1
|
Mizobuchi H, Yamagishi J, Sanjoba C, Goto Y. Vertical transmission of Leishmania donovani with placental degeneration in the pregnant mouse model of visceral leishmaniasis. PLoS Negl Trop Dis 2025; 19:e0012650. [PMID: 40489555 DOI: 10.1371/journal.pntd.0012650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 05/27/2025] [Indexed: 06/11/2025] Open
Abstract
Visceral leishmaniasis (VL) is a zoonotic disease caused by infection of macrophages by Leishmania donovani or L. infantum, and exhibits symptoms such as fever, anemia, and hepatosplenomegaly. VL during pregnancy has been reported to have negative effects such as miscarriage and vertical infection, but the mechanism is not clear. Here, we aimed to establish a pregnant VL mouse model and elucidate its immunopathology. Female BALB/c mice mated 6 months after L. donovani infection showed reduced pregnancy rates. The fetus was removed by caesarean section on the 18th day of pregnancy, and Leishmania parasite DNA was detected from fetal spleens and livers. As a result, the PCR positive rate was 68.9% (71/103 fetus), and vertical transmission was suspected in 66.7% of infected mothers (12/18 dams). Immunohistochemistry in the fetal livers detected cells positive for the Leishmania antigen, kinetoplastid membrane protein 11 (KMP11). In addition, pathological analysis of the VL placenta revealed trophoblast cell atrophy and vasodilation accompanied by CD3+ cell infiltration in the infected group. On the other hand, few KMP11+ cells were observed in the placenta of the infected group. Furthermore, RNA-Seq analysis revealed that IFN signal activation and cellular immune suppression were induced in the placenta of the infected group. These results suggest that VL in pregnancy induces suppression of placental cellular immunity through IFN and collapse of the placental barrier through trophoblast degeneration, leading to vertical transmission. Because few infected macrophages were observed in the placenta, it is possible that free Leishmania parasites in the blood contribute to transmission across the placenta.
Collapse
Affiliation(s)
- Haruka Mizobuchi
- Laboratory of Molecular Immunology, Department of Animal Resource Sciences, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Junya Yamagishi
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
- Division of Collaboration and Education, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Chizu Sanjoba
- Laboratory of Molecular Immunology, Department of Animal Resource Sciences, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Yasuyuki Goto
- Laboratory of Molecular Immunology, Department of Animal Resource Sciences, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
2
|
Gu Y, Xu J, Gu X, Lu X, Lewis DF, Scott RS, Zoorob D, Wang Y. Increased DNA damage/repair response in placentas from women infected with COVID-19 in pregnancy. Placenta 2025; 167:152-160. [PMID: 40393216 DOI: 10.1016/j.placenta.2025.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 04/01/2025] [Accepted: 05/12/2025] [Indexed: 05/22/2025]
Abstract
INTRODUCTION SARS-CoV-2 may induce DNA damage in infected cells. The objective of the study is to determine if maternal COVID-19 infection can induce cellular DNA damage in the placenta. We examined phospho-Histone H2AX (pH2AX) foci-positive cells, MRE11 expression, and telomere length (markers of DNA damage/repair) in placental specimens from women with or without COVID-19 infection during pregnancy. METHODS A total of 61 placental formalin-fixed paraffin embedded (FFPE) tissue specimens were used in the study, 33 in the COVID-19 group and 28 in the control group. Expression of pH2AX (Ser-139) and MRE11 was determined by immunostaining. Telomere length was assessed using the relative Human Telomere Length Quantification qPCR Assay Kit. Maternal demographic data was obtained via electronic medical record review. Data were analyzed by unpaired t-test or Chi-square test. A p level <0.05 was set statistically significant. RESULTS pH2AX foci-positive cells in the placentas were significantly higher in the COVID group than in the control group, p < 0.01; Relative expression of MRE11 was significantly increased in the COVID group (15.68 ± 1.57) than in the control group (5.74 ± 1.21), p < 0.01. In contrast, relative telomere length was significantly shorter in the COVID group (0.839 ± 0.123) than in the control group (1.652 ± 0.285), p < 0.05. The correlation of telomere length with placental weight and gestational age at delivery and the correlation of placental/newborn ratio were also analyzed. CONCLUSIONS Both pH2AX and MRE11 are biomarkers of DNA double-strand breaks. Telomeres are targets of a persistent DNA damage response, and reduced telomere length is an indicator of cellular senescence and aging. Our findings of increased pH2AX foci-positive cells and MRE11 expression and reduced telomere length in placental cells from women infected with COVID-19 in pregnancy provide plausible evidence that increased placental cellular DNA damage could be associated with maternal COVID-19 infection in pregnancy.
Collapse
Affiliation(s)
- Yang Gu
- Departments of Obstetrics and Gynecology, Louisiana State University Health Sciences Center - Shreveport, Shreveport, LA, 71103, USA.
| | - Jie Xu
- Departments of Obstetrics and Gynecology, Louisiana State University Health Sciences Center - Shreveport, Shreveport, LA, 71103, USA.
| | - Xin Gu
- Department of Pathology,Louisiana State University Health Sciences Center - Shreveport, Shreveport, LA, 71103, USA.
| | - Xiaohong Lu
- Department of Pharmacology, Toxicology and Neuroscience, Louisiana State University Health Sciences Center - Shreveport, Shreveport, LA,71103, USA.
| | - David F Lewis
- Departments of Obstetrics and Gynecology, Louisiana State University Health Sciences Center - Shreveport, Shreveport, LA, 71103, USA.
| | - Rona S Scott
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center - Shreveport, Shreveport, LA, 71103, USA.
| | - Dani Zoorob
- Departments of Obstetrics and Gynecology, Louisiana State University Health Sciences Center - Shreveport, Shreveport, LA, 71103, USA.
| | - Yuping Wang
- Departments of Obstetrics and Gynecology, Louisiana State University Health Sciences Center - Shreveport, Shreveport, LA, 71103, USA.
| |
Collapse
|
3
|
Pallathadka H, Khaleel AQ, Hjazi A, Kumar A, Aloraibi F, Kadhum WR, Pramanik A, Hamzah HF, Mohammed SK, Mustafa YF. Decoding immune tolerance in infertility: Exploring immune pathways and non-coding RNAs as pioneering biomarkers and therapeutic targets. Hum Immunol 2025; 86:111264. [PMID: 39978249 DOI: 10.1016/j.humimm.2025.111264] [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: 08/14/2024] [Revised: 01/23/2025] [Accepted: 02/12/2025] [Indexed: 02/22/2025]
Abstract
Infertility, impacting a significant number of couples, is characterized by the failure to conceive after one year of consistent, unprotected sexual intercourse. It is multifactorial, with etiological contributors including ovulatory dysfunction, male reproductive anomalies, and tubal patency issues. Approximately 15% of infertility cases are classified as "unexplained," highlighting the complexity of this condition. Lifestyle determinants such as obesity and smoking further complicate reproductive outcomes, while infertility can also indicate underlying chronic health conditions. A specialized category, immune infertility, arises from a breakdown of immunological tolerance, an essential aspect for conception and the maintenance of pregnancy. The role of various immunological components, including immune cells, cytokines, chemokines, factors like HLA-G, etc., is pivotal in this context. Moreover, non-coding RNAs (ncRNAs) have emerged as critical regulators of immune tolerance within the reproductive axis. This review synthesizes the complex immunological pathways vital for successful implantation and the early stages of pregnancy alongside the regulatory roles of ncRNAs in these processes. Offering an integrated view of molecular and immunological interactions associated with infertility seeks to enhance our understanding of potential strategies to facilitate successful conception and sustain early pregnancy.
Collapse
Affiliation(s)
| | - Abdulrahman Qais Khaleel
- Department of Medical Instruments Engineering, Al-Maarif University College, Al Anbar, 31001, Iraq.
| | - Ahmed Hjazi
- Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia.
| | - Ashwani Kumar
- Department of Life Sciences, School of Sciences, Jain (Deemed-to-be) University, Bengaluru, Karnataka 560069, India; Department of Pharmacy, Vivekananda Global University, Jaipur, Rajasthan 303012, India.
| | - Farah Aloraibi
- Department of Density, Al-Manara College for Medical Sciences, Maysan, Iraq.
| | - Wesam R Kadhum
- Department of Pharmacy, Kut University College, Kut 52001, Wasit, Iraq; Advanced Research Center, Kut University College, Kut 52001, Wasit, Iraq.
| | - Atreyi Pramanik
- School of Applied and Life Sciences, Division of Research and Innovation, Uttaranchal University, Dehradun, Uttarakhand, India.
| | - Hamza Fadhel Hamzah
- Department of Medical Laboratories Technology, Al-Nisour University College, Nisour Seq. Karkh, Baghdad, Iraq.
| | - Saad Khudhur Mohammed
- College of Pharmacy, National University of Science and Technology, Dhi Qar 64001, Iraq.
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul 41001, Iraq.
| |
Collapse
|
4
|
Mouillet JF, Ouyang Y, Sadovsky E, Kothnadan VK, Sorenson HL, Badeau LJ, Sarkar SN, Chu T, Sorkin A, Sadovsky Y. The Chromosome 19 miRNA Cluster Guards Trophoblasts Against Overacting Innate Immunity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.03.647038. [PMID: 40236003 PMCID: PMC11996509 DOI: 10.1101/2025.04.03.647038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
To maintain pregnancy health, the human placenta delicately balances protection of the developing fetus from invading pathogens with suppression of excessive inflammation that could lead to fetal and neonatal autoimmune disorders. Previous research, including our own, has shown that small RNA products of the Chromosome 19 MicroRNA Cluster (C19MC) promote viral resistance in non-trophoblastic cells. However, the role of C19MC products in placental trophoblasts remained unclear. Here, we analyzed chromatin accessibility in the C19MC enhancer and identified a previously unknown regulatory domain. Deletion of this domain silenced the expression of C19MC microRNA and Alu elements in trophoblasts. This silencing unexpectedly led to marked activation of cellular innate immune response and strikingly increased Toll-like receptor 3 (TLR3)-mediated sensitivity to poly(I:C), a viral RNA mimic. Our data suggest that C19MC non-coding RNAs interfere with endosomal TLR3 activation in trophoblasts, highlighting a previously unrecognized mechanism for hindrance of excessive innate immune activation.
Collapse
|
5
|
Wang L, Mu Q, Zhang W, Zheng W, Zhu X, Yu Y, Wang Y, Xu W, Lu Z, Han X. Placental targeted drug delivery: a review of recent progress. NANOSCALE 2025; 17:8316-8335. [PMID: 40070242 DOI: 10.1039/d4nr05338a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2025]
Abstract
The placenta plays a crucial role in mediating nutrient and gas exchange between the mother and fetus during pregnancy. Targeting therapeutic agents to the placenta presents significant opportunities for treating placental disorders and enhancing fetal outcomes. However, the unique structural complexity and selective permeability of the placenta pose substantial challenges for effective drug delivery. This review provides a comprehensive overview of current strategies for placental targeting, including lipid nanoparticle (LNP) delivery systems, targeted peptide modifications, specific antibody targeting of placental receptors, and the use of viral vectors. We critically analyze the advantages and limitations of each approach, emphasizing recent advancements in enhancing targeting specificity and delivery efficiency. By consolidating the latest research developments, this review aims to foster further innovation in placental drug delivery methods and contribute significantly to the advancement of therapeutic strategies for placental disorders, ultimately improving outcomes for both mother and fetus.
Collapse
Affiliation(s)
- Linjian Wang
- Department of Obstetrics, Haining Maternal and Child Health Hospital, Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, No. 309, East Shuiyueting Road, Xiashi Street, Haining, Zhejiang, 314400, China
| | - Qiuqiu Mu
- Third Affliated Hospital of Wenzhou Medical University, WanSong Road No. 108, Ruian, Wenzhou, Zhejiang, 325200, China
| | - Wenjing Zhang
- Department of Obstetrics, Women's Hospital, Zhejiang University School of Medicine, Xueshi Road No. 1, Hangzhou, Zhejiang, 310006, China.
| | - Weiqian Zheng
- Department of Obstetrics, Women's Hospital, Zhejiang University School of Medicine, Xueshi Road No. 1, Hangzhou, Zhejiang, 310006, China.
| | - Xiaojun Zhu
- Department of Obstetrics, Women's Hospital, Zhejiang University School of Medicine, Xueshi Road No. 1, Hangzhou, Zhejiang, 310006, China.
| | - Ying Yu
- Department of Obstetrics, Women's Hospital, Zhejiang University School of Medicine, Xueshi Road No. 1, Hangzhou, Zhejiang, 310006, China.
| | - YuPeng Wang
- Department of Obstetrics, Women's Hospital, Zhejiang University School of Medicine, Xueshi Road No. 1, Hangzhou, Zhejiang, 310006, China.
| | - Wenli Xu
- Department of Obstetrics, Haining Maternal and Child Health Hospital, Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, No. 309, East Shuiyueting Road, Xiashi Street, Haining, Zhejiang, 314400, China
| | - Zhimin Lu
- Department of Obstetrics, Women's Hospital, Zhejiang University School of Medicine, Xueshi Road No. 1, Hangzhou, Zhejiang, 310006, China.
| | - Xiujun Han
- Department of Obstetrics, Women's Hospital, Zhejiang University School of Medicine, Xueshi Road No. 1, Hangzhou, Zhejiang, 310006, China.
| |
Collapse
|
6
|
Ding X, Zhou Y, Qiu X, Xu X, Hu X, Qin J, Chen Y, Zhang M, Ke J, Liu Z, Zhou Y, Ding C, Shen N, Tian Z, Fu B, Wei H. RSAD2: A pathogenic interferon-stimulated gene at the maternal-fetal interface of patients with systemic lupus erythematosus. Cell Rep Med 2025; 6:101974. [PMID: 39983716 PMCID: PMC11970327 DOI: 10.1016/j.xcrm.2025.101974] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 11/21/2024] [Accepted: 01/27/2025] [Indexed: 02/23/2025]
Abstract
Pregnancy disorders in patients with autoimmune diseases or viral infections are often associated with an excessive response of type I interferons. We identify radical S-adenosyl methionine domain containing 2 (RSAD2) as a pathogenic interferon-stimulated gene (ISG) associated with pregnancy complications in systemic lupus erythematosus (SLE). The increased expression of RSAD2 mainly occurs in macrophages and structural cell populations at the maternal-fetal interface of pregnant patients with SLE. The elevation of RSAD2 leads to the accumulation of diacylglycerol lipids in the placenta, impairing the necessary vascular development for the fetus. Depletion of Rsad2 in pregnant mice models exposed to type I interferon inducers significantly reduces lipid accumulation, vascular injury, and embryo development disorders. An RSAD2 inhibitor, L-chicoric acid (LCA), alleviates lipid accumulation and vascular damage, improving pregnancy outcomes in SLE-induced and spontaneous mouse models. This study proposes the potential of targeting RSAD2 to improve pregnancy outcomes in individuals with heightened type I interferon response.
Collapse
Affiliation(s)
- Xiaoyu Ding
- The National Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China; Institute of Immunology, University of Science and Technology of China, Hefei, China
| | - Yonggang Zhou
- The National Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China; Institute of Immunology, University of Science and Technology of China, Hefei, China; Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China.
| | - Xiaofeng Qiu
- The National Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China; Institute of Immunology, University of Science and Technology of China, Hefei, China
| | - Xiuxiu Xu
- The National Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China; Institute of Immunology, University of Science and Technology of China, Hefei, China
| | - Xinyu Hu
- The National Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China; Institute of Immunology, University of Science and Technology of China, Hefei, China
| | - Jingkun Qin
- The National Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China; Institute of Immunology, University of Science and Technology of China, Hefei, China
| | - Yulan Chen
- Department of Rheumatology and Immunology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, China
| | - Min Zhang
- Department of Rheumatology and Immunology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Jieqi Ke
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Zhenbang Liu
- The National Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - Ying Zhou
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Chen Ding
- Institute of Biomedical Sciences, State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Nan Shen
- Shanghai Institute of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200001, China
| | - Zhigang Tian
- The National Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China; Institute of Immunology, University of Science and Technology of China, Hefei, China
| | - Binqing Fu
- The National Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China; Institute of Immunology, University of Science and Technology of China, Hefei, China; Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China.
| | - Haiming Wei
- The National Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China; Institute of Immunology, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
7
|
Gerovasili E, Sarantaki A, Bothou A, Deltsidou A, Dimitrakopoulou A, Diamanti A. The role of vitamin D deficiency in placental dysfunction: A systematic review. Metabol Open 2025; 25:100350. [PMID: 40034802 PMCID: PMC11874864 DOI: 10.1016/j.metop.2025.100350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 01/24/2025] [Accepted: 01/24/2025] [Indexed: 03/05/2025] Open
Abstract
Introduction Vitamin D plays a critical role in pregnancy, supporting placental function via angiogenesis, immune regulation, and nutrient transport. Deficiency in vitamin D during gestation is associated with complications such as preeclampsia, intrauterine growth restriction (IUGR), and preterm birth. However, the mechanisms linking vitamin D deficiency to placental dysfunction remain inadequately understood, highlighting the need for systematic evaluation. Methods A systematic review was conducted in adherence to Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines, with searches in PubMed, Scopus, and Web of Science for studies published within the last 20 years. Inclusion criteria targeted human studies examining the association between vitamin D and placental function, including randomized controlled trials, cohort studies, and case-control studies. A total of 10 studies were included following rigorous screening and quality assessment. Results Findings from human studies indicate that maternal vitamin D deficiency significantly impairs placental function by reducing vascular integrity, downregulating nutrient transporters, and promoting inflammation. Mechanistic evidence highlights decreased expression of vascular endothelial growth factor (VEGF) and increased inflammatory cytokines in vitamin D-deficient pregnancies. Supplementation with active vitamin D [1α,25(OH)2D3] mitigated these adverse effects, restoring placental growth, improving nutrient transport, and reducing inflammation. Notably, population-specific differences and sex-specific responses to vitamin D sufficiency were observed. Conclusions Vitamin D is essential for optimal placental function and pregnancy outcomes. This review underscores the need for standardized supplementation protocols and further research into long-term and population-specific effects of vitamin D. Addressing these gaps can inform targeted interventions to reduce pregnancy complications and improve maternal-fetal health.
Collapse
Affiliation(s)
- Eleni Gerovasili
- Department of Midwifery, Faculty of Health and Caring Sciences, University of West Attica, Egaleo, Greece
| | - Antigoni Sarantaki
- Department of Midwifery, Faculty of Health and Caring Sciences, University of West Attica, Egaleo, Greece
| | - Anastasia Bothou
- Department of Midwifery, Faculty of Health and Caring Sciences, University of West Attica, Egaleo, Greece
| | - Anna Deltsidou
- Department of Midwifery, Faculty of Health and Caring Sciences, University of West Attica, Egaleo, Greece
| | - Aikaterini Dimitrakopoulou
- Department of Midwifery, Faculty of Health and Caring Sciences, University of West Attica, Egaleo, Greece
| | - Athina Diamanti
- Department of Midwifery, Faculty of Health and Caring Sciences, University of West Attica, Egaleo, Greece
| |
Collapse
|
8
|
Freedman AN, Hartwell H, Fry R. Using transcriptomic signatures to elucidate individual and mixture effects of inorganic arsenic and manganese in human placental trophoblast HTR-8/SVneo cells. Toxicol Sci 2025; 203:216-226. [PMID: 39836092 PMCID: PMC11775420 DOI: 10.1093/toxsci/kfae147] [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: 01/22/2025] Open
Abstract
Prenatal exposure to the toxic metal inorganic arsenic (iAs) is associated with adverse pregnancy and fetal growth outcomes. These adverse outcomes are tied to physiological disruptions in the placenta. Although iAs co-occurs in the environment with other metals such as manganese (Mn), there is a gap in the knowledge of the effects of metal mixtures on the placenta. To address this, we exposed human placental trophoblast cells to iAs, Mn, and an iAs-Mn mixture at 3 concentrations and evaluated transcriptome-wide gene expression and placental migration. We hypothesized that co-exposure to iAs-Mn in a mixture would result in a synergistic/enhanced transcriptomic effect compared to either metal alone. We also anticipated that genes involved in inflammatory or immune-related pathways would be differentially expressed in relation to the mixture compared to single-metals. The results highlight that iAs exposure alone had a stronger genomic response than Mn exposure, with 2-fold the number of differentially expressed genes (DEGs). When analyzing DEGs present across all concentrations of study, the iAs-Mn mixture resulted in the greatest number of DEGs. The results highlight that iAs exposure alone influences the expression of toll-like receptor-initiated response pathways including Triggering Receptor Expressed on Myeloid Cells-1. Exposure to Mn alone influenced the expression of Nicotinamide adenine dinucleotide biosynthesis pathways. In contrast, exposure to the iAs-Mn mixtures resulted in altered expression of inflammatory and immune response-related pathways, including the Nuclear factor erythroid 2-related factor 2 (NRF2)-mediated oxidative stress response pathway. Migration was unaffected by iAs, Mn, or the iAs-Mn mixture. These findings provide novel toxicogenomic insights into iAs- and Mn-induced placental transcriptomic dysregulations at environmentally relevant concentrations, with implications that in utero exposure to metal mixtures can influence inflammatory and immune pathways within the placenta.
Collapse
Affiliation(s)
- Anastasia N Freedman
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC 27599, United States
- Institute for Environmental Health Solutions, University of North Carolina, Chapel Hill, NC 27599, United States
| | - Hadley Hartwell
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC 27599, United States
- Institute for Environmental Health Solutions, University of North Carolina, Chapel Hill, NC 27599, United States
| | - Rebecca Fry
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC 27599, United States
- Institute for Environmental Health Solutions, University of North Carolina, Chapel Hill, NC 27599, United States
| |
Collapse
|
9
|
Christian C, Ana L, Alejandro FM, Jesús GM, Sebastian A, Gabriela C, Sebastián A, Christian G, Juan Diego M, Marioly M, Ulrike K. Congenital Chagas disease: The importance of Trypanosoma cruzi-placenta interactions. Placenta 2025:S0143-4004(25)00011-6. [PMID: 39855996 DOI: 10.1016/j.placenta.2025.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 12/19/2024] [Accepted: 01/06/2025] [Indexed: 01/27/2025]
Affiliation(s)
- Castillo Christian
- Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Liempi Ana
- Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | | | - Guerrero-Muñoz Jesús
- Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Araneda Sebastian
- Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile; Departamento de Patología y Medicina Oral, Facultad de Odontología, Universidad de Chile, Santiago, Chile
| | - Cáceres Gabriela
- Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Alfaro Sebastián
- Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Gallardo Christian
- Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Maya Juan Diego
- Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Müller Marioly
- Departamento de Tecnología Médica Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Kemmerling Ulrike
- Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile.
| |
Collapse
|
10
|
Guma E, Chakravarty MM. Immune Alterations in the Intrauterine Environment Shape Offspring Brain Development in a Sex-Specific Manner. Biol Psychiatry 2025; 97:12-27. [PMID: 38679357 PMCID: PMC11511788 DOI: 10.1016/j.biopsych.2024.04.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 03/20/2024] [Accepted: 04/17/2024] [Indexed: 05/01/2024]
Abstract
Exposure to immune dysregulation in utero or in early life has been shown to increase risk for neuropsychiatric illness. The sources of inflammation can be varied, including acute exposures due to maternal infection or acute stress, or persistent exposures due to chronic stress, obesity, malnutrition, or autoimmune diseases. These exposures may cause subtle alteration in brain development, structure, and function that can become progressively magnified across the lifespan, potentially increasing the likelihood of developing a neuropsychiatric conditions. There is some evidence that males are more susceptible to early-life inflammatory challenges than females. In this review, we discuss the various sources of in utero or early-life immune alteration and the known effects on fetal development with a sex-specific lens. To do so, we leveraged neuroimaging, behavioral, cellular, and neurochemical findings. Gaining clarity about how the intrauterine environment affects offspring development is critically important for informing preventive and early intervention measures that may buffer against the effects of these early-life risk factors.
Collapse
Affiliation(s)
- Elisa Guma
- Section on Developmental Neurogenomics, Human Genetics Branch, National Institute of Mental Health, Bethesda, Maryland; Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts.
| | - M Mallar Chakravarty
- Computational Brain Anatomy Laboratory, Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec, Canada; Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec, Canada; Department of Psychiatry, McGill University, Montreal, Quebec, Canada; Department of Biomedical Engineering, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
11
|
Chen H, Shao LZ, Wang YX, Han ZJ, Wang YH, Li X, Chen JY, Liu TH. Causal Relationships Between Leukocyte Subsets and Adverse Fetal Outcomes: A Mendelian Randomization Study. Mediators Inflamm 2024; 2024:6349687. [PMID: 39748887 PMCID: PMC11695084 DOI: 10.1155/mi/6349687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 07/28/2024] [Accepted: 11/07/2024] [Indexed: 01/04/2025] Open
Abstract
Background: The tolerance and dynamic regulation of the maternal immune system during pregnancy are pivotal for ensuring fetal health. Immune cell subsets play a complex and crucial role in this process, closely linked to the neonatal health status. Despite recognizing the significance of dysregulation in the quantity and activity of immune cells in neonatal disease occurrence, their specific roles remain elusive, resulting in a dearth of clinically viable interventions for immune-mediated neonatal diseases. Materials and Methods: Employing two-sample Mendelian randomization (MR) methodology, this study systematically investigated 446 leukocyte features (N = 500,675), including leukocyte subsets, absolute cell (AC) counts, and morphological parameters (MP) and their correlation with seven adverse fetal outcomes (N = 1,100,458), encompassing fetal growth restriction (FGR), preterm birth (PTB), neonatal jaundice (NNJ), digestive system disorders of fetus and newborn (DSDFN), hemorrhagic and hematological disorders of fetus and newborn (HDFN), respiratory distress of newborn (RDN), and transitory disorders of metabolism specific to fetus and newborn (TDMSFN). Results: The results unveiled significant causal relationships between 301 leukocyte subsets and these seven adverse fetal outcomes, with 259, 245, 15, 44, 11, 32, and 68 pairs of notable associations for each adverse outcome, respectively. Furthermore, the study highlighted potential pathogenic mechanisms underlying the mutual influence among neonatal diseases. MR results indicated FGR as a robustly correlated risk factor for PTB and NNJ and showed a reciprocal causal relationship between NNJ and FGR. PTB exhibited a positive correlation with HDFN. Conclusions: This study provided profound insights into the intricate regulatory mechanisms of leukocyte subsets in neonatal diseases, paving the way for new avenues in the diagnosis and treatment of associated disorders.
Collapse
Affiliation(s)
- Hong Chen
- Department of Bioinformatics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, China
- Joint International Research Laboratory of Reproduction and Development, Chongqing Medical University, Chongqing 400016, China
| | - Li-Zhen Shao
- Department of Bioinformatics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, China
- Joint International Research Laboratory of Reproduction and Development, Chongqing Medical University, Chongqing 400016, China
| | - Ying-Xiong Wang
- Department of Bioinformatics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, China
- Joint International Research Laboratory of Reproduction and Development, Chongqing Medical University, Chongqing 400016, China
| | - Zhi-Jie Han
- Department of Bioinformatics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Yong-Heng Wang
- Department of Bioinformatics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, China
- Joint International Research Laboratory of Reproduction and Development, Chongqing Medical University, Chongqing 400016, China
| | - Xia Li
- Department of Bioinformatics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, China
- Joint International Research Laboratory of Reproduction and Development, Chongqing Medical University, Chongqing 400016, China
| | - Jing-Yu Chen
- Department of Ultrasound, Children's Hospital of Chongqing Medical University, 136 Zhongshan 2nd Road, Chongqing 400014, China
| | - Tai-Hang Liu
- Department of Bioinformatics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, China
- Joint International Research Laboratory of Reproduction and Development, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
12
|
Brooker IA, Fisher JJ, Sutherland JM, Pringle KG. Understanding the impact of placental oxidative and nitrative stress in pregnancies complicated by fetal growth restriction. Placenta 2024; 158:318-328. [PMID: 39577026 DOI: 10.1016/j.placenta.2024.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 11/12/2024] [Accepted: 11/13/2024] [Indexed: 11/24/2024]
Abstract
Fetal growth restriction (FGR) impacts approximately 10 % of all pregnancies worldwide and is associated with major adverse effects on fetal health in both the short- and long-term [1]. FGR most commonly arises as a result of impaired placentation, occurring in up to 60 % of cases in developed countries [2]. This narrative review outlines the impact of defective placentation on the placenta, focusing on redox imbalance, how this leads to placental oxidative and nitrative stress, and the implications of these stressors on placental nutrient transfer, premature replicative senescence, and trophoblast cell death. Furthermore, this review highlights the pivotal role of antioxidants in protecting against oxidative and nitrative damage by reducing the burden of reactive species. We explore how targeting antioxidants in pregnancy provides a promising strategy for preventing or treating FGR, to ultimately reduce the devastating burden of FGR on infant health.
Collapse
Affiliation(s)
- India A Brooker
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia; Women's Health Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Joshua J Fisher
- School of Medicine and Public Health, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia; Mothers and Babies Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Jessie M Sutherland
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia; Women's Health Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Kirsty G Pringle
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia; Women's Health Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.
| |
Collapse
|
13
|
Aljani B, Garbe AI, Sedlmeier EM, Lickert R, Rost F, Ziegler AG, Bonifacio E, Eugster A. Gene expression profiles in placenta and their association with anesthesia, delivery mode and maternal diabetes. Placenta 2024; 158:126-135. [PMID: 39427562 DOI: 10.1016/j.placenta.2024.10.008] [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: 04/15/2024] [Revised: 09/11/2024] [Accepted: 10/13/2024] [Indexed: 10/22/2024]
Abstract
INTRODUCTION Fetal development is dependent on placenta and affected by multiple factors including maternal diabetes. Here we aimed to identify maternal diabetes-associated changes in placentas and analyzed placental gene expression to understand its modulation by maternal diabetes and birth mode. METHODS Placental RNAseq transcriptome analyses were performed on maternally-derived decidua and fetal-derived villous tissue from pregnancies of mothers with type 1 diabetes (n = 14), gestational diabetes (n = 6) and without diabetes (n = 14). Information on delivery mode and anesthesia were included as covariables. Analyses were performed separately for decidua and fetal tissues and adjusted for sex. RESULTS Substantial placenta gene expression variation was associated with factors other than maternal diabetes, including site, sex, anesthesia type and delivery mode. Two dominant gene expression clusters aligned to anesthesia and delivery mode were observed for decidua and villous tissue. Upregulation of genes within pathways related to organ morphogenesis and downregulation of immune response to steroid- and hypoxia pathway genes was characteristic of placentas from primary cesarean section deliveries with spinal anesthesia. Opposite profiles were observed for placentas from secondary cesarean and epidural anesthesia deliveries. Placentas from vaginal delivery had intermediate gene expression profiles. More subtle changes were associated with maternal diabetes: upregulation of ribosome activity, down-regulation of maternally-derived decidua chemokine signaling pathways and for gestational diabetes, alteration in hypoxia response genes. DISCUSSION The findings reveal suppression of immune pathways and upregulation of ribosome activity in the placenta by maternal diabetes highlighting the importance of confounding factors when examining cell and tissue expression profiles. Further studies should determine whether the observed gene expression differences are related to underlying causes for cesarean section deliveries.
Collapse
Affiliation(s)
- Bassam Aljani
- Center for Regenerative Therapies Dresden, Faculty of Medicine, TU Dresden, Dresden, Germany.
| | - Annette I Garbe
- Center for Regenerative Therapies Dresden, Faculty of Medicine, TU Dresden, Dresden, Germany.
| | - Eva-Maria Sedlmeier
- Institute of Diabetes Research, Helmholtz Munich, German Center for Environmental Health, Munich, Germany.
| | - Ramona Lickert
- Institute of Diabetes Research, Helmholtz Munich, German Center for Environmental Health, Munich, Germany.
| | - Fabian Rost
- Dresden-CONCEPT Genome Center, Center for Molecular and Cellular Bioenginee-ring (CMCB), TU Dresden, Dresden, 01062, Germany; Max Planck Institute of Molecular Cell Biology and Genetics, 01307, Dresden, Germany.
| | - Anette-Gabriele Ziegler
- Institute of Diabetes Research, Helmholtz Munich, German Center for Environmental Health, Munich, Germany; Forschergruppe Diabetes, School of Medicine, Klinikum rechts der Isar, Technical University Munich, Munich, Germany; Forschergruppe Diabetes e.V. at Helmholtz Munich, German Research Center for Environmental Health, Munich, Germany.
| | - Ezio Bonifacio
- Center for Regenerative Therapies Dresden, Faculty of Medicine, TU Dresden, Dresden, Germany; German Center for Diabetes Research (DZD), Paul Langerhans Institute Dresden of Helmholtz Centre Munich at University Clinic Carl Gustav Carus of TU Dresden, Faculty of Medicine, Dresden, Germany.
| | - Anne Eugster
- Center for Regenerative Therapies Dresden, Faculty of Medicine, TU Dresden, Dresden, Germany.
| |
Collapse
|
14
|
Yang C, Chen W, Huang Y. Long non-coding RNA SUN2-AS1 acts as a negative regulator of ISGs transcription to promote flavivirus infection. Virology 2024; 600:110245. [PMID: 39288611 DOI: 10.1016/j.virol.2024.110245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 09/04/2024] [Accepted: 09/12/2024] [Indexed: 09/19/2024]
Abstract
Recent studies highlight the critical involvement of long non-coding RNAs (lncRNAs) in modulating viral replication and immune responses, yet their specific roles in flavivirus infections remain underexplored. Our study has identified lncRNA SUN2-AS1, which is significantly upregulated in response to flavivirus infection in A549, Huh7 cells, and monocyte-differentiated macrophages (MDMs). SUN2-AS1 interacts with the transcription factors NF-κB and STAT1, andits expression is induced by ZIKV RNA via the type I interferon (IFN) pathway. Notably, SUN2-AS1 enhances the infection of flaviviruses, including ZIKV, DENV2, and JEV, while showing no effect on VSV or HSV-1 infections. Mechanistically, SUN2-AS1 exerts a proviral effect by inhibiting the transcription of interferon-stimulated genes (ISGs). These findings uncover a novel mechanism by which lncRNAs facilitate flavivirus propagation and highlight SUN2-AS1 as a potential target for antiviral therapeutic strategies.
Collapse
Affiliation(s)
- Chao Yang
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China; Guangxi Hospital Division of the First Affiliated Hospital, Sun Yat-sen University, Nanning, 530022, China
| | - Weikang Chen
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China.
| | - Yanxia Huang
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China.
| |
Collapse
|
15
|
Valensin C, Côté EJM, Pereira-Carvalho D, Gardner RA, Nishku G, Giles CL, Gill C, Brockbank A, Story L, Shennan AH, Suff N, Gibbons DL, Tribe RM. INSIGHT-2: mechanistic studies into pregnancy complications and their impact on maternal and child health-study protocol. Reprod Health 2024; 21:177. [PMID: 39609862 PMCID: PMC11605920 DOI: 10.1186/s12978-024-01911-0] [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: 10/09/2024] [Accepted: 11/14/2024] [Indexed: 11/30/2024] Open
Abstract
BACKGROUND Pregnancy and early childhood cohorts provide a framework for investigating the complex interplay between early-life exposures and health outcomes, thereby informing prevention strategies and interventions to improve maternal and child health. In this paper, we outline the objectives, methodologies and expected contributions of INSIGHT-2, a comprehensive cohort study dedicated to advancing our understanding of pregnancy and pregnancy complications towards improving the health and well-being of mothers and their offspring. METHODS Over the course of 5 years, the study aims to establish a diverse cohort of 1700 pregnant women and to follow up their children up to 2 years of age. Recruitment targets participants with healthy pregnancies, preexisting conditions, and/or risk factors for pregnancy complications or later child health problems. Clinical and lifestyle data and a range of biological samples will be collected, providing a comprehensive resource for biomarker investigations and cross-sectional analyses. It is anticipated that the cohort will continue beyond this initial 5-year plan. DISCUSSION By gathering a wide range of biological samples and using diverse analytical techniques, this study supports broad participation, potential replication and collaboration across various sites. The extensive collection of longitudinal data and samples not only facilitates current investigations but also establishes a biobank for future research. The exploration of pre-pregnancy and pregnancy factors that may contribute to disease processes and impact fetal well-being and future health will provide a comprehensive picture of disease mechanisms in both mothers and children, facilitating the identification of biomarkers for the prediction, diagnosis, and management of pregnancy complications. Additionally, our diverse population allows for the capture of various pregnancy complications and outcomes, enhancing external validity and addressing health disparities. This comprehensive design ultimately aims to improve maternal and child health outcomes by providing a valuable longitudinal study of the relationships among the in utero environment, pregnancy management, and long-term maternal and child health, ensuring that findings are relevant and beneficial to a broader population.
Collapse
Affiliation(s)
- Carlotta Valensin
- Department of Women and Children's Health, St Thomas' Hospital, King's College London, London, UK.
| | - Emilie J M Côté
- Department of Women and Children's Health, St Thomas' Hospital, King's College London, London, UK
| | - Daniela Pereira-Carvalho
- Department of Women and Children's Health, St Thomas' Hospital, King's College London, London, UK
- Peter Gorer Department of Immunobiology, Guy's Hospital, King's College London, London, UK
| | - Rachael A Gardner
- Guy's and St Thomas' National Health Service Foundation Trust, London, UK
| | - Glen Nishku
- Guy's and St Thomas' National Health Service Foundation Trust, London, UK
| | - Caitlin L Giles
- Department of Women and Children's Health, St Thomas' Hospital, King's College London, London, UK
| | - Carolyn Gill
- Department of Women and Children's Health, St Thomas' Hospital, King's College London, London, UK
| | - Anna Brockbank
- Department of Women and Children's Health, St Thomas' Hospital, King's College London, London, UK
| | - Lisa Story
- Department of Women and Children's Health, St Thomas' Hospital, King's College London, London, UK
- Department of Perinatal Imaging, St Thomas' Hospital, King's College London, London, UK
- Guy's and St Thomas' National Health Service Foundation Trust, London, UK
| | - Andrew H Shennan
- Department of Women and Children's Health, St Thomas' Hospital, King's College London, London, UK
- Guy's and St Thomas' National Health Service Foundation Trust, London, UK
| | - Natalie Suff
- Department of Women and Children's Health, St Thomas' Hospital, King's College London, London, UK
| | - Deena L Gibbons
- Peter Gorer Department of Immunobiology, Guy's Hospital, King's College London, London, UK
| | - Rachel M Tribe
- Department of Women and Children's Health, St Thomas' Hospital, King's College London, London, UK
| |
Collapse
|
16
|
Nakamura K, Shigeyasu K, Vu TH, Maki J, Okamoto K, Masuyama H. ADAR1 could be a potential diagnostic target for intrauterine infection patients. Sci Rep 2024; 14:29419. [PMID: 39592688 PMCID: PMC11599928 DOI: 10.1038/s41598-024-81097-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 11/25/2024] [Indexed: 11/28/2024] Open
Abstract
Intrauterine infection (IUI) is mainly an ascending infection in which vaginal and cervical pathogens ascend to the uterus and can affect the fetus. Until now, there is still no effective diagnostic biomarker for IUI, such as chorioamnionitis (CAM) and funisitis (FUN). Deoxyribonucleic acid (DNA)/Ribonucleic acid (RNA) editing molecules such as apolipoprotein-B mRNA-editing complex (APOBEC) 3 families and Adenosine deaminase family acting on RNA (ADAR)1 were examined in chorioamniotic membranes and umbilical cord of 83 patient samples. Furthermore, Ureaplasma parvum induced ADAR1 was investigated in human HTR-8/SVneo EVT cell line. ADAR1 had a significantly higher area under the curve (AUC) (0.721 and 0.745) than other APOBEC3s or cytokines in CAM and FUN patients. In vitro, ureaplasma parvum was demonstrated to activate ADAR1 (p = 0.025) and reduce RIG-I, IRF3, IFN-[Formula: see text], and IFN-β expression in EVT cell line (p = 0.005, p = 0.010, p < 0.001, and p = 0.018, respectively). High expression of ADAR1 was strongly associated with CAM and FUN patients (multivariate analyses; p = 0.035 and p = 0.002). ADAR1 could be a potential diagnostic target for IUI, such as CAM and FUN patients.
Collapse
Affiliation(s)
- Keiichiro Nakamura
- Department of Obstetrics and Gynecology, Dentistry and Pharmaceutical Sciences, Okayama University Graduate School of Medicine, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan.
| | - Kunitoshi Shigeyasu
- Department of Gastroenterological Surgery, Dentistry and Pharmaceutical Sciences, Okayama University Graduate School of Medicine, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Thuy Ha Vu
- Department of Obstetrics and Gynecology, Dentistry and Pharmaceutical Sciences, Okayama University Graduate School of Medicine, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Jota Maki
- Department of Obstetrics and Gynecology, Dentistry and Pharmaceutical Sciences, Okayama University Graduate School of Medicine, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Kazuhiro Okamoto
- Department of Obstetrics and Gynecology, Dentistry and Pharmaceutical Sciences, Okayama University Graduate School of Medicine, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Hisashi Masuyama
- Department of Obstetrics and Gynecology, Dentistry and Pharmaceutical Sciences, Okayama University Graduate School of Medicine, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| |
Collapse
|
17
|
Obeagu EI, Obeagu GU. Neutrophil phenotypes in prolonged labor: Implications for therapeutic strategies. Medicine (Baltimore) 2024; 103:e40611. [PMID: 39560573 PMCID: PMC11576028 DOI: 10.1097/md.0000000000040611] [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: 12/19/2023] [Accepted: 11/01/2024] [Indexed: 11/20/2024] Open
Abstract
Prolonged labor, defined as labor extending beyond 20 hours for nulliparas and 14 hours for multiparas, poses significant risks to both maternal and neonatal health. The inflammatory response plays a crucial role in the pathophysiology of prolonged labor, with neutrophils being key players in this process. Neutrophils, the most abundant leukocytes, exhibit diverse phenotypes and functions in response to prolonged labor, influencing both the onset and progression of labor through their inflammatory actions. Classical neutrophils (N1) are involved in acute inflammatory responses, aiding in tissue remodeling and labor onset, but their prolonged activation can lead to tissue damage. Regulatory neutrophils (N2), which produce anti-inflammatory cytokines, help resolve inflammation and facilitate labor progression. Low-density granulocytes and aged neutrophils, associated with chronic inflammation and impaired function respectively, contribute to labor complications. The balance among these neutrophil phenotypes is crucial for maintaining a controlled inflammatory response during labor. Therapeutic strategies targeting neutrophil recruitment, NETosis, and cytokine production hold promise for managing prolonged labor. Modulating chemokine pathways, regulating NET formation, and balancing cytokine profiles may reduce inflammation and improve labor outcomes. Further research into the mechanisms of neutrophil regulation and the development of targeted therapies is essential for mitigating the adverse effects of prolonged labor and enhancing maternal and neonatal health.
Collapse
|
18
|
Szukiewicz D. Insights into Reproductive Immunology and Placental Pathology. Int J Mol Sci 2024; 25:12135. [PMID: 39596208 PMCID: PMC11594814 DOI: 10.3390/ijms252212135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 11/09/2024] [Accepted: 11/10/2024] [Indexed: 11/28/2024] Open
Abstract
The formation of a daughter organism as a result of the fusion of an egg and a sperm cell, followed by the implantation of the embryo, the formation of the placenta, and the further growth of the embryo and then fetus until delivery, poses particular challenges for the immune system [...].
Collapse
Affiliation(s)
- Dariusz Szukiewicz
- Department of Biophysics, Physiology & Pathophysiology, Faculty of Health Sciences, Medical University of Warsaw, 02-004 Warsaw, Poland
| |
Collapse
|
19
|
Maxwell A, Swanson G, Thy Nguyen A, Hu A, Richards D, You Y, Stephan L, Manaloto M, Liao A, Ding J, Mor G. Hydroquinone impairs trophoblast migration and invasion via AHR-twist-IFITM1 axis. Placenta 2024; 155:88-99. [PMID: 39173312 PMCID: PMC11421844 DOI: 10.1016/j.placenta.2024.07.315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 07/21/2024] [Accepted: 07/31/2024] [Indexed: 08/24/2024]
Abstract
INTRODUCTION Embryo implantation is a tightly regulated process, critical for a successful pregnancy. After attachment of the blastocyst to the surface epithelium of the endometrium trophoblast migrate from the trophectoderm and invade into the stromal component of endometrium. Alterations on either process will lead to implantation failure or miscarriage. Volatile organic compounds (VOCs) such as benzene induce pregnancy complications, including preterm birth and miscarriages. The mechanism of this effect is unknown. The objective of this study was to elucidate the impact of benzene metabolite, Hydroquinone, on trophoblast function. We tested the hypothesis that Hydroquinone activates the Aryl hydrocarbon receptor (AhR) pathway modulating trophoblast migration and invasion. METHODS First-trimester trophoblast cells (Sw.71) were treated with hydroquinone (6 and 25 μM). Trophoblast migration and invasion was evaluated using a 3D invasion/migration model. Gene expression was quantified by q-PCR and Western blot analysis. RESULTS Hydroquinone impairs trophoblast migration and invasion. This loss is associated with the activation of the AhR pathway which reduced the expression of Twist1and IFITM1. IFITM1 overexpression can rescue impaired trophoblast migration. DISCUSSION Our study highlights that hydroquinone treatment induces the activation of the AhR pathway in trophoblast cells, which impairs trophoblast invasion and migration. We postulate that activation of the AhR pathway in trophoblast suppress Twist1 and a subsequent IFITM1. Thus, the AhR-Twist1-IFITM1 axis represent a critical pathway involved in the regulation of trophoblast migration and it is sensitive to benzene exposure. These findings provide crucial insights into the molecular mechanisms underlying pregnancy complications induced by air pollution.
Collapse
Affiliation(s)
- Anthony Maxwell
- C.S Mott Center for Human Growth and Development, Wayne State University, Detroit, MI, USA
| | - Grace Swanson
- C.S Mott Center for Human Growth and Development, Wayne State University, Detroit, MI, USA
| | - Annie Thy Nguyen
- C.S Mott Center for Human Growth and Development, Wayne State University, Detroit, MI, USA
| | - Anna Hu
- C.S Mott Center for Human Growth and Development, Wayne State University, Detroit, MI, USA
| | - Darby Richards
- C.S Mott Center for Human Growth and Development, Wayne State University, Detroit, MI, USA
| | - Yuan You
- C.S Mott Center for Human Growth and Development, Wayne State University, Detroit, MI, USA
| | - Laura Stephan
- C.S Mott Center for Human Growth and Development, Wayne State University, Detroit, MI, USA
| | - Marcia Manaloto
- C.S Mott Center for Human Growth and Development, Wayne State University, Detroit, MI, USA
| | - Aihua Liao
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Jiahui Ding
- C.S Mott Center for Human Growth and Development, Wayne State University, Detroit, MI, USA; Department of Obstetrics and Gynecology, School of Medicine, Wayne State University, Detroit, MI, USA
| | - Gil Mor
- C.S Mott Center for Human Growth and Development, Wayne State University, Detroit, MI, USA; Department of Obstetrics and Gynecology, School of Medicine, Wayne State University, Detroit, MI, USA.
| |
Collapse
|
20
|
McMillen CM, Megli C, Radisic R, Skvarca LB, Hoehl RM, Boyles DA, McGaughey JJ, Bird BH, McElroy AK, Hartman AL. Vaccine strains of Rift Valley fever virus exhibit attenuation at the maternal-fetal placental interface. J Virol 2024; 98:e0098324. [PMID: 39016561 PMCID: PMC11334480 DOI: 10.1128/jvi.00983-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 06/17/2024] [Indexed: 07/18/2024] Open
Abstract
Rift Valley fever virus (RVFV) infection causes abortions in ruminant livestock and is associated with an increased likelihood of miscarriages in women. Using sheep and human placenta explant cultures, we sought to identify tissues at the maternal-fetal interface targeted by RVFV. Sheep villi and fetal membranes were highly permissive to RVFV infection resulting in markedly higher virus titers than human cultures. Sheep cultures were most permissive to wild-type RVFV and ΔNSm infection, while live-attenuated RVFV vaccines (LAVs; MP-12, ΔNSs, and ΔNSs/ΔNSm) exhibited reduced replication. The human fetal membrane restricted wild-type and LAV replication, and when infection occurred, it was prominent on the maternal-facing side. Type I and type III interferons were induced in human villi exposed to LAVs lacking the NSs protein. This study supports the use of sheep and human placenta explants to understand vertical transmission of RVFV in mammals and whether LAVs are attenuated at the maternal-fetal interface.IMPORTANCEA direct comparison of replication of Rift Valley fever virus (RVFV) in sheep and human placental explants reveals comparative efficiencies and permissivity to infection and replication. Vaccine strains of RVFV demonstrated reduced infection and replication capacity in the mammalian placenta. This study represents the first direct cross-host comparison of the vertical transmission capacity of this high-priority emerging mosquito-transmitted virus.
Collapse
Affiliation(s)
- Cynthia M. McMillen
- University of Pittsburgh, Center for Vaccine Research, Pittsburgh, Pennsylvania, USA
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, School of Public Health, Pittsburgh, Pennsylvania, USA
| | - Christina Megli
- Division of Maternal-Fetal Medicine, Division of Reproductive Infectious Disease, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine and the Magee-Womens Research Institute, Pittsburgh, Pennsylvania, USA
| | - Rebecca Radisic
- One Health Institute, School of Veterinary Medicine, University of California, Davis, California, USA
| | - Lauren B. Skvarca
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Ryan M. Hoehl
- University of Pittsburgh, Center for Vaccine Research, Pittsburgh, Pennsylvania, USA
| | - Devin A. Boyles
- University of Pittsburgh, Center for Vaccine Research, Pittsburgh, Pennsylvania, USA
| | - Jackson J. McGaughey
- University of Pittsburgh, Center for Vaccine Research, Pittsburgh, Pennsylvania, USA
| | - Brian H. Bird
- One Health Institute, School of Veterinary Medicine, University of California, Davis, California, USA
| | - Anita K. McElroy
- University of Pittsburgh, Center for Vaccine Research, Pittsburgh, Pennsylvania, USA
- Department of Pediatrics, Division of Pediatric Infectious Disease, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Amy L. Hartman
- University of Pittsburgh, Center for Vaccine Research, Pittsburgh, Pennsylvania, USA
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, School of Public Health, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
21
|
Nakamura K, Shigeyasu K, Maki J, Eto E, Masuyama H. The adenosine deaminase family acting on RNA 1 can be a useful diagnostic biomarker in chorioamnionitis. Placenta 2024; 153:53-58. [PMID: 38820942 DOI: 10.1016/j.placenta.2024.05.133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 05/06/2024] [Accepted: 05/22/2024] [Indexed: 06/02/2024]
Abstract
INTRODUCTION Chorioamnionitis (CAM) involves infection and inflammation of the chorion and amniotic membrane, but there are still no effective diagnostic biomarkers for CAM. METHODS We investigated the correlation between RNA editing enzyme Adenosine deaminase family acting on RNA 1 (ADAR1) and CAM in chorion and amniotic membrane specimens derived from premature rupture of the membrane (PROM), CAM (pathologically diagnosed), and clinical CAM (clinically diagnosed) patients using reverse transcription polymerase chain reaction (RT-PCR). RESULTS ADAR1 was upregulated in the chorion and amniotic membrane specimens of CAM and clinical CAM patients (p < 0.001 and p = 0.005). ADAR1 had a significantly higher area under the curve (AUC) (0.735 and 0.828) than markers of inflammation characteristics in diagnosing CAM and clinical CAM patients. ADAR1 also had significantly higher AUC (0.701 and 0.837) than clinical characteristics for CAM and clinical CAM patients. DISCUSSION ADAR1 can be a useful diagnostic biomarker in CAM patients.
Collapse
Affiliation(s)
- Keiichiro Nakamura
- Department of Obstetrics and Gynecology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan.
| | - Kunitoshi Shigeyasu
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Jota Maki
- Department of Obstetrics and Gynecology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Eriko Eto
- Department of Obstetrics and Gynecology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Hisashi Masuyama
- Department of Obstetrics and Gynecology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| |
Collapse
|
22
|
Ye M, Nguyen S, Kim MJ, Hwang JS, Bae GW, Yang KHS, Shim I. Antidepressant Effect of Enzymatic Porcine Placenta Hydrolysate in Repeated Immobilization Stress-Induced Ovariectomized Female Mice. Curr Issues Mol Biol 2024; 46:6121-6138. [PMID: 38921037 PMCID: PMC11202803 DOI: 10.3390/cimb46060366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 06/04/2024] [Accepted: 06/12/2024] [Indexed: 06/27/2024] Open
Abstract
When postmenopausal women are under stress conditions, this exacerbates mood disorders and issues with neuroimmune systems. The porcine placenta is known to relieve menopausal depression in clinical trials, but its underlying mechanisms for depression and anti-inflammatory functions remain poorly defined. The present study was designed to examine the anti-inflammatory effects of enzymatic porcine placenta hydrolysate (EPPH) on LPS-induced levels of nitric oxide (NO), prostaglandin E2 (PGE2), corticosterone (CORT), and pro-inflammatory cytokine interleukin-1 beta (IL-1β) in RAW 264.7 macrophage cells. In addition, the neurite outgrowth of PC12 cells was evaluated to examine the effects of EPPH on neurite growth. To mimic the symptoms of women with menopause-related depression, a stressed ovariectomized (OVX) female mouse model was used to evaluate the antidepressant effects of EPPH. The female mice were randomly divided into five groups: (1) the sham-operated (Sham) group, (2) the OVX + repeated stress + saline-treated (OVX + ST) group, (3) the OVX + repeated stress + estradiol (0.2 mg/kg)-treated (positive control) group, (4) the OVX + repeated stress + EPPH (300 mg/kg)-treated (300) group, and (5) the OVX + repeated stress + EPPH (1500 mg/kg)-treated (1500) group. Female mice were OVX and repeatedly immobilization-stressed for 2 weeks (2 h/day). A tail suspension test was conducted on the 13th day, followed by the forced swimming test on the 14th day to assess the antidepressant effects of EPPH. After the behavioral tests, the levels of CORT, PGE2, and IL-1β were evaluated. In addition, c-Fos expression in the paraventricular nucleus (PVN) was evaluated using immunohistochemistry. The concentrations of NO, PGE2, and IL-1β stimulated by LPS were significantly reduced via the addition of EPPH to RAW 264.7 cells. EPPH significantly promoted neurite outgrowth in PC12 cells compared to that of the controls. In the tail suspension test, the duration of immobility was reduced in mice treated with EPPH 1500 compared to the OVX + ST group. The EPPH 1500 group had significantly decreased levels of c-Fos-positive neurons in the PVN and reduced levels of CORT and IL-1β in the serum of the Sham group. These results suggested that the high dose of EPPH administration induced the antidepressant-like effect in the ovariectomized mice with repeated stress via downregulating the levels of CORT, IL-1β, and PGE2 in the serum through reducing the expression of c-Fos in the PVN regions.
Collapse
Affiliation(s)
- Minsook Ye
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Sharon Nguyen
- Biological Sciences Program, Schmid College of Science and Technology, Chapman University, Orange, CA 92866, USA
| | - Min Ju Kim
- Department of R&D, Unimed Pharmaceuticals Inc., Unimed Bldg., Seoul 05567, Republic of Korea (J.S.H.); (G.W.B.)
| | - Jee Sun Hwang
- Department of R&D, Unimed Pharmaceuticals Inc., Unimed Bldg., Seoul 05567, Republic of Korea (J.S.H.); (G.W.B.)
| | - Gun Won Bae
- Department of R&D, Unimed Pharmaceuticals Inc., Unimed Bldg., Seoul 05567, Republic of Korea (J.S.H.); (G.W.B.)
| | - Keun-Hang Susan Yang
- Biological Sciences Program, Schmid College of Science and Technology, Chapman University, Orange, CA 92866, USA
- Institute for Earth, Computing, Human and Observing (ECHO), Chapman University, Orange, CA 92866, USA
| | - Insop Shim
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
23
|
Hansen SSK, Krautz R, Rago D, Havelund J, Stigliani A, Færgeman NJ, Prézelin A, Rivière J, Couturier-Tarrade A, Akimov V, Blagoev B, Elfving B, Neess D, Vogel U, Khodosevich K, Hougaard KS, Sandelin A. Pulmonary maternal immune activation does not cross the placenta but leads to fetal metabolic adaptation. Nat Commun 2024; 15:4711. [PMID: 38830841 PMCID: PMC11148039 DOI: 10.1038/s41467-024-48492-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 04/29/2024] [Indexed: 06/05/2024] Open
Abstract
The fetal development of organs and functions is vulnerable to perturbation by maternal inflammation which may increase susceptibility to disorders after birth. Because it is not well understood how the placenta and fetus respond to acute lung- inflammation, we characterize the response to maternal pulmonary lipopolysaccharide exposure across 24 h in maternal and fetal organs using multi-omics, imaging and integrative analyses. Unlike maternal organs, which mount strong inflammatory immune responses, the placenta upregulates immuno-modulatory genes, in particular the IL-6 signaling suppressor Socs3. Similarly, we observe no immune response in the fetal liver, which instead displays metabolic changes, including increases in lipids containing docosahexaenoic acid, crucial for fetal brain development. The maternal liver and plasma display similar metabolic alterations, potentially increasing bioavailability of docosahexaenoic acid for the mother and fetus. Thus, our integrated temporal analysis shows that systemic inflammation in the mother leads to a metabolic perturbation in the fetus.
Collapse
Affiliation(s)
- Signe Schmidt Kjølner Hansen
- Department of Biology, University of Copenhagen, Copenhagen, Denmark.
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark.
- National Research Centre for the Working Environment, Copenhagen, Denmark.
| | - Robert Krautz
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Daria Rago
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Jesper Havelund
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Arnaud Stigliani
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Nils J Færgeman
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Audrey Prézelin
- Université Paris-Saclay, UVSQ, INRAE, BREED, 78350, Jouy-en-Josas, France
- Ecole Nationale Vétérinaire d'Alfort, BREED, 94700, Maisons-Alfort, France
| | - Julie Rivière
- Paris-Saclay University, INRAE, AgroParisTech, GABI, 78350, Jouy-en-Josas, France
- Paris-Saclay University, INRAE, AgroParisTech, Micalis Institute, 78350, Jouy-en-Josas, France
| | - Anne Couturier-Tarrade
- Université Paris-Saclay, UVSQ, INRAE, BREED, 78350, Jouy-en-Josas, France
- Ecole Nationale Vétérinaire d'Alfort, BREED, 94700, Maisons-Alfort, France
| | - Vyacheslav Akimov
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Blagoy Blagoev
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Betina Elfving
- Translational Neuropsychiatry Unit, Aarhus University, Aarhus, Denmark
| | - Ditte Neess
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Ulla Vogel
- National Research Centre for the Working Environment, Copenhagen, Denmark
| | - Konstantin Khodosevich
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Karin Sørig Hougaard
- National Research Centre for the Working Environment, Copenhagen, Denmark.
- Department of Public Health, University of Copenhagen, Copenhagen, Denmark.
| | - Albin Sandelin
- Department of Biology, University of Copenhagen, Copenhagen, Denmark.
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
24
|
McMillen CM, Megli C, Radisic R, Skvarca LB, Hoehl RM, Boyles DA, McGaughey JJ, Bird BH, McElroy AK, Hartman AL. Vaccine strains of Rift Valley fever virus exhibit attenuation at the maternal-fetal placental interface. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.31.596800. [PMID: 38854055 PMCID: PMC11160702 DOI: 10.1101/2024.05.31.596800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Rift Valley fever virus (RVFV) infection causes abortions in ruminant livestock and is associated with an increased likelihood of miscarriages in women. Using sheep and human placenta explant cultures, we sought to identify tissues at the maternal-fetal interface targeted by RVFV. Sheep villi and fetal membranes were highly permissive to RVFV infection resulting in markedly higher virus titers than human cultures. Sheep cultures were most permissive to wild-type RVFV and ΔNSm infection, while live attenuated RVFV vaccines (LAVs; MP-12, ΔNSs, and ΔNSs/ΔNSm) exhibited reduced replication. The human fetal membrane restricted wild-type and LAV replication, and when infection occurred, it was prominent in the maternal-facing side. Type-I and type-III interferons were induced in human villi exposed to LAVs lacking the NSs protein. This study supports the use of sheep and human placenta explants to understand vertical transmission of RVFV in mammals and whether LAVs are attenuated at the maternal-fetal interface.
Collapse
Affiliation(s)
- Cynthia M. McMillen
- University of Pittsburgh, Center for Vaccine Research, Pittsburgh, PA, USA
- University of Pittsburgh, Department of Infectious Diseases and Microbiology, School of Public Health, Pittsburgh, PA, USA
| | - Christina Megli
- University of Pittsburgh School of Medicine and the Magee-Womens Research Institute, Division of Maternal-Fetal Medicine, Division of Reproductive Infectious Disease, Department of Obstetrics, Gynecology and Reproductive Sciences, Pittsburgh, PA, USA
| | - Rebecca Radisic
- One Health Institute, School of Veterinary Medicine, University of California, Davis, CA 95616, USA
| | - Lauren B. Skvarca
- University of Pittsburgh School of Medicine, Department of Pathology, Pittsburgh, PA, USA
| | - Ryan M. Hoehl
- University of Pittsburgh, Center for Vaccine Research, Pittsburgh, PA, USA
| | - Devin A. Boyles
- University of Pittsburgh, Center for Vaccine Research, Pittsburgh, PA, USA
| | | | - Brian H. Bird
- One Health Institute, School of Veterinary Medicine, University of California, Davis, CA 95616, USA
| | - Anita K. McElroy
- University of Pittsburgh, Center for Vaccine Research, Pittsburgh, PA, USA
- University of Pittsburgh School of Medicine, Department of Pediatrics, Division of Pediatric Infectious Disease, Pittsburgh, PA, USA
| | - Amy L. Hartman
- University of Pittsburgh, Center for Vaccine Research, Pittsburgh, PA, USA
- University of Pittsburgh, Department of Infectious Diseases and Microbiology, School of Public Health, Pittsburgh, PA, USA
| |
Collapse
|
25
|
Chen Z, Zhou X, Qu H, Zhang X, Kwak-Kim J, Wang W. Characteristics and functions of memory regulatory T cells in normal pregnancy cycle and pregnancy complications. J Reprod Immunol 2024; 163:104235. [PMID: 38574576 DOI: 10.1016/j.jri.2024.104235] [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: 08/09/2023] [Revised: 02/21/2024] [Accepted: 03/07/2024] [Indexed: 04/06/2024]
Abstract
Regulatory T cells (Tregs) are activated and expanded after exposure to fetal-specific (paternal) antigens. A proportion of Tregs differentiate into memory Tregs (mTregs), exhibiting immune memory function and exerting more potent immunosuppression than naive Tregs (nTregs). However, it is unclear how mTregs are regulated during normal and pathological pregnancies (e.g., gestational diabetes mellitus (GDM) and preeclampsia (PE)). In this study, PD-1, HLA-G, and HLA-DR expressions on memory CD4+ T cells, naive CD4+ T cells, Tregs, mTregs, and nTregs in healthy non-pregnant women (n=20), healthy first (n=20), second (n=20), and third-trimester women (n=20), postpartum women (n=20), GDM (n=20), and PE patients (n=20) were analyzed. The proportion of mTregs out of Tregs was increased (P<0.05) in the first trimester compared with that in non-pregnancy and reduced in the second and third trimesters. The proportions of PD-1+ Tregs and mTregs were significantly increased during the first trimester compared to those of non-pregnancy (P<0.01), reached their maximum in the second trimester. Moreover, the proportions of HLA-G+ memory CD4+ T cells, Tregs, and mTregs were increased in the first and second trimesters (P<0.01), reached their maximum in the third trimester. GDM patients were characterized by significantly lower percentages of PD-1+ and HLA-G+ mTregs (P<0.01), while PE patients were characterized by significantly lower percentages of HLA-G+ mTregs (P<0.01), compared with the healthy third-trimester women. In general, as demonstrated by this study, mTregs increase in number and enhance maternal-fetal immunoregulation during pregnancy, and their dysfunction can result in pregnancy complications such as GMD or PE.
Collapse
Affiliation(s)
- Zeyang Chen
- Reproduction Medical Center, Xinhua Hospital affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200092, PR China; School of Medicine, Qingdao University, Qingdao 266000, PR China
| | - Xiaojiao Zhou
- Department of Pharmacy, the Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, PR China
| | - Hongmei Qu
- Department of Obstetrics and Gynecology, the Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, PR China
| | - Xiaolu Zhang
- Department of Clinical Laboratory, the Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, PR China
| | - Joanne Kwak-Kim
- Reproductive Medicine and Immunology, Obstetrics and Gynecology, Clinical Sciences Department, Chicago Medical School, Rosalind Franklin University of Medicine and Science, Vernon Hills, IL 60061, USA; Center for Cancer Cell Biology, Immunology and Infection, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
| | - Wenjuan Wang
- Reproduction Medical Center, Xinhua Hospital affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200092, PR China.
| |
Collapse
|
26
|
Hoo R, Ruiz-Morales ER, Kelava I, Rawat M, Mazzeo CI, Tuck E, Sancho-Serra C, Chelaghma S, Predeus AV, Murray S, Fernandez-Antoran D, Waller RF, Álvarez-Errico D, Lee MCS, Vento-Tormo R. Acute response to pathogens in the early human placenta at single-cell resolution. Cell Syst 2024; 15:425-444.e9. [PMID: 38703772 DOI: 10.1016/j.cels.2024.04.002] [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: 05/19/2023] [Revised: 12/01/2023] [Accepted: 04/16/2024] [Indexed: 05/06/2024]
Abstract
The placenta is a selective maternal-fetal barrier that provides nourishment and protection from infections. However, certain pathogens can attach to and even cross the placenta, causing pregnancy complications with potential lifelong impacts on the child's health. Here, we profiled at the single-cell level the placental responses to three pathogens associated with intrauterine complications-Plasmodium falciparum, Listeria monocytogenes, and Toxoplasma gondii. We found that upon exposure to the pathogens, all placental lineages trigger inflammatory responses that may compromise placental function. Additionally, we characterized the responses of fetal macrophages known as Hofbauer cells (HBCs) to each pathogen and propose that they are the probable niche for T. gondii. Finally, we revealed how P. falciparum adapts to the placental microenvironment by modulating protein export into the host erythrocyte and nutrient uptake pathways. Altogether, we have defined the cellular networks and signaling pathways mediating acute placental inflammatory responses that could contribute to pregnancy complications.
Collapse
Affiliation(s)
- Regina Hoo
- Wellcome Sanger Institute, Cambridge, UK; Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
| | | | - Iva Kelava
- Wellcome Sanger Institute, Cambridge, UK
| | - Mukul Rawat
- Wellcome Sanger Institute, Cambridge, UK; Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, University of Dundee, Dundee, UK
| | | | | | | | - Sara Chelaghma
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | | | | | - David Fernandez-Antoran
- Wellcome/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge, UK; Department of Pathology, University of Cambridge, Cambridge, UK
| | - Ross F Waller
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | | | - Marcus C S Lee
- Wellcome Sanger Institute, Cambridge, UK; Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, University of Dundee, Dundee, UK.
| | - Roser Vento-Tormo
- Wellcome Sanger Institute, Cambridge, UK; Centre for Trophoblast Research, University of Cambridge, Cambridge, UK.
| |
Collapse
|
27
|
Khorami-Sarvestani S, Vanaki N, Shojaeian S, Zarnani K, Stensballe A, Jeddi-Tehrani M, Zarnani AH. Placenta: an old organ with new functions. Front Immunol 2024; 15:1385762. [PMID: 38707901 PMCID: PMC11066266 DOI: 10.3389/fimmu.2024.1385762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 04/08/2024] [Indexed: 05/07/2024] Open
Abstract
The transition from oviparity to viviparity and the establishment of feto-maternal communications introduced the placenta as the major anatomical site to provide nutrients, gases, and hormones to the developing fetus. The placenta has endocrine functions, orchestrates maternal adaptations to pregnancy at different periods of pregnancy, and acts as a selective barrier to minimize exposure of developing fetus to xenobiotics, pathogens, and parasites. Despite the fact that this ancient organ is central for establishment of a normal pregnancy in eutherians, the placenta remains one of the least studied organs. The first step of pregnancy, embryo implantation, is finely regulated by the trophoectoderm, the precursor of all trophoblast cells. There is a bidirectional communication between placenta and endometrium leading to decidualization, a critical step for maintenance of pregnancy. There are three-direction interactions between the placenta, maternal immune cells, and the endometrium for adaptation of endometrial immune system to the allogeneic fetus. While 65% of all systemically expressed human proteins have been found in the placenta tissues, it expresses numerous placenta-specific proteins, whose expression are dramatically changed in gestational diseases and could serve as biomarkers for early detection of gestational diseases. Surprisingly, placentation and carcinogenesis exhibit numerous shared features in metabolism and cell behavior, proteins and molecular signatures, signaling pathways, and tissue microenvironment, which proposes the concept of "cancer as ectopic trophoblastic cells". By extensive researches in this novel field, a handful of cancer biomarkers has been discovered. This review paper, which has been inspired in part by our extensive experiences during the past couple of years, highlights new aspects of placental functions with emphasis on its immunomodulatory role in establishment of a successful pregnancy and on a potential link between placentation and carcinogenesis.
Collapse
Affiliation(s)
- Sara Khorami-Sarvestani
- Reproductive Immunology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
- Monoclonal Antibody Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Negar Vanaki
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Sorour Shojaeian
- Department of Biochemistry, School of Medical Sciences, Alborz University of Medical Sciences, Karaj, Iran
| | - Kayhan Zarnani
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Allan Stensballe
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
- Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
| | - Mahmood Jeddi-Tehrani
- Monoclonal Antibody Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Amir-Hassan Zarnani
- Reproductive Immunology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
28
|
Chowdhury SF, Prout N, Rivera-Núñez Z, Barrett E, Brunner J, Duberstein Z, Kannan K, Salafia CM, Shah R, Miller RK, O'Connor TG. PFAS alters placental arterial vasculature in term human placentae: A prospective pregnancy cohort study. Placenta 2024; 149:54-63. [PMID: 38518389 PMCID: PMC10997442 DOI: 10.1016/j.placenta.2024.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 03/04/2024] [Accepted: 03/06/2024] [Indexed: 03/24/2024]
Abstract
INTRODUCTION Perfluoroalkyl substances (PFAS) are synthetic chemicals used in industrial and consumer goods that are widely detected in human populations and are associated with adverse health outcomes, including perinatal health risks and child health. One mechanism of influence may be the impact of PFAS exposure on placental structure and function. OBJECTIVES The objective of this study is to investigate the relationship between maternal prenatal exposure to PFAS and measures of placental vascularization, and to assess whether changes in vascularization play a role in mediating the impact of PFAS on birth outcomes. METHODS Using data from a prospective cohort study, we examined associations between second trimester PFAS (individually and as mixtures using Bayesian kernel machine regression) and placental arterial vasculature in term placentae (N = 158); secondarily we evaluated the degree to which alterations in placental arterial vasculature explained associations between PFAS exposure and birth outcomes. Placental arterial vasculature features were collected from arterial tracings of each placental image. RESULTS In both linear regression and mixture models, natural log-transformed perfluorooctanoic acid concentrations were negatively associated with surface vasculature, indexed by the mean distance from arterial end point to perimeter (β = -0.23, 95% CI: -0.41, -0.041); additionally, maximum arterial tortuosity was negatively associated with placental weight (β = -0.19, 95% CI: -0.34, -0.051). There were no reliable differences in effect by fetal sex. DISCUSSION The findings provide some of the first evidence of PFAS exposure shaping a key measure of placental vascular function, which may underlie the impact of PFAS on perinatal and child health risks.
Collapse
Affiliation(s)
- Sadia Firoza Chowdhury
- Wynne Center for Family Research, University of Rochester, 601 Elmwood Avenue., Rochester, NY, 14642, USA; Translational Biomedical Sciences Program, University of Rochester, 601 Elmwood Avenue., Rochester, NY, 14642, USA.
| | - Nashae Prout
- Wynne Center for Family Research, University of Rochester, 601 Elmwood Avenue., Rochester, NY, 14642, USA; Toxicology Graduate Program, University of Rochester, 601 Elmwood Avenue., Rochester, NY, 14642, USA.
| | - Zorimar Rivera-Núñez
- Department of Biostatistics and Epidemiology, Rutgers School of Public Health, 683 Hoes Lane West, Piscataway, NJ, 08854, USA; Environmental and Occupational Health Sciences Institute, Rutgers University, 170 Frelinghuysen Rd., Piscataway, NJ, 08854, USA.
| | - Emily Barrett
- Department of Biostatistics and Epidemiology, Rutgers School of Public Health, 683 Hoes Lane West, Piscataway, NJ, 08854, USA; Environmental and Occupational Health Sciences Institute, Rutgers University, 170 Frelinghuysen Rd., Piscataway, NJ, 08854, USA; Department of Obstetrics and Gynecology, University of Rochester School of Medicine and Dentistry, USA.
| | - Jessica Brunner
- Department of Obstetrics and Gynecology, University of Rochester School of Medicine and Dentistry, USA.
| | - Zoe Duberstein
- Wynne Center for Family Research, University of Rochester, 601 Elmwood Avenue., Rochester, NY, 14642, USA; Psychology, University of Rochester, Meliora Hall, P.O. Box 270266, Rochester, NY, 14627, USA.
| | - Kurunthachalam Kannan
- Department of Pediatrics and Department of Environmental Medicine, New York University Grossman School of Medicine, 550 1st Ave., New York, NY, 10016, USA.
| | - Carolyn M Salafia
- Placental Analytics LLC, 187 Overlook Circle, New Rochelle, NY, 10804, USA; Institute for Basic Research, 1550 Forest Hill Road, Staten Island, NY 10314, USA; New York Presbyterian- Brooklyn Methodist Hospital, 550 6th Street, Brooklyn, NY, 11215, USA; Queens Hospital Center, 82-68 164th Street, Queens, New York, 11432, USA.
| | - Ruchit Shah
- Placental Analytics LLC, 187 Overlook Circle, New Rochelle, NY, 10804, USA.
| | - Richard K Miller
- Department of Obstetrics and Gynecology, University of Rochester School of Medicine and Dentistry, USA.
| | - Thomas G O'Connor
- Wynne Center for Family Research, University of Rochester, 601 Elmwood Avenue., Rochester, NY, 14642, USA; Department of Obstetrics and Gynecology, University of Rochester School of Medicine and Dentistry, USA; Psychology, University of Rochester, Meliora Hall, P.O. Box 270266, Rochester, NY, 14627, USA; Department of Psychiatry, University of Rochester, 300 Crittenden Blvd., Rochester, NY, 14642, USA; Department of Neuroscience, University of Rochester, 601 Elmwood Avenue., Rochester, NY, 14642, USA.
| |
Collapse
|
29
|
Cruz-Holguín VJ, González-García LD, Velázquez-Cervantes MA, Arévalo-Romero H, De Jesús-González LA, Helguera-Repetto AC, León-Reyes G, Salazar MI, Cedillo-Barrón L, León-Juárez M. Collateral Damage in the Placenta during Viral Infection in Pregnancy: A Possible Mechanism for Vertical Transmission and an Adverse Pregnancy Outcome. Diseases 2024; 12:59. [PMID: 38534983 PMCID: PMC10969698 DOI: 10.3390/diseases12030059] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/03/2024] [Accepted: 03/06/2024] [Indexed: 11/11/2024] Open
Abstract
In mammals, the placenta is a connection between a mother and a new developing organism. This tissue has a protective function against some microorganisms, transports nutrients, and exchanges gases and excretory substances between the mother and the fetus. Placental tissue is mainly composed of chorionic villi functional units called trophoblasts (cytotrophoblasts, the syncytiotrophoblast, and extravillous trophoblasts). However, some viruses have developed mechanisms that help them invade the placenta, causing various conditions such as necrosis, poor perfusion, and membrane rupture which, in turn, can impact the development of the fetus and put the mother's health at risk. In this study, we collected the most relevant information about viral infection during pregnancy which can affect both the mother and the fetus, leading to an increase in the probability of vertical transmission. Knowing these mechanisms could be relevant for new research in the maternal-fetal context and may provide options for new therapeutic targets and biomarkers in fetal prognosis.
Collapse
Affiliation(s)
- Victor Javier Cruz-Holguín
- Laboratorio de Virologia Perinatal y Diseño Molecular de Antigenos y Biomarcadores, Departamento de Inmunobioquimica, Instituto Nacional de Perinatología, Mexico City 11000, Mexico; (V.J.C.-H.); (L.D.G.-G.); (M.A.V.-C.)
- Departamento de Biomedicina Molecular, Centro de Investigación y Estudios Avanzados del IPN (CINVESTAV), Mexico City 07360, Mexico;
| | - Luis Didier González-García
- Laboratorio de Virologia Perinatal y Diseño Molecular de Antigenos y Biomarcadores, Departamento de Inmunobioquimica, Instituto Nacional de Perinatología, Mexico City 11000, Mexico; (V.J.C.-H.); (L.D.G.-G.); (M.A.V.-C.)
- Departamento de Biomedicina Molecular, Centro de Investigación y Estudios Avanzados del IPN (CINVESTAV), Mexico City 07360, Mexico;
- Posgrado de Inmunología, Escuela Nacional de Ciencias Biologócas (ENCB), Instituto Politecnico Naciona, Mexico City 11350, Mexico;
| | - Manuel Adrián Velázquez-Cervantes
- Laboratorio de Virologia Perinatal y Diseño Molecular de Antigenos y Biomarcadores, Departamento de Inmunobioquimica, Instituto Nacional de Perinatología, Mexico City 11000, Mexico; (V.J.C.-H.); (L.D.G.-G.); (M.A.V.-C.)
| | - Haruki Arévalo-Romero
- Laboratorio de Inmunologia y Microbiologia Molecular, Division Academica Multidisciplinaria de Jalpa de Méndez, Jalpa de Mendez 86205, Mexico;
| | | | | | - Guadalupe León-Reyes
- Laboratorio de Nutrigenética y Nutrigenómica, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City 14610, Mexico;
| | - Ma. Isabel Salazar
- Posgrado de Inmunología, Escuela Nacional de Ciencias Biologócas (ENCB), Instituto Politecnico Naciona, Mexico City 11350, Mexico;
- Laboratorio Nacional de Vacunología y Virus Tropicales (LNVyVT), Escuela Nacional de Ciencias Biologócas (ENCB), Instituto Politecnico Naciona, Mexico City 11350, Mexico
| | - Leticia Cedillo-Barrón
- Departamento de Biomedicina Molecular, Centro de Investigación y Estudios Avanzados del IPN (CINVESTAV), Mexico City 07360, Mexico;
| | - Moisés León-Juárez
- Laboratorio de Virologia Perinatal y Diseño Molecular de Antigenos y Biomarcadores, Departamento de Inmunobioquimica, Instituto Nacional de Perinatología, Mexico City 11000, Mexico; (V.J.C.-H.); (L.D.G.-G.); (M.A.V.-C.)
| |
Collapse
|
30
|
Kong X, Li R, Chen M, Zheng R, Wang J, Sun C, Qu Y. Endogenous retrovirus HERVH-derived lncRNA UCA1 controls human trophoblast development. Proc Natl Acad Sci U S A 2024; 121:e2318176121. [PMID: 38483994 PMCID: PMC10962953 DOI: 10.1073/pnas.2318176121] [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: 10/19/2023] [Accepted: 02/12/2024] [Indexed: 03/19/2024] Open
Abstract
Endogenous retroviruses (ERVs) are frequently reactivated in mammalian placenta. It has been proposed that ERVs contribute to shaping the gene regulatory network of mammalian trophoblasts, dominantly acting as species- and placental-specific enhancers. However, whether and how ERVs control human trophoblast development through alternative pathways remains poorly understood. Besides the well-recognized function of human endogenous retrovirus-H (HERVH) in maintaining pluripotency of early human epiblast, here we present a unique role of HERVH on trophoblast lineage development. We found that the LTR7C/HERVH subfamily exhibits an accessible chromatin state in the human trophoblast lineage. Particularly, the LTR7C/HERVH-derived Urothelial Cancer Associated 1 (UCA1), a primate-specific long non-coding RNA (lncRNA), is transcribed in human trophoblasts and promotes the proliferation of human trophoblast stem cells (hTSCs), whereas its ectopic expression compromises human trophoblast syncytialization coinciding with increased interferon signaling pathway. Importantly, UCA1 upregulation is detectable in placental samples from early-onset preeclampsia (EO-PE) patients and the transcriptome of EO-PE placenta exhibits considerable similarities to that of the syncytiotrophoblasts differentiated from UCA1-overexpressing hTSCs, supporting up-regulated UCA1 as a potential biomarker of this disease. Altogether, our data shed light on the versatile regulatory role of HERVH in early human development and provide a unique mechanism whereby ERVs exert a function in human placentation and placental syndromes.
Collapse
Affiliation(s)
- Xuhui Kong
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou510080, China
- Key Laboratory for Stem Cells and Tissue Engineering, Sun Yat-sen University, Ministry of Education, Guangzhou510080, China
| | - Ruiqi Li
- Reproductive and Genetic Hospital of Kapok, Hainan571400, China
- Department of Obstetrics and Gynecology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou510120, China
- The First People’s Hospital of Kashgar, Kashgar844000, China
| | - Manqi Chen
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou510080, China
- Key Laboratory for Stem Cells and Tissue Engineering, Sun Yat-sen University, Ministry of Education, Guangzhou510080, China
| | - Rongyan Zheng
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou510080, China
- Key Laboratory for Stem Cells and Tissue Engineering, Sun Yat-sen University, Ministry of Education, Guangzhou510080, China
| | - Jichang Wang
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou510080, China
- Key Laboratory for Stem Cells and Tissue Engineering, Sun Yat-sen University, Ministry of Education, Guangzhou510080, China
| | - Chuanbo Sun
- Laboratory of Medical Systems Biology, Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou510623, China
| | - Yuliang Qu
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou510080, China
- Key Laboratory for Stem Cells and Tissue Engineering, Sun Yat-sen University, Ministry of Education, Guangzhou510080, China
| |
Collapse
|
31
|
Noguchi S, Ohkura S, Negishi Y, Tozawa S, Takizawa T, Morita R, Takahashi H, Ohkuchi A, Takizawa T. Cytoplasmic and nuclear DROSHA in human villous trophoblasts. J Reprod Immunol 2024; 162:104189. [PMID: 38241848 DOI: 10.1016/j.jri.2023.104189] [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: 11/01/2023] [Revised: 12/21/2023] [Accepted: 12/28/2023] [Indexed: 01/21/2024]
Abstract
In villous trophoblasts, DROSHA is a key ribonuclease III enzyme that processes pri-microRNAs (pri-miRNAs) into pre-miRNAs at the placenta-specific, chromosome 19 miRNA cluster (C19MC) locus. However, little is known of its other functions. We performed formaldehyde crosslinking, immunoprecipitation, and sequencing (fCLIP-seq) analysis of terminal chorionic villi to identify DROSHA-binding RNAs in villous trophoblasts. In villous trophoblasts, DROSHA predominantly generated placenta-specific C19MC pre-miRNAs, including antiviral C19MC pre-miRNAs. The fCLIP-seq analysis also identified non-miRNA transcripts with hairpin structures potentially capable of binding to DROSHA (e.g., SNORD100 and VTRNA1-1). Moreover, in vivo immunohistochemical analysis revealed DROSHA in the cytoplasm of villous trophoblasts. DROSHA was abundant in the cytoplasm of villous trophoblasts, particularly in the apical region of syncytiotrophoblast, in the full-term placenta. Furthermore, in BeWo trophoblasts infected with Sindbis virus (SINV), DROSHA translocated to the cytoplasm and recognized the genomic RNA of SINV. Therefore, in trophoblasts, DROSHA not only regulates RNA metabolism, including the biogenesis of placenta-specific miRNAs, but also recognizes viral RNAs. After SINV infection, BeWo DROSHA-binding VTRNA1-1 was significantly upregulated, and cellular VTRNA1-1 was significantly downregulated, suggesting that DROSHA soaks up VTRNA1-1 in response to viral infection. These results suggest that the DROSHA-mediated recognition of RNAs defends against viral infection in villous trophoblasts. Our data provide insight into the antiviral functions of DROSHA in villous trophoblasts of the human placenta.
Collapse
Affiliation(s)
- Syunya Noguchi
- Department of Molecular Medicine and Anatomy, Nippon Medical School, Tokyo 113-8602, Japan
| | - Sadayuki Ohkura
- Department of Microbiology and Immunology, Nippon Medical School, Tokyo 113-8602, Japan
| | - Yasuyuki Negishi
- Department of Microbiology and Immunology, Nippon Medical School, Tokyo 113-8602, Japan
| | - Shohei Tozawa
- Department of Molecular Medicine and Anatomy, Nippon Medical School, Tokyo 113-8602, Japan; Department of Obstetrics and Gynecology, Jichi Medical University, Tochigi 329-0498, Japan
| | - Takami Takizawa
- Department of Molecular Medicine and Anatomy, Nippon Medical School, Tokyo 113-8602, Japan
| | - Rimpei Morita
- Department of Microbiology and Immunology, Nippon Medical School, Tokyo 113-8602, Japan
| | - Hironori Takahashi
- Department of Obstetrics and Gynecology, Jichi Medical University, Tochigi 329-0498, Japan
| | - Akihide Ohkuchi
- Department of Obstetrics and Gynecology, Jichi Medical University, Tochigi 329-0498, Japan
| | - Toshihiro Takizawa
- Department of Molecular Medicine and Anatomy, Nippon Medical School, Tokyo 113-8602, Japan.
| |
Collapse
|
32
|
Lin J, Xie Q, Xu C, Wan Q. Live Births in Women over 40 Years of Age Correlate with Obesity Rates. Endocr Metab Immune Disord Drug Targets 2024; 24:255-264. [PMID: 37282645 DOI: 10.2174/1871530323666230606120744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 04/01/2023] [Accepted: 04/14/2023] [Indexed: 06/08/2023]
Abstract
AIMS This cross-sectional study aimed to analyze the relationship between live birth and the prevalence of obesity in Chinese women over 40 years of age. METHODS From April to November 2011, the Endocrinology Branch of the Chinese Medical Association conducted the REACTION project, a national, multicenter, cross-sectional study of Chinese adults aged 40 years and older. Demographic and medical data were collected through validated questionnaires and equipment. Anthropometric indicators, blood pressure, and biochemical data were measured by professional medical personnel. Data were analyzed using descriptive statistics and logistic analysis. Multivariate regression models were used to analyze obesity-related risk factors. RESULTS The prevalence of obesity among women increased gradually from 3.8% to 6.0% with an increasing number of live births. Women with two live births had the highest prevalence of overweight at 34.3%. Overall, the obesity and overweight rates were slightly higher in premenopausal women than in postmenopausal women. Univariate regression analysis showed that the risk of obesity in women increased with an increasing number of live births. In addition, multivariate regression analysis showed that the risk of obesity increased with an increasing number of live births in women with systolic blood pressure (SBP) < 121 mmHg or current smoking (P < 0.05). CONCLUSION The risk of obesity increases with the number of live births in Chinese women over 40 years of age with SBP < 121 mmHg or current smoking. Our findings may facilitate the development of interventions to prevent obesity in this population.
Collapse
Affiliation(s)
- Jie Lin
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, China
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, China
| | - Qian Xie
- The people's Hospital of Leshan, Leshan, China
| | - Chaoran Xu
- The people's Hospital of Leshan, Leshan, China
| | - Qin Wan
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, China
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, China
| |
Collapse
|
33
|
Lingasamy P, Modhukur V, Mändar R, Salumets A. Exploring Immunome and Microbiome Interplay in Reproductive Health: Current Knowledge, Challenges, and Novel Diagnostic Tools. Semin Reprod Med 2023; 41:172-189. [PMID: 38262441 PMCID: PMC10846929 DOI: 10.1055/s-0043-1778017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2024]
Abstract
The dynamic interplay between the immunome and microbiome in reproductive health is a complex and rapidly advancing research field, holding tremendously vast possibilities for the development of reproductive medicine. This immunome-microbiome relationship influences the innate and adaptive immune responses, thereby affecting the onset and progression of reproductive disorders. However, the mechanisms governing these interactions remain elusive and require innovative approaches to gather more understanding. This comprehensive review examines the current knowledge on reproductive microbiomes across various parts of female reproductive tract, with special consideration of bidirectional interactions between microbiomes and the immune system. Additionally, it explores innate and adaptive immunity, focusing on immunoglobulin (Ig) A and IgM antibodies, their regulation, self-antigen tolerance mechanisms, and their roles in immune homeostasis. This review also highlights ongoing technological innovations in microbiota research, emphasizing the need for standardized detection and analysis methods. For instance, we evaluate the clinical utility of innovative technologies such as Phage ImmunoPrecipitation Sequencing (PhIP-Seq) and Microbial Flow Cytometry coupled to Next-Generation Sequencing (mFLOW-Seq). Despite ongoing advancements, we emphasize the need for further exploration in this field, as a deeper understanding of immunome-microbiome interactions holds promise for innovative diagnostic and therapeutic strategies for reproductive health, like infertility treatment and management of pregnancy.
Collapse
Affiliation(s)
| | - Vijayachitra Modhukur
- Competence Centre on Health Technologies, Tartu, Estonia
- Department of Obstetrics and Gynecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
| | - Reet Mändar
- Competence Centre on Health Technologies, Tartu, Estonia
- Department of Microbiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Andres Salumets
- Competence Centre on Health Technologies, Tartu, Estonia
- Department of Obstetrics and Gynecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
34
|
You Y, Grasso E, Alvero A, Condon J, Dimova T, Hu A, Ding J, Alexandrova M, Manchorova D, Dimitrova V, Liao A, Mor G. Twist1-IRF9 Interaction Is Necessary for IFN-Stimulated Gene Anti-Zika Viral Infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:1899-1912. [PMID: 37144865 PMCID: PMC10615665 DOI: 10.4049/jimmunol.2300081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 04/10/2023] [Indexed: 05/06/2023]
Abstract
An efficient immune defense against pathogens requires sufficient basal sensing mechanisms that can deliver prompt responses. Type I IFNs are protective against acute viral infections and respond to viral and bacterial infections, but their efficacy depends on constitutive basal activity that promotes the expression of downstream genes known as IFN-stimulated genes (ISGs). Type I IFNs and ISGs are constitutively produced at low quantities and yet exert profound effects essential for numerous physiological processes beyond antiviral and antimicrobial defense, including immunomodulation, cell cycle regulation, cell survival, and cell differentiation. Although the canonical response pathway for type I IFNs has been extensively characterized, less is known regarding the transcriptional regulation of constitutive ISG expression. Zika virus (ZIKV) infection is a major risk for human pregnancy complications and fetal development and depends on an appropriate IFN-β response. However, it is poorly understood how ZIKV, despite an IFN-β response, causes miscarriages. We have uncovered a mechanism for this function specifically in the context of the early antiviral response. Our results demonstrate that IFN regulatory factor (IRF9) is critical in the early response to ZIKV infection in human trophoblast. This function is contingent on IRF9 binding to Twist1. In this signaling cascade, Twist1 was not only a required partner that promotes IRF9 binding to the IFN-stimulated response element but also an upstream regulator that controls basal levels of IRF9. The absence of Twist1 renders human trophoblast cells susceptible to ZIKV infection.
Collapse
Affiliation(s)
- Yuan You
- C. S Mott Center for Human Development, Wayne State University, 275 E Hancock St, Detroit, MI, 48093
| | - Esteban Grasso
- C. S Mott Center for Human Development, Wayne State University, 275 E Hancock St, Detroit, MI, 48093
- School of Science, University of Buenos Aires, Intendente Guiraldes 2160, Buenos Aires, 1428
| | - Ayesha Alvero
- C. S Mott Center for Human Development, Wayne State University, 275 E Hancock St, Detroit, MI, 48093
| | - Jennifer Condon
- C. S Mott Center for Human Development, Wayne State University, 275 E Hancock St, Detroit, MI, 48093
| | - Tanya Dimova
- Institute of Biology and Immunology of Reproduction “Acad. K. Bratanov”, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Anna Hu
- C. S Mott Center for Human Development, Wayne State University, 275 E Hancock St, Detroit, MI, 48093
| | - Jiahui Ding
- C. S Mott Center for Human Development, Wayne State University, 275 E Hancock St, Detroit, MI, 48093
| | - Marina Alexandrova
- Institute of Biology and Immunology of Reproduction “Acad. K. Bratanov”, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Diana Manchorova
- Institute of Biology and Immunology of Reproduction “Acad. K. Bratanov”, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Violeta Dimitrova
- Institute of Biology and Immunology of Reproduction “Acad. K. Bratanov”, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Aihua Liao
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Gil Mor
- C. S Mott Center for Human Development, Wayne State University, 275 E Hancock St, Detroit, MI, 48093
| |
Collapse
|
35
|
Zhang Y, Liu Z, Sun H. Fetal-maternal interactions during pregnancy: a 'three-in-one' perspective. Front Immunol 2023; 14:1198430. [PMID: 37350956 PMCID: PMC10282753 DOI: 10.3389/fimmu.2023.1198430] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 05/24/2023] [Indexed: 06/24/2023] Open
Abstract
A successful human pregnancy requires the maternal immune system to recognize and tolerate the semi-allogeneic fetus, allowing for appropriate trophoblasts invasion and protecting the fetus from invading pathogens. Therefore, maternal immunity is critical for the establishment and maintenance of pregnancy, especially at the maternal-fetal interface. Anatomically, the maternal-fetal interface has both maternally- and fetally- derived cells, including fetal originated trophoblasts and maternal derived immune cells and stromal cells. Besides, a commensal microbiota in the uterus was supposed to aid the unique immunity in pregnancy. The appropriate crosstalk between fetal derived and maternal originated cells and uterine microbiota are critical for normal pregnancy. Dysfunctional maternal-fetal interactions might be associated with the development of pregnancy complications. This review elaborates the latest knowledge on the interactions between trophoblasts and decidual immune cells, highlighting their critical roles in maternal-fetal tolerance and pregnancy development. We also characterize the role of commensal bacteria in promoting pregnancy progression. Furthermore, this review may provide new thought on future basic research and the development of clinical applications for pregnancy complications.
Collapse
Affiliation(s)
- Yonghong Zhang
- Center for Reproductive Medicine and Obstetrics and Gynecology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Zhaozhao Liu
- Reproduction Center, The Third Affiliated Hospital of ZhengZhou University, ZhengZhou, China
| | - Haixiang Sun
- Center for Reproductive Medicine and Obstetrics and Gynecology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| |
Collapse
|
36
|
Waugh KA, Minter R, Baxter J, Chi C, Galbraith MD, Tuttle KD, Eduthan NP, Kinning KT, Andrysik Z, Araya P, Dougherty H, Dunn LN, Ludwig M, Schade KA, Tracy D, Smith KP, Granrath RE, Busquet N, Khanal S, Anderson RD, Cox LL, Estrada BE, Rachubinski AL, Lyford HR, Britton EC, Fantauzzo KA, Orlicky DJ, Matsuda JL, Song K, Cox TC, Sullivan KD, Espinosa JM. Triplication of the interferon receptor locus contributes to hallmarks of Down syndrome in a mouse model. Nat Genet 2023; 55:1034-1047. [PMID: 37277650 PMCID: PMC10260402 DOI: 10.1038/s41588-023-01399-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 04/14/2023] [Indexed: 06/07/2023]
Abstract
Down syndrome (DS), the genetic condition caused by trisomy 21, is characterized by variable cognitive impairment, immune dysregulation, dysmorphogenesis and increased prevalence of diverse co-occurring conditions. The mechanisms by which trisomy 21 causes these effects remain largely unknown. We demonstrate that triplication of the interferon receptor (IFNR) gene cluster on chromosome 21 is necessary for multiple phenotypes in a mouse model of DS. Whole-blood transcriptome analysis demonstrated that IFNR overexpression associates with chronic interferon hyperactivity and inflammation in people with DS. To define the contribution of this locus to DS phenotypes, we used genome editing to correct its copy number in a mouse model of DS, which normalized antiviral responses, prevented heart malformations, ameliorated developmental delays, improved cognition and attenuated craniofacial anomalies. Triplication of the Ifnr locus modulates hallmarks of DS in mice, suggesting that trisomy 21 elicits an interferonopathy potentially amenable to therapeutic intervention.
Collapse
Affiliation(s)
- Katherine A Waugh
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Ross Minter
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Jessica Baxter
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Congwu Chi
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Gates Center for Regenerative Medicine and Stem Cell Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- The Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Matthew D Galbraith
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kathryn D Tuttle
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Neetha P Eduthan
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kohl T Kinning
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Zdenek Andrysik
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Paula Araya
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Hannah Dougherty
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Lauren N Dunn
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Pediatrics, Section of Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Michael Ludwig
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kyndal A Schade
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Dayna Tracy
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Keith P Smith
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Ross E Granrath
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Nicolas Busquet
- Animal Behavior Core, NeuroTechnology Center, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Santosh Khanal
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Ryan D Anderson
- Department of Oral and Craniofacial Sciences, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Liza L Cox
- Department of Oral and Craniofacial Sciences, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Belinda Enriquez Estrada
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Angela L Rachubinski
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Pediatrics, Section of Developmental Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Hannah R Lyford
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Eleanor C Britton
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Katherine A Fantauzzo
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - David J Orlicky
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Jennifer L Matsuda
- Department of Immunology and Genomic Medicine, National Jewish Health, Denver, CO, USA
| | - Kunhua Song
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Gates Center for Regenerative Medicine and Stem Cell Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- The Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Timothy C Cox
- Department of Oral and Craniofacial Sciences, University of Missouri-Kansas City, Kansas City, MO, USA
- Department of Pediatrics, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Kelly D Sullivan
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
- Department of Pediatrics, Section of Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| | - Joaquin M Espinosa
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
37
|
Maxwell A, Adzibolosu N, Hu A, You Y, Stemmer PM, Ruden DM, Petriello MC, Sadagurski M, Debarba LK, Koshko L, Ramadoss J, Nguyen AT, Richards D, Liao A, Mor G, Ding J. Intrinsic sexual dimorphism in the placenta determines the differential response to benzene exposure. iScience 2023; 26:106287. [PMID: 37153445 PMCID: PMC10156617 DOI: 10.1016/j.isci.2023.106287] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 12/09/2022] [Accepted: 02/22/2023] [Indexed: 03/02/2023] Open
Abstract
Maternal immune activation (MIA) by environmental challenges is linked to severe developmental complications, such as neurocognitive disorders, autism, and even fetal/maternal death. Benzene is a major toxic compound in air pollution that affects the mother as well as the fetus and has been associated with reproductive complications. Our objective was to elucidate whether benzene exposure during gestation triggers MIA and its impact on fetal development. We report that benzene exposure during pregnancy leads MIA associated with increased fetal resorptions, fetal growth, and abnormal placenta development. Furthermore, we demonstrate the existence of a sexual dimorphic response to benzene exposure in male and female placentas. The sexual dimorphic response is a consequence of inherent differences between male and female placenta. These data provide crucial information on the origins or sexual dimorphism and how exposure to environmental factors can have a differential impact on the development of male and female offspring.
Collapse
Affiliation(s)
- Anthony Maxwell
- C.S Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, USA
| | - Nicholas Adzibolosu
- C.S Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, USA
| | - Anna Hu
- C.S Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, USA
| | - Yuan You
- C.S Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, USA
| | - Paul M. Stemmer
- Institute of Environmental Health Sciences, Wayne State University, Detroit, MI, USA
| | - Douglas M. Ruden
- C.S Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, USA
| | - Michael C. Petriello
- Institute of Environmental Health Sciences and Department of Pharmacology, Wayne State University, Detroit, MI, USA
| | - Marianna Sadagurski
- Department of Biological Sciences, Integrative Biosciences Center, Wayne State University, Detroit, MI, USA
| | - Lucas K. Debarba
- Department of Biological Sciences, Integrative Biosciences Center, Wayne State University, Detroit, MI, USA
| | - Lisa Koshko
- Department of Biological Sciences, Integrative Biosciences Center, Wayne State University, Detroit, MI, USA
| | - Jayanth Ramadoss
- C.S Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, USA
| | | | - Darby Richards
- School of Medicine, Wayne State University, Detroit, MI, USA
| | - Aihua Liao
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Gil Mor
- C.S Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, USA
| | - Jiahui Ding
- C.S Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, USA
| |
Collapse
|
38
|
Arfuso F, Minuti A, Liotta L, Giannetto C, Trevisi E, Piccione G, Lopreiato V. Stress and inflammatory response of cows and their calves during peripartum and early neonatal period. Theriogenology 2023; 196:157-166. [PMID: 36423510 DOI: 10.1016/j.theriogenology.2022.11.019] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 11/09/2022] [Accepted: 11/10/2022] [Indexed: 11/16/2022]
Abstract
Stress, inflammatory response, and their relationship were investigated in Simmental cows during the transition period (N = 8; 5 multiparous and 3 primiparous) and in their calves (N = 8; 5 heifers and 3 bulls). From cows, blood was collected at days -21 (±4), 0, +1, +7, and +21 days relative to calving. From calves, blood was collected after birth before colostrum intake (0) and then at 1, 7, and 15 days of age. Cortisol, Interleukin 6 (IL-6) and haptoglobin concentration was assessed by ELISA technique; white blood cells (WBC) were assessed using an ADVIA 2120 Hematology System machine. One-way ANOVA showed an effect of time for all the investigated parameters (P < 0.001) except for lymphocytes in peripartal cows. At calving and 1 d after, cortisol concentration was negatively correlated with levels of IL-6, WBC, and monocytes, whereas levels of IL-6 were positively correlated with WBC, neutrophils, and monocytes count. Cortisol, IL-6, haptoglobin, WBC and all leukocyte populations were affected by the age of neonatal calves (P < 0.001) except for neutrophils. A negative correlation between cortisol and IL-6, neutrophils, monocytes and haptoglobin was found at 15 days of age. A positive correlation between IL-6 and haptoglobin at day 15 of age, and with neutrophils and monocytes at days 7 and 15 of age was found. A positive correlation was obtained between cortisol levels measured in cows around calving and those obtained in calves after birth before colostrum intake (r = 0.83), and between IL-6 concentrations obtained from cows at calving and 1 d after and those obtained in calves at day 1 of age, after the colostrum intake (r = 0.93 and 0.79, respectively). The study suggests that immune function of peripartal cows is in an active state and that, in addition to other well-known factors driving the changes of parameters herein investigated, cortisol could have a role in the immune-modulatory adjustment during peripartum in cows. Furthermore, it can be hypothesized that cortisol is transferred from the cow to newborn calf through the placenta only and not through colostrum, whereas IL-6 levels in calves during the 24 h after birth seem to be influenced by IL-6 values measured in cows around calving due to its transfer through colostrum.
Collapse
Affiliation(s)
- Francesca Arfuso
- Department of Veterinary Sciences, University of Messina, Polo University Annunziata, 98168, Messina, Italy
| | - Andrea Minuti
- Department of Animal Sciences, Food and Nutrition (DiANA), Faculty of Agriculture, Food and Environmental Sciences, Università Cattolica del Sacro Cuore, 29122, Piacenza, Italy
| | - Luigi Liotta
- Department of Veterinary Sciences, University of Messina, Polo University Annunziata, 98168, Messina, Italy
| | - Claudia Giannetto
- Department of Veterinary Sciences, University of Messina, Polo University Annunziata, 98168, Messina, Italy.
| | - Erminio Trevisi
- Department of Animal Sciences, Food and Nutrition (DiANA), Faculty of Agriculture, Food and Environmental Sciences, Università Cattolica del Sacro Cuore, 29122, Piacenza, Italy
| | - Giuseppe Piccione
- Department of Veterinary Sciences, University of Messina, Polo University Annunziata, 98168, Messina, Italy
| | - Vincenzo Lopreiato
- Department of Veterinary Sciences, University of Messina, Polo University Annunziata, 98168, Messina, Italy
| |
Collapse
|
39
|
Ju L, Li C, Hua L, Xu H, Hu Y, Zhou X, Sun S, Zhang Q, Cheng H, Yang M, Cao J, Ding R. Uterine decidual stromal cell-derived exosomes mediate the indirect effects of 1-nitropyrene on trophoblast biological behaviors. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 248:114288. [PMID: 36371887 DOI: 10.1016/j.ecoenv.2022.114288] [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: 08/17/2022] [Revised: 11/01/2022] [Accepted: 11/09/2022] [Indexed: 06/16/2023]
Abstract
1-nitropyrene (1-NP) is representative nitropolycyclic aromatic hydrocarbon pollutant widely present in exhaust particles of internal combustion engine, which is known for its carcinogenicity and mutagenicity. Previous studies have demonstrated that 1-NP has reproductive toxicity, but the specific mechanism is unknown. In this study, Human decidual stromal cells (HDSCs) were treated by 1-NP, exosomes were extracted from the conditioned medium of HDSCs, which were then used to treat human chorionic trophoblast cells (HTR8/SVneo) for 24 h. The findings showed that human decidual stromal cell-derived exosomes (HDSC-EXOs) can promote the proliferation, migration, invasion and epithelial-mesenchymal transition (EMT; Vimentin and N-cadherin) of HTR8/SVneo by about 64%, 17%, 23%, 81% and 13%. The process of regulating the biological behaviors of embryonic trophoblast cells by maternal decidual stromal cells during pregnancy was simulated. Further investigations showed that HDSC-EXOs treatment activated the Wnt/β-catenin signaling pathway in HTR8/SVneo. Co-treatment by dickkopf-1 (DKK-1) significantly suppressed the activation of Wnt/β-catenin signaling pathway in HTR8/SVneo, and inhibited the proliferation, migration, invasion and EMT (N-cadherin and E-cadherin) of HTR8/SVneo by about 60%, 22%, 42%, 25%, 55% and 21%. These findings indicated that 1-NP exposure could induce the secretion of HDSC-EXOs from HDSCs, which in turn activate the Wnt/β-catenin signaling pathway and enhance the proliferation, migration, invasion and EMT of HTR8/SVneo.
Collapse
Affiliation(s)
- Liangliang Ju
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China.
| | - Changlian Li
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China; Heifei Center for Disease Control and Prevention, Hefei, Anhui, China.
| | - Lei Hua
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China.
| | - Hanbing Xu
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China.
| | - Yingyu Hu
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China.
| | - Xinyu Zhou
- The First Clinical College of Anhui Medical University, Hefei, Anhui 230032, China.
| | - Shu Sun
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China.
| | - Qi Zhang
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China.
| | - Han Cheng
- The First Clinical College of Anhui Medical University, Hefei, Anhui 230032, China.
| | - Mingwei Yang
- Department of Radiation Oncology, The First Affiliated Hospital of Anhui Medical University, Heifei, Anhui, China.
| | - Jiyu Cao
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China; Department of Teaching Center for Preventive Medicine, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China.
| | - Rui Ding
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China.
| |
Collapse
|