1
|
Sehgal N, Li L, Goin DE, Chen J, Jigmeddagva U, Morello-Frosch R, Woodruff TJ, Gaw SL, Robinson JF, Eick SM. Psychosocial stress and associations with inflammation in mid-gestation maternal, fetal, and placental tissue. Reprod Toxicol 2025; 135:108922. [PMID: 40254104 DOI: 10.1016/j.reprotox.2025.108922] [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: 01/28/2025] [Revised: 04/14/2025] [Accepted: 04/15/2025] [Indexed: 04/22/2025]
Abstract
BACKGROUND Inflammation has been implicated as an intermediary between psychosocial stress and adverse birth outcomes. However, prior work has mostly relied on maternal inflammation as a proxy for fetal inflammation mid-gestation or measured fetal inflammation in cord blood and placenta obtained at delivery. No studies have examined psychosocial stress in relation to fetal inflammation mid-gestation. METHODS Twenty cytokines were measured in matched maternal blood, cord blood, and placenta obtained mid-gestation from a socio-demographically diverse group of pregnant participants undergoing elective second-trimester pregnancy terminations (N = 106). Corticotropin-releasing hormone, a proposed biomarker of gestational length, was measured in maternal blood. Perceived stress, and exposure to stressful life events, job strain, and social support were measured via questionnaires. We used linear regression to estimate associations between individual stressors and inflammatory biomarkers in each biomatrix and principal component analysis to assess groups of inflammatory biomarkers. RESULTS We observed many matrix-specific associations between psychosocial stressors and inflammatory biomarkers. For example, low versus high social support was associated with significantly decreased levels of maternal blood CCL3 (β=-0.53; 95 % confidence interval [CI]=-0.98,-0.07), CCL4 (β=-0.26; 95 % CI=-0.47,-0.04), IL8 (β=-0.79; 95 % CI=-1.47,-0.11), CXCL9 (β=-0.47; 95 % CI=-0.89,-0.06), IFNγ (β=-2.28; 95 % CI=-3.60,-0.96), IL4 (β=-1.07; 95 % CI=-1.88,-0.26); and cord blood IFNγ (β=-0.83; 95 % CI=-1.52,-0.14). Social support was not associated with placental inflammation. CONCLUSIONS During mid-pregnancy, psychosocial stress─ particularly low social support─ was associated with maternal blood levels of select cytokines, suggesting a potential pathway linking social stress and inflammation. Our results indicate that the placenta may buffer these inflammatory effects on the fetus.
Collapse
Affiliation(s)
- Neha Sehgal
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Lin Li
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, CA, USA
| | - Dana E Goin
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Jessica Chen
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, CA, USA
| | - Unurzul Jigmeddagva
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, CA, USA
| | - Rachel Morello-Frosch
- Program on Reproductive Health and the Environment, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, CA, USA; Department of Environmental Science, Policy and Management and School of Public Health, University of California, Berkeley, CA, USA
| | - Tracey J Woodruff
- Program on Reproductive Health and the Environment, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, CA, USA
| | - Stephanie L Gaw
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, CA, USA; Division of Maternal Fetal Medicine, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, CA, USA
| | - Joshua F Robinson
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, CA, USA; Program on Reproductive Health and the Environment, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, CA, USA
| | - Stephanie M Eick
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA.
| |
Collapse
|
2
|
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: 0] [Impact Index Per Article: 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
|
3
|
Wang J, Sun Y, Wu R. ACSM3 Suppresses Ovarian Cancer Progression by Inactivating the IFN-γ/JAK/STAT3 Signaling Pathway. Adv Biol (Weinh) 2025; 9:e2400093. [PMID: 39913127 DOI: 10.1002/adbi.202400093] [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: 02/19/2024] [Revised: 11/12/2024] [Indexed: 02/07/2025]
Abstract
This study aimed to investigate the role of Acyl-CoA medium-chain synthetase 3 (ACSM3) in ovarian cancer (OC) progression. ACSM3 expression in OC tissues and cells is detected by real-time quantitative polymerase chain reaction, immunohistochemistry, or western blot. The influences of ACSM3 on OC progression are assessed in vitro and in vivo experiments. Gene set enrichment analysis is performed to find significantly enriched pathways related to ACSM3. In this study, ACSM3 expression in OC tissues and cells is downregulated. ACSM3 inhibited tumor growth in vivo. Knockdown of ACSM3 promoted cell proliferation, migration, and invasion, inhibited cell apoptosis, and activated JAK/STAT3 signaling pathway in SKOV3 cells, while overexpression of ACSM3 in A2780 cells led to the opposite results. Moreover, treatment with interferon-gamma (IFN-γ) in A2780 cells reversed the effects of ACSM3 on cell proliferation, migration, invasion, and apoptosis, but IFN-γ further enhanced the effects of ACSM3 knockdown on SKOV3 cell proliferation, migration, invasion, and apoptosis. In conclusion, ACSM3 inhibited OC cell proliferation, migration, and invasion, and promoted cell apoptosis by suppressing the IFN-γ/JAK/STAT3 signaling pathway, which might provide a promising therapeutic target for OC treatment.
Collapse
Affiliation(s)
- Juan Wang
- Department of Gynecology, Bin Zhou Medical University Hospital, Binzhou, Shandong, 256603, P. R. China
| | - Yanqiu Sun
- Department of obstetrics and gynecology, Bin Zhou Medical University Hospital, Binzhou, Shandong, 256603, P. R. China
| | - Ruixue Wu
- Department of Gynecology, Bin Zhou Medical University Hospital, Binzhou, Shandong, 256603, P. R. China
| |
Collapse
|
4
|
Liu Z, Zhang J, Li L, Zhang T, Huang L, Yin Q. Shoutai Pill Enhances Endometrial Receptivity in Controlled Ovarian Hyperstimulation Mice by Improving the In-Vivo Immune Environment. Comb Chem High Throughput Screen 2025; 28:711-723. [PMID: 37929727 DOI: 10.2174/0113862073274708231028185333] [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: 07/29/2023] [Revised: 09/07/2023] [Accepted: 09/22/2023] [Indexed: 11/07/2023]
Abstract
BACKGROUND The Shoutai pill (STP) is a classic formulation in traditional Chinese medicine. Preliminary experimental observations from our study suggest that it is effective in enhancing endometrial receptivity. However, the underlying mechanisms by which STP influences endometrial receptivity remain to be elucidated. OBJECTIVE The objective of this study is to investigate the effects and mechanisms of the STP formulation in enhancing endometrial receptivity in controlled ovarian hyperstimulation (COH) model mice. METHODS The network pharmacology analysis identified target proteins associated with the reduction of endometrial receptivity by STP. The COH mouse model was established using the GnRHa+PMSG+HCG protocol. The levels of MHC-1 and MHC-2 in mouse serum were measured using the ELISA method, while the levels of IL-1β, IL-4, IL-10, IP-10, IL-1a, IL-2, IL-17, TNF-a, and IFN-y were measured using liquid chip technology. RESULTS STP exhibited a significant improvement in the immune environment of COH model mice. The major active components of STP were identified as beta-sitosterol and quercetin, among others. Furthermore, AKT1, VEGFA, and several immune factors, such as TNF, IFN, IL- 1β, and IL-10, were identified as key targets for regulating endometrial receptivity. STP enhanced the expression of IL-10, IL-4, and IP-10 in the mice while reducing the expression levels of IL-2, IL-17, TNF-α, and IFN-γ in COH mice. These effects led to the modulation of early high expression of IL-1β and an improvement in endometrial receptivity. CONCLUSION This study demonstrates that STP can modulate in-vivo immune factors throughout the COH process, subsequently restoring the immune equilibrium within the endometrium, thereby enhancing the endometrial receptivity in the COH model mice.
Collapse
Affiliation(s)
- Ziping Liu
- Chengdu University of Traditional Chinese Medicine, Chengdu Sichuan, China
| | - Jizhong Zhang
- Southwest University for Nationalities, Chengdu Sichuan, China
| | - Liming Li
- 3Sichuan Academy of Traditional Chinese Medicine Science, Chengdu Sichuan China
| | - Tiane Zhang
- Chengdu University of Traditional Chinese Medicine, Chengdu Sichuan, China
| | - Li Huang
- Sichuan Academy of Traditional Chinese Medicine Science, Chengdu Sichuan China
| | - Qiaozhi Yin
- Chengdu University of Traditional Chinese Medicine, Chengdu Sichuan, China
| |
Collapse
|
5
|
Li J, Zhu X, Zhu W, Li L, Wei H, Zhang S. Research Progress on the Impact of Human Chorionic Gonadotropin on Reproductive Performance in Sows. Animals (Basel) 2024; 14:3266. [PMID: 39595318 PMCID: PMC11591456 DOI: 10.3390/ani14223266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 11/01/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
Human chorionic gonadotropin is a glycoprotein hormone produced by human or humanoid syncytiotrophoblasts that differentiate during pregnancy. Due to its superior stability and long-lasting effects compared to luteinizing hormone, it is often used to replace luteinizing hormone to regulate reproductive performance in sows. Human chorionic gonadotropin promotes oocyte maturation, follicle development, and luteinization, thereby increasing conception rates and supporting early embryonic development. In sow reproductive management, the application of human chorionic gonadotropin not only enhances ovulation synchrony but also improves the success rate of embryo implantation by regulating endometrial receptivity and immune mechanisms, significantly enhancing overall reproductive performance. This article primarily reviews the application of human chorionic gonadotropin in sow follicle development, luteal maintenance, and embryo implantation, providing theoretical support for its use in improving reproductive performance in sows.
Collapse
Affiliation(s)
| | | | | | | | | | - Shouquan Zhang
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agroanimal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510640, China; (J.L.); (X.Z.); (W.Z.); (L.L.); (H.W.)
| |
Collapse
|
6
|
Moghaddami R, Mahdipour M, Ahmadpour E. Inflammatory pathways of Toxoplasmagondii infection in pregnancy. Travel Med Infect Dis 2024; 62:102760. [PMID: 39293589 DOI: 10.1016/j.tmaid.2024.102760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 08/07/2024] [Accepted: 09/13/2024] [Indexed: 09/20/2024]
Abstract
Toxoplasma gondii (T. gondii), an obligate intracellular parasite, is considered as an opportunistic infection and causes toxoplasmosis in humans and animals. Congenital toxoplasmosis can influence pregnancy and cause mild to severe consequences for the fetal and neonatal. During early T. gondii infection, neutrophils as the most abundant white blood cells provide a front line of defense mechanism against infection. The activated dendritic cells are then responsible for initiating an inflammatory response via T-helper 1 (Th1) cells. As part of its robust immune response, the infected host cells produce interferon (IFN-γ). IFN-γ inhibits T. gondii replication and promotes its transformation from an active form to tissue cysts. Although anti- T. gondii antibodies play an important role in infection control, T-helper 2 (Th2) immune response, can facilitate the growth and proliferation of T. gondii in the host cell. In pregnant women infected with T. gondii, the expression of cytokines may vary and in response diverse outcomes are expected. Cytokine profiles serve as valuable indicators for estimating the patho-immunological effects of T. gondii infection. This demonstrates the intricate relationship between pro-inflammatory and anti-inflammatory cytokines, as well as their influence on the various pregnancy outcomes in T. gondii infection.
Collapse
Affiliation(s)
- Reyhaneh Moghaddami
- Department of Plant, Cell and Molecular Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Mahdi Mahdipour
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ehsan Ahmadpour
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
7
|
Wu Y, Su K, Zhang Y, Liang L, Wang F, Chen S, Gao L, Zheng Q, Li C, Su Y, Mao Y, Zhu S, Chai C, Lan Q, Zhai M, Jin X, Zhang J, Xu X, Zhang Y, Gao Y, Huang H. A spatiotemporal transcriptomic atlas of mouse placentation. Cell Discov 2024; 10:110. [PMID: 39438452 PMCID: PMC11496649 DOI: 10.1038/s41421-024-00740-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 09/10/2024] [Indexed: 10/25/2024] Open
Abstract
The placenta, a temporary but essential organ for gestational support, undergoes intricate morphological and functional transformations throughout gestation. However, the spatiotemporal patterns of gene expression underlying placentation remain poorly understood. Utilizing Stereo-seq, we constructed a Mouse Placentation Spatiotemporal Transcriptomic Atlas (MPSTA) spanning from embryonic day (E) 7.5 to E14.5, which includes the transcriptomes of large trophoblast cells that were not captured in previous single-cell atlases. We defined four distinct strata of the ectoplacental cone, an early heterogeneous trophectoderm structure, and elucidated the spatial trajectory of trophoblast differentiation during early postimplantation stages before E9.5. Focusing on the labyrinth region, the interface of nutrient exchange in the mouse placenta, our spatiotemporal ligand-receptor interaction analysis unveiled pivotal modulators essential for trophoblast development and placental angiogenesis. We also found that paternally expressed genes are exclusively enriched in the placenta rather than in the decidual regions, including a cluster of genes enriched in endothelial cells that may function in placental angiogenesis. At the invasion front, we identified interface-specific transcription factor regulons, such as Atf3, Jun, Junb, Stat6, Mxd1, Maff, Fos, and Irf7, involved in gestational maintenance. Additionally, we revealed that maternal high-fat diet exposure preferentially affects this interface, exacerbating inflammatory responses and disrupting angiogenic homeostasis. Collectively, our findings furnish a comprehensive, spatially resolved atlas that offers valuable insights and benchmarks for future explorations into placental morphogenesis and pathology.
Collapse
Affiliation(s)
- Yanting Wu
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China.
- Key Laboratory of Reproductive Genetics (Ministry of Education), Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences (No. 2019RU056), Shanghai, China.
- Shanghai Key Laboratory of Reproduction and Development, Shanghai, China.
| | - Kaizhen Su
- Key Laboratory of Reproductive Genetics (Ministry of Education), Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ying Zhang
- BGI Research, Shenzhen, Guangdong, China
- Shanxi Medical University - BGI Collaborative Center for Future Medicine, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Langchao Liang
- BGI Research, Qingdao, Shandong, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Fei Wang
- BGI Research, Shenzhen, Guangdong, China
| | - Siyue Chen
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China
| | - Ling Gao
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China
| | - Qiutong Zheng
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China
| | - Cheng Li
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China
| | - Yunfei Su
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China
| | - Yiting Mao
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China
| | - Simeng Zhu
- Department of Cardiology, Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chaochao Chai
- BGI Research, Qingdao, Shandong, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Qing Lan
- BGI Research, Shenzhen, Guangdong, China
| | - Man Zhai
- BGI Research, Shenzhen, Guangdong, China
| | - Xin Jin
- BGI Research, Shenzhen, Guangdong, China
| | - Jinglan Zhang
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences (No. 2019RU056), Shanghai, China
- Shanghai Key Laboratory of Reproduction and Development, Shanghai, China
| | - Xun Xu
- BGI Research, Shenzhen, Guangdong, China
- Guangdong Provincial Key Laboratory of Genome Read and Write, Shenzhen, Guangdong, China
| | - Yu Zhang
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China.
| | - Ya Gao
- BGI Research, Shenzhen, Guangdong, China.
- Shanxi Medical University - BGI Collaborative Center for Future Medicine, Shanxi Medical University, Taiyuan, Shanxi, China.
- Shenzhen Engineering Laboratory for Birth Defects Screening, BGI Research, Shenzhen, Guangdong, China.
| | - Hefeng Huang
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China.
- Key Laboratory of Reproductive Genetics (Ministry of Education), Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences (No. 2019RU056), Shanghai, China.
- Shanghai Key Laboratory of Reproduction and Development, Shanghai, China.
- Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
8
|
Stavros S, Panagopoulos P, Machairiotis N, Potiris A, Mavrogianni D, Sfakianakis A, Drakaki E, Christodoulaki C, Panagiotopoulos D, Sioutis D, Karampitsakos T, Antonakopoulos N, Christopoulos P, Drakakis P. Association between cytokine polymorphisms and recurrent pregnancy loss: A review of current evidence. Int J Gynaecol Obstet 2024; 167:45-57. [PMID: 38706379 DOI: 10.1002/ijgo.15575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 04/09/2024] [Accepted: 04/20/2024] [Indexed: 05/07/2024]
Abstract
Cytokines are a type of protein that play an important role in the immune response and can also affect many physiological processes in the body. Cytokine polymorphisms refer to genetic variations or mutations that occur within the genes that code for cytokines, which may affect the level of cytokine production and function. Some cytokine polymorphisms have been associated with an increased risk of developing certain diseases, while others may be protective or have no significant effect on health. In recent years, the role of cytokine polymorphisms in the development of recurrent pregnancy loss (RPL) has been studied. RPL or miscarriage is defined as the occurrence of two or more consecutive pregnancy losses before the 20th week of gestation. There are diverse causes leading to RPL, including genetic, anatomical, hormonal, and immunological factors. With regard to cytokine polymorphisms, a few of them have been found to be associated with an increased risk of RPL, for instance, variations in the genes that code for interleukin-6, tumor necrosis factor-alpha, and interleukin-10. The exact mechanisms by which cytokine polymorphisms affect the risk of recurrent miscarriage are still being studied, and further research is essential to fully understand this complex condition. This brief review aims to summarize the recent literature on the association between cytokine polymorphisms and RPL.
Collapse
Affiliation(s)
- Sofoklis Stavros
- Third Department of Obstetrics and Gynecology, University General Hospital "ATTIKON", Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Periklis Panagopoulos
- Third Department of Obstetrics and Gynecology, University General Hospital "ATTIKON", Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Nikolaos Machairiotis
- Third Department of Obstetrics and Gynecology, University General Hospital "ATTIKON", Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Anastasios Potiris
- Third Department of Obstetrics and Gynecology, University General Hospital "ATTIKON", Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Despoina Mavrogianni
- First Department of Obstetrics and Gynecology, Alexandra Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Eirini Drakaki
- First Department of Obstetrics and Gynecology, Alexandra Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Chrysi Christodoulaki
- Department of Obstetrics and Gynecology, Chania General Hospital "St. George", Chania, Greece
| | - Dimitrios Panagiotopoulos
- Third Department of Obstetrics and Gynecology, University General Hospital "ATTIKON", Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Dimos Sioutis
- Third Department of Obstetrics and Gynecology, University General Hospital "ATTIKON", Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Theodoros Karampitsakos
- Third Department of Obstetrics and Gynecology, University General Hospital "ATTIKON", Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Panagiotis Christopoulos
- Second Department of Obstetrics and Gynecology, University Hospital Aretaieion, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Peter Drakakis
- Third Department of Obstetrics and Gynecology, University General Hospital "ATTIKON", Medical School, National and Kapodistrian University of Athens, Athens, Greece
- First Department of Obstetrics and Gynecology, Alexandra Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
9
|
Peralta MB, Cainelli S, Stassi AF, Angeli E, Rey F, Ortega HH, Salvetti NR, Velázquez M. Association between endometrial protein expression of IFN-γ and delayed conception after parturition in dairy cows. Vet Res Commun 2024; 48:3187-3196. [PMID: 39141287 DOI: 10.1007/s11259-024-10490-1] [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: 04/23/2024] [Accepted: 08/01/2024] [Indexed: 08/15/2024]
Abstract
The cytokine context present in the reproductive tract of cows is closely involved in normal uterine functions, including the estrous cycle and the establishment and maintenance of pregnancy. However, the roles of some cytokines in the uterus, and their relation with reproductive performance remain to be elucidated. Thus, this study aimed to examine the protein expression of several cytokines such as TNFα, IL-6, IL-8, IFNγ, IL-4, and TGF-β3 in endometrial biopsies previous to conception, to evaluate the possible association with delayed conception in dairy cows. Protein expression levels were evaluated by immunohistochemistry. Results showed that the protein expression levels of TNFα, IL-6, IL-4 and TGF-β3 were not associated with the parturition-conception interval, whereas the high protein expression levels of IFNγ were associated with the parturition-conception interval. Finally, the low protein expression of IL-8 showed a statistical tendency to be associated with delayed conception. This is the first report about the protein expression of IFN-γ in the endometrium of dairy cows and also, this cytokine could enhance the favorable conditions to achieve an early pregnancy.
Collapse
Affiliation(s)
- M B Peralta
- Laboratorio de Biología Celular y Molecular Aplicada, Instituto de Ciencias Veterinarias del Litoral (ICiVet-Litoral), Universidad Nacional del Litoral (UNL), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Esperanza, Santa Fe, Argentina
| | - S Cainelli
- Laboratorio de Biología Celular y Molecular Aplicada, Instituto de Ciencias Veterinarias del Litoral (ICiVet-Litoral), Universidad Nacional del Litoral (UNL), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Esperanza, Santa Fe, Argentina
| | - A F Stassi
- Laboratorio de Biología Celular y Molecular Aplicada, Instituto de Ciencias Veterinarias del Litoral (ICiVet-Litoral), Universidad Nacional del Litoral (UNL), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Esperanza, Santa Fe, Argentina
- Facultad de Ciencias Veterinarias, Universidad Nacional del Litoral (UNL), Esperanza, Santa Fe, Argentina
| | - E Angeli
- Laboratorio de Biología Celular y Molecular Aplicada, Instituto de Ciencias Veterinarias del Litoral (ICiVet-Litoral), Universidad Nacional del Litoral (UNL), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Esperanza, Santa Fe, Argentina
- Facultad de Ciencias Veterinarias, Universidad Nacional del Litoral (UNL), Esperanza, Santa Fe, Argentina
| | - F Rey
- Laboratorio de Biología Celular y Molecular Aplicada, Instituto de Ciencias Veterinarias del Litoral (ICiVet-Litoral), Universidad Nacional del Litoral (UNL), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Esperanza, Santa Fe, Argentina
- Facultad de Ciencias Veterinarias, Universidad Nacional del Litoral (UNL), Esperanza, Santa Fe, Argentina
| | - H H Ortega
- Laboratorio de Biología Celular y Molecular Aplicada, Instituto de Ciencias Veterinarias del Litoral (ICiVet-Litoral), Universidad Nacional del Litoral (UNL), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Esperanza, Santa Fe, Argentina
- Facultad de Ciencias Veterinarias, Universidad Nacional del Litoral (UNL), Esperanza, Santa Fe, Argentina
| | - N R Salvetti
- Laboratorio de Biología Celular y Molecular Aplicada, Instituto de Ciencias Veterinarias del Litoral (ICiVet-Litoral), Universidad Nacional del Litoral (UNL), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Esperanza, Santa Fe, Argentina
- Facultad de Ciencias Veterinarias, Universidad Nacional del Litoral (UNL), Esperanza, Santa Fe, Argentina
| | - Mml Velázquez
- Facultad de Ciencias Veterinarias, Universidad Nacional del Litoral (UNL), Esperanza, Santa Fe, Argentina.
- Instituto de Ecología Humana y Desarrollo Sustentable, Universidad Católica de Santa Fe (UCSF). Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Cngo. Echagüe 7151, Santa Fe, Argentina.
| |
Collapse
|
10
|
Singh S, Goel I, Tripathi S, Ahirwar A, Kumar M, Rana A, Dhar R, Karmakar S. Effect of environmental air pollutants on placental function and pregnancy outcomes: a molecular insight. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2024; 31:59819-59851. [PMID: 39388084 DOI: 10.1007/s11356-024-35016-9] [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: 06/26/2024] [Accepted: 09/13/2024] [Indexed: 10/15/2024]
Abstract
Air pollution has become a major health concern, particularly for vulnerable populations such as the elderly, children, and pregnant women. Studies have reported a strong association between prenatal exposure to air pollutants and adverse pregnancy outcomes, including lower birth weight, reduced fetal growth, and an increased frequency of preterm births. This review summarizes the harmful effects of air pollutants, such as particulate matter, on pregnancy and outlines the mechanistic details associated with these adverse outcomes. Particulate pollutant matter may be able to cross the placenta barrier, and alterations in placental functions are central to the detrimental effects of these pollutants. In addition to associations with preeclampsia and gestational hypertension, air pollutants also induce oxidative stress, inflammation, and epigenetic alteration in the placenta. These pollutants can also affect placental homeostasis and endocrine function, contributing to pregnancy complications and possible transgenerational effects. Prenatal air pollution exposure has been linked to reduced cognitive and motor function in infants and newborns, increasing the predisposition to autism spectrum disorders and other neuropsychiatric disorders. This review also summarizes the use of various animal models to study the harmful effects of air pollution on pregnancy and postnatal outcomes. These findings provide valuable insight into the molecular events associated with the process and can aid in risk mitigation and adopting safety measures. Implementing effective environmental protocols and taking appropriate steps may reduce the global disease burden, particularly for developing nations with poor regulatory compliance and large populations of pregnant women.
Collapse
Affiliation(s)
- Sunil Singh
- Department of Biochemistry, All India Institute of Medical Sciences, Room 3020, New Delhi, 110029, India
| | - Isha Goel
- Department of Psychiatry, All India Institute of Medical Sciences, New Delhi, India
| | - Smita Tripathi
- Department of Biochemistry, Lady Harding Medical College, New Delhi, India
| | - Ashok Ahirwar
- Department of Lab Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Megha Kumar
- CSIR-Centre for Cellular and Molecular Biology (CSIR-CCMB), Habsiguda, Hyderabad, India
| | - Anubhuti Rana
- Department of Obstetrics and Gynaecology, All India Institute of Medical Sciences, New Delhi, India
| | - Ruby Dhar
- Department of Biochemistry, All India Institute of Medical Sciences, Room 3020, New Delhi, 110029, India
| | - Subhradip Karmakar
- Department of Biochemistry, All India Institute of Medical Sciences, Room 3020, New Delhi, 110029, India.
| |
Collapse
|
11
|
Abdelbaky HH, Shimoda N, Akthar I, Nakamura S, Hasan MH, Ushio N, Miyamoto A, Nishikawa Y. In vitro regulation of gene expression of pregnancy-associated proteins and cytokines in bovine endometrial epithelial cells and bovine trophoblastic cells by infection with Neospora caninum. Parasitol Int 2024; 101:102898. [PMID: 38648880 DOI: 10.1016/j.parint.2024.102898] [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: 10/18/2023] [Revised: 04/17/2024] [Accepted: 04/19/2024] [Indexed: 04/25/2024]
Abstract
Abortion caused by the parasite Neospora caninum is an important threat to the livestock industry worldwide. Trophoblasts and caruncular cells play major roles in initiating innate immune responses and controlling parasite infection at the fetal-maternal interface. In the present study, bovine uterine epithelial cells (BUECs) and bovine trophoblastic (BT) cells treated with bovine interferon-gamma (IFN-γ), IFN-alpha (IFN-α) and IFN-tau (IFN-τ) followed by infection with N. caninum were examined by measuring the mRNA expression levels of numerous pregnancy-associated proteins and observing parasite growth to elucidate the host-parasite interaction at the uteroplacental region. N. caninum infection increased the expression of prolactin-related protein 1 (PRP1), pregnancy-associated glycoprotein 1 (PAG1), and cytokines (TNF-α, IL-8 and IL-10) in BUECs and of IL-8 in BT cells. Bovine IFN-γ inhibited IL-8 and TNF-α expression in BUECs and IL-8 in BT cells. In contrast, the expression of the interferon-stimulated gene OAS1 was significantly increased by treatment of the infected BT cells with IFN-γ. However, treatment with bovine IFNs did not inhibit N. caninum growth in either cell line. In conclusion, our results suggest that bovine IFN-γ plays a crucial role in control of pathogenesis in uterus and induction of inflammatory response in the placental region following N. caninum infection, rather than growth inhibition of the parasites.
Collapse
Affiliation(s)
- Hanan H Abdelbaky
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-Cho, Obihiro, Hokkaido 080-8555, Japan
| | - Naomi Shimoda
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-Cho, Obihiro, Hokkaido 080-8555, Japan
| | - Ihshan Akthar
- Global AgroMedicine Research Center (GAMRC), Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan
| | - Shu Nakamura
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-Cho, Obihiro, Hokkaido 080-8555, Japan
| | - Md Hasibul Hasan
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-Cho, Obihiro, Hokkaido 080-8555, Japan
| | - Nanako Ushio
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-Cho, Obihiro, Hokkaido 080-8555, Japan
| | - Akio Miyamoto
- Global AgroMedicine Research Center (GAMRC), Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan
| | - Yoshifumi Nishikawa
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-Cho, Obihiro, Hokkaido 080-8555, Japan.
| |
Collapse
|
12
|
Wang H, Wang L, Gong G, Lin X, Luo J, Liu C, Mor G, Liao A. Interleukin-10: a novel metabolic inducer of macrophage differentiation and subsequently contributing to improved pregnancy outcomes of mice by orchestrating oxidative phosphorylation metabolism†. Biol Reprod 2024; 111:76-91. [PMID: 38501817 PMCID: PMC11466864 DOI: 10.1093/biolre/ioae041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 12/14/2023] [Accepted: 03/08/2024] [Indexed: 03/20/2024] Open
Abstract
Metabolism regulates the phenotype and function of macrophages. After recruitment to local tissues, monocytes are influenced by the local microenvironment and differentiate into various macrophages depending on different metabolic pathways. However, the metabolic mechanisms underlying decidual macrophage differentiation remain unknown. Interleukin-10 (IL-10) is an important decidual macrophage inducer and promotes oxidative phosphorylation (OXPHOS) of bone marrow-derived macrophages. In this study, we mainly investigate the metabolic changes involved in IL-10-generated macrophages from monocytes using in vitro models. We demonstrate that exposure of monocytes (either peripheral or THP-1) to IL-10 altered the phenotype and function of resultant macrophages that are linked with OXPHOS changes. Interleukin-10 enhanced the mitochondrial complex I and III activity of THP-1 cell-differentiated macrophages and increased the mitochondrial membrane potential, intracellular adenosine triphosphate, and reactive oxygen species levels. Oxidative phosphorylation blockage with oligomycin changed the cell morphology of IL-10-generated macrophages and the expression levels of cytokines, such as transforming growth factor beta, tumor necrosis factor-alpha, interferon gamma, and IL-10, apart from changes in the expression level of the surface markers CD206, CD209, and CD163. Moreover, in vivo IL-10 administration reduced the lipopolysaccharide (LPS)-induced embryo resorption rate, and this effect was diminished when OXPHOS was inhibited, demonstrating that OXPHOS is important for the improved pregnancy outcomes of IL-10 in LPS-induced abortion-prone mice. Our findings provide deep insights into the roles of IL-10 in macrophage biology and pregnancy maintenance. Nevertheless, the direct evidence that OXPHOS is involved in decidual macrophage differentiation needs further investigations.
Collapse
Affiliation(s)
- Huan Wang
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P.R. China
| | - Liling Wang
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P.R. China
| | - Guangshun Gong
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P.R. China
| | - Xinxiu Lin
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P.R. China
| | - Jing Luo
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P.R. China
| | - Chunyan Liu
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P.R. China
| | - Gil Mor
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P.R. China
- C.S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Aihua Liao
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P.R. China
| |
Collapse
|
13
|
Piemonti L, Vettor L, Contro E. A Case Report of Metastatic Gastric Cancer Treated with Pembrolizumab during Pregnancy. Fetal Diagn Ther 2024; 51:493-499. [PMID: 38934141 PMCID: PMC11446398 DOI: 10.1159/000540000] [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: 04/07/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024]
Abstract
INTRODUCTION Immune checkpoint inhibitors are extensively used in present-day clinical practice for treating many types of cancers at different stages. To date, there are scarce data on the use of immunotherapy in pregnancy. Immune-related adverse events are a typical consequence of this therapy miming autoimmune diseases. CASE PRESENTATION A 35-year-old woman (G1P0) diagnosed with gastric carcinoma underwent neoadjuvant chemotherapy followed by surgery. During follow-up, axillary metastasis was discovered, radiotherapy failed, and consequently immunotherapy was started. Concurrently, pregnancy ensued. Despite potential risks, the patient opted to continue immunotherapy and the pregnancy. At 31 weeks, fetal bowel dilation was noted. Subsequently, the fetus also developed fetal growth restriction. A cesarean section was performed at 35 weeks. The newborn required repeated bowel resections for necrotizing enterocolitis, necessitating extensive medical intervention. The mother continues pembrolizumab treatment with a positive response. CONCLUSION To the best of our knowledge, this might constitute a possible case of a fetal immune-related adverse event after immunotherapy in utero exposure.
Collapse
Affiliation(s)
- Linda Piemonti
- Obstetric Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy,
| | - Laura Vettor
- Department of Women's and Children's Health Gynecologic and Obstetrics Clinic, University of Padua, Padua, Italy
| | - Elena Contro
- Obstetric Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| |
Collapse
|
14
|
Kirkham MN, Cooper C, Broberg E, Robertson P, Clarke D, Pickett BE, Bikman B, Reynolds PR, Arroyo JA. Different Lengths of Gestational Exposure to Secondhand Smoke or e-Cigarette Vapor Induce the Development of Placental Disease Symptoms. Cells 2024; 13:1009. [PMID: 38920640 PMCID: PMC11201565 DOI: 10.3390/cells13121009] [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: 05/06/2024] [Revised: 05/28/2024] [Accepted: 06/07/2024] [Indexed: 06/27/2024] Open
Abstract
Exposure to cigarette smoke is known to induce disease during pregnancy. Recent evidence showed that exposure to secondhand smoke (SHS) negatively impacts fetal and placental weights, leading to the development of intrauterine growth restriction (IUGR). Electronic cigarettes (eCigs) represent a phenomenon that has recently emerged, and their use is also steadily rising. Even so, the effects of SHS or eCigs during gestation remain limited. In the present study, we wanted to characterize the effects of SHS or eCig exposure at two different important gestational points during mouse pregnancy. C57/Bl6 mice were exposed to SHS or eCigs via a nose-only delivery system for 4 days (from 14.5 to 17.5 gestational days (dGA) or for 6 days (from 12.5 dGA to 17.5 dGA)). At the time of necropsy (18.5 dGA), placental and fetal weights were recorded, maternal blood pressure was determined, and a dipstick test to measure proteinuria was performed. Placental tissues were collected, and inflammatory molecules in the placenta were identified. Treatment with SHS showed the following: (1) a significant decrease in placental and fetal weights following four days of exposure, (2) higher systolic and diastolic blood pressure following six days of exposure, and (3) increased proteinuria after six days of exposure. Treatment with eCigs showed the following: (1) a significant decrease in placental weight and fetal weight following four or six days of exposure, (2) higher systolic and diastolic blood pressure following six days of exposure, and (3) increased proteinuria after six days of exposure. We also observed different inflammatory markers associated with the development of IUGR or PE. We conclude that the detrimental effects of SHS or eCig treatment coincide with the length of maternal exposure. These results could be beneficial in understanding the long-term effects of SHS or eCig exposure in the development of placental diseases.
Collapse
Affiliation(s)
- Madison N. Kirkham
- Lung and Placenta Laboratory, Department of Cell Biology and Physiology, Brigham Young University, Provo, UT 84602, USA (E.B.)
| | - Christian Cooper
- Lung and Placenta Laboratory, Department of Cell Biology and Physiology, Brigham Young University, Provo, UT 84602, USA (E.B.)
| | - Emily Broberg
- Lung and Placenta Laboratory, Department of Cell Biology and Physiology, Brigham Young University, Provo, UT 84602, USA (E.B.)
| | - Peter Robertson
- Lung and Placenta Laboratory, Department of Cell Biology and Physiology, Brigham Young University, Provo, UT 84602, USA (E.B.)
| | - Derek Clarke
- Lung and Placenta Laboratory, Department of Cell Biology and Physiology, Brigham Young University, Provo, UT 84602, USA (E.B.)
| | - Brett E. Pickett
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT 84602, USA;
| | - Benjamin Bikman
- Lung and Placenta Laboratory, Department of Cell Biology and Physiology, Brigham Young University, Provo, UT 84602, USA (E.B.)
| | - Paul R. Reynolds
- Lung and Placenta Laboratory, Department of Cell Biology and Physiology, Brigham Young University, Provo, UT 84602, USA (E.B.)
| | - Juan A. Arroyo
- Lung and Placenta Laboratory, Department of Cell Biology and Physiology, Brigham Young University, Provo, UT 84602, USA (E.B.)
| |
Collapse
|
15
|
Linck Moroni J, Tsoi S, Wenger II, Plastow GS, Dyck MK. Placental Transcriptome Analysis in Connection with Low Litter Birth Weight Phenotype (LBWP) Sows. Genes (Basel) 2024; 15:703. [PMID: 38927639 PMCID: PMC11203121 DOI: 10.3390/genes15060703] [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: 04/07/2024] [Revised: 05/16/2024] [Accepted: 05/21/2024] [Indexed: 06/28/2024] Open
Abstract
It is possible to identify sub-populations of sows in every pig herd that consistently give birth to low birth weight (BW) piglets, irrespective of the litter size. A previous study from our group demonstrated that placental development is a main factor affecting the litter birth weight phenotype (LBWP) in sows, thereby impacting the BW of entire litters, but the biological and molecular pathways behind this phenomenon are largely unknown. The aim of this study was to investigate the differential gene expression in placental tissues at day 30 of gestation between low LBWP (LLBWP) vs. high LBWP (HLBWP) sows from a purebred Large White maternal line. Using mRNA sequencing, we found 45 differentially expressed genes (DEGs) in placental tissues of LLBWP and HLBWP sows. Furthermore, (GO) enrichment of upregulated DEGs predicted that there were two biological processes significantly related to cornification and regulation of cell population proliferation. To better understand the molecular interaction between cell proliferation and cornification, we conducted transcriptional factor binding site (TFBS) prediction analysis. The results indicated that a highly significant TFBS was located at the 5' upstream of all four upregulated genes (CDSN, DSG3, KLK14, KRT17), recognized by transcription factors EGR4 and FOSL1. Our findings provide novel insight into how transcriptional regulation of two different biological processes interact in placental tissues of LLBWP sows.
Collapse
Affiliation(s)
| | | | | | | | - Michael K. Dyck
- Department of Agricultural, Food & Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada; (J.L.M.); (S.T.); (I.I.W.); (G.S.P.)
| |
Collapse
|
16
|
Vincze M, Sikovanyecz J, Földesi I, Surányi A, Várbíró S, Németh G, Kozinszky Z, Sikovanyecz J. How the Soluble Human Leukocyte Antigen-G levels in Amniotic Fluid and Maternal Serum Correlate with the Feto-Placental Growth in Uncomplicated Pregnancies. Bioengineering (Basel) 2024; 11:509. [PMID: 38790375 PMCID: PMC11117885 DOI: 10.3390/bioengineering11050509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 04/18/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024] Open
Abstract
Introduction: Trophoblast-derived angiogenic factors are considered to play an important role in the pathophysiology of various complications of pregnancy. Human Leukocyte Antigen-G (HLA-G) belongs to the non-classical human major histocompatibility complex (MHC-I) molecule and has membrane-bound and soluble forms. HLA-G is primarily expressed by extravillous cytotrophoblasts located in the placenta between the maternal and fetal compartments and plays a pivotal role in providing immune tolerance. The aim of this study was to establish a relationship between concentrations of soluble HLA-G (sHLA-G) in maternal serum and amniotic fluid at 16-22 weeks of gestation and the sonographic measurements of fetal and placental growth. Materials and methods: sHLA-G in serum and amniotic fluid, as well as fetal biometric data and placental volume and perfusion indices, were determined in 41 singleton pregnancies with no complications. The level of sHLA-G (U/mL) was tested with a sandwich enzyme-linked immunosorbent assay (ELISA) kit. Results: The sHLA-G levels were unchanged both in amniotic fluid and serum during mid-pregnancy. The sHLA-G level in serum correlated positively with amniotic sHLA-G level (β = 0.63, p < 0.01). Serum sHLA-G level was significantly correlated with abdominal measurements (β = 0.41, p < 0.05) and estimated fetal weight (β = 0.41, p < 0.05). Conversely, amniotic sHLA-G level and placental perfusion (VI: β = -0.34, p < 0.01 and VFI: β = -0.44, p < 0.01, respectively) were negatively correlated. A low amniotic sHLA-G level was significantly associated with nuchal translucency (r = -0.102, p < 0.05). Conclusions: sHLA-G assayed in amniotic fluid might be a potential indicator of placental function, whereas the sHLA-G level in serum can be a prognostic factor for feto-placental insufficiency.
Collapse
Affiliation(s)
- Márió Vincze
- Department of Obstetrics and Gynecology, Albert Szent-Gyorgyi Medical School, University of Szeged, H-6725 Szeged, Hungary; (M.V.); (J.S.J.); (A.S.); (S.V.); (G.N.); (J.S.)
| | - János Sikovanyecz
- Department of Obstetrics and Gynecology, Albert Szent-Gyorgyi Medical School, University of Szeged, H-6725 Szeged, Hungary; (M.V.); (J.S.J.); (A.S.); (S.V.); (G.N.); (J.S.)
| | - Imre Földesi
- Department of Laboratory Medicine, Albert Szent-Gyorgyi Medical School, University of Szeged, H-6720 Szeged, Hungary;
| | - Andrea Surányi
- Department of Obstetrics and Gynecology, Albert Szent-Gyorgyi Medical School, University of Szeged, H-6725 Szeged, Hungary; (M.V.); (J.S.J.); (A.S.); (S.V.); (G.N.); (J.S.)
| | - Szabolcs Várbíró
- Department of Obstetrics and Gynecology, Albert Szent-Gyorgyi Medical School, University of Szeged, H-6725 Szeged, Hungary; (M.V.); (J.S.J.); (A.S.); (S.V.); (G.N.); (J.S.)
| | - Gábor Németh
- Department of Obstetrics and Gynecology, Albert Szent-Gyorgyi Medical School, University of Szeged, H-6725 Szeged, Hungary; (M.V.); (J.S.J.); (A.S.); (S.V.); (G.N.); (J.S.)
| | - Zoltan Kozinszky
- Capio Specialized Center for Gynecology, Postgången 53, 171 45 Solna, Sweden
| | - János Sikovanyecz
- Department of Obstetrics and Gynecology, Albert Szent-Gyorgyi Medical School, University of Szeged, H-6725 Szeged, Hungary; (M.V.); (J.S.J.); (A.S.); (S.V.); (G.N.); (J.S.)
| |
Collapse
|
17
|
Geisert RD, Bazer FW, Lucas CG, Pfeiffer CA, Meyer AE, Sullivan R, Johns DN, Sponchiado M, Prather RS. Maternal recognition of pregnancy in the pig: A servomechanism involving sex steroids, cytokines and prostaglandins. Anim Reprod Sci 2024; 264:107452. [PMID: 38522133 DOI: 10.1016/j.anireprosci.2024.107452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/07/2024] [Accepted: 03/08/2024] [Indexed: 03/26/2024]
Abstract
Maternal recognition of pregnancy (MRP) is a term utilized in mammals to describe pathways in which the conceptus alters the endometrial environment to prevent regression of corpora lutea to ensure continued production of progesterone (P4) required for establishment and maintenance of pregnancy. For nearly 40 years after publication of the endocrine/exocrine theory, conceptus estrogen (E2) was considered the primary maternal recognition signal in the pig. Conceptus production of prostaglandin E2 (PGE2) was also considered to be a major factor in preventing luteolysis. An addition to E2 and PGE2, pig conceptuses produce interleukin 1B2 (IL1B2) and interferons (IFN) delta (IFND) and gamma (IFNG). The present review provides brief history of the discovery of E2, PGs and IFNS which led to research investigating the role of these conceptus secreted factors in establishing and maintaining pregnancy in the pig. The recent utilization of gene editing technology allowed a more direct approach to investigate the in vivo roles of IL1B2, E2, PGE2, AND IFNG for establishment of pregnancy. These studies revealed unknown functions for IFNG and ILB2 in addition to PGE2 and E2. Thus, pregnancy recognition signal is via a servomechanism in requiring sequential effects of P4, E2, IL1B2, PGE2 and IFNG. Results indicate that the original established dogma for the role of conceptus E2 and PGs in MRP is a far too simplified model that involves the interplay of numerous mechanisms for inhibiting luteolysis, inducing critical elongation of the conceptuses and resolution of inflammation in pigs.
Collapse
Affiliation(s)
- Rodney D Geisert
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211, USA.
| | - Fuller W Bazer
- Department of Animal Science, Texas A&M University, College Station, TX 77843, USA
| | - Caroline G Lucas
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211, USA
| | - Caroline A Pfeiffer
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211, USA
| | - Ashley E Meyer
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211, USA
| | - Riley Sullivan
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211, USA
| | - Destiny N Johns
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211, USA
| | - Mariana Sponchiado
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211, USA
| | - Randall S Prather
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211, USA
| |
Collapse
|
18
|
Aghajanpour S, Mehraein F, Amjadi F, Zandieh Z, Ghaffari F, Aflatoonian K, Hosseini E, Bakhtiyari M, Aflatoonian R. Endometrial scratching in unexplained repeated implantation failure causes two competing forces, angiogenesis and anti-angiogenesis: An RCT study. Int J Reprod Biomed 2024; 22:253-268. [PMID: 39035633 PMCID: PMC11255460 DOI: 10.18502/ijrm.v22i4.16387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 02/10/2024] [Accepted: 03/06/2024] [Indexed: 07/23/2024] Open
Abstract
Background A significant association between endometrial vascularity and pregnancy has been shown in previous research, while poor vascularization was attributed to repeated implantation failure (RIF). One possible approach to enhance angiogenesis for successful implantation is endometrial scratching (ES). Objective The purpose was to investigate endometrial responses to scratching by profiling angiogenesis-related gene expression in unexplained RIF participants. Materials and Methods In this randomized controlled trial study, 20 infertile women with unexplained RIF were assigned to 2 groups by the balanced block randomization method (n = 10/each group): the intervention group (group A) (who received ES in the follicular phase) and the control group (group B). Endometrial biopsy was performed in the secretory phase. Gene expression profiling was performed using a polymerase chain reaction-array kit for human-angiogenic growth factors. The implantation and clinical pregnancy rates were also assessed. Results Among the angiogenesis-promoting genes, FGF1, FGF13, FGF2, TGFA, ANG, ANGPT1, and VEGFA were significantly upregulated (p < 0.05). IL12A (an angiogenesis-inhibiting cytokine) was significantly upregulated (p < 0.01). In contrast, 15 genes with angiogenesis-related functions, including CXCL11, CXCL13, CXCL3, CXCL5, CXCL6, EREG, FIGF, FST, IL10, LEP, PPBP, PROK1, RHOB, TNF, and TYMP, were downregulated after ES. No significant differences were observed between the intervention (group A) and control (group B) groups in terms of implantation (43.75% vs. 28.57%) or clinical pregnancy rates (75% vs. 57.1%). Conclusion ES induced significant alterations in the expression of angiogenesis-related genes, with notable up/downregulation of key angiogenic/antiangiogenic factors. These findings enhance our understanding of the molecular responses triggered by ES, underscoring the potential influence of ES on the complex processes of angiogenesis crucial for implantation.
Collapse
Affiliation(s)
- Samaneh Aghajanpour
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Fereshteh Mehraein
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Minimally Invasive Surgery Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemehsadat Amjadi
- Reproductive Sciences and Technology Research Center, Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Zandieh
- Reproductive Sciences and Technology Research Center, Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Firouzeh Ghaffari
- Department of Endocrinology and Female Infertility, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | | | - Elham Hosseini
- Department of Obstetrics and Gynecology, Mousavi Hospital, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
- Zanjan Metabolic Diseases Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mehrdad Bakhtiyari
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Reza Aflatoonian
- Department of Endocrinology and Female Infertility, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| |
Collapse
|
19
|
Candia AA, Lean SC, Zhang CXW, McKeating DR, Cochrane A, Gulacsi E, Herrera EA, Krause BJ, Sferruzzi-Perri AN. Obesogenic Diet in Mice Leads to Inflammation and Oxidative Stress in the Mother in Association with Sex-Specific Changes in Fetal Development, Inflammatory Markers and Placental Transcriptome. Antioxidants (Basel) 2024; 13:411. [PMID: 38671859 PMCID: PMC11047652 DOI: 10.3390/antiox13040411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 03/21/2024] [Accepted: 03/26/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND Obesity during pregnancy is related to adverse maternal and neonatal outcomes. Factors involved in these outcomes may include increased maternal insulin resistance, inflammation, oxidative stress, and nutrient mishandling. The placenta is the primary determinant of fetal outcomes, and its function can be impacted by maternal obesity. The aim of this study on mice was to determine the effect of obesity on maternal lipid handling, inflammatory and redox state, and placental oxidative stress, inflammatory signaling, and gene expression relative to female and male fetal growth. METHODS Female mice were fed control or obesogenic high-fat/high-sugar diet (HFHS) from 9 weeks prior to, and during, pregnancy. On day 18.5 of pregnancy, maternal plasma, and liver, placenta, and fetal serum were collected to examine the immune and redox states. The placental labyrinth zone (Lz) was dissected for RNA-sequencing analysis of gene expression changes. RESULTS the HFHS diet induced, in the dams, hepatic steatosis, oxidative stress (reduced catalase, elevated protein oxidation) and the activation of pro-inflammatory pathways (p38-MAPK), along with imbalanced circulating cytokine concentrations (increased IL-6 and decreased IL-5 and IL-17A). HFHS fetuses were asymmetrically growth-restricted, showing sex-specific changes in circulating cytokines (GM-CSF, TNF-α, IL-6 and IFN-γ). The morphology of the placenta Lz was modified by an HFHS diet, in association with sex-specific alterations in the expression of genes and proteins implicated in oxidative stress, inflammation, and stress signaling. Placental gene expression changes were comparable to that seen in models of intrauterine inflammation and were related to a transcriptional network involving transcription factors, LYL1 and PLAG1. CONCLUSION This study shows that fetal growth restriction with maternal obesity is related to elevated oxidative stress, inflammatory pathways, and sex-specific placental changes. Our data are important, given the marked consequences and the rising rates of obesity worldwide.
Collapse
Affiliation(s)
- Alejandro A. Candia
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK; (A.A.C.); (C.X.W.Z.); (D.R.M.); (A.C.); (E.G.)
- Institute of Health Sciences, University of O’Higgins, Rancagua 2841959, Chile;
- Pathophysiology Program, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago 7500922, Chile;
- Department for the Woman and Newborn Health Promotion, Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile
| | - Samantha C. Lean
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK; (A.A.C.); (C.X.W.Z.); (D.R.M.); (A.C.); (E.G.)
| | - Cindy X. W. Zhang
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK; (A.A.C.); (C.X.W.Z.); (D.R.M.); (A.C.); (E.G.)
| | - Daniel R. McKeating
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK; (A.A.C.); (C.X.W.Z.); (D.R.M.); (A.C.); (E.G.)
| | - Anna Cochrane
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK; (A.A.C.); (C.X.W.Z.); (D.R.M.); (A.C.); (E.G.)
| | - Edina Gulacsi
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK; (A.A.C.); (C.X.W.Z.); (D.R.M.); (A.C.); (E.G.)
| | - Emilio A. Herrera
- Pathophysiology Program, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago 7500922, Chile;
| | - Bernardo J. Krause
- Institute of Health Sciences, University of O’Higgins, Rancagua 2841959, Chile;
| | - Amanda N. Sferruzzi-Perri
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK; (A.A.C.); (C.X.W.Z.); (D.R.M.); (A.C.); (E.G.)
| |
Collapse
|
20
|
Bhatia Z, Kumar S, Seshadri S. Composition and interaction of maternal microbiota with immune mediators during pregnancy and their outcome: A narrative review. Life Sci 2024; 340:122440. [PMID: 38278350 DOI: 10.1016/j.lfs.2024.122440] [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: 10/18/2023] [Revised: 12/26/2023] [Accepted: 01/14/2024] [Indexed: 01/28/2024]
Abstract
The connection between maternal microbiota and infant health has been greatly garnered interest for therapeutic purposes. The early resident microbiota perpetually exhibits much more flexibility as compared to that of the adults, and therefore, constant need of understanding the infant as well as maternal microbiota and their implications however has increased. In this review, we focus mainly on the diversity of overall maternal microbiota including the gut, vaginal, colostrum microbiota and how inflammatory markers fluctuate throughout the normal pregnancy as well in pregnancy with complications. The maternal body undergoes a cascade of physiological changes including hormonal, immunological and metabolic events to support the fetal development. These changes at the time of pregnancy have been correlated with alteration in the composition and diversity of maternal microbiota. Along with alteration in microbiome, the levels of circulatory cytokines fluctuate by complex network of inflammation, in order to prevent the fetal allograft throughout the pregnancy. The dynamic relationship of gut microbiota with the host and its immune system allows one to have greater insights of their role in pregnancy and newborn's health. Emerging evidence suggests that the vertical transmission of bacterial community from mother to newborn may begin in-utero which contributes in developing the immune system and infant gut microbiota.
Collapse
Affiliation(s)
- Zeel Bhatia
- Institute of Science, Nirma University, Ahmedabad, Gujarat 382481, India
| | - Sunny Kumar
- Institute of Science, Nirma University, Ahmedabad, Gujarat 382481, India
| | - Sriram Seshadri
- Institute of Science, Nirma University, Ahmedabad, Gujarat 382481, India.
| |
Collapse
|
21
|
Ravelojaona M, Girouard J, Kana Tsapi ES, Chambers M, Vaillancourt C, Van Themsche C, Thornton CA, Reyes-Moreno C. Oncostatin M and STAT3 Signaling Pathways Support Human Trophoblast Differentiation by Inhibiting Inflammatory Stress in Response to IFNγ and GM-CSF. Cells 2024; 13:229. [PMID: 38334621 PMCID: PMC10854549 DOI: 10.3390/cells13030229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 01/19/2024] [Accepted: 01/23/2024] [Indexed: 02/10/2024] Open
Abstract
Interleukin-6 (IL-6) superfamily cytokines play critical roles during human pregnancy by promoting trophoblast differentiation, invasion, and endocrine function, and maintaining embryo immunotolerance and protection. In contrast, the unbalanced activity of pro-inflammatory factors such as interferon gamma (IFNγ) and granulocyte-macrophage colony-stimulating factor (GM-CSF) at the maternal-fetal interface have detrimental effects on trophoblast function and differentiation. This study demonstrates how the IL-6 cytokine family member oncostatin M (OSM) and STAT3 activation regulate trophoblast fusion and endocrine function in response to pro-inflammatory stress induced by IFNγ and GM-CSF. Using human cytotrophoblast-like BeWo (CT/BW) cells, differentiated in villous syncytiotrophoblast (VST/BW) cells, we show that beta-human chorionic gonadotrophin (βhCG) production and cell fusion process are affected in response to IFNγ or GM-CSF. However, those effects are abrogated with OSM by modulating the activation of IFNγ-STAT1 and GM-CSF-STAT5 signaling pathways. OSM stimulation enhances the expression of STAT3, the phosphorylation of STAT3 and SMAD2, and the induction of negative regulators of inflammation (e.g., IL-10 and TGFβ1) and cytokine signaling (e.g., SOCS1 and SOCS3). Using STAT3-deficient VST/BW cells, we show that STAT3 expression is required for OSM to regulate the effects of IFNγ in βhCG and E-cadherin expression. In contrast, OSM retains its modulatory effect on GM-CSF-STAT5 pathway activation even in STAT3-deficient VST/BW cells, suggesting that OSM uses STAT3-dependent and -independent mechanisms to modulate the activation of pro-inflammatory pathways IFNγ-STAT1 and GM-CSF-STAT5. Moreover, STAT3 deficiency in VST/BW cells leads to the production of both a large amount of βhCG and an enhanced expression of activated STAT5 induced by GM-CSF, independently of OSM, suggesting a key role for STAT3 in βhCG production and trophoblast differentiation through STAT5 modulation. In conclusion, our study describes for the first time the critical role played by OSM and STAT3 signaling pathways to preserve and regulate trophoblast biological functions during inflammatory stress.
Collapse
Affiliation(s)
- Marion Ravelojaona
- Groupe de Recherche en Signalisation Cellulaire (GRSC), Département de Biologie Médicale, Université du Québec à Trois-Rivières, 3351 Boul. des Forges, Trois-Rivières, QC G8Z 4M3, Canada
- Centre de Recherche Interuniversitaire en Reproduction et Développement-Réseau Québécois en Reproduction (CIRD-RQR), Université de Montréal, St-Hyacinthe, QC J2S 2M2, Canada;
- Regroupement Intersectoriel de Recherche en Santé de l’Université du Québec (RISUQ), Université du Québec, Québec, QC G1K 9H7, Canada
| | - Julie Girouard
- Groupe de Recherche en Signalisation Cellulaire (GRSC), Département de Biologie Médicale, Université du Québec à Trois-Rivières, 3351 Boul. des Forges, Trois-Rivières, QC G8Z 4M3, Canada
- Centre de Recherche Interuniversitaire en Reproduction et Développement-Réseau Québécois en Reproduction (CIRD-RQR), Université de Montréal, St-Hyacinthe, QC J2S 2M2, Canada;
- Regroupement Intersectoriel de Recherche en Santé de l’Université du Québec (RISUQ), Université du Québec, Québec, QC G1K 9H7, Canada
| | - Emmanuelle Stella Kana Tsapi
- Groupe de Recherche en Signalisation Cellulaire (GRSC), Département de Biologie Médicale, Université du Québec à Trois-Rivières, 3351 Boul. des Forges, Trois-Rivières, QC G8Z 4M3, Canada
| | | | - Cathy Vaillancourt
- Centre de Recherche Interuniversitaire en Reproduction et Développement-Réseau Québécois en Reproduction (CIRD-RQR), Université de Montréal, St-Hyacinthe, QC J2S 2M2, Canada;
- Regroupement Intersectoriel de Recherche en Santé de l’Université du Québec (RISUQ), Université du Québec, Québec, QC G1K 9H7, Canada
- Centre Armand Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique (INRS), Laval, QC H7V 1B7, Canada
| | - Céline Van Themsche
- Groupe de Recherche en Signalisation Cellulaire (GRSC), Département de Biologie Médicale, Université du Québec à Trois-Rivières, 3351 Boul. des Forges, Trois-Rivières, QC G8Z 4M3, Canada
- Centre de Recherche Interuniversitaire en Reproduction et Développement-Réseau Québécois en Reproduction (CIRD-RQR), Université de Montréal, St-Hyacinthe, QC J2S 2M2, Canada;
- Regroupement Intersectoriel de Recherche en Santé de l’Université du Québec (RISUQ), Université du Québec, Québec, QC G1K 9H7, Canada
| | | | - Carlos Reyes-Moreno
- Groupe de Recherche en Signalisation Cellulaire (GRSC), Département de Biologie Médicale, Université du Québec à Trois-Rivières, 3351 Boul. des Forges, Trois-Rivières, QC G8Z 4M3, Canada
- Centre de Recherche Interuniversitaire en Reproduction et Développement-Réseau Québécois en Reproduction (CIRD-RQR), Université de Montréal, St-Hyacinthe, QC J2S 2M2, Canada;
- Regroupement Intersectoriel de Recherche en Santé de l’Université du Québec (RISUQ), Université du Québec, Québec, QC G1K 9H7, Canada
| |
Collapse
|
22
|
Velez C, Williamson D, Cánovas ML, Giai LR, Rutland C, Pérez W, Barbeito CG. Changes in Immune Response during Pig Gestation with a Focus on Cytokines. Vet Sci 2024; 11:50. [PMID: 38275932 PMCID: PMC10819333 DOI: 10.3390/vetsci11010050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 12/28/2023] [Accepted: 01/08/2024] [Indexed: 01/27/2024] Open
Abstract
Pigs have the highest percentage of embryonic death not associated with specific diseases of all livestock species, at 20-45%. During gestation processes, a series of complex alterations can arise, including embryonic migration and elongation, maternal immunological recognition of pregnancy, and embryonic competition for implantation sites and subsequent nutrition requirements and development. Immune cells and cytokines act as mediators between other molecules in highly complex interactions between various cell types. However, other non-immune cells, such as trophoblast cells, are important in immune pregnancy regulation. Numerous studies have shed light on the crucial roles of several cytokines that regulate the inflammatory processes that characterize the interface between the fetus and the mother throughout normal porcine gestation, but most of these reports are limited to the implantational and peri-implantational periods. Increase in some proinflammatory cytokines have been found in other gestational periods, such as placental remodeling. Porcine immune changes during delivery have not been studied as deeply as in other species. This review details some of the immune system cells actively involved in the fetomaternal interface during porcine gestation, as well as the principal cells, cytokines, and molecules, such as antibodies, that play crucial roles in sow pregnancy, both in early and mid-to-late gestation.
Collapse
Affiliation(s)
- Carolina Velez
- Laboratory of Histology, Faculty of Veterinary Science, National University of La Pampa (UNLPam), Santa Rosa 6300, Argentina; (C.V.); (D.W.); (L.R.G.)
- National Scientific and Technical Research Council of Argentina (CONICET), Buenos Aires 2690, Argentina;
| | - Delia Williamson
- Laboratory of Histology, Faculty of Veterinary Science, National University of La Pampa (UNLPam), Santa Rosa 6300, Argentina; (C.V.); (D.W.); (L.R.G.)
| | - Mariela Lorena Cánovas
- Laboratory of Histology, Faculty of Veterinary Science, National University of La Pampa (UNLPam), Santa Rosa 6300, Argentina; (C.V.); (D.W.); (L.R.G.)
| | - Laura Romina Giai
- Laboratory of Histology, Faculty of Veterinary Science, National University of La Pampa (UNLPam), Santa Rosa 6300, Argentina; (C.V.); (D.W.); (L.R.G.)
| | - Catrin Rutland
- Sutton Bonington Campus, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham NG7 2RD, UK
| | - William Pérez
- Department of Veterinary Anatomy, University of Montevideo, Montevideo 11600, Uruguay
| | - Claudio Gustavo Barbeito
- National Scientific and Technical Research Council of Argentina (CONICET), Buenos Aires 2690, Argentina;
- Laboratory of Descriptive, Comparative and Experimental Histology and Embriology (LHYEDEC), Department of Basic Sciences, Faculty of Veterinary Science, National University of La Plata (UNLP), La Plata 1900, Argentina
| |
Collapse
|
23
|
PALA Ş, ATILGAN R, İLHAN N. High amniotic fluid fractalkine and MIP-1β levels are associated with intrauterine growth restriction: a prospective cohort study. Turk J Med Sci 2023; 54:280-290. [PMID: 38812616 PMCID: PMC11031143 DOI: 10.55730/1300-0144.5789] [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: 05/12/2023] [Revised: 02/15/2024] [Accepted: 12/16/2023] [Indexed: 05/31/2024] Open
Abstract
Background/aim Proinflammatory chemokines have been shown to play crucial roles in implantation, spiral artery invasion, and the fetomaternal immunological response. In this context, we investigated the levels of fractalkine (CX3CL1) and chemokine CC motif ligand 4 (CCL4 or MIP-1β) in maternal serum and amniotic fluids in pregnant women with intrauterine growth restriction (IUGR). Materials and methods This prospective cohort study was carried out at Fırat University Obstetrics Clinic between January 1, 2022 and July 1, 2022. Group (G) 1: The control group consisted of 40 pregnant women who underwent elective cesarean section (CS) at 38-40 weeks of gestation. G2: A total of 40 pregnant women with IUGR at 28-37 weeks of gestation were included in the study group. Levels of tumor necrosis factor-alpha (TNF-α), interleukin-1 beta (IL-1β), interferon-gamma (IFN-γ), hypoxia-inducible factor-1 alpha (HIF-1α), macrophage inflammatory protein-1 beta (MIP-1β), and fractalkine were measured in maternal serum and amniotic fluid samples obtained during CS. Results When maternal age was compared, no statistically significant difference was observed between G1 and G2 (p = 0.374). The number of gravidity was found to be statistically higher in G1 compared to G2 (p = 0.003). The mean gestational week was statistically higher in G1 (p < 0.001). Maternal serum MIP-1β (p = 0.03) and IFN-γ (p = 0.006) levels were higher in G1. The birth weight of the baby (p < 0.001) and umbilical cord blood gas pH value (p < 0.001) at birth were higher in G1. HIF-1α (p < 0.001), fractalkine (p < 0.001), MIP-1β (p < 0.001), TNF-α (p = 0.007), IL-1β (p < 0.001), and IFN-γ levels (p = 0.007) in amniotic fluid were higher in G2. Conclusion Elevated levels of proinflammatory factors, including fractalkine and MIP-1β, along with inflammatory factors such as TNF-α, IL-1β, and IFN-γ, as well as increased HIF-1α levels in amniotic fluid, are associated with intrauterine growth restriction (IUGR) attributed to a hypoxic amniotic environment.
Collapse
Affiliation(s)
- Şehmus PALA
- Department of Obstetrics and Gynecology, Fırat University School of Medicine, Elazığ,
Turkiye
| | - Remzi ATILGAN
- Department of Obstetrics and Gynecology, Fırat University School of Medicine, Elazığ,
Turkiye
| | - Nevin İLHAN
- Department of Biochemistry, Fırat University School of Medicine, Elazığ,
Turkiye
| |
Collapse
|
24
|
Rios LE, Lokugamage N, Choudhuri S, Chowdhury IH, Garg NJ. Subunit nanovaccine elicited T cell functional activation controls Trypanosoma cruzi mediated maternal and placental tissue damage and improves pregnancy outcomes in mice. NPJ Vaccines 2023; 8:188. [PMID: 38104118 PMCID: PMC10725459 DOI: 10.1038/s41541-023-00782-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 11/10/2023] [Indexed: 12/19/2023] Open
Abstract
This study investigated a candidate vaccine effect against maternal Trypanosoma cruzi (Tc) infection and improved pregnancy outcomes. For this, TcG2 and TcG4 were cloned in a nanoplasmid optimized for delivery, antigen expression, and regulatory compliance (nano2/4 vaccine). Female C57BL/6 mice were immunized with nano2/4, infected (Tc SylvioX10), and mated 7-days post-infection to enable fetal development during the maternal acute parasitemia phase. Females were euthanized at E12-E17 (gestation) days. Splenic and placental T-cell responses were monitored by flow cytometry. Maternal and placental/fetal tissues were examined for parasites by qPCR and inflammatory infiltrate by histology. Controls included age/immunization-matched non-pregnant females. Nano2/4 exhibited no toxicity and elicited protective IgG2a/IgG1 response in mice. Nano2/4 signaled a splenic expansion of functionally active CD4+ effector/effector memory (Tem) and central memory (Tcm) cells in pregnant mice. Upon challenge infection, nano2/4 increased the splenic CD4+ and CD8+T cells in all mice and increased the proliferation of CD4+Tem, CD4+Tcm, and CD8+Tcm subsets producing IFNγ and cytolytic molecules (PRF1, GZB) in pregnant mice. A balanced serum cytokines/chemokines response and placental immune characteristics indicated that pregnancy prevented the overwhelming damaging immune response in mice. Importantly, pregnancy itself resulted in a significant reduction of parasites in maternal and fetal tissues. Nano2/4 was effective in arresting the Tc-induced tissue inflammatory infiltrate, necrosis, and fibrosis in maternal and placental tissues and improving maternal fertility, placental efficiency, and fetal survival. In conclusion, we show that maternal nano2/4 vaccination is beneficial in controlling the adverse effects of Tc infection on maternal health, fetal survival, and pregnancy outcomes.
Collapse
Affiliation(s)
- Lizette Elaine Rios
- Department of Microbiology and Immunology, University of Texas Medical Branch (UTMB), Galveston, TX, USA
- Department of Biochemistry and Molecular Biology, UTMB, Galveston, TX, USA
| | - Nandadeva Lokugamage
- Department of Microbiology and Immunology, University of Texas Medical Branch (UTMB), Galveston, TX, USA
| | - Subhadip Choudhuri
- Department of Microbiology and Immunology, University of Texas Medical Branch (UTMB), Galveston, TX, USA
| | - Imran Hussain Chowdhury
- Department of Microbiology and Immunology, University of Texas Medical Branch (UTMB), Galveston, TX, USA
| | - Nisha Jain Garg
- Department of Microbiology and Immunology, University of Texas Medical Branch (UTMB), Galveston, TX, USA.
- Institute for Human Infections and Immunity (IHII), UTMB, Galveston, TX, USA.
- Sealy Institute for Vaccine Sciences (SIVS), UTMB, Galveston, TX, USA.
| |
Collapse
|
25
|
Kouthouridis S, Sotra A, Khan Z, Alvarado J, Raha S, Zhang B. Modeling the Progression of Placental Transport from Early- to Late-Stage Pregnancy by Tuning Trophoblast Differentiation and Vascularization. Adv Healthc Mater 2023; 12:e2301428. [PMID: 37830445 PMCID: PMC11468690 DOI: 10.1002/adhm.202301428] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 09/18/2023] [Indexed: 10/14/2023]
Abstract
The early-stage placental barrier is characterized by a lack of fetal circulation and by a thick trophoblastic barrier, whereas the later-stage placenta consists of vascularized chorionic villi encased in a thin, differentiated trophoblast layer, ideal for nutrient transport. In this work, predictive models of early- and late-stage placental transport are created using blastocyst-derived placental stem cells (PSCs) by modulating PSC differentiation and model vascularization. PSC differentiation results in a thinner, fused trophoblast layer, as well as an increase in human chorionic gonadotropin secretion, barrier permeability, and secretion of certain inflammatory cytokines, which are consistent with in vivo findings. Further, gene expression confirms this shift toward a differentiated trophoblast subtype. Vascularization results in a molecule type- and size-dependent change in dextran and insulin permeability. These results demonstrate that trophoblast differentiation and vascularization have critical effects on placental barrier permeability and that this model can be used as a predictive measure to assess fetal toxicity of xenobiotic substances at different stages of pregnancy.
Collapse
Affiliation(s)
- Sonya Kouthouridis
- Department of Chemical EngineeringMcMaster UniversityHamiltonONL8S 4L8Canada
| | - Alexander Sotra
- School of Biomedical EngineeringMcMaster UniversityHamiltonONL8S 4L8Canada
| | - Zaim Khan
- Department of Biochemistry and Biomedical SciencesMcMaster UniversityHamiltonONL8S 4L8Canada
| | - Justin Alvarado
- Department of Biochemistry and Biomedical SciencesMcMaster UniversityHamiltonONL8S 4L8Canada
| | - Sandeep Raha
- Department of Pediatrics and the Graduate Programme in Medical SciencesMcMaster UniversityHamiltonONL8S 4L8Canada
| | - Boyang Zhang
- Department of Chemical EngineeringMcMaster UniversityHamiltonONL8S 4L8Canada
- School of Biomedical EngineeringMcMaster UniversityHamiltonONL8S 4L8Canada
| |
Collapse
|
26
|
Piekarska K, Dratwa M, Radwan P, Radwan M, Bogunia-Kubik K, Nowak I. Pro- and anti-inflammatory cytokines and growth factors in patients undergoing in vitro fertilization procedure treated with prednisone. Front Immunol 2023; 14:1250488. [PMID: 37744353 PMCID: PMC10511889 DOI: 10.3389/fimmu.2023.1250488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 08/23/2023] [Indexed: 09/26/2023] Open
Abstract
Embryo implantation is a key moment in pregnancy. Abnormal production of pro- and anti-inflammatory cytokines, their receptors and other immune factors may result in embryo implantation failure and pregnancy loss. The aim of this study was to determine the profile of selected pro- and anti-inflammatory factors in the blood plasma of patients undergoing in vitro fertilization (IVF) and control women who achieved pregnancy after natural conception. The examined patients were administered steroid prednisone. We present results concern the plasma levels of IFN-ɣ, BDNF, LIF, VEGF-A, sTNFR1 and IL-10. We found that IVF patients receiving steroids differed significantly from patients who were not administered such treatment in terms of IFN-γ and IL-10 levels. Moreover, IVF patients differed in secretion of all tested factors with the fertile controls. Our results indicated that women who secrete at least 1409 pg/ml of sTNFR1 have a chance to become pregnant naturally and give birth to a child, while patients after IVF must achieve a concentration of 962.3 pg/ml sTNFR1 in blood plasma for successful pregnancy. In addition, IVF patients secreting VEGF-A above 43.28 pg/ml have a greater risk of miscarriage or a failed transfer in comparison to women secreting below this value. In conclusion, fertile women present a different profile of pro- and anti-inflammatory cytokines, and growth factors compared to patients with recurrent implantation failure (RIF).
Collapse
Affiliation(s)
- Karolina Piekarska
- Laboratory of Immunogenetics and Tissue Immunology, Department of Clinical Immunology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Marta Dratwa
- Laboratory of Clinical Immunogenetics and Pharmacogenetics, Department of Clinical Immunology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Paweł Radwan
- Department of Reproductive Medicine, Gameta Hospital, Rzgów, Poland
| | - Michał Radwan
- Department of Reproductive Medicine, Gameta Hospital, Rzgów, Poland
- Faculty of Health Sciences, The Mazovian Academy in Plock, Płock, Poland
| | - Katarzyna Bogunia-Kubik
- Laboratory of Clinical Immunogenetics and Pharmacogenetics, Department of Clinical Immunology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Izabela Nowak
- Laboratory of Immunogenetics and Tissue Immunology, Department of Clinical Immunology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| |
Collapse
|
27
|
Erdő-Bonyár S, Simon D, Bajnok A, Nörenberg J, Serény-Litvai T, Várnagy Á, Kovács K, Hantosi E, Mezősi E, Berki T. Physiological Changes in the Levels of Anti-Cytokine Autoantibodies in Early Pregnancy Are Missing in Pregnant Women with Hashimoto's Thyroiditis. J Immunol Res 2023; 2023:5221658. [PMID: 37663050 PMCID: PMC10473897 DOI: 10.1155/2023/5221658] [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: 04/18/2023] [Revised: 07/21/2023] [Accepted: 08/09/2023] [Indexed: 09/05/2023] Open
Abstract
T helper type 1 (Th1) and inflammatory cytokines play essential roles in early pregnancy and also in the pathogenesis of Hashimoto's thyroiditis (HT). Changes in the serum level of autoantibodies to cytokines, which may be able to modulate their availability and actions have been described in several autoimmune disorders. Yet, no data are available on anti-cytokine autoantibodies either during early pregnancy or in patients with HT. The aim of the study was to measure autoantibodies to inflammatory-, Th1- and Th22-cytokines in serum samples in healthy pregnancy (HP) and in pregnant women with HT (HTP). As pathological autoantibodies are hallmarks of HT, in addition we also measured anti-B-cell activator factor (BAFF) autoantibodies. The measurement was carried out with a Luminex multiplex assay and the Luminex MAGPIX Instrument, age-matched healthy women (HC) and women with HT (HT) were used as controls. In the first trimester of HP, anti-TNFα, anti-IL-8, and anti-IFNγ autoantibodies were significantly decreased, while autoantibodies to BAFF were significantly elevated compared to the HC. However, these alterations were not present in the HTP. Moreover, the levels of autoantibodies to IL-22 and TNFα were significantly increased in HTP compared to the HP. All differences in the levels of the investigated autoantibodies could be detected in the first trimester of pregnancies except for anti-IL-22 autoantibodies. According to our results we can conclude that alterations in the levels of autoantibodies to inflammatory and Th1 cytokines are physiological in the first trimester of pregnancy and their disturbance can be associated with autoimmune conditions such as HT.
Collapse
Affiliation(s)
- Szabina Erdő-Bonyár
- Department of Immunology and Biotechnology, Clinical Center, University of Pécs Medical School, Pécs, Hungary
- National Laboratory on Human Reproduction, University of Pécs, Pécs, Hungary
| | - Diána Simon
- Department of Immunology and Biotechnology, Clinical Center, University of Pécs Medical School, Pécs, Hungary
- National Laboratory on Human Reproduction, University of Pécs, Pécs, Hungary
| | - Anna Bajnok
- National Laboratory on Human Reproduction, University of Pécs, Pécs, Hungary
- Szentágothai Research Centre, University of Pécs, Pécs, Hungary
- Department of Obstetrics and Gynecology, Clinical Center, University of Pécs Medical School, Pécs, Hungary
| | - Jasper Nörenberg
- National Laboratory on Human Reproduction, University of Pécs, Pécs, Hungary
- Department of Obstetrics and Gynecology, Clinical Center, University of Pécs Medical School, Pécs, Hungary
| | - Tímea Serény-Litvai
- Department of Immunology and Biotechnology, Clinical Center, University of Pécs Medical School, Pécs, Hungary
- National Laboratory on Human Reproduction, University of Pécs, Pécs, Hungary
- Szentágothai Research Centre, University of Pécs, Pécs, Hungary
| | - Ákos Várnagy
- National Laboratory on Human Reproduction, University of Pécs, Pécs, Hungary
- Department of Obstetrics and Gynecology, Clinical Center, University of Pécs Medical School, Pécs, Hungary
| | - Kálmán Kovács
- National Laboratory on Human Reproduction, University of Pécs, Pécs, Hungary
- Department of Obstetrics and Gynecology, Clinical Center, University of Pécs Medical School, Pécs, Hungary
| | - Eszter Hantosi
- Department of Obstetrics and Gynecology, Clinical Center, University of Pécs Medical School, Pécs, Hungary
| | - Emese Mezősi
- National Laboratory on Human Reproduction, University of Pécs, Pécs, Hungary
- First Department of Internal Medicine, Clinical Center, University of Pécs Medical School, Pécs, Hungary
| | - Tímea Berki
- Department of Immunology and Biotechnology, Clinical Center, University of Pécs Medical School, Pécs, Hungary
- National Laboratory on Human Reproduction, University of Pécs, Pécs, Hungary
| |
Collapse
|
28
|
Shi Y, Wang CC, Wu L, Zhang Y, Xu A, Wang Y. Pathophysiological Insight into Fatty Acid-Binding Protein-4: Multifaced Roles in Reproduction, Pregnancy, and Offspring Health. Int J Mol Sci 2023; 24:12655. [PMID: 37628833 PMCID: PMC10454382 DOI: 10.3390/ijms241612655] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 08/04/2023] [Accepted: 08/06/2023] [Indexed: 08/27/2023] Open
Abstract
Fatty acid-binding protein-4 (FABP4), commonly known as adipocyte-fatty acid-binding protein (A-FABP), is a pleiotropic adipokine that broadly affects immunity and metabolism. It has been increasingly recognized that FABP4 dysfunction is associated with various metabolic syndromes, including obesity, diabetes, cardiovascular diseases, and metabolic inflammation. However, its explicit roles within the context of women's reproduction and pregnancy remain to be investigated. In this review, we collate recent studies probing the influence of FABP4 on female reproduction, pregnancy, and even fetal health. Elevated circulating FABP4 levels have been found to correlate with impaired reproductive function in women, such as polycystic ovary syndrome and endometriosis. Throughout pregnancy, FABP4 affects maternal-fetal interface homeostasis by affecting both glycolipid metabolism and immune tolerance, leading to adverse pregnancy outcomes, including miscarriage, gestational obesity, gestational diabetes, and preeclampsia. Moreover, maternal FABP4 levels exhibit a substantial linkage with the metabolic health of offspring. Herein, we discuss the emerging significance and potential application of FABP4 in reproduction and pregnancy health and delve into its underlying mechanism at molecular levels.
Collapse
Affiliation(s)
- Yue Shi
- The Second Clinical Medical School, Beijing University of Chinese Medicine, Beijing 100078, China; (Y.S.); (Y.Z.)
| | - Chi-Chiu Wang
- Department of Obstetrics and Gynaecology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong;
- Li Ka Shing Institute of Health Sciences, School of Biomedical Sciences, Chinese University of Hong Kong-Sichuan University Joint Laboratory in Reproductive Medicine, The Chinese University of Hong Kong, Hong Kong
| | - Liqun Wu
- Department of Pediatrics, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing 100078, China;
| | - Yunqing Zhang
- The Second Clinical Medical School, Beijing University of Chinese Medicine, Beijing 100078, China; (Y.S.); (Y.Z.)
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong;
- Department of Medicine, The University of Hong Kong, Hong Kong
| | - Yao Wang
- Department of Obstetrics and Gynaecology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong;
| |
Collapse
|
29
|
Kalinina A, Semenova M, Bruter A, Varlamova E, Kubekina M, Pavlenko N, Silaeva Y, Deikin A, Antoshina E, Gorkova T, Trukhanova L, Salmina A, Novikova S, Voronkov D, Kazansky D, Khromykh L. Cyclophilin A as a Pro-Inflammatory Factor Exhibits Embryotoxic and Teratogenic Effects during Fetal Organogenesis. Int J Mol Sci 2023; 24:11279. [PMID: 37511039 PMCID: PMC10380070 DOI: 10.3390/ijms241411279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 07/05/2023] [Accepted: 07/08/2023] [Indexed: 07/30/2023] Open
Abstract
The precise balance of Th1, Th2, and Th17 cytokines is a key factor in successful pregnancy and normal embryonic development. However, to date, not all humoral factors that regulate and influence physiological pregnancy have been completely studied. Our data here pointed out cyclophilin A (CypA) as the adverse pro-inflammatory factor negatively affecting fetal development and associated with pregnancy complications. In different mouse models in vivo, we demonstrated dramatic embryotoxicity and teratogenicity of increased CypA levels during pregnancy. Using generated transgenic models, we showed that CypA overexpression in fetal tissues induced the death of all transgenic fetuses and complete miscarriage. Administration of recombinant human CypA in a high dose to pregnant females during fetal organogenesis (6.5-11.5 dpc) exhibited teratogenic effects, causing severe defects in the brain and bone development that could lead to malformations and postnatal behavioral and cognitive disorders in the offspring. Embryotoxic and teratogenic effects could be mediated by CypA-induced up-regulation of M1 macrophage polarization via activation of the STAT1/3 signaling pathways. Here, we propose secreted CypA as a novel marker of complicated pregnancy and a therapeutic target for the correction of pregnancy complications.
Collapse
Affiliation(s)
- Anastasiia Kalinina
- N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, 24 Kashirskoe Shosse, Moscow 115478, Russia
| | - Maria Semenova
- Department of Embryology, Faculty of Biology, Moscow State University, 1/12 Leninskie Gory, Moscow 119992, Russia
| | - Alexandra Bruter
- N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, 24 Kashirskoe Shosse, Moscow 115478, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilov Street, Moscow 119334, Russia
| | - Ekaterina Varlamova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilov Street, Moscow 119334, Russia
| | - Marina Kubekina
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilov Street, Moscow 119334, Russia
| | - Natalia Pavlenko
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilov Street, Moscow 119334, Russia
| | - Yulia Silaeva
- Core Facility Center, Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilova Street, Moscow 119334, Russia
| | - Alexey Deikin
- United Center for Genetic Technologies, Belgorod State National Research University, 85 Pobedi Street, Belgorod 308001, Russia
| | - Elena Antoshina
- N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, 24 Kashirskoe Shosse, Moscow 115478, Russia
| | - Tatyana Gorkova
- N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, 24 Kashirskoe Shosse, Moscow 115478, Russia
| | - Lubov Trukhanova
- N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, 24 Kashirskoe Shosse, Moscow 115478, Russia
| | - Alla Salmina
- Research Center of Neurology, 80 Volokolamskoye Shosse, Moscow 125367, Russia
| | - Svetlana Novikova
- Research Center of Neurology, 80 Volokolamskoye Shosse, Moscow 125367, Russia
| | - Dmitry Voronkov
- Research Center of Neurology, 80 Volokolamskoye Shosse, Moscow 125367, Russia
| | - Dmitry Kazansky
- N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, 24 Kashirskoe Shosse, Moscow 115478, Russia
| | - Ludmila Khromykh
- N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, 24 Kashirskoe Shosse, Moscow 115478, Russia
| |
Collapse
|
30
|
Hasegawa Y, Kim DHJ, Zhang Z, Taha AY, Capitanio JP, Hogrefe CE, Bauman MD, Golub MS, Van de Water J, VandeVoort CA, Walker CK, Slupsky CM. Calorie restriction and pravastatin administration during pregnancy in obese rhesus macaques modulates maternal and infant metabolism and infant brain and behavioral development. Front Nutr 2023; 10:1146804. [PMID: 37255938 PMCID: PMC10225656 DOI: 10.3389/fnut.2023.1146804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 04/26/2023] [Indexed: 06/01/2023] Open
Abstract
Background Maternal obesity has been associated with a higher risk of pregnancy-related complications in mothers and offspring; however, effective interventions have not yet been developed. We tested two interventions, calorie restriction and pravastatin administration, during pregnancy in a rhesus macaque model with the hypothesis that these interventions would normalize metabolic dysregulation in pregnant mothers leading to an improvement in infant metabolic and cognitive/social development. Methods A total of 19 obese mothers were assigned to either one of the two intervention groups (n = 5 for calorie restriction; n = 7 for pravastatin) or an obese control group (n = 7) with no intervention, and maternal gestational samples and postnatal infant samples were compared with lean control mothers (n = 6) using metabolomics methods. Results Gestational calorie restriction normalized one-carbon metabolism dysregulation in obese mothers, but altered energy metabolism in her offspring. Although administration of pravastatin during pregnancy tended to normalize blood cholesterol in the mothers, it potentially impacted the gut microbiome and kidney function of their offspring. In the offspring, both calorie restriction and pravastatin administration during pregnancy tended to normalize the activity of AMPK in the brain at 6 months, and while results of the Visual Paired-Comparison test, which measures infant recognition memory, was not significantly impacted by either of the interventions, gestational pravastatin administration, but not calorie restriction, tended to normalize anxiety assessed by the Human Intruder test. Conclusions Although the two interventions tested in a non-human primate model led to some improvements in metabolism and/or infant brain development, negative impacts were also found in both mothers and infants. Our study emphasizes the importance of assessing gestational interventions for maternal obesity on both maternal and offspring long-term outcomes.
Collapse
Affiliation(s)
- Yu Hasegawa
- Department of Food Science and Technology, University of California-Davis, Davis, CA, United States
| | - Danielle H J Kim
- Department of Internal Medicine, Division of Rheumatology, Allergy, and Clinical Immunology, University of California-Davis, Davis, CA, United States
| | - Zhichao Zhang
- Department of Food Science and Technology, University of California-Davis, Davis, CA, United States
| | - Ameer Y Taha
- Department of Food Science and Technology, University of California-Davis, Davis, CA, United States
| | - John P Capitanio
- California National Primate Research Center, University of California-Davis, Davis, CA, United States
| | - Casey E Hogrefe
- California National Primate Research Center, University of California-Davis, Davis, CA, United States
| | - Melissa D Bauman
- California National Primate Research Center, University of California-Davis, Davis, CA, United States
- The UC Davis MIND Institute, University of California-Davis, Sacramento, CA, United States
- Department of Psychiatry and Behavioral Sciences, University of California-Davis, Sacramento, CA, United States
| | - Mari S Golub
- California National Primate Research Center, University of California-Davis, Davis, CA, United States
| | - Judy Van de Water
- The UC Davis MIND Institute, University of California-Davis, Sacramento, CA, United States
- Department of Internal Medicine, University of California-Davis, Sacramento, CA, United States
| | - Catherine A VandeVoort
- California National Primate Research Center, University of California-Davis, Davis, CA, United States
- Department of Obstetrics and Gynecology, University of California-Davis, Davis, CA, United States
| | - Cheryl K Walker
- California National Primate Research Center, University of California-Davis, Davis, CA, United States
- The UC Davis MIND Institute, University of California-Davis, Sacramento, CA, United States
- Department of Obstetrics and Gynecology, University of California-Davis, Davis, CA, United States
| | - Carolyn M Slupsky
- Department of Food Science and Technology, University of California-Davis, Davis, CA, United States
- Department of Nutrition, University of California-Davis, Davis, CA, United States
| |
Collapse
|
31
|
Beltrami S, Rizzo S, Caccuri F, Rizzo R, Bortolotti D, Schiuma G. SARS-CoV-2 Systemic Effects: New Clues. Microorganisms 2023; 11:1209. [PMID: 37317183 DOI: 10.3390/microorganisms11051209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 04/07/2023] [Indexed: 06/16/2023] Open
Abstract
To date, much discussion has been had on severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) lung infection associated with COVID-19 onset, of which the major manifestation is characterized by a "cytokine storm" [...].
Collapse
Affiliation(s)
- Silvia Beltrami
- Department of Chemical, Pharmaceutical and Agricultural Science, University of Ferrara, 44121 Ferrara, Italy
| | - Sabrina Rizzo
- Department of Chemical, Pharmaceutical and Agricultural Science, University of Ferrara, 44121 Ferrara, Italy
| | - Francesca Caccuri
- Department of Microbiology and Virology, "Spedali Civili", 25126 Brescia, Italy
| | - Roberta Rizzo
- Department of Chemical, Pharmaceutical and Agricultural Science, University of Ferrara, 44121 Ferrara, Italy
| | - Daria Bortolotti
- Department of Chemical, Pharmaceutical and Agricultural Science, University of Ferrara, 44121 Ferrara, Italy
| | - Giovanna Schiuma
- Department of Chemical, Pharmaceutical and Agricultural Science, University of Ferrara, 44121 Ferrara, Italy
| |
Collapse
|
32
|
Motomura K, Miller D, Galaz J, Liu TN, Romero R, Gomez-Lopez N. The effects of progesterone on immune cellular function at the maternal-fetal interface and in maternal circulation. J Steroid Biochem Mol Biol 2023; 229:106254. [PMID: 36681283 PMCID: PMC10038932 DOI: 10.1016/j.jsbmb.2023.106254] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 01/16/2023] [Accepted: 01/16/2023] [Indexed: 01/19/2023]
Abstract
Progesterone is a sex steroid hormone that plays a critical role in the establishment and maintenance of pregnancy. This hormone drives numerous maternal physiological adaptations to ensure the continuation of pregnancy and to facilitate fetal growth, including broad and potent modulation of the maternal immune system to promote maternal-fetal tolerance. In this brief review, we provide an overview of the immunomodulatory functions of progesterone in the decidua, placenta, myometrium, and maternal circulation during pregnancy. Specifically, we summarize current evidence of the regulated functions of innate and adaptive immune cells induced by progesterone and its downstream effector molecules in these compartments, including observations in human pregnancy and in animal models. Our review highlights the gaps in knowledge of interactions between progesterone and maternal cellular immunity that may direct future research.
Collapse
Affiliation(s)
- Kenichiro Motomura
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, and Detroit, MI, USA; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Derek Miller
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, and Detroit, MI, USA; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Jose Galaz
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, and Detroit, MI, USA; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA; Division of Obstetrics and Gynecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Tzu Ning Liu
- Wayne State University School of Medicine, Detroit, MI, USA
| | - Roberto Romero
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, and Detroit, MI, USA; Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA; Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, USA; Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA; Detroit Medical Center, Detroit, MI, USA
| | - Nardhy Gomez-Lopez
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, and Detroit, MI, USA; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA; Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA; Department of Biochemistry, Microbiology, and Immunology, Wayne State University School of Medicine, Detroit, MI, USA.
| |
Collapse
|
33
|
Arrighi F, Granese A, Chimenti P, Guglielmi P. Novel therapeutic opportunities for Toxoplasma gondii, Trichomonas vaginalis and Giardia intestinalis infections. Expert Opin Ther Pat 2023; 33:211-245. [PMID: 37099697 DOI: 10.1080/13543776.2023.2206017] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2023]
Abstract
INTRODUCTION Toxoplasma gondii, Trichomonas vaginalis and Giardia intestinalis are the causative agents of Toxoplasmosis, Trichomoniasis and Giardiasis, three important infections threatening human health and affecting millions of people worldwide. Although drugs and treatment are available to fight these protozoan parasites, side-effects and increasing drug resistance, require continuous efforts for the development of novel effective drugs. AREAS COVERED The patents search was carried out in September/October 2022 with four official scientific databases (Espacenet, Scifinder, Reaxys, Google Patents). Treatments for Toxoplasmosis, Trichomoniasis and Giardiasis (2015-2022) have been grouped according to their chemotypes. In particular, novel chemical entities have been reported and investigated for their structure-activity relationship, when accessible. On the other hand, drug repurposing, extensively exploited to obtain novel anti-protozoal treatment, has been in-depth described. Finally, natural metabolites and extracts have also been reported. EXPERT OPINION T. gondii, T. vaginalis and G. intestinalis are protozoan infections usually controlled by immune system in immunocompetent patients; however, they could represent a threatening health for immunocompromised people. The needs of novel effective drugs, endowed with new mechanisms of actions arises from the increasing drug resistance affecting antibiotic as well as antiprotozoal therapies. In this review different therapeutic approaches to treat protozoan infections have been reported.
Collapse
Affiliation(s)
- Francesca Arrighi
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, Rome, Italy
| | - Arianna Granese
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, Rome, Italy
| | - Paola Chimenti
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, Rome, Italy
| | - Paolo Guglielmi
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
34
|
Santos LC, de Souza CA, Silva JF, Ocarino NM, Serakides R. Maternal hyperthyroidism alters the immunological mediators profile and population of natural killers cells in decidua of rats. Acta Histochem 2023; 125:152026. [PMID: 37058857 DOI: 10.1016/j.acthis.2023.152026] [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: 02/27/2023] [Revised: 03/27/2023] [Accepted: 03/29/2023] [Indexed: 04/16/2023]
Abstract
Decidual immunological mediators modulate placental formation, decidualization and fetal development. However, the effect of maternal hyperthyroidism on decidual immunology needs further research. The aim of this study was to evaluate the population of uterine natural killer cells (uNKs) and the expression of immunological mediators in the decidua of female rats throughout pregnancy. Wistar rats were used and hyperthyroidism was induced by daily administration of L-thyroxine (T4) throughout pregnancy. The population of uNK cells in decidua was evaluated by immunostaining Lectin DBA, as well as the expression of interferon γ (INFγ), macrophage migration inhibitory factor (MIF), interleukin 15 (IL-15) and inducible nitric oxide synthase (iNOS) at 7, 10, 12, 14 and 19 days of gestation (DG). Maternal hyperthyroidism reduced the DBA+ uNK cell population in the decidua at 7 (P < 0.05) and 10 (P < 0.01) DGs compared to that in the control group, while it increased in the basal decidua (P < 0.05) and metrial gland (P < 0.0001) at the 12th DG. Hyperthyroidism also increased immunostaining of IL-15 (P < 0.0001), INFγ (P < 0.05), and MIF (P < 0.05) in the 7th DG, and increased immunostaining of IL-15 (P < 0.0001) and MIF (P < 0.01) in the 10th DG. However, excess thyroxine reduced IL-15 expression in the metrial gland and/or basal decidua in the 12th (P < 0.05), 14th (P < 0.01), and 19th (P < 0.001) DGs, as was also observed for INFγ in the basal decidua (P<0.001) and metrial gland (P < 0.0001) in the 12th DG. Regarding iNOS, an antiinflammatory cytokine, lower expression was observed in the basal decidua of hyperthyroid animals at 7 and 12 DGs (P < 0.05), whereas an increase occurred in the 10th DG (P < 0.05). These data demonstrate that maternal hyperthyroidism in female rats, particularly between 7 and 10 DGs, reduces the population of DBA+ uNKs in the decidua and increases the expression of inflammatory cytokines, suggesting a more proinflammatory environment in early pregnancy caused by this gestational disease.
Collapse
Affiliation(s)
- Luciano Cardoso Santos
- Centro de Microscopia Eletronica, Departamento de Ciencias Biologicas, Universidade Estadual de Santa Cruz, Campus Soane Nazare de Andrade, 45662-900 Ilheus, Brazil
| | - Cíntia Almeida de Souza
- Departamento de Clinica e Cirurgia Veterinarias, Escola de Veterinaria, Universidade Federal de Minas Gerais, 31270-901 Belo Horizonte, Brazil
| | - Juneo Freitas Silva
- Centro de Microscopia Eletronica, Departamento de Ciencias Biologicas, Universidade Estadual de Santa Cruz, Campus Soane Nazare de Andrade, 45662-900 Ilheus, Brazil
| | - Natália Melo Ocarino
- Departamento de Clinica e Cirurgia Veterinarias, Escola de Veterinaria, Universidade Federal de Minas Gerais, 31270-901 Belo Horizonte, Brazil
| | - Rogéria Serakides
- Departamento de Clinica e Cirurgia Veterinarias, Escola de Veterinaria, Universidade Federal de Minas Gerais, 31270-901 Belo Horizonte, Brazil.
| |
Collapse
|
35
|
Campisciano G, Zanotta N, Quadrifoglio M, Careri A, Torresani A, Cason C, De Seta F, Ricci G, Comar M, Stampalija T. The Bacterial DNA Profiling of Chorionic Villi and Amniotic Fluids Reveals Overlaps with Maternal Oral, Vaginal, and Gut Microbiomes. Int J Mol Sci 2023; 24:ijms24032873. [PMID: 36769194 PMCID: PMC9917689 DOI: 10.3390/ijms24032873] [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: 12/16/2022] [Revised: 01/27/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
The in utero microbiome hypothesis has been long debated. This hypothesis will change our comprehension of the pioneer human microbiome if proved correct. In 60 uncomplicated pregnancies, we profiled the microbiome of chorionic villi (CV) and amniotic fluids (AF) in relation to maternal saliva, rectum, and vagina and the soluble cytokines cascade in the vagina, CV and AF. In our series, 12/37 (32%) AF and 10/23 (44%) CV tested positive for bacterial DNA. CV and AF harbored bacterial DNA of Streptococcus and Lactobacillus, overlapping that of the matched oral and vaginal niches, which showed a dysbiotic microbiome. In these pregnant women, the immune profiling revealed an immune hyporesponsiveness in the vagina and a high intraamniotic concentration of inflammatory cytokines. To understand the eventual role of bacterial colonization of the CV and AF and the associated immune response in the pregnancy outcome, further appropriate studies are needed. In this context, further studies should highlight if the hematogenous route could justify the spread of bacterial DNA from the oral microbiome to the placenta and if vaginal dysbiosis could favor the likelihood of identifying CV and AF positive for bacterial DNA.
Collapse
Affiliation(s)
- Giuseppina Campisciano
- Department of Advanced Translational Microbiology, Institute for Maternal and Child Health—IRCCS Burlo Garofolo, Via dell’Istria, 65, 34137 Trieste, Italy
- Correspondence:
| | - Nunzia Zanotta
- Department of Advanced Translational Microbiology, Institute for Maternal and Child Health—IRCCS Burlo Garofolo, Via dell’Istria, 65, 34137 Trieste, Italy
| | - Mariachiara Quadrifoglio
- Unit of Fetal Medicine and Prenatal Diagnosis, Institute for Maternal and Child Health—IRCCS Burlo Garofolo, Via dell’Istria, 65, 34137 Trieste, Italy
| | - Annalisa Careri
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Strada di Fiume 447, 34149 Trieste, Italy
| | - Alessandra Torresani
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Strada di Fiume 447, 34149 Trieste, Italy
| | - Carolina Cason
- Department of Advanced Translational Microbiology, Institute for Maternal and Child Health—IRCCS Burlo Garofolo, Via dell’Istria, 65, 34137 Trieste, Italy
| | - Francesco De Seta
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Strada di Fiume 447, 34149 Trieste, Italy
- Department of Obstetrics and Gynecology, Institute for Maternal and Child Health—IRCCS Burlo Garofolo, Via dell’Istria, 65, 34137 Trieste, Italy
| | - Giuseppe Ricci
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Strada di Fiume 447, 34149 Trieste, Italy
- Department of Obstetrics and Gynecology, Institute for Maternal and Child Health—IRCCS Burlo Garofolo, Via dell’Istria, 65, 34137 Trieste, Italy
| | - Manola Comar
- Department of Advanced Translational Microbiology, Institute for Maternal and Child Health—IRCCS Burlo Garofolo, Via dell’Istria, 65, 34137 Trieste, Italy
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Strada di Fiume 447, 34149 Trieste, Italy
| | - Tamara Stampalija
- Unit of Fetal Medicine and Prenatal Diagnosis, Institute for Maternal and Child Health—IRCCS Burlo Garofolo, Via dell’Istria, 65, 34137 Trieste, Italy
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Strada di Fiume 447, 34149 Trieste, Italy
| |
Collapse
|
36
|
Meyyazhagan A, Kuchi Bhotla H, Pappuswamy M, Tsibizova V, Al Qasem M, Di Renzo GC. Cytokine see-saw across pregnancy, its related complexities and consequences. Int J Gynaecol Obstet 2023; 160:516-525. [PMID: 35810391 DOI: 10.1002/ijgo.14333] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 06/17/2022] [Accepted: 07/07/2022] [Indexed: 01/20/2023]
Abstract
During pregnancy, a woman's immune system adapts to the changing hormonal concentrations, causing immunologic transition. These immunologic changes are required for a full-term pregnancy, preserving the fetus' innate and adaptive immunity. Preterm labor, miscarriage, gestational diabetes mellitus, and pre-eclampsia are all caused by abnormal cytokine expression during pregnancy and childbirth. A disruption in the cytokine balance can lead to autoimmune diseases or microbiologic infections, or to autoimmune illness remission during pregnancy with postpartum recurrence. The cytokine treatments are essential and damaging to the developing fetus. The current review summarizes the known research on cytokine changes during pregnancy and their possible consequences for pregnant women. Studies suggest that customizing medication for each woman and her progesterone levels should be based on the cytokine profile of each pregnant woman. Immune cells and chemicals play an important function in development of the placenta and embryo. During pregnancy, T cells divide and move, and a careful balance between proinflammatory and anti-inflammatory cytokines is necessary. The present review focuses on the mother's endurance in generating fetal cells and the immunologic mechanism involved.
Collapse
Affiliation(s)
- Arun Meyyazhagan
- Center for Perinatal and Reproductive Medicine, University of Perugia, Perugia, Italy.,PREIS International and European School of Perinatal, Neonatal and Reproductive Medicine, Florence, Italy
| | - Haripriya Kuchi Bhotla
- Department of Life Sciences, CHRIST (Deemed to be University), Bengaluru, Karnataka, India
| | - Manikantan Pappuswamy
- Department of Life Sciences, CHRIST (Deemed to be University), Bengaluru, Karnataka, India
| | - Valentina Tsibizova
- PREIS International and European School of Perinatal, Neonatal and Reproductive Medicine, Florence, Italy.,Institute of Perinatology and Pediatrics, Almazov National Medical Research Center, Saint-Petersburg, Russia
| | - Malek Al Qasem
- PREIS International and European School of Perinatal, Neonatal and Reproductive Medicine, Florence, Italy.,Department of Obstetrics and Gynecology, Faculty of Medicine, Mutah University, Al-Karak, Jordan
| | - Gian Carlo Di Renzo
- Center for Perinatal and Reproductive Medicine, University of Perugia, Perugia, Italy.,PREIS International and European School of Perinatal, Neonatal and Reproductive Medicine, Florence, Italy.,Department of Obstetrics and Gynecology, I.M. Sechenov First State University of Moscow, Moscow, Russia
| |
Collapse
|
37
|
Li QH, Kim K, Shresta S. Mouse models of Zika virus transplacental transmission. Antiviral Res 2023; 210:105500. [PMID: 36567026 PMCID: PMC9852097 DOI: 10.1016/j.antiviral.2022.105500] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 12/14/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022]
Abstract
Seven years after the onset of the Zika virus (ZIKV) epidemic in the Americas, longitudinal studies are beginning to demonstrate that children infected in utero and born without severe birth defects exhibit motor skill deficits at up to 3 years of age. Long term health and socioeconomic impacts of fetal ZIKV infection appear imminent. ZIKV continues to circulate in low levels much as the virus did for decades prior to the 2015 epidemic, and the timing of the ZIKV outbreak is unknown. Thus, in the continued absence of ZIKV vaccines or antivirals, small animal models of ZIKV transplacental transmission have never been more necessary to test antiviral strategies for both mother and fetuses, and to elucidate mechanisms of immunity at the maternal-fetal interface. Here we review the state of ZIKV transplacental transmission models, highlight key unanswered questions, and set goals for the next generation of mouse models.
Collapse
Affiliation(s)
- Qin Hui Li
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA, 92037, USA
| | - Kenneth Kim
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA, 92037, USA
| | - Sujan Shresta
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA, 92037, USA.
| |
Collapse
|
38
|
Carolina V, Mariangeles C, Delia W, Monica G, Mirta K, Claudio B. Integrins and ligands, are correlated at pig placental interface during pregnancy? REPRODUCTION AND FERTILITY 2023; 4:RAF-22-0079. [PMID: 36795678 PMCID: PMC10083652 DOI: 10.1530/raf-22-0079] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 02/16/2023] [Indexed: 02/17/2023] Open
Abstract
In the present work, we emphasize the studies about integrins and their receptors in pig placental interface at different times of gestation. Uterine placental interface (n=24) of 17-, 30-, 60- and 70-days gestation (dg) and non-pregnant uterus (n=4) of crossbred sows were used. The presence of αvβ3 and α5β1 integrins, and their ligands fibronectin (FN), and osteopontin (OPN) were detected by immunohistochemistry, and the immunolabelled area percentage (IAP) and the optical density (OD) were determined. The integrins and its ligands analyzed have presented peaks of expression in early and mid-gestation, both in IAP and the OD area decreasing at 70 dg. These temporal changes showed us that the molecules studied in this work participate in embryo/feto-maternal attachment, variably. Besides, we found a significant correlation both in the intensity and in the extension of immunostaining for trophoblastic FN and endometrial αvβ3, and trophoblastic OPN and endometrial α5β1, throughout the entire pig pregnancy. At late gestation, take place a notable placental remodelation with subsequent removal or renewal of folds at the uterine-placental interface that results in the loss of focal adhesions. The decrease of the expression of some integrins and their ligands in late gestation, particularly at 70 dg, would demonstrate that there would be other adhesion molecules and other ligands that could be participating in the establishment of the maternal-fetal interface.
Collapse
Affiliation(s)
- Velez Carolina
- Faculty of Veterinary Science, National University of La Pampa (UNLPam), Argentina
- National Scientific and Technical Research Council of Argentina (CONICET), Argentina
| | - Clauzure Mariangeles
- Faculty of Veterinary Science, National University of La Pampa (UNLPam), Argentina
- National Scientific and Technical Research Council of Argentina (CONICET), Argentina
| | - Williamson Delia
- Faculty of Veterinary Science, National University of La Pampa (UNLPam), Argentina
| | - Garcia Monica
- Faculty of Veterinary Science, National University of La Pampa (UNLPam), Argentina
| | - Koncurat Mirta
- Faculty of Veterinary Science, National University of La Pampa (UNLPam), Argentina
| | - Barbeito Claudio
- National Scientific and Technical Research Council of Argentina (CONICET), Argentina
- Laboratory of Descriptive, Comparative and Experimental Histology and Embriology (LHYEDEC). Faculty of Veterinary Science, National University of La Plata (UNLP), Argentina
| |
Collapse
|
39
|
Low Interferon-γ Levels in Cord and Peripheral Blood of Pregnant Women Infected with SARS-CoV-2. Microorganisms 2023; 11:microorganisms11010223. [PMID: 36677515 PMCID: PMC9861455 DOI: 10.3390/microorganisms11010223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/12/2023] [Accepted: 01/12/2023] [Indexed: 01/19/2023] Open
Abstract
COVID-19 is characterized by the immune system's overreaction resulting in a 'cytokine storm', consisting in a massive release of cytokine into the bloodstream, leading to local and systemic inflammatory response. This clinical picture is further complicated in case of infection of patients with a peculiar immunological status, such as pregnancy. In this paper, we focused on Interferon-γ, which plays a pivotal immunomodulatory role in normal pregnancy and fetal development, as well as in defense against pathogens. In this study, we compared the levels of Interferon-γ and the Interferon autoantibodies of the peripheral and cord blood of pregnant women with confirmed mild COVID-19 and healthy pregnant women. The Interferon-γ was significantly lower both in the peripheral and cord blood of SARS-CoV-2-positive mothers, suggesting that infection can affect the fetal microenvironment even without severe maternal symptoms. In conclusion, further studies are needed to clarify whether lower levels of Interferon-γ due to SARS-CoV-2 infection affect the development or infection susceptibility of infants born to SARS-CoV-2-infected mothers.
Collapse
|
40
|
Harrison TD, Chaney EM, Brandt KJ, Ault-Seay TB, Payton RR, Schneider LG, Strickland LG, Schrick FN, McLean KJ. The effects of nutritional level and body condition score on cytokines in seminal plasma of beef bulls. FRONTIERS IN ANIMAL SCIENCE 2023. [DOI: 10.3389/fanim.2022.1078960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
IntroductionHigh quality semen is essential for reproductive efficiency. Nutrition and environmental factors impact the ejaculate components, like cytokines, that are essential for pregnancy establishment. We hypothesized that differing nutritional periods and body condition scores would affect seminal plasma cytokine concentrations of bulls.MethodsMature Angus bulls (n=11) were individually housed and randomly assigned to either over-fed (n=5) or restricted (n=6) treatment pathways. Bulls were fed different volumes of a single ration creating 8 individual treatment periods. Body weight and body condition scores were taken every 14 d to manage intake volumes. Ejaculates were collected every 84 d to determine seminal plasma cytokine profiles. A complete randomized design was used to evaluate seminal plasma cytokines after each nutritional treatment. Initial cytokine concentrations and volume of the ejaculate were included as covariates.ResultsAll cytokines returned to initial concentrations following maintenance treatments at an ideal body condition score of 6. Nutritional treatments affected (P ≤ 0.05) IFN-γ, IL-8, MIP-1α, MIP-1β, TNF-α, IL-1β, and VEGF-A. However, TNF-α, IFN-γ, and MIP-1α had the greatest impact on cytokine profile.DiscussionNutritional levels and adiposity altered seminal plasma cytokine concentrations which could potentially impact the inflammatory balance of the uterus and the immune responses necessary for pregnancy establishment.
Collapse
|
41
|
Bensussen A, Torres-Magallanes JA, Roces de Álvarez-Buylla E. Molecular tracking of insulin resistance and inflammation development on visceral adipose tissue. Front Immunol 2023; 14:1014778. [PMID: 37026009 PMCID: PMC10070947 DOI: 10.3389/fimmu.2023.1014778] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 02/27/2023] [Indexed: 04/08/2023] Open
Abstract
Background Visceral adipose tissue (VAT) is one of the most important sources of proinflammatory molecules in obese people and it conditions the appearance of insulin resistance and diabetes. Thus, understanding the synergies between adipocytes and VAT-resident immune cells is essential for the treatment of insulin resistance and diabetes. Methods We collected information available on databases and specialized literature to construct regulatory networks of VAT resident cells, such as adipocytes, CD4+ T lymphocytes and macrophages. These networks were used to build stochastic models based on Markov chains to visualize phenotypic changes on VAT resident cells under several physiological contexts, including obesity and diabetes mellitus. Results Stochastic models showed that in lean people, insulin produces inflammation in adipocytes as a homeostatic mechanism to downregulate glucose intake. However, when the VAT tolerance to inflammation is exceeded, adipocytes lose insulin sensitivity according to severity of the inflammatory condition. Molecularly, insulin resistance is initiated by inflammatory pathways and sustained by intracellular ceramide signaling. Furthermore, our data show that insulin resistance potentiates the effector response of immune cells, which suggests its role in the mechanism of nutrient redirection. Finally, our models show that insulin resistance cannot be inhibited by anti-inflammatory therapies alone. Conclusion Insulin resistance controls adipocyte glucose intake under homeostatic conditions. However, metabolic alterations such as obesity, enhances insulin resistance in adipocytes, redirecting nutrients to immune cells, permanently sustaining local inflammation in the VAT.
Collapse
Affiliation(s)
- Antonio Bensussen
- *Correspondence: Antonio Bensussen, ; Elena Roces de Álvarez-Buylla,
| | | | | |
Collapse
|
42
|
Vélez C, Clauzure M, Williamson D, Koncurat MA, Barbeito C. IFN-γ and IL-10: seric and placental profile during pig gestation Seric and placental cytokines in pig gestation. AN ACAD BRAS CIENC 2023; 95:e20201160. [PMID: 37075349 DOI: 10.1590/0001-3765202320201160] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 10/25/2020] [Indexed: 04/21/2023] Open
Abstract
Concentration of interferon-gamma and interleukin-10 in maternal serum and in maternal and fetal porcine placental extracts from different gestation periods was determined. Crossbred pigs' placental samples of 17, 30, 60, 70, and 114 days gestation and non-pregnant uteri were used. Interferon-gamma concentration was increased at the placental interface at 17 days, in maternal and fetal placenta, and decreased significantly in the remaining gestation periods. Interferon-gamma showed a peak in serum at 60 days. Regarding interleukin-10, placental tissue concentrations were unaltered, there were no significant differences with non-gestating uteri samples. In serum interleukin-10 increased at 17, 60, and 114 days gestation. At 17 days there are uterus structural and molecular changes that allow the embryos implantation and placenta development. The presence of interferon-gamma found at this moment in the interface would favor that placental growth. Moreover, its significant increase in serum at 60 days, would generate a proinflammatory cytokine pattern that facility the placental remodeling characteristic of this moment of porcine gestation. On the other hand, a significant interleukin-10 increase in serum at 17, 60 and 114 days could indicate its immunoregulatory role at a systemic level during pig gestation.
Collapse
Affiliation(s)
- Carolina Vélez
- Faculty of Veterinary Science, National University of La Pampa (UNLPam), Calle 5, 116, General Pico, 6360 La Pampa, Argentina
- National Scientific and Technical Research Council (CONICET), Godoy Cruz 2290, C1425FQB Buenos Aires, Argentina
| | - Mariángeles Clauzure
- Faculty of Veterinary Science, National University of La Pampa (UNLPam), Calle 5, 116, General Pico, 6360 La Pampa, Argentina
- National Scientific and Technical Research Council (CONICET), Godoy Cruz 2290, C1425FQB Buenos Aires, Argentina
| | - Delia Williamson
- Faculty of Veterinary Science, National University of La Pampa (UNLPam), Calle 5, 116, General Pico, 6360 La Pampa, Argentina
| | - Mirta A Koncurat
- Faculty of Veterinary Science, National University of La Pampa (UNLPam), Calle 5, 116, General Pico, 6360 La Pampa, Argentina
| | - Claudio Barbeito
- National Scientific and Technical Research Council (CONICET), Godoy Cruz 2290, C1425FQB Buenos Aires, Argentina
- Laboratory of Descriptive, Comparative and Experimental Histology and Embryology, School of Veterinary Science, National University of La Plata, Av. 60, 118, B1900 La Plata, Buenos Aires, Argentina
| |
Collapse
|
43
|
Oravecz O, Romero R, Tóth E, Kapitány J, Posta M, Gallo DM, Rossi SW, Tarca AL, Erez O, Papp Z, Matkó J, Than NG, Balogh A. Placental galectins regulate innate and adaptive immune responses in pregnancy. Front Immunol 2022; 13:1088024. [PMID: 36643922 PMCID: PMC9832025 DOI: 10.3389/fimmu.2022.1088024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 12/05/2022] [Indexed: 12/29/2022] Open
Abstract
Introduction Galectins are master regulators of maternal immune responses and placentation in pregnancy. Galectin-13 (gal-13) and galectin-14 (gal-14) are expressed solely by the placenta and contribute to maternal-fetal immune tolerance by inducing the apoptosis of activated T lymphocytes and the polarization of neutrophils toward an immune-regulatory phenotype.Furthermore, their decreased placental expression is associated with pregnancy complications, such as preeclampsia and miscarriage. Yet, our knowledge of the immunoregulatory role of placental galectins is incomplete. Methods This study aimed to investigate the effects of recombinant gal-13 and gal-14 on cell viability, apoptosis, and cytokine production of peripheral blood mononuclear cells (PBMCs) and the signaling pathways involved. Results Herein, we show that gal-13 and gal-14 bind to the surface of non-activated PBMCs (monocytes, natural killer cells, B cells, and T cells) and increase their viability while decreasing the rate of their apoptosis without promoting cell proliferation. We also demonstrate that gal-13 and gal-14 induce the production of interleukin (IL)-8, IL-10, and interferon-gamma cytokines in a concentration-dependent manner in PBMCs. The parallel activation of Erk1/2, p38, and NF-ĸB signaling evidenced by kinase phosphorylation in PBMCs suggests the involvement of these pathways in the regulation of the galectin-affected immune cell functions. Discussion These findings provide further evidence on how placenta-specific galectins assist in the establishment and maintenance of a proper immune environment during a healthy pregnancy.
Collapse
Affiliation(s)
- Orsolya Oravecz
- Systems Biology of Reproduction Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary,Doctoral School of Biology, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Roberto Romero
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Detroit, MI, United States,Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, United States,Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, United States,Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, United States,Detroit Medical Center, Detroit, MI, United States
| | - Eszter Tóth
- Systems Biology of Reproduction Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Judit Kapitány
- Systems Biology of Reproduction Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Máté Posta
- Systems Biology of Reproduction Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary,Károly Rácz Doctoral School of Clinical Medicine, Semmelweis University, Budapest, Hungary
| | - Dahiana M. Gallo
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Detroit, MI, United States,Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, United States,Department of Obstetrics and Gynecology, Universidad Del Valle, Cali, Colombia
| | | | - Adi L. Tarca
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Detroit, MI, United States,Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, United States,Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, United States,Genesis Theranostix Group, Budapest, Hungary
| | - Offer Erez
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Detroit, MI, United States,Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, United States,Genesis Theranostix Group, Budapest, Hungary,Department of Obstetrics and Gynecology, Soroka University Medical Center, Beer Sheva, Israel
| | - Zoltán Papp
- Department of Obstetrics and Gynecology, Semmelweis University, Budapest, Hungary,Maternity Private Clinic of Obstetrics and Gynecology, Budapest, Hungary
| | - János Matkó
- Systems Biology of Reproduction Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Nándor Gábor Than
- Systems Biology of Reproduction Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary,Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Detroit, MI, United States,Genesis Theranostix Group, Budapest, Hungary,Department of Obstetrics and Gynecology, Semmelweis University, Budapest, Hungary,Maternity Private Clinic of Obstetrics and Gynecology, Budapest, Hungary,*Correspondence: Nándor Gábor Than,
| | - Andrea Balogh
- Systems Biology of Reproduction Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| |
Collapse
|
44
|
Wang W, Cao C, Zhang B, Wang F, Deng D, Cao J, Li H, Yu M. Integrating Transcriptomic and ChIP-Seq Reveals Important Regulatory Regions Modulating Gene Expression in Myometrium during Implantation in Pigs. Biomolecules 2022; 13:biom13010045. [PMID: 36671430 PMCID: PMC9856092 DOI: 10.3390/biom13010045] [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: 10/12/2022] [Revised: 12/17/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022] Open
Abstract
The myometrium is the outer layer of the uterus. Its contraction and steroidogenic activities are required for embryo implantation. However, the molecular mechanisms underlying its functions remain unknown in pigs. The myometrium includes the inner circular muscle (CM) and the outer longitudinal muscle (LM) layers. In this study, we collected the CM and LM samples from the mesometrial side (named M) of the uterus on days 12 (pre-implantation stage) and 15 (implantation stage) of pregnancy and day 15 of the estrous cycle. The transcriptomic results revealed distinct differences between the uterine CM and LM layers in early pregnancy: the genes expressed in the LM layer were mainly related to contraction pathways, whereas the transcriptional signatures in the CM layer on day 15 of pregnancy were primarily involved in the immune response processes. Subsequent comparisons in the CM layer between pregnant and cyclic gilts show that the transcriptional signatures of the CM layer are implantation-dependent. Next, we investigated the genome-wide profiling of histone H3 lysine 27 acetylation (H3K27ac) and histone H3 lysine 4 trimethylation (H3K4me3) in pig uterine CM and LM layers. The genomic regions that had transcriptional activity and were associated with the expression of genes in the two layers were characterized. Taken together, the regulatory regions identified in the study may contribute to modulating the gene expression in pig uterine CM and LM layers during implantation.
Collapse
Affiliation(s)
- Weiwei Wang
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Caiqin Cao
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Botao Zhang
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, Foshan University, Foshan 528225, China
| | - Feiyu Wang
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Dadong Deng
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Jianhua Cao
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Hua Li
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, Foshan University, Foshan 528225, China
| | - Mei Yu
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
- Correspondence:
| |
Collapse
|
45
|
Unravelling the genetics of non-random fertilization associated with gametic incompatibility. Sci Rep 2022; 12:22314. [PMID: 36566278 PMCID: PMC9789956 DOI: 10.1038/s41598-022-26910-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 12/21/2022] [Indexed: 12/25/2022] Open
Abstract
In the dairy industry, mate allocation is dependent on the producer's breeding goals and the parents' breeding values. The probability of pregnancy differs among sire-dam combinations, and the compatibility of a pair may vary due to the combination of gametic haplotypes. Under the hypothesis that incomplete incompatibility would reduce the odds of fertilization, and complete incompatibility would lead to a non-fertilizing or lethal combination, deviation from Mendelian inheritance expectations would be observed for incompatible pairs. By adding an interaction to a transmission ratio distortion (TRD) model, which detects departure from the Mendelian expectations, genomic regions linked to gametic incompatibility can be identified. This study aimed to determine the genetic background of gametic incompatibility in Holstein cattle. A total of 283,817 genotyped Holstein trios were used in a TRD analysis, resulting in 422 significant regions, which contained 2075 positional genes further investigated for network, overrepresentation, and guilt-by-association analyses. The identified biological pathways were associated with immunology and cellular communication and a total of 16 functional candidate genes were identified. Further investigation of gametic incompatibility will provide opportunities to improve mate allocation for the dairy cattle industry.
Collapse
|
46
|
The Update Immune-Regulatory Role of Pro- and Anti-Inflammatory Cytokines in Recurrent Pregnancy Losses. Int J Mol Sci 2022; 24:ijms24010132. [PMID: 36613575 PMCID: PMC9820098 DOI: 10.3390/ijms24010132] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 12/12/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022] Open
Abstract
Recurrent pregnancy losses (RPL) is a common reproductive disorder with various underlying etiologies. In recent years, rapid progress has been made in exploring the immunological mechanisms for RPL. A propensity toward Th2 over Th1 and regulatory T (Treg) over Th17 immune responses may be advantageous for reproductive success. In women with RPL and animals prone to abortion, an inordinate expression of cytokines associated with implantation and early embryo development is present in the endometrium or decidua secreted from immune and non-immune cells. Hence, an adverse cytokine milieu at the maternal-fetal interface assaults immunological tolerance, leading to fetal rejection. Similar to T cells, NK cells can be categorized based on the characteristics of cytokines they secrete. Decidual NK (dNK) cells of RPL patients exhibited an increased NK1/NK2 ratio (IFN-γ/IL-4 producing NK cell ratios), leading to pro-inflammatory cytokine milieu and increased NK cell cytotoxicity. Genetic polymorphism may be the underlying etiologies for Th1 and Th17 propensity since it alters cytokine production. In addition, various hormones participate in cytokine regulations, including progesterone and estrogen, controlling cytokine balance in favor of the Th2 type. Consequently, the intricate regulation of cytokines and hormones may prevent the RPL of immune etiologies. Local or systemic administration of cytokines or their antagonists might help maintain adequate cytokine milieu, favoring Th2 over Th1 response or Treg over Th17 immune response in women with RPL. Herein, we provided an updated comprehensive review regarding the immune-regulatory role of pro- and anti-inflammatory cytokines in RPL. Understanding the roles of cytokines involved in RPL might significantly advance the early diagnosis, monitoring, and treatment of RPL.
Collapse
|
47
|
Wang L, Yuan X, Zhou X. Expression pattern and clinical significance of microRNA-let-7a and IFN-gamma in placental tissue of patients with preeclampsia with severe features. J Perinat Med 2022; 50:1142-1149. [PMID: 35596257 DOI: 10.1515/jpm-2021-0133] [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: 03/24/2021] [Accepted: 04/03/2022] [Indexed: 11/15/2022]
Abstract
OBJECTIVES Preeclampsia with severe features (PECsf) is a common disease in pregnant women. let-7a and IFN-gamma (interferon-gamma) are involved in diagnosis and prognosis of preeclampsia. This study explored effects of let-7a and IFN-gamma on PECsf patients. METHODS The placental tissue of 21 PECsf, 19 preeclampsia without severe features (PEC), and 20 normal pregnant women were collected, and clinical data were recorded. let-7a and IFN-gamma expressions in placental tissue were detected. The correlation between let-7a/IFN-gamma expression and clinical indexes was analyzed. According to let-7a and IFN-gamma expressions, PECsf patients were assigned into Hlet-7a group (let-7a high expression group), Llet-7a group (let-7a low expression group), HIFN-gamma group (IFN-gamma high expression group) and LIFN-gamma group (IFN-gamma low expression group). The incidence of adverse prognosis was compared. RESULTS let-7a and IFN-gamma were highly expressed in placental tissue of preeclampsia patients, with significant differences between PEC and PECsf. The high expressions of let-7a and IFN-gamma were positively correlated with mean arterial pressure, lactate dehydrogenase, and 24 h urinary protein in placental tissues of PECsf patients. High let-7a and IFN-gamma expressions were correlated with adverse outcomes of PECsf. CONCLUSIONS High let-7a and IFN-gamma expressions were correlated with clinical features, and could be used as biomarkers for treatment and poor prognosis of PECsf.
Collapse
Affiliation(s)
- Liping Wang
- Department of Obstetrics and Gynecology, Daqing Longnan Hospital, Daqing, Heilongjiang, P.R. China
| | - Xiaojie Yuan
- Department of Obstetrics and Gynecology, Daqing Longnan Hospital, Daqing, Heilongjiang, P.R. China
| | - Xuewu Zhou
- Department of Obstetrics and Gynecology, Daqing Longnan Hospital, Daqing, Heilongjiang, P.R. China
| |
Collapse
|
48
|
Park Y, Han SJ. Interferon Signaling in the Endometrium and in Endometriosis. Biomolecules 2022; 12:biom12111554. [PMID: 36358904 PMCID: PMC9687697 DOI: 10.3390/biom12111554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 10/21/2022] [Indexed: 12/04/2022] Open
Abstract
Endometriosis is an estrogen-dependent inflammatory disease that develops in reproductive-aged women who experience pelvic pain and infertility. Even though endometriosis is not a new disease, its molecular etiology has not been clearly elucidated. Defects in the immune system might be one of the factors that promote endometriosis progression. For example, elevated levels of proinflammatory cytokines are associated with endometriosis. Interferon is one of the cytokines that is elevated in endometriotic tissues compared with normal endometrium. Therefore, high interferon levels play a crucial role in endometriosis progression. In addition to endometriosis, however, interferon has a critical role in endometrial function, particularly in the initiation and maintenance of pregnancy. Therefore, this review describes the double-edged sword of interferon signaling in normal endometrial function versus endometriosis progression and also discusses interferon targeting as a new nonhormonal therapy for endometriosis. This approach may increase the efficacy of endometriosis treatment and reduce the adverse effects associated with current hormonal therapy for this disease.
Collapse
Affiliation(s)
- Yuri Park
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Sang Jun Han
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
- Reproductive Medicine, Baylor College of Medicine, Houston, TX 77030, USA
- Correspondence:
| |
Collapse
|
49
|
Rosini AM, Teixeira SC, Milian ICB, Silva RJ, de Souza G, Luz LC, Gomes AO, Mineo JR, Mineo TWP, Ferro EAV, Barbosa BF. LPS-mediated activation of TLR4 controls Toxoplasma gondii growth in human trophoblast cell (BeWo) and human villous explants in a dependent-manner of TRIF, MyD88, NF-κB and cytokines. Tissue Cell 2022; 78:101907. [DOI: 10.1016/j.tice.2022.101907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 06/22/2022] [Accepted: 08/22/2022] [Indexed: 02/07/2023]
|
50
|
Rudolf Vegas A, Hamdi M, Podico G, Bollwein H, Fröhlich T, Canisso IF, Bauersachs S, Almiñana C. Uterine extracellular vesicles as multi-signal messengers during maternal recognition of pregnancy in the mare. Sci Rep 2022; 12:15616. [PMID: 36114358 PMCID: PMC9481549 DOI: 10.1038/s41598-022-19958-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 09/07/2022] [Indexed: 11/09/2022] Open
Abstract
In contrast to other domestic mammals, the embryo-derived signal(s) leading to maternal recognition of pregnancy (MRP) are still unknow in the mare. We hypothesize that these embryonic signals could be packed into uterine extracellular vesicles (uEVs), acting as multi-signal messengers between the conceptus and the maternal tract, and contributing to MRP. To unveil these signals, the RNA and protein cargos of uEVs isolated from uterine lavages collected from pregnant mares (P; day 10, 11, 12 and 13 after ovulation) and cyclic control mares (C; day 10 and 13 after ovulation) were analyzed. Our results showed a fine-tuned regulation of the uEV cargo (RNAs and proteins), by the day of pregnancy, the estrous cycle, and even the size of the embryo. A particular RNA pattern was identified with specific increase on P12 related to immune system and hormonal response. Besides, a set of proteins as well as RNAs was highly enriched in EVs on P12 and P13. Differential abundance of miRNAs was also identified in P13-derived uEVs. Their target genes were linked to down- or upregulated genes in the embryo and the endometrium, exposing their potential origin. Our study identified for first time specific molecules packed in uEVs, which were previously associated to MRP in the mare, and thus bringing added value to the current knowledge. Further integrative and functional analyses will help to confirm the role of these molecules in uEVs during MRP in the mare.
Collapse
Affiliation(s)
- Alba Rudolf Vegas
- Functional Genomics Group, Institute of Veterinary Anatomy, Vetsuisse Faculty Zurich, University of Zurich, 8315, Lindau, ZH, Switzerland
| | - Meriem Hamdi
- Functional Genomics Group, Institute of Veterinary Anatomy, Vetsuisse Faculty Zurich, University of Zurich, 8315, Lindau, ZH, Switzerland
| | - Giorgia Podico
- Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, 61802, USA
| | - Heinrich Bollwein
- Clinic of Reproductive Medicine, Vetsuisse-Faculty, University of Zurich, 8315, Lindau, ZH, Switzerland
| | - Thomas Fröhlich
- Gene Center, Laboratory for Functional Genome Analysis, LMU Munich, 81377, Munich, Germany
| | - Igor F Canisso
- Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, 61802, USA
| | - Stefan Bauersachs
- Functional Genomics Group, Institute of Veterinary Anatomy, Vetsuisse Faculty Zurich, University of Zurich, 8315, Lindau, ZH, Switzerland
| | - Carmen Almiñana
- Functional Genomics Group, Institute of Veterinary Anatomy, Vetsuisse Faculty Zurich, University of Zurich, 8315, Lindau, ZH, Switzerland.
| |
Collapse
|