1
|
Afrose D, Alfonso-Sánchez S, McClements L. Targeting oxidative stress in preeclampsia. Hypertens Pregnancy 2025; 44:2445556. [PMID: 39726411 DOI: 10.1080/10641955.2024.2445556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Accepted: 12/16/2024] [Indexed: 12/28/2024]
Abstract
Preeclampsia is a complex condition characterized by elevated blood pressure and organ damage involving kidneys or liver, resulting in significant morbidity and mortality for both the mother and the fetus. Increasing evidence suggests that oxidative stress, often caused by mitochondrial dysfunction within fetal trophoblast cells may play a major role in the development and progression of preeclampsia. Oxidative stress occurs as a result of an imbalance between the production of reactive oxygen species (ROS) and the capacity of antioxidant defenses, which can lead to placental cellular damage and endothelial cell dysfunction. Targeting oxidative stress appears to be a promising therapeutic approach that has the potential to improve both short- and long-term maternal and fetal outcomes, thus reducing the global burden of preeclampsia. The purpose of this review is to provide a comprehensive account of the mechanisms of oxidative stress in preeclampsia. Furthermore, it also examines potential interventions for reducing oxidative stress in preeclampsia, including natural antioxidant supplements, lifestyle modifications, mitochondrial targeting antioxidants, and pharmacological agents.A better understanding of the mechanism of action of proposed therapeutic strategies targeting oxidative stress is essential for the identification of companion biomarkers and personalized medicine approaches for the development of effective treatments of preeclampsia.
Collapse
Affiliation(s)
- Dinara Afrose
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Sofía Alfonso-Sánchez
- School of Biomedical Engineering, Faculty of Engineering and Information Technology, University of Technology Sydney, Sydney, NSW, Australia
| | - Lana McClements
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
- Institute for Biomedical Materials and Devices, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| |
Collapse
|
2
|
Viana-Mattioli S, Fonseca-Alaniz MH, Pinheiro-de-Sousa I, Junior RR, Mastella MH, de Carvalho Cavalli R, Sandrim VC. Plasma from hypertensive pregnancy patients induce endothelial dysfunction even under atheroprotective shear stress. Sci Rep 2025; 15:4675. [PMID: 39920219 PMCID: PMC11805971 DOI: 10.1038/s41598-025-88902-8] [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: 06/29/2024] [Accepted: 01/31/2025] [Indexed: 02/09/2025] Open
Abstract
Preeclampsia (PE) is a challenge in maternal healthcare due to its complex nature, characterized by high blood pressure, protein in the urine, and damage to various organs. There is evidence linking PE to endothelial dysfunction (ED), triggered by substances released from an oxygen-deprived placenta. Previous in vitro studies have not considered the impact of in vivo elements, such as the different patterns of blood flow, and laminar (LSS) vs. oscillatory (OSS) shear stress, on the development of ED. We investigated the impact of plasma from healthy pregnant women (HP), subjects with gestational hypertension (GH), and PE patients on global gene expression of human coronary endothelial cells (HCAECs) under LSS and OSS. Our findings revealed a unique transcriptional profile of endothelial cells induced by plasma incubation in LSS. Notably, OSS resulted in similar transcriptomes irrespective of plasma treatment. Under LSS, GH plasma resulted in a proliferative profile, whereas PE plasma was linked to pro-inflammatory and antioxidant profiles compared to HP plasma. Our findings demonstrate that shear stress levels influence the endothelial cell transcriptome in response to plasma from hypertensive pregnancy patients. Both PE and GH can induce endothelial dysfunction under atheroprotective LSS, with a more significant effect observed with PE-derived plasma.
Collapse
Affiliation(s)
- Sarah Viana-Mattioli
- Department of Biophysics and Pharmacology, Institute of Biosciences of Botucatu, Universidade Estadual Paulista (UNESP), Distrito Rubião Júnior, Botucatu, São Paulo, SP, Brazil
- Laboratorio de Genetica e Cardiologia Molecular, Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Miriam Helena Fonseca-Alaniz
- Laboratorio de Genetica e Cardiologia Molecular, Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Iguaracy Pinheiro-de-Sousa
- Laboratorio de Genetica e Cardiologia Molecular, Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton, UK
| | - Ricardo Rosa Junior
- Laboratorio de Genetica e Cardiologia Molecular, Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Moises Henrique Mastella
- Department of Biophysics and Pharmacology, Institute of Biosciences of Botucatu, Universidade Estadual Paulista (UNESP), Distrito Rubião Júnior, Botucatu, São Paulo, SP, Brazil
| | - Ricardo de Carvalho Cavalli
- Department of Gynecology and Obstetrics, Hospital das Clínicas, Medical School of Ribeirão Preto, University of São Paulo, São Paulo, SP, Brazil
| | - Valeria Cristina Sandrim
- Department of Biophysics and Pharmacology, Institute of Biosciences of Botucatu, Universidade Estadual Paulista (UNESP), Distrito Rubião Júnior, Botucatu, São Paulo, SP, Brazil.
| |
Collapse
|
3
|
Mischke J, Klein S, Cornberg M, Kraft ARM. MitoTempo treatment as an approach to cure persistent viral infections? Virology 2024; 600:110280. [PMID: 39492087 DOI: 10.1016/j.virol.2024.110280] [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: 06/25/2024] [Revised: 10/24/2024] [Accepted: 10/28/2024] [Indexed: 11/05/2024]
Abstract
Chronic viral infections are characterized by exhausted virus-specific T cells. Exhaustion is associated with mitochondrial dysfunction, revealing a possible target for treatment. Targeting these metabolic processes may interfere with the exhaustion process of immune cells during infection. It has been shown that the mitochondria-targeted antioxidant MitoTempo could restore hepatitis-B-virus-specific T cells in vitro. Thus, we investigated MitoTempo as a treatment option using the chronic lymphocytic choriomeningitis virus (LCMVcl13) mouse model. MitoTempo treatment of chronically LCMVcl13 infected mice resulted in a transient reduction of LCMV titer. However, no obvious restoration of functional LCMV-specific T cells was observed, beside subtle changes in phenotype of GP33- and NP205-specific T cells. However, these changes did not translate into significantly more functional responses. Our study showed a transient antiviral effect of MitoTempo, but no profound effect on exhausted T cell responses, although further studies are needed to further elucidate the mechanism and use of MitoTempo.
Collapse
Affiliation(s)
- Jasmin Mischke
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany; Twincore, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research (HZI) and the Hannover Medical School, Hannover, Germany; German Center for Infection Research (DZIF), Partner-site Hannover-Braunschweig, Hannover, Germany; Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz Centre for Infection Research (HZI) and Hannover Medical School (MHH), Hannover, Germany
| | - Sebastian Klein
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany; Twincore, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research (HZI) and the Hannover Medical School, Hannover, Germany; German Center for Infection Research (DZIF), Partner-site Hannover-Braunschweig, Hannover, Germany; Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz Centre for Infection Research (HZI) and Hannover Medical School (MHH), Hannover, Germany
| | - Markus Cornberg
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany; Twincore, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research (HZI) and the Hannover Medical School, Hannover, Germany; German Center for Infection Research (DZIF), Partner-site Hannover-Braunschweig, Hannover, Germany; Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz Centre for Infection Research (HZI) and Hannover Medical School (MHH), Hannover, Germany
| | - Anke R M Kraft
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany; Twincore, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research (HZI) and the Hannover Medical School, Hannover, Germany; German Center for Infection Research (DZIF), Partner-site Hannover-Braunschweig, Hannover, Germany; Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz Centre for Infection Research (HZI) and Hannover Medical School (MHH), Hannover, Germany.
| |
Collapse
|
4
|
Suksai M, Romero R, Bosco M, Gotsch F, Jung E, Chaemsaithong P, Tarca AL, Gudicha DW, Gomez-Lopez N, Arenas-Hernandez M, Meyyazhagan A, Grossman LI, Aras S, Chaiworapongsa T. A mitochondrial regulator protein, MNRR1, is elevated in the maternal blood of women with preeclampsia. J Matern Fetal Neonatal Med 2024; 37:2297158. [PMID: 38220225 DOI: 10.1080/14767058.2023.2297158] [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: 06/29/2023] [Accepted: 12/15/2023] [Indexed: 01/16/2024]
Abstract
OBJECTIVE Preeclampsia, one of the most serious obstetric complications, is a heterogenous disorder resulting from different pathologic processes. However, placental oxidative stress and an anti-angiogenic state play a crucial role. Mitochondria are a major source of cellular reactive oxygen species. Abnormalities in mitochondrial structures, proteins, and functions have been observed in the placentae of patients with preeclampsia, thus mitochondrial dysfunction has been implicated in the mechanism of the disease. Mitochondrial nuclear retrograde regulator 1 (MNRR1) is a newly characterized bi-organellar protein with pleiotropic functions. In the mitochondria, this protein regulates cytochrome c oxidase activity and reactive oxygen species production, whereas in the nucleus, it regulates the transcription of a number of genes including response to tissue hypoxia and inflammatory signals. Since MNRR1 expression changes in response to hypoxia and to an inflammatory signal, MNRR1 could be a part of mitochondrial dysfunction and involved in the pathologic process of preeclampsia. This study aimed to determine whether the plasma MNRR1 concentration of women with preeclampsia differed from that of normal pregnant women. METHODS This retrospective case-control study included 97 women with preeclampsia, stratified by gestational age at delivery into early (<34 weeks, n = 40) and late (≥34 weeks, n = 57) preeclampsia and by the presence or absence of placental lesions consistent with maternal vascular malperfusion (MVM), the histologic counterpart of an anti-angiogenic state. Women with an uncomplicated pregnancy at various gestational ages who delivered at term served as controls (n = 80) and were further stratified into early (n = 25) and late (n = 55) controls according to gestational age at venipuncture. Maternal plasma MNRR1 concentrations were determined by an enzyme-linked immunosorbent assay. RESULTS 1) Women with preeclampsia at the time of diagnosis (either early or late disease) had a significantly higher median (interquartile range, IQR) plasma MNRR1 concentration than the controls [early preeclampsia: 1632 (924-2926) pg/mL vs. 630 (448-4002) pg/mL, p = .026, and late preeclampsia: 1833 (1441-5534) pg/mL vs. 910 (526-6178) pg/mL, p = .021]. Among women with early preeclampsia, those with MVM lesions in the placenta had the highest median (IQR) plasma MNRR1 concentration among the three groups [with MVM: 2066 (1070-3188) pg/mL vs. without MVM: 888 (812-1781) pg/mL, p = .03; and with MVM vs. control: 630 (448-4002) pg/mL, p = .04]. There was no significant difference in the median plasma MNRR1 concentration between women with early preeclampsia without MVM lesions and those with an uncomplicated pregnancy (p = .3). By contrast, women with late preeclampsia, regardless of MVM lesions, had a significantly higher median (IQR) plasma MNRR1 concentration than women in the control group [with MVM: 1609 (1392-3135) pg/mL vs. control: 910 (526-6178), p = .045; and without MVM: 2023 (1578-8936) pg/mL vs. control, p = .01]. CONCLUSIONS MNRR1, a mitochondrial regulator protein, is elevated in the maternal plasma of women with preeclampsia (both early and late) at the time of diagnosis. These findings may reflect some degree of mitochondrial dysfunction, intravascular inflammation, or other unknown pathologic processes that characterize this obstetrical syndrome.
Collapse
Affiliation(s)
- Manaphat Suksai
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
- Department of Obstetrics and Gynecology, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand
| | - Roberto Romero
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, USA
| | - Mariachiara Bosco
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
- Department of Obstetrics and Gynecology, AOUI Verona, University of Verona, Verona, Italy
| | - Francesca Gotsch
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Eunjung Jung
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
- Department of Obstetrics and Gynecology, Busan Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea
| | - Piya Chaemsaithong
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
- Department of Obstetrics and Gynecology, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Adi L Tarca
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
- Department of Computer Science, Wayne State University College of Engineering, Detroit, MI, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA
| | - Dereje W Gudicha
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Nardhy Gomez-Lopez
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA
- Department of Biochemistry, Microbiology and Immunology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Marcia Arenas-Hernandez
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Arun Meyyazhagan
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
- Centre of Perinatal and Reproductive Medicine, University of Perugia, Perugia, Italy
| | - Lawrence I Grossman
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA
| | - Siddhesh Aras
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA
| | - Tinnakorn Chaiworapongsa
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| |
Collapse
|
5
|
Manoharan MM, Montes GC, Acquarone M, Swan KF, Pridjian GC, Nogueira Alencar AK, Bayer CL. Metabolic theory of preeclampsia: implications for maternal cardiovascular health. Am J Physiol Heart Circ Physiol 2024; 327:H582-H597. [PMID: 38968164 PMCID: PMC11442029 DOI: 10.1152/ajpheart.00170.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 07/02/2024] [Accepted: 07/02/2024] [Indexed: 07/07/2024]
Abstract
Preeclampsia (PE) is a multisystemic disorder of pregnancy that not only causes perinatal mortality and morbidity but also has a long-term toll on the maternal and fetal cardiovascular system. Women diagnosed with PE are at greater risk for the subsequent development of hypertension, ischemic heart disease, cardiomyopathy, cerebral edema, seizures, and end-stage renal disease. Although PE is considered heterogeneous, inefficient extravillous trophoblast (EVT) migration leading to deficient spiral artery remodeling and increased uteroplacental vascular resistance is the likely initiation of the disease. The principal pathophysiology is placental hypoxia, causing subsequent oxidative stress, leading to mitochondrial dysfunction, mitophagy, and immunological imbalance. The damage imposed on the placenta in turn results in the "stress response" categorized by the dysfunctional release of vasoactive components including oxidative stressors, proinflammatory factors, and cytokines into the maternal circulation. These bioactive factors have deleterious effects on systemic endothelial cells and coagulation leading to generalized vascular dysfunction and hypercoagulability. A better understanding of these metabolic factors may lead to novel therapeutic approaches to prevent and treat this multisystemic disorder. In this review, we connect the hypoxic-oxidative stress and inflammation involved in the pathophysiology of PE to the resulting persistent cardiovascular complications in patients with preeclampsia.
Collapse
Affiliation(s)
- Mistina M Manoharan
- Department of Biomedical Engineering, Tulane University, New Orleans, Louisiana, United States
| | - Guilherme C Montes
- Department of Pharmacology and Psychobiology, Roberto Alcântara Gomes Institute Biology (IBRAG), Rio de Janeiro State University (UERJ), Rio de Janeiro, Brazil
| | - Mariana Acquarone
- Department of Neurology, Tulane University, New Orleans, Louisiana, United States
| | - Kenneth F Swan
- Department of Obstetrics and Gynecology, Tulane University, New Orleans, Louisiana, United States
| | - Gabriella C Pridjian
- Department of Obstetrics and Gynecology, Tulane University, New Orleans, Louisiana, United States
| | | | - Carolyn L Bayer
- Department of Biomedical Engineering, Tulane University, New Orleans, Louisiana, United States
- Department of Obstetrics and Gynecology, Tulane University, New Orleans, Louisiana, United States
| |
Collapse
|
6
|
Toledano JM, Puche-Juarez M, Galvez-Navas JM, Moreno-Fernandez J, Diaz-Castro J, Ochoa JJ. Pregnancy Disorders: A Potential Role for Mitochondrial Altered Homeostasis. Antioxidants (Basel) 2024; 13:979. [PMID: 39199225 PMCID: PMC11351112 DOI: 10.3390/antiox13080979] [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: 06/14/2024] [Revised: 08/02/2024] [Accepted: 08/10/2024] [Indexed: 09/01/2024] Open
Abstract
Pregnancy is a complex and challenging process associated with physiological changes whose objective is to adapt the maternal organism to the increasing energetic requirements due to embryo and fetal development. A failed adaptation to these demands may lead to pregnancy complications that threaten the health of both mothers and their offspring. Since mitochondria are the main organelle responsible for energy generation in the form of ATP, the adequate state of these organelles seems crucial for proper pregnancy development and healthy pregnancy outcomes. The homeostasis of these organelles depends on several aspects, including their content, biogenesis, energy production, oxidative stress, dynamics, and signaling functions, such as apoptosis, which can be modified in relation to diseases during pregnancy. The etiology of pregnancy disorders like preeclampsia, fetal growth restriction, and gestational diabetes mellitus is not yet well understood. Nevertheless, insufficient placental perfusion and oxygen transfer are characteristic of many of them, being associated with alterations in the previously cited different aspects of mitochondrial homeostasis. Therefore, and due to the capacity of these multifactorial organelles to respond to physiological and pathophysiological stimuli, it is of great importance to gather the currently available scientific information regarding the relationship between main pregnancy complications and mitochondrial alterations. According to this, the present review is intended to show clear insight into the possible implications of mitochondria in these disorders, thus providing relevant information for further investigation in relation to the investigation and management of pregnancy diseases.
Collapse
Affiliation(s)
- Juan M. Toledano
- Department of Physiology, Faculty of Pharmacy, Campus Universitario de Cartuja, University of Granada, 18071 Granada, Spain; (J.M.T.); (J.D.-C.); (J.J.O.)
- Institute of Nutrition and Food Technology “José Mataix Verdú”, University of Granada, 18071 Granada, Spain
- Nutrition and Food Sciences Ph.D. Program, University of Granada, 18071 Granada, Spain
| | - María Puche-Juarez
- Department of Physiology, Faculty of Pharmacy, Campus Universitario de Cartuja, University of Granada, 18071 Granada, Spain; (J.M.T.); (J.D.-C.); (J.J.O.)
- Institute of Nutrition and Food Technology “José Mataix Verdú”, University of Granada, 18071 Granada, Spain
- Nutrition and Food Sciences Ph.D. Program, University of Granada, 18071 Granada, Spain
| | - Jose Maria Galvez-Navas
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), 28029 Madrid, Spain;
- Cáncer Registry of Granada, Andalusian School of Public Health, Cuesta del Observatorio 4, Campus Universitario de Cartuja, 18011 Granada, Spain
- Department of Biochemistry and Molecular Biology II, Faculty of Pharmacy, Campus Universitario de Cartuja, University of Granada, 18071 Granada, Spain
- Instituto de Investigación Biosanitaria (IBS), 18016 Granada, Spain
| | - Jorge Moreno-Fernandez
- Department of Physiology, Faculty of Pharmacy, Campus Universitario de Cartuja, University of Granada, 18071 Granada, Spain; (J.M.T.); (J.D.-C.); (J.J.O.)
- Institute of Nutrition and Food Technology “José Mataix Verdú”, University of Granada, 18071 Granada, Spain
- Instituto de Investigación Biosanitaria (IBS), 18016 Granada, Spain
| | - Javier Diaz-Castro
- Department of Physiology, Faculty of Pharmacy, Campus Universitario de Cartuja, University of Granada, 18071 Granada, Spain; (J.M.T.); (J.D.-C.); (J.J.O.)
- Institute of Nutrition and Food Technology “José Mataix Verdú”, University of Granada, 18071 Granada, Spain
- Instituto de Investigación Biosanitaria (IBS), 18016 Granada, Spain
| | - Julio J. Ochoa
- Department of Physiology, Faculty of Pharmacy, Campus Universitario de Cartuja, University of Granada, 18071 Granada, Spain; (J.M.T.); (J.D.-C.); (J.J.O.)
- Institute of Nutrition and Food Technology “José Mataix Verdú”, University of Granada, 18071 Granada, Spain
- Instituto de Investigación Biosanitaria (IBS), 18016 Granada, Spain
| |
Collapse
|
7
|
Torres-Torres J, Espino-y-Sosa S, Martinez-Portilla R, Borboa-Olivares H, Estrada-Gutierrez G, Acevedo-Gallegos S, Ruiz-Ramirez E, Velasco-Espin M, Cerda-Flores P, Ramirez-Gonzalez A, Rojas-Zepeda L. A Narrative Review on the Pathophysiology of Preeclampsia. Int J Mol Sci 2024; 25:7569. [PMID: 39062815 PMCID: PMC11277207 DOI: 10.3390/ijms25147569] [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: 06/15/2024] [Revised: 07/06/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
Preeclampsia (PE) is a multifactorial pregnancy disorder characterized by hypertension and proteinuria, posing significant risks to both maternal and fetal health. Despite extensive research, its complex pathophysiology remains incompletely understood. This narrative review aims to elucidate the intricate mechanisms contributing to PE, focusing on abnormal placentation, maternal systemic response, oxidative stress, inflammation, and genetic and epigenetic factors. This review synthesizes findings from recent studies, clinical trials, and meta-analyses, highlighting key molecular and cellular pathways involved in PE. The review integrates data on oxidative stress biomarkers, angiogenic factors, immune interactions, and mitochondrial dysfunction. PE is initiated by poor placentation due to inadequate trophoblast invasion and improper spiral artery remodeling, leading to placental hypoxia. This triggers the release of anti-angiogenic factors such as soluble fms-like tyrosine kinase-1 (sFlt-1) and soluble endoglin (sEng), causing widespread endothelial dysfunction and systemic inflammation. Oxidative stress, mitochondrial abnormalities, and immune dysregulation further exacerbate the condition. Genetic and epigenetic modifications, including polymorphisms in the Fms-like tyrosine kinase 1 (FLT1) gene and altered microRNA (miRNA) expression, play critical roles. Emerging therapeutic strategies targeting oxidative stress, inflammation, angiogenesis, and specific molecular pathways like the heme oxygenase-1/carbon monoxide (HO-1/CO) and cystathionine gamma-lyase/hydrogen sulfide (CSE/H2S) pathways show promise in mitigating preeclampsia's effects. PE is a complex disorder with multifactorial origins involving abnormal placentation, endothelial dysfunction, systemic inflammation, and oxidative stress. Despite advances in understanding its pathophysiology, effective prevention and treatment strategies remain limited. Continued research is essential to develop targeted therapies that can improve outcomes for both mothers and their babies.
Collapse
Affiliation(s)
- Johnatan Torres-Torres
- Clinical Research Branch, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico; (R.M.-P.)
- Obstetric and Gynecology Department, Hospital General de México Dr. Eduardo Liceaga, Mexico City 06720, Mexico (P.C.-F.)
| | - Salvador Espino-y-Sosa
- Clinical Research Branch, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico; (R.M.-P.)
| | - Raigam Martinez-Portilla
- Clinical Research Branch, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico; (R.M.-P.)
| | - Hector Borboa-Olivares
- Clinical Research Branch, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico; (R.M.-P.)
| | - Guadalupe Estrada-Gutierrez
- Clinical Research Branch, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico; (R.M.-P.)
| | - Sandra Acevedo-Gallegos
- Clinical Research Branch, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico; (R.M.-P.)
| | - Erika Ruiz-Ramirez
- Obstetric and Gynecology Department, Hospital General de México Dr. Eduardo Liceaga, Mexico City 06720, Mexico (P.C.-F.)
| | - Martha Velasco-Espin
- Obstetric and Gynecology Department, Hospital General de México Dr. Eduardo Liceaga, Mexico City 06720, Mexico (P.C.-F.)
| | - Pablo Cerda-Flores
- Obstetric and Gynecology Department, Hospital General de México Dr. Eduardo Liceaga, Mexico City 06720, Mexico (P.C.-F.)
| | - Andrea Ramirez-Gonzalez
- Obstetric and Gynecology Department, Hospital General de México Dr. Eduardo Liceaga, Mexico City 06720, Mexico (P.C.-F.)
| | - Lourdes Rojas-Zepeda
- Maternal-Fetal Medicine Department, Instituto Materno Infantil del Estado de Mexico, Toluca 50170, Mexico
| |
Collapse
|
8
|
Vélez EJ, Schnebert S, Goguet M, Balbuena-Pecino S, Dias K, Beauclair L, Fontagné-Dicharry S, Véron V, Depincé A, Beaumatin F, Herpin A, Seiliez I. Chaperone-mediated autophagy protects against hyperglycemic stress. Autophagy 2024; 20:752-768. [PMID: 37798944 PMCID: PMC11062381 DOI: 10.1080/15548627.2023.2267415] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 09/26/2023] [Accepted: 10/02/2023] [Indexed: 10/07/2023] Open
Abstract
Chaperone-mediated autophagy (CMA) is a major pathway of lysosomal proteolysis critical for cellular homeostasis and metabolism, and whose defects have been associated with several human pathologies. While CMA has been well described in mammals, functional evidence has only recently been documented in fish, opening up new perspectives to tackle this function under a novel angle. Now we propose to explore CMA functions in the rainbow trout (RT, Oncorhynchus mykiss), a fish species recognized as a model organism of glucose intolerance and characterized by the presence of two paralogs of the CMA-limiting factor Lamp2A (lysosomal associated membrane protein 2A). To this end, we validated a fluorescent reporter (KFERQ-PA-mCherry1) previously used to track functional CMA in mammalian cells, in an RT hepatoma-derived cell line (RTH-149). We found that incubation of cells with high-glucose levels (HG, 25 mM) induced translocation of the CMA reporter to lysosomes and/or late endosomes in a KFERQ- and Lamp2A-dependent manner, as well as reduced its half-life compared to the control (5 mM), thus demonstrating increased CMA flux. Furthermore, we observed that activation of CMA upon HG exposure was mediated by generation of mitochondrial reactive oxygen species, and involving the antioxidant transcription factor Nfe2l2/Nrf2 (nfe2 like bZIP transcription factor 2). Finally, we demonstrated that CMA plays an important protective role against HG-induced stress, primarily mediated by one of the two RT Lamp2As. Together, our results provide unequivocal evidence for CMA activity existence in RT and highlight both the role and regulation of CMA during glucose-related metabolic disorders.Abbreviations: AREs: antioxidant response elements; CHC: α-cyano -4-hydroxycinnamic acid; Chr: chromosome; CMA: chaperone-mediated autophagy; CT: control; DMF: dimethyl fumarate; Emi: endosomal microautophagy; HG: high-glucose; HMOX1: heme oxygenase 1; H2O2: hydrogen peroxide; KFERQ: lysine-phenylalanine-glutamate-arginine-glutamine; LAMP1: lysosomal associated membrane protein 1; LAMP2A: lysosomal associated membrane protein 2A; MCC: Manders' correlation coefficient; Manders' correlation coefficient Mo: morpholino oligonucleotide; NAC: N-acetyl cysteine; NFE2L2/NRF2: NFE2 like bZIP transcription factor 2; PA-mCherry: photoactivable mCherry; PCC: Pearson's correlation coefficient; ROS: reactive oxygen species; RT: rainbow trout; siRNAs: small interfering RNAs; SOD: superoxide dismutase; Tsg101: tumor susceptibility 101; TTFA: 2-thenoyltrifluoroacetone; WGD: whole-genome duplication.
Collapse
Affiliation(s)
- Emilio J. Vélez
- Université de Pau et des Pays de l‘Adour, E2S UPPA, INRAE, UMR1419 Nutrition Métabolisme et Aquaculture, Saint-Pée-sur-Nivelle, France
| | - Simon Schnebert
- Université de Pau et des Pays de l‘Adour, E2S UPPA, INRAE, UMR1419 Nutrition Métabolisme et Aquaculture, Saint-Pée-sur-Nivelle, France
| | - Maxime Goguet
- Université de Pau et des Pays de l‘Adour, E2S UPPA, INRAE, UMR1419 Nutrition Métabolisme et Aquaculture, Saint-Pée-sur-Nivelle, France
| | - Sara Balbuena-Pecino
- Université de Pau et des Pays de l‘Adour, E2S UPPA, INRAE, UMR1419 Nutrition Métabolisme et Aquaculture, Saint-Pée-sur-Nivelle, France
| | - Karine Dias
- Université de Pau et des Pays de l‘Adour, E2S UPPA, INRAE, UMR1419 Nutrition Métabolisme et Aquaculture, Saint-Pée-sur-Nivelle, France
| | - Linda Beauclair
- Université de Pau et des Pays de l‘Adour, E2S UPPA, INRAE, UMR1419 Nutrition Métabolisme et Aquaculture, Saint-Pée-sur-Nivelle, France
| | - Stéphanie Fontagné-Dicharry
- Université de Pau et des Pays de l‘Adour, E2S UPPA, INRAE, UMR1419 Nutrition Métabolisme et Aquaculture, Saint-Pée-sur-Nivelle, France
| | - Vincent Véron
- Université de Pau et des Pays de l‘Adour, E2S UPPA, INRAE, UMR1419 Nutrition Métabolisme et Aquaculture, Saint-Pée-sur-Nivelle, France
| | - Alexandra Depincé
- INRAE, UR1037 Laboratory of Fish Physiology and Genomics, Campus de Beaulieu, Rennes, France
| | - Florian Beaumatin
- Université de Pau et des Pays de l‘Adour, E2S UPPA, INRAE, UMR1419 Nutrition Métabolisme et Aquaculture, Saint-Pée-sur-Nivelle, France
| | - Amaury Herpin
- INRAE, UR1037 Laboratory of Fish Physiology and Genomics, Campus de Beaulieu, Rennes, France
| | - Iban Seiliez
- Université de Pau et des Pays de l‘Adour, E2S UPPA, INRAE, UMR1419 Nutrition Métabolisme et Aquaculture, Saint-Pée-sur-Nivelle, France
| |
Collapse
|
9
|
Abramicheva PA, Andrianova NV, Babenko VA, Zorova LD, Zorov SD, Pevzner IB, Popkov VA, Semenovich DS, Yakupova EI, Silachev DN, Plotnikov EY, Sukhikh GT, Zorov DB. Mitochondrial Network: Electric Cable and More. BIOCHEMISTRY. BIOKHIMIIA 2023; 88:1596-1607. [PMID: 38105027 DOI: 10.1134/s0006297923100140] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 09/20/2023] [Accepted: 09/22/2023] [Indexed: 12/19/2023]
Abstract
Mitochondria in a cell can unite and organize complex, extended structures that occupy the entire cellular volume, providing an equal supply with energy in the form of ATP synthesized in mitochondria. In accordance with the chemiosmotic concept, the oxidation energy of respiratory substrates is largely stored in the form of an electrical potential difference on the inner membrane of mitochondria. The theory of the functioning of extended mitochondrial structures as intracellular electrical wires suggests that mitochondria provide the fastest delivery of electrical energy through the cellular volume, followed by the use of this energy for the synthesis of ATP, thereby accelerating the process of ATP delivery compared to the rather slow diffusion of ATP in the cell. This analytical review gives the history of the cable theory, lists unsolved critical problems, describes the restructuring of the mitochondrial network and the role of oxidative stress in this process. In addition to the already proven functioning of extended mitochondrial structures as electrical cables, a number of additional functions are proposed, in particular, the hypothesis is put forth that mitochondrial networks maintain the redox potential in the cellular volume, which may vary depending on the physiological state, as a result of changes in the three-dimensional organization of the mitochondrial network (fragmentation/fission-fusion). A number of pathologies accompanied by a violation of the redox status and the participation of mitochondria in them are considered.
Collapse
Affiliation(s)
- Polina A Abramicheva
- Belozersky Research Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Nadezda V Andrianova
- Belozersky Research Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Valentina A Babenko
- Belozersky Research Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
- Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow, 117997, Russia
| | - Ljubava D Zorova
- Belozersky Research Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
- Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow, 117997, Russia
| | - Savva D Zorov
- Belozersky Research Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Irina B Pevzner
- Belozersky Research Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
- Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow, 117997, Russia
| | - Vasily A Popkov
- Belozersky Research Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
- Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow, 117997, Russia
| | - Dmitry S Semenovich
- Belozersky Research Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Elmira I Yakupova
- Belozersky Research Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Denis N Silachev
- Belozersky Research Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
- Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow, 117997, Russia
| | - Egor Y Plotnikov
- Belozersky Research Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
- Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow, 117997, Russia
| | - Gennady T Sukhikh
- Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow, 117997, Russia
| | - Dmitry B Zorov
- Belozersky Research Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia.
- Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow, 117997, Russia
| |
Collapse
|
10
|
Placental Mitochondrial Function and Dysfunction in Preeclampsia. Int J Mol Sci 2023; 24:ijms24044177. [PMID: 36835587 PMCID: PMC9963167 DOI: 10.3390/ijms24044177] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 02/09/2023] [Accepted: 02/14/2023] [Indexed: 02/22/2023] Open
Abstract
The placenta is a vital organ of pregnancy, regulating adaptation to pregnancy, gestational parent/fetal exchange, and ultimately, fetal development and growth. Not surprisingly, in cases of placental dysfunction-where aspects of placental development or function become compromised-adverse pregnancy outcomes can result. One common placenta-mediated disorder of pregnancy is preeclampsia (PE), a hypertensive disorder of pregnancy with a highly heterogeneous clinical presentation. The wide array of clinical characteristics observed in pregnant individuals and neonates of a PE pregnancy are likely the result of distinct forms of placental pathology underlying the PE diagnosis, explaining why no one common intervention has proven effective in the prevention or treatment of PE. The historical paradigm of placental pathology in PE highlights an important role for utero-placental malperfusion, placental hypoxia and oxidative stress, and a critical role for placental mitochondrial dysfunction in the pathogenesis and progression of the disease. In the current review, the evidence of placental mitochondrial dysfunction in the context of PE will be summarized, highlighting how altered mitochondrial function may be a common feature across distinct PE subtypes. Further, advances in this field of study and therapeutic targeting of mitochondria as a promising intervention for PE will be discussed.
Collapse
|
11
|
Barron A, Manna S, McElwain CJ, Musumeci A, McCarthy FP, O’Keeffe GW, McCarthy CM. Maternal pre-eclampsia serum increases neurite growth and mitochondrial function through a potential IL-6-dependent mechanism in differentiated SH-SY5Y cells. Front Physiol 2023; 13:1043481. [PMID: 36714304 PMCID: PMC9877349 DOI: 10.3389/fphys.2022.1043481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 12/28/2022] [Indexed: 01/15/2023] Open
Abstract
Introduction: Pre-eclampsia (PE) is a common and serious hypertensive disorder of pregnancy, which affects 3%-5% of first-time pregnancies and is a leading cause of maternal and neonatal morbidity and mortality. Prenatal exposure to PE is associated with an increased risk of neurodevelopmental disorders in affected offspring, although the cellular and molecular basis of this increased risk is largely unknown. Methods: Here, we examined the effects of exposure to maternal serum from women with PE or a healthy uncomplicated pregnancy on the survival, neurite growth and mitochondrial function of neuronally differentiated human SH-SY5Y neuroblastoma cells, which are commonly used to study neurite growth. Neurite growth and mitochondrial function are two strongly linked neurodevelopmental parameters in which alterations have been implicated in neurodevelopmental disorders. Following this, we investigated the pleiotropic cytokine interleukin-6 (IL-6) levels as a potential mechanism. Results: Cells exposed to 3% (v/v) PE serum for 72 h exhibited increased neurite growth (p < 0.05), which was validated in the human neural progenitor cell line, ReNcell® VM (p < 0.01), and mitochondrial respiration (elevated oxygen consumption rate (p < 0.05), basal mitochondrial respiration, proton leak, ATP synthesis, and non-mitochondrial respiration) compared to control serum-treated cells. ELISA analysis showed elevations in maternal IL-6 in PE sera (p < 0.05) and placental explants (p < 0.05). In support of this, SH-SY5Y cells exposed to 3% (v/v) PE serum for 24 h had increased phospho-STAT3 levels, which is a key intracellular mediator of IL-6 signalling (p < 0.05). Furthermore, treatment with anti-IL-6 neutralizing antibody blocked the effects of PE serum on neurite growth (p < 0.05), and exposure to IL-6 promoted neurite growth in SH-SY5Y cells (p < 0.01). Discussion: Collectively these data show elevated serum levels of maternal IL-6 in PE, which increases neurite growth and mitochondrial function in SH-SY5Y cells. This rationalizes the further study of IL-6 as a potential mediator between PE exposure and neurodevelopmental outcome in the offspring.
Collapse
Affiliation(s)
- Aaron Barron
- Department of Anatomy and Neuroscience, University College, Cork, Ireland,Department of Pharmacology and Therapeutics, University College Cork, Cork, Ireland
| | - Samprikta Manna
- Department of Pharmacology and Therapeutics, University College Cork, Cork, Ireland,Department of Obstetrics and Gynaecology, Cork University Maternity Hospital, Cork, Ireland
| | - Colm J. McElwain
- Department of Pharmacology and Therapeutics, University College Cork, Cork, Ireland
| | - Andrea Musumeci
- Department of Pharmacology and Therapeutics, University College Cork, Cork, Ireland
| | - Fergus P. McCarthy
- Department of Obstetrics and Gynaecology, Cork University Maternity Hospital, Cork, Ireland
| | - Gerard W. O’Keeffe
- Department of Anatomy and Neuroscience, University College, Cork, Ireland,Cork Neuroscience Centre, University College Cork, Cork, Ireland,*Correspondence: Gerard W. O’Keeffe, ; Cathal M. McCarthy,
| | - Cathal M. McCarthy
- Department of Pharmacology and Therapeutics, University College Cork, Cork, Ireland,*Correspondence: Gerard W. O’Keeffe, ; Cathal M. McCarthy,
| |
Collapse
|
12
|
Hu XQ, Zhang L. Oxidative Regulation of Vascular Ca v1.2 Channels Triggers Vascular Dysfunction in Hypertension-Related Disorders. Antioxidants (Basel) 2022; 11:antiox11122432. [PMID: 36552639 PMCID: PMC9774363 DOI: 10.3390/antiox11122432] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 11/28/2022] [Accepted: 12/06/2022] [Indexed: 12/13/2022] Open
Abstract
Blood pressure is determined by cardiac output and peripheral vascular resistance. The L-type voltage-gated Ca2+ (Cav1.2) channel in small arteries and arterioles plays an essential role in regulating Ca2+ influx, vascular resistance, and blood pressure. Hypertension and preeclampsia are characterized by high blood pressure. In addition, diabetes has a high prevalence of hypertension. The etiology of these disorders remains elusive, involving the complex interplay of environmental and genetic factors. Common to these disorders are oxidative stress and vascular dysfunction. Reactive oxygen species (ROS) derived from NADPH oxidases (NOXs) and mitochondria are primary sources of vascular oxidative stress, whereas dysfunction of the Cav1.2 channel confers increased vascular resistance in hypertension. This review will discuss the importance of ROS derived from NOXs and mitochondria in regulating vascular Cav1.2 and potential roles of ROS-mediated Cav1.2 dysfunction in aberrant vascular function in hypertension, diabetes, and preeclampsia.
Collapse
|
13
|
Chatre L, Ducat A, Spradley FT, Palei AC, Chéreau C, Couderc B, Thomas KC, Wilson AR, Amaral LM, Gaillard I, Méhats C, Lagoutte I, Jacques S, Miralles F, Batteux F, Granger JP, Ricchetti M, Vaiman D. Increased NOS coupling by the metabolite tetrahydrobiopterin (BH4) reduces preeclampsia/IUGR consequences. Redox Biol 2022; 55:102406. [PMID: 35964341 PMCID: PMC9389306 DOI: 10.1016/j.redox.2022.102406] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 07/04/2022] [Accepted: 07/11/2022] [Indexed: 11/24/2022] Open
Abstract
Preeclampsia (PE) is a high-prevalence pregnancy disease characterized by placental insufficiency, gestational hypertension, and proteinuria. Overexpression of the A isoform of the STOX1 transcription factor (STOX1A) recapitulates PE in mice, and STOX1A overexpressing trophoblasts recapitulate PE patients hallmarks in terms of gene expression and pathophysiology. STOX1 overexpression induces nitroso-redox imbalance and mitochondrial hyper-activation. Here, by a thorough analysis on cell models, we show that STOX1 overexpression in trophoblasts alters inducible nitric oxide synthase (iNOS), nitric oxide (NO) content, the nitroso-redox balance, the antioxidant defense, and mitochondrial function. This is accompanied by specific alterations of the Krebs cycle leading to reduced l-malate content. By increasing NOS coupling using the metabolite tetrahydrobiopterin (BH4) we restore this multi-step pathway in vitro. Moving in vivo on two different rodent models (STOX1 mice and RUPP rats, alike early onset and late onset preeclampsia, respectively), we show by transcriptomics that BH4 directly reverts STOX1-deregulated gene expression including glutathione metabolism, oxidative phosphorylation, cholesterol metabolism, inflammation, lipoprotein metabolism and platelet activation, successfully treating placental hypotrophy, gestational hypertension, proteinuria and heart hypertrophy. In the RUPP rats we show that the major fetal issue of preeclampsia, Intra Uterine Growth Restriction (IUGR), is efficiently corrected. Our work posits on solid bases BH4 as a novel potential therapy for preeclampsia.
Collapse
Affiliation(s)
- Laurent Chatre
- Institut Pasteur, Department of Developmental & Stem Cell Biology, Stem Cell & Development, 25-28 Rue du Dr. Roux, Paris, France; UMR 3738 CNRS, 25 Rue du Dr. Roux, Paris, 75015, France
| | - Aurélien Ducat
- Institut Cochin U1016, INSERM UMR8104 CNRS, 24, rue du Fg St Jacques, Paris, France
| | - Frank T Spradley
- Department of Surgery, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Ana C Palei
- Department of Surgery, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Christiane Chéreau
- Institut Cochin U1016, INSERM UMR8104 CNRS, 24, rue du Fg St Jacques, Paris, France
| | - Betty Couderc
- Institut Cochin U1016, INSERM UMR8104 CNRS, 24, rue du Fg St Jacques, Paris, France
| | - Kamryn C Thomas
- Department of Surgery, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Anna R Wilson
- Department of Surgery, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Lorena M Amaral
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Irène Gaillard
- Institut Cochin U1016, INSERM UMR8104 CNRS, 24, rue du Fg St Jacques, Paris, France
| | - Céline Méhats
- Institut Cochin U1016, INSERM UMR8104 CNRS, 24, rue du Fg St Jacques, Paris, France
| | - Isabelle Lagoutte
- Institut Cochin U1016, INSERM UMR8104 CNRS, 24, rue du Fg St Jacques, Paris, France
| | - Sébastien Jacques
- Institut Cochin U1016, INSERM UMR8104 CNRS, 24, rue du Fg St Jacques, Paris, France
| | - Francisco Miralles
- Institut Cochin U1016, INSERM UMR8104 CNRS, 24, rue du Fg St Jacques, Paris, France
| | - Frédéric Batteux
- Institut Cochin U1016, INSERM UMR8104 CNRS, 24, rue du Fg St Jacques, Paris, France
| | - Joey P Granger
- Department of Physiology & Biophysics, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Miria Ricchetti
- Institut Pasteur, Department of Developmental & Stem Cell Biology, Stem Cell & Development, 25-28 Rue du Dr. Roux, Paris, France; UMR 3738 CNRS, 25 Rue du Dr. Roux, Paris, 75015, France; Institut Pasteur, Molecular Mechanisms of Pathological and Physiological Ageing, 25-28 Rue du Dr. Roux, Paris, France
| | - Daniel Vaiman
- Institut Cochin U1016, INSERM UMR8104 CNRS, 24, rue du Fg St Jacques, Paris, France.
| |
Collapse
|
14
|
Workalemahu T, Enquobahrie DA, Gelaye B, Tadesse MG, Sanchez SE, Tekola-Ayele F, Hajat A, Thornton TA, Ananth CV, Williams MA. Maternal-fetal genetic interactions, imprinting, and risk of placental abruption. J Matern Fetal Neonatal Med 2022; 35:3473-3482. [PMID: 32972274 PMCID: PMC8601203 DOI: 10.1080/14767058.2020.1822314] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Revised: 09/02/2020] [Accepted: 09/08/2020] [Indexed: 12/17/2022]
Abstract
RESULTS Abruption cases were more likely to experience preeclampsia, have shorter gestational age, and deliver infants with lower birthweight compared with controls. Models with MFGI effects provided improved fit than models with only maternal and fetal genotype main effects for SNP rs12530904 (p-value = 1.2e-04) in calcium/calmodulin-dependent protein kinase [CaM kinase] II beta (CAMK2B), and, SNP rs73136795 (p-value = 1.9e-04) in peroxisome proliferator-activated receptor-gamma (PPARG), both MB genes. We identified 320 SNPs in 45 maternally-imprinted genes (including potassium voltage-gated channel subfamily Q member 1 [KCNQ1], neurotrimin [NTM], and, ATPase phospholipid transporting 10 A [ATP10A]) associated with abruption. Top hits included rs2012323 (p-value = 1.6E-16) and rs12221520 (p-value1.3e-13) in KCNQ1, rs8036892 (p-value = 9.3E-17) and rs188497582 in ATP10A, rs12589854 (p-value = 2.9E-11) and rs80203467 (p-value = 4.6e-11) in maternally expressed 8, small nucleolar RNA host (MEG8), and rs138281088 in solute carrier family 22 member 2 (SLC22A2) (p-value = 6.8e-9). CONCLUSIONS We identified novel PA-related maternal-fetal MB gene interactions and imprinting effects that highlight the role of the fetus in PA risk development. Findings can inform mechanistic investigations to understand the pathogenesis of PA.
Collapse
Affiliation(s)
- Tsegaselassie Workalemahu
- Department of Epidemiology, School of Public Health, University of Washington, Seattle, Washington
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, University of Utah, Salt Lake City, Utah
| | - Daniel A. Enquobahrie
- Department of Epidemiology, School of Public Health, University of Washington, Seattle, Washington
- Center for Perinatal Studies, Swedish Medical Center, Seattle, Washington
| | - Bizu Gelaye
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Mahlet G. Tadesse
- Department of Mathematics and Statistics, Georgetown University, Washington, District of Columbia
| | - Sixto E. Sanchez
- Facultad de Medicina Humana, Universidad de San Martín de Porres, Lima, Peru
- Asociación Civil PROESA, Lima, Peru
| | - Fasil Tekola-Ayele
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, University of Utah, Salt Lake City, Utah
| | - Anjum Hajat
- Department of Epidemiology, School of Public Health, University of Washington, Seattle, Washington
| | | | - Cande V. Ananth
- Division of Epidemiology and Biostatistics, Department of Obstetrics, Gynecology and Reproductive Sciences, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ
- Department of Biostatistics and Epidemiology, Rutgers School of Public Health, Piscataway, NJ
- Cardiovascular Institute of New Jersey (CVI-NJ), Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ
- Environmental and Occupational Health Sciences Institute (EOHSI), Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ
| | - Michelle A. Williams
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| |
Collapse
|
15
|
Oxidative and Inflammatory Imbalance in Placenta and Kidney of sFlt1-Induced Early-Onset Preeclampsia Rat Model. Antioxidants (Basel) 2022; 11:antiox11081608. [PMID: 36009326 PMCID: PMC9405374 DOI: 10.3390/antiox11081608] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 08/13/2022] [Accepted: 08/15/2022] [Indexed: 12/13/2022] Open
Abstract
Preeclampsia (PE) is a pregnancy-specific disorder characterized by the new onset of hypertension plus proteinuria and/or end-organ dysfunction. Here, we investigate the role of the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase system as a major component of reactive oxygen species generation, in a rodent model of early-onset preeclampsia induced by excess sFlt1 (soluble fms-like tyrosine kinase 1). Placenta and kidney samples were obtained from normal pregnant and PE rats to measure the sFlt1/PlGF (placental growth factor) ratio in addition to oxidative stress-related parameters, including the activities and expressions of NADPH oxidase isoforms (NOX1, NOX2, and NOX4), components of nitric oxide (NO) metabolism, and antioxidant enzymes. Peroxisome proliferator-activated receptors (PPARα, PPARγ) and cytokines IL1β, IL3, IL6, IL10, and IL18 were also measured to evaluate the inflammation status in our experimental setting. Excessive O2●− production was found in rats that were treated with sFlt1; interestingly, this alteration appears to be mediated mainly by NOX2 in the placenta and by NOX4 in the kidney. Altered NO metabolism and antioxidant defense systems, together with mitochondrial dysfunction, were observed in this model of PE. Preeclamptic animals also exhibited overexpression of proinflammatory biomarkers as well as increased collagen deposition. Our results highlight the role of NADPH oxidase in mediating oxidative stress and possibly inflammatory processes in the placenta and kidney of an sFlt1-based model of early-onset preeclampsia.
Collapse
|
16
|
Abstract
PURPOSE OF REVIEW Preeclampsia complicates 5-10% of all pregnancies and is a leading cause of maternal and perinatal mortality and morbidity. The placenta plays a pivotal role in determining pregnancy outcome by supplying the fetus with oxygen and nutrients and by synthesizing hormones. Placental function is highly dependent on energy supplied by mitochondria. It is well-known that preeclampsia is originated from placental dysfunction, although the etiology of it remains elusive. RECENT FINDINGS During the last three decades, substantial evidence suggests that mitochondrial abnormality is a major contributor to placental dysfunction. In addition, mitochondrial damage caused by circulating bioactive factors released from the placenta may cause endothelial dysfunction and subsequent elevation in maternal blood pressure. In this review, we summarize the current knowledge of mitochondrial abnormality in the pathogenesis of preeclampsia and discuss therapeutic approaches targeting mitochondria for treatment of preeclampsia.
Collapse
|
17
|
Di Fabrizio C, Giorgione V, Khalil A, Murdoch CE. Antioxidants in Pregnancy: Do We Really Need More Trials? Antioxidants (Basel) 2022; 11:812. [PMID: 35624676 PMCID: PMC9137466 DOI: 10.3390/antiox11050812] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 04/13/2022] [Accepted: 04/19/2022] [Indexed: 02/01/2023] Open
Abstract
Human pregnancy can be affected by numerous pathologies, from those which are mild and reversible to others which are life-threatening. Among these, gestational diabetes mellitus and hypertensive disorders of pregnancy with subsequent consequences stand out. Health problems experienced by women during pregnancy and postpartum are associated with significant costs to health systems worldwide and contribute largely to maternal mortality and morbidity. Major risk factors for mothers include obesity, advanced maternal age, cardiovascular dysfunction, and endothelial damage; in these scenarios, oxidative stress plays a major role. Markers of oxidative stress can be measured in patients with preeclampsia, foetal growth restriction, and gestational diabetes mellitus, even before their clinical onset. In consequence, antioxidant supplements have been proposed as a possible therapy; however, results derived from large scale randomised clinical trials have been disappointing as no positive effects were demonstrated. This review focuses on the latest evidence on oxidative stress in pregnancy complications, their early diagnosis, and possible therapies to prevent or treat these pathologies.
Collapse
Affiliation(s)
- Carolina Di Fabrizio
- Vascular Biology Research Center, Molecular and Clinical Sciences Research Institute, St George’s University of London, London SW17 0QT, UK; (C.D.F.); (V.G.); (A.K.)
- Systems Medicine, School of Medicine, University of Dundee, Dundee DD1 9SY, UK
| | - Veronica Giorgione
- Vascular Biology Research Center, Molecular and Clinical Sciences Research Institute, St George’s University of London, London SW17 0QT, UK; (C.D.F.); (V.G.); (A.K.)
| | - Asma Khalil
- Vascular Biology Research Center, Molecular and Clinical Sciences Research Institute, St George’s University of London, London SW17 0QT, UK; (C.D.F.); (V.G.); (A.K.)
- Fetal Medicine Unit, St George’s University Hospitals NHS Foundation Trust, London SW17 0QT, UK
| | - Colin E. Murdoch
- Systems Medicine, School of Medicine, University of Dundee, Dundee DD1 9SY, UK
| |
Collapse
|
18
|
Manna S, Ruano CSM, Hegenbarth JC, Vaiman D, Gupta S, McCarthy FP, Méhats C, McCarthy C, Apicella C, Scheel J. Computational Models on Pathological Redox Signalling Driven by Pregnancy: A Review. Antioxidants (Basel) 2022; 11:585. [PMID: 35326235 PMCID: PMC8945226 DOI: 10.3390/antiox11030585] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 03/15/2022] [Accepted: 03/16/2022] [Indexed: 02/04/2023] Open
Abstract
Oxidative stress is associated with a myriad of diseases including pregnancy pathologies with long-term cardiovascular repercussions for both the mother and baby. Aberrant redox signalling coupled with deficient antioxidant defence leads to chronic molecular impairment. Abnormal placentation has been considered the primary source for reactive species; however, placental dysfunction has been deemed secondary to maternal cardiovascular maladaptation in pregnancy. While various therapeutic interventions, aimed at combating deregulated oxidative stress during pregnancy have shown promise in experimental models, they often result as inconclusive or detrimental in clinical trials, warranting the need for further research to identify candidates. The strengths and limitations of current experimental methods in redox research are discussed. Assessment of redox status and oxidative stress in experimental models and in clinical practice remains challenging; the state-of-the-art of computational models in this field is presented in this review, comparing static and dynamic models which provide functional information such as protein-protein interactions, as well as the impact of changes in molecular species on the redox-status of the system, respectively. Enhanced knowledge of redox biology in during pregnancy through computational modelling such as generation of Systems Biology Markup Language model which integrates existing models to a larger network in the context of placenta physiology.
Collapse
Affiliation(s)
- Samprikta Manna
- Department of Obstetrics and Gynaecology, Cork University Maternity Hospital, University College Cork, T12 YE02 Cork, Ireland;
| | - Camino S. M. Ruano
- Institut Cochin, Inserm U1016, UMR8104 CNRS, Université de Paris, 75014 Paris, France; (C.S.M.R.); (D.V.); (C.M.); (C.A.)
| | - Jana-Charlotte Hegenbarth
- Department of Molecular Genetics, Faculty of Science and Engineering, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6211 KH Maastricht, The Netherlands;
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Daniel Vaiman
- Institut Cochin, Inserm U1016, UMR8104 CNRS, Université de Paris, 75014 Paris, France; (C.S.M.R.); (D.V.); (C.M.); (C.A.)
| | - Shailendra Gupta
- Department of Systems Biology and Bioinformatics, Rostock University, 18051 Rostock, Germany; (S.G.); (J.S.)
| | - Fergus P. McCarthy
- Department of Obstetrics and Gynaecology, Cork University Maternity Hospital, University College Cork, T12 YE02 Cork, Ireland;
| | - Céline Méhats
- Institut Cochin, Inserm U1016, UMR8104 CNRS, Université de Paris, 75014 Paris, France; (C.S.M.R.); (D.V.); (C.M.); (C.A.)
| | - Cathal McCarthy
- Department of Pharmacology and Therapeutics, Western Gateway Building, University College Cork, T12 K8AF Cork, Ireland;
| | - Clara Apicella
- Institut Cochin, Inserm U1016, UMR8104 CNRS, Université de Paris, 75014 Paris, France; (C.S.M.R.); (D.V.); (C.M.); (C.A.)
| | - Julia Scheel
- Department of Systems Biology and Bioinformatics, Rostock University, 18051 Rostock, Germany; (S.G.); (J.S.)
| |
Collapse
|
19
|
Clément L, Testoni B. [Mitochondrial dysfunction as a new therapeutic target to restore exhausted immune responses against HBV]. Med Sci (Paris) 2022; 38:223-226. [PMID: 35179480 DOI: 10.1051/medsci/2022011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Dans le cadre d’un partenariat avec médecine/sciences, et pour la quatrième année consécutive, des étudiants du module d’enseignement « d’immunologie virologie et cancer » du Master de cancérologie de Lyon présentent une analyse d’articles scientifiques récents faisant état d’observations innovantes et importantes. Ce travail a été encadré par des chercheurs confirmés du Centre de Recherche en Cancérologie de Lyon (CRCL). Le Master de cancérologie (université Claude Bernard Lyon1 - VetAgroSup) accueille chaque année 40 étudiants en M1 et environ 80 en M2. Ce master dit « d’excellence » (master international labellisé Université de Lyon) assure aux étudiants de M1 une formation à la cancérologie reposant sur un socle de base commun (biologie cellulaire, moléculaire, immunologie, bio-statistique, épidémiologie, recherche translationnelle, etc.). Les étudiants de M2 peuvent choisir l’un des parcours suivants : 1) Biologie du Cancer ; 2) Innovations thérapeutiques en Cancérologie ; 3) Médecine de précision en Cancérologie ; 4) Cancer Bio-engineering.
Le Master de cancérologie de Lyon repose sur une forte implication des chercheurs et enseignants-chercheurs du laboratoire d'excellence en développement et cancérologie (LabEx DEVweCAN), ainsi que sur un partenariat solide avec plusieurs instituts à travers le monde dont le MIT (Massachusetts Institute of Technology, Cambridge, États-Unis), l'université d'Harvard (Boston, États-Unis), l'université de San Diego (États-Unis), University of City of London (UCL), le Beatson Institute de Glasgow (Royaume-Uni), les universités de Shanghai Jiao Tong (République populaire de Chine, RPC), de Tokyo et de Tohoku (Japon), de Melbourne (Australie) et d'Auckland (Nouvelle-Zélande).
Collapse
Affiliation(s)
- Léa Clément
- University de Lyon, UMR_S1052, CRCL (Cancer Research Center of Lyon), 69008 Lyon, France
| | | |
Collapse
|
20
|
Is Mitochondrial Oxidative Stress a Viable Therapeutic Target in Preeclampsia? Antioxidants (Basel) 2022; 11:antiox11020210. [PMID: 35204094 PMCID: PMC8868187 DOI: 10.3390/antiox11020210] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/15/2022] [Accepted: 01/18/2022] [Indexed: 01/31/2023] Open
Abstract
Despite considerable research efforts over the past few decades, the pathology of preeclampsia (PE) remains poorly understood with no new FDA-approved treatments. There is a substantial amount of work being conducted by investigators around the world to identify targets to develop therapies for PE. Oxidative stress has been identified as one of the crucial players in pathogenesis of PE and has garnered a great deal of attention by several research groups including ours. While antioxidants have shown therapeutic benefit in preclinical models of PE, the clinical trials evaluating antioxidants (vitamin E and vitamin C) were found to be disappointing. Although the idea behind contribution of mitochondrial oxidative stress in PE is not new, recent years have seen an enormous interest in exploring mitochondrial oxidative stress as an important pathological mediator in PE. We and others using animals, cell models, and preeclamptic patient samples have shown the evidence for placental, renal, and endothelial cell mitochondrial oxidative stress, and its significance in PE. These studies offer promising results; however, the important and relevant question is can we translate these results into clinical efficacy in treating PE. Hence, the purpose of this review is to review the existing literature and offer our insights on the potential of mitochondrial antioxidants in treating PE.
Collapse
|
21
|
Cushen SC, Ricci CA, Bradshaw JL, Silzer T, Blessing A, Sun J, Zhou Z, Scroggins SM, Santillan MK, Santillan DA, Phillips NR, Goulopoulou S. Reduced Maternal Circulating Cell-Free Mitochondrial DNA Is Associated With the Development of Preeclampsia. J Am Heart Assoc 2022; 11:e021726. [PMID: 35014857 PMCID: PMC9238514 DOI: 10.1161/jaha.121.021726] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Background Circulating cell-free mitochondrial DNA (ccf-mtDNA) is a damage-associated molecular pattern that reflects cell stress responses and tissue damage, but little is known about ccf-mtDNA in preeclampsia. The main objectives of this study were to determine (1) absolute concentrations of ccf-mtDNA in plasma and mitochondrial DNA content in peripheral blood mononuclear cells and (2) forms of ccf-mtDNA transport in blood from women with preeclampsia and healthy controls. In addition, we sought to establish the association between aberrance in circulating DNA-related metrics, including ccf-mtDNA and DNA clearance mechanisms, and the clinical diagnosis of preeclampsia using bootstrapped penalized logistic regression. Methods and Results Absolute concentrations of ccf-mtDNA were reduced in plasma from women with preeclampsia compared with healthy controls (P≤0.02), while mtDNA copy number in peripheral blood mononuclear cells did not differ between groups (P>0.05). While the pattern of reduced ccf-mtDNA in patients with preeclampsia remained, DNA isolation from plasma using membrane lysis buffer resulted in 1000-fold higher ccf-mtDNA concentrations in the preeclampsia group (P=0.0014) and 430-fold higher ccf-mtDNA concentrations in the control group (P<0.0001). Plasma from women with preeclampsia did not induce greater Toll-like receptor-9-induced nuclear factor kappa-light-chain enhancer of activated B cells-dependent responses in human embryonic kidney 293 cells overexpressing the human TLR-9 gene (P>0.05). Penalized regression analysis showed that women with preeclampsia were more likely to have lower concentrations of ccf-mtDNA as well as higher concentrations of nuclear DNA and DNase I compared with their matched controls. Conclusions Women with preeclampsia have aberrant circulating DNA dynamics, including reduced ccf-mtDNA concentrations and DNA clearance mechanisms, compared with gestational age-matched healthy pregnant women.
Collapse
Affiliation(s)
- Spencer C Cushen
- Department of Physiology and Anatomy University of North Texas Health Science Center Fort Worth TX.,Texas College of Osteopathic Medicine University of North Texas Health Science Center Fort Worth TX
| | - Contessa A Ricci
- Department of Physiology and Anatomy University of North Texas Health Science Center Fort Worth TX
| | - Jessica L Bradshaw
- Department of Physiology and Anatomy University of North Texas Health Science Center Fort Worth TX
| | - Talisa Silzer
- Department of Microbiology, Immunology and Genetics University of North Texas Health Science Center Fort Worth TX
| | - Alexandra Blessing
- Department of Microbiology, Immunology and Genetics University of North Texas Health Science Center Fort Worth TX
| | - Jie Sun
- Department of Microbiology, Immunology and Genetics University of North Texas Health Science Center Fort Worth TX
| | - Zhengyang Zhou
- Department of Biostatistics and Epidemiology University of North Texas Health Science Center Fort Worth TX
| | - Sabrina M Scroggins
- Department of Obstetrics and Gynecology University of Iowa Carver College of Medicine Iowa City IA
| | - Mark K Santillan
- Department of Obstetrics and Gynecology University of Iowa Carver College of Medicine Iowa City IA
| | - Donna A Santillan
- Department of Obstetrics and Gynecology University of Iowa Carver College of Medicine Iowa City IA
| | - Nicole R Phillips
- Department of Microbiology, Immunology and Genetics University of North Texas Health Science Center Fort Worth TX
| | - Styliani Goulopoulou
- Department of Physiology and Anatomy University of North Texas Health Science Center Fort Worth TX
| |
Collapse
|
22
|
Bradshaw JL, Cushen SC, Phillips NR, Goulopoulou S. Circulating cell-free mitochondrial DNA in pregnancy. Physiology (Bethesda) 2022; 37:0. [PMID: 35001655 DOI: 10.1152/physiol.00037.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Circulating cell-free mitochondrial DNA (ccf-mtDNA) released upon cell injury or death stimulates diverse pattern recognition receptors to activate innate immune responses and initiate systemic inflammation. In this review, we discuss the temporal changes of ccf-mtDNA during pregnancy and its potential contribution to adverse pregnancy outcomes in pregnancy complications.
Collapse
Affiliation(s)
- Jessica L Bradshaw
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Spencer C Cushen
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas, United States.,Texas College of Osteopathic Medicine, University of North Texas Health Science Center, Fort Worth, United States
| | - Nicole R Phillips
- Department of Microbiology, Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, TX, United States
| | - Styliani Goulopoulou
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas, United States
| |
Collapse
|
23
|
Amelio GS, Provitera L, Raffaeli G, Tripodi M, Amodeo I, Gulden S, Cortesi V, Manzoni F, Cervellini G, Tomaselli A, Pravatà V, Garrido F, Villamor E, Mosca F, Cavallaro G. Endothelial dysfunction in preterm infants: The hidden legacy of uteroplacental pathologies. Front Pediatr 2022; 10:1041919. [PMID: 36405831 PMCID: PMC9671930 DOI: 10.3389/fped.2022.1041919] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Accepted: 10/14/2022] [Indexed: 11/06/2022] Open
Abstract
Millions of infants are born prematurely every year worldwide. Prematurity, particularly at lower gestational ages, is associated with high mortality and morbidity and is a significant global health burden. Pregnancy complications and preterm birth syndrome strongly impact neonatal clinical phenotypes and outcomes. The vascular endothelium is a pivotal regulator of fetal growth and development. In recent years, the key role of uteroplacental pathologies impairing endothelial homeostasis is emerging. Conditions leading to very and extremely preterm birth can be classified into two main pathophysiological patterns or endotypes: infection/inflammation and dysfunctional placentation. The first is frequently related to chorioamnionitis, whereas the second is commonly associated with hypertensive disorders of pregnancy and fetal growth restriction. The nature, timing, and extent of prenatal noxa may alter fetal and neonatal endothelial phenotype and functions. Changes in the luminal surface, oxidative stress, growth factors imbalance, and dysregulation of permeability and vascular tone are the leading causes of endothelial dysfunction in preterm infants. However, the available evidence regarding endothelial physiology and damage is limited in neonates compared to adults. Herein, we discuss the current knowledge on endothelial dysfunction in the infectious/inflammatory and dysfunctional placentation endotypes of prematurity, summarizing their molecular features, available biomarkers, and clinical impact. Furthermore, knowledge gaps, shadows, and future research perspectives are highlighted.
Collapse
Affiliation(s)
- Giacomo Simeone Amelio
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Livia Provitera
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Genny Raffaeli
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Clinical Sciences and Community Health, Università Degli Studi di Milano, Milan, Italy
| | - Matteo Tripodi
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Ilaria Amodeo
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Silvia Gulden
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Valeria Cortesi
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Clinical Sciences and Community Health, Università Degli Studi di Milano, Milan, Italy
| | - Francesca Manzoni
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Clinical Sciences and Community Health, Università Degli Studi di Milano, Milan, Italy
| | - Gaia Cervellini
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Clinical Sciences and Community Health, Università Degli Studi di Milano, Milan, Italy
| | - Andrea Tomaselli
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Clinical Sciences and Community Health, Università Degli Studi di Milano, Milan, Italy
| | - Valentina Pravatà
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Felipe Garrido
- Department of Pediatrics, Clínica Universidad de Navarra, Madrid, Spain
| | - Eduardo Villamor
- Department of Pediatrics, Maastricht University Medical Center (MUMC+), School for Oncology and Reproduction (GROW), University of Maastricht, Maastricht, Netherlands
| | - Fabio Mosca
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Clinical Sciences and Community Health, Università Degli Studi di Milano, Milan, Italy
| | - Giacomo Cavallaro
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
24
|
The effects of real and simulated microgravity on cellular mitochondrial function. NPJ Microgravity 2021; 7:44. [PMID: 34750383 PMCID: PMC8575887 DOI: 10.1038/s41526-021-00171-7] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 10/07/2021] [Indexed: 11/22/2022] Open
Abstract
Astronauts returning from space shuttle missions or the International Space Station have been diagnosed with various health problems such as bone demineralization, muscle atrophy, cardiovascular deconditioning, and vestibular and sensory imbalance including visual acuity, altered metabolic and nutritional status, and immune system dysregulation. These health issues are associated with oxidative stress caused by a microgravity environment. Mitochondria are a source of reactive oxygen species (ROS). However, the molecular mechanisms through which mitochondria produce ROS in a microgravity environment remain unclear. Therefore, this review aimed to explore the mechanism through which microgravity induces oxidative damage in mitochondria by evaluating the expression of genes and proteins, as well as relevant metabolic pathways. In general, microgravity-induced ROS reduce mitochondrial volume by mainly affecting the efficiency of the respiratory chain and metabolic pathways. The impaired respiratory chain is thought to generate ROS through premature electron leakage in the electron transport chain. The imbalance between ROS production and antioxidant defense in mitochondria is the main cause of mitochondrial stress and damage, which leads to mitochondrial dysfunction. Moreover, we discuss the effects of antioxidants against oxidative stress caused by the microgravity environment space microgravity in together with simulated microgravity (i.e., spaceflight or ground-based spaceflight analogs: parabolic flight, centrifugal force, drop towers, etc.). Further studies should be taken to explore the effects of microgravity on mitochondrial stress-related diseases, especially for the development of new therapeutic drugs that can help increase the health of astronauts on long space missions.
Collapse
|
25
|
Characterization of Mitochondrial Bioenergetics in Preeclampsia. J Clin Med 2021; 10:jcm10215063. [PMID: 34768583 PMCID: PMC8584662 DOI: 10.3390/jcm10215063] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 10/19/2021] [Accepted: 10/24/2021] [Indexed: 01/07/2023] Open
Abstract
Preeclampsia (PE) is characterized by new onset hypertension during pregnancy and is associated with oxidative stress, placental ischemia, and autoantibodies to the angiotensin II type I receptor (AT1-AA). Mitochondrial (mt) dysfunction in PE and various sources of oxidative stress, such as monocytes, neutrophils, and CD4 + T cells, have been identified as important players in the pathophysiology of PE. We have established the significance of AT1-AA, TNF-α, and CD4 + T cells in causing mitochondrial (mt) dysfunction in renal and placental tissues in pregnant rats. Although the role of mt dysfunction from freshly isolated intact placental mitochondria has been compared in human PE and normally pregnant (NP) controls, variations among preterm PE or term PE have not been compared and mechanisms contributing to mt ROS during PE are unclear. Therefore, we hypothesized PE placentas would exhibit impaired placental mt function, which would be worse in preterm PE patients than in those of later gestational ages. Immediately after delivery, PE and NP patient’s placentas were collected, mt were isolated and mt respiration and ROS were measured. PE patients at either < or >34 weeks gestational age (GA) exhibited elevated blood pressure and decreased placental mt respiration rates (state 3 and maximal). Patients delivering at >34 weeks exhibited decreased Complex IV activity and expression. Placental mtROS was significantly reduced in both PE groups, compared to NP placental mitochondria. Collectively, the study demonstrates that PE mt dysfunction occurs in the placenta, with mtROS being lower than that seen in NP controls. These data indicate why antioxidants, as a potential target or new therapeutic agent, may not be ideal in treating the oxidative stress associated with PE.
Collapse
|
26
|
Deer E, Jones J, Cornelius DC, Comley K, Herrock O, Campbell N, Fitzgerald S, Ibrahim T, LaMarca B, Amaral LM. Progesterone Induced Blocking Factor Reduces Hypertension and Placental Mitochondrial Dysfunction in Response to sFlt-1 during Pregnancy. Cells 2021; 10:2817. [PMID: 34831040 PMCID: PMC8616090 DOI: 10.3390/cells10112817] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 10/06/2021] [Accepted: 10/10/2021] [Indexed: 01/12/2023] Open
Abstract
Preeclampsia (PE) is characterized by new onset hypertension in association with placental ischemia, reduced fetal weight, elevated soluble fms-like tyrosine kinase-1 (sFlt-1), and placental mitochondrial (mt) dysfunction and oxidative stress (ROS). Progesterone induced blocking factor (PIBF) is a product of progesterone signaling that blocks inflammatory processes and we have previously shown PIBF to lower mean arterial blood pressure (MAP) and sFlt-1 in a rat model of PE. Infusion of sFlt-1 causes hypertension and many characteristics of PE in pregnant rodents, however, its role in causing mt dysfunction is unknown. Therefore, we hypothesize that PIBF will improve mt function and MAP in response to elevated sFlt-1 during pregnancy. We tested our hypothesis by infusing sFlt-1 via miniosmotic pumps in normal pregnant (NP) Sprague-Dawley rats (3.7 μg·kg-1·day-1) on gestation days (GD) 13-19 in the presence or absence of PIBF (2.0 µg/mL) injected intraperitoneally on GD 15 and examined mean arterial blood pressure (MAP) and placental mt ROS on GD 19. sFlt-1 increased MAP to 112 + 2 (n = 11) compared to NP rats (98 + 2 mmHg, n = 15, p < 0.05), which was lowered in the presence of sFlt-1 (100 + 1 mmHg, n = 5, p < 0.05). Placental mtATP was reduced in sFlt-1 infused rats versus NP controls, but was improved with PIBF. Placental mtROS was elevated with sFlt-1 compared to NP controls, but was reduced with PIBF. Sera from NP + sFlt-1 increased endothelial cell mtROS, which was attenuated with PIBF. These data demonstrate sFlt-1 induced HTN during pregnancy reduces placental mt function. Importantly, PIBF improved placental mt function and HTN, indicating the efficacy of improved progesterone signaling as potential therapeutics for PE.
Collapse
Affiliation(s)
- Evangeline Deer
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA; (E.D.); (J.J.); (K.C.); (O.H.); (N.C.); (S.F.); (T.I.); (B.L.)
| | - Jalisa Jones
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA; (E.D.); (J.J.); (K.C.); (O.H.); (N.C.); (S.F.); (T.I.); (B.L.)
| | - Denise C. Cornelius
- Department of Emergency Medicine, University of Mississippi Medical Center, Jackson, MS 39126, USA;
| | - Kyleigh Comley
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA; (E.D.); (J.J.); (K.C.); (O.H.); (N.C.); (S.F.); (T.I.); (B.L.)
| | - Owen Herrock
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA; (E.D.); (J.J.); (K.C.); (O.H.); (N.C.); (S.F.); (T.I.); (B.L.)
| | - Nathan Campbell
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA; (E.D.); (J.J.); (K.C.); (O.H.); (N.C.); (S.F.); (T.I.); (B.L.)
| | - Sarah Fitzgerald
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA; (E.D.); (J.J.); (K.C.); (O.H.); (N.C.); (S.F.); (T.I.); (B.L.)
| | - Tarek Ibrahim
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA; (E.D.); (J.J.); (K.C.); (O.H.); (N.C.); (S.F.); (T.I.); (B.L.)
| | - Babbette LaMarca
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA; (E.D.); (J.J.); (K.C.); (O.H.); (N.C.); (S.F.); (T.I.); (B.L.)
- Department of Obstetrics and Gynecology, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Lorena M. Amaral
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA; (E.D.); (J.J.); (K.C.); (O.H.); (N.C.); (S.F.); (T.I.); (B.L.)
| |
Collapse
|
27
|
Deer E, Amaral LM, Campbell N, Fitzgerald S, Herrock O, Ibrahim T, LaMarca B. Low Dose of IL-2 Normalizes Hypertension and Mitochondrial Function in the RUPP Rat Model of Placental Ischemia. Cells 2021; 10:2797. [PMID: 34685775 PMCID: PMC8534834 DOI: 10.3390/cells10102797] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 10/08/2021] [Accepted: 10/12/2021] [Indexed: 12/30/2022] Open
Abstract
IL-2 is a cytokine released from CD4+T cells with dual actions and can either potentiate the inflammatory response or quell a chronic inflammatory response depending on its circulating concentration. IL-2 is elevated in many chronic inflammatory conditions and is increased during preeclampsia (PE). PE is characterized by new-onset hypertension during pregnancy and organ dysfunction and increasing evidence indicates that proinflammatory cytokines cause hypertension and mitochondrial (mt) dysfunction during pregnancy. The reduced uterine perfusion pressure (RUPP) model of placental ischemia is a rat model of PE that we commonly use in our laboratory and we have previously shown that low doses of recombinant IL-2 can decrease blood pressure in RUPP rats. The objective of this study was to determine the effects of a low dose of recombinant IL-2 on multi-organ mt dysfunction in the RUPP rat model of PE. We tested our hypothesis by infusing recombinant IL-2 (0.05 ng/mL) into RUPP rats on GD14 and examined mean arterial pressure (MAP), renal, placental and endothelial cell mt function compared to control RUPP. MAP was elevated in RUPP rats (n = 6) compared to controls (n = 5) (122 ± 5 vs. 102 ± 3 mmHg, p < 0.05), but was reduced by administration of LD recombinant IL-2 (107 ± 1 vs. 122 ± 5 mmHg, n = 9, p < 0.05). Renal, placental and endothelial mt ROS were significantly increased in RUPP rats compared to RUPP+ IL-2 and controls. Placental and renal respiration rates were reduced in RUPP rats compared to control rats but were normalized with IL-2 administration to RUPPs. These data indicate that low-dose IL-2 normalized multi-organ mt function and hypertension in response to placental ischemia.
Collapse
Affiliation(s)
- Evangeline Deer
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA; (E.D.); (L.M.A.); (N.C.); (S.F.); (O.H.); (T.I.)
| | - Lorena M. Amaral
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA; (E.D.); (L.M.A.); (N.C.); (S.F.); (O.H.); (T.I.)
| | - Nathan Campbell
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA; (E.D.); (L.M.A.); (N.C.); (S.F.); (O.H.); (T.I.)
| | - Sarah Fitzgerald
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA; (E.D.); (L.M.A.); (N.C.); (S.F.); (O.H.); (T.I.)
| | - Owen Herrock
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA; (E.D.); (L.M.A.); (N.C.); (S.F.); (O.H.); (T.I.)
| | - Tarek Ibrahim
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA; (E.D.); (L.M.A.); (N.C.); (S.F.); (O.H.); (T.I.)
| | - Babbette LaMarca
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA; (E.D.); (L.M.A.); (N.C.); (S.F.); (O.H.); (T.I.)
- Department of Obstetrics and Gynecology, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Departments of Pharmacology, Physiology, and Obstetrics and Gynecology, Center for Excellence in Cardiovascular and Renal Research, University of Mississippi Medical Center, Jackson, MS 39216, USA
| |
Collapse
|
28
|
Impaired placental mitophagy and oxidative stress are associated with dysregulated BNIP3 in preeclampsia. Sci Rep 2021; 11:20469. [PMID: 34650122 PMCID: PMC8516954 DOI: 10.1038/s41598-021-99837-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 09/29/2021] [Indexed: 12/19/2022] Open
Abstract
Preeclampsia (PE) is a severe multisystem pregnancy complication characterized by gestational hypertension and proteinuria. Bcl-2/adenovirus E1B 19-kDa interacting protein 3 (BNIP3) is a mediator of mitophagy and has been proven to be associated with PE, but the mechanism is not well understood. This study aimed to investigate the role of BNIP3 in PE. Placentae from preeclamptic and normal pregnancies were analyzed by western-blot and transmission electron microscopy to quantify the level of BNIP3 expression and observe the organelle morphologies. Trophoblast cells with knockdown BNIP3 were analyzed by western-blot, immunofluorescence, flow cytometry, migration and invasion assays. BNIP3 expression was suppressed in PE patients. Impaired autophagy and increased mitochondrial damage were observed in PE placentae when compared with normal placentae. Suppression of BNIP3 inhibited Beclin-1 expression and reduced the transformation of LC3-I to LC3-II. In the knockdown BNIP3 group, p62 was overexpressed, ROS accumulated and the apoptotic process was elevated under oxidative stress condition. The knockdown of BNIP3 reduced the colocalization of GFP-LC3 and mitochondria. The findings of this study suggest that dysregulated BNIP3 is associated with impaired mitophagy, oxidative stress, and apoptosis in PE. The study provides new insights into the role of BNIP3 in the pathophysiology of PE.
Collapse
|
29
|
Effect of Endogenic and Exogenic Oxidative Stress Triggers on Adverse Pregnancy Outcomes: Preeclampsia, Fetal Growth Restriction, Gestational Diabetes Mellitus and Preterm Birth. Int J Mol Sci 2021; 22:ijms221810122. [PMID: 34576285 PMCID: PMC8468091 DOI: 10.3390/ijms221810122] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 09/10/2021] [Accepted: 09/16/2021] [Indexed: 12/12/2022] Open
Abstract
Oxidative stress is caused by an imbalance between the production of reactive oxygen species (ROS) in cells and tissues and the ability of a biological system to detoxify them. During a normal pregnancy, oxidative stress increases the normal systemic inflammatory response and is usually well-controlled by the balanced body mechanism of the detoxification of anti-oxidative products. However, pregnancy is also a condition in which this adaptation and balance can be easily disrupted. Excessive ROS is detrimental and associated with many pregnancy complications, such as preeclampsia (PE), fetal growth restriction (FGR), gestational diabetes mellitus (GDM), and preterm birth (PTB), by damaging placentation. The placenta is a tissue rich in mitochondria that produces the majority of ROS, so it is important to maintain normal placental function and properly develop its vascular network to ensure a safe and healthy pregnancy. Antioxidants may ameliorate these diseases, and related research is progressing. This review aimed to determine the association between oxidative stress and adverse pregnancy outcomes, especially PE, FGR, GDM, and PTB, and explore how to overcome this oxidative stress in these unfavorable conditions.
Collapse
|
30
|
Correia Y, Scheel J, Gupta S, Wang K. Placental mitochondrial function as a driver of angiogenesis and placental dysfunction. Biol Chem 2021; 402:887-909. [PMID: 34218539 DOI: 10.1515/hsz-2021-0121] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 06/09/2021] [Indexed: 12/12/2022]
Abstract
The placenta is a highly vascularized and complex foetal organ that performs various tasks, crucial to a healthy pregnancy. Its dysfunction leads to complications such as stillbirth, preeclampsia, and intrauterine growth restriction. The specific cause of placental dysfunction remains unknown. Recently, the role of mitochondrial function and mitochondrial adaptations in the context of angiogenesis and placental dysfunction is getting more attention. The required energy for placental remodelling, nutrient transport, hormone synthesis, and the reactive oxygen species leads to oxidative stress, stemming from mitochondria. Mitochondria adapt to environmental changes and have been shown to adjust their oxygen and nutrient use to best support placental angiogenesis and foetal development. Angiogenesis is the process by which blood vessels form and is essential for the delivery of nutrients to the body. This process is regulated by different factors, pro-angiogenic factors and anti-angiogenic factors, such as sFlt-1. Increased circulating sFlt-1 levels have been linked to different preeclamptic phenotypes. One of many effects of increased sFlt-1 levels, is the dysregulation of mitochondrial function. This review covers mitochondrial adaptations during placentation, the importance of the anti-angiogenic factor sFlt-1in placental dysfunction and its role in the dysregulation of mitochondrial function.
Collapse
Affiliation(s)
- Yolanda Correia
- Aston Medical School, College of Health & Life Sciences, Aston University, Aston Triangle, Birmingham B4 7ET, UK
| | - Julia Scheel
- Department of Systems Biology and Bioinformatics, University of Rostock, D-18051 Rostock, Germany
| | - Shailendra Gupta
- Department of Systems Biology and Bioinformatics, University of Rostock, D-18051 Rostock, Germany
| | - Keqing Wang
- Aston Medical School, College of Health & Life Sciences, Aston University, Aston Triangle, Birmingham B4 7ET, UK
| |
Collapse
|
31
|
Pravda J. Sepsis: Evidence-based pathogenesis and treatment. World J Crit Care Med 2021; 10:66-80. [PMID: 34316443 PMCID: PMC8291008 DOI: 10.5492/wjccm.v10.i4.66] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 04/13/2021] [Accepted: 06/02/2021] [Indexed: 02/06/2023] Open
Abstract
Sepsis can develop during the body’s response to a critical illness leading to multiple organ failure, irreversible shock, and death. Sepsis has been vexing health care providers for centuries due to its insidious onset, generalized metabolic dysfunction, and lack of specific therapy. A common factor underlying sepsis is the characteristic hypermetabolic response as the body ramps up every physiological system in its fight against the underlying critical illness. A hypermetabolic response requires supraphysiological amounts of energy, which is mostly supplied via oxidative phosphorylation generated ATP. A by-product of oxidative phosphorylation is hydrogen peroxide (H2O2), a toxic, membrane-permeable oxidizing agent that is produced in far greater amounts during a hypermetabolic state. Continued production of mitochondrial H2O2 can overwhelm cellular reductive (antioxidant) capacity leading to a build-up within cells and eventual diffusion into the bloodstream. H2O2 is a metabolic poison that can inhibit enzyme systems leading to organ failure, microangiopathic dysfunction, and irreversible septic shock. The toxic effects of H2O2 mirror the clinical and laboratory abnormalities observed in sepsis, and toxic levels of blood H2O2 have been reported in patients with septic shock. This review provides evidence to support a causal role for H2O2 in the pathogenesis of sepsis, and an evidence-based therapeutic intervention to reduce H2O2 levels in the body and restore redox homeostasis, which is necessary for normal organ function and vascular responsiveness.
Collapse
Affiliation(s)
- Jay Pravda
- Inflammatory Disease Research Centre, Therashock LLC, Palm Beach Gardens, FL 33410, United States
| |
Collapse
|
32
|
Sanchez-Aranguren L, Nadeem S. Bioenergetics adaptations and redox homeostasis in pregnancy and related disorders. Mol Cell Biochem 2021; 476:4003-4018. [PMID: 34196872 PMCID: PMC8473347 DOI: 10.1007/s11010-021-04215-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 06/21/2021] [Indexed: 12/14/2022]
Abstract
Pregnancy is a challenging physiological process that involves maternal adaptations to the increasing energetics demands imposed by the growing conceptus. Failure to adapt to these requirements may result in serious health complications for the mother and the baby. The mitochondria are biosynthetic and energy-producing organelles supporting the augmented energetic demands of pregnancy. Evidence suggests that placental mitochondria display a dynamic phenotype through gestation. At early stages of pregnancy placental mitochondria are mainly responsible for the generation of metabolic intermediates and reactive oxygen species (ROS), while at later stages of gestation, the placental mitochondria exhibit high rates of oxygen consumption. This review describes the metabolic fingerprint of the placental mitochondria at different stages of pregnancy and summarises key signs of mitochondrial dysfunction in pathological pregnancy conditions, including preeclampsia, gestational diabetes and intrauterine growth restriction (IUGR). So far, the effects of placental-driven metabolic changes governing the metabolic adaptations occurring in different maternal tissues in both, healthy and pathological pregnancies, remain to be uncovered. Understanding the function and molecular aspects of the adaptations occurring in placental and maternal tissue's mitochondria will unveil potential targets for further therapeutic exploration that could address pregnancy-related disorders. Targeting mitochondrial metabolism is an emerging approach for regulating mitochondrial bioenergetics. This review will also describe the potential therapeutic use of compounds with a recognised effect on mitochondria, for the management of preeclampsia.
Collapse
Affiliation(s)
| | - Sarah Nadeem
- College of Health and Life Sciences, Aston Medical School, Aston University, Birmingham, UK
| |
Collapse
|
33
|
Resveratrol and endothelial function: A literature review. Pharmacol Res 2021; 170:105725. [PMID: 34119624 DOI: 10.1016/j.phrs.2021.105725] [Citation(s) in RCA: 106] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/08/2021] [Accepted: 06/08/2021] [Indexed: 12/12/2022]
Abstract
Endothelial dysfunction is a major contributing factor to diseases such as atherosclerosis, diabetes mellitus, obesity, hypertension, acute lung injury, preeclampsia, among others. Resveratrol (RSV) is a naturally occurring bioactive polyphenol found in grapes and red wine. According to experimental studies, RSV modulates several events involved in endothelial dysfunction such as impaired vasorelaxation, eNOS uncoupling, leukocyte adhesion, endothelial senescence, and endothelial mesenchymal transition. The endothelial protective effects of RSV are found to be mediated by numerous molecular targets (e.g. Silent Information Regulator 1 (SIRT1), 5' AMP-activated protein kinase (AMPK), endothelial nitric oxide synthase (eNOS), nuclear factor-erythroid-derived 2-related factor-2 (Nrf2), peroxisome proliferator-activated receptor (PPAR), Krüppel-like factor-2 (KLF2), and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kB)). Herein, we present an updated review addressing pharmacological effects and molecular targets of RSV in maintaining endothelial function, and the potential of this phytochemical for endothelial dysfunction-associated disorders.
Collapse
|
34
|
Mitochondrial dynamics and reactive oxygen species initiate thrombopoiesis from mature megakaryocytes. Blood Adv 2021; 5:1706-1718. [PMID: 33720340 DOI: 10.1182/bloodadvances.2020002847] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 01/20/2021] [Indexed: 02/07/2023] Open
Abstract
Blood platelets are essential for controlling hemostasis. They are released by megakaryocytes (MKs) located in the bone marrow, upon extension of cytoplasmic protrusions into the lumen of bone marrow sinusoids. Their number increases in postpulmonary capillaries, suggesting a role for oxygen gradient in thrombopoiesis (ie, platelet biogenesis). In this study, we show that initiation of thrombopoiesis from human mature MKs was enhanced under hyperoxia or during pro-oxidant treatments, whereas antioxidants dampened it. Quenching mitochondrial reactive oxygen species (mtROS) with MitoTEMPO decreased thrombopoiesis, whereas genetically enhancing mtROS by deacetylation-null sirtuin-3 expression increased it. Blocking cytosolic ROS production by NOX inhibitors had no impact. Classification according to the cell roundness index delineated 3 stages of thrombopoiesis in mature MKs. Early-stage round MKs exhibited the highest index, which correlated with low mtROS levels, a mitochondrial tubular network, and the mitochondrial recruitment of the fission activator Drp1. Intermediate MKs at the onset of thrombopoiesis showed high mtROS levels and small, well-delineated mitochondria. Terminal MKs showed the lowest roundness index and long proplatelet extensions. Inhibiting Drp1-dependent mitochondrial fission of mature MKs by Mdivi-1 favored a tubular mitochondrial network and lowered both mtROS levels and intermediate MKs proportion, whereas enhancing Drp1 activity genetically had opposite effects. Reciprocally, quenching mtROS limited mitochondrial fission in round MKs. These data demonstrate a functional coupling between ROS and mitochondrial fission in MKs, which is crucial for the onset of thrombopoiesis. They provide new molecular cues that control initiation of platelet biogenesis and may help elucidate some unexplained thrombocytopenia.
Collapse
|
35
|
Barron A, McCarthy CM, O'Keeffe GW. Preeclampsia and Neurodevelopmental Outcomes: Potential Pathogenic Roles for Inflammation and Oxidative Stress? Mol Neurobiol 2021; 58:2734-2756. [PMID: 33492643 DOI: 10.1007/s12035-021-02290-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 01/12/2021] [Indexed: 12/13/2022]
Abstract
Preeclampsia (PE) is a common and serious hypertensive disorder of pregnancy that occurs in approximately 3-5% of first-time pregnancies and is a well-known leading cause of maternal and neonatal mortality and morbidity. In recent years, there has been accumulating evidence that in utero exposure to PE acts as an environmental risk factor for various neurodevelopmental disorders, particularly autism spectrum disorder and ADHD. At present, the mechanism(s) mediating this relationship are uncertain. In this review, we outline the most recent evidence implicating a causal role for PE exposure in the aetiology of various neurodevelopmental disorders and provide a novel interpretation of neuroanatomical alterations in PE-exposed offspring and how these relate to their sub-optimal neurodevelopmental trajectory. We then postulate that inflammation and oxidative stress, two prominent features of the pathophysiology of PE, are likely to play a major role in mediating this association. The increased inflammation in the maternal circulation, placenta and fetal circulation in PE expose the offspring to both prenatal maternal immune activation-a risk factor for neurodevelopmental disorders, which has been well-characterised in animal models-and directly higher concentrations of pro-inflammatory cytokines, which adversely affect neuronal development. Similarly, the exaggerated oxidative stress in the mother, placenta and foetus induces the placenta to secrete factors deleterious to neurons, and exposes the fetal brain to directly elevated oxidative stress and thus adversely affects neurodevelopmental processes. Finally, we describe the interplay between inflammation and oxidative stress in PE, and how both systems interact to potentially alter neurodevelopmental trajectory in exposed offspring.
Collapse
Affiliation(s)
- Aaron Barron
- Department of Anatomy and Neuroscience, University College, Cork, Ireland.,Department of Pharmacology and Therapeutics, University College Cork, Cork, Ireland
| | - Cathal M McCarthy
- Department of Pharmacology and Therapeutics, University College Cork, Cork, Ireland.
| | - Gerard W O'Keeffe
- Department of Anatomy and Neuroscience, University College, Cork, Ireland. .,Cork Neuroscience Centre, University College Cork, Cork, Ireland.
| |
Collapse
|
36
|
Deer E, Vaka VR, McMaster KM, Wallace K, Cornelius DC, Amaral LM, Cunningham MW, LaMarca B. Vascular endothelial mitochondrial oxidative stress in response to preeclampsia: a role for angiotension II type 1 autoantibodies. Am J Obstet Gynecol MFM 2021; 3:100275. [PMID: 33451592 PMCID: PMC8092846 DOI: 10.1016/j.ajogmf.2020.100275] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 10/15/2020] [Accepted: 10/26/2020] [Indexed: 01/10/2023]
Abstract
BACKGROUND Preeclampsia is characterized by a new onset of hypertension during pregnancy and is associated with autoantibodies against the angiotensin II type 1 receptor and oxidative stress. There is growing evidence for mitochondrial dysfunction in preeclampsia, however, the culprits for mitochondrial dysfunction are still being defined. We previously demonstrated that angiotensin II type 1 autoantibodies cause renal, placental, and endothelial mitochondrial dysfunction in pregnant rats. However, the role of the angiotensin II type 1 autoantibodies in endothelial mitochondrial function in response to sera from preeclamptics is unknown. Thus, we hypothesized that circulating factors, such as the angiotensin II type 1 autoantibodies, during preeclampsia would negatively impact the vascular endothelial mitochondrial function in human umbilical vein endothelial cells. OBJECTIVE The objective of the study was to determine a role for circulating angiotensin II type 1 autoantibodies to cause endothelial mitochondrial reactive oxygen species and dysfunction in preeclampsia compared to normal pregnant controls. STUDY DESIGN Immediately after delivery, sera was collected from preeclamptic patients and normal pregnant controls. The mitochondrial reactive oxygen species were determined from the cells treated overnight with 10% sera from either the control or preeclamptic patients with and without the antiotension II type 1 autoantibodies inhibitor peptide ('n7AAc'). RESULTS Preeclampsia patients at <34 weeks' gestation exhibited an elevated mean arterial blood pressure. Cells treated with serum from the preeclampsia patients at <34 weeks gestational age showed significantly (P<0.05) greater mitochondrial oxidative stress and reduced respiration than cells treated with the control sera, and these abnormalities were restored with 'n7AAc'. CONCLUSION This study demonstrates that endothelial mitochondrial dysfunction occurs in response to circulating factors, especially in response to serum from preterm preeclampsia patients, and can be restored by blocking circulating angiotensin II type 1 autoantibodies, thereby indicating a potential new therapeutic target for preeclampsia.
Collapse
Affiliation(s)
- Evangeline Deer
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS
| | - V Ramana Vaka
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS
| | - Kristen M McMaster
- Department of Obstetrics and Gynecology, University of Mississippi Medical Center, Jackson, MS
| | - Kedra Wallace
- Department of Obstetrics and Gynecology, University of Mississippi Medical Center, Jackson, MS
| | - Denise C Cornelius
- Department of Emergency Medicine, University of Mississippi Medical Center, Jackson, MS
| | - Lorena M Amaral
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS
| | - Mark W Cunningham
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS
| | - Babbette LaMarca
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS; Department of Obstetrics and Gynecology, University of Mississippi Medical Center, Jackson, MS.
| |
Collapse
|
37
|
Pandey D, Yevale A, Naha R, Kuthethur R, Chakrabarty S, Satyamoorthy K. Mitochondrial DNA copy number variation - A potential biomarker for early onset preeclampsia. Pregnancy Hypertens 2020; 23:1-4. [PMID: 33160129 DOI: 10.1016/j.preghy.2020.10.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 09/27/2020] [Accepted: 10/02/2020] [Indexed: 10/23/2022]
Abstract
OBJECTIVES Oxidative stress has been hypothesized as a central component of both placental and endothelial dysfunction, leading to PE. This oxidative stress leading to mitochondrial dysfunction may be due to variations in mtDNA copy numbers as an adaptive response. In the present study we aimed to analyse mtDNA copy numbers in the placenta obtained after delivery from the women with PE as compared to the controls. STUDY DESIGN It was a prospective case control study. A total of 32 placental samples were analyzed (Cases: 17; Controls: 15). Samples were collected ex vivo, after childbirth. MtDNA content was determined useing real-time quantitative PCR qRT-PCR) using TaqMan probes designed for two genes: MT-ND1 and a mitochondrial gene encoding for the NADH dehydrogenase 1 protein. RESULTS We found that the median (IQR) mtDNA copy number was higher in PE cases 24.32 (9.260-33.51) as compared with controls 20.32 (13.33-26.22). On subgroup analysis, the median (IQR) mtDNA copy number was higher in early onset PE 28.06 (20.80-36.87) as compared to late onset PE 9.215 (4.150-56.45) as well as the controls 20.32 (13.33-26.22). CONCLUSION Our findings support a higher mtDNA copy number in early onset PE as compared to late onset PE and control population. Although, mtDNA may only be increased in very severe cases of early onset preeclampsia. Future research may be directed to ascertain if mtDNA copy numbers can be a novel biomarker to predict or prognosticate early onset preeclampsia.
Collapse
|
38
|
Brand MD. Riding the tiger - physiological and pathological effects of superoxide and hydrogen peroxide generated in the mitochondrial matrix. Crit Rev Biochem Mol Biol 2020; 55:592-661. [PMID: 33148057 DOI: 10.1080/10409238.2020.1828258] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Elevated mitochondrial matrix superoxide and/or hydrogen peroxide concentrations drive a wide range of physiological responses and pathologies. Concentrations of superoxide and hydrogen peroxide in the mitochondrial matrix are set mainly by rates of production, the activities of superoxide dismutase-2 (SOD2) and peroxiredoxin-3 (PRDX3), and by diffusion of hydrogen peroxide to the cytosol. These considerations can be used to generate criteria for assessing whether changes in matrix superoxide or hydrogen peroxide are both necessary and sufficient to drive redox signaling and pathology: is a phenotype affected by suppressing superoxide and hydrogen peroxide production; by manipulating the levels of SOD2, PRDX3 or mitochondria-targeted catalase; and by adding mitochondria-targeted SOD/catalase mimetics or mitochondria-targeted antioxidants? Is the pathology associated with variants in SOD2 and PRDX3 genes? Filtering the large literature on mitochondrial redox signaling using these criteria highlights considerable evidence that mitochondrial superoxide and hydrogen peroxide drive physiological responses involved in cellular stress management, including apoptosis, autophagy, propagation of endoplasmic reticulum stress, cellular senescence, HIF1α signaling, and immune responses. They also affect cell proliferation, migration, differentiation, and the cell cycle. Filtering the huge literature on pathologies highlights strong experimental evidence that 30-40 pathologies may be driven by mitochondrial matrix superoxide or hydrogen peroxide. These can be grouped into overlapping and interacting categories: metabolic, cardiovascular, inflammatory, and neurological diseases; cancer; ischemia/reperfusion injury; aging and its diseases; external insults, and genetic diseases. Understanding the involvement of mitochondrial matrix superoxide and hydrogen peroxide concentrations in these diseases can facilitate the rational development of appropriate therapies.
Collapse
|
39
|
Bioenergetic effects of hydrogen sulfide suppress soluble Flt-1 and soluble endoglin in cystathionine gamma-lyase compromised endothelial cells. Sci Rep 2020; 10:15810. [PMID: 32978411 PMCID: PMC7519095 DOI: 10.1038/s41598-020-72371-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 08/26/2020] [Indexed: 12/14/2022] Open
Abstract
Endothelial dysfunction is a hallmark of preeclampsia, a life-threatening complication of pregnancy characterised by hypertension and elevated soluble Fms-Like Tyrosine Kinase-1 (sFlt-1). Dysregulation of hydrogen sulfide (H2S) by inhibition of cystathionine γ-lyase (CSE) increases sFlt-1 and soluble endoglin (sEng) release. We explored whether compromise in CSE/H2S pathway is linked to dysregulation of the mitochondrial bioenergetics and oxidative status. We investigated whether these effects were linked to CSE-induced sFlt-1 and sEng production in endothelial cells. Here, we demonstrate that CSE/H2S pathway sustain endothelial mitochondrial bioenergetics and loss of CSE increases the production of mitochondrial-specific superoxide. As a compensatory effect, low CSE environment enhances the reliance on glycolysis. The mitochondrial-targeted H2S donor, AP39, suppressed the antiangiogenic response and restored the mitochondrial bioenergetics in endothelial cells. AP39 revealed that upregulation of sFlt-1, but not sEng, is independent of the mitochondrial H2S metabolising enzyme, SQR. These data provide new insights into the molecular mechanisms for antiangiogenic upregulation in a mitochondrial-driven environment. Targeting H2S to the mitochondria may be of therapeutic benefit in the prevention of endothelial dysfunction associated with preeclampsia.
Collapse
|
40
|
Giller A, Andrawus M, Gutman D, Atzmon G. Pregnancy as a model for aging. Ageing Res Rev 2020; 62:101093. [PMID: 32502628 DOI: 10.1016/j.arr.2020.101093] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 04/21/2020] [Accepted: 05/23/2020] [Indexed: 12/12/2022]
Abstract
The process of aging can be defined as the sum accumulation of damages and changes in metabolism during the life of an organism, due to both genetic predisposition and stochastic damage. During the gestational period and following parturition, similar damage can be seen due to the strenuous effect on the maternal body, exhibited on both the physiological and cellular level. In this review, we will focus on the similar physiological and cellular characteristics exhibited during pregnancy and aging, including induction of and response to oxidative stress, inflammation, and degradation of telomeres. We will evaluate any similar processes between aging and pregnancy by comparing common biomarkers, pathologies, and genetic and epigenetic effects, to establish the pregnant body as a model for aging. This review will approach the connection both in respect to current theories on aging as a byproduct of natural selection, and regarding unrelated biochemical similarities between the two, drawing on existing studies and models in humans and other species where relevant alike. Furthermore, we will show the response of the pregnant body to these changes, and through that illuminate unique areas of potential study to advance our knowledge of the maladies relating to aging and pregnancy, and an avenue for solutions.
Collapse
Affiliation(s)
- Abram Giller
- Faculty of Natural Sciences, University of Haifa, 199 Aba Khoushy Ave., Mount Carmel, Haifa, 349888, Israel
| | - Mariana Andrawus
- Faculty of Natural Sciences, University of Haifa, 199 Aba Khoushy Ave., Mount Carmel, Haifa, 349888, Israel
| | - Danielle Gutman
- Faculty of Natural Sciences, University of Haifa, 199 Aba Khoushy Ave., Mount Carmel, Haifa, 349888, Israel
| | - Gil Atzmon
- Faculty of Natural Sciences, University of Haifa, 199 Aba Khoushy Ave., Mount Carmel, Haifa, 349888, Israel; Departments of Genetics and Medicine, Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, New York, 10461, USA.
| |
Collapse
|
41
|
Mishra JS, Blesson CS, Kumar S. Testosterone Decreases Placental Mitochondrial Content and Cellular Bioenergetics. BIOLOGY 2020; 9:biology9070176. [PMID: 32698476 PMCID: PMC7407169 DOI: 10.3390/biology9070176] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 07/09/2020] [Accepted: 07/16/2020] [Indexed: 01/13/2023]
Abstract
Placental mitochondrial dysfunction plays a central role in the pathogenesis of preeclampsia. Since preeclampsia is a hyperandrogenic state, we hypothesized that elevated maternal testosterone levels induce damage to placental mitochondria and decrease bioenergetic profiles. To test this hypothesis, pregnant Sprague–Dawley rats were injected with vehicle or testosterone propionate (0.5 mg/kg/day) from gestation day (GD) 15 to 19. On GD20, the placentas were isolated to assess mitochondrial structure, copy number, ATP/ADP ratio, and biogenesis (Pgc-1α and Nrf1). In addition, in vitro cultures of human trophoblasts (HTR-8/SVneo) were treated with dihydrotestosterone (0.3, 1.0, and 3.0 nM), and bioenergetic profiles using seahorse analyzer were assessed. Testosterone exposure in pregnant rats led to a 2-fold increase in plasma testosterone levels with an associated decrease in placental and fetal weights compared with controls. Elevated maternal testosterone levels induced structural damage to the placental mitochondria and decreased mitochondrial copy number. The ATP/ADP ratio was reduced with a parallel decrease in the mRNA and protein expression of Pgc-1α and Nrf1 in the placenta of testosterone-treated rats compared with controls. In cultured trophoblasts, dihydrotestosterone decreased the mitochondrial copy number and reduced PGC-1α, NRF1 mRNA, and protein levels without altering the expression of mitochondrial fission/fusion genes. Dihydrotestosterone exposure induced significant mitochondrial energy deficits with a dose-dependent decrease in basal respiration, ATP-linked respiration, maximal respiration, and spare respiratory capacity. In summary, our study suggests that the placental mitochondrial dysfunction induced by elevated maternal testosterone might be a potential mechanism linking preeclampsia to feto-placental growth restriction.
Collapse
Affiliation(s)
- Jay S. Mishra
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA;
| | - Chellakkan S. Blesson
- Reproductive Endocrinology and Infertility Division, Department of Obstetrics and Gynecology, Baylor College of Medicine and Family Fertility Center, Texas Children’s Hospital, Houston, TX 77030, USA;
| | - Sathish Kumar
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA;
- Department of Obstetrics and Gynecology, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53792, USA
- Endocrinology-Reproductive Physiology Program, University of Wisconsin, Madison, WI 53715, USA
- Correspondence:
| |
Collapse
|
42
|
Mitochondrial function in immune cells in health and disease. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165845. [PMID: 32473386 DOI: 10.1016/j.bbadis.2020.165845] [Citation(s) in RCA: 143] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 05/18/2020] [Accepted: 05/21/2020] [Indexed: 02/06/2023]
Abstract
One of the main functions of mitochondria is production of ATP for cellular energy needs, however, it becomes more recognized that mitochondria are involved in differentiation and activation processes of immune cells. Upon activation, immune cells have a high need for energy. Immune cells have different strategies to generate this energy. In pro-inflammatory cells, such as activated monocytes and activated T and B cells, the energy is generated by increasing glycolysis, while in regulatory cells, such as regulatory T cells or M2 macrophages, energy is generated by increasing mitochondrial function and beta-oxidation. Except for being important for energy supply during activation, mitochondria also induce immune responses. During an infection, they release mitochondrial danger associated molecules (DAMPs) that resemble structures of bacterial derived pathogen associated molecular patterns (PAMPs). Such mitochondrial DAMPS are for instance mitochondrial DNA with hypomethylated CpG motifs or a specific lipid that is only present in prokaryotic bacteria and mitochondria, i.e. cardiolipin. Via release of such DAMPs, mitochondria guide the immune response towards an inflammatory response against pathogens. This is an important mechanism in early detection of an infection and in stimulating and sustaining immune responses to fight infections. However, mitochondrial DAMPs may also have a negative impact. If mitochondrial DAMPs are released by damaged cells, without the presence of an infection, such as after a trauma, mitochondrial DAMPs may induce an undesired inflammatory response, resulting in tissue damage and organ dysfunction. Thus, immune cells have developed mechanisms to prevent such undesired immune activation by mitochondrial components. In the present narrative review, we will describe the current view of mitochondria in regulation of immune responses. We will also discuss the current knowledge on disturbed mitochondrial function in immune cells in various immunological diseases.
Collapse
|
43
|
Chen YF, Hebert VY, Stadler K, Xue SY, Slaybaugh K, Luttrell-Williams E, Glover MC, Krzywanski DM, Dugas TR. Coenzyme Q10 Alleviates Chronic Nucleoside Reverse Transcriptase Inhibitor-Induced Premature Endothelial Senescence. Cardiovasc Toxicol 2020; 19:500-509. [PMID: 31020509 DOI: 10.1007/s12012-019-09520-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Human immunodeficiency virus (HIV)-infected patients undergoing antiretroviral therapy are afforded an increased lifespan but also exhibit an elevated incidence of cardiovascular disease. HIV therapy uses a combination drug approach, and nucleoside reverse transcriptase inhibitors (NRTI) are a backbone of this therapy. Endothelial dysfunction is an initiating event in cardiovascular disease etiology, and in our prior studies, NRTIs induced an endothelial dysfunction that was dependent upon mitochondrial oxidative stress. Moreover, short-term NRTI administration induced a mitophagy-associated endothelial toxicity and increased reactive oxygen species (ROS) production that was rescued by coenzyme Q10 (Q10) or overexpression of a mitochondrial antioxidant enzyme. Thus, our objective was to examine mitochondrial toxicity in endothelial cells after chronic NRTI treatment and evaluate Q10 as a potential adjunct therapy for preventing NRTI-induced mitochondrial toxicity. Human aortic endothelial cells (HAEC) were exposed to chronic NRTI treatment, with or without Q10. ROS production, cell proliferation rate, levels of senescence, and mitochondrial bioenergetic function were determined. Chronic NRTI increased ROS production but decreased population doubling. In addition, NRTI increased the accumulation of β-galactosidase, indicative of an accelerated rate of senescence. Moreover, ATP-linked respiration was diminished. Co-treatment with Q10 delayed the onset of NRTI-induced senescence, decreased ROS production and rescued the cells' mitochondrial respiration rate. Thus, our findings may suggest antioxidant enrichment approaches for reducing the cardiovascular side effects of NRTI therapy.
Collapse
Affiliation(s)
- Yi-Fan Chen
- Comparative Biomedical Sciences, LSU School of Veterinary Medicine, Skip Bertman Drive, Baton Rouge, LA, 70808, USA
| | - Valeria Y Hebert
- Pharmacology, Toxicology and Neuroscience, LSU Health Sciences Center, 1501 Kings Highway, Shreveport, LA, 71103, USA
| | - Krisztian Stadler
- Oxidative Stress and Disease Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA
| | - Stephen Y Xue
- Pharmacology, Toxicology and Neuroscience, LSU Health Sciences Center, 1501 Kings Highway, Shreveport, LA, 71103, USA
| | - Kate Slaybaugh
- Pharmacology, Toxicology and Neuroscience, LSU Health Sciences Center, 1501 Kings Highway, Shreveport, LA, 71103, USA
| | - Elliot Luttrell-Williams
- Pharmacology, Toxicology and Neuroscience, LSU Health Sciences Center, 1501 Kings Highway, Shreveport, LA, 71103, USA
| | - Mitzi C Glover
- Clinical Laboratory Sciences, School of Allied Health Professions, LSU Health Sciences Center, 1900 Gravier Street, New Orleans, LA, 70112, USA
| | - David M Krzywanski
- Cellular Biology and Anatomy, LSU Health Sciences Center, 1501 Kings Highway, Shreveport, LA, 71103, USA
| | - Tammy R Dugas
- Comparative Biomedical Sciences, LSU School of Veterinary Medicine, Skip Bertman Drive, Baton Rouge, LA, 70808, USA.
| |
Collapse
|
44
|
Erlich JR, To EE, Liong S, Brooks R, Vlahos R, O'Leary JJ, Brooks DA, Selemidis S. Targeting Evolutionary Conserved Oxidative Stress and Immunometabolic Pathways for the Treatment of Respiratory Infectious Diseases. Antioxid Redox Signal 2020; 32:993-1013. [PMID: 32008371 PMCID: PMC7426980 DOI: 10.1089/ars.2020.8028] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Significance: Up until recently, metabolism has scarcely been referenced in terms of immunology. However, emerging evidence has shown that immune cells undergo an adaptation of metabolic processes, known as the metabolic switch. This switch is key to the activation, and sustained inflammatory phenotype in immune cells, which includes the production of cytokines and reactive oxygen species (ROS) that underpin infectious diseases, respiratory and cardiovascular disease, neurodegenerative disease, as well as cancer. Recent Advances: There is a burgeoning body of evidence that immunometabolism and redox biology drive infectious diseases. For example, influenza A virus (IAV) utilizes endogenous ROS production via NADPH oxidase (NOX)2-containing NOXs and mitochondria to circumvent antiviral responses. These evolutionary conserved processes are promoted by glycolysis, the pentose phosphate pathway, and the tricarboxylic acid (TCA) cycle that drive inflammation. Such metabolic products involve succinate, which stimulates inflammation through ROS-dependent stabilization of hypoxia-inducible factor-1α, promoting interleukin-1β production by the inflammasome. In addition, itaconate has recently gained significant attention for its role as an anti-inflammatory and antioxidant metabolite of the TCA cycle. Critical Issues: The molecular mechanisms by which immunometabolism and ROS promote viral and bacterial pathology are largely unknown. This review will provide an overview of the current paradigms with an emphasis on the roles of immunometabolism and ROS in the context of IAV infection and secondary complications due to bacterial infection such as Streptococcus pneumoniae. Future Directions: Molecular targets based on metabolic cell processes and ROS generation may provide novel and effective therapeutic strategies for IAV and associated bacterial superinfections.
Collapse
Affiliation(s)
- Jonathan R. Erlich
- Program in Chronic Infectious and Inflammatory Diseases, Oxidant and Inflammation Biology Group, School of Health and Biomedical Sciences, College of Science, Engineering & Health, RMIT University, Bundoora, Australia
| | - Eunice E. To
- Program in Chronic Infectious and Inflammatory Diseases, Oxidant and Inflammation Biology Group, School of Health and Biomedical Sciences, College of Science, Engineering & Health, RMIT University, Bundoora, Australia
| | - Stella Liong
- Program in Chronic Infectious and Inflammatory Diseases, Oxidant and Inflammation Biology Group, School of Health and Biomedical Sciences, College of Science, Engineering & Health, RMIT University, Bundoora, Australia
| | - Robert Brooks
- School of Pharmacy and Medical Sciences, University of South Australia Cancer Research Institute, University of South Australia, Adelaide, Australia
| | - Ross Vlahos
- Program in Chronic Infectious and Inflammatory Diseases, Oxidant and Inflammation Biology Group, School of Health and Biomedical Sciences, College of Science, Engineering & Health, RMIT University, Bundoora, Australia
| | - John J. O'Leary
- School of Pharmacy and Medical Sciences, University of South Australia Cancer Research Institute, University of South Australia, Adelaide, Australia
- Department of Histopathology, Trinity College Dublin, Dublin, Ireland
- Sir Patrick Dun's Laboratory, Central Pathology Laboratory, St James's Hospital, Dublin, Ireland
| | - Doug A. Brooks
- School of Pharmacy and Medical Sciences, University of South Australia Cancer Research Institute, University of South Australia, Adelaide, Australia
- Molecular Pathology Laboratory, Coombe Women and Infants' University Hospital, Dublin, Ireland
| | - Stavros Selemidis
- Program in Chronic Infectious and Inflammatory Diseases, Oxidant and Inflammation Biology Group, School of Health and Biomedical Sciences, College of Science, Engineering & Health, RMIT University, Bundoora, Australia
- Address correspondence to: Prof. Stavros Selemidis, Program in Chronic Infectious and Inflammatory Diseases, Oxidant and Inflammation Biology Group, School of Health and Biomedical Sciences, College of Science, Engineering & Health, RMIT University, Bundoora, VIC 3083, Australia
| |
Collapse
|
45
|
Travis OK, White D, Baik C, Giachelli C, Thompson W, Stubbs C, Greer M, Lemon JP, Williams JM, Cornelius DC. Interleukin-17 signaling mediates cytolytic natural killer cell activation in response to placental ischemia. Am J Physiol Regul Integr Comp Physiol 2020; 318:R1036-R1046. [PMID: 32320265 DOI: 10.1152/ajpregu.00285.2019] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
T-helper (TH)17s, IL-17, and cytolytic natural killer cells (cNKs) are increased in preeclampsia and contribute to the hypertension, inflammation, and fetal growth restriction that occurs in response to placental ischemia in the reduced uterine perfusion pressure (RUPP) rat model of preeclampsia. As IL-17 stimulates NK cytotoxicity in vitro, we tested the hypothesis that IL-17 inhibition in RUPP rats would decrease cNK activation as a mechanism to improve maternal and fetal outcomes. On gestation day (GD) 14, rats undergoing RUPP received a miniosmotic pump infusing IL-17RC (100 pg/day), a soluble IL-17 receptor (RUPP + IL-17RC). On GD19, mean arterial pressure (MAP) was measured in normal pregnant (NP), RUPP, and RUPP + IL-17RC rats (n = 10-12/group), animals were euthanized, and blood and tissues were collected for analysis. MAP was 30% higher in RUPP compared with NP (P < 0.0001) and was 12% lower in RUPP + IL-17RC (P = 0.0007 vs. RUPP). Placental cytolytic NK cells were 132% higher in RUPP than in NP (P = 0.04 vs. NP) and were normalized in RUPP + IL-17RC (P = 0.03 vs. RUPP). Placental levels of TNF-α, a cNK-secreted cytokine, and macrophage inflammatory protein-3α (MIP-3α), a cNK chemokine, were higher in RUPP vs. NP and lower after IL-17 blockade. Placental VEGF was lower in RUPP vs. NP and was normalized in RUPP + IL-17RC. In vitro cytolytic activity of RUPP placental NKs was higher compared with NP and was blunted in RUPP + IL-17RC NKs. Finally, both fetal weight and placental weight were lower in RUPP compared with NP, and were improved in RUPP + IL-17RC. These data identify IL-17 as a mediator of cNK activation in response to placental ischemia during pregnancy.
Collapse
Affiliation(s)
- Olivia K Travis
- Department of Pharmacology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Dakota White
- Department of Emergency Medicine, University of Mississippi Medical Center, Jackson, Mississippi
| | - Cedar Baik
- Department of Emergency Medicine, University of Mississippi Medical Center, Jackson, Mississippi
| | - Chelsea Giachelli
- Department of Emergency Medicine, University of Mississippi Medical Center, Jackson, Mississippi
| | - Willie Thompson
- Department of Pharmacology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Cassandra Stubbs
- Department of Pharmacology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Mallory Greer
- Department of Pharmacology, University of Mississippi Medical Center, Jackson, Mississippi
| | - James P Lemon
- Department of Pharmacology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Jan Michael Williams
- Department of Pharmacology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Denise C Cornelius
- Department of Pharmacology, University of Mississippi Medical Center, Jackson, Mississippi.,Department of Emergency Medicine, University of Mississippi Medical Center, Jackson, Mississippi
| |
Collapse
|
46
|
Mukosera GT, Clark TC, Ngo L, Liu T, Schroeder H, Power GG, Yellon SM, Parast MM, Blood AB. Nitric oxide metabolism in the human placenta during aberrant maternal inflammation. J Physiol 2020; 598:2223-2241. [PMID: 32118291 DOI: 10.1113/jp279057] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 02/25/2020] [Indexed: 12/27/2022] Open
Abstract
KEY POINTS Nitric oxide (NO) is a gasotransmitter with important physiological and pathophysiological roles in pregnancy. There is limited information available about the sources and metabolism of NO and its bioactive metabolites (NOx) in both normal and complicated pregnancies. The present study characterized and quantified endogenous NOx in human and mouse placenta following determination of the stability of exogenous NOx in placental homogenates. NOx have differential stability in placental homogenates. NO and iron nitrosyl species (FeNOs), are relatively unstable in placental homogenates from normal placentas. Exogenous NO, nitrite and nitrosothiols react with placental homogenates to form iron nitrosyl complexes. FeNOs were also detected endogenously in mouse and human placenta. NOx levels in placental villous tissue are increased in fetal growth restriction vs. placentas from women with normal pregnancies, particularly in fetal growth restriction associated with pre-eclampsia. Villitis was not associated, however, with an increase in NOx levels in either normotensive or pre-eclamptic placentas. The results call for further investigation of FeNOs in normal and complicated pregnancies. ABSTRACT Nitric oxide (NO) is a gasotransmitter with important roles in pregnancy under both physiological and pathophysiological conditions. Although products of NO metabolism (NOx) also have significant bioactivity, little is known about the role of NO and NOx under conditions of aberrant placental inflammation during pregnancy. An ozone-based chemiluminescence approach was used to investigate the stability and metabolic fate of NOx in human placental homogenates from uncomplicated pregnancies in healthy mothers compared to that in placental tissue from normotensive and pre-eclamptic pregnancies complicated with fetal growth restriction (FGR) with and without villitis of unknown aetiology. We hypothesized that placental NOx would be increased in FGR vs. normal tissue, and be further increased in villitis vs. non-villitis placentas. Findings indicate that nitrate, nitrite and nitrosothiols, but not NO or iron nitrosyl species (FeNOs), are relatively stable in placental homogenates from normal placentas, and that NO, nitrite and nitrosothiols react with placental homogenates to form iron nitrosyl complexes. Furthermore, NOx levels in placental villous tissue are increased in FGR vs. placentas from women with normal pregnancies, particularly in FGR associated with pre-eclampsia. However, in contrast to our hypothesis, villitis was not associated with an increase in NOx levels in either normotensive or pre-eclamptic placentas. Our results also strongly support the involvement of FeNOs in both mouse and human placenta, and call for their further study as a critical mechanistic link between pre-eclampsia and fetal growth restriction.
Collapse
Affiliation(s)
- George T Mukosera
- Lawrence D Longo Center for Perinatal Biology and Department of Pediatrics, Loma Linda University, 11175 Campus Street, Loma Linda, CA, 92354, USA
| | - Tatianna C Clark
- Lawrence D Longo Center for Perinatal Biology and Department of Pediatrics, Loma Linda University, 11175 Campus Street, Loma Linda, CA, 92354, USA
| | - Larry Ngo
- Lawrence D Longo Center for Perinatal Biology and Department of Pediatrics, Loma Linda University, 11175 Campus Street, Loma Linda, CA, 92354, USA
| | - Taiming Liu
- Lawrence D Longo Center for Perinatal Biology and Department of Pediatrics, Loma Linda University, 11175 Campus Street, Loma Linda, CA, 92354, USA
| | - Hobe Schroeder
- Lawrence D Longo Center for Perinatal Biology and Department of Pediatrics, Loma Linda University, 11175 Campus Street, Loma Linda, CA, 92354, USA
| | - Gordon G Power
- Lawrence D Longo Center for Perinatal Biology and Department of Pediatrics, Loma Linda University, 11175 Campus Street, Loma Linda, CA, 92354, USA
| | - Steven M Yellon
- Lawrence D Longo Center for Perinatal Biology and Department of Pediatrics, Loma Linda University, 11175 Campus Street, Loma Linda, CA, 92354, USA
| | - Mana M Parast
- Department of Pathology, University of California San Diego, 200 W Arbor Dr, San Diego, CA, 92103, USA
| | - Arlin B Blood
- Lawrence D Longo Center for Perinatal Biology and Department of Pediatrics, Loma Linda University, 11175 Campus Street, Loma Linda, CA, 92354, USA
| |
Collapse
|
47
|
Suvakov S, Richards C, Nikolic V, Simic T, McGrath K, Krasnodembskaya A, McClements L. Emerging Therapeutic Potential of Mesenchymal Stem/Stromal Cells in Preeclampsia. Curr Hypertens Rep 2020; 22:37. [PMID: 32291521 DOI: 10.1007/s11906-020-1034-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW Preeclampsia is a dangerous pregnancy condition affecting both the mother and offspring. It is a multifactorial disease with poorly understood pathogenesis, lacking effective treatments. Maternal immune response, inflammation and oxidative stress leading to endothelial dysfunction are the most prominent pathogenic processes implicated in preeclampsia development. Here, we give a detailed overview of the therapeutic applications and mechanisms of mesenchymal stem/stromal cells (MSCs) as a potential new treatment for preeclampsia. RECENT FINDINGS MSCs have gained growing attention due to low immunogenicity, easy cultivation and expansion in vitro. Accumulating evidence now suggests that MSCs act primarily through their secretomes facilitating paracrine signalling that leads to potent immunomodulatory, pro-angiogenic and regenerative therapeutic effects. MSCs have been studied in different animal models of preeclampsia demonstrating promising result, which support further investigations into the therapeutic effects and mechanisms of MSC-based therapies in preeclampsia, steering these therapies into clinical trials.
Collapse
Affiliation(s)
- S Suvakov
- Department of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA.,Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - C Richards
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW, Australia
| | - V Nikolic
- Department of Pharmacology and Toxicology, Medical Faculty, University of Nis, Nis, Serbia
| | - T Simic
- Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade, Belgrade, Serbia.,Serbian Academy of Sciences and Arts, Belgrade, Serbia
| | - K McGrath
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW, Australia
| | - A Krasnodembskaya
- The Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - L McClements
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW, Australia.
| |
Collapse
|
48
|
Morillon AC, Williamson RD, Baker PN, Kell DB, Kenny LC, English JA, McCarthy FP, McCarthy C. Effect of L-Ergothioneine on the metabolic plasma profile of the RUPP rat model of pre-eclampsia. PLoS One 2020; 15:e0230977. [PMID: 32231385 PMCID: PMC7108727 DOI: 10.1371/journal.pone.0230977] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 03/12/2020] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION Pre-eclampsia is a major cause of maternal and fetal mortality and morbidity worldwide. Its pathophysiology remains unclear, but mitochondrial dysfunction and oxidative stress have been implicated. L-Ergothioneine is a naturally occurring, water-soluble betaine, that has demonstrated antioxidant properties. Using the reduced uterine perfusion pressure (RUPP) rat model of pre-eclampsia, this study aimed to define the plasma metabolic profile following treatment with L-Ergothioneine. METHODS The effect of L-Ergothioneine (ET) treatment was explored using in vivo treatment in rats: Sham control (SC, n = 5), RUPP control (RC, n = 5), Sham +ET (ST, n = 5), RUPP +ET (RT, n = 5). Differential expression of plasma metabolites were obtained using untargeted liquid chromatography coupled to mass spectrometry. Statistical analysis was performed on normalised data comparing RC to SC, RT to RC, and RT to ST. Metabolites significantly altered (FDR < 0.05) were identified through database search. RESULTS We report significantly lower levels of L-palmitoylcarnitine in RC compared to SC, a fatty acyl substrate involved in beta-oxidation in the mitochondria. We report that a metabolite that has been associated with oxidative stress (Glutamylcysteine) was detected at significantly higher levels in RT vs RC and RT vs ST. Five metabolites associated with inflammation were significantly lower in RT vs RC and three metabolites in RT vs ST, demonstrating the anti-inflammatory effects of ET in the RUPP rat model of pre-eclampsia. CONCLUSIONS L-Ergothioneine may help preserve mitochondrial function by increasing antioxidant levels, and reducing inflammatory responses associated with pre-eclampsia. This study shows the potential of L-Ergothioneine as a treatment for pre-eclampsia.
Collapse
Affiliation(s)
- Aude-Claire Morillon
- INFANT Research Centre, Cork, Ireland
- Department of Obstetrics and Gynecology, University College Cork, Cork, Ireland
| | | | - Philip N. Baker
- College of Life Sciences, University of Leicester, Leicester, United Kingdom
| | - Douglas B. Kell
- Dept of Biochemistry, Institute of Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Crown St, Liverpool, United Kingdom
- Novo Nordisk Foundation Centre for Biosustainability, Technical University of Denmark, Lyngby, Denmark
| | - Louise C. Kenny
- Department of Women’s and Children’s Health, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Jane A. English
- INFANT Research Centre, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Fergus P. McCarthy
- INFANT Research Centre, Cork, Ireland
- Department of Obstetrics and Gynecology, University College Cork, Cork, Ireland
| | - Cathal McCarthy
- Department of Pharmacology and Therapeutics, University College Cork, Cork, Ireland
| |
Collapse
|
49
|
McElwain CJ, Tuboly E, McCarthy FP, McCarthy CM. Mechanisms of Endothelial Dysfunction in Pre-eclampsia and Gestational Diabetes Mellitus: Windows Into Future Cardiometabolic Health? Front Endocrinol (Lausanne) 2020; 11:655. [PMID: 33042016 PMCID: PMC7516342 DOI: 10.3389/fendo.2020.00655] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 08/11/2020] [Indexed: 12/12/2022] Open
Abstract
Placental insufficiency and adipose tissue dysregulation are postulated to play key roles in the pathophysiology of both pre-eclampsia (PE) and gestational diabetes mellitus (GDM). A dysfunctional release of deleterious signaling motifs can offset an increase in circulating oxidative stressors, pro-inflammatory factors and various cytokines. It has been previously postulated that endothelial dysfunction, instigated by signaling from endocrine organs such as the placenta and adipose tissue, may be a key mediator of the vasculopathy that is evident in both adverse obstetric complications. These signaling pathways also have significant effects on long term maternal cardiometabolic health outcomes, specifically cardiovascular disease, hypertension, and type II diabetes. Recent studies have noted that both PE and GDM are strongly associated with lower maternal flow-mediated dilation, however the exact pathways which link endothelial dysfunction to clinical outcomes in these complications remains in question. The current diagnostic regimen for both PE and GDM lacks specificity and consistency in relation to clinical guidelines. Furthermore, current therapeutic options rely largely on clinical symptom control such as antihypertensives and insulin therapy, rather than that of early intervention or prophylaxis. A better understanding of the pathogenic origin of these obstetric complications will allow for more targeted therapeutic interventions. In this review we will explore the complex signaling relationship between the placenta and adipose tissue in PE and GDM and investigate how these intricate pathways affect maternal endothelial function and, hence, play a role in acute pathophysiology and the development of future chronic maternal health outcomes.
Collapse
Affiliation(s)
- Colm J. McElwain
- Department of Pharmacology and Therapeutics, Western Gateway Building, University College Cork, Cork, Ireland
- *Correspondence: Colm J. McElwain
| | - Eszter Tuboly
- Department of Pharmacology and Therapeutics, Western Gateway Building, University College Cork, Cork, Ireland
| | - Fergus P. McCarthy
- Department of Obstetrics and Gynaecology, Cork University Maternity Hospital, Cork, Ireland
| | - Cathal M. McCarthy
- Department of Pharmacology and Therapeutics, Western Gateway Building, University College Cork, Cork, Ireland
| |
Collapse
|
50
|
Williamson RD, McCarthy FP, Manna S, Groarke E, Kell DB, Kenny LC, McCarthy CM. L-(+)-Ergothioneine Significantly Improves the Clinical Characteristics of Preeclampsia in the Reduced Uterine Perfusion Pressure Rat Model. Hypertension 2019; 75:561-568. [PMID: 31865793 DOI: 10.1161/hypertensionaha.119.13929] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Preeclampsia is a multifactorial hypertensive disorder of pregnancy founded on abnormal placentation, and the resultant placental ischemic microenvironment is thought to play a crucial role in its pathophysiology. Placental ischemia because of fluctuations in the delivery of oxygen results in oxidative stress, and recent evidence suggests that mitochondrial dysfunction may be a prime mediator. However, large clinical trials of therapeutic antioxidants such as vitamins C and E for the treatment of preeclampsia have been disappointing. L-(+)-ergothioneine (ERG)-an unusual amino acid betaine derived from histidine-has important cytoprotective and antioxidant properties under conditions of high oxidative stress. In this study, we investigated the potential therapeutic effects of administration of ERG in the reduced uterine perfusion pressure (RUPP) rat model of preeclampsia. ERG (25 mg/kg per day) was administered to rats on gestational day 11. On gestational day 14, RUPP surgery was performed, and on gestational day 19, blood pressure (mean arterial pressure) and fetal growth were measured. Production of mitochondria-specific H2O2 was analyzed in vivo in kidney samples. ERG ameliorated the hypertension (129±3 versus 115±4 mm Hg; P=0.01; n=8) and significantly increased pup weight in RUPP rats. ERG also significantly decreased circulating levels of antiangiogenic sFlt-1 (soluble fms-like tyrosine kinase-1) in RUPP rats (1367±245 pg/mL; P=0.04). Mitochondria-specific H2O2 (0.022±0.003 versus 0.029±0.001; MitoP/B ratio, n=3; P=0.05) was also significantly decreased in kidney tissue in RUPP rats treated with ERG. These data support the potential use of ERG for the treatment of preeclampsia.
Collapse
Affiliation(s)
- Rachel D Williamson
- From the Irish Centre for Fetal and Neonatal Translational Research, Cork University Maternity Hospital, Ireland (R.D.W., F.P.M., S.M., C.M.)
| | - Fergus P McCarthy
- From the Irish Centre for Fetal and Neonatal Translational Research, Cork University Maternity Hospital, Ireland (R.D.W., F.P.M., S.M., C.M.)
| | - Samprikta Manna
- From the Irish Centre for Fetal and Neonatal Translational Research, Cork University Maternity Hospital, Ireland (R.D.W., F.P.M., S.M., C.M.)
| | - Emer Groarke
- Clinical Biochemistry, Cork University Hospital, Ireland (E.G.)
| | - Douglas B Kell
- Department of Biochemistry, Faculty of Health and Life Sciences, Institute of Integrative Biology, University of Liverpool, United Kingdom (D.B.K.).,Novo Nordisk Foundation Centre for Biosustainability, Technical University of Denmark, Lyngby (D.B.K.)
| | - Louise C Kenny
- Department of Women's and Children's Health, Institute of Translational Medicine, University of Liverpool, United Kingdom (L.C.K.)
| | - Cathal M McCarthy
- From the Irish Centre for Fetal and Neonatal Translational Research, Cork University Maternity Hospital, Ireland (R.D.W., F.P.M., S.M., C.M.).,Department of Pharmacology and Therapeutics, Western Gateway Building, University College Cork, Ireland (C.M.)
| |
Collapse
|