1
|
Ma F, Feng X, Feng S, Liu J, Li J, Mo L, Xu L, Liu Y, Wu J, Yang P, Ning Y. Impaired inducibility of immune regulatory capacity of peripheral B cells of patients with recurrent pregnancy loss. Immunol Res 2024:10.1007/s12026-024-09549-7. [PMID: 39495420 DOI: 10.1007/s12026-024-09549-7] [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: 05/21/2024] [Accepted: 09/25/2024] [Indexed: 11/05/2024]
Abstract
The pathogenesis of recurrent pregnancy loss (RPL) is unclear. RPL may have an association with disruption of immune tolerance. The aim of this study is to characterize the inducibility of immune regulatory ability in peripheral naïve B cells of patients with RPL. In this study, blood samples were taken from patients with RPL. B220+ B cells were isolated by flow cytometry cell sorting. The gene profile of B cells was analyzed using RNA sequencing (RNAseq). The results showed that peripheral B220+ B cells of RPL patients had lower expression of IL10 and exacerbated ER stress. The induction of IL10 expression in peripheral B220+ B cells of RPL patients were impaired. High ubiquitination of c-Maf inducing protein (CMIP) was detected in RPL B cells. Exposure to thapsigargin (an ER stress agonist) decreased the amount of CMIP in B cells. The effects of ER stress on reducing CMIP quantity in B cells were mediated by the histone H2B E3 ubiquitin ligase ring finger protein 20 (RNF20). Inhibition of RNF20 or ER stress restored the inducibility of immune regulatory functions of B220+ B cells of RPL patients. In summary, peripheral B cells in patients with RPL show impaired immune regulation capacity, in which exacerbated ER stress plays a crucial role. Regulation of ER stress or inhibition of RNF20 can restore the immune regulatory capacity in the B cells.
Collapse
Affiliation(s)
- Fei Ma
- Department of Chinese Traditional Medicine, Affiliated Shenzhen Maternity & Child Healthcare Hospital, Southern Medical University, Shenzhen, China.
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China.
| | - Xiaoyang Feng
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Shiyu Feng
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Jin Liu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Jia Li
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Lihua Mo
- Department of General Practice Medicine, Third Affiliated Hospital of Shenzhen University, Shenzhen, China
- Institute of Allergy & Immunology of Shenzhen University, State Key Laboratory of Respiratory Diseases Allergy Division, Shenzhen University, Room A7-509 at Lihu Campus of Shenzhen University, 1066 Xueyuan Blvd, Shenzhen, 518055, China
| | - Lingzhi Xu
- Department of Immunology, School of Basic Medical Sciences, Weifang Medical University, Weifang, China
| | - Yulei Liu
- Department of Chinese Traditional Medicine, Affiliated Shenzhen Maternity & Child Healthcare Hospital, Southern Medical University, Shenzhen, China
| | - Jiaman Wu
- Department of Chinese Traditional Medicine, Affiliated Shenzhen Maternity & Child Healthcare Hospital, Southern Medical University, Shenzhen, China
| | - Pingchang Yang
- Institute of Allergy & Immunology of Shenzhen University, State Key Laboratory of Respiratory Diseases Allergy Division, Shenzhen University, Room A7-509 at Lihu Campus of Shenzhen University, 1066 Xueyuan Blvd, Shenzhen, 518055, China.
| | - Yan Ning
- Department of Chinese Traditional Medicine, Affiliated Shenzhen Maternity & Child Healthcare Hospital, Southern Medical University, Shenzhen, China.
| |
Collapse
|
2
|
Zhao X, Hu Y, Xiao W, Ma Y, Shen D, Jiang Y, Shen Y, Wang S, Ma J. Efficacy of mesenchymal stromal cells in the treatment of unexplained recurrent spontaneous abortion in mice: An analytical and systematic review of meta-analyses. PLoS One 2023; 18:e0294855. [PMID: 38011163 PMCID: PMC10681256 DOI: 10.1371/journal.pone.0294855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 11/08/2023] [Indexed: 11/29/2023] Open
Abstract
OBJECTIVES Unexplained recurrent spontaneous abortion (URSA) remains an intractable reproductive dilemma due to the lack of understanding of the pathogenesis. This study aimed to evaluate the preclinical evidence for the mesenchymal stromal cell (MSC) treatment for URSA. METHODS A meticulous literature search was independently performed by two authors across the Cochrane Library, EMBASE, and PubMed databases from inception to April 9, 2023. Each study incorporated was assessed using the Systematic Review Centre for Laboratory Animal Experimentation (SYRCLE) risk of bias tool. The amalgamated standardized mean difference (SMD) accompanied by 95% confidence interval (CI) were deduced through a fixed-effects or random-effects model analysis. RESULTS A total of ten studies incorporating 140 mice were subjected to data analysis. The MSC treatment yielded a significant reduction in the abortion rate within the URSA model (OR = 0.23, 95%CI [0.17, 0.3], P<0.00001). Moreover, it elicited a positive modulatory impact on the expression profiles of several inflammatory cytokines in the decidual tissue of URSA murine models, inclusive of IL4 (SMD 1.63, 95% CI [0.39, 2.86], P = 0.01), IL10 (SMD 1.60, 95% CI [0.58, 2.61], P = 0.002), IFN-γ (SMD -1.66, 95%CI [-2.79, -0.52], P = 0.004), and TNF-α (SMD -1.98, 95% CI [-2.93, -1.04], P< 0.0001). Subgroup analyses underscored that the administration mode of intraperitoneal and uterine horn injections, and sources of bone MSCs and adipose-derived MSCs contributed positively to the expression of IL4, IL10, and decreased the expression of IFN-γ in decidual tissue of URSA (P<0.05). Conversely, the tail vein injections subgroup was observed with no statistical significance (P>0.05). CONCLUSIONS The findings underscore the considerable potential of MSCs in URSA therapy. Nonetheless, the demand for enhanced transparency in research design and direct comparisons between various MSC sources and administration routes in URSA is paramount to engendering robust evidence that could pave the way for successful clinical translation.
Collapse
Affiliation(s)
- Xiaoxuan Zhao
- Department of Traditional Chinese Medicine (TCM) Gynecology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Zhejiang Province, Hangzhou, 310007, China
| | - Yijie Hu
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Zhejiang Province, Hangzhou, 310053, China
| | - Wenjun Xiao
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Zhejiang Province, Hangzhou, 310053, China
| | - Yiming Ma
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Zhejiang Province, Hangzhou, 310053, China
| | - Dan Shen
- Department of Traditional Chinese Medicine (TCM) Gynecology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Zhejiang Province, Hangzhou, 310007, China
| | - Yuepeng Jiang
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Zhejiang Province, Hangzhou, 310053, China
| | - Yi Shen
- Department of Traditional Chinese Medicine (TCM) Gynecology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Zhejiang Province, Hangzhou, 310007, China
| | - Suxia Wang
- Department of Traditional Chinese Medicine (TCM) Gynecology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Zhejiang Province, Hangzhou, 310007, China
| | - Jing Ma
- Department of Traditional Chinese Medicine (TCM) Gynecology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Zhejiang Province, Hangzhou, 310007, China
| |
Collapse
|
3
|
Gallino L, Hauk V, Castagnola L, Vota D, Pascuali N, Parborell F, May M, Fontana V, Merech F, Naguila Z, Waschek J, Leirós CP, Ramhorst R. Vasoactive intestinal peptide deficiency promotes ovarian dysfunction associated to a proinflammatory microenvironment reminiscent of premature aging. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166585. [PMID: 36423894 DOI: 10.1016/j.bbadis.2022.166585] [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: 03/07/2022] [Revised: 10/15/2022] [Accepted: 10/20/2022] [Indexed: 11/23/2022]
Abstract
Complex immune regulation during pregnancy is required to ensure a successful pregnancy outcome. Vasoactive intestinal peptide (VIP) has local immunoregulatory effects on the ovary, uterus and maternal-fetal interface that favor a tolerogenic maternal microenvironment. Since the VIP Knockout (KO) mice are subfertile, we investigated the mechanisms underlying the effects of VIP deficiency on ovarian physiology and immune homeostasis. Therefore, we studied VIP KO, deficient (HT) and wild type (WT) female mice in estrus at 3 or 8 months of age. Young KO mice showed abnormal cycle timing and regularity associated with dysfunctional ovaries. Ovaries presented higher number of atretic follicles and reduced number of corpora lutea leading to a lower ovulation rates. Part of the VIP KO mice (25 %) failed to ovulate or ovulated oocytes incompetent to be fertilized (50 %). In particular, ovaries of young KO mice exhibited features of premature aging accompanied by a pro-inflammatory milieu with increased levels of IL-1β. A unique macrophage subpopulation identified as "foamy macrophages" was found. On the other hand, aged VIP KO females did not gain body weight probably due to the sustained production of E2. Finally, the adoptive transfer of FOXP3+ cells to infertile VIP KO females resulted in their selective recruitment to the ovary. It increased FOXP3/RORγt and TGFβ/IL-6 ratio improving ovarian microenvironment and pregnancy rate. The present results suggest that VIP contributes to ovarian homeostatic mechanisms required for a successful pregnancy.
Collapse
Affiliation(s)
- Lucila Gallino
- CONICET, Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires, Argentina
| | - Vanesa Hauk
- CONICET, Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires, Argentina
| | - Lara Castagnola
- CONICET, Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires, Argentina
| | - Daiana Vota
- CONICET, Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires, Argentina
| | | | | | - Maria May
- ININFA, Universidad de Buenos Aires, CONICET, Argentina
| | - Vanina Fontana
- CONICET, Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires, Argentina
| | - Fatima Merech
- CONICET, Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires, Argentina
| | - Zaira Naguila
- CONICET, Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires, Argentina
| | - James Waschek
- The David Geffen School of Medicine, University of California, Los Angeles, USA
| | - Claudia Perez Leirós
- CONICET, Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires, Argentina
| | - Rosanna Ramhorst
- CONICET, Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires, Argentina.
| |
Collapse
|
4
|
Zhang D, Ye Y, Hu X. A non-invasive piTreg-related gene signature for spontaneous tolerance in renal transplantation. Gene X 2023; 848:146901. [DOI: 10.1016/j.gene.2022.146901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 09/11/2022] [Accepted: 09/13/2022] [Indexed: 10/14/2022] Open
|
5
|
The Role and Function of Regulatory T Cells in Toxoplasma gondii-Induced Adverse Pregnancy Outcomes. J Immunol Res 2021; 2021:8782672. [PMID: 34458378 PMCID: PMC8390175 DOI: 10.1155/2021/8782672] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/22/2021] [Accepted: 07/29/2021] [Indexed: 12/31/2022] Open
Abstract
Infection with Toxoplasma gondii (T. gondii) during the pregnant period and its potentially miserable outcomes for the fetus, newborn, and even adult offspring continuously occur worldwide. People acquire infection through the consumption of infected and undercooked meat or contaminated food or water. T. gondii infection in pregnant women primarily during the gestation causes microcephaly, mental and psychomotor retardation, or death. Abnormal pregnancy outcomes are mainly associated with regulatory T cell (Treg) dysfunction. Tregs, a special subpopulation of T cells, function as a vital regulator in maintaining immune homeostasis. Tregs exert a critical effect on forming and maintaining maternal-fetal tolerance and promoting fetal development during the pregnancy period. Forkhead box P3 (Foxp3), a significant functional factor of Tregs, determines the status of Tregs. In this review, we summarize the effects of T. gondii infection on host Tregs and its critical transcriptional factor, Foxp3.
Collapse
|
6
|
Walia B, Kmush BL, Lane SD, Endy T, Montresor A, Larsen DA. Routine deworming during antenatal care decreases risk of neonatal mortality and low birthweight: A retrospective cohort of survey data. PLoS Negl Trop Dis 2021; 15:e0009282. [PMID: 33914732 PMCID: PMC8084140 DOI: 10.1371/journal.pntd.0009282] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 03/02/2021] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Soil transmitted helminths (STH) are a common infection among pregnant women in areas with poor access to sanitation. Deworming medications are cheap and safe; however, the health benefit of deworming during pregnancy is not clear. METHODS / PRINCIPAL FINDINGS We created a retrospective cohort of more than 800,000 births from 95 Demographic and Health Survey datasets to estimate the impact of deworming medicine during routine antenatal care (ANC) on neonatal mortality and low birthweight. We first matched births on the probability of receiving deworming during ANC. We then modeled the birth outcomes with the matched group as a random intercept to estimate the effect of deworming during antenatal care after accounting for various risk factors. We also tested for effect modification of soil transmitted helminth prevalence on the impact of deworming during ANC. Receipt of deworming medication during ANC was associated with a 14% reduction in the risk of neonatal mortality (95% confidence interval = 10-17%, n = 797,772 births), with no difference between high and low transmission countries. In low transmission countries, we found an 11% reduction in the odds of low birth weight (95% confidence interval = 8-13%) for women receiving deworming medicine, and in high transmission countries, we found a 2% reduction in the odds of low birthweight (95% confidence interval = 0-5%). CONCLUSIONS / SIGNIFICANCE These results suggest a substantial health benefit for deworming during ANC that may be even greater in countries with low STH transmission.
Collapse
Affiliation(s)
- Bhavneet Walia
- Syracuse University Department of Public Health, Syracuse, New York, United States of America
| | - Brittany L. Kmush
- Syracuse University Department of Public Health, Syracuse, New York, United States of America
| | - Sandra D. Lane
- Syracuse University Department of Public Health, Syracuse, New York, United States of America
| | - Timothy Endy
- State University of New York Upstate Medical University Department of Infectious Disease, Syracuse, New York, United States of America
| | - Antonio Montresor
- Department of Control of Neglected Tropical Diseases, World Health Organization, Geneva, Switzerland
| | - David A. Larsen
- Syracuse University Department of Public Health, Syracuse, New York, United States of America
| |
Collapse
|
7
|
On use of animal models. Emerg Top Life Sci 2020; 4:207-227. [PMID: 32691841 DOI: 10.1042/etls20200042] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 06/25/2020] [Accepted: 06/29/2020] [Indexed: 12/16/2022]
Abstract
Human pregnancy, critical for our species survival, is inefficient and prone to complications such as infertility, spontaneous miscarriages and preeclampsia (PE). Immunological factors may be important as the embryo is 50% paternal and foreign to the mother. Mouse pregnancy models, and in particular the murine CBA/J x DBA/2 mating combination, has been widely used to investigate mechanisms causing and preventing partner-specific recurrent miscarriages (RM) and PE. Occult losses can represent T cell-mediated rejection, and antigen-specific regulatory T cells (Tregs) with classical αβ T cell receptors (TcR) activated by semen antigens at the time of mating are protective. If there is no occult loss, an inadequate Treg response can also predispose to RM. In RM, proinflammatory cytokines from natural killer (NK)-type cells and macrophages of the innate immune system are responsible and cells with γδ TcR protect via release of TGF-β-type molecules. Immunization of abortion-prone female CBA/J mice or administration of cell-associated or soluble CD200, an immune check point inhibitor, can prevent abortions by augmenting uterine decidual suppressor cell activity. Human studies suggest that is also true in couples with RM. Environmental activators of the innate immune system, such as bacterial LPS and stress, can cause abortions as well as occult losses. The endogenous level of Tregs and activation of Tregs specific for the male H-Y antigen may determine success rates and alter the male:female birth ratio. Intralipid alters LPS clearance, prevents abortions in the CBAxDBA/2 model, and is effective in increasing live birth rates in couples undergoing IVF treatment.
Collapse
|
8
|
Plowden TC, Connell MT, Hill MJ, Mendola P, Kim K, Nobles CJ, Kuhr DL, Galai N, Gibbins KJ, Silver RM, Wilcox B, Sjaarda L, Perkins NJ, Schisterman EF, Mumford SL. Family history of autoimmune disease in relation to time-to-pregnancy, pregnancy loss, and live birth rate. J Transl Autoimmun 2020; 3:100059. [PMID: 32743539 PMCID: PMC7388372 DOI: 10.1016/j.jtauto.2020.100059] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 05/27/2020] [Indexed: 12/27/2022] Open
Abstract
Several autoimmune conditions have adverse effects on reproductive outcomes, but the relationship between family history of autoimmune disease in women without these conditions and pregnancy is uncertain. The objective of this study was to determine if there is an association between a family history of an autoimmune condition and time-to-pregnancy (TTP), pregnancy loss, and live birth. This was a prospective cohort study from a RCT of 1228 adult women ages 18-40, who were healthy, had no history of infertility, were actively attempting to conceive, and had one or two prior pregnancy losses. Of these, 1172 women had data available regarding family history of autoimmune conditions. Women with an affected first-degree relative had similar TTP when compared to those without a FHx (fecundability odds ratio 0.90, 95% confidence interval [CI] 0.70, 1.15). Women with an affected first-degree relative had a lower likelihood of live birth (relative risk [RR] 0.83, 95% CI 0.69, 0.99). Among women who achieved pregnancy, FHx of autoimmune disease was associated with a higher likelihood of pregnancy loss (RR 1.49, 95% CI 1.10, 2.03). Women who had a first-degree relative with an autoimmune disease had a similar TTP as unaffected women but a lower likelihood of live birth and higher risk of pregnancy loss. This information may encourage clinicians to evaluate women with a family history of autoimmune conditions prior to pregnancy and highlights the need for further studies to ascertain the effects of autoimmunity and pregnancy.
Collapse
Affiliation(s)
- Torie C Plowden
- Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, 6710B Rockledge Dr. MSC 7004, Bethesda, MD 20892, United States.,Program in Reproductive Endocrinology and Gynecology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, 10 Center Drive, Bethesda, MD 20892, United States
| | - Matthew T Connell
- Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, 6710B Rockledge Dr. MSC 7004, Bethesda, MD 20892, United States.,Program in Reproductive Endocrinology and Gynecology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, 10 Center Drive, Bethesda, MD 20892, United States
| | - Micah J Hill
- Program in Reproductive Endocrinology and Gynecology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, 10 Center Drive, Bethesda, MD 20892, United States
| | - Pauline Mendola
- Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, 6710B Rockledge Dr. MSC 7004, Bethesda, MD 20892, United States
| | - Keewan Kim
- Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, 6710B Rockledge Dr. MSC 7004, Bethesda, MD 20892, United States
| | - Carrie J Nobles
- Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, 6710B Rockledge Dr. MSC 7004, Bethesda, MD 20892, United States
| | - Daniel L Kuhr
- Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, 6710B Rockledge Dr. MSC 7004, Bethesda, MD 20892, United States.,Department of Obstetrics and Gynecology, University Hospitals Cleveland Medical Center, 11100 Euclid Ave, Mail Stop MAC 5034, Cleveland, OH 44106, United States
| | - Noya Galai
- Department of Statistics, University of Haifa, Mt Carmel, Haifa, 31905, Israel
| | - Karen J Gibbins
- OB/GYN, University of Utah, Salt Lake City, UT, United States
| | - Robert M Silver
- OB/GYN, University of Utah, Salt Lake City, UT, United States
| | - Brian Wilcox
- Department of Obstetrics and Gynecology, University of Utah Health Sciences Center, Room 2B200 SOM, 50 North Medical Drive, Salt Lake City, UT 84132, United States
| | - Lindsey Sjaarda
- Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, 6710B Rockledge Dr. MSC 7004, Bethesda, MD 20892, United States
| | - Neil J Perkins
- Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, 6710B Rockledge Dr. MSC 7004, Bethesda, MD 20892, United States
| | - Enrique F Schisterman
- Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, 6710B Rockledge Dr. MSC 7004, Bethesda, MD 20892, United States
| | - Sunni L Mumford
- Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, 6710B Rockledge Dr. MSC 7004, Bethesda, MD 20892, United States
| |
Collapse
|
9
|
Spinelli M, Boucard C, Di Nicuolo F, Haesler V, Castellani R, Pontecorvi A, Scambia G, Granieri C, Barnea ER, Surbek D, Mueller M, Di Simone N. Synthetic PreImplantation Factor (sPIF) reduces inflammation and prevents preterm birth. PLoS One 2020; 15:e0232493. [PMID: 32511256 PMCID: PMC7279576 DOI: 10.1371/journal.pone.0232493] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 04/15/2020] [Indexed: 01/05/2023] Open
Abstract
Preterm birth (PTB) is the leading cause of neonatal morbidity and mortality and spontaneous PTB is a major contributor. The preceding inflammation/infection contributes not only to spontaneous PTB but is associated with neonatal morbidities including impaired brain development. Therefore, control of exaggerated immune response during pregnancy is an attractive strategy. A potential candidate is synthetic PreImplantation Factor (sPIF) as sPIF prevents inflammatory induced fetal loss and has neuroprotective properties. Here, we tested maternal sPIF prophylaxis in pregnant mice subjected to a lipopolysaccharides (LPS) insult, which results in PTB. Additionally, we evaluated sPIF effects in placental and microglial cell lines. Maternal sPIF application reduced the LPS induced PTB rate significantly. Consequently, sPIF reduced microglial activation (Iba-1 positive cells) and preserved neuronal migration (Cux-2 positive cells) in fetal brains. In fetal brain lysates sPIF decreased IL-6 and INFγ concentrations. In-vitro, sPIF reduced Iba1 and TNFα expression in microglial cells and reduced the expression of pro-apoptotic (Bad and Bax) and inflammatory (IL-6 and NLRP4) genes in placental cell lines. Together, maternal sPIF prophylaxis prevents PTB in part by controlling exaggerated immune response. Given the sPIF`FDA Fast Track approval in non-pregnant subjects, we envision sPIF therapy in pregnancy.
Collapse
Affiliation(s)
- Marialuigia Spinelli
- Department of Obstetrics and Gynecology and Department of Biomedical Research, University Hospital Bern, University of Bern, Bern, Switzerland
| | - Céline Boucard
- Department of Obstetrics and Gynecology and Department of Biomedical Research, University Hospital Bern, University of Bern, Bern, Switzerland
| | - Fiorella Di Nicuolo
- Università Cattolica del Sacro Cuore, Istituto di Clinica Ostetrica e Ginecologica, Roma, Italia
- International Scientific Institute Paolo VI, Università Cattolica Del Sacro Cuore, A. Gemelli Universitary Hospital, Rome, Italia
| | - Valerie Haesler
- Department of Obstetrics and Gynecology and Department of Biomedical Research, University Hospital Bern, University of Bern, Bern, Switzerland
| | - Roberta Castellani
- Università Cattolica del Sacro Cuore, Istituto di Clinica Ostetrica e Ginecologica, Roma, Italia
| | - Alfredo Pontecorvi
- Università Cattolica del Sacro Cuore, Istituto di Clinica Ostetrica e Ginecologica, Roma, Italia
- U.O.C di Endocrinologia e Diabetologia, Dipartimento di Scienze Gastroenterologiche, Endocrino-Metaboliche e Nefro-Urologiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italia
| | - Giovanni Scambia
- Università Cattolica del Sacro Cuore, Istituto di Clinica Ostetrica e Ginecologica, Roma, Italia
- U.O.C. di Ginecologia Oncologica, Dipartimento di Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italia
| | - Chiara Granieri
- Università Cattolica del Sacro Cuore, Istituto di Clinica Ostetrica e Ginecologica, Roma, Italia
| | - Eytan R. Barnea
- The Society for The Investigation of Early Pregnancy (SIEP), Cherry Hill, NJ, United States of America
- BioIncept LLC, Cherry Hill, NJ, United States of America
| | - Daniel Surbek
- Department of Obstetrics and Gynecology and Department of Biomedical Research, University Hospital Bern, University of Bern, Bern, Switzerland
| | - Martin Mueller
- Department of Obstetrics and Gynecology and Department of Biomedical Research, University Hospital Bern, University of Bern, Bern, Switzerland
- * E-mail: (MM); (NDS)
| | - Nicoletta Di Simone
- Università Cattolica del Sacro Cuore, Istituto di Clinica Ostetrica e Ginecologica, Roma, Italia
- Dipartimento di Scienze della Salute della Donna e del Bambino, Fondazione Policlinico Universitario A. Gemelli IRCCS, U.O.C. di Ostetricia e Patologia Ostetrica, Roma, Italia
- * E-mail: (MM); (NDS)
| |
Collapse
|
10
|
The Role of Decidual PD-1 + Treg Cells in Adverse Pregnancy Outcomes due to Toxoplasma gondii Infection. Inflammation 2020; 42:2119-2128. [PMID: 31468303 DOI: 10.1007/s10753-019-01075-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Toxoplasma gondii infection during pregnancy can result in adverse pregnancy outcomes. Previously, we have reported that these outcomes are associated with the impaired function of decidual Treg cells; however, the detailed mechanisms involved were unclear. It has been reported that the suppressive capacity of Treg cells is dependent on PD-1 expression. The present study explored the role of decidual PD-1+ Treg cell function in adverse pregnancy outcomes due to T. gondii infection. Toxoplasma gondii-infected pregnant mice were sacrificed on gestational day 14 and their pregnancy outcomes were observed. The expression of PD-1 on decidual Treg cells and expressions of Foxp3, CTLA-4, TGF-β, and IL-10 on decidual PD-1+ and PD-1- Treg cells were determined using flow cytometry. The results showed that the expression of PD-1 on decidual Treg cells was clearly higher in the T. gondii-infected mice than in the normal mice. Meanwhile, the expressions of Foxp3, CTLA-4, TGF-β, and IL-10 on decidual PD-1+ Treg cells were higher in the infected mice than in the normal mice. The expressions were higher in decidual PD1+ Treg cells than in PD-1- Treg cells in the infected mice. However, these expressions on PD-1- Treg cells did not significantly differ between the infected and normal mice. Nonetheless, the absolute percentages of decidual PD-1+ Treg cells decreased significantly in the infected mice compared with those in the normal mice. These results suggest that T. gondii infection mainly influences the function of decidual PD-1+ Treg cells, which would result in an insufficiently immunotolerant microenvironment and consequently in adverse pregnancy outcomes.
Collapse
|
11
|
Li G, Yang L, Li D, Zhang J, Du L, Xia L, Liu Y, Hu W. Effects of combined treatment with PD‑L1 Ig and CD40L mAb on immune tolerance in the CBA/J x DBA/2 mouse model. Mol Med Rep 2020; 21:1789-1798. [PMID: 32319625 PMCID: PMC7057827 DOI: 10.3892/mmr.2020.10977] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 01/01/2020] [Indexed: 12/31/2022] Open
Abstract
The embryo is a natural allograft and is the only exception to immune rejection, which reflects maternal immune tolerance towards the embryo. However, pregnancy loss is primarily caused by maternal immune rejection of the embryo. The aim of the present study was to explore the effects of combined treatment of programmed death-ligand 1 (PD-L1) immunoglobulin (Ig) and CD40-ligand (CD40L) monoclonal antibody (mAb) on immune tolerance in an abortion-prone mating model. Mice were divided into the normal, spontaneous abortion, PD-L1 Ig, CD40L mAb and the PD-L1 Ig + CD40L mAb groups. On day 14 of gestation, the embryo resorption abortion rates of all the groups was observed. The maternal hypo-responsiveness to paternal antigens was determined using a mixed lymphocyte response and the splenic CD4+CD25+ T-cell population, major histocompatibility complex (MHC)-II+, CD80+ and CD86+ cell populations in pregnant female CBA/J mice were analyzed using flow cytometry. The expression levels of intracellular cytokines in the splenic tissues of pregnant CBA/J female mice were analyzed using western blotting. The PD-L1 Ig + CD40L group displayed the lowest resorption rate compared with the other groups. A significant decrease in the proliferative response of maternal splenic immunocompetent cells against paternal antigens, and a significant increase in the proliferative response of maternal splenic CD4+CD25+ T cells was observed in the PD-L1 Ig + CD40L group compared with the spontaneous abortion group. The number of MHC-II+, CD80+ and CD86+ bone marrow-derived dendritic cells (DCs) generated by female mice, and the levels of tumor necrosis factor-α and interferon-γ in the spleens of female mice were significantly decreased in the PD-L1 Ig + CD40L mAb group compared with the spontaneous abortion group. By contrast, interleukin-4 levels were significantly increased in the PD-L1 Ig + CD40L mAb group compared with the spontaneous abortion group. The results suggested that the administration of PD-L1 Ig + CD40L mAb on day 4 of gestation, the period of peri-implantation, may induce paternal antigen-specific immunotolerance, leading to the embryo resorption rate of the abortion-prone model being similar to that of the normal pregnancy model. The results indicate that the combined treatment of PD-L1 Ig and anti-CD40L mAbs may serve as a potential therapeutic for pregnancy loss.
Collapse
Affiliation(s)
- Guanfei Li
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, P.R. China
| | - Lihua Yang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, P.R. China
| | - Dan Li
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, P.R. China
| | - Jinhong Zhang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, P.R. China
| | - Ling Du
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, P.R. China
| | - Libin Xia
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, P.R. China
| | - Yunhua Liu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, P.R. China
| | - Wanqin Hu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, P.R. China
| |
Collapse
|
12
|
Xiang YJ, Hou YY, Yan HL, Liu H, Ge YX, Chen N, Xiang JF, Hao CF. Mesenchymal stem cells-derived exosomes improve pregnancy outcome through inducing maternal tolerance to the allogeneic fetus in abortion-prone mating mouse. Kaohsiung J Med Sci 2020; 36:363-370. [PMID: 31943723 DOI: 10.1002/kjm2.12178] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 12/10/2019] [Indexed: 12/20/2022] Open
Abstract
Recurrent pregnancy loss (RPL) is three or more times of consecutive spontaneous loss of pregnancy. The underlying cause is complicated and the etiology of over 50% of RPL patients is unclear. In the present study, bone marrow mesenchymal stem cells were isolated from CBA/J female mice and exosomes were isolated from cell culture medium by ultracentrifugation. CBA/J female mice were paired with male DBA/2 to generate abortion prone mouse model, and CBA/J females paired with male BALB/c mice were used as control. Exosomes were injected through uterine horns into pregnant CBA/J mice on day 4.5 of gestation in abortion-prone matting. On day 13.5 of pregnancy, abortion rates were calculated and the level of transforming growth factor-β (TGF-β), interleukin 10 (IL-10), interferon g (IFN-γ), and tumor necrosis factor a (TNF-α) in CD4+ T cells and macrophages in deciduas were evaluated by flow cytometry. Exosomes injection improved the pregnancy outcomes in abortion prone mice. The IL-4 and IL-10 levels on CD4+ T cells were upregulated in the maternal-fetal interface; meanwhile, the TNF-α and IFN-γ levels on CD4+ T cells were reduced. The IL-10 level was increased and IL-12 was reduced on the monocytes that separated from deciduas. miR-101 level was increased in the CD4+ T cells in the deciduas. In conclusion, the treatment of ESCs-derived exosomes modulates T cells' function and macrophages activities in the maternal-fetal interface that resulted in a decreased embryo resorption rate, and provides a therapeutic potential to treat RPL.
Collapse
Affiliation(s)
- Yan-Jie Xiang
- School of Medicine, Shandong University, Jinan, Shandong Province, People's Republic of China.,Reproductive Medicine Center, The Affiliated Changhai Hospital of China Naval Military Medical University, Shanghai, People's Republic of China
| | - Yan-Yan Hou
- Maternity Department of International Maternal and Child Peace Hospital, The Affiliated Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Hong-Li Yan
- Reproductive Medicine Center, The Affiliated Changhai Hospital of China Naval Military Medical University, Shanghai, People's Republic of China
| | - Hui Liu
- Department of Reproductive Medicine, People's Hospital of Rizhao, Rizhao, Shandong, People's Republic of China
| | - Yan-Xin Ge
- Department of Reproductive Medicine, People's Hospital of Rizhao, Rizhao, Shandong, People's Republic of China
| | - Na Chen
- Department of Obstetrics, Traditional Chinese Medicine Hospital of The West Coast of Qingdao New District, Qingdao, Shandong, People's Republic of China
| | - Jian-Feng Xiang
- Department of Intervention, Shanghai Fengxian District Central Hospital, Shanghai, People's Republic of China
| | - Cui-Fang Hao
- Center for Reproductive Medicine, Qingdao Women and Children's Hospital (QWCH) affiliated to Qingdao University, Qingdao, Shandong, People's Republic of China
| |
Collapse
|
13
|
Gallino L, Hauk V, Fernández L, Soczewski E, Gori S, Grasso E, Calo G, Saraco N, Berensztein E, Waschek JA, Pérez Leirós C, Ramhorst R. VIP Promotes Recruitment of Tregs to the Uterine-Placental Interface During the Peri-Implantation Period to Sustain a Tolerogenic Microenvironment. Front Immunol 2020; 10:2907. [PMID: 31969877 PMCID: PMC6960177 DOI: 10.3389/fimmu.2019.02907] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Accepted: 11/26/2019] [Indexed: 01/02/2023] Open
Abstract
Uterine receptivity and embryo implantation are two main processes that need a finely regulated balance between pro-inflammatory and tolerogenic mediators to allow a successful pregnancy. The neuroimmune peptide vasoactive intestinal peptide (VIP) is a key regulator, and it is involved in the induction of regulatory T cells (Tregs), which are crucial in both processes. Here, we analyzed the ability of endogenous and exogenous VIP to sustain a tolerogenic microenvironment during the peri-implantation period, particularly focusing on Treg recruitment. Wild-type (WT) and VIP-deficient mice [heterozygous (HT, +/-), knockout (KO, -/-)], and FOXP3-knock-in-GFP mice either pregnant or in estrus were used. During the day of estrus, we found significant histological differences between the uterus of WT mice vs. VIP-deficient mice, with the latter exhibiting undetectable levels of FOXP3 expression, decreased expression of interleukin (IL)-10, and vascular endothelial growth factor (VEGF)c, and increased gene expression of the Th17 proinflammatory transcription factor RORγt. To study the implantation window, we mated WT and VIP (+/-) females with WT males and observed altered FOXP3, VEGFc, IL-10, and transforming growth factor (TGF)β gene expression at the implantation sites at day 5.5 (d5.5), demonstrating a more inflammatory environment in VIP (+/-) vs. VIP (+/+) females. A similar molecular profile was observed at implantation sites of WT × WT mice treated with VIP antagonist at d3.5. We then examined the ability GFP-sorted CD4+ cells from FOXP3-GFP females to migrate toward conditioned media (CM) obtained from d5.5 implantation sites cultured in the absence/presence of VIP or VIP antagonist. VIP treatment increased CD4+FOXP3+ and decreased CD4+ total cell migration towards implantation sites, and VIP antagonist prevented these effects. Finally, we performed adoptive cell transfer of Tregs (sorted from FOXP3-GFP females) in VIP-deficient-mice, and we observed that FOXP3-GFP cells were mainly recruited into the uterus/implantation sites compared to all other tested tissues. In addition, after Treg transfer, we found an increase in IL-10 expression and VEGFc in HT females and allowed embryo implantation in KO females. In conclusion, VIP contributes to a local tolerogenic response necessary for successful pregnancy, preventing the development of a hostile uterine microenvironment for implantation by the selective recruitment of Tregs during the peri-implantation period.
Collapse
Affiliation(s)
- Lucila Gallino
- CONICET, Laboratorio de Inmunofarmacología, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Vanesa Hauk
- CONICET, Laboratorio de Inmunofarmacología, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Laura Fernández
- CONICET, Laboratorio de Inmunofarmacología, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Elizabeth Soczewski
- CONICET, Laboratorio de Inmunofarmacología, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Soledad Gori
- CONICET, Laboratorio de Inmunofarmacología, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Esteban Grasso
- CONICET, Laboratorio de Inmunofarmacología, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Guillermina Calo
- CONICET, Laboratorio de Inmunofarmacología, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Nora Saraco
- Servicio de Endocrinología, Hospital Pediátrico Dr. Juan P. Garrahan, CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Esperanza Berensztein
- Servicio de Endocrinología, Hospital Pediátrico Dr. Juan P. Garrahan, CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - James A Waschek
- Department of Psychiatry and Biobehavioral Sciences, The David Geffen School of Medicine, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, United States
| | - Claudia Pérez Leirós
- CONICET, Laboratorio de Inmunofarmacología, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Rosanna Ramhorst
- CONICET, Laboratorio de Inmunofarmacología, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
14
|
Daimon A, Morihara H, Tomoda K, Morita N, Koishi Y, Kanki K, Ohmichi M, Asahi M. Intravenously Injected Pluripotent Stem Cell-derived Cells Form Fetomaternal Vasculature and Prevent Miscarriage in Mouse. Cell Transplant 2020; 29:963689720970456. [PMID: 33349053 PMCID: PMC7873769 DOI: 10.1177/0963689720970456] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 09/26/2020] [Accepted: 10/13/2020] [Indexed: 01/22/2023] Open
Abstract
Miscarriage is the most common complication of pregnancy, and about 1% of pregnant women suffer a recurrence. Using a widely used mouse miscarriage model, we previously showed that intravenous injection of bone marrow (BM)-derived endothelial progenitor cells (EPCs) may prevent miscarriage. However, preparing enough BM-derived EPCs to treat a patient might be problematic. Here, we demonstrated the generation of mouse pluripotent stem cells (PSCs), propagation of sufficient PSC-derived cells with endothelial potential (PSC-EPs), and intravenous injection of the PSC-EPs into the mouse miscarriage model. We found that the injection prevented miscarriage. Three-dimensional reconstruction images of the decidua after tissue cleaning revealed robust fetomaternal neovascularization induced by the PSC-EP injection. Additionally, the injected PSC-EPs directly formed spiral arteries. These findings suggest that intravenous injection of PSC-EPs could become a promising remedy for recurrent miscarriage.
Collapse
Affiliation(s)
- Atsushi Daimon
- Department of Obstetrics and Gynecology, Osaka Medical College,
Takatsuki, Japan
- Department of Pharmacology, Osaka Medical College, Takatsuki,
Japan
- These authors contributed equally to
this article
| | - Hirofumi Morihara
- Department of Pharmacology, Osaka Medical College, Takatsuki,
Japan
- These authors contributed equally to
this article
| | - Kiichiro Tomoda
- Department of Pharmacology, Osaka Medical College, Takatsuki,
Japan
- Department of Life Science Frontiers, Center for iPS Cell Research
and Application, Kyoto University, Japan
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA,
USA
- These authors contributed equally to
this article
| | - Natsuko Morita
- Department of Obstetrics and Gynecology, Osaka Medical College,
Takatsuki, Japan
- Department of Pharmacology, Osaka Medical College, Takatsuki,
Japan
| | - Yoshinori Koishi
- Division of Research Animal Laboratory and Translational Medicine,
Research and Development Center, Takatsuki, Osaka, Japan
| | - Kazuyoshi Kanki
- Department of Obstetrics and Gynecology, Osaka Medical College,
Takatsuki, Japan
| | - Masahide Ohmichi
- Department of Obstetrics and Gynecology, Osaka Medical College,
Takatsuki, Japan
| | - Michio Asahi
- Department of Pharmacology, Osaka Medical College, Takatsuki,
Japan
| |
Collapse
|
15
|
Baek H, Yang H, Lee JH, Kang NH, Lee J, Bae H, Hwang DS. Prophylactic Effects of Bee Venom Phospholipase A2 in Lipopolysaccharide-Induced Pregnancy Loss. Toxins (Basel) 2019; 11:toxins11070404. [PMID: 31336883 PMCID: PMC6669565 DOI: 10.3390/toxins11070404] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 07/11/2019] [Accepted: 07/11/2019] [Indexed: 11/23/2022] Open
Abstract
Spontaneous abortion represents a common form of embryonic loss caused by early pregnancy failure. In the present study, we investigated the prophylactic effects of bee venom phospholipase A2 (bvPLA2), a regulatory T cell (Treg) inducer, on a lipopolysaccharide (LPS)-induced abortion mouse model. Fetal loss, including viable implants, the fetal resorption rate, and the fetal weight, were measured after LPS and bvPLA2 treatment. The levels of serum and tissue inflammatory cytokines were determined. To investigate the involvement of the Treg population in bvPLA2-mediated protection against fetal loss, the effect of Treg depletion was evaluated following bvPLA2 and LPS treatment. The results clearly revealed that bvPLA2 can prevent fetal loss accompanied by growth restriction in the remaining viable fetus. When the LPS-induced abortion mice were treated with bvPLA2, Treg cells were significantly increased compared with those in the non-pregnant, PBS, and LPS groups. After LPS injection, the levels of proinflammatory cytokines were markedly increased compared with those in the PBS mouse group, while bvPLA2 treatment showed significantly decreased TNF-α and IFN-γ expression compared with that in the LPS group. The protective effects of bvPLA2 treatment were not detected in Treg-depleted abortion-prone mice. These findings suggest that bvPLA2 has protective effects in the LPS-induced abortion mouse model by regulating Treg populations.
Collapse
Affiliation(s)
- Hyunjung Baek
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul 02453, Korea
| | - HyeJin Yang
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul 02453, Korea
| | - Jong Hoon Lee
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul 02453, Korea
| | - Na-Hoon Kang
- Department of Clinical Korean Medicine, Graduate School, Kyung Hee University, Seoul 02453, Korea
| | - Jinwook Lee
- Department of Clinical Korean Medicine, Graduate School, Kyung Hee University, Seoul 02453, Korea
| | - Hyunsu Bae
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul 02453, Korea.
| | - Deok-Sang Hwang
- Department of Clinical Korean Medicine, Graduate School, Kyung Hee University, Seoul 02453, Korea.
| |
Collapse
|
16
|
Thiele K, Ahrendt LS, Hecher K, Arck PC. The mnemonic code of pregnancy: Comparative analyses of pregnancy success and complication risk in first and second human pregnancies. J Reprod Immunol 2019; 134-135:11-20. [PMID: 31374263 DOI: 10.1016/j.jri.2019.06.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 06/21/2019] [Accepted: 06/23/2019] [Indexed: 11/16/2022]
Abstract
Obstetrical complications such as spontaneous abortion/miscarriage, fetal growth restriction, preeclampsia or preterm birth occur in approx. 15% of human pregnancies. Clinical experts often state that a previous uncomplicated pregnancy reduces the risk for complications in subsequent pregnancies. Vice versa, a prior pregnancy affected by obstetrical complications increases the risk for reoccurrence. However, published evidence directly underpinning these clinical statements is sparse. Considering that the maternal immune adaptation may be causally involved in determining the outcome of subsequent pregnancies, a comprehensive analysis of clinical data was long overdue. We here present a systematic analysis of clinical data using a PubMed-based approach to identify human studies with relevant information on birth weight and incidences of pregnancy complications in first and second pregnancies. From initially 18,592 publications, 37 studies were included in the quantitative data analysis. Women with a previous pregnancy affected by complications where a derailed immune response can be inferred have a 2.2-3.2-fold increased risk to be affected again in a subsequent pregnancy. Conversely, a normally progressing primary pregnancy reduced the risk for complications in a subsequent pregnancy by 35-65%. Moreover, an uncomplicated primary pregnancy was associated with a 4.2% increased birth weight in a following pregnancy without a difference in gestational age at delivery. In conclusion, the increased birth weight after previously uncomplicated pregnancies suggests that an immune memory is mounted during primary pregnancies. This immune memory may promote the successful outcome of subsequent pregnancies or - if missing or compromised - account for a risk perpetuation of pregnancy complications.
Collapse
Affiliation(s)
- Kristin Thiele
- Division of Experimental Feto-Maternal Medicine, Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - Lisa Sophie Ahrendt
- Division of Experimental Feto-Maternal Medicine, Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kurt Hecher
- Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Petra Clara Arck
- Division of Experimental Feto-Maternal Medicine, Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
17
|
Clark DA. Oral tolerance and the materno-fetal relationship. J Reprod Immunol 2019; 134-135:36-39. [PMID: 31300171 DOI: 10.1016/j.jri.2019.07.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 07/03/2019] [Indexed: 01/14/2023]
Abstract
Oral administration of antigen is a potent route for induction of systemic tolerance. Regulatory T cells and TH3 cellsare generated at intestinal mucosal sites. Oral exposure to antigens in seminal plasma has been suggested to be ableto generate 'tolerance' at the materno-fetal interface that may reduce the risk of pre-eclampsia and recurrent miscarriage. Issues relating to assessing the applicability of oral exposure to seminal plasma antigens in recurrentmiscarriage are discussed. Such tolerance could represent an unappreciated confounder in immunotherapy trials.
Collapse
|
18
|
Soluble CD200 in secretory phase endometriosis endometrial venules may explain endometriosis pathophysiology and provide a novel treatment target. J Reprod Immunol 2018; 129:59-67. [DOI: 10.1016/j.jri.2018.05.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 05/11/2018] [Accepted: 05/12/2018] [Indexed: 11/19/2022]
|
19
|
Zahran AM, Zharan KM, Hetta HF. Significant correlation between regulatory T cells and vitamin D status in term and preterm labor. J Reprod Immunol 2018; 129:15-22. [PMID: 30029057 DOI: 10.1016/j.jri.2018.07.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 07/06/2018] [Accepted: 07/11/2018] [Indexed: 12/18/2022]
Abstract
BACKGROUND AND AIM Vitamin D insufficiency and deficiency have been associated with an increased risk of adverse pregnancy outcomes. Also, vitamin D is known to play a role in promoting the function of regulatory T-cells (Tregs). Tregs play an important role in suppressing the immune response during pregnancy. Our study aimed to investigate Tregs phenotypes in preterm and tem laboring women and its association with vitamin D level. METHODS This cross-sectional study included 82 pregnant women, divided into 46 term and 36 preterm laboring women in addition to 30 healthy non-pregnant women. The percentage of CD4+CD25+Foxp3+Treg cells and their composition of four different Treg subsets were evaluated using flow cytometric analysis. Also, serum vitamin D levels were measured by ELISA. RESULTS The percentage of the CD4+ CD25+ FoxP3+ Tregs were significantly decreased in term and preterm laboring women compared to the non-pregnant controls. The percentage of CD45RA+ Tregs, was significantly increased in term laboring women than preterm laboring women and non-pregnant women. Also, term labor women had increased proportion of HLA-DRhighTregs. Preterm labor women had significant increased proportion of HLA-DRnegative Tregs compared to term labor women. The overall prevalence of vitamin D deficiency and vitamin D insufficiency was higher in preterm than term laboring women and non-pregnant women. Significant positive correlations were found between serum level of 25 (OH)D and percentage of CD4+ CD25+ Foxp3+Tregs and percentage HLA-DRhighTregs among term and preterm laboring women with vitamin D deficiency. CONCLUSION There is a strong association between the percentage of Treg phenotypes and vitamin D level in term and preterm labor women with vitamin D deficiency. Also, the onset of term and preterm labor is associated with changes in the composition of the total Treg pool with different Treg subsets which in turn may be responsible for immunologic mechanisms that associated with labor induction.
Collapse
Affiliation(s)
- Asmaa M Zahran
- Department of Clinical Pathology, South Egypt Cancer institute, Assiut, Egypt
| | - Kamal M Zharan
- Department of Obstetrics and Gynecology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Helal F Hetta
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Assiut University, Assiut, Egypt.
| |
Collapse
|
20
|
Paiola M, Knigge T, Duflot A, Pinto PIS, Farcy E, Monsinjon T. Oestrogen, an evolutionary conserved regulator of T cell differentiation and immune tolerance in jawed vertebrates? DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2018; 84:48-61. [PMID: 29408048 DOI: 10.1016/j.dci.2018.01.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 01/18/2018] [Accepted: 01/18/2018] [Indexed: 06/07/2023]
Abstract
In teleosts, as in mammals, the immune system is tightly regulated by sexual steroid hormones, such as oestrogens. We investigated the effects of 17β-oestradiol on the expression of several genes related to T cell development and resulting T cell subpopulations in sea bass, Dicentrarchus labrax, for a primary lymphoid organ, the thymus, and two secondary lymphoid organs, the head-kidney and the spleen. In parallel, the oxidative burst capacity was assessed in leucocytes of the secondary lymphoid organs. Apoptosis- and proliferation-related genes, indicative of B and T cell clonal selection and lymphoid progenitor activity, were not affected by elevated oestrogen-levels. Sex-related oestrogen-responsiveness in T cell and antigen-presenting cell markers was observed, the expression of which was differentially induced by oestrogen-exposure in the three lymphoid organs. Remarkably, in the spleen, oestrogen increased regulatory T cell-related gene expression was associated with a decrease in oxidative burst capacity. To the best of our knowledge, this study indicates for the first time that physiological levels of oestrogen are likely to promote immune tolerance by modulating thymic function (i.e., T cell development and output) and peripheral T cells in teleosts, similar to previously reported oestrogenic effects in mammals.
Collapse
Affiliation(s)
- Matthieu Paiola
- Normandy University, FR CNRS 3730 SCALE, UMR-I 02 INERIS-URCA-ULH Environmental Stress and Aquatic Biomonitoring (SEBIO), University of Le Havre Normandy, 76600 Le Havre, France
| | - Thomas Knigge
- Normandy University, FR CNRS 3730 SCALE, UMR-I 02 INERIS-URCA-ULH Environmental Stress and Aquatic Biomonitoring (SEBIO), University of Le Havre Normandy, 76600 Le Havre, France
| | - Aurélie Duflot
- Normandy University, FR CNRS 3730 SCALE, UMR-I 02 INERIS-URCA-ULH Environmental Stress and Aquatic Biomonitoring (SEBIO), University of Le Havre Normandy, 76600 Le Havre, France
| | - Patricia I S Pinto
- Laboratory of Comparative Endocrinology and Integrative Biology, CCMAR - Centre of Marine Sciences, University of Algarve, 8005-139 Faro, Portugal
| | - Emilie Farcy
- Montpellier University, UMR MARBEC (UM, CNRS, Ifremer, IRD), 34095 Montpellier, France
| | - Tiphaine Monsinjon
- Normandy University, FR CNRS 3730 SCALE, UMR-I 02 INERIS-URCA-ULH Environmental Stress and Aquatic Biomonitoring (SEBIO), University of Le Havre Normandy, 76600 Le Havre, France.
| |
Collapse
|
21
|
Trophoblast CD200 expression in successful human pregancies and missed abortions. J Reprod Immunol 2018; 127:55-57. [DOI: 10.1016/j.jri.2018.03.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 03/02/2018] [Accepted: 03/16/2018] [Indexed: 01/09/2023]
|
22
|
Schumacher A, Ehrentraut S, Scharm M, Wang H, Hartig R, Morse HC, Zenclussen AC. Plasma Cell Alloantigen 1 and IL-10 Secretion Define Two Distinct Peritoneal B1a B Cell Subsets With Opposite Functions, PC1 high Cells Being Protective and PC1 low Cells Harmful for the Growing Fetus. Front Immunol 2018; 9:1045. [PMID: 29868008 PMCID: PMC5962664 DOI: 10.3389/fimmu.2018.01045] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 04/26/2018] [Indexed: 12/18/2022] Open
Abstract
B cells possess various immuno regulatory functions. However, research about their participation in tolerance induction toward the fetus is just emerging. Accumulating evidence supports the idea that B cells can play seemingly conflicting roles during pregnancy, either protecting or harming the fetus. Previous findings indicated the presence of two different peritoneal B cell subsets, defined by the expression of the plasma cell alloantigen 1 (PC1) and with distinct immune modulatory functions. Here, we aimed to study the participation of these two B cell subsets, on pregnancy outcome in a murine model of disturbed fetal tolerance. The frequencies and cell numbers of peritoneal and splenic CD19+IL-10+ and CD19+CD5+IL-10+PC1+ cells were assessed in virgin as well as normal pregnant (NP) and abortion-prone (AP) females during the course of gestation. Peritoneal PC1low or PC1high B1a B cells were sorted, analyzed for their ability to secrete IL-10 and adoptively transferred into NP or AP females. On gestation day (gd) 12, the abortion rate as well as the frequencies and cell numbers of regulatory T cells, TH1 and TH17 cells were determined in spleens and decidua. In addition, mRNA expression of IL-10, TGF-β, IFN-γ, and TNF-α was analyzed in decidual tissue. Peritoneal CD19+IL-10+ and CD19+CD5+IL-10+PC1+ frequencies fluctuated during the progression of normal pregnancies while no significant changes were observed in spleen. AP females showed significantly reduced frequencies of both B cell populations and exhibited an altered peritoneal PC1high/PC1low ratio at gd10. Adoptive transfers of PC1low B1a B cells into NP females increased the abortion rate in association with a reduced splenic regulatory T/TH17 ratio. By contrast, the transfer of PC1high B1a B cells into AP females significantly diminished the fetal rejection rate and significantly reduced the numbers of splenic TH17 cells. Our results suggest that the peritoneum harbors two distinct B1a B cell subsets that can be distinguished by their PC1 expression. Whereas PC1high B1a B cells seem to support fetal survival, PC1low cells B1a B cells may compromise fetal well-being.
Collapse
Affiliation(s)
- Anne Schumacher
- Department of Experimental Obstetrics and Gynecology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Stefanie Ehrentraut
- Department of Experimental Obstetrics and Gynecology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Markus Scharm
- Department of Experimental Obstetrics and Gynecology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Hongsheng Wang
- Virology and Cellular Immunology Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, NIH, Rockville, MD, United States
| | - Roland Hartig
- Core Facility Multidimensional Microscopy and Cellular Diagnostics, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Herbert C Morse
- Virology and Cellular Immunology Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, NIH, Rockville, MD, United States
| | - Ana Claudia Zenclussen
- Department of Experimental Obstetrics and Gynecology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| |
Collapse
|
23
|
Ozen M, Novak C, Burd I. Placenta immune infiltrates and perinatal outcomes. Am J Reprod Immunol 2018; 79:e12850. [PMID: 29577494 DOI: 10.1111/aji.12850] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 02/22/2018] [Indexed: 12/19/2022] Open
Abstract
Pregnancy is a state of immunotolerance and loss of this immunotolerance may lead to fetal rejection, pregnancy complications, and neonatal complications. Immunobiology of pregnancy is complex and involves unique immune cell populations specific to pregnancy, changes in mucosal immune cells and peripheral immune system, and reciprocal adaptations between the mother and the fetus. The mechanisms required for sustaining a healthy feto-placental barrier and a healthy pregnancy such as activation of regulatory immune responses with a predominance of regulatory T cells lead to immune evasion and propagation of cancer. It is intriguing to note that the immune pathways which are effective in limiting or eliminating cancer form the very basis for loss of feto-maternal tolerance. In this article, we aim to compare and contrast immunobiology of healthy and pathological pregnancies mirroring with cancer immunobiology with a focus on immune checkpoint receptors.
Collapse
Affiliation(s)
- Maide Ozen
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Integrated Research Center for Fetal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Christopher Novak
- Integrated Research Center for Fetal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Division of Maternal Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Irina Burd
- Integrated Research Center for Fetal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Division of Maternal Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
24
|
Kenny LC, Kell DB. Immunological Tolerance, Pregnancy, and Preeclampsia: The Roles of Semen Microbes and the Father. Front Med (Lausanne) 2018; 4:239. [PMID: 29354635 PMCID: PMC5758600 DOI: 10.3389/fmed.2017.00239] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 12/12/2017] [Indexed: 12/18/2022] Open
Abstract
Although it is widely considered, in many cases, to involve two separable stages (poor placentation followed by oxidative stress/inflammation), the precise originating causes of preeclampsia (PE) remain elusive. We have previously brought together some of the considerable evidence that a (dormant) microbial component is commonly a significant part of its etiology. However, apart from recognizing, consistent with this view, that the many inflammatory markers of PE are also increased in infection, we had little to say about immunity, whether innate or adaptive. In addition, we focused on the gut, oral and female urinary tract microbiomes as the main sources of the infection. We here marshall further evidence for an infectious component in PE, focusing on the immunological tolerance characteristic of pregnancy, and the well-established fact that increased exposure to the father's semen assists this immunological tolerance. As well as these benefits, however, semen is not sterile, microbial tolerance mechanisms may exist, and we also review the evidence that semen may be responsible for inoculating the developing conceptus (and maybe the placenta) with microbes, not all of which are benign. It is suggested that when they are not, this may be a significant cause of PE. A variety of epidemiological and other evidence is entirely consistent with this, not least correlations between semen infection, infertility and PE. Our view also leads to a series of other, testable predictions. Overall, we argue for a significant paternal role in the development of PE through microbial infection of the mother via insemination.
Collapse
Affiliation(s)
- Louise C. Kenny
- The Irish Centre for Fetal and Neonatal Translational Research (INFANT), University College Cork, Cork, Ireland
- Department of Obstetrics and Gynecology, University College Cork, Cork, Ireland
- Faculty of Health and Life Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Douglas B. Kell
- School of Chemistry, The University of Manchester, Manchester, United Kingdom
- The Manchester Institute of Biotechnology, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
25
|
Ali SB, Jeelall Y, Pennell CE, Hart R, McLean-Tooke A, Lucas M. The role of immunological testing and intervention in reproductive medicine: A fertile collaboration? Am J Reprod Immunol 2017; 79. [DOI: 10.1111/aji.12784] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 10/25/2017] [Indexed: 12/13/2022] Open
Affiliation(s)
- Syed B. Ali
- Department of Clinical Immunology; Sir Charles Gairdner Hospital; Perth WA Australia
| | - Yogesh Jeelall
- School of Medicine and Pharmacology; University of Western Australia; Perth WA Australia
- Harry Perkins Institute of Medical Research; Perth WA Australia
| | - Craig E. Pennell
- School of Women's and Infants’ Health; The University of Western Australia; Perth WA Australia
- The Women and Infants Research Foundation; Perth WA Australia
| | - Roger Hart
- School of Women's and Infants’ Health; The University of Western Australia; Perth WA Australia
- Fertility Specialists of Western; Bethesda Hospital; Claremont WA Australia
| | - Andrew McLean-Tooke
- Department of Clinical Immunology; Sir Charles Gairdner Hospital; Perth WA Australia
- Pathwest Laboratory Medicine; Queen Elizabeth II Medical Centre; Perth WA Australia
| | - Michaela Lucas
- Department of Clinical Immunology; Sir Charles Gairdner Hospital; Perth WA Australia
- Pathwest Laboratory Medicine; Queen Elizabeth II Medical Centre; Perth WA Australia
- School of Medicine and Pharmacology; School of Pathology and Laboratory Medicine; University of Western Australia; Perth WA Australia
- Institute for Immunology and Infectious Diseases; Murdoch University; Perth WA Australia
| |
Collapse
|
26
|
Nakajima A, Kaga N, Nakanishi Y, Ohno H, Miyamoto J, Kimura I, Hori S, Sasaki T, Hiramatsu K, Okumura K, Miyake S, Habu S, Watanabe S. Maternal High Fiber Diet during Pregnancy and Lactation Influences Regulatory T Cell Differentiation in Offspring in Mice. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2017; 199:3516-3524. [PMID: 29021375 DOI: 10.4049/jimmunol.1700248] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 09/18/2017] [Indexed: 12/16/2023]
Abstract
Short-chain fatty acids (SCFAs), the end products of dietary fiber, influence the immune system. Moreover, during pregnancy the maternal microbiome has a great impact on the development of the offspring's immune system. However, the exact mechanisms by which maternal SCFAs during pregnancy and lactation influence the immune system of offspring are not fully understood. We investigated the molecular mechanisms underlying regulatory T cell (Treg) differentiation in offspring regulated by a maternal high fiber diet (HFD). Plasma levels of SCFAs in offspring from HFD-fed mice were higher than in those from no fiber diet-fed mice. Consequently, the offspring from HFD-fed mice had higher frequencies of thymic Treg (tTreg) and peripheral Tregs We found that the offspring of HFD-fed mice exhibited higher autoimmune regulator (Aire) expression, a transcription factor expressed in the thymic microenvironment, suggesting SCFAs promote tTreg differentiation through increased Aire expression. Notably, the receptor for butyrate, G protein-coupled receptor 41 (GPR41), is highly expressed in the thymic microenvironment and Aire expression is not increased by stimulation with butyrate in GPR41-deficient mice. Our studies highlight the significance of SCFAs produced by a maternal HFD for Treg differentiation in the thymus of offspring. Given that Aire expression is associated with the induction of tTregs, the maternal microbiome influences Treg differentiation in the thymus of offspring through GPR41-mediated Aire expression.
Collapse
Affiliation(s)
- Akihito Nakajima
- Department of Gastroenterology, School of Medicine, Juntendo University, Tokyo 113-8421, Japan;
| | - Naoko Kaga
- Laboratory of Proteomics and Biomolecular Science, Biomedical Research Center, School of Medicine, Juntendo University, Tokyo 113-8421, Japan
| | - Yumiko Nakanishi
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Science, Yokohama 230-0045, Japan
| | - Hiroshi Ohno
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Science, Yokohama 230-0045, Japan
| | - Junki Miyamoto
- Department of Applied Biological Science, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Fuchu 183-8509, Japan
| | - Ikuo Kimura
- Department of Applied Biological Science, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Fuchu 183-8509, Japan
| | - Shohei Hori
- Laboratory for Immune Homeostasis, RIKEN Center for Integrative Medical Science, Yokohama 230-0045, Japan
- Laboratory of Immunology and Microbiology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-8654, Japan
| | - Takashi Sasaki
- Department of Bacteriology, School of Medicine, Juntendo University, Tokyo 113-8421, Japan
| | - Keiichi Hiramatsu
- Department of Bacteriology, School of Medicine, Juntendo University, Tokyo 113-8421, Japan
| | - Ko Okumura
- Atopic Research Center, School of Medicine, Juntendo University, Tokyo 113-8421, Japan; and
| | - Sachiko Miyake
- Department of Immunology, School of Medicine, Juntendo University, Tokyo 113-8421, Japan
| | - Sonoko Habu
- Atopic Research Center, School of Medicine, Juntendo University, Tokyo 113-8421, Japan; and
| | - Sumio Watanabe
- Department of Gastroenterology, School of Medicine, Juntendo University, Tokyo 113-8421, Japan
| |
Collapse
|
27
|
Li F, Dang J, Jiang M, He M, Yang M, Li J, Hao H, Zhou Y, Zuo W, Xie Y, Deng D. Upregulation of Tim-3 expression at feto-maternal interface may explain embryo survival in the CBAxDBA/2 model of abortion. Am J Reprod Immunol 2017; 79. [PMID: 29083087 DOI: 10.1111/aji.12775] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 10/02/2017] [Indexed: 01/08/2023] Open
Abstract
PROBLEM To understand the mechanisms of action of Tim-3 at the maternal-fetal interface and explore how Tim-3 might be involved in the pathogenesis of abortion by constructing an in vitro trophoblast-lymphocyte system. METHODS OF STUDY Female CBA/J × male DBA/2 matings were used as the abortion-prone model and CBA/J × male BALB/c matings as control. The expression of Tim-3 at the maternal-fetal interface and in the peripheral blood lymphocytes was measured by immunohistochemistry and Western blotting. The proliferation index of lymphocytes and levels of Th1/Th2-derived cytokines in peripheral blood and in the co-culture system were determined using CCK-8 assay and ELISA, respectively. RESULTS The expression level of Tim-3 was higher in abortion-prone matings than that of control (P < .05). A preponderance of Th1 was observed in the co-culture system in the abortion-prone mating group. Recombinant Tim-3 Ig reversed the imbalance of Th1/Th2 immunity of abortion-prone matings by suppressing the secretion of IFN-γ and IL-2 but had no direct effect on the generation of IL-4. CONCLUSION Tim-3 might contribute to successful pregnancy by restraining Th1 bias, and the maternal immune system might develop a strategy including upregulation of Tim-3 at the maternal-fetal interface and in peripheral blood so as to maintain moderate inflammatory responses against miscarriage.
Collapse
Affiliation(s)
- Fanfan Li
- Department of Gynecology and Obstetrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jing Dang
- Department of Gynecology and Obstetrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Min Jiang
- Department of Gynecology and Obstetrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.,Department of Gynecology and Obstetrics, Northern Jiangsu Province Hospital, Clinical Medical College, Yangzhou University, Jiangsu, China
| | - Mengzhou He
- Department of Gynecology and Obstetrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Meitao Yang
- Department of Gynecology and Obstetrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jing Li
- Department of Gynecology and Obstetrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.,Faculty of Reproductive Medical Center of the Third Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Haiyan Hao
- Department of Gynecology and Obstetrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yuan Zhou
- Department of Gynecology and Obstetrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.,Department of Reproductive Medical Center, Tangdu Hospital, The Fourth Military Medical University, xi'an, China
| | - Wei Zuo
- Department of Gynecology and Obstetrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.,Faculty of Department of Orthopedics, Pu Ai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yin Xie
- Department of Gynecology and Obstetrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Dongrui Deng
- Department of Gynecology and Obstetrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
28
|
Immunometabolism, pregnancy, and nutrition. Semin Immunopathol 2017; 40:157-174. [PMID: 29071391 DOI: 10.1007/s00281-017-0660-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 10/18/2017] [Indexed: 12/16/2022]
Abstract
The emerging field of immunometabolism has substantially progressed over the last years and provided pivotal insights into distinct metabolic regulators and reprogramming pathways of immune cell populations in various immunological settings. However, insights into immunometabolic reprogramming in the context of reproduction are still enigmatic. During pregnancy, the maternal immune system needs to actively adapt to the presence of the fetal antigens, i.e., by functional modifications of distinct innate immune cell subsets, the generation of regulatory T cells, and the suppression of an anti-fetal effector T cell response. Considering that metabolic pathways have been shown to affect the functional role of such immune cells in a number of settings, we here review the potential role of immunometabolism with regard to the molecular and cellular mechanisms necessary for successful reproduction. Since immunometabolism holds the potential for a therapeutic approach to alter the course of immune diseases, we further highlight how a targeted metabolic reprogramming of immune cells may be triggered by maternal anthropometric or nutritional aspects.
Collapse
|
29
|
Synthetic PreImplantation Factor (PIF) prevents fetal loss by modulating LPS induced inflammatory response. PLoS One 2017; 12:e0180642. [PMID: 28704412 PMCID: PMC5507516 DOI: 10.1371/journal.pone.0180642] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 06/19/2017] [Indexed: 12/31/2022] Open
Abstract
Maternal control of inflammation is essential during pregnancy and an exaggerated response is one of the underlying causes of fetal loss. Inflammatory response is mediated by multiple factors and Toll-like receptors (TLRs) are central. Activation of TLRs results in NALP-3 mediated assembly of apoptosis-associated speck-like protein containing a CARD (ASC) and caspase-1 into the inflammasome and production of pro-inflammatory cytokines IL-1β and IL-18. Given that preventing measures are lacking, we investigated PreImplantation Factor (PIF) as therapeutic option as PIF modulates Inflammation in pregnancy. Additionally, synthetic PIF (PIF analog) protects against multiple immune disorders. We used a LPS induced murine model of fetal loss and synthetic PIF reduced this fetal loss and increased the embryo weight significantly. We detected increased PIF expression in the placentae after LPS insult. The LPS induced serum and placenta cytokines were abolished by synthetic PIF treatment and importantly synthetic PIF modulated key members of inflammasome complex NALP-3, ASC, and caspase-1 as well. In conclusion our results indicate that synthetic PIF protects against LPS induced fetal loss, likely through modulation of inflammatory response especially the inflammasome complex. Given that synthetic PIF is currently tested in autoimmune diseases of non-pregnant subjects (clinicaltrials.gov, NCT02239562), therapeutic approach during pregnancy can be envisioned.
Collapse
|
30
|
de Barros IBL, Malvezzi H, Gueuvoghlanian-Silva BY, Piccinato CA, Rizzo LV, Podgaec S. “What do we know about regulatory T cells and endometriosis? A systematic review”. J Reprod Immunol 2017; 120:48-55. [DOI: 10.1016/j.jri.2017.04.003] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 04/05/2017] [Accepted: 04/06/2017] [Indexed: 12/31/2022]
|
31
|
Clark DA, Dmetrichuk JM, McCready E, Dhesy-Thind S, Arredondo JL. Changes in expression of the CD200 tolerance-signaling molecule and its receptor (CD200R) by villus trophoblasts during first trimester missed abortion and in chronic histiocytic intervillositis. Am J Reprod Immunol 2017; 78. [PMID: 28326648 DOI: 10.1111/aji.12665] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Accepted: 02/17/2017] [Indexed: 12/20/2022] Open
Abstract
PROBLEM Expression of CD200 at the feto-maternal interface is associated with successful murine and human pregnancy. CD200 binding to CD200 receptors on lymphomyeloid cells suppresses inflammation and induces Tregs. CD200 receptors are also expressed on mouse and human placental trophoblast cells. What is the expression of CD200 and CD200R in human missed abortions which have preserved Treg levels and in chronic histiocytic intervillositis (CHI) where maternal inflammatory cells cause IUGR? METHODS Immunohistiochemistry for CD200, CD200R, and Ki67 using human placental sections from missed abortions, term placenta, and CHI. PCR testing was done for trisomy in missed abortion. RESULTS CD200 and CD200R were expressed by human villus trophoblasts from 2 weeks post-implantation to term. Cytotrophoblast proliferation (Ki-67+ count) decreased at term. In first trimester missed abortion cases, CD200>CD200R villus trophoblasts accompanied missed abortion of non-trisomic male fetuses. CD200 and Ki67+ trophoblast proliferation was preserved in CHI with maternal inflammatory cell infiltration but CD200R was greatly decreased. CONCLUSION Residual CD200 activity may prevent completion of abortions via induction of Treg cells. In CHI, infiltrating maternal effector T cells may block Treg induction. An autocrine role for CD200-CD200R interaction versus inhibition of soluble CD200 by soluble CD200R is discussed.
Collapse
Affiliation(s)
- David A Clark
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Jennifer M Dmetrichuk
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Elizabeth McCready
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada.,Genetics Services, Hamilton Regional Laboratory Medicine Program, Hamilton Health Sciences, Hamilton, Ontario, Canada
| | | | - Jorge L Arredondo
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
32
|
Meng YH, Zhu XH, Yan LY, Zhang Y, Jin HY, Xia X, Li R, Qiao J. Bone mesenchymal stem cells improve pregnancy outcome by inducing maternal tolerance to the allogeneic fetus in abortion-prone matings in mouse. Placenta 2016; 47:29-36. [PMID: 27780537 DOI: 10.1016/j.placenta.2016.08.089] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 08/23/2016] [Accepted: 08/27/2016] [Indexed: 12/14/2022]
Abstract
INTRODUCTION The successful pregnancy depends on maternal immune tolerance against the fetus. It has been reported that MSCs (mesenchymal stem cells) could play a regulatory role on immune cells such as CD4+T cells, macrophages and NK cells, but their effect on recurrent miscarriage is unknown. STUDY DESIGN In a prospective study, the abortion-prone (CBA/J × DBA/2) H-2d × H-2k mice were utilized. Female CBA/J mice (8-10 weeks old) were injected with vehicle or MSCs via tail vein or uterine horns, and 14 days later, they were mated with DBA/2 males for the following experiments. RESULTS Comparing with the control group, the embryo resorption rate in MSCs-horn injection group was dramatically decreased. MSCs were mainly located at the maternal-fetal interface, indicating that the reduction of resorption rate was due to MSCs' local effect. No matter which treatment was given, there was no significant difference in the levels of IL-4, IL-10, TNF-α and IFN-γ in CD4+T cells and IL-10 and IL-12 in macrophages in spleens among each group. However, in contrast to other groups, the levels of IL-4 and IL-10 in CD4+T cells localized at the maternal-fetal interface in MSCs-horn injection group were dramatically increased, and TNF-α and IFN-γ levels were notably decreased. While IL-10 expressed in macrophages was obviously higher than other groups and IL-12 in macrophages was significantly lower than other groups. CONCLUSIONS The findings indicate that MSCs injection through uterine horns could decrease embryo resorption rate.
Collapse
Affiliation(s)
- Yu-Han Meng
- Reproductive Medical Center, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China; Reproductive Medical Center, Affiliated Hospital of Weifang Medical University, Weifang, China; Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China
| | - Xiao-Hui Zhu
- Reproductive Medical Center, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China; Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China
| | - Li-Ying Yan
- Reproductive Medical Center, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China; Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China
| | - Yan Zhang
- Reproductive Medical Center, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China; Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China
| | - Hong-Yan Jin
- Reproductive Medical Center, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China; Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China
| | - Xi Xia
- Reproductive Medical Center, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China; Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China
| | - Rong Li
- Reproductive Medical Center, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China; Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Jie Qiao
- Reproductive Medical Center, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China; Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China.
| |
Collapse
|
33
|
Pregnancy and multiple sclerosis: from molecular mechanisms to clinical application. Semin Immunopathol 2016; 38:709-718. [PMID: 27501960 DOI: 10.1007/s00281-016-0584-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 07/19/2016] [Indexed: 12/14/2022]
Abstract
Translational research generally refers to a "bench to bedside" approach where basic science discoveries in models move to clinical trials in humans. However, a "bedside to bench to bedside" approach may be more promising with respect to clinical relevance, since it starts with a clinical observation that can serve as a research paradigm to elucidate mechanisms and translate them back into novel therapeutic approaches. The effect of pregnancy on human autoimmune disorders in general, and multiple sclerosis (MS) in particular, serves as an intriguing example of how this can be used to understand disease pathobiology and discover new therapeutic targets. Disease activity in MS undergoes pronounced shifts in the time before, during, and after pregnancy. The most well-known and established example is a reduction in relapse rates in the last trimester by 70-80 %. However, disease activity reappears in the first few months after delivery, temporarily overshooting pre-pregnancy levels. This phenomenon has since its first description served as a model for investigating novel treatment options in animal models and has cumulated in successful phase 2a and 2b trials in female MS patients. However, recently, a number of other clinical observations have been made that might be similarly suitable to offer additional insights into pathobiological mechanisms of MS activity, progression, and possibly even incidence. Here, we outline the various changes in the clinical course of MS that have been described in relation to pregnancy, both short term and long term, and discuss how these may inform the development of novel treatments for autoimmune diseases.
Collapse
|