nuclear Industry, China
| | - Yu Liu
- Department of Obstetrics and Gynecology, Xijing Hospital,the Military Medical University of PLA Airforce (Fourth Military Medical University), China
| | - Ying Guo
- Department of Obstetrics and Gynecology, Xijing Hospital,the Military Medical University of PLA Airforce (Fourth Military Medical University), China
| | - Yongkang Yang
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Shaanxi University of Chinese Medicine, China.
| | - Biliang Chen
- Department of Obstetrics and Gynecology, Xijing Hospital,the Military Medical University of PLA Airforce (Fourth Military Medical University), China.
| |
Collapse
|
48
|
Nalivaeva NN, Turner AJ, Zhuravin IA. Role of Prenatal Hypoxia in Brain Development, Cognitive Functions, and Neurodegeneration. Front Neurosci 2018; 12:825. [PMID: 30510498 PMCID: PMC6254649 DOI: 10.3389/fnins.2018.00825] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 10/22/2018] [Indexed: 12/15/2022] Open
Abstract
This review focuses on the role of prenatal hypoxia in the development of brain functions in the postnatal period and subsequent increased risk of neurodegenerative disorders in later life. Accumulating evidence suggests that prenatal hypoxia in critical periods of brain formation results in significant changes in development of cognitive functions at various stages of postnatal life which correlate with morphological changes in brain structures involved in learning and memory. Prenatal hypoxia also leads to a decrease in brain adaptive potential and plasticity due to the disturbance in the process of formation of new contacts between cells and propagation of neuronal stimuli, especially in the cortex and hippocampus. On the other hand, prenatal hypoxia has a significant impact on expression and processing of a variety of genes involved in normal brain function and their epigenetic regulation. This results in changes in the patterns of mRNA and protein expression and their post-translational modifications, including protein misfolding and clearance. Among proteins affected by prenatal hypoxia are a key enzyme of the cholinergic system-acetylcholinesterase, and the amyloid precursor protein (APP), both of which have important roles in brain function. Disruption of their expression and metabolism caused by prenatal hypoxia can also result, apart from early cognitive dysfunctions, in development of neurodegeneration in later life. Another group of enzymes affected by prenatal hypoxia are peptidases involved in catabolism of neuropeptides, including amyloid-β peptide (Aβ). The decrease in the activity of neprilysin and other amyloid-degrading enzymes observed after prenatal hypoxia could result over the years in an Aβ clearance deficit and accumulation of its toxic species which cause neuronal cell death and development of neurodegeneration. Applying various approaches to restore expression of neuronal genes disrupted by prenatal hypoxia during postnatal development opens an avenue for therapeutic compensation of cognitive dysfunctions and prevention of Aβ accumulation in the aging brain and the model of prenatal hypoxia in rodents can be used as a reliable tool for assessment of their efficacy.
Collapse
Affiliation(s)
- Natalia N. Nalivaeva
- I. M. Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, Russia
- Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, United Kingdom
| | - Anthony J. Turner
- Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, United Kingdom
| | - Igor A. Zhuravin
- I. M. Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, Russia
- Research Centre, Saint-Petersburg State Pediatric Medical University, St. Petersburg, Russia
| |
Collapse
|
49
|
Keshavarzi F, Mehdizadeh S, Khazaie H, Ghadami MR. Objective assessment of obstructive sleep apnea in normal pregnant and preeclamptic women. Hypertens Pregnancy 2018; 37:154-159. [DOI: 10.1080/10641955.2018.1498879] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Farahnaz Keshavarzi
- Department of obstetrics and gynecology, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Shervin Mehdizadeh
- Department of obstetrics and gynecology, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Habibolah Khazaie
- Sleep Disorders Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mohammad Rasoul Ghadami
- Sleep Disorders Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
50
|
Khatami M. Cancer; an induced disease of twentieth century! Induction of tolerance, increased entropy and 'Dark Energy': loss of biorhythms (Anabolism v. Catabolism). Clin Transl Med 2018; 7:20. [PMID: 29961900 PMCID: PMC6026585 DOI: 10.1186/s40169-018-0193-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2018] [Accepted: 05/29/2018] [Indexed: 12/15/2022] Open
Abstract
Maintenance of health involves a synchronized network of catabolic and anabolic signals among organs/tissues/cells that requires differential bioenergetics from mitochondria and glycolysis (biological laws or biorhythms). We defined biological circadian rhythms as Yin (tumoricidal) and Yang (tumorigenic) arms of acute inflammation (effective immunity) involving immune and non-immune systems. Role of pathogens in altering immunity and inducing diseases and cancer has been documented for over a century. However, in 1955s decision makers in cancer/medical establishment allowed public (current baby boomers) to consume million doses of virus-contaminated polio vaccines. The risk of cancer incidence and mortality sharply rose from 5% (rate of hereditary/genetic or innate disease) in 1900s, to its current scary status of 33% or 50% among women and men, respectively. Despite better hygiene, modern detection technologies and discovery of antibiotics, baby boomers and subsequent 2-3 generations are sicker than previous generations at same age. American health status ranks last among other developed nations while America invests highest amount of resources for healthcare. In this perspective we present evidence that cancer is an induced disease of twentieth century, facilitated by a great deception of cancer/medical establishment for huge corporate profits. Unlike popularized opinions that cancer is 100, 200 or 1000 diseases, we demonstrate that cancer is only one disease; the severe disturbances in biorhythms (differential bioenergetics) or loss of balance in Yin and Yang of effective immunity. Cancer projects that are promoted and funded by decision makers are reductionist approaches, wrong and unethical and resulted in loss of millions of precious lives and financial toxicity to society. Public vaccination with pathogen-specific vaccines (e.g., flu, hepatitis, HPV, meningitis, measles) weakens, not promotes, immunity. Results of irresponsible projects on cancer sciences or vaccines are increased population of drug-dependent sick society. Outcome failure rates of claimed 'targeted' drugs, 'precision' or 'personalized' medicine are 90% (± 5) for solid tumors. We demonstrate that aging, frequent exposures to environmental hazards, infections and pathogen-specific vaccines and ingredients are 'antigen overload' for immune system, skewing the Yin and Yang response profiles and leading to induction of 'mild', 'moderate' or 'severe' immune disorders. Induction of decoy or pattern recognition receptors (e.g., PRRs), such as IRAK-M or IL-1dRs ('designer' molecules) and associated genomic instability and over-expression of growth promoting factors (e.g., pyruvate kinases, mTOR and PI3Ks, histamine, PGE2, VEGF) could lead to immune tolerance, facilitating cancer cells to hijack anabolic machinery of immunity (Yang) for their increased growth requirements. Expression of constituent embryonic factors would negatively regulate differentiation of tumor cells through epithelial-mesenchymal-transition and create "dual negative feedback loop" that influence tissue metabolism under hypoxic conditions. It is further hypothesized that induction of tolerance creates 'dark energy' and increased entropy and temperature in cancer microenvironment allowing disorderly cancer proliferation and mitosis along with increased glucose metabolism via Crabtree and Pasteur Effects, under mitophagy and ribophagy, conditions that are toxic to host survival. Effective translational medicine into treatment requires systematic and logical studies of complex interactions of tumor cells with host environment that dictate clinical outcomes. Promoting effective immunity (biological circadian rhythms) are fundamental steps in correcting host differential bioenergetics and controlling cancer growth, preventing or delaying onset of diseases and maintaining public health. The author urges independent professionals and policy makers to take a closer look at cancer dilemma and stop the 'scientific/medical ponzi schemes' of a powerful group that control a drug-dependent sick society before all hopes for promoting public health evaporate.
Collapse
Affiliation(s)
- Mahin Khatami
- Inflammation, Aging and Cancer, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA.
| |
Collapse
|