1
|
Liu Y, Yu W, Tang J, Zhang Y, Yang J, Wu Q, Li H, Zhu Z, Li A, Han J, Zhang X, Luo Y, Zhang Y, Liu J. Evolutionary dynamics of PEG10 and its interacting proteins during early and late-stage placental development in ruminants. Int J Biol Macromol 2025; 309:142761. [PMID: 40180104 DOI: 10.1016/j.ijbiomac.2025.142761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 03/02/2025] [Accepted: 03/31/2025] [Indexed: 04/05/2025]
Abstract
Ruminants play a crucial role in dairy farming, pharmaceuticals, and embryonic stem cell research; thus, it is vital to prevent pregnancy loss and improve reproductive outcomes through a better understanding of placental development. Paternally-expressed gene 10 (PEG10) is a conserved gene essential for placental development in mammals, but its function in ruminants is not well understood. To develop insights into its role in placental development, this study investigated the gene structure and expression of PEG10 in cattle and goats. We found that PEG10's structure was conserved across species, and in the placenta, it retained the ability to bind to its own mRNA. Transcript analysis revealed differential expression patterns of PEG10 at early and late stages of placental development. We identified 70 proteins potentially interacting with PEG10 that were involved in biological processes like metabolism, signal transduction, cell proliferation, and immune responses. These proteins were grouped into seven clusters, associated with pathways such as amino acid degradation, the TCA cycle, longevity regulation, cardiomyopathy, proteasome function, and biosynthesis. Our findings suggest that PEG10 regulates placental development in ruminants by interacting with key proteins like CAST, ITGA6, and FTL, which are responsible for critical cellular processes in placental function.
Collapse
Affiliation(s)
- Yayi Liu
- Hainan Institute of Northwest A&F University, Sanya 572000, China; College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China; Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Yangling 712100, Shaanxi, China
| | - Wei Yu
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China; Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Yangling 712100, Shaanxi, China
| | - Jiaomei Tang
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China; Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Yangling 712100, Shaanxi, China
| | - Yuanyuan Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China; Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Yangling 712100, Shaanxi, China
| | - Jing Yang
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China; Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Yangling 712100, Shaanxi, China
| | - Qingyan Wu
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China; Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Yangling 712100, Shaanxi, China
| | - Huijia Li
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China; Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Yangling 712100, Shaanxi, China
| | - Zhenliang Zhu
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China; Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Yangling 712100, Shaanxi, China
| | - Aicong Li
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China; Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Yangling 712100, Shaanxi, China
| | - Jing Han
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China; Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Yangling 712100, Shaanxi, China
| | - Xinyan Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China; Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Yangling 712100, Shaanxi, China
| | - Yan Luo
- College of Animal Engineering, Yangling Vocational and Technical College, Key Laboratory for Efficient Ruminant Breeding Technology of Higher Education Institutions in Shaanxi Province, Yangling 712100, China.
| | - Yong Zhang
- Hainan Institute of Northwest A&F University, Sanya 572000, China; College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China; Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Yangling 712100, Shaanxi, China.
| | - Jun Liu
- Hainan Institute of Northwest A&F University, Sanya 572000, China; College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China; Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Yangling 712100, Shaanxi, China.
| |
Collapse
|
2
|
Pei Z, Tang H, Wu J, Wang J, Liu D, Cao C, Pan W, Li T, Duan H, Wang Z, Zheng M, Hu Y, Zhao G. Identification of syncytiotrophoblast-derived cf-RNA OPA1 to predict the occurrence of preeclampsia. Placenta 2025; 160:1-10. [PMID: 39742547 DOI: 10.1016/j.placenta.2024.12.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 12/19/2024] [Accepted: 12/28/2024] [Indexed: 01/03/2025]
Abstract
BACKGROUND Pre-eclampsia (PE) poses a significant threat to mothers and infants worldwide. Studies indicate that taking low-dose aspirin before the 16th week of pregnancy may prevent approximately 70 % of PE cases, highlighting the importance of predicting PE. Cell-free RNA (cf-RNA) exhibits significant changes in the maternal peripheral blood during early pregnancy, making cf-RNA analysis a promising and less invasive method for predicting PE. METHODS The two datasets, GSE192902 and GSE149440, were analyzed to identify differentially expressed cf-RNAs, followed by the calculation of their AUC values Subsequently, these cf-RNAs were validated using placental tissues, as well as late- and early-stage plasma samples collected from both healthy individuals and patients with PE. Furthermore, we performed tissue localization and functional analyses on the ultimate candidate gene. RESULTS Mitochondrial Dynamin-Like GTPase (OPA1) emerged as the molecule with the most consistent and statistically significant alterations in placental tissues and serum samples from patients with PE across various gestational weeks. Notably, the combination of OPA1 levels and mean arterial pressure (MAP) yielded an AUC of 0.825 (95 % CI: 0.759-0.879) for predicting PE. Additionally, we verified that OPA1 is predominantly expressed in placental syncytiotrophoblast (STB) cells, and its downregulation negatively impacts STB mitochondrial function, angiogenic potential, and cell proliferation. CONCLUSIONS OPA1 holds the potential to emerge as a novel cf-RNA for predicting PE.
Collapse
Affiliation(s)
- Zhongrui Pei
- Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Huirong Tang
- Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Jing Wu
- Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jie Wang
- Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Dan Liu
- Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Chenrui Cao
- Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Weichen Pan
- Department of Obstetrics and Gynecology, Drum Tower Clinical Medical College, Nanjing Medical University, Nanjing, China
| | - Taishun Li
- Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Honglei Duan
- Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Zhiyin Wang
- Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Mingming Zheng
- Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| | - Yali Hu
- Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| | - Guangfeng Zhao
- Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China; Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| |
Collapse
|
3
|
Merech F, Lara B, Rios D, Paparini D, Ramhorst R, Hauk V, Pérez Leirós C, Vota D. Vasoactive intestinal peptide induces metabolic rewiring of human-derived cytotrophoblast cells to promote cell migration. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119886. [PMID: 39653085 DOI: 10.1016/j.bbamcr.2024.119886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 11/28/2024] [Accepted: 12/02/2024] [Indexed: 12/16/2024]
Abstract
The placenta has an extraordinary metabolic rate with high oxygen consumption. Extravillous cytotrophoblast cells (EVT) metabolism and function are critical to sustain their invasive phenotype supporting fetal development. Deficient EVT function underlies pregnancy complications as preeclampsia (PE) and fetal growth restriction (FGR). The vasoactive intestinal peptide (VIP) promotes human cytotrophoblast cell migration and invasion through mTOR signaling pathways suggesting its crucial role during placentation. Here we explored fatty acid uptake as well as lipid and glucose metabolism in human-derived cytotrophoblast cell function upon VIP stimulation. We found that VIP induced long chain fatty acid (LCFAs) uptake along with the expression of FATP2 transporter, CPT1 fatty acid oxidation (FAO)-rate limiting step importer, and lipid droplet accumulation. VIP induced the expression of glucose 6-P-dehydrogenase, a rate-limiting enzyme of the pentose phosphate pathway (PPP) and pyruvate dehydrogenase complex enzyme DLAT E2, without altering lactate secretion. This metabolic rewiring of trophoblast cells induced by VIP takes place without compromising mitochondrial function or reactive oxygen species (ROS) production. Moreover, cytotrophoblast cell migration induced by VIP required the three glycolysis, oxidative phosphorylation (OXPHOS) and FAO pathways. Our results provide evidence supporting VIP as a metabolic regulatory peptide in cytotrophoblast cells sustaining proper placentation and fetal growth.
Collapse
Affiliation(s)
- Fátima Merech
- Universidad de Buenos Aires (UBA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN-CONICET), Buenos Aires, Argentina
| | - Brenda Lara
- Universidad de Buenos Aires (UBA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN-CONICET), Buenos Aires, Argentina
| | - Daiana Rios
- Universidad de Buenos Aires (UBA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN-CONICET), Buenos Aires, Argentina
| | - Daniel Paparini
- Universidad de Buenos Aires (UBA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN-CONICET), Buenos Aires, Argentina
| | - Rosanna Ramhorst
- Universidad de Buenos Aires (UBA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN-CONICET), Buenos Aires, Argentina
| | - Vanesa Hauk
- Universidad de Buenos Aires (UBA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN-CONICET), Buenos Aires, Argentina
| | - Claudia Pérez Leirós
- Universidad de Buenos Aires (UBA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN-CONICET), Buenos Aires, Argentina.
| | - Daiana Vota
- Universidad de Buenos Aires (UBA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN-CONICET), Buenos Aires, Argentina.
| |
Collapse
|
4
|
Nicheperovich A, Schuster-Böckler B, Ní Leathlobhair M. Gestational trophoblastic disease: understanding the molecular mechanisms of placental tumours. Dis Model Mech 2025; 18:DMM052010. [PMID: 39873178 PMCID: PMC11810044 DOI: 10.1242/dmm.052010] [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] [Indexed: 01/30/2025] Open
Abstract
Gestational trophoblastic disease (GTD) describes a group of rare benign and cancerous lesions originating from the trophoblast cells of the placenta. These neoplasms are unconventional entities, being one of the few instances in which cancer develops from the cells of another organism, the foetus. Although this condition was first described over 100 years ago, the specific genetic and non-genetic drivers of this disease remain unknown to this day. However, recent findings have provided valuable insights into the potential mechanisms underlying this rare condition. Unlike previous reviews focused primarily on the clinical and diagnostic aspects of disease development, this Review consolidates the latest research concerning the role of genetics, epigenetics and microRNAs in the initiation and progression of GTD. By examining GTD from a molecular perspective, this Review provides a unique framework for understanding the pathogenesis and progression of this rare disease.
Collapse
Affiliation(s)
- Alina Nicheperovich
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7DQ, UK
| | - Benjamin Schuster-Böckler
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7DQ, UK
| | | |
Collapse
|
5
|
Carrasco-Wong I, Sanchez JM, Gutierrez JA, Chiarello DI. Trained innate immunity as a potential link between preeclampsia and future cardiovascular disease. Front Endocrinol (Lausanne) 2024; 15:1500772. [PMID: 39741876 PMCID: PMC11685753 DOI: 10.3389/fendo.2024.1500772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 11/27/2024] [Indexed: 01/03/2025] Open
Abstract
Preeclampsia (PE) is a complex pregnancy syndrome characterized by hypertension with or without proteinuria, affecting 2-6% of pregnancies globally. PE is characterized by excessive release of damage-associated molecular patterns (DAMPs) into the maternal circulation. This DAMP-rich milieu acts on innate immune cells, inducing a proinflammatory state characterized by elevated cytokines such as IL-1β and IL-18. This proinflammatory state in the mother and placenta results in the endothelial dysfunction strongly associated with cardiovascular disorders. While the immediate maternal and fetal risks of PE are well-documented, accumulating evidence indicates that PE also confers long-term cardiovascular risks to the mother, including hypertension, coronary heart disease, stroke, and heart failure. The underlying mechanisms connecting PE to these chronic cardiovascular conditions remain unclear. This article explores the potential role of trained innate immunity (TRIM) as a mechanistic link between PE and increased long-term cardiovascular risk. We propose that the persistent exposure to DAMPs during PE may epigenetically reprogram maternal innate immune cells and their progenitors, leading to TRIM. This reprogramming enhances the inflammatory response to subsequent stimuli, potentially contributing to endothelial dysfunction and chronic inflammation that predispose women to cardiovascular diseases later in life. Understanding the role of TRIM in PE could provide novel insights into the pathophysiology of PE-related cardiovascular complications and identify potential targets for therapeutic intervention. Further research is warranted to investigate the epigenetic and metabolic alterations in innate immune cells induced by PE and to determine how these changes may influence long-term maternal cardiovascular health.
Collapse
Affiliation(s)
| | | | - Jaime A. Gutierrez
- Escuela de Tecnología Médica, Facultad de Medicina y Ciencia,
Universidad San Sebastián, Santiago, Chile
| | - Delia I. Chiarello
- Escuela de Tecnología Médica, Facultad de Medicina y Ciencia,
Universidad San Sebastián, Santiago, Chile
| |
Collapse
|
6
|
Parameshwar PK, Vaillancourt C, Moraes C. Engineering placental trophoblast fusion: A potential role for biomechanics in syncytialization. Placenta 2024; 157:50-54. [PMID: 38448351 DOI: 10.1016/j.placenta.2024.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 02/07/2024] [Accepted: 02/21/2024] [Indexed: 03/08/2024]
Abstract
The process by which placental trophoblasts fuse to form the syncytiotrophoblast around the chorionic villi is not fully understood. Mechanical features of the in vivo and in vitro culture environments have recently emerged as having the potential to influence fusion efficiency, and considering these mechanical cues may ultimately allow predictive control of trophoblast syncytialization. Here, we review recent studies that suggest that biomechanical factors such as shear stress, tissue stiffness, and dimensionally-related stresses affect villous trophoblast fusion efficiency. We then discuss how these stimuli might arise in vivo and how they can be incorporated in cultures to study and enhance villous trophoblast fusion. We believe that this mechanical paradigm will provide novel insight into manipulating the syncytialization process to better engineer improved models, understand disease progression, and ultimately develop novel therapeutic strategies.
Collapse
Affiliation(s)
| | - Cathy Vaillancourt
- Institut National de la Recherche Scientifique (INRS)-Centre Armand-Frappier Santé Biotechnologie, Laval, QC, H7B 1B7, Canada; Department of Obstetrics and Gynecology, Université de Montréal, and Research Center Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) du Nord-de-l'Île-de-Montréal, Montréal, QC, H3L 1K5, Canada
| | - Christopher Moraes
- Department of Biological and Biomedical Engineering, McGill University, Montréal, QC, H3A 2B4, Canada; Department of Chemical Engineering, McGill University, Montréal, QC, H3A 0C5, Canada; Goodman Cancer Research Centre, McGill University, Montréal, QC, H3A 1A3, Canada; Division of Experimental Medicine, McGill University, Montréal, QC, H4A 3J1, Canada.
| |
Collapse
|
7
|
Chen A, Tian M, Luo Z, Cao X, Gu Y. Analysis of the evolution of placental oxidative stress research from a bibliometric perspective. Front Pharmacol 2024; 15:1475244. [PMID: 39484166 PMCID: PMC11524950 DOI: 10.3389/fphar.2024.1475244] [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: 08/03/2024] [Accepted: 10/07/2024] [Indexed: 11/03/2024] Open
Abstract
Background Research on placental oxidative stress is pivotal for comprehending pregnancy-related physiological changes and disease mechanisms. Despite recent advancements, a comprehensive review of current status, hotspots, and trends remains challenging. This bibliometric study systematically analyzes the evolution of placental oxidative stress research, offering a reference for future studies. Objective To conduct a comprehensive bibliometric analysis of the literature on placental oxidative stress to identify research hotspots, trends, and key contributors, thereby providing guidance for future research. Methods Relevant data were retrieved from the Web of Science Core Collection database and analyzed using VOSviewer, CiteSpace, and the bibliometrix package. An in-depth analysis of 4,796 publications was conducted, focusing on publication year, country/region, institution, author, journal, references, and keywords. Data collection concluded on 29 April 2024. Results A total of 4,796 papers were retrieved from 1,173 journals, authored by 18,835 researchers from 4,257 institutions across 103 countries/regions. From 1991 to 2023, annual publications on placental oxidative stress increased from 7 to 359. The United States (1,222 publications, 64,158 citations), the University of Cambridge (125 publications, 13,562 citations), and Graham J. Burton (73 publications, 11,182 citations) were the most productive country, institution, and author, respectively. The journal Placenta had the highest number of publications (329) and citations (17,152), followed by the International Journal of Molecular Sciences (122 publications). The most frequent keywords were "oxidative stress," "expression," "pregnancy," "preeclampsia," and "lipid peroxidation." Emerging high-frequency keywords included "gestational diabetes mellitus," "health," "autophagy," "pathophysiology," "infection," "preterm birth," "stem cell," and "inflammation." Conclusion Over the past 3 decades, research has concentrated on oxidative stress processes, antioxidant mechanisms, pregnancy-related diseases, and gene expression regulation. Current research frontiers involve exploring pathophysiology and mechanisms, assessing emerging risk factors and environmental impacts, advancing cell biology and stem cell research, and understanding the complex interactions of inflammation and immune regulation. These studies elucidate the mechanisms of placental oxidative stress, offering essential scientific evidence for future intervention strategies, therapeutic approaches, and public health policies.
Collapse
Affiliation(s)
| | | | | | - Xiaohui Cao
- Department of Obstetrics and Gynecology, Women’s Hospital of Jiangnan University, Wuxi Maternity and Child Health Care Hospital, Wuxi, China
| | - Yanfang Gu
- Department of Obstetrics and Gynecology, Women’s Hospital of Jiangnan University, Wuxi Maternity and Child Health Care Hospital, Wuxi, China
| |
Collapse
|
8
|
Júnior JPDL, Teixeira SC, de Souza G, Faria GV, Almeida MPO, Franco PS, Luz LC, Paschoalino M, Dos Santos NCL, de Oliveira RM, Martínez AFF, Rosini AM, Ambrosio MALV, Veneziani RCS, Bastos JK, Gomes AO, Alves RN, da Silva CV, Martins CHG, Ferro EAV, Barbosa BF. Copaifera spp. oleoresins control Trypanosoma cruzi infection in human trophoblast cells (BeWo) and placental explants. Biomed Pharmacother 2024; 179:117425. [PMID: 39265235 DOI: 10.1016/j.biopha.2024.117425] [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: 06/11/2024] [Revised: 08/25/2024] [Accepted: 09/04/2024] [Indexed: 09/14/2024] Open
Abstract
Congenital Chagas disease (CCD) is a worldwide neglected problem with significant treatment limitations. This study aimed to evaluate the potential of Copaifera spp. oleoresins (ORs) against Trypanosoma cruzi infection in trophoblast cells (BeWo lineage) and human chorionic villous explants (HCVE). The cytotoxicity of ORs was investigated using LDH and MTT assays. T. cruzi (Y strain) proliferation, invasion and reversibility were assessed in OR-treated BeWo cells, and proliferation was evaluated in OR-treated HCVE. The ultrastructure of T. cruzi trypomastigotes and amastigotes treated with ORs were analyzed by scanning and transmission electronic microscopy. ROS production in infected and treated BeWo cells and cytokines in BeWo and HCVE were measured. The ORs irreversibly decreased T. cruzi invasion, proliferation and release in BeWo cells by up to 70 %, 82 % and 80 %, respectively, and reduced parasite load in HCVE by up to 80 %. Significant structural changes in treated parasites were observed. ORs showed antioxidant capacity in BeWo cells, reducing ROS production induced by T. cruzi infection. Also, T. cruzi infection modulated the cytokine profile in both BeWo cells and HCVE; however, treatment with ORs upregulated cytokines decreased by T. cruzi infection in BeWo cells, while downregulated cytokines increased by the T. cruzi infection in HCVE. In conclusion, non-cytotoxic concentrations of Copaifera ORs demonstrated promising potential for controlling T. cruzi infection in models of the human maternal-fetal interface.
Collapse
Affiliation(s)
- Joed Pires de Lima Júnior
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil.
| | - Samuel Cota Teixeira
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil.
| | - Guilherme de Souza
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil.
| | - Guilherme Vieira Faria
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil.
| | - Marcos Paulo Oliveira Almeida
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil.
| | - Priscila Silva Franco
- Department of Parasitology, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil.
| | - Luana Carvalho Luz
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Marina Paschoalino
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Natália Carine Lima Dos Santos
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil.
| | - Rafael Martins de Oliveira
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Aryani Felixa Fajardo Martínez
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Alessandra Monteiro Rosini
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | | | | | - Jairo Kenupp Bastos
- School of Pharmaceutical Sciences of Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brazil.
| | - Angelica Oliveira Gomes
- Institute of Natural and Biological Sciences, Universidade Federal do Triângulo Mineiro, Uberaba, MG, Brazil.
| | - Rosiane Nascimento Alves
- Department of Agricultural and Natural Science, Universidade do Estado de Minas Gerais, Ituiutaba, MG, Brazil.
| | - Claudio Vieira da Silva
- Laboratory of Trypanosomatids, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil.
| | - Carlos Henrique Gomes Martins
- Laboratory of Antimicrobial Testing, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil.
| | - Eloisa Amália Vieira Ferro
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil.
| | - Bellisa Freitas Barbosa
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil.
| |
Collapse
|
9
|
Xie W, Koppula S, Kale MB, Ali LS, Wankhede NL, Umare MD, Upaganlawar AB, Abdeen A, Ebrahim EE, El-Sherbiny M, Behl T, Shen B, Singla RK. Unraveling the nexus of age, epilepsy, and mitochondria: exploring the dynamics of cellular energy and excitability. Front Pharmacol 2024; 15:1469053. [PMID: 39309002 PMCID: PMC11413492 DOI: 10.3389/fphar.2024.1469053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 08/20/2024] [Indexed: 09/25/2024] Open
Abstract
Epilepsy, a complex neurological condition marked by recurring seizures, is increasingly recognized for its intricate relationship with mitochondria, the cellular powerhouses responsible for energy production and calcium regulation. This review offers an in-depth examination of the interplay between epilepsy, mitochondrial function, and aging. Many factors might account for the correlation between epilepsy and aging. Mitochondria, integral to cellular energy dynamics and neuronal excitability, perform a critical role in the pathophysiology of epilepsy. The mechanisms linking epilepsy and mitochondria are multifaceted, involving mitochondrial dysfunction, reactive oxygen species (ROS), and mitochondrial dynamics. Mitochondrial dysfunction can trigger seizures by compromising ATP production, increasing glutamate release, and altering ion channel function. ROS, natural byproducts of mitochondrial respiration, contribute to oxidative stress and neuroinflammation, critical factors in epileptogenesis. Mitochondrial dynamics govern fusion and fission processes, influence seizure threshold and calcium buffering, and impact seizure propagation. Energy demands during seizures highlight the critical role of mitochondrial ATP generation in maintaining neuronal membrane potential. Mitochondrial calcium handling dynamically modulates neuronal excitability, affecting synaptic transmission and action potential generation. Dysregulated mitochondrial calcium handling is a hallmark of epilepsy, contributing to excitotoxicity. Epigenetic modifications in epilepsy influence mitochondrial function through histone modifications, DNA methylation, and non-coding RNA expression. Potential therapeutic avenues targeting mitochondria in epilepsy include mitochondria-targeted antioxidants, ketogenic diets, and metabolic therapies. The review concludes by outlining future directions in epilepsy research, emphasizing integrative approaches, advancements in mitochondrial research, and ethical considerations. Mitochondria emerge as central players in the complex narrative of epilepsy, offering profound insights and therapeutic potential for this challenging neurological disorder.
Collapse
Affiliation(s)
- Wen Xie
- Department of Pharmacy and Institutes for Systems Genetics, Center for High Altitude Medicine, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Sushruta Koppula
- College of Biomedical and Health Sciences, Konkuk University, Chungju-Si, Republic of Korea
| | - Mayur B. Kale
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, India
| | - Lashin S. Ali
- Department of Basic Medical Sciences, Faculty of Dentistry, Al-Ahliyya Amman University, Amman, Jordan
| | | | - Mohit D. Umare
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, India
| | | | - Ahmed Abdeen
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Benha University, Toukh, Egypt
| | - Elturabi E. Ebrahim
- Medical-Surgical Nursing Department, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Mohamed El-Sherbiny
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, Riyadh, Saudi Arabia
- Department of Anatomy, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Tapan Behl
- Amity School of Pharmaceutical Sciences, Amity University, Mohali, India
| | - Bairong Shen
- Institutes for Systems Genetics, West China Tianfu Hospital, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Rajeev K. Singla
- Department of Pharmacy and Institutes for Systems Genetics, Center for High Altitude Medicine, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, India
| |
Collapse
|
10
|
Hijam AC, Tongbram YC, Nongthombam PD, Meitei HN, Koijam AS, Rajashekar Y, Haobam R. Neuroprotective potential of traditionally used medicinal plants of Manipur against rotenone-induced neurotoxicity in SH-SY5Y neuroblastoma cells. JOURNAL OF ETHNOPHARMACOLOGY 2024; 330:118197. [PMID: 38636579 DOI: 10.1016/j.jep.2024.118197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 04/08/2024] [Accepted: 04/12/2024] [Indexed: 04/20/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Alternanthera sessilis (L.) R. Br. ex DC., Eryngium foetidum L., and Stephania japonica (Thunb.) Miers plants are traditionally used to treat various central nervous system disorders like paralysis, epilepsy, seizure, convulsion, chronic pain, headache, sleep disturbances, sprain, and mental disorders. However, their possible neuroprotective effects have not been evaluated experimentally so far. AIM OF THE STUDY The study aims to examine the neuroprotective potential of the three plants against cytotoxicity induced by rotenone in SH-SY5Y neuroblastoma cells and assess its plausible mechanisms of neuroprotection. MATERIALS AND METHODS The antioxidant properties of the plant extracts were determined chemically by DPPH and ABTS assay methods. The cytotoxicity of rotenone and the cytoprotective activities of the extracts were evaluated using MTT assays. Microtubule-associated protein 2 (MAP2) expression studies in cells were performed to assess neuronal survival after rotenone and extract treatments. Mitochondrial membrane potential and intracellular levels of reactive oxygen species were evaluated using Rhodamine 123 and DCF-DA dye, respectively. Catalase, glutathione peroxidase, and superoxide dismutase activities were also measured. Apoptotic nuclei were examined using DAPI staining. Liquid chromatography-quadrupole time-of-flight tandem mass spectrometry (LC-QTOF-MS) analysis of the plant extracts was also performed. RESULTS The methanol extracts of A. sessilis, S. japonica, and E. foetidum showed excellent free radical scavenging activities. MAP2 expression studies show that A. sessilis and S. japonica have higher neuroprotective effects against rotenone-induced neurotoxicity in SH-SY5Y cells than E. foetidum. Pre-treating cells with the plant extracts reverses the rotenone-induced increase in intracellular ROS. The plant extracts could also restore the reduced mitochondrial membrane potential induced by rotenone treatment and reinstate rotenone-induced increases in catalase, glutathione peroxidase, and superoxide dismutase activities. All the extracts inhibited rotenone-induced changes in nuclear morphology and DNA condensation, an early event of cellular apoptosis. LC-QTOF-MS analysis of the plant extracts shows the presence of neuroprotective compounds. CONCLUSIONS The plant extracts showed neuroprotective activities against rotenone-treated SH-SY5Y cells through antioxidant and anti-apoptotic mechanisms. These findings support the ethnopharmacological uses of these plants in treating neurological disorders. They probably are a good source of neuroprotective compounds that could be further explored to develop treatment strategies for neurodegenerative diseases like Parkinson's disease.
Collapse
Affiliation(s)
- Aruna Chanu Hijam
- Department of Biotechnology, Manipur University, Canchipur, Imphal, 795003, Manipur, India
| | | | - Pooja Devi Nongthombam
- Department of Biotechnology, Manipur University, Canchipur, Imphal, 795003, Manipur, India
| | | | - Arunkumar Singh Koijam
- Insect Bioresources Laboratory, Animal Bioresources Programme, Institute of Bioresources & Sustainable Development, Department of Biotechnology, Govt. of India, Takyelpat, Imphal, 795001, Manipur, India
| | - Yallapa Rajashekar
- Insect Bioresources Laboratory, Animal Bioresources Programme, Institute of Bioresources & Sustainable Development, Department of Biotechnology, Govt. of India, Takyelpat, Imphal, 795001, Manipur, India
| | - Reena Haobam
- Department of Biotechnology, Manipur University, Canchipur, Imphal, 795003, Manipur, India.
| |
Collapse
|
11
|
Gardner JJ, Cushen SC, Oliveira da Silva RDN, Bradshaw JL, Hula N, Gorham IK, Tucker SM, Zhou Z, Cunningham RL, Phillips NR, Goulopoulou S. Oxidative stress induces release of mitochondrial DNA into the extracellular space in human placental villous trophoblast BeWo cells. Am J Physiol Cell Physiol 2024; 326:C1776-C1788. [PMID: 38738304 PMCID: PMC11371324 DOI: 10.1152/ajpcell.00091.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 04/26/2024] [Accepted: 04/26/2024] [Indexed: 05/14/2024]
Abstract
Circulating cell-free mitochondrial DNA (ccf-mtDNA) is an indicator of cell death, inflammation, and oxidative stress. ccf-mtDNA in pregnancies with placental dysfunction differs from that in healthy pregnancies, and the direction of this difference depends on gestational age and method of mtDNA quantification. Reactive oxygen species (ROS) trigger release of mtDNA, yet it is unknown whether trophoblast cells release mtDNA in response to oxidative stress, a common feature of pregnancies with placental pathology. We hypothesized that oxidative stress would induce cell death and release of mtDNA from trophoblast cells. BeWo cells were treated with antimycin A (10-320 µM) or rotenone (0.2-50 µM) to induce oxidative stress. A multiplex real-time quantitative PCR (qPCR) assay was used to quantify mtDNA and nuclear DNA in membrane-bound, non-membrane-bound, and vesicle-bound forms in cell culture supernatants and cell lysates. Treatment with antimycin A increased ROS (P < 0.0001), induced cell necrosis (P = 0.0004) but not apoptosis (P = 0.6471), and was positively associated with release of membrane-bound and non-membrane-bound mtDNA (P < 0.0001). Antimycin A increased mtDNA content in exosome-like extracellular vesicles (vesicle-bound form; P = 0.0019) and reduced autophagy marker expression (LC3A/B, P = 0.0002; p62, P < 0.001). Rotenone treatment did not influence mtDNA release or cell death (P > 0.05). Oxidative stress induces release of mtDNA into the extracellular space and causes nonapoptotic cell death and a reduction in autophagy markers in BeWo cells, an established in vitro model of human trophoblast cells. Intersection between autophagy and necrosis may mediate the release of mtDNA from the placenta in pregnancies exposed to oxidative stress.NEW & NOTEWORTHY This is the first study to test whether trophoblast cells release mitochondrial (mt)DNA in response to oxidative stress and to identify mechanisms of release and biological forms of mtDNA from this cellular type. This research identifies potential cellular mechanisms that can be used in future investigations to establish the source and biomarker potential of circulating mtDNA in preclinical experimental models and humans.
Collapse
Affiliation(s)
- Jennifer J Gardner
- 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, Texas, United States
| | - Reneé de Nazaré Oliveira da Silva
- Lawrence D. Longo, MD Center for Perinatal Biology, Departments of Basic Sciences, Gynecology, and Obstetrics, Loma Linda University School of Medicine, Loma Linda, California, United States
| | - Jessica L Bradshaw
- Department of Pharmaceutical Sciences, System College of Pharmacy, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Nataliia Hula
- Lawrence D. Longo, MD Center for Perinatal Biology, Departments of Basic Sciences, Gynecology, and Obstetrics, Loma Linda University School of Medicine, Loma Linda, California, United States
| | - Isabelle K Gorham
- Department of Microbiology, Immunology, and Genetics, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Selina M Tucker
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Zhengyang Zhou
- Department of Population & Community Health, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Rebecca L Cunningham
- Department of Pharmaceutical Sciences, System College of Pharmacy, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Nicole R Phillips
- Department of Microbiology, Immunology, and Genetics, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Styliani Goulopoulou
- Lawrence D. Longo, MD Center for Perinatal Biology, Departments of Basic Sciences, Gynecology, and Obstetrics, Loma Linda University School of Medicine, Loma Linda, California, United States
| |
Collapse
|
12
|
Álvarez-Sánchez A, Grinat J, Doria-Borrell P, Mellado-López M, Pedrera-Alcócer É, Malenchini M, Meseguer S, Hemberger M, Pérez-García V. The GPI-anchor biosynthesis pathway is critical for syncytiotrophoblast differentiation and placental development. Cell Mol Life Sci 2024; 81:246. [PMID: 38819479 PMCID: PMC11143174 DOI: 10.1007/s00018-024-05284-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 05/08/2024] [Accepted: 05/16/2024] [Indexed: 06/01/2024]
Abstract
The glycosylphosphatidylinositol (GPI) biosynthetic pathway in the endoplasmic reticulum (ER) is crucial for generating GPI-anchored proteins (GPI-APs), which are translocated to the cell surface and play a vital role in cell signaling and adhesion. This study focuses on two integral components of the GPI pathway, the PIGL and PIGF proteins, and their significance in trophoblast biology. We show that GPI pathway mutations impact on placental development impairing the differentiation of the syncytiotrophoblast (SynT), and especially the SynT-II layer, which is essential for the establishment of the definitive nutrient exchange area within the placental labyrinth. CRISPR/Cas9 knockout of Pigl and Pigf in mouse trophoblast stem cells (mTSCs) confirms the role of these GPI enzymes in syncytiotrophoblast differentiation. Mechanistically, impaired GPI-AP generation induces an excessive unfolded protein response (UPR) in the ER in mTSCs growing in stem cell conditions, akin to what is observed in human preeclampsia. Upon differentiation, the impairment of the GPI pathway hinders the induction of WNT signaling for early SynT-II development. Remarkably, the transcriptomic profile of Pigl- and Pigf-deficient cells separates human patient placental samples into preeclampsia and control groups, suggesting an involvement of Pigl and Pigf in establishing a preeclamptic gene signature. Our study unveils the pivotal role of GPI biosynthesis in early placentation and uncovers a new preeclampsia gene expression profile associated with mutations in the GPI biosynthesis pathway, providing novel molecular insights into placental development with implications for enhanced patient stratification and timely interventions.
Collapse
Affiliation(s)
- Andrea Álvarez-Sánchez
- Centro de Investigación Príncipe Felipe, Calle de Eduardo Primo Yúfera, 3, 46012, Valencia, Spain
| | - Johanna Grinat
- Epigenetics Programme, The Babraham Institute, Babraham Research Campus, Cambridge, UK
| | - Paula Doria-Borrell
- Centro de Investigación Príncipe Felipe, Calle de Eduardo Primo Yúfera, 3, 46012, Valencia, Spain
| | - Maravillas Mellado-López
- Centro de Investigación Príncipe Felipe, Calle de Eduardo Primo Yúfera, 3, 46012, Valencia, Spain
| | - Érica Pedrera-Alcócer
- Centro de Investigación Príncipe Felipe, Calle de Eduardo Primo Yúfera, 3, 46012, Valencia, Spain
| | - Marta Malenchini
- Centro de Investigación Príncipe Felipe, Calle de Eduardo Primo Yúfera, 3, 46012, Valencia, Spain
| | - Salvador Meseguer
- Centro de Investigación Príncipe Felipe, Calle de Eduardo Primo Yúfera, 3, 46012, Valencia, Spain
| | - Myriam Hemberger
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Canada
| | - Vicente Pérez-García
- Centro de Investigación Príncipe Felipe, Calle de Eduardo Primo Yúfera, 3, 46012, Valencia, Spain.
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Madrid, Spain.
| |
Collapse
|
13
|
Dos Santos Silva P, Kra G, Butenko Y, Daddam JR, Levin Y, Zachut M. Maternal supplementation with n-3 fatty acids affects placental lipid metabolism, inflammation, oxidative stress, the endocannabinoid system, and the neonate cytokine concentrations in dairy cows. J Anim Sci Biotechnol 2024; 15:74. [PMID: 38769527 PMCID: PMC11106909 DOI: 10.1186/s40104-024-01033-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 04/06/2024] [Indexed: 05/22/2024] Open
Abstract
BACKGROUND The placenta plays a crucial role in supporting and influencing fetal development. We compared the effects of prepartum supplementation with omega-3 (n-3) fatty acid (FA) sources, flaxseed oil (FLX) and fish oil (FO), on the expression of genes and proteins related to lipid metabolism, inflammation, oxidative stress, and the endocannabinoid system (ECS) in the expelled placenta, as well as on FA profile and inflammatory response of neonates. Late-pregnant Holstein dairy cows were supplemented with saturated fat (CTL), FLX, or FO. Placental cotyledons (n = 5) were collected immediately after expulsion, and extracted RNA and proteins were analyzed by RT-PCR and proteomic analysis. Neonatal blood was assessed for FA composition and concentrations of inflammatory markers. RESULTS FO increased the gene expression of fatty acid binding protein 4 (FABP4), interleukin 10 (IL-10), catalase (CAT), cannabinoid receptor 1 (CNR1), and cannabinoid receptor 2 (CNR2) compared with CTL placenta. Gene expression of ECS-enzyme FA-amide hydrolase (FAAH) was lower in FLX and FO than in CTL. Proteomic analysis identified 3,974 proteins; of these, 51-59 were differentially abundant between treatments (P ≤ 0.05, |fold change| ≥ 1.5). Top canonical pathways enriched in FLX vs. CTL and in FO vs. CTL were triglyceride metabolism and inflammatory processes. Both n-3 FA increased the placental abundance of FA binding proteins (FABPs) 3 and 7. The abundance of CNR1 cannabinoid-receptor-interacting-protein-1 (CNRIP1) was reduced in FO vs. FLX. In silico modeling affirmed that bovine FABPs bind to endocannabinoids. The FLX increased the abundance of inflammatory CD44-antigen and secreted-phosphoprotein-1, whereas prostaglandin-endoperoxide synthase 2 was decreased in FO vs. CTL placenta. Maternal FO enriched neonatal plasma with n-3 FAs, and both FLX and FO reduced interleukin-6 concentrations compared with CTL. CONCLUSION Maternal n-3 FA from FLX and FO differentially affected the bovine placenta; both enhanced lipid metabolism and modulated oxidative stress, however, FO increased some transcriptional ECS components, possibly related to the increased FABPs. Maternal FO induced a unique balance of pro- and anti-inflammatory components in the placenta. Taken together, different sources of n-3 FA during late pregnancy enhanced placental immune and metabolic processes, which may affect the neonatal immune system.
Collapse
Affiliation(s)
- Priscila Dos Santos Silva
- Department of Ruminant Science, Institute of Animal Sciences, ARO Volcani Institute, Rishon LeZion, Israel
| | - Gitit Kra
- Department of Ruminant Science, Institute of Animal Sciences, ARO Volcani Institute, Rishon LeZion, Israel
- Department of Animal Science, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Yana Butenko
- Department of Ruminant Science, Institute of Animal Sciences, ARO Volcani Institute, Rishon LeZion, Israel
| | | | - Yishai Levin
- The Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot, Israel
| | - Maya Zachut
- Department of Ruminant Science, Institute of Animal Sciences, ARO Volcani Institute, Rishon LeZion, Israel.
| |
Collapse
|
14
|
Podinic T, Limoges L, Monaco C, MacAndrew A, Minhas M, Nederveen J, Raha S. Cannabidiol Disrupts Mitochondrial Respiration and Metabolism and Dysregulates Trophoblast Cell Differentiation. Cells 2024; 13:486. [PMID: 38534330 PMCID: PMC10968792 DOI: 10.3390/cells13060486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 03/01/2024] [Accepted: 03/06/2024] [Indexed: 03/28/2024] Open
Abstract
Trophoblast differentiation is a crucial process in the formation of the placenta where cytotrophoblasts (CTs) differentiate and fuse to form the syncytiotrophoblast (ST). The bioactive components of cannabis, such as Δ9-THC, are known to disrupt trophoblast differentiation and fusion, as well as mitochondrial dynamics and respiration. However, less is known about the impact of cannabidiol (CBD) on trophoblast differentiation. Due to the central role of mitochondria in stem cell differentiation, we evaluated the impact of CBD on trophoblast mitochondrial function and differentiation. Using BeWo b30 cells, we observed decreased levels of mRNA for markers of syncytialization (GCM1, ERVW1, hCG) following 20 µM CBD treatment during differentiation. In CTs, CBD elevated transcript levels for the mitochondrial and cellular stress markers HSP60 and HSP70, respectively. Furthermore, CBD treatment also increased the lipid peroxidation and oxidative damage marker 4-hydroxynonenal. Mitochondrial membrane potential, basal respiration and ATP production were diminished with the 20 µM CBD treatment in both sub-lineages. mRNA levels for endocannabinoid system (ECS) components (FAAH, NAPEPLD, TRPV1, CB1, CB2, PPARγ) were altered differentially by CBD in CTs and STs. Overall, we demonstrate that CBD impairs trophoblast differentiation and fusion, as well as mitochondrial bioenergetics and redox homeostasis.
Collapse
Affiliation(s)
- Tina Podinic
- Graduate Program in Medical Sciences, Department of Pediatrics, McMaster University, 1280 Main St. W., Hamilton, ON L8S 4K1, Canada; (T.P.); (L.L.); (C.M.); (A.M.)
| | - Louise Limoges
- Graduate Program in Medical Sciences, Department of Pediatrics, McMaster University, 1280 Main St. W., Hamilton, ON L8S 4K1, Canada; (T.P.); (L.L.); (C.M.); (A.M.)
| | - Cristina Monaco
- Graduate Program in Medical Sciences, Department of Pediatrics, McMaster University, 1280 Main St. W., Hamilton, ON L8S 4K1, Canada; (T.P.); (L.L.); (C.M.); (A.M.)
| | - Andie MacAndrew
- Graduate Program in Medical Sciences, Department of Pediatrics, McMaster University, 1280 Main St. W., Hamilton, ON L8S 4K1, Canada; (T.P.); (L.L.); (C.M.); (A.M.)
| | - Mahek Minhas
- Department of Pediatrics, McMaster University, 1280 Main St. W., Hamilton, ON L8S 4K1, Canada; (M.M.); (J.N.)
- Department of Kinesiology, McMaster University, 1280 Main St. W., Hamilton, ON L8S 4K1, Canada
| | - Joshua Nederveen
- Department of Pediatrics, McMaster University, 1280 Main St. W., Hamilton, ON L8S 4K1, Canada; (M.M.); (J.N.)
- Department of Kinesiology, McMaster University, 1280 Main St. W., Hamilton, ON L8S 4K1, Canada
| | - Sandeep Raha
- Graduate Program in Medical Sciences, Department of Pediatrics, McMaster University, 1280 Main St. W., Hamilton, ON L8S 4K1, Canada; (T.P.); (L.L.); (C.M.); (A.M.)
| |
Collapse
|
15
|
Gardner JJ, Cushen SC, Oliveira da Silva RDN, Bradshaw JL, Hula N, Gorham IK, Tucker SM, Zhou Z, Cunningham RL, Phillips NR, Goulopoulou S. Oxidative stress induces release of mitochondrial DNA into the extracellular space in human placental villous trophoblast BeWo cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.02.578433. [PMID: 38352590 PMCID: PMC10862877 DOI: 10.1101/2024.02.02.578433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2024]
Abstract
Circulating cell-free mitochondrial DNA (ccf-mtDNA) is an indicator of cell death, inflammation, and oxidative stress. ccf-mtDNA differs in pregnancies with placental dysfunction from healthy pregnancies and the direction of this difference depends on gestational age and method of mtDNA quantification. Reactive oxygen species (ROS) trigger release of mtDNA from non-placental cells; yet it is unknown whether trophoblast cells release mtDNA in response to oxidative stress, a common feature of pregnancies with placental pathology. We hypothesized that oxidative stress would induce cell death and release of mtDNA from trophoblast cells. BeWo cells were treated with antimycin A (10-320 μM) or rotenone (0.2-50 μM) to induce oxidative stress. A multiplex real-time quantitative PCR (qPCR) assay was used to quantify mtDNA and nuclear DNA in membrane bound, non-membrane bound, and vesicular-bound forms in cell culture supernatants and cell lysates. Treatment with antimycin A increased ROS (p<0.0001), induced cell necrosis (p=0.0004) but not apoptosis (p=0.6471) and was positively associated with release of membrane-bound and non-membrane bound mtDNA (p<0.0001). Antimycin A increased mtDNA content in exosome-like extracellular vesicles (vesicular-bound form; p=0.0019) and reduced autophagy marker expression (LC3A/B, p=0.0002; p62, p<0.001). Rotenone treatment did not influence mtDNA release or cell death (p>0.05). Oxidative stress induces release of mtDNA into the extracellular space and causes non-apoptotic cell death and a reduction in autophagy markers in BeWo cells, an established in vitro model of human trophoblast cells. Intersection between autophagy and necrosis may mediate the release of mtDNA from the placenta in pregnancies exposed to oxidative stress. NEW & NOTEWORTHY This is the first study to test whether trophoblast cells release mitochondrial DNA in response to oxidative stress and to identify mechanisms of release and biological forms of mtDNA from this cellular type. This research identifies potential cellular mechanisms that can be used in future investigations to establish the source and biomarker potential of circulating mitochondrial DNA in preclinical experimental models and humans.
Collapse
|
16
|
Mangrulkar SV, Wankhede NL, Kale MB, Upaganlawar AB, Taksande BG, Umekar MJ, Anwer MK, Dailah HG, Mohan S, Behl T. Mitochondrial Dysfunction as a Signaling Target for Therapeutic Intervention in Major Neurodegenerative Disease. Neurotox Res 2023; 41:708-729. [PMID: 37162686 DOI: 10.1007/s12640-023-00647-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 12/29/2022] [Accepted: 04/08/2023] [Indexed: 05/11/2023]
Abstract
Neurodegenerative diseases (NDD) are incurable and the most prevalent cognitive and motor disorders of elderly. Mitochondria are essential for a wide range of cellular processes playing a pivotal role in a number of cellular functions like metabolism, intracellular signaling, apoptosis, and immunity. A plethora of evidence indicates the central role of mitochondrial functions in pathogenesis of many aging related NDD. Considering how mitochondria function in neurodegenerative diseases, oxidative stress, and mutations in mtDNA both contribute to aging. Many substantial reports suggested the involvement of numerous contributing factors including, mitochondrial dysfunction, oxidative stress, mitophagy, accumulation of somatic mtDNA mutations, compromised mitochondrial dynamics, and transport within axons in neurodegenerative disorders including Alzheimer's disease, Parkinson's disease, Huntington's disease, and Amyotrophic Lateral Sclerosis. Therapies therefore target fundamental mitochondrial processes such as energy metabolism, free-radical generation, mitochondrial biogenesis, mitochondrial redox state, mitochondrial dynamics, mitochondrial protein synthesis, mitochondrial quality control, and metabolism hold great promise to develop pharmacological based therapies in NDD. By emphasizing the most efficient pharmacological strategies to target dysfunction of mitochondria in the treatment of neurodegenerative diseases, this review serves the scientific community engaged in translational medical science by focusing on the establishment of novel, mitochondria-targeted treatment strategies.
Collapse
Affiliation(s)
| | - Nitu L Wankhede
- Smt. Shantabai Patil College of Diploma in Pharmacy, Kamptee, Nagpur, Maharashtra, India
| | - Mayur B Kale
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra, India
| | - Aman B Upaganlawar
- SNJB's Shriman Sureshdada Jain College of Pharmacy, Neminagar, Chandwad, Nasik, Maharashta, India
| | - Brijesh G Taksande
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra, India
| | - Milind J Umekar
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra, India
| | - Md Khalid Anwer
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, 16278, Saudi Arabia
| | - Hamad Ghaleb Dailah
- Research and Scientific Studies Unit, College of Nursing, Jazan University, Jazan, Saudi Arabia
| | - Syam Mohan
- Substance Abuse and Toxicology Research Center, Jazan University, Jazan, Saudi Arabia
- School of Health Sciences and Technology, University of Petroleum and Energy Studies, Dehradun, Uttarakhand, India
- Centre for Transdisciplinary Research, Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Science, Saveetha University, Chennai, India
| | - Tapan Behl
- School of Health Sciences and Technology, University of Petroleum and Energy Studies, Dehradun, Uttarakhand, India.
| |
Collapse
|
17
|
Opichka MA, Livergood MC, Balapattabi K, Ritter ML, Brozoski DT, Wackman KK, Lu KT, Kozak KN, Wells C, Fogo AB, Gibson-Corley KN, Kwitek AE, Sigmund CD, McIntosh JJ, Grobe JL. Mitochondrial-targeted antioxidant attenuates preeclampsia-like phenotypes induced by syncytiotrophoblast-specific Gαq signaling. SCIENCE ADVANCES 2023; 9:eadg8118. [PMID: 38039359 PMCID: PMC10691776 DOI: 10.1126/sciadv.adg8118] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 10/31/2023] [Indexed: 12/03/2023]
Abstract
Syncytiotrophoblast stress is theorized to drive development of preeclampsia, but its molecular causes and consequences remain largely undefined. Multiple hormones implicated in preeclampsia signal via the Gαq cascade, leading to the hypothesis that excess Gαq signaling within the syncytiotrophoblast may contribute. First, we present data supporting increased Gαq signaling and antioxidant responses within villous and syncytiotrophoblast samples of human preeclamptic placenta. Second, Gαq was activated in mouse placenta using Cre-lox and DREADD methodologies. Syncytiotrophoblast-restricted Gαq activation caused hypertension, kidney damage, proteinuria, elevated circulating proinflammatory factors, decreased placental vascularization, diminished spiral artery diameter, and augmented responses to mitochondrial-derived superoxide. Administration of the mitochondrial-targeted antioxidant Mitoquinone attenuated maternal proteinuria, lowered circulating inflammatory and anti-angiogenic mediators, and maintained placental vascularization. These data demonstrate a causal relationship between syncytiotrophoblast stress and the development of preeclampsia and identify elevated Gαq signaling and mitochondrial reactive oxygen species as a cause of this stress.
Collapse
Affiliation(s)
- Megan A. Opichka
- Department of Physiology, Medical College of Wisconsin, Milwaukee, USA
| | | | | | | | | | - Kelsey K. Wackman
- Department of Physiology, Medical College of Wisconsin, Milwaukee, USA
| | - Ko-Ting Lu
- Department of Physiology, Medical College of Wisconsin, Milwaukee, USA
| | - Kaleigh N. Kozak
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, USA
| | - Clive Wells
- Electron Microscopy Core Facility, Medical College of Wisconsin, Milwaukee, USA
| | - Agnes B. Fogo
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, USA
| | - Katherine N. Gibson-Corley
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, USA
| | - Anne E. Kwitek
- Department of Physiology, Medical College of Wisconsin, Milwaukee, USA
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, USA
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, USA
| | - Curt D. Sigmund
- Department of Physiology, Medical College of Wisconsin, Milwaukee, USA
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, USA
- Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, USA
| | - Jennifer J. McIntosh
- Department of Physiology, Medical College of Wisconsin, Milwaukee, USA
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, USA
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, USA
| | - Justin L. Grobe
- Department of Physiology, Medical College of Wisconsin, Milwaukee, USA
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, USA
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, USA
- Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, USA
- Comprehensive Rodent Metabolic Phenotyping Core, Medical College of Wisconsin, Milwaukee, USA
| |
Collapse
|
18
|
Podinić T, Werstuck G, Raha S. The Implications of Cannabinoid-Induced Metabolic Dysregulation for Cellular Differentiation and Growth. Int J Mol Sci 2023; 24:11003. [PMID: 37446181 DOI: 10.3390/ijms241311003] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 06/27/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023] Open
Abstract
The endocannabinoid system (ECS) governs and coordinates several physiological processes through an integrated signaling network, which is responsible for inducing appropriate intracellular metabolic signaling cascades in response to (endo)cannabinoid stimulation. This intricate cellular system ensures the proper functioning of the immune, reproductive, and nervous systems and is involved in the regulation of appetite, memory, metabolism, and development. Cannabinoid receptors have been observed on both cellular and mitochondrial membranes in several tissues and are stimulated by various classes of cannabinoids, rendering the ECS highly versatile. In the context of growth and development, emerging evidence suggests a crucial role for the ECS in cellular growth and differentiation. Indeed, cannabinoids have the potential to disrupt key energy-sensing metabolic signaling pathways requiring mitochondrial-ER crosstalk, whose functioning is essential for successful cellular growth and differentiation. This review aims to explore the extent of cannabinoid-induced cellular dysregulation and its implications for cellular differentiation.
Collapse
Affiliation(s)
- Tina Podinić
- The Department of Pediatrics and the Graduate Program in Medical Sciences, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Geoff Werstuck
- Department of Medicine and the Thrombosis and Atherosclerosis Research Institute, David Braley Research Institute, McMaster University, Hamilton, ON L8L 2X2, Canada
| | - Sandeep Raha
- The Department of Pediatrics and the Graduate Program in Medical Sciences, McMaster University, Hamilton, ON L8S 4K1, Canada
| |
Collapse
|
19
|
Malledevarahalli Chandrappa S, Pascottini OB, Opsomer G, Meineri G, Martino NA, Banchi P, Vincenti L, Ricci A. Circulating and endometrial cell oxidative stress in dairy cows diagnosed with metritis. Theriogenology 2023; 198:217-223. [PMID: 36610371 DOI: 10.1016/j.theriogenology.2022.12.045] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 12/29/2022] [Accepted: 12/30/2022] [Indexed: 01/01/2023]
Abstract
Dairy cows diagnosed with metritis may experience a greater degree of oxidative stress (OS) and a deficit in the antioxidative capacity compared to healthy cows. We aimed to assess circulating OS markers and endometrial cell mitochondrial function, intracellular reactive oxygen species (ROS) production, and mean endometrial nuclear cell area in postpartum cows diagnosed with metritis or as healthy. From an initial pool of 121 Holstein cows, we retrospectively selected 34 cows and balanced for metritis (n = 17) or healthy (n = 17). Metritis was defined as an enlarged uterus with red-brown watery or thick off-white purulent discharge occurring within 21 days postpartum. Cows with no signs of clinical disease (including dystocia or retained placenta) were referred to as healthy. Blood samples for serum reactive oxygen metabolites (d-ROM), antioxidants (OXY), and oxidative status index (OSI) tests, evaluated via photometric determination of plasma thiols, were performed at 7, 14, 21, 28, and 35 days postpartum. Furthermore, from the initial pool, a random subset of 5 cows diagnosed with metritis and 6 diagnosed as healthy we collected (at the same time points as for the blood samples) endometrial cytology samples using the cytobrush technique. From the uterine samples, we evaluated the endometrial cell mitochondrial function, intracellular ROS levels, and the endometrial cell nuclear area using MitoTracker Orange, dichlorodihydrofluorescein diacetate, and Hoechst 33258, respectively. Mixed linear regression models, accounting for repeated measurements, were fitted to assess the effect of metritis versus healthy on circulating and endometrial cell OS parameters and endometrial cell size. The effect of days postpartum and its interaction with uterine health status were forced into each model. Serum concentrations of d-ROMs and OSI were greater in metritis at 7, 14, and 35 days postpartum than in healthy cows. Interestingly, the mean endometrial cell nuclear area was lower in metritis than healthy cows at 14 and 21 days postpartum. We found no differences between metritis and healthy for endometrial cell mitochondrial function and intracellular ROS production. In conclusion, cows diagnosed with metritis experienced greater systemic OS levels than healthy cows, but their OS was not higher in the uterine milieu.
Collapse
Affiliation(s)
- Sanjana Malledevarahalli Chandrappa
- Department of Veterinary Sciences, University of Turin, 10095, Grugliasco, Italy; Department of Internal Medicine, Reproduction and Population Medicine, Faculty of Veterinary Medicine, Ghent University, 9820, Merelbeke, Belgium.
| | - Osvaldo Bogado Pascottini
- Department of Internal Medicine, Reproduction and Population Medicine, Faculty of Veterinary Medicine, Ghent University, 9820, Merelbeke, Belgium; Laboratory for Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, University of Antwerp, 2610, Wilrijk, Belgium
| | - Geert Opsomer
- Department of Internal Medicine, Reproduction and Population Medicine, Faculty of Veterinary Medicine, Ghent University, 9820, Merelbeke, Belgium
| | - Giorgia Meineri
- Department of Veterinary Sciences, University of Turin, 10095, Grugliasco, Italy
| | - Nicola Antonio Martino
- Department of Biosciences, Biotechnologies & Environment, University of Bari Aldo Moro, Valenzano, Bari, Italy
| | - Penelope Banchi
- Department of Veterinary Sciences, University of Turin, 10095, Grugliasco, Italy; Department of Internal Medicine, Reproduction and Population Medicine, Faculty of Veterinary Medicine, Ghent University, 9820, Merelbeke, Belgium
| | - Leila Vincenti
- Department of Veterinary Sciences, University of Turin, 10095, Grugliasco, Italy
| | - Alessandro Ricci
- Department of Veterinary Sciences, University of Turin, 10095, Grugliasco, Italy
| |
Collapse
|
20
|
Yang W, Kang Q, Li C, Bo S, Wang Y. Matrine promotes trophoblast invasion and reduces inflammation via miR-19a-3p to prevent preeclampsia. Mol Cell Toxicol 2022. [DOI: 10.1007/s13273-022-00293-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
21
|
Micro-RNAs in Human Placenta: Tiny Molecules, Immense Power. Molecules 2022; 27:molecules27185943. [PMID: 36144676 PMCID: PMC9501247 DOI: 10.3390/molecules27185943] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 08/30/2022] [Accepted: 09/09/2022] [Indexed: 12/06/2022] Open
Abstract
Micro-RNAs (miRNAs) are short non-coding single-stranded RNAs that modulate the expression of various target genes after transcription. The expression and distribution of kinds of miRNAs have been characterized in human placenta during different gestational stages. The identified miRNAs are recognized as key mediators in the regulation of placental development and in the maintenance of human pregnancy. Aberrant expression of miRNAs is associated with compromised pregnancies in humans, and dysregulation of those miRNAs contributes to the occurrence and development of related diseases during pregnancy, such as pre-eclampsia (PE), fetal growth restriction (FGR), gestational diabetes mellitus (GDM), recurrent miscarriage, preterm birth (PTB) and small-for-gestational-age (SGA). Thus, having a better understanding of the expression and functions of miRNAs in human placenta during pregnancy and thereby developing novel drugs targeting the miRNAs could be a potentially promising method in the prevention and treatment of relevant diseases in future. Here, we summarize the current knowledge of the expression pattern and function regulation of miRNAs in human placental development and related diseases.
Collapse
|
22
|
Rathod S, Arya S, Kanike S, Shah SA, Bahadur P, Tiwari S. Advances on nanoformulation approaches for delivering plant-derived antioxidants: A case of quercetin. Int J Pharm 2022; 625:122093. [PMID: 35952801 DOI: 10.1016/j.ijpharm.2022.122093] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 07/21/2022] [Accepted: 08/04/2022] [Indexed: 10/15/2022]
Abstract
Oxidative stress has been implicated in tumorigenic, cardiovascular, neuro-, and age-related degenerative changes. Antioxidants minimize the oxidative damage through neutralization of reactive oxygen species (ROS) and other causative agents. Ever since the emergence of COVID-19, plant-derived antioxidants have received enormous attention, particularly in the Indian subcontinent. Quercetin (QCT), a bio-flavonoid, exists in the glycosylated form in fruits, berries and vegetables. The antioxidant potential of QCT analogs relates to the number of free hydroxyl groups in their structure. Despite presence of these groups, QCT exhibits substantial hydrophobicity. Formulation scientists have tested nanotechnology-based approaches for its improved solubilization and delivery to the intended site of action. By the virtue of its hydrophobicity, QCT gets encapsulated in nanocarriers carrying hydrophobic domains. Apart from passive accumulation, active uptake of such formulations into the target cells can be facilitated through well-studied functionalization strategies. In this review, we have discussed the approaches of improving solubilization and bioavailability of QCT with the use of nanoformulations.
Collapse
Affiliation(s)
- Sachin Rathod
- UKA Tarsadia University, Maliba Pharmacy College, Gopal-Vidyanagar Campus, Surat 394350, India
| | - Shristi Arya
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) - Raebareli, Lucknow 226002, India
| | - Shirisha Kanike
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) - Raebareli, Lucknow 226002, India
| | - Shailesh A Shah
- UKA Tarsadia University, Maliba Pharmacy College, Gopal-Vidyanagar Campus, Surat 394350, India
| | - Pratap Bahadur
- Department of Chemistry, Veer Narmad South Gujarat University, Surat 395007, India
| | - Sanjay Tiwari
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) - Raebareli, Lucknow 226002, India.
| |
Collapse
|
23
|
Renaud SJ, Jeyarajah MJ. How trophoblasts fuse: an in-depth look into placental syncytiotrophoblast formation. Cell Mol Life Sci 2022; 79:433. [PMID: 35859055 PMCID: PMC11072895 DOI: 10.1007/s00018-022-04475-z] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 05/07/2022] [Accepted: 07/06/2022] [Indexed: 11/24/2022]
Abstract
In humans, cell fusion is restricted to only a few cell types under normal conditions. In the placenta, cell fusion is a critical process for generating syncytiotrophoblast: the giant multinucleated trophoblast lineage containing billions of nuclei within an interconnected cytoplasm that forms the primary interface separating maternal blood from fetal tissue. The unique morphology of syncytiotrophoblast ensures that nutrients and gases can be efficiently transferred between maternal and fetal tissue while simultaneously restricting entry of potentially damaging substances and maternal immune cells through intercellular junctions. To maintain integrity of the syncytiotrophoblast layer, underlying cytotrophoblast progenitor cells terminate their capability for self-renewal, upregulate expression of genes needed for differentiation, and then fuse into the overlying syncytium. These processes are disrupted in a variety of obstetric complications, underscoring the importance of proper syncytiotrophoblast formation for pregnancy health. Herein, an overview of key mechanisms underlying human trophoblast fusion and syncytiotrophoblast development is discussed.
Collapse
Affiliation(s)
- Stephen J Renaud
- Department of Anatomy and Cell Biology and Children's Health Research Institute, University of Western Ontario, London, ON, N6A5C1, Canada.
| | - Mariyan J Jeyarajah
- Department of Anatomy and Cell Biology and Children's Health Research Institute, University of Western Ontario, London, ON, N6A5C1, Canada
| |
Collapse
|
24
|
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
|
25
|
Differences and Interactions in Placental Manganese and Iron Transfer across an In Vitro Model of Human Villous Trophoblasts. Int J Mol Sci 2022; 23:ijms23063296. [PMID: 35328723 PMCID: PMC8951728 DOI: 10.3390/ijms23063296] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 03/03/2022] [Accepted: 03/14/2022] [Indexed: 12/10/2022] Open
Abstract
Manganese (Mn) as well as iron (Fe) are essential trace elements (TE) important for the maintenance of physiological functions including fetal development. However, in the case of Mn, evidence suggests that excess levels of intrauterine Mn are associated with adverse pregnancy outcomes. Although Mn is known to cross the placenta, the fundamentals of Mn transfer kinetics and mechanisms are largely unknown. Moreover, exposure to combinations of TEs should be considered in mechanistic transfer studies, in particular for TEs expected to share similar transfer pathways. Here, we performed a mechanistic in vitro study on the placental transfer of Mn across a BeWo b30 trophoblast layer. Our data revealed distinct differences in the placental transfer of Mn and Fe. While placental permeability to Fe showed a clear inverse dose-dependency, Mn transfer was largely independent of the applied doses. Concurrent exposure of Mn and Fe revealed transfer interactions of Fe and Mn, indicating that they share common transfer mechanisms. In general, mRNA and protein expression of discussed transporters like DMT1, TfR, or FPN were only marginally altered in BeWo cells despite the different exposure scenarios highlighting that Mn transfer across the trophoblast layer likely involves a combination of active and passive transport processes.
Collapse
|
26
|
Mandakh Y, Oudin A, Erlandsson L, Isaxon C, Hansson SR, Broberg K, Malmqvist E. Association of Prenatal Ambient Air Pollution Exposure With Placental Mitochondrial DNA Copy Number, Telomere Length and Preeclampsia. FRONTIERS IN TOXICOLOGY 2022; 3:659407. [PMID: 35295138 PMCID: PMC8915808 DOI: 10.3389/ftox.2021.659407] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 04/19/2021] [Indexed: 12/22/2022] Open
Abstract
Background: Studies have shown that ambient air pollution is linked to preeclampsia (PE), possibly via generation of oxidative stress in the placenta. Telomere length and mitochondrial DNA copy number (mtDNAcn) are sensitive to oxidative stress damage. Objective: To study the association between prenatal exposure to ambient nitrogen oxides (NOx, a marker for traffic-related air pollution), and PE, as well as potential mediation effects by placental telomere length and mtDNAcn. Methods: This is a cross-sectional study of 42 preeclamptic and 95 arbitrarily selected normotensive pregnant women with gestational ambient NOx exposure assessment in southern Scania, Sweden. Hourly concentrations of NOx were estimated at the residential addresses by a Gaussian-plume dispersion model with 100 × 100 m spatial resolutions and aggregated into trimester-specific mean concentrations. Placental relative mtDNAcn and telomere length were measured using qPCR. Linear and logistic regression models were used to investigate associations, adjusted for perinatal and seasonal characteristics. Results: Exposure was categorized into low and high exposures by median cut-offs during first [11.9 μg/m3; interquartile range (IQR) 7.9, 17.9], second (11.6 μg/m3; IQR: 7.1, 21.1), third trimesters (11.9 μg/m3; IQR: 7.7, 19.5) and entire pregnancy (12.0 μg/m3; IQR: 7.6, 20.1). Increased risk of PE was found for high prenatal NOx exposure during the first trimester (OR 4.0; 95% CI: 1.4, 11.1; p = 0.008), and entire pregnancy (OR 3.7; 95% CI: 1.3, 10.4; p = 0.012). High exposed group during the first trimester had lower placental relative mtDNAcn compared with low exposed group (-0.20; 95% CI: -0.36, -0.04; p = 0.01). Changes in relative mtDNAcn did not mediate the association between prenatal NOx exposure and PE. No statistically significant association was found between placental relative telomere length, prenatal NOx exposure and PE. Conclusion: In this region with relatively low levels of air pollution, ambient NOx exposure during the first trimester was associated with reduced placental relative mtDNAcn and an increased risk of PE. However, we did not find any evidence that mtDNAcn or TL mediated the association between air pollution and PE. Future research should further investigate the role of mtDNAcn for pregnancy complications in relation to exposure to ambient air pollution during pregnancy.
Collapse
Affiliation(s)
- Yumjirmaa Mandakh
- Environment Society and Health, Division of Occupational and Environmental Medicine, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Anna Oudin
- Environment Society and Health, Division of Occupational and Environmental Medicine, Department of Laboratory Medicine, Lund University, Lund, Sweden.,Section of Sustainable Health, Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Lena Erlandsson
- Division of Obstetrics and Gynecology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Christina Isaxon
- Ergonomics and Aerosol Technology, Department of Design Sciences, Lund University, Lund, Sweden
| | - Stefan R Hansson
- Division of Obstetrics and Gynecology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden.,Department of Obstetrics & Gynaecology, Skåne University Hospital, Malmö, Sweden
| | - Karin Broberg
- Division of Occupational and Environmental Medicine, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Ebba Malmqvist
- Environment Society and Health, Division of Occupational and Environmental Medicine, Department of Laboratory Medicine, Lund University, Lund, Sweden
| |
Collapse
|
27
|
Wang X, Ma H, Sun J, Zheng T, Zhao P, Li H, Yang M. Mitochondrial Ferritin Deficiency Promotes Osteoblastic Ferroptosis Via Mitophagy in Type 2 Diabetic Osteoporosis. Biol Trace Elem Res 2022; 200:298-307. [PMID: 33594527 DOI: 10.1007/s12011-021-02627-z] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 02/03/2021] [Indexed: 12/15/2022]
Abstract
The incidence of type 2 diabetic osteoporosis (T2DOP), which seriously threatens elderly people's health, is rapidly increasing in recent years. However, the specific mechanism of the T2DOP is still unclear. Studies have shown the relationship between iron overload and T2DOP. Mitochondrial ferritin (FtMt) is a protein that stores iron ions and intercepts toxic ferrous ions in cells mitochondria. Ferroptosis, an iron-dependent cell injured way, may be related to the pathogenesis of T2DOP. In this study, we intend to elucidate the effect of FtMt on ferroptosis in osteoblasts and explain the possible mechanism. We first detected the occurrence of ferroptosis in bone tissue and the expression of FtMt after inducing T2DOP rat model. Then we used hFOB1.19 cells to study the influence of high glucose on FtMt, ferroptosis, and osteogenic function of osteoblasts. Then we observed the effect of FtMt on ferroptosis and osteoblast function by lentiviral silencing and overexpression of FtMt. We found ferroptosis in T2DOP rats bone. Overexpression of FtMt reduced osteoblastic ferroptosis under high glucose condition while silent FtMt induced mitophagy through ROS / PINK1/Parkin pathway. Then we found increased ferroptosis in osteoblasts after activating mitophagy by carbonyl cyanide-m-chlorophenyl-hydrazine (CCCP, a mitophagy agonist). Our study demonstrated that FtMt inhibited the occurrence of ferroptosis in osteoblasts by reducing oxidative stress caused by excess ferrous ions, and FtMt deficiency induced mitophagy in the pathogenesis of T2DOP. This study suggested that FtMt might serve as a potential target for T2DOP therapy.
Collapse
Affiliation(s)
- XinDong Wang
- Department of Orthopedics, The First Hospital of China Medical University, 155 North Nanjing Street, Shenyang, 110001, Liaoning, China
| | - HongDong Ma
- Department of Orthopedics, The First Hospital of China Medical University, 155 North Nanjing Street, Shenyang, 110001, Liaoning, China
| | - Jun Sun
- Department of Orthopedics, The Third Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - TianYu Zheng
- The VIP Department, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, Liaoning, China
| | - Peng Zhao
- Department of Orthopedics, The First Hospital of China Medical University, 155 North Nanjing Street, Shenyang, 110001, Liaoning, China
| | - HaiTian Li
- Department of Orthopedics, The First Hospital of China Medical University, 155 North Nanjing Street, Shenyang, 110001, Liaoning, China
| | - MaoWei Yang
- Department of Orthopedics, The First Hospital of China Medical University, 155 North Nanjing Street, Shenyang, 110001, Liaoning, China.
| |
Collapse
|
28
|
Chae SA, Son JS, Du M. Prenatal exercise in fetal development: a placental perspective. FEBS J 2021; 289:3058-3071. [PMID: 34449982 DOI: 10.1111/febs.16173] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 08/09/2021] [Accepted: 08/26/2021] [Indexed: 02/06/2023]
Abstract
Maternal obesity (MO) and gestational diabetes mellitus (GDM) are common in Western societies, which impair fetal development and predispose offspring to metabolic dysfunction. Placenta is the organ linking the mother to her fetus, and MO suppresses the development of vascular system and expression of nutrient transporters in placenta, thereby affecting fetal development. For maintaining its proper physiological function, placenta is energy demanding, which is met through extensive oxidative phosphorylation. However, the oxidative capacity of placenta is suppressed due to MO and GDM. Recently, several studies showed that physical activity during pregnancy enhances oxidative metabolism and improves placental function, which might be partially mediated by exerkines, referring to cytokines elicited by exercise. In addition, as an endocrine organ, placenta secretes cytokines, termed placentokines, including apelin, superoxide dismutase 3, irisin, and adiponectin, which mediate fetal development and maternal metabolism. Possible molecular mechanisms linking maternal exercise and placentokines to placental and fetal development are further discussed. As an emerging field, up to now, available studies are limited, mostly conducted in rodents. Given the epidemics of obesity and metabolic disorders, as well as the prevalence of maternal sedentary lifestyle, the effects of exercise of pregnant women on placental function and placentokine secretion, as well as their impacts on fetal development, need to be further examined.
Collapse
Affiliation(s)
- Song Ah Chae
- Nutrigenomics and Growth Biology Laboratory, Department of Animal Sciences, Washington State University, Pullman, WA, USA
| | - Jun Seok Son
- Laboratory of Perinatal Kinesioepigenetics, Department of Obstetrics, Gynecology & Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Min Du
- Nutrigenomics and Growth Biology Laboratory, Department of Animal Sciences, Washington State University, Pullman, WA, USA
| |
Collapse
|
29
|
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
|
30
|
Syncytiotrophoblast stress in early onset preeclampsia: The issues perpetuating the syndrome. Placenta 2021; 113:57-66. [PMID: 34053733 DOI: 10.1016/j.placenta.2021.05.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 04/26/2021] [Accepted: 05/14/2021] [Indexed: 01/08/2023]
Abstract
Preeclampsia is a pregnancy-specific syndrome characterized by a sudden increase in blood pressure accompanied by proteinuria and/or maternal multi-system damage associated to poor fetal outcome. In early-onset preeclampsia, utero-placental perfusion is altered, causing constant and progressive damage to the syncytiotrophoblast, generating syncytiotrophoblast stress. The latter leads to the detachment and release of syncytiotrophoblast fragments, anti-angiogenic factors and pro-inflammatory molecules into maternal circulation, resulting in the emergence and persistence of the characteristic symptoms of this syndrome during pregnancy. Therefore, understanding the origin and consequences of syncytiotrophoblast stress in preeclampsia is vital to develop new therapeutic alternatives, focused on reducing the burden of this syndrome. In this review, we describe five central characteristics of syncytial stress that should be targeted or prevented in order to reduce preeclampsia symptoms: histological alterations, syncytiotrophoblast damage, antiangiogenic protein export, placental deportation, and altered syncytiotrophoblast turnover. Therapeutic management of these characteristics may improve maternal and fetal outcomes.
Collapse
|
31
|
Hu XQ, Zhang L. Hypoxia and Mitochondrial Dysfunction in Pregnancy Complications. Antioxidants (Basel) 2021; 10:antiox10030405. [PMID: 33800426 PMCID: PMC7999178 DOI: 10.3390/antiox10030405] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/02/2021] [Accepted: 03/03/2021] [Indexed: 02/07/2023] Open
Abstract
Hypoxia is a common and severe stress to an organism's homeostatic mechanisms, and hypoxia during gestation is associated with significantly increased incidence of maternal complications of preeclampsia, adversely impacting on the fetal development and subsequent risk for cardiovascular and metabolic disease. Human and animal studies have revealed a causative role of increased uterine vascular resistance and placental hypoxia in preeclampsia and fetal/intrauterine growth restriction (FGR/IUGR) associated with gestational hypoxia. Gestational hypoxia has a major effect on mitochondria of uteroplacental cells to overproduce reactive oxygen species (ROS), leading to oxidative stress. Excess mitochondrial ROS in turn cause uteroplacental dysfunction by damaging cellular macromolecules, which underlies the pathogenesis of preeclampsia and FGR. In this article, we review the current understanding of hypoxia-induced mitochondrial ROS and their role in placental dysfunction and the pathogenesis of pregnancy complications. In addition, therapeutic approaches selectively targeting mitochondrial ROS in the placental cells are discussed.
Collapse
|
32
|
Walker OS, Gurm H, Sharma R, Verma N, May LL, Raha S. Delta-9-tetrahydrocannabinol inhibits invasion of HTR8/SVneo human extravillous trophoblast cells and negatively impacts mitochondrial function. Sci Rep 2021; 11:4029. [PMID: 33597628 PMCID: PMC7889882 DOI: 10.1038/s41598-021-83563-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Accepted: 01/29/2021] [Indexed: 01/31/2023] Open
Abstract
Prenatal cannabis use is a significant problem and poses important health risks for the developing fetus. The molecular mechanisms underlying these changes are not fully elucidated but are thought to be attributed to delta-9-tetrahydrocannabinol (THC), the main bioactive constituent of cannabis. It has been reported that THC may target the mitochondria in several tissue types, including placental tissue and trophoblast cell lines, and alter their function. In the present study, in response to 48-h THC treatment of the human extravillous trophoblast cell line HTR8/SVneo, we demonstrate that cell proliferation and invasion are significantly reduced. We further demonstrate THC-treatment elevated levels of cellular reactive oxygen species and markers of lipid damage. This was accompanied by evidence of increased mitochondrial fission. We also observed increased expression of cellular stress markers, HSP70 and HSP60, following exposure to THC. These effects were coincident with reduced mitochondrial respiratory function and a decrease in mitochondrial membrane potential. Taken together, our results suggest that THC can induce mitochondrial dysfunction and reduce trophoblast invasion; outcomes that have been previously linked to poor placentation. We also demonstrate that these changes in HTR8/SVneo biology may be variably mediated by cannabinoid receptors CB1 and CB2.
Collapse
Affiliation(s)
- O’Llenecia S. Walker
- grid.25073.330000 0004 1936 8227Graduate Program in Medical Sciences, Department of Pediatrics, McMaster University, HSC 4H7, Hamilton, ON L8S 4K1 Canada
| | - Harmeet Gurm
- grid.25073.330000 0004 1936 8227Graduate Program in Medical Sciences, Department of Pediatrics, McMaster University, HSC 4H7, Hamilton, ON L8S 4K1 Canada
| | - Reeti Sharma
- grid.25073.330000 0004 1936 8227 Department of Pediatrics, McMaster University, HSC 4H7, Hamilton, ON L8S 4K1 Canada
| | - Navkiran Verma
- grid.25073.330000 0004 1936 8227 Department of Pediatrics, McMaster University, HSC 4H7, Hamilton, ON L8S 4K1 Canada
| | - Linda L. May
- grid.25073.330000 0004 1936 8227 Department of Pediatrics, McMaster University, HSC 4H7, Hamilton, ON L8S 4K1 Canada
| | - Sandeep Raha
- grid.25073.330000 0004 1936 8227Graduate Program in Medical Sciences, Department of Pediatrics, McMaster University, HSC 4H7, Hamilton, ON L8S 4K1 Canada
| |
Collapse
|
33
|
Lu M, Sferruzzi-Perri AN. Placental mitochondrial function in response to gestational exposures. Placenta 2021; 104:124-137. [PMID: 33338764 DOI: 10.1016/j.placenta.2020.11.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 11/25/2020] [Accepted: 11/29/2020] [Indexed: 02/07/2023]
Abstract
Poor environmental conditions, including malnutrition, hypoxia and obesity in the mother increase the risk of pregnancy complications, such as pre-eclampsia and gestational diabetes mellitus, which impacts the lifelong health of the mother and her offspring. The placenta plays an important role in determining pregnancy outcome by acting as an exchange interface and endocrine hub to support fetal growth. Mitochondria are energy powerhouses of cells that fuel placental physiology throughout pregnancy, including placental development, substrate exchange and hormone secretion. They are responsive to environmental cues and changes in mitochondrial function may serve to mediate or mitigate the impacts of poor gestational environments on placental physiology and hence, the risks of pregnancy complications. Thus, a more integrated understanding about the role of placental mitochondria in orchestrating changes in relation to environmental conditions and pregnancy outcome is paramount. This review summarises the functions of mitochondria in the placenta and findings from humans and experimental animals that demonstrate how mitochondrial structure and function are altered in different gestational environments (namely complicated pregnancies and adverse environmental conditions). Together the available data suggest that mitochondria in the placenta play a major role in determining placental physiology, fetal growth and pregnancy outcome.
Collapse
Affiliation(s)
- Minhui Lu
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Amanda Nancy Sferruzzi-Perri
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK.
| |
Collapse
|
34
|
Fraichard C, Bonnet-Serrano F, Laguillier-Morizot C, Hebert-Schuster M, Lai-Kuen R, Sibiude J, Fournier T, Cohen M, Guibourdenche J. Protease Inhibitor Anti-HIV, Lopinavir, Impairs Placental Endocrine Function. Int J Mol Sci 2021; 22:E683. [PMID: 33445576 PMCID: PMC7827556 DOI: 10.3390/ijms22020683] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/06/2021] [Accepted: 01/08/2021] [Indexed: 12/12/2022] Open
Abstract
Protease Inhibitors (PI e.g., ritonavir (RTV) and lopinavir (LPV)) used to treat pregnant mothers infected by HIV induce prematurity and endocrine dysfunctions. The maintenance of pregnancy relies on placental hormone production (human Chorionic Gonadotrophin (hCG) and progesterone (P4)). Those functions are ensured by the villous trophoblast and are mainly regulated by the Unfolded Protein Response (UPR) pathway and mitochondria. We investigated, in vitro, if PI impair hCG and P4 production and the potential intracellular mechanisms involved. Term villous cytotrophoblast (VCT) were cultured with or without RTV or LPV from 6 to 48 h. VCT differentiation into syncytiotrophoblast (ST) was followed measuring hCG and P4 secretion. We evaluated the expression of P4 synthesis partners (Metastatic Lymph Node 64 (MLN64), cholesterol side-chain cleavage (P450SCC), Hydroxy-delta-5-Steroid Dehydrogenase and 3 Beta-and steroid delta-isomerase 1 (HSD3B1)), of mitochondrial pro-fusion factors (Mitofusin 2 (Mfn2), Optic Atrophy 1 (OPA1)) and of UPR factors (Glucose-Regulated Protein 78 (GRP78), Activating Transcription Factor 4 (ATF4), Activating Transcription Factor 6 (ATF6), spliced X-box Binding Protein 1 (sXBP1)). RTV had no significant effect on hCG and P4 secretion, whereas lopinavir significantly decreased both secretions. LPV also decreased P450SCC and HSD3B1 expression, whereas it increased Mfn2, GRP78 and sXBP1 expression in ST. RTV has no effect on the endocrine placenta. LPV impairs both villous trophoblast differentiation and P4 production. It is likely to act via mitochondrial fusion and UPR pathway activation. These trophoblastic alterations may end in decreased P4 levels in maternal circulation, inducing prematurity.
Collapse
Affiliation(s)
- Camille Fraichard
- INSERM UMR-S 1139, Faculté de Pharmacie, Université de Paris, 75006 Paris, France; (C.F.); (C.L.-M.); (T.F.)
| | | | - Christelle Laguillier-Morizot
- INSERM UMR-S 1139, Faculté de Pharmacie, Université de Paris, 75006 Paris, France; (C.F.); (C.L.-M.); (T.F.)
- Service d’Hormonologie, CHU Cochin, HUPC, AP-HP, 75014 Paris, France;
| | - Marylise Hebert-Schuster
- Service de Gynécologie-Obstétrique, Faculté de Médecine, Université de Genève, 1206 Genève, Suisse; (M.H.-S.); (M.C.)
| | - René Lai-Kuen
- INSERM UMS 025—CNRS UMS 3612, Faculté de Pharmacie, Université de Paris, 75006 Paris, France;
| | - Jeanne Sibiude
- Service de Gynécologie-Obstétrique, CHU Louis Mourier, HUPN, AP-HP, 92700 Colombes, France;
| | - Thierry Fournier
- INSERM UMR-S 1139, Faculté de Pharmacie, Université de Paris, 75006 Paris, France; (C.F.); (C.L.-M.); (T.F.)
| | - Marie Cohen
- Service de Gynécologie-Obstétrique, Faculté de Médecine, Université de Genève, 1206 Genève, Suisse; (M.H.-S.); (M.C.)
| | - Jean Guibourdenche
- INSERM UMR-S 1139, Faculté de Pharmacie, Université de Paris, 75006 Paris, France; (C.F.); (C.L.-M.); (T.F.)
- Service d’Hormonologie, CHU Cochin, HUPC, AP-HP, 75014 Paris, France;
| |
Collapse
|
35
|
Colson A, Sonveaux P, Debiève F, Sferruzzi-Perri AN. Adaptations of the human placenta to hypoxia: opportunities for interventions in fetal growth restriction. Hum Reprod Update 2020; 27:531-569. [PMID: 33377492 DOI: 10.1093/humupd/dmaa053] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 10/15/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The placenta is the functional interface between the mother and the fetus during pregnancy, and a critical determinant of fetal growth and life-long health. In the first trimester, it develops under a low-oxygen environment, which is essential for the conceptus who has little defense against reactive oxygen species produced during oxidative metabolism. However, failure of invasive trophoblasts to sufficiently remodel uterine arteries toward dilated vessels by the end of the first trimester can lead to reduced/intermittent blood flow, persistent hypoxia and oxidative stress in the placenta with consequences for fetal growth. Fetal growth restriction (FGR) is observed in ∼10% of pregnancies and is frequently seen in association with other pregnancy complications, such as preeclampsia (PE). FGR is one of the main challenges for obstetricians and pediatricians, as smaller fetuses have greater perinatal risks of morbidity and mortality and postnatal risks of neurodevelopmental and cardio-metabolic disorders. OBJECTIVE AND RATIONALE The aim of this review was to examine the importance of placental responses to changing oxygen environments during abnormal pregnancy in terms of cellular, molecular and functional changes in order to highlight new therapeutic pathways, and to pinpoint approaches aimed at enhancing oxygen supply and/or mitigating oxidative stress in the placenta as a mean of optimizing fetal growth. SEARCH METHODS An extensive online search of peer-reviewed articles using PubMed was performed with combinations of search terms including pregnancy, placenta, trophoblast, oxygen, hypoxia, high altitude, FGR and PE (last updated in May 2020). OUTCOMES Trophoblast differentiation and placental establishment are governed by oxygen availability/hypoxia in early pregnancy. The placental response to late gestational hypoxia includes changes in syncytialization, mitochondrial functions, endoplasmic reticulum stress, hormone production, nutrient handling and angiogenic factor secretion. The nature of these changes depends on the extent of hypoxia, with some responses appearing adaptive and others appearing detrimental to the placental support of fetal growth. Emerging approaches that aim to increase placental oxygen supply and/or reduce the impacts of excessive oxidative stress are promising for their potential to prevent/treat FGR. WIDER IMPLICATIONS There are many risks and challenges of intervening during pregnancy that must be considered. The establishment of human trophoblast stem cell lines and organoids will allow further mechanistic studies of the effects of hypoxia and may lead to advanced screening of drugs for use in pregnancies complicated by placental insufficiency/hypoxia. Since no treatments are currently available, a better understanding of placental adaptations to hypoxia would help to develop therapies or repurpose drugs to optimize placental function and fetal growth, with life-long benefits to human health.
Collapse
Affiliation(s)
- Arthur Colson
- Pole of Obstetrics, Institute of Experimental and Clinical Research (IREC), Université catholique de Louvain, Brussels, Belgium.,Pole of Pharmacology & Therapeutics, Institute of Experimental and Clinical Research (IREC), Université catholique de Louvain, Brussels, Belgium.,Department of Obstetrics, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Pierre Sonveaux
- Pole of Pharmacology & Therapeutics, Institute of Experimental and Clinical Research (IREC), Université catholique de Louvain, Brussels, Belgium
| | - Frédéric Debiève
- Pole of Obstetrics, Institute of Experimental and Clinical Research (IREC), Université catholique de Louvain, Brussels, Belgium.,Department of Obstetrics, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Amanda N Sferruzzi-Perri
- Department of Physiology, Development and Neuroscience, Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
| |
Collapse
|
36
|
Zhang M, He Q, Chen G, Li PA. Suppression of NLRP3 Inflammasome, Pyroptosis, and Cell Death by NIM811 in Rotenone-Exposed Cells as an in vitro Model of Parkinson's Disease. NEURODEGENER DIS 2020; 20:73-83. [PMID: 33176317 DOI: 10.1159/000511207] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 08/28/2020] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Parkinson's disease (PD) is characterized by the selective death of dopaminergic neurons in the substantia nigra. Recently, NLRP3 inflammasome and pyroptosis were found to be associated with PD. Cyclosporine A (CsA), an immunosuppressant, reduces neuronal death in PD. However, CsA could hardly pass through the blood-brain barrier (BBB) and high dose is associated with severe side effects and toxicity. N-methyl-4-isoleucine-cyclosporine (NIM811) is a CsA derivate that can pass through the BBB. However, little is known about its effect on PD. OBJECTIVE The objectives of this study were to explore the mechanism of rotenone-induced cell damage and to examine the protective effects of NIM811 on the neurotoxicity of a Parkinson-like in vitro model induced by rotenone. METHODS Murine hippocampal HT22 cells were cultured with the mitochondrial complex I inhibitor rotenone, a widely used pesticide that has been used for many years as a tool to induce a PD model in vitro and in vivo and proven to be reproducible. NIM811 was added to the culture media 3 h prior to the rotenone incubation. Cell viability was determined by resazurin assay, reactive oxygen species (ROS) production by dihydroethidine (DHE), and mitochondrial membrane potential by tetramethyl rhodamine methyl ester (TMRM). TUNEL and caspase-1 immunofluorescent double staining was used to detect pyroptosis. NLRP3, caspase-1, pro-caspase-1, GSDMD, and interleukin-18 (IL-18) were measured using Western blotting after 24 h of rotenone incubation. The reactivity of interleukin-1β (IL-1β) was determined by ELISA. RESULTS Our results demonstrated that rotenone caused more than 40% of cell death, increased ROS production, and reduced mitochondrial membrane potential, while NIM811 reversed these alterations. Immunofluorescent double staining showed that rotenone increased the percentage of caspase-1 and TUNEL double-labelled cells, an indication of pyroptosis, after 24 h of incubation. The protein expression of NLRP3, caspase-1, pro-caspase-1, GSDMD, IL-18, and IL-1β was significantly increased after 24 h of rotenone incubation. NIM811 suppressed rotenone-induced pyroptosis and downregulated the protein expression of NLRP3, caspase-1, pro-caspase-1, GSDMD, IL-1β, and IL-18. CONCLUSION These results provide evidence that rotenone activates the NLRP3 inflammomere and induces pyroptosis. NIM811 protects the cell from rotenone-induced damage and inhibits NLRP3 inflammasome and pyroptosis. NIM811 might serve as a potential therapeutic drug in the treatment of PD.
Collapse
Affiliation(s)
- Minghao Zhang
- Department of Pathophysiology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China.,Department of Pharmaceutical Sciences, Biomanufacturing Research Institute Biotechnology Enterprise (BRITE), College of Arts and Sciences, North Carolina Central University, Durham, North Carolina, USA.,Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Qingping He
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute Biotechnology Enterprise (BRITE), College of Arts and Sciences, North Carolina Central University, Durham, North Carolina, USA
| | - Guisheng Chen
- Department of Neurology, Neuroscience Center, General Hospital of Ningxia Medical University, Key Laboratory of Craniocerebral Diseases, Yinchuan, China
| | - P Andy Li
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute Biotechnology Enterprise (BRITE), College of Arts and Sciences, North Carolina Central University, Durham, North Carolina, USA,
| |
Collapse
|
37
|
Mitochondrial dysfunction in the fetoplacental unit in gestational diabetes mellitus. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165948. [PMID: 32866635 DOI: 10.1016/j.bbadis.2020.165948] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 08/17/2020] [Accepted: 08/20/2020] [Indexed: 12/12/2022]
Abstract
Gestational diabetes mellitus (GDM) is a disease of pregnancy that is associated with d-glucose intolerance and foeto-placental vascular dysfunction. GMD causes mitochondrial dysfunction in the placental endothelium and trophoblast. Additionally, GDM is associated with reduced placental oxidative phosphorylation due to diminished activity of the mitochondrial F0F1-ATP synthase (complex V). This phenomenon may result from a higher generation of reactive superoxide anion and nitric oxide. Placental mitochondrial biogenesis and mitophagy work in concert to maintain cell homeostasis and are vital mechanisms securing the efficient generation of ATP, whose demand is higher in pregnancy, ensuring foetal growth and development. Additional factors disturbing placental ATP synthase activity in GDM include pre-gestational maternal obesity or overweight, intracellular pH, miRNAs, fatty acid oxidation, and foetal (and 'placental') sex. GDM is also associated with maternal and foetal hyperinsulinaemia, altered circulating levels of adiponectin and leptin, and the accumulation of extracellular adenosine. Here, we reviewed the potential interplay between these molecules or metabolic conditions on the mechanisms of mitochondrial dysfunction in the foeto-placental unit in GDM pregnancies.
Collapse
|
38
|
Walker OS, Ragos R, Gurm H, Lapierre M, May LL, Raha S. Delta-9-tetrahydrocannabinol disrupts mitochondrial function and attenuates syncytialization in human placental BeWo cells. Physiol Rep 2020; 8:e14476. [PMID: 32628362 PMCID: PMC7336740 DOI: 10.14814/phy2.14476] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 05/11/2020] [Accepted: 05/12/2020] [Indexed: 12/20/2022] Open
Abstract
The psychoactive component in cannabis, delta-9-tetrahydrocannabinol, can restrict fetal growth and development. Delta-9-tetrahydrocannabinol has been shown to negatively impact cellular proliferation and target organelles like the mitochondria resulting in reduced cellular respiration. In the placenta, mitochondrial dysfunction leading to oxidative stress prevents proper placental development and function. A key element of placental development is the proliferation and fusion of cytotrophoblasts to form the syncytium that comprises the materno-fetal interface. The impact of delta-9-tetrahydrocannabinol on this process is not well understood. To elucidate the nature of the mitochondrial dysfunction and its consequences on trophoblast fusion, we treated undifferentiated and differentiated BeWo human trophoblast cells, with 20 µM delta-9-tetrahydrocannabinol for 48 hr. At this concentration, delta-9-tetrahydrocannabinol on BeWo cells reduced the expression of markers involved in syncytialization and mitochondrial dynamics, but had no effect on cell viability. Delta-9-tetrahydrocannabinol significantly attenuated the process of syncytialization and induced oxidative stress responses in BeWo cells. Importantly, delta-9-tetrahydrocannabinol also caused a reduction in the secretion of human chorionic gonadotropin and the production of human placental lactogen and insulin growth factor 2, three hormones known to be important in facilitating fetal growth. Furthermore, we also demonstrate that delta-9-tetrahydrocannabinol attenuated mitochondrial respiration, depleted adenosine triphosphate, and reduced mitochondrial membrane potential. These changes were also associated with an increase in cellular reactive oxygen species, and the expression of stress responsive chaperones, HSP60 and HSP70. These findings have important implications for understanding the role of delta-9-tetrahydrocannabinol-induced mitochondrial injury and the role this might play in compromising human pregnancies.
Collapse
Affiliation(s)
- O’Llenecia S. Walker
- Department of PediatricsMcMaster UniversityHamiltonONCanada
- The Graduate Program in Medical SciencesMcMaster UniversityHamiltonONCanada
| | | | - Harmeet Gurm
- Department of PediatricsMcMaster UniversityHamiltonONCanada
| | | | - Linda L. May
- Department of PediatricsMcMaster UniversityHamiltonONCanada
| | - Sandeep Raha
- Department of PediatricsMcMaster UniversityHamiltonONCanada
- The Graduate Program in Medical SciencesMcMaster UniversityHamiltonONCanada
| |
Collapse
|