1
|
Wekema L, Schoenmakers S, Schenkelaars N, Laskewitz A, Huurman RH, Liu L, Walters L, Harmsen HJM, Steegers-Theunissen RPM, Faas MM. Diet-Induced Obesity in Mice Affects the Maternal Gut Microbiota and Immune Response in Mid-Pregnancy. Int J Mol Sci 2024; 25:9076. [PMID: 39201761 PMCID: PMC11354285 DOI: 10.3390/ijms25169076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 08/10/2024] [Accepted: 08/17/2024] [Indexed: 09/03/2024] Open
Abstract
Maternal obesity during pregnancy is associated with adverse pregnancy outcomes. This might be due to undesired obesity-induced changes in the maternal gut microbiota and related changes in the maternal immune adaptations during pregnancy. The current study examines how obesity affects gut microbiota and immunity in pregnant obese and lean mice during mid-pregnancy (gestational day 12 (GD12)). C57BL/6 mice were fed a high-fat diet or low-fat diet from 8 weeks before mating and during pregnancy. At GD12, we analyzed the gut microbiota composition in the feces and immune responses in the intestine (Peyer's patches, mesenteric lymph nodes) and the peripheral circulation (spleen and peripheral blood). Maternal obesity reduced beneficial bacteria (e.g., Bifidobacterium and Akkermansia) and changed intestinal and peripheral immune responses (e.g., dendritic cells, Th1/Th2/Th17/Treg axis, monocytes). Numerous correlations were found between obesity-associated bacterial genera and intestinal/peripheral immune anomalies. This study shows that maternal obesity impacts the abundance of specific bacterial gut genera as compared to lean mice and deranges maternal intestinal immune responses that subsequently change peripheral maternal immune responses in mid-pregnancy. Our findings underscore the opportunities for early intervention strategies targeting maternal obesity, ideally starting in the periconceptional period, to mitigate these obesity-related pregnancy effects.
Collapse
Affiliation(s)
- Lieske Wekema
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands; (A.L.); (R.H.H.)
| | - Sam Schoenmakers
- Department of Obstetrics and Gynaecology, Erasmus Medical Center, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands; (S.S.); (N.S.); (R.P.M.S.-T.)
| | - Nicole Schenkelaars
- Department of Obstetrics and Gynaecology, Erasmus Medical Center, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands; (S.S.); (N.S.); (R.P.M.S.-T.)
| | - Anne Laskewitz
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands; (A.L.); (R.H.H.)
| | - Romy H. Huurman
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands; (A.L.); (R.H.H.)
| | - Lei Liu
- Department of Medical Microbiology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands; (L.L.); (L.W.); (H.J.M.H.)
| | - Lisa Walters
- Department of Medical Microbiology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands; (L.L.); (L.W.); (H.J.M.H.)
| | - Hermie J. M. Harmsen
- Department of Medical Microbiology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands; (L.L.); (L.W.); (H.J.M.H.)
| | - Régine P. M. Steegers-Theunissen
- Department of Obstetrics and Gynaecology, Erasmus Medical Center, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands; (S.S.); (N.S.); (R.P.M.S.-T.)
| | - Marijke M. Faas
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands; (A.L.); (R.H.H.)
- Department of Obstetrics and Gynaecology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| |
Collapse
|
2
|
Wekema L, Schoenmakers S, Schenkelaars N, Laskewitz A, Liu L, Walters L, Harmsen HJM, Steegers-Theunissen RPM, Faas MM. Obesity and diet independently affect maternal immunity, maternal gut microbiota and pregnancy outcome in mice. Front Immunol 2024; 15:1376583. [PMID: 39072322 PMCID: PMC11272480 DOI: 10.3389/fimmu.2024.1376583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 06/24/2024] [Indexed: 07/30/2024] Open
Abstract
Introduction Maternal obesity poses risks for both mother and offspring during pregnancy, with underlying mechanisms remaining largely unexplored. Obesity is associated with microbial gut dysbiosis and low-grade inflammation, and also the diet has a major impact on these parameters. This study aimed to investigate how maternal obesity and diet contribute to changes in immune responses, exploring potential associations with gut microbiota dysbiosis and adverse pregnancy outcomes in mice. Methods Before mating, C57BL/6 mice were assigned to either a high-fat-diet (HFD) or low-fat-diet (LFD) to obtain obese (n=17) and lean (n=10) mice. To distinguish between the effects of obesity and diet, 7 obese mice were switched from the HFD to the LFD from day 7 until day 18 of pregnancy ("switch group"), which was the endpoint of the study. T helper (Th) cell subsets were studied in the spleen, mesenteric lymph nodes (MLN) and Peyer's patches (PP), while monocyte subsets and activation status were determined in maternal blood (flow cytometry). Feces were collected before and during pregnancy (day 7,14,18) for microbiota analysis (16S rRNA sequencing). Pregnancy outcome included determination of fetal and placental weight. Results Obesity increased splenic Th1 and regulatory T cells, MLN Th1 and PP Th17 cells and enhanced IFN-γ and IL-17A production by splenic Th cells upon ex vivo stimulation. Switching diet decreased splenic and PP Th2 cells and classical monocytes, increased intermediate monocytes and activation of intermediate/nonclassical monocytes. Obesity and diet independently induced changes in the gut microbiota. Various bacterial genera were increased or decreased by obesity or the diet switch. These changes correlated with the immunological changes. Fetal weight was lower in the obese than the lean group, while placental weight was lower in the switch than the obese group. Discussion This study demonstrates that obesity and diet independently impact peripheral and intestinal immune responses at the end of pregnancy. Simultaneously, both factors affect specific bacterial gut genera and lead to reduced fetal or placental weight. Our data suggest that switching diet during pregnancy to improve maternal health is not advisable and it supports pre/probiotic treatment of maternal obesity-induced gut dysbiosis to improve maternal immune responses and pregnancy outcome.
Collapse
Affiliation(s)
- Lieske Wekema
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Sam Schoenmakers
- Department of Obstetrics and Gynaecology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Nicole Schenkelaars
- Department of Obstetrics and Gynaecology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Anne Laskewitz
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Lei Liu
- Department of Medical Microbiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Lisa Walters
- Department of Medical Microbiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Hermie J. M. Harmsen
- Department of Medical Microbiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | | | - Marijke M. Faas
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- Department of Obstetrics and Gynaecology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
3
|
Okuyama MW, Moriyoshi M, Katagiri S. Changes in interleukin-2, -4, -6 and -8 expression in the postovulatory sow endometrium after artificial insemination based on conceived or failed to conceive. J Reprod Dev 2024; 70:192-196. [PMID: 38616120 PMCID: PMC11153119 DOI: 10.1262/jrd.2023-094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 03/24/2024] [Indexed: 04/16/2024] Open
Abstract
The establishment and maintenance of a pregnancy requires proper interaction between the endocrine and immune systems in the uterus. Therefore, it is crucial to understand how changes in endometrial cytokine levels facilitate reproduction. This study aimed to investigate how representative cytokines sequentially changed in the endometrium and whether conception could be attributed to these changes. In this study, artificial insemination was performed twice in 160 sows and ovulation was examined every 3 h using transrectal ultrasonography. Uterine endometrial tissues were obtained via repeated biopsies at 2, 4, 6, 8, 12, 16, and 20 h after ovulation and interleukin (IL)-2, IL-4, IL-6, and IL-8 expression was examined using real-time polymerase chain reaction. The conception rate was 91.9%. The IL-2 levels showed no differences in conception or time. The expression peaks of IL-4 and IL-6 were delayed in sows that failed to conceive within 4-6 h and 2 h, respectively, compared to those that did conceive. In sows that conceived, IL-8 was highest after 2 h, and no difference was observed at other time point, regardless of conception. In sows that failed to conceive, the increase in IL-8 levels might have been cancelled or terminated before the first sampling time. These results highlight the importance of timely increases and subsequent declines in the levels of some cytokines for the establishment of pregnancy. Differences in uterine capacity start just after ovulation; detection and correction of these deviations can improve the reproductive efficiency of sows.
Collapse
Affiliation(s)
- Minami W Okuyama
- Faculty of Medicine, Oita University, Yufu 879-5593, Japan
- Laboratory of Theriogenology, Department of Large Animal Clinical Sciences, School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu 069-8501, Japan
| | - Masaharu Moriyoshi
- Laboratory of Theriogenology, Department of Large Animal Clinical Sciences, School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu 069-8501, Japan
| | - Seiji Katagiri
- Laboratory of Theriogenology, Department of Veterinary Clinical Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
- Laboratory of Theriogenology, Department of Large Animal Clinical Sciences, School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu 069-8501, Japan
| |
Collapse
|
4
|
Rokhsartalab Azar P, Karimi S, Haghtalab A, Taram S, Hejazi M, Sadeghpour S, Pashaei MR, Ghasemnejad-Berenji H, Taheri-Anganeh M. The role of the endometrial microbiome in embryo implantation and recurrent implantation failure. J Reprod Immunol 2024; 162:104192. [PMID: 38215650 DOI: 10.1016/j.jri.2024.104192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 12/21/2023] [Accepted: 01/01/2024] [Indexed: 01/14/2024]
Abstract
There is a suggested pathophysiology associated with endometrial microbiota in cases where repeated implantation failure of high-quality embryos is observed. However, there is a suspected association between endometrial microbiota and the pathogenesis of implantation failure. However, there is still a lack of agreement on the fundamental composition of the physiological microbiome within the uterine cavity. This is primarily due to various limitations in the studies conducted, including small sample sizes and variations in experimental designs. As a result, the impact of bacterial communities in the endometrium on human reproduction is still a subject of debate. In this discourse, we undertake a comprehensive examination of the existing body of research pertaining to the uterine microbiota and its intricate interplay with the process of embryo implantation.
Collapse
Affiliation(s)
| | - Sarmad Karimi
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| | - Arian Haghtalab
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| | - Saman Taram
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| | - Milad Hejazi
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| | - Sonia Sadeghpour
- Reproductive Health Research Center, Clinical Research Institute, Urmia University of Medical Sciences, Urmia, Iran; Department of Obstetrics and Gynecology, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Mohammad Reza Pashaei
- Department of Internal Medicine, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Hojat Ghasemnejad-Berenji
- Reproductive Health Research Center, Clinical Research Institute, Urmia University of Medical Sciences, Urmia, Iran.
| | - Mortaza Taheri-Anganeh
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|
5
|
Omoto T, Kyozuka H, Murata T, Fukuda T, Isogami H, Okoshi C, Yasuda S, Yamaguchi A, Sato A, Ogata Y, Nagasaka Y, Hosoya M, Yasumura S, Hashimoto K, Nishigori H, Fujimori K, The Japan Environment And Children's Study Group. Association between Preconception Dietary Fiber Intake and Preterm Birth: The Japan Environment and Children's Study. Nutrients 2024; 16:713. [PMID: 38474840 DOI: 10.3390/nu16050713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 02/27/2024] [Accepted: 02/28/2024] [Indexed: 03/14/2024] Open
Abstract
Preterm birth (PTB) is a leading cause of neonatal morbidity and mortality. Therefore, this study aimed to determine whether preconception dietary fiber intake is associated with PTB. This was a prospective cohort Japan Environmental and Children's Study (JECS). The study population comprised 85,116 singleton live-birth pregnancies from the JECS database delivered between 2011 and 2014. The participants were categorized into five groups based on their preconception dietary fiber intake quintiles (Q1 and Q5 were the lowest and highest groups, respectively). Multiple logistic regression analysis was performed to determine the association between preconception dietary fiber intake and PTB. Multiple logistic regression analysis revealed that the risk for PTB before 34 weeks was lower in the Q3, Q4, and Q5 groups than in the Q1 group (Q3: adjusted odds ratio [aOR] 0.78, 95% confidence interval [CI] 0.62-0.997; Q4: aOR 0.74, 95% CI 0.57-0.95; Q5: aOR 0.68, 95% CI 0.50-0.92). However, there was no significant difference between preconception dietary fiber intake and PTB before 37 weeks. In conclusion, higher preconception dietary fiber intake correlated with a reduced the risk for PTB before 34 weeks. Therefore, new recommendations on dietary fiber intake as part of preconception care should be considered.
Collapse
Affiliation(s)
- Takahiro Omoto
- Fukushima Regional Center for the Japan Environmental and Children's Study, Fukushima 960-1295, Japan
- Department of Obstetrics and Gynecology, School of Medicine, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Hyo Kyozuka
- Fukushima Regional Center for the Japan Environmental and Children's Study, Fukushima 960-1295, Japan
- Department of Obstetrics and Gynecology, School of Medicine, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Tsuyoshi Murata
- Fukushima Regional Center for the Japan Environmental and Children's Study, Fukushima 960-1295, Japan
- Department of Obstetrics and Gynecology, School of Medicine, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Toma Fukuda
- Fukushima Regional Center for the Japan Environmental and Children's Study, Fukushima 960-1295, Japan
- Department of Obstetrics and Gynecology, School of Medicine, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Hirotaka Isogami
- Fukushima Regional Center for the Japan Environmental and Children's Study, Fukushima 960-1295, Japan
- Department of Obstetrics and Gynecology, School of Medicine, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Chihiro Okoshi
- Fukushima Regional Center for the Japan Environmental and Children's Study, Fukushima 960-1295, Japan
- Department of Obstetrics and Gynecology, School of Medicine, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Shun Yasuda
- Fukushima Regional Center for the Japan Environmental and Children's Study, Fukushima 960-1295, Japan
- Department of Obstetrics and Gynecology, School of Medicine, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Akiko Yamaguchi
- Fukushima Regional Center for the Japan Environmental and Children's Study, Fukushima 960-1295, Japan
- Department of Obstetrics and Gynecology, School of Medicine, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Akiko Sato
- Fukushima Regional Center for the Japan Environmental and Children's Study, Fukushima 960-1295, Japan
| | - Yuka Ogata
- Fukushima Regional Center for the Japan Environmental and Children's Study, Fukushima 960-1295, Japan
| | - Yuichi Nagasaka
- Fukushima Regional Center for the Japan Environmental and Children's Study, Fukushima 960-1295, Japan
| | - Mitsuaki Hosoya
- Fukushima Regional Center for the Japan Environmental and Children's Study, Fukushima 960-1295, Japan
- Department of Pediatrics, School of Medicine, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Seiji Yasumura
- Fukushima Regional Center for the Japan Environmental and Children's Study, Fukushima 960-1295, Japan
- Department of Public Health, School of Medicine, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Koichi Hashimoto
- Fukushima Regional Center for the Japan Environmental and Children's Study, Fukushima 960-1295, Japan
- Department of Pediatrics, School of Medicine, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Hidekazu Nishigori
- Fukushima Regional Center for the Japan Environmental and Children's Study, Fukushima 960-1295, Japan
- Fukushima Medical Center for Children and Women, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Keiya Fujimori
- Fukushima Regional Center for the Japan Environmental and Children's Study, Fukushima 960-1295, Japan
- Department of Obstetrics and Gynecology, School of Medicine, Fukushima Medical University, Fukushima 960-1295, Japan
| | | |
Collapse
|
6
|
Sharon Y, Adler A, Tiosano A, Sanchez JM, Amer R, Kramer M. The effect of pregnancy on the course of uveitis in single and multiple pregnancies. Graefes Arch Clin Exp Ophthalmol 2023; 261:803-808. [PMID: 36094584 DOI: 10.1007/s00417-022-05829-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 08/31/2022] [Accepted: 09/02/2022] [Indexed: 11/04/2022] Open
Abstract
PURPOSE To explore the effect of pregnancy on the clinical course, outcome, and treatment in multiparous women with non-infectious uveitis. METHODS Retrospective study of women with a history of non-infectious uveitis and pregnancies prior to and during disease course. Disease activity and severity 1-year prior pregnancy, during pregnancy, and 1-year postpartum were recorded as well as patients' and diseases' characteristics. The main outcome measures included the rate and severity of uveitis attacks and the effect on ocular complications and therapies. RESULTS Included were 32 women (70 pregnancies, mean of 2.6 pregnancies/patient), with a mean follow-up time of 6.5 years. The most common uveitis types were anterior (31%) and pan-uveitis (31%). Flare-ups were more frequent in the year prior to pregnancy, in the first trimester, and in the postpartum period and decreased markedly during pregnancy. Women who experienced a flare-up during pregnancy had a higher rate of flare-ups in the year prior pregnancy than those who did not experience a flare-up during pregnancy (p-0.047). The rate of flare-ups 12 months' postpartum was also higher compared to women without any flare-up during pregnancy (p = 0.01). Severity of flare-ups in the postpartum period was worse in women who experienced a flare-up during pregnancy compared to women without flare-ups (p = 0.001). The severity of flare-ups was higher in the first pregnancy compared to subsequent pregnancies. CONCLUSIONS Women who had active or non-controlled uveitis prior to pregnancy have higher disease activity and severity during pregnancy as well. The first pregnancy seems to behave differently from subsequent pregnancies, in terms of disease severity.
Collapse
Affiliation(s)
- Yael Sharon
- Department of Ophthalmology, Rabin Medical Center-Beilinson Hospital, 39 Jabotinsky St., 49100, Petach Tikva, Israel. .,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - Avital Adler
- Department of Ophthalmology, Rabin Medical Center-Beilinson Hospital, 39 Jabotinsky St., 49100, Petach Tikva, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Alon Tiosano
- Department of Ophthalmology, Rabin Medical Center-Beilinson Hospital, 39 Jabotinsky St., 49100, Petach Tikva, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Juan M Sanchez
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Department of Ophthalmology, Hadassah University Hospital, Jerusalem, Israel
| | - Radgonde Amer
- Department of Ophthalmology, Hadassah University Hospital, Jerusalem, Israel
| | - Michal Kramer
- Department of Ophthalmology, Rabin Medical Center-Beilinson Hospital, 39 Jabotinsky St., 49100, Petach Tikva, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
7
|
Parker J, Dubin A, Schneider R, Wagner KS, Jentoft S, Böhne A, Bayer T, Roth O. Immunological tolerance in the evolution of male pregnancy. Mol Ecol 2023; 32:819-840. [PMID: 34951070 DOI: 10.1111/mec.16333] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 12/12/2021] [Accepted: 12/14/2021] [Indexed: 11/29/2022]
Abstract
The unique male pregnancy in pipefishes and seahorses ranges from basic attachment (pouch-less species: Nerophinae) of maternal eggs to specialized internal gestation in pouched species (e.g. Syngnathus and Hippocampus) with many transitions in between. Due to this diversity, male pregnancy offers a unique platform for assessing physiological and molecular adaptations in pregnancy evolution. These insights will contribute to answering long-standing questions of why and how pregnancy evolved convergently in so many vertebrate systems. To understand the molecular congruencies and disparities in male pregnancy evolution, we compared transcriptome-wide differentially expressed genes in four syngnathid species, at four pregnancy stages (nonpregnant, early, late and parturition). Across all species and pregnancy forms, metabolic processes and immune dynamics defined pregnancy stages, especially pouched species shared expression features akin to female pregnancy. The observed downregulation of adaptive immune genes in early-stage pregnancy and its reversed upregulation during late/parturition in pouched species, most notably in Hippocampus, combined with directionless expression in the pouch-less species, suggests immune modulation to be restricted to pouched species that evolved placenta-like systems. We propose that increased foeto-paternal intimacy in pouched syngnathids commands immune suppression processes in early gestation, and that the elevated immune response during parturition coincides with pouch opening and reduced progeny reliance. Immune response regulation in pouched species supports the recently described functional MHC II pathway loss as critical in male pregnancy evolution. The independent co-option of similar genes and pathways both in male and female pregnancy highlights immune modulation as crucial for the evolutionary establishment of pregnancy.
Collapse
Affiliation(s)
- Jamie Parker
- Marine Evolutionary Ecology, GEOMAR Helmholtz Centre for Ocean Research Kiel, Kiel, Germany
| | - Arseny Dubin
- Marine Evolutionary Ecology, GEOMAR Helmholtz Centre for Ocean Research Kiel, Kiel, Germany
| | - Ralf Schneider
- Marine Evolutionary Ecology, GEOMAR Helmholtz Centre for Ocean Research Kiel, Kiel, Germany
| | - Kim Sara Wagner
- Marine Evolutionary Ecology, GEOMAR Helmholtz Centre for Ocean Research Kiel, Kiel, Germany
| | - Sissel Jentoft
- Department of Biosciences, Centre for Ecological and Evolutionary Synthesis, University of Oslo, Oslo, Norway
| | - Astrid Böhne
- Center for Molecular Biodiversity Research, Zoological Research Museum Alexander Koenig, Bonn, Germany
| | - Till Bayer
- Marine Evolutionary Ecology, GEOMAR Helmholtz Centre for Ocean Research Kiel, Kiel, Germany
| | - Olivia Roth
- Marine Evolutionary Ecology, GEOMAR Helmholtz Centre for Ocean Research Kiel, Kiel, Germany
| |
Collapse
|
8
|
Mohammed RHA, Mumtaz H, Sangah AB, Shaikh SS, Nasir N, Jabeen S. Pregnancy in lupus: an updated consensus to guide best practice strategies. EGYPTIAN RHEUMATOLOGY AND REHABILITATION 2022. [DOI: 10.1186/s43166-022-00167-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Abstract
Background
Systemic lupus erythematosus is a multifaceted chronic relapsing autoimmune disease of unknown etiology. The disease has always been a serious diagnosis in women being a multisystem pathology that is classically encountered during the childbearing age posing serious systemic comorbidities with a potential impact on the functional performance, psychosocial status, and survival. In this article, we review critical issues related to the decision to conceive in female with lupus highlighting the impact of the diagnosis and disease activity status on the mother and the fetus, attempting to suggest a consensus to guide safe decision making for pregnancy with SLE.
Main body
The pleomorphic dysregulated immune nature of lupus in the presence of uncontrolled disease carries a higher risk of complicated pregnancy. Therefore, SLE pregnancies should be well planned and are usually encouraged if the disease is inactive (at least 6 months prior to conception) to ensure immune quiescence towards a safer outcome.
Conclusion
With the proper implementation of preconception counseling strategy, choice of the correct timing of conception, close monitoring of SLE flares with tight control, and the appreciation of the value of multidisciplinary management to best practice most young women with SLE can carry on successful pregnancies with favorable outcome.
Collapse
|
9
|
Mora-Palazuelos C, Bermúdez M, Aguilar-Medina M, Ramos-Payan R, Ayala-Ham A, Romero-Quintana JG. Cytokine-polymorphisms associated with Preeclampsia: A review. Medicine (Baltimore) 2022; 101:e30870. [PMID: 36181055 PMCID: PMC9524891 DOI: 10.1097/md.0000000000030870] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Preeclampsia (PE) is a syndromic disorder that affects 2% to 8% of pregnancies and is diagnosed principally when hypertension appears in the second-d half of pregnancy. WHO estimates the incidence of PE to be seven times higher in developing countries than in developed countries. Severe preeclampsia/eclampsia is one of the most important causes of maternal mortality, associated with 50,000 to 100,000 annual deaths globally as well as serious fetal and neonatal morbidity and mortality, especially in developing countries. Even though evidence from family-based studies suggest PE has a heritable component, its etiology, and specific genetic contributions remain unclear. Many studies examining the genetic factors contributing to PE have been conducted, most of them are focused on single nucleotide polymorphisms (SNPs). Given that PE has a very important inflammatory component, is mandatory to examine cytokine-SNPs for elucidating all mechanisms involved in this pathology. In this review, we describe the most important cytokine-polymorphisms associated with the onset and development of PE. We aim to provide current and relevant evidence in this regard. METHODS We searched English databases such as PubMed and the National Center for Biotechnology Information. The publication time of the papers was set from the establishment of the databases to February 2022. All studies about Th1/Th2/Th17 cytokines polymorphisms were included in our study. RESULTS SNPs in IFN-γ, TNF-α, IL-4, IL-6, IL-10, IL-17A, and IL-22 are associated with the development, early-onset and severity of PE, being the Th1/Th2/Th17 responses affected by the presence of these SNPs. CONCLUSIONS The changes in Th1/Th2/Th17 response modify processes such as placentation, control of inflammation, and vascular function. Nonetheless, association studies have shown different results depending on sample size, diagnostic, and population.
Collapse
Affiliation(s)
| | - Mercedes Bermúdez
- Facultad de Odontología, Universidad Autónoma de Chihuahua, Chihuahua, México
| | - Maribel Aguilar-Medina
- Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Sinaloa, Culiacán, México
| | - Rosalío Ramos-Payan
- Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Sinaloa, Culiacán, México
| | - Alfredo Ayala-Ham
- Facultad de Biología, Universidad Autónoma de Sinaloa, Culiacán, México
| | - Jose Geovanni Romero-Quintana
- Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Sinaloa, Culiacán, México
- *Correspondence: Jose Geovanni Romero-Quintana, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Sinaloa, Josefa Ortiz DE Domínguez S/N y Avenida DE las Américas, CP. 80010, Culiacán, Sinaloa, México (e-mail: )
| |
Collapse
|
10
|
Azadehrah M, Vosoogh S, Azadehrah M. The roles and therapeutic applications of cytokines in endometrial cancer. J Reprod Immunol 2022; 152:103652. [PMID: 35753146 DOI: 10.1016/j.jri.2022.103652] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 05/21/2022] [Accepted: 06/05/2022] [Indexed: 12/26/2022]
Abstract
Endometrial cancer (EC) is a common gynecological cancer globally and the most frequent gynecologic malignancy in industrialized countries. Patients are typically diagnosed when the disease is still restricted to the uterus. 5-year overall survival ranges from 70 % to 90 % in patients without metastatic disease; however, the metastatic form of the disease affects 16 % of EC patients, with a 5-year survival rate of 16.8 %. The immune system can detect abnormal cells as non-self in the early stages of carcinogenesis, producing the appropriate pro-inflammatory environment to eliminate cancer cells. In a second phase, cancer cells use various immune-editing systems to alter the profile of the immune response from pro to anti-inflammatory, resulting in immune escape. The directors of this immune switching mechanism are cytokines. Studies have reported the increased expression of several pro-and anti-inflammatory cytokines in EC tissues and cell lines, including Interleukin (IL)- 6, IL-8, IL-31, IL-33, IL-10, TGF-β, VEGF, and IL-1Ra. Immune cells producing these cytokines have also been reported to be present in EC tissues. Therefore, in this study, we aimed to show the possible mechanisms of the mentioned cytokines on EC progression, as well as the most current and prospective advancements in cytokine-based therapeutic applications.
Collapse
Affiliation(s)
- Malihe Azadehrah
- Cancer Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Shohre Vosoogh
- Clinical Research Development Unit (CRDU), Sayad Shirazi Hospital, Golestan university of Medical Sciences, Gorgan, Iran
| | - Mahboobeh Azadehrah
- Cancer Research Center, Golestan University of Medical Sciences, Gorgan, Iran.
| |
Collapse
|
11
|
Sanchez-Vicente JL, Moruno-Rodríguez A, De las Morenas-Iglesias J, Gonzalez-Jauregui B, Franco-Ruedas C, Lechon-Caballero B, Talego-Sancha A, Rueda-Rueda T, Del Estad-Cabello A, López-Herrero F. Clinical Manifestations and Treatment of Vogt–Koyanagi–Harada Disease during Pregnancy and after Birth: A Case Report. Ocul Immunol Inflamm 2022; 31:830-837. [PMID: 35404732 DOI: 10.1080/09273948.2022.2049314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
PURPOSE To describe the clinical manifestations of Vogt-Koyanagi-Harada (VKH) disease during pregnancy and after birth and the therapeutic challenge of treating patients with this condition. METHODS We describe the clinical manifestations of this disease, as well as the diagnostic tests and treatments performed. RESULTS The patient was referred for evaluation due to a persistent headache. Examination revealed bilateral anterior uveitis, papillitis and yellowish-white choroidal lesions in both eyes. A tentative diagnosis of VKH disease was made. A multimodal imaging study was performed at the time of presentation and throughout the disease course. The patient was initially treated with intravenous corticosteroids, and subsequently, oral corticosteroids and cyclosporine were administered. Clinical manifestations increased in severity after childbirth. CONCLUSIONS The clinical course of VKH disease can be modified by pregnancy. While clinical manifestations during gestation may be mild, these may be exacerbated after birth. Treatment with corticosteroids and cyclosporine can be effective.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Arturo Talego-Sancha
- Department of Ophthalmology, Virgen Del Rocío University Hospital, Seville, Spain
| | - Trinidad Rueda-Rueda
- Department of Ophthalmology, Virgen Del Rocío University Hospital, Seville, Spain
| | | | | |
Collapse
|
12
|
Fransson E. Psychoneuroimmunology in the context of perinatal depression - Tools for improved clinical practice. Brain Behav Immun Health 2021; 17:100332. [PMID: 34589817 PMCID: PMC8474604 DOI: 10.1016/j.bbih.2021.100332] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 08/17/2021] [Accepted: 08/18/2021] [Indexed: 12/15/2022] Open
Abstract
Maternal mental health spans in a temporary manner from pre-conception through the phases of pregnancy, childbirth, and the postpartum period (i.e., perinatal). The psychoneuroimmunology (PNI) field has made important contributions to the knowledge of the pathophysiology of poor perinatal mental health, but the PNI lens could be used more broadly to inform clinical practice. This review argues that PNI holds the key to several important aspects of variations in mental health for pregnant and postpartum women. This review describes existing knowledge from studies on immune activation in maternal depression during pregnancy and postpartum, and other important features such as stress reactivity, the microbiome, and its metabolites. The importance of objective measures for screening and prediction is discussed as well as the need for novel therapeutics to treat poor mental health in the perinatal period. The PNI framework could thus be further applied to inform research about the mechanisms of perinatal psychiatric morbidity, which could pave the way for future precision medicine for perinatal mental health issues.
Collapse
Affiliation(s)
- Emma Fransson
- Centre for Translational Microbiome Research, Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
- Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden
| |
Collapse
|
13
|
Günther V, Alkatout I, Meyerholz L, Maass N, Görg S, von Otte S, Ziemann M. Live Birth Rates after Active Immunization with Partner Lymphocytes. Biomedicines 2021; 9:biomedicines9101350. [PMID: 34680467 PMCID: PMC8533392 DOI: 10.3390/biomedicines9101350] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 09/23/2021] [Accepted: 09/26/2021] [Indexed: 12/30/2022] Open
Abstract
Although many potential causes have been established for recurrent implantation failure (RIF) and recurrent miscarriage (RM), about 50% of these remain idiopathic. Scientific research is focused on immunological risk factors. In the present study, we aim to evaluate live birth rates after immunization with paternal lymphocytes (lymphocyte immunotherapy (LIT)). This retrospective study consisted of 148 couples with a history of RM and/or RIF. The women underwent immunization with lymphocytes of their respective partners from November 2017 to August 2019. Fifty-five patients (43%) had live births. Stratified by indication (RM, RIF, combined), live birth rates in the RM and the combined group were significantly higher than that in the RIF group (53%, 59% and 33%, respectively, p = 0.02). The difference was especially noticeable during the first 90 days after immunization (conception rate leading to live births: 31%, 23% and 8% for RM, the combined group and RIF, respectively; p = 0.005), while there was no difference between groups during the later follow-up. LIT was associated with high live birth rates, especially in women with recurrent miscarriage. In view of the limited data from randomized studies, LIT cannot be recommended as routine therapy. However, it may be considered in individual cases.
Collapse
Affiliation(s)
- Veronika Günther
- Department of Obstetrics and Gynecology, University Hospitals Schleswig-Holstein, Campus Kiel, Arnold-Heller-Straße 3 (House C), 24105 Kiel, Germany; (I.A.); (L.M.); (N.M.)
- University Fertility Center, Ambulanzzentrum gGmbH, University Hospitals Schleswig-Holstein, Campus Kiel, Arnold-Heller-Straße 3 (House C), 24105 Kiel, Germany;
- Institute for Transfusion Medicine and Transplant Center, University Hospitals Schleswig-Holstein, Campus Kiel, Arnold-Heller-Straße 3 (House 17), 24105 Kiel, Germany; (S.G.); (M.Z.)
- Institute for Transfusion Medicine and Transplant Center, University Hospitals Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160 (House 31), 23538 Lübeck, Germany
- Correspondence: ; Tel.: +49-(0)431-500-21401
| | - Ibrahim Alkatout
- Department of Obstetrics and Gynecology, University Hospitals Schleswig-Holstein, Campus Kiel, Arnold-Heller-Straße 3 (House C), 24105 Kiel, Germany; (I.A.); (L.M.); (N.M.)
| | - Lisa Meyerholz
- Department of Obstetrics and Gynecology, University Hospitals Schleswig-Holstein, Campus Kiel, Arnold-Heller-Straße 3 (House C), 24105 Kiel, Germany; (I.A.); (L.M.); (N.M.)
| | - Nicolai Maass
- Department of Obstetrics and Gynecology, University Hospitals Schleswig-Holstein, Campus Kiel, Arnold-Heller-Straße 3 (House C), 24105 Kiel, Germany; (I.A.); (L.M.); (N.M.)
| | - Siegfried Görg
- Institute for Transfusion Medicine and Transplant Center, University Hospitals Schleswig-Holstein, Campus Kiel, Arnold-Heller-Straße 3 (House 17), 24105 Kiel, Germany; (S.G.); (M.Z.)
- Institute for Transfusion Medicine and Transplant Center, University Hospitals Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160 (House 31), 23538 Lübeck, Germany
| | - Sören von Otte
- University Fertility Center, Ambulanzzentrum gGmbH, University Hospitals Schleswig-Holstein, Campus Kiel, Arnold-Heller-Straße 3 (House C), 24105 Kiel, Germany;
| | - Malte Ziemann
- Institute for Transfusion Medicine and Transplant Center, University Hospitals Schleswig-Holstein, Campus Kiel, Arnold-Heller-Straße 3 (House 17), 24105 Kiel, Germany; (S.G.); (M.Z.)
- Institute for Transfusion Medicine and Transplant Center, University Hospitals Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160 (House 31), 23538 Lübeck, Germany
| |
Collapse
|
14
|
Adur MK, Braundmeier-Fleming AG, Lessey BA, Nowak RA. Altered eutopic endometrial T-regulatory and T-helper 17 lymphocyte ratio in women with unexplained subfertility. JOURNAL OF ENDOMETRIOSIS AND PELVIC PAIN DISORDERS 2021; 13:185-194. [PMID: 34354965 PMCID: PMC8330881 DOI: 10.1177/22840265211018544] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
PROBLEM Perturbations in T-helper lymphocyte profiles have previously been associated with endometriosis related subfertility and conception failure. Hence a retrospective in vitro study was conducted to evaluate the relationship between T-regulatory (Treg) and T-helper 17 (Th17) lymphocytes in the eutopic endometrium of women with unexplained subfertility and correlate these profiles to their conception status. METHOD OF STUDY Eutopic endometrial biopsies were collected during the mid-secretory phase of the menstrual cycle, from women with unexplained subfertility. These samples were evaluated immunohistochemically for Treg and Th17 lymphocytes as well as the related proinflammatory cytokine, Interleukin-17 (IL-17). These eutopic endometrial T lymphocyte subpopulations were compared to the patients' conception status in subsequent cycles. RESULTS Though Treg cells were not indicative of conception success in subsequent cycles, patients who maintained their subfertile (no conception) status were observed to have a higher Th17 cell count in their eutopic endometrium. The ratio of Treg:Th17 cell counts was significantly correlated to patient conception status as well. These trends stayed consistent irrespective of concurrent endometriosis. CONCLUSION Patients with a high proinflammatory Th17 lymphocyte profile and low Treg:Th17 ratio in their eutopic endometrium during the secretory phase of their menstrual cycle are more likely to not conceive in subsequent cycles.
Collapse
Affiliation(s)
- Malavika K Adur
- Department of Animal Science, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Department of Animal Science, Iowa State University, Ames, IA, USA
| | - Andrea G Braundmeier-Fleming
- Department of Animal Science, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Department of Medical Microbiology, Immunology and Cell Biology, SIU School of Medicine, Springfield, IL, USA
- Department of Obstetrics and Gynecology, SIU School of Medicine, Springfield, IL, USA
| | - Bruce A Lessey
- Department of Obstetrics and Gynecology, Greenville Hospital System, Greenville, SC, USA
- Department of Obstetrics and Gynecology, Wake Forest Baptist Health, Winston-Salem, NC, USA
| | - Romana A Nowak
- Department of Animal Science, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| |
Collapse
|
15
|
Mayoral Andrade G, Vásquez Martínez G, Pérez-Campos Mayoral L, Hernández-Huerta MT, Zenteno E, Pérez-Campos Mayoral E, Martínez Cruz M, Martínez Cruz R, Matias-Cervantes CA, Meraz Cruz N, Romero Díaz C, Cruz-Parada E, Pérez-Campos E. Molecules and Prostaglandins Related to Embryo Tolerance. Front Immunol 2020; 11:555414. [PMID: 33329514 PMCID: PMC7710691 DOI: 10.3389/fimmu.2020.555414] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 10/19/2020] [Indexed: 12/20/2022] Open
Abstract
It is generally understood that the entry of semen into the female reproductive tract provokes molecular and cellular changes facilitating conception and pregnancy. We show a broader picture of the participation of prostaglandins in the fertilization, implantation and maintenance of the embryo. A large number of cells and molecules are related to signaling networks, which regulate tolerance to implantation and maintenance of the embryo and fetus. In this work, many of those cells and molecules are analyzed. We focus on platelets, polymorphonuclear leukocytes, and group 2 innate lymphoid cells involved in embryo tolerance in order to have a wider view of how prostaglandins participate. The combination of platelets and neutrophil extracellular traps (Nets), uterine innate lymphoid cells (uILC), Treg cells, NK cells, and sex hormones have an important function in immunological tolerance. In both animals and humans, the functions of these cells can be regulated by prostaglandins and soluble factors in seminal plasma to achieve an immunological balance, which maintains fetal-maternal tolerance. Prostaglandins, such as PGI2 and PGE2, play an important role in the suppression of the previously mentioned cells. PGI2 inhibits platelet aggregation, in addition to IL-5 and IL-13 expression in ILC2, and PGE2 inhibits some neutrophil functions, such as chemotaxis and migration processes, leukotriene B4 (LTB4) biosynthesis, ROS production, and the formation of extracellular traps, which could help prevent trophoblast injury and fetal loss. The implications are related to fertility in female when seminal fluid is deposited in the vagina or uterus.
Collapse
Affiliation(s)
- Gabriel Mayoral Andrade
- Research Centre Medicine National Autonomous University of Mexico-Benito Juárez Autonomous University of Oaxaca (UNAM-UABJO), Faculty of Medicine, Benito Juárez Autonomous University of Oaxaca, Oaxaca, Mexico
| | | | - Laura Pérez-Campos Mayoral
- Research Centre Medicine National Autonomous University of Mexico-Benito Juárez Autonomous University of Oaxaca (UNAM-UABJO), Faculty of Medicine, Benito Juárez Autonomous University of Oaxaca, Oaxaca, Mexico
| | | | - Edgar Zenteno
- Department of Biochemistry, School of Medicine, UNAM, Mexico City, México
| | - Eduardo Pérez-Campos Mayoral
- Research Centre Medicine National Autonomous University of Mexico-Benito Juárez Autonomous University of Oaxaca (UNAM-UABJO), Faculty of Medicine, Benito Juárez Autonomous University of Oaxaca, Oaxaca, Mexico
| | | | - Ruth Martínez Cruz
- Research Centre Medicine National Autonomous University of Mexico-Benito Juárez Autonomous University of Oaxaca (UNAM-UABJO), Faculty of Medicine, Benito Juárez Autonomous University of Oaxaca, Oaxaca, Mexico
| | | | - Noemi Meraz Cruz
- School of Medicine, Branch at National Institute of Genomic Medicine, Mexico City, Mexico
| | - Carlos Romero Díaz
- Research Centre Medicine National Autonomous University of Mexico-Benito Juárez Autonomous University of Oaxaca (UNAM-UABJO), Faculty of Medicine, Benito Juárez Autonomous University of Oaxaca, Oaxaca, Mexico
| | - Eli Cruz-Parada
- Biochemistry and Immunology Unit, National Technological of Mexico/ITOaxaca, Oaxaca, Mexico
| | - Eduardo Pérez-Campos
- Biochemistry and Immunology Unit, National Technological of Mexico/ITOaxaca, Oaxaca, Mexico
| |
Collapse
|
16
|
Naim N, Amrit FRG, McClendon TB, Yanowitz JL, Ghazi A. The molecular tug of war between immunity and fertility: Emergence of conserved signaling pathways and regulatory mechanisms. Bioessays 2020; 42:e2000103. [PMID: 33169418 DOI: 10.1002/bies.202000103] [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: 05/04/2020] [Revised: 09/11/2020] [Accepted: 09/17/2020] [Indexed: 12/21/2022]
Abstract
Reproduction and immunity are energy intensive, intimately linked processes in most organisms. In women, pregnancy is associated with widespread immunological adaptations that alter immunity to many diseases, whereas, immune dysfunction has emerged as a major cause for infertility in both men and women. Deciphering the molecular bases of this dynamic association is inherently challenging in mammals. This relationship has been traditionally studied in fast-living, invertebrate species, often in the context of resource allocation between life history traits. More recently, these studies have advanced our understanding of the mechanistic underpinnings of the immunity-fertility dialogue. Here, we review the molecular connections between reproduction and immunity from the perspective of human pregnancy to mechanistic discoveries in laboratory organisms. We focus particularly on recent invertebrate studies identifying conserved signaling pathways and transcription factors that regulate resource allocation and shape the balance between reproductive status and immune health.
Collapse
Affiliation(s)
- Nikki Naim
- Departments of Pediatrics, Developmental Biology and Cell Biology and Physiology, John, G. Rangos Sr. Research Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Francis R G Amrit
- Departments of Pediatrics, Developmental Biology and Cell Biology and Physiology, John, G. Rangos Sr. Research Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - T Brooke McClendon
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Judith L Yanowitz
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Arjumand Ghazi
- Departments of Pediatrics, Developmental Biology and Cell Biology and Physiology, John, G. Rangos Sr. Research Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
17
|
Bruno V, Lindau R, Jenmalm MC, Ticconi C, Piccione E, Pietropolli A, Ernerudh J. First-trimester trophoblasts obtained by chorionic villus sampling maintain tolerogenic and proteomic features in successful pregnancies despite a history of unexplained recurrent pregnancy loss. Am J Reprod Immunol 2020; 84:e13314. [PMID: 32734710 DOI: 10.1111/aji.13314] [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: 05/17/2020] [Revised: 07/03/2020] [Accepted: 07/21/2020] [Indexed: 01/06/2023] Open
Abstract
PROBLEM While there are several known causes for recurrent pregnancy loss (RPL), about 50% are unexplained (uRPL), and in these cases, an aberrant immune regulation seems to be involved. Although fetally derived trophoblast cells have a key role in immune regulation, it is difficult to study their immune function during pregnancy, and it is not known whether trophoblast function may be an inherent aberration in uRPL or whether it is associated with the outcome of the current pregnancy. METHOD OF STUDY Chorionic villus sampling (CVS) was performed for clinical indications at 12 weeks of gestation. Superfluous materials, divided in small explants, were cultured for 20-24 hours, and supernatants (conditioned medium) were collected from 36 women with singleton normal pregnancies, of whom 9 women had a history of RPL. The secreted immune protein profile was measured by proximity extension assay, and the conditioned medium was further used in functional ex vivo models to assess ability to polarize blood monocytes and CD4+ T cells into immune regulatory phenotypes, as detected by flow cytometry. RESULTS Conditioned medium from chorionic villi, human fetally derived placental tissue, was able to induce a decidual-type of M2-like macrophages, as well as an expansion of Treg cells ex vivo, both in women with uRPL and in control women. The preserved immunological properties were confirmed by a maintained immune protein profile in RPL compared with controls. CONCLUSION Trophoblasts in an ex vivo model maintain tolerogenic and proteomic profile features in successful pregnancies, despite a previous history of RPL.
Collapse
Affiliation(s)
- Valentina Bruno
- Section of Gynecology, Academic Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy.,Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden.,Academic Department of Surgical Sciences, Section of Gynecology, Tor Vergata University, Rome, Italy
| | - Robert Lindau
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Maria C Jenmalm
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Carlo Ticconi
- Academic Department of Surgical Sciences, Section of Gynecology, Tor Vergata University, Rome, Italy
| | - Emilio Piccione
- Academic Department of Surgical Sciences, Section of Gynecology, Tor Vergata University, Rome, Italy
| | - Adalgisa Pietropolli
- Academic Department of Surgical Sciences, Section of Gynecology, Tor Vergata University, Rome, Italy
| | - Jan Ernerudh
- Department of Clinical Immunology and Transfusion Medicine, Linköping University, Linköping, Sweden.,Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
18
|
Mitochondrial function in immune cells in health and disease. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165845. [PMID: 32473386 DOI: 10.1016/j.bbadis.2020.165845] [Citation(s) in RCA: 111] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 05/18/2020] [Accepted: 05/21/2020] [Indexed: 02/06/2023]
Abstract
One of the main functions of mitochondria is production of ATP for cellular energy needs, however, it becomes more recognized that mitochondria are involved in differentiation and activation processes of immune cells. Upon activation, immune cells have a high need for energy. Immune cells have different strategies to generate this energy. In pro-inflammatory cells, such as activated monocytes and activated T and B cells, the energy is generated by increasing glycolysis, while in regulatory cells, such as regulatory T cells or M2 macrophages, energy is generated by increasing mitochondrial function and beta-oxidation. Except for being important for energy supply during activation, mitochondria also induce immune responses. During an infection, they release mitochondrial danger associated molecules (DAMPs) that resemble structures of bacterial derived pathogen associated molecular patterns (PAMPs). Such mitochondrial DAMPS are for instance mitochondrial DNA with hypomethylated CpG motifs or a specific lipid that is only present in prokaryotic bacteria and mitochondria, i.e. cardiolipin. Via release of such DAMPs, mitochondria guide the immune response towards an inflammatory response against pathogens. This is an important mechanism in early detection of an infection and in stimulating and sustaining immune responses to fight infections. However, mitochondrial DAMPs may also have a negative impact. If mitochondrial DAMPs are released by damaged cells, without the presence of an infection, such as after a trauma, mitochondrial DAMPs may induce an undesired inflammatory response, resulting in tissue damage and organ dysfunction. Thus, immune cells have developed mechanisms to prevent such undesired immune activation by mitochondrial components. In the present narrative review, we will describe the current view of mitochondria in regulation of immune responses. We will also discuss the current knowledge on disturbed mitochondrial function in immune cells in various immunological diseases.
Collapse
|
19
|
Major Variation in the Incidence of Appendicitis Before, During and After Pregnancy: A Population-Based Cohort Study. World J Surg 2020; 44:2601-2608. [PMID: 32328784 DOI: 10.1007/s00268-020-05524-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Previous studies indicate a low incidence of appendicitis in third-trimester pregnancy, suggesting a protecting effect of pregnancy. This large population-based cohort study analyzes the association of appendicitis with pregnancy in more detail. The aim of the study was to investigate the incidence of appendicitis and negative appendectomy before, during and after pregnancy. METHODS Cross-linking between two Swedish health registries provided data on appendectomy for all women in Sweden giving birth between 1973 and 2013. We analyzed the incidence rates (IR) of perforated and non-perforated appendicitis and negative appendectomy before, during and after pregnancy, and secular trends during the study period. Standardized incidence ratios (SIR) were estimated using age-, sex- and period-specific IR from the background population in Sweden. RESULTS Some 3,888,452 pregnancies resulted in birth during the study period. An appendectomy was registered for 27,575 women in the interval starting one year before and ending two years after pregnancy. The incidence of appendicitis varied substantially during and after pregnancy. SIR for perforated appendicitis was 0.47 (95% CI 0.38-0.59) in the third trimester, 3.89 (2.92-5.18) peripartum, 2.20 (1.89-2.55) in the puerperium and 1.27 (1.19-1.36) in the year postpartum. The pattern was similar for non-perforated appendicitis. Negative appendectomy decreased postpartum. Incidence rate of non-perforated appendicitis and negative appendectomy decreased for both pregnant and non-pregnant women during the study period. CONCLUSIONS The findings in this study suggest a protecting effect of pregnancy on the development of appendicitis, which is followed by a rebound effect after birth.
Collapse
|
20
|
Liu N, Chen J, He Y, Jia H, Jiang D, Li S, Yang Y, Dai Z, Wu Z, Wu G. Effects of maternal L-proline supplementation on inflammatory cytokines at the placenta and fetus interface of mice. Amino Acids 2020; 52:587-596. [PMID: 32170468 DOI: 10.1007/s00726-020-02837-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 03/06/2020] [Indexed: 12/12/2022]
Abstract
Dietary L-proline (proline) supplementation during gestation enhances fetal survival and placental development in mice. The objective of the present study was to test the hypothesis that this beneficial effect of proline was associated with alterations in inflammatory response at the placenta and fetus interface. Populations of immune cells present in peripheral blood mononuclear cells (PBMC) were determined by flow cytometry analysis. The concentrations of immunoglobulins in plasma, and the concentrations of cytokines in plasma, uterus, placenta, and amniotic fluid were measured using a bead-based immunoassay. The data showed that proline supplementation led to higher (P < 0.05) populations of B lymphocytes (CD3-CD19+), natural killer (NK) cells (CD3-NK1.1+), and dendritic cells (DCs, CD11c+MHCII+) in peripheral blood, as compared with the controls. Conversely, mice fed a proline-supplemented diet had a lower population of neutrophils (CD11b+F4/80-). Further study showed that proline supplementation decreased (P < 0.05) the concentrations of (1) interleukin (IL)-23, IL-1α, and IL-6 in plasma; (2) IL-6 in the uterus; and (3) tumor necrosis factor alpha (TNF-α), monocyte chemotactic protein (MCP)-1, and IL-17 in the placenta; and (4) interferon (IFN)-γ in amniotic fluid, compared with controls. Conversely, proline supplementation resulted in higher (P < 0.05) concentrations of (1) IL-10, IL-17 and granulocyte-macrophage colony-stimulating factor (GM-CSF) in plasma; (2) IL-10 and IL-1α in the uterus; and (3) IL-1α, IL-1β, IL-10, IL-27, and IFN-β in amniotic fluid, compared with controls. Moreover, concentrations of immunoglobulin (Ig) G2b and IgM were enhanced (P < 0.05) by proline administration. Taken together, our results reveal a regulatory effect of proline in the immunological response at the maternal-fetal interface, which is critical for embryonic development and fetal survival.
Collapse
Affiliation(s)
- Ning Liu
- Department of Animal Nutrition and Feed Science, State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, 100193, China
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Jingqing Chen
- Department of Animal Nutrition and Feed Science, State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, 100193, China
| | - Yu He
- Department of Animal Nutrition and Feed Science, State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, 100193, China
| | - Hai Jia
- Department of Animal Nutrition and Feed Science, State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, 100193, China
| | - Da Jiang
- Department of Animal Nutrition and Feed Science, State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, 100193, China
| | - Shuai Li
- Department of Animal Nutrition and Feed Science, State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, 100193, China
| | - Ying Yang
- Department of Animal Nutrition and Feed Science, State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, 100193, China
| | - Zhaolai Dai
- Department of Animal Nutrition and Feed Science, State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, 100193, China
| | - Zhenlong Wu
- Department of Animal Nutrition and Feed Science, State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, 100193, China.
| | - Guoyao Wu
- Department of Animal Nutrition and Feed Science, State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, 100193, China.
- Department of Animal Science, Texas A&M University, College Station, TX, 77843, USA.
| |
Collapse
|
21
|
Bruno V, Corrado G, Baci D, Chiofalo B, Carosi MA, Ronchetti L, Piccione E, Albini A, Noonan DM, Piaggio G, Vizza E. Endometrial Cancer Immune Escape Mechanisms: Let Us Learn From the Fetal-Maternal Interface. Front Oncol 2020; 10:156. [PMID: 32226771 PMCID: PMC7080858 DOI: 10.3389/fonc.2020.00156] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 01/29/2020] [Indexed: 12/14/2022] Open
Abstract
The immune escape mechanisms at the base of tumor progression in endometrial cancer mimic immune tolerance mechanisms occurring at the maternal-fetal interface. The biological and immunological processes behind the maternal-fetal interface are finely tuned in time and space during embryo implantation and subsequent pregnancy stages; conversely, those behind cancer progression are often aberrant. The environment composition at the maternal-fetal interface parallels the pro-tumor microenvironment identified in many cancers, pointing to the possibility for the use of the maternal-fetal interface as a model to depict immune therapeutic targets in cancer. The framework of cancer environment signatures involved in immune adaptations, precisely timed in cancer progression, could reveal a specific "immune clock" in endometrial cancer, which might guide clinicians in patient risk class assessment, diagnostic workup, management, surgical and therapeutic approach, and surveillance strategies. Here, we review studies approaching this hypothesis, focusing on what is known so far about oncofetal similarities in immunity with the idea to individualize personalized immunotherapy targets, through the downregulation of the immune escape stage or the reactivation of the pro-inflammatory processes suppressed by the tumor.
Collapse
Affiliation(s)
- Valentina Bruno
- Gynecologic Oncology Unit, Department of Experimental Clinical Oncology, IRCCS-Regina Elena National Cancer Institute, Rome, Italy
| | - Giacomo Corrado
- Gynecologic Oncology Unit, Department of Women and Children Health, Fondazione Policlinico Universitario A. Gemelli, IRCCS-Università Cattolica del Sacro Cuore, Rome, Italy
| | - Denisa Baci
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - Benito Chiofalo
- Gynecologic Oncology Unit, Department of Experimental Clinical Oncology, IRCCS-Regina Elena National Cancer Institute, Rome, Italy
| | - Maria Antonia Carosi
- Anatomy Pathology Unit, Department of Research, Diagnosis and Innovative Technologies, IRCCS-Regina Elena National Cancer Institute, Rome, Italy
| | - Livia Ronchetti
- Anatomy Pathology Unit, Department of Research, Diagnosis and Innovative Technologies, IRCCS-Regina Elena National Cancer Institute, Rome, Italy
| | - Emilio Piccione
- Section of Gynecology, Academic Department of Surgical Sciences, Tor Vergata University Hospital, University of Rome "Tor Vergata", Rome, Italy
| | - Adriana Albini
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy.,Vascular Biology and Angiogenesis Laboratory, Science and Technology Pole (PST), IRCCS MultiMedica, Milan, Italy
| | - Douglas M Noonan
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy.,Vascular Biology and Angiogenesis Laboratory, Science and Technology Pole (PST), IRCCS MultiMedica, Milan, Italy
| | - Giulia Piaggio
- Department of Research, Diagnosis and Innovative Technologies, UOSD SAFU, IRCCS-Regina Elena National Cancer Institute, Rome, Italy
| | - Enrico Vizza
- Gynecologic Oncology Unit, Department of Experimental Clinical Oncology, IRCCS-Regina Elena National Cancer Institute, Rome, Italy
| |
Collapse
|
22
|
Xu L, Yu Y, Sang R, Ge B, Wang M, Zhou H, Zhang X. Inonotus obliquus polysaccharide protects against adverse pregnancy caused by Toxoplasma gondii infection through regulating Th17/Treg balance via TLR4/NF-κB pathway. Int J Biol Macromol 2020; 146:832-840. [DOI: 10.1016/j.ijbiomac.2019.10.051] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 09/25/2019] [Accepted: 10/03/2019] [Indexed: 02/07/2023]
|
23
|
Ali-Hassanzadeh M, Hosseini MS, Ahmadi M, Zare M, Akbarzadeh-Jahromi M, Derakhshanfar A, Gharesi-Fard B. Analysis of the frequency of type 2 innate lymphoid cells and regulatory T cells in abortion-prone mice. Immunol Lett 2020; 220:1-10. [PMID: 31945403 DOI: 10.1016/j.imlet.2020.01.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 01/06/2020] [Accepted: 01/10/2020] [Indexed: 02/07/2023]
Abstract
Recurrent spontaneous abortion (RSA) is the most common pregnancy related complication, affecting 1-5 % of pregnancies. Despite hormonal, genetic and anatomical factors that result in abortion, impairment of immune response at the feto-maternal interface during the first trimester of pregnancy is also one of the main causes of RSA. In the present study, we evaluated the frequency of blood and uterine group 2 innate lymphoid cells (ILC2s), their subsets and regulatory T cells (Tregs) in CBA/J × DBA/2 J as an abortion-prone model compared to normal pregnant (NP) mice using immunophenotyping. Results indicated that the percentages of ILC2s were significantly decreased in the AP group compared to the NP group at mid-gestation (P ≤ 0.01). Moreover, the percentages of both blood and uterine nILC2s were increased in NP mice at mid-gestation (P ≤ 0.01, and P ≤ 0.05, respectively), while iILC2s significantly increased in AP mice at mid-gestation (P ≤ 0.01, and P ≤ 0.05, respectively). Tregs were reduced in AP mice at both early and mid-gestation stages (P ≤ 0.01). Overall, our findings suggest that the changes in blood and uterine ILC2s might be associated with abortion in mice.
Collapse
Affiliation(s)
- Mohammad Ali-Hassanzadeh
- Department of Immunology, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Immunology, School of Medicine, Jiroft University of Medical Sciences, Jiroft, Iran
| | | | - Moslem Ahmadi
- Department of Immunology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maryam Zare
- Department of Immunology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mojgan Akbarzadeh-Jahromi
- Maternal-Fetal Medicine Research Center, Pathology Department, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amin Derakhshanfar
- DiagnosticLaboratory Sciences and Technology Research Center, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran; Center of Comparative and Experimental Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Behrouz Gharesi-Fard
- Department of Immunology, Shiraz University of Medical Sciences, Shiraz, Iran; Infertility Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
24
|
Faas MM, Liu Y, Borghuis T, van Loo-Bouwman CA, Harmsen H, de Vos P. Microbiota Induced Changes in the Immune Response in Pregnant Mice. Front Immunol 2020; 10:2976. [PMID: 31998293 PMCID: PMC6962187 DOI: 10.3389/fimmu.2019.02976] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 12/04/2019] [Indexed: 01/14/2023] Open
Abstract
Pregnancy is associated with adaptations of the immune response and with changes in the gutmicrobiota. We hypothesized the gut microbiota are involved in inducing (part of) the immunological adaptations during pregnancy. To test this hypothesis, we collected feces from pregnant conventional mice before and during pregnancy (days 7, 14, and 18) and microbiota were measured using 16S RNA sequencing. At day 18, mice were sacrificed and splenic (various Th cell populations) and blood immune cells (monocyte subsets) were measured by flow cytometry. The data were compared with splenic and blood immune cell populations from pregnant (day 18) germfree mice and non-pregnant conventional and germfree mice. Finally, the abundances of the individual gut bacteria in the microbiota of each conventional pregnant mouse were correlated to the parameters of the immune response of the same mouse. The microbiota of conventional mice were significantly different at the end of pregnancy (day 18) as compared with pre-pregnancy (Permanova, p < 0.05). The Shannon index was decreased and the Firmicutes/Bacteroidetes ratio was increased (Friedman followed by Dunn's test, p < 0.05), while abundances of various species (such as Allobaculum stercoricanis, Barnesiella intestihominis, and Roseburia faecis) were significantly different at day 18 compared with pre-pregnancy. In pregnant conventional mice, the percentage of Th1 cells was decreased, while the percentages of Treg cells and Th2 cells were or tended to be increased vs. non-pregnant mice. In germfree mice, only the percentage of Th1 cells was decreased in pregnant vs. non-pregnant mice, with no effect of pregnancy on Treg and Th2 cells. The percentages of monocyte subsets were affected by pregnancy similarly in conventional and germfree mice. However, the activation status of monocytes (expression of CD80 and MHCII) was affected by pregnancy mainly in conventional mice, and not in germfree mice. Correlation (Spearman's coefficient) of pregnancy affected microbiota with pregnancy affected immune cells, i.e., immune cells that were only affected differently in conventional mice and germfree mice, showed 4 clusters of bacteria and 4 clusters of immune cells, some of these clusters were correlated with each other. For instance, the microbiota in cluster 1 and 2 (in which there were various short chain fatty acid producing microbiota) are positively correlated with immune cells in cluster B, containing Treg cells and Th2 cells. Microbiota and immune cells are affected by pregnancy in mice. The different immunological adaptations to pregnancy between conventional and germfree mice, such as the increase in Treg and tendency to an increase in Th2 cells in conventional pregnant mice only, may suggest that the microbiota may play a role in adapting the maternal immune response to pregnancy.
Collapse
Affiliation(s)
- Marijke M Faas
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Yuanrui Liu
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Theo Borghuis
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | | | - Hermie Harmsen
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Paul de Vos
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
25
|
Rasmark Roepke E, Bruno V, Nedstrand E, Boij R, Strid CP, Piccione E, Berg G, Svensson-Arvelund J, Jenmalm MC, Rubér M, Ernerudh J. Low-molecular-weight-heparin increases Th1- and Th17-associated chemokine levels during pregnancy in women with unexplained recurrent pregnancy loss: a randomised controlled trial. Sci Rep 2019; 9:12314. [PMID: 31444404 PMCID: PMC6707182 DOI: 10.1038/s41598-019-48799-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 08/13/2019] [Indexed: 11/29/2022] Open
Abstract
Low-molecular-weight heparin (LMWH) is widely used to treat recurrent pregnancy loss (RPL) because of its anti-coagulant effects. Although in vitro studies have suggested additional immunological effects, these are debated. We therefore investigated whether LMWH could modulate immune responses in vivo during pregnancy of women with unexplained RPL. A Swedish open multi-centre randomised controlled trial included 45 women treated with tinzaparin and 42 untreated women. Longitudinally collected plasma samples were obtained at gestational weeks (gw) 6, 18, 28 and 34 and analysed by multiplex bead technology for levels of 11 cytokines and chemokines, chosen to represent inflammation and T-helper subset-associated immunity. Mixed linear models test on LMWH-treated and untreated women showed differences during pregnancy of the Th1-associated chemokines CXCL10 (p = 0.01), CXCL11 (p < 0.001) and the Th17-associated chemokine CCL20 (p = 0.04), while CCL2, CCL17, CCL22, CXCL1, CXCL8, CXCL12, CXCL13 and IL-6 did not differ. Subsequent Student's t-test showed significantly higher plasma levels of CXCL10 and CXCL11 in treated than untreated women at gw 28 and 34. The consistent increase in the two Th1-associated chemokines suggests a potential proinflammatory and unfavourable effect of LMWH treatment during later stages of pregnancy, when Th1 immunity is known to disrupt immunological tolerance.
Collapse
Affiliation(s)
- E Rasmark Roepke
- Department of Obstetrics and Gynecology, Skåne University Hospital, Malmö and Lund University, Lund, Sweden.
| | - V Bruno
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
- Section of Gynecology and Obstetrics, Academic Department of Biomedicine and Prevention, and Clinical Department of Surgery, Tor Vergata University Hospital, Rome, Italy
| | - E Nedstrand
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - R Boij
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - C Petersson Strid
- Departmen of Obstetrics and Gynecology, Kalmar Hospital, Kalmar, Sweden
| | - E Piccione
- Section of Gynecology and Obstetrics, Academic Department of Biomedicine and Prevention, and Clinical Department of Surgery, Tor Vergata University Hospital, Rome, Italy
| | - G Berg
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - J Svensson-Arvelund
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - M C Jenmalm
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - M Rubér
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - J Ernerudh
- Department of Clinical Immunology and Transfusion Medicine, and Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| |
Collapse
|
26
|
Li Y, Lopez GE, Vazquez J, Sun Y, Chavarria M, Lindner PN, Fredrickson S, Karst N, Stanic AK. Decidual-Placental Immune Landscape During Syngeneic Murine Pregnancy. Front Immunol 2018; 9:2087. [PMID: 30283441 PMCID: PMC6156255 DOI: 10.3389/fimmu.2018.02087] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 08/23/2018] [Indexed: 01/08/2023] Open
Abstract
Adaptive immune system, principally governed by the T cells-dendritic cells (DCs) nexus, is an essential mediator of gestational fetal tolerance and protection against infection. However, the exact composition and dynamics of DCs and T cell subsets in gestational tissues are not well understood. These are controlled in human physiology by a complex interplay of alloantigen distribution and presentation, cellular/humoral active and passive tolerance, hormones/chemokines/angiogenic factors and their gradients, systemic and local microbial communities. Reductive discrimination of these factors in physiology and pathology of model systems and humans requires simplification of the model and increased resolution of interrogative technologies. As a baseline, we have studied the gestational tissue dynamics in the syngeneic C57BL/6 mice, as the simplest immunological environment, and focused on validating the approach to increased data density and computational analysis pipeline afforded by highly polychromatic flow cytometry and machine learning interpretation. We mapped DC and T cell subsets, and comprehensively examined their maternal (decidual)-fetal (placental) interface dynamics. Both frequency and composition of decidual DCs changed across gestation, with a dramatic increase in myeloid DCs in early pregnancy, and exclusion of plasmacytoid DCs. CD4+ T cells, in contrast, were lower at all gestational ages and an unusual CD4-CD8-TCRαβ+group was prominent at mid-pregnancy. Dimensionality reduction with machine learning-aided clustering revealed that CD4-CD8- T cells were phenotypically different from CD4+ and CD8+ T cells. Additionally, divergence between maternal decidual and fetal placental compartment was prominent, with absence of DCs from the placenta, but not decidua or embryo. These results provide a novel framework and a syngeneic baseline on which the specific role of alloantigen/tolerance, polymicrobial environment, and models of pregnancy pathology can be precisely modeled and analyzed.
Collapse
Affiliation(s)
- Yan Li
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI, United States
| | - Gladys E. Lopez
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI, United States
| | - Jessica Vazquez
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI, United States
| | - Yan Sun
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI, United States
- Reproductive Medicine Center, Fujian Provincial Maternity and Children's Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Melina Chavarria
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI, United States
| | - Payton N. Lindner
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI, United States
| | - Samantha Fredrickson
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI, United States
| | - Nathan Karst
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI, United States
| | - Aleksandar K. Stanic
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI, United States
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
27
|
Hauk V, Vota D, Gallino L, Calo G, Paparini D, Merech F, Ochoa F, Zotta E, Ramhorst R, Waschek J, Leirós CP. Trophoblast VIP deficiency entails immune homeostasis loss and adverse pregnancy outcome in mice. FASEB J 2018; 33:1801-1810. [PMID: 30204500 DOI: 10.1096/fj.201800592rr] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Immune homeostasis maintenance throughout pregnancy is critical for normal fetal development. Trophoblast cells differentiate into an invasive phenotype and contribute to the transformation of maternal arteries and the functional shaping of decidual leukocyte populations. Insufficient trophoblast invasion, inadequate vascular remodeling, and a loss of immunologic homeostasis are associated with pregnancy complications, such as preeclampsia and intrauterine growth restriction. Vasoactive intestinal peptide (VIP) is a pleiotropic neuropeptide synthetized in trophoblasts at the maternal-placental interface. It regulates the function of trophoblast cells and their interaction with decidual leukocytes. By means of a murine model of pregnancy in normal maternal background with VIP-deficient trophoblast cells, here we demonstrate that trophoblast VIP is critical for trophoblast function: VIP gene haploinsufficiency results in lower matrix metalloproteinase 9 expression, and reduced migration and invasion capacities. A reduced number of regulatory T cells at the implantation sites along with a lower expression of proangiogenic and antiinflammatory markers were also observed. Findings detected in the implantation sites at early stages were followed by an abnormal placental structure and lower fetal weight. This effect was overcome by VIP treatment of the early pregnant mice. Our results support the relevance of trophoblast-synthesized VIP as a critical factor in vivo for trophoblast-cell function and immune homeostasis maintenance in mouse pregnancy.-Hauk, V., Vota, D., Gallino, L., Calo, G., Paparini, D., Merech, F., Ochoa, F., Zotta, E., Ramhorst, R., Waschek, J., Leirós, C. P. Trophoblast VIP deficiency entails immune homeostasis loss and adverse pregnancy outcome in mice.
Collapse
Affiliation(s)
- Vanesa Hauk
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN)
| | - Daiana Vota
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN)
| | - Lucila Gallino
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN)
| | - Guillermina Calo
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN)
| | - Daniel Paparini
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN)
| | - Fátima Merech
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN)
| | - Federico Ochoa
- Departamento de Ciencias Fisiológicas, Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO-Houssay), Universidad de Buenos Aires, Facultad de Medicina, Buenos Aires, Argentina
| | - Elsa Zotta
- Departamento de Ciencias Fisiológicas, Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO-Houssay), Universidad de Buenos Aires, Facultad de Medicina, Buenos Aires, Argentina.,Catedra de Fisiopatología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Rosanna Ramhorst
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN)
| | - James Waschek
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, The David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Claudia Pérez Leirós
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN)
| |
Collapse
|
28
|
An Y, Gao S, Zhao WC, Qiu BA, Xia NX, Zhang PJ, Fan ZP. Transforming growth factor-β and peripheral regulatory cells are negatively correlated with the overall survival of hepatocellular carcinoma. World J Gastroenterol 2018; 24:2733-2740. [PMID: 29991878 PMCID: PMC6034152 DOI: 10.3748/wjg.v24.i25.2733] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 04/14/2018] [Accepted: 06/01/2018] [Indexed: 02/06/2023] Open
Abstract
AIM To understand the cellular and molecular changes in peripheral blood that can lead to the development of hepatocellular carcinoma (HCC) and provide new methods for its diagnosis and treatment.
METHODS Peripheral blood mononuclear cells were isolated from the peripheral blood of HCC patients and normal controls and then analyzed by flow cytometry. The percentage of transforming growth factor-β (TGF-β)+ regulatory cells (Tregs) in the peripheral blood was measured, and the expression of TGF-β was also determined. Then, the relationship between the changes and the 5-year survival of patients was analyzed. In addition, recombinant human TGF-β (rhTGF-β) and recombinant human interleukin-6 were added to stimulate the cultured cells, and their effects on HCC were evaluated.
RESULTS The expression of TGF-β and the percentage of TGF-β+ Tregs in the peripheral blood of HCC patients increased significantly compared with normal controls. Compared with the low TGF-β expression group, the high TGF-β expression group had a significantly lower 5-year survival rate, and the same result was found in the two TGF-β+ Treg groups, suggesting that TGF-β and TGF-β+ Tregs were negatively correlated with the overall survival of the patients. In addition, rhTGF-β promoted the growth of tumor cells and induced high expression levels of IL-6, which further promoted tumor proliferation.
CONCLUSION The results showed that TGF-β may promote tumor growth and proliferation by inducing the production of IL-6, and TGF-β and TGF-β+ Tregs may serve as new markers for predicting a poor prognosis in HCC.
Collapse
Affiliation(s)
- Yang An
- Department of Hepato-Biliary-Pancreatic Surgery, Navy General Hospital of Chinese People’s Liberation Army, Beijing 100048, China
| | - Song Gao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Interventional Therapy, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Wen-Chao Zhao
- Department of Hepato-Biliary-Pancreatic Surgery, Navy General Hospital of Chinese People’s Liberation Army, Beijing 100048, China
| | - Bao-An Qiu
- Department of Hepato-Biliary-Pancreatic Surgery, Navy General Hospital of Chinese People’s Liberation Army, Beijing 100048, China
| | - Nian-Xin Xia
- Department of Hepato-Biliary-Pancreatic Surgery, Navy General Hospital of Chinese People’s Liberation Army, Beijing 100048, China
| | - Peng-Jun Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Interventional Therapy, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Zhen-Ping Fan
- Liver Disease Center for Cadre Medical Care, Beijing 302 Military Hospital, Beijing 100039, China
| |
Collapse
|
29
|
Hosseini A, Dolati S, Hashemi V, Abdollahpour‐Alitappeh M, Yousefi M. Regulatory T and T helper 17 cells: Their roles in preeclampsia. J Cell Physiol 2018; 233:6561-6573. [DOI: 10.1002/jcp.26604] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 03/16/2018] [Indexed: 12/27/2022]
Affiliation(s)
- Arezoo Hosseini
- Stem Cell Research CenterTabriz University of Medical SciencesTabrizIran
- Drug Applied Research CenterTabriz University of Medical SciencesTabrizIran
- Department of ImmunologyFaculty of MedicineTabriz University of Medical SciencesTabrizIran
- Student's Research CommitteeTabriz University of Medical SciencesTabrizIran
| | - Sanam Dolati
- Stem Cell Research CenterTabriz University of Medical SciencesTabrizIran
- Drug Applied Research CenterTabriz University of Medical SciencesTabrizIran
- Department of ImmunologyFaculty of MedicineTabriz University of Medical SciencesTabrizIran
- Student's Research CommitteeTabriz University of Medical SciencesTabrizIran
| | - Vida Hashemi
- Department of Basic ScienceFaculty of MedicineMaragheh University of Medical SciencesMaraghehIran
| | - Meghdad Abdollahpour‐Alitappeh
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver DiseasesShahid Beheshti University of Medical SciencesTehranIran
| | - Mehdi Yousefi
- Drug Applied Research CenterTabriz University of Medical SciencesTabrizIran
- Department of ImmunologyFaculty of MedicineTabriz University of Medical SciencesTabrizIran
| |
Collapse
|
30
|
Effects of low molecular weight heparin on the polarization and cytokine profile of macrophages and T helper cells in vitro. Sci Rep 2018. [PMID: 29520033 PMCID: PMC5843640 DOI: 10.1038/s41598-018-22418-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Low molecular weight heparin (LMWH) is widely used in recurrent miscarriage treatment. The anti-coagulant effects are established, while immunological effects are not fully known. Our aim was to assess LMWH effects on activation and polarization of central regulatory immune cells from healthy women, and on placenta tissues from women undergoing elective abortions. Isolated blood monocytes and T helper (Th) cells under different activation and polarizing conditions were cultured with or without LMWH. Flow cytometry showed that LMWH exposure induced increased expression of HLA-DR and CD206 in macrophages. This phenotype was associated with increased secretion of Th17-associated CCL20, and decreased secretion of CCL2 (M2-associated) and CCL22 (Th2), as measured by multiplex bead array. In accordance, LMWH exposure to Th cells reduced the proportion of CD25highFoxp3+ regulatory T-cells, intensified IFN-γ secretion and showed a tendency to increase the lymphoblast proportions. Collectively, a mainly pro-inflammatory effect was noted on two essential tolerance-promoting cells. Although the biological significancies of these in vitro findings are uncertain and need to be confirmed in vivo, they suggest the possibility that immunological effects of LMWH may be beneficial mainly at an earlier gestational age to provide an appropriate implantation process in women with recurrent miscarriage.
Collapse
|
31
|
Kempe P, Eklund D, Hallin A, Hammar M, Olsson T, Brynhildsen J, Ernerudh J. Immune profile in relation to sex steroid cyclicity in healthy women and women with multiple sclerosis. J Reprod Immunol 2018; 126:53-59. [PMID: 29501895 DOI: 10.1016/j.jri.2018.02.006] [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: 08/26/2017] [Revised: 02/11/2018] [Accepted: 02/24/2018] [Indexed: 10/17/2022]
Abstract
To prospectively study systemic in vivo immunological effects of sex hormones, using different phases of oral combined hormonal contraceptives (CHC), and the natural menstrual cycles in both healthy women and in women with multiple sclerosis (MS), blood samples from sixty female MS patients and healthy controls with and without CHC were drawn in high and low estrogenic/progestogenic phases. Expression of Th-associated genes in blood cells was determined by qPCR and a panel of cytokines and chemokines was measured in plasma. High hormone level phases were associated with increases in Th1 (TBX21) and Th2 (GATA3) associated markers, as well as the B cell-associated chemokine CXCL13, while the inhibitory regulator CTLA-4 was decreased. These changes were not observed in MS patients, of whom most were treated with immunomodulatory drugs. Our data indicate immune activating properties in vivo of high steroid sex hormone levels during both CHC and normal menstrual cyclicity.
Collapse
Affiliation(s)
- Per Kempe
- Obstetrics and Gynaecology, Department of Clinical and Experimental Medicine, Linköping University, SE-58185, Linköping, Sweden; Department of Obstetrics & Gynaecology, County Hospital Sundsvall, SE-85643, Sundsvall, Sweden
| | - Daniel Eklund
- Department of Clinical and Experimental Medicine, Linköping University, SE-58185, Linköping, Sweden
| | - Agnes Hallin
- Department of Clinical and Experimental Medicine, Linköping University, SE-58185, Linköping, Sweden
| | - Mats Hammar
- Obstetrics and Gynaecology, Department of Clinical and Experimental Medicine, Linköping University, SE-58185, Linköping, Sweden
| | - Tomas Olsson
- Department of Clinical Neuroscience, Karolinska Institutet, SE-17177, Stockholm, Sweden
| | - Jan Brynhildsen
- Obstetrics and Gynaecology, Department of Clinical and Experimental Medicine, Linköping University, SE-58185, Linköping, Sweden.
| | - Jan Ernerudh
- Clinical Immunology and Transfusion Medicine, Department of Clinical and Experimental Medicine, Linköping University, SE-58185, Linköping, Sweden
| |
Collapse
|
32
|
Varese A, Remes Lenicov F, Gonzalez Prinz M, Paletta A, Ernst G, Maeto C, Merlotti A, Sabatte J, Símula S, Holgado MP, Dantas E, Geffner J, Ceballos A. Seminal vesicle fluid increases the efficacy of intravaginal HSV-2 vaccination. Mucosal Immunol 2018; 11:536-548. [PMID: 28745327 DOI: 10.1038/mi.2017.57] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 05/16/2017] [Indexed: 02/04/2023]
Abstract
Once considered merely as a vehicle for spermatozoa, it is now clear that seminal plasma (SP) induces a variety of biological actions on the female reproductive tissues able to modulate the immune response against paternal antigens. To our knowledge, the influence of SP on the immune response against sexually transmitted pathogens has not been yet evaluated. We here analyzed whether the seminal vesicle fluid (SVF), which contributes almost 60% of the SP volume in mice, could modulate the immune response against herpes simplex virus type 2 (HSV-2). We found that SVF does not modify the course of primary infection, but markedly improved protection conferred by vaginal vaccination with inactivated HSV-2 against a lethal challenge. This protective effect was shown to be associated to a robust memory immune response mediated by CD4+ and CD8+ T cells in both the lymph nodes draining the vagina and the vaginal mucosa, the site of viral replication. In contrast with the widespread notion that SP acts as an immunosuppressive agent, our results suggest that SVF might improve the female immune response against sexually transmitted pathogens.
Collapse
Affiliation(s)
- A Varese
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Universidad de Buenos Aires (UBA) and Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - F Remes Lenicov
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Universidad de Buenos Aires (UBA) and Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - M Gonzalez Prinz
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Universidad de Buenos Aires (UBA) and Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - A Paletta
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Universidad de Buenos Aires (UBA) and Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - G Ernst
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Universidad de Buenos Aires (UBA) and Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - C Maeto
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Universidad de Buenos Aires (UBA) and Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - A Merlotti
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Universidad de Buenos Aires (UBA) and Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - J Sabatte
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Universidad de Buenos Aires (UBA) and Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - S Símula
- Instituto de Microbiología y Parasitología Médica (IMPaM), UBA-CONICET, Buenos Aires, Argentina
| | - M P Holgado
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Universidad de Buenos Aires (UBA) and Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - E Dantas
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Universidad de Buenos Aires (UBA) and Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - J Geffner
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Universidad de Buenos Aires (UBA) and Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - A Ceballos
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Universidad de Buenos Aires (UBA) and Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| |
Collapse
|
33
|
Abstract
Preeclampsia is characterized by blood pressure greater than 140/90 mmHg in the second half of pregnancy. This disease is a major contributor to preterm and low birth weight babies. The early delivery of the baby, which becomes necessary for maintaining maternal well-being, makes preeclampsia the leading cause for preterm labor and infant mortality and morbidity. Currently, there is no cure for this pregnancy disorder. The current clinical management of PE is hydralazine with labetalol and magnesium sulfate to slow disease progression and prevent maternal seizure, and hopefully prolong the pregnancy. This review will highlight factors implicated in the pathophysiology of preeclampsia and current treatments for the management of this disease.
Collapse
|
34
|
Washington K, Ghosh S, Reeves IV. A Review: Molecular Concepts and Common Pathways Involving Vitamin D in the Pathophysiology of Preeclampsia. ACTA ACUST UNITED AC 2018. [DOI: 10.4236/ojog.2018.83023] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
35
|
Lymphocytes in Placental Tissues: Immune Regulation and Translational Possibilities for Immunotherapy. Stem Cells Int 2017; 2017:5738371. [PMID: 29348758 PMCID: PMC5733952 DOI: 10.1155/2017/5738371] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 10/11/2017] [Indexed: 02/07/2023] Open
Abstract
Immune modulation at the fetomaternal interface is crucial to ensure that the fetal allograft is not rejected. In the present review, the focus is to describe basic functions of lymphocyte populations and how they may contribute to fetomaternal immune regulation, as well as determining what proportions and effector functions of these cells are reported to be present in placental tissues in humans. Also explored is the possibility that unique cell populations at the fetomaternal interface may be targets for adoptive cell therapy. Increasing the understanding of immune modulation during pregnancy can give valuable insight into other established fields such as allogeneic hematopoietic stem cell transplantation and solid organ transplantation. In these settings, lymphocytes are key components that contribute to inflammation and rejection of either patient or donor tissues following transplantation. In contrast, an allogeneic fetus eludes rejection by the maternal immune system.
Collapse
|
36
|
The pathogenesis of microcephaly resulting from congenital infections: why is my baby’s head so small? Eur J Clin Microbiol Infect Dis 2017; 37:209-226. [DOI: 10.1007/s10096-017-3111-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 09/17/2017] [Indexed: 02/07/2023]
|
37
|
Shah NM, Herasimtschuk AA, Boasso A, Benlahrech A, Fuchs D, Imami N, Johnson MR. Changes in T Cell and Dendritic Cell Phenotype from Mid to Late Pregnancy Are Indicative of a Shift from Immune Tolerance to Immune Activation. Front Immunol 2017; 8:1138. [PMID: 28966619 PMCID: PMC5605754 DOI: 10.3389/fimmu.2017.01138] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Accepted: 08/29/2017] [Indexed: 12/12/2022] Open
Abstract
During pregnancy, the mother allows the immunologically distinct fetoplacental unit to develop and grow. Opinions are divided as to whether this represents a state of fetal-specific tolerance or of a generalized suppression of the maternal immune system. We hypothesized that antigen-specific T cell responses are modulated by an inhibitory T cell phenotype and modified dendritic cell (DC) phenotype in a gestation-dependent manner. We analyzed changes in surface markers of peripheral blood T cells, ex vivo antigen-specific T cell responses, indoleamine 2,3-dioxygenase (IDO) activity (kynurenine/tryptophan ratio, KTR), plasma neopterin concentration, and the in vitro expression of progesterone-induced blocking factor (PIBF) in response to peripheral blood mononuclear cell culture with progesterone. We found that mid gestation is characterized by reduced antigen-specific T cell responses associated with (1) predominance of effector memory over other T cell subsets; (2) upregulation of inhibitory markers (programmed death ligand 1); (3) heightened response to progesterone (PIBF); and (4) reduced proportions of myeloid DC and concurrent IDO activity (KTR). Conversely, antigen-specific T cell responses normalized in late pregnancy and were associated with increased markers of T cell activation (CD38, neopterin). However, these changes occur with a simultaneous upregulation of immune suppressive mechanisms including apoptosis (CD95), coinhibition (TIM-3), and immune regulation (IL-10) through the course of pregnancy. Together, our data suggest that immune tolerance dominates in the second trimester and that it is gradually reversed in the third trimester in association with immune activation as the end of pregnancy approaches.
Collapse
Affiliation(s)
- Nishel Mohan Shah
- Department of Surgery and Cancer, Imperial College London, Chelsea and Westminster Hospital, London, United Kingdom
| | - Anna A Herasimtschuk
- Department of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, United Kingdom
| | - Adriano Boasso
- Department of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, United Kingdom
| | - Adel Benlahrech
- Medical Research Council Human Immunology Unit, Weatherall Institute of Molecular Medicine and Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Dietmar Fuchs
- Division of Biological Chemistry, Biocenter, Innsbruck Medical University, Innsbruck, Austria
| | - Nesrina Imami
- Department of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, United Kingdom
| | - Mark R Johnson
- Department of Surgery and Cancer, Imperial College London, Chelsea and Westminster Hospital, London, United Kingdom
| |
Collapse
|
38
|
Faas MM, de Vos P. Maternal monocytes in pregnancy and preeclampsia in humans and in rats. J Reprod Immunol 2016; 119:91-97. [PMID: 27396500 DOI: 10.1016/j.jri.2016.06.009] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 06/17/2016] [Accepted: 06/27/2016] [Indexed: 12/20/2022]
Abstract
Monocytes are short-lived cells, arising from the bone marrow and maturing in the circulation. They play an important role in immune responses and are thought to be important for healthy pregnancy. In humans, 3 subpopulations of monocytes have been identified: classical, intermediate and non-classical monocytes. These subpopulations have different functions and phenotypical characteristics. Healthy pregnancy is characterized by a pro-inflammatory condition, with increased numbers of monocytes and monocyte activation as well as with increased numbers of intermediate monocytes and decreased numbers of classical monocytes. This may suggest monocyte maturation. Preeclampsia is an important pregnancy complication characterized by hypertension and proteinuria developing in the second half of pregnancy. The pathophysiology of preeclampsia is associated with further activation of the inflammatory response, further activation of monocytes and further monocyte maturation. In the present review we focus on the role of monocyte activation and maturation in healthy and preeclamptic pregnancy.
Collapse
Affiliation(s)
- M M Faas
- Section of Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen and University Medical Center Groningen, The Netherlands; Department of Obstetrics and Gynecology, University of Groningen and University Medical Center Groningen, The Netherlands.
| | - P de Vos
- Section of Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen and University Medical Center Groningen, The Netherlands
| |
Collapse
|
39
|
Leshansky L, Aberdam D, Itskovitz-Eldor J, Berrih-Aknin S. Human embryonic stem cells prevent T-cell activation by suppressing dendritic cells function via TGF-beta signaling pathway. Stem Cells 2015; 32:3137-49. [PMID: 25186014 DOI: 10.1002/stem.1833] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Accepted: 07/23/2014] [Indexed: 12/25/2022]
Abstract
Human embryonic stem cells (hESCs) represent a potential source of transplantable cells for regenerative medicine, but development of teratoma even in syngenic recipients represents a critical obstacle to safe stem cell-based therapies. We hypothesized that hESCs escape the immune surveillance by regulating the environmental immune system. Using cocultures of hESCs with allogenic peripheral blood mononuclear cells, we demonstrated that hESCs prevent proliferation and activation of human CD4+ T lymphocytes, an effect dependent upon monocytes. Altered expression of key signaling molecules responsible for the crosstalk of monocytes with T cells was detected in the presence of hESCs. Analyzing the mechanism of action, we demonstrated that hESCs were able to downregulate intracellular glutathione levels in both monocytes and CD4+ cells by suppressing glutamate cysteine ligase expression and to alter MHCII and CD80 expression in monocytes. These effects were achieved at least partially via TGF-beta signaling, and both monocyte phenotype and GCLC expression were affected by Caspase-3 proteolytic activity. Altogether, our results demonstrate a novel immune-suppressive mechanism used by hESCs.
Collapse
Affiliation(s)
- Lucy Leshansky
- INSERTECH Stem Cell Center, Ruth and Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel; Stem Cell Center, Ruth and Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | | | | | | |
Collapse
|
40
|
Boij R, Mjösberg J, Svensson-Arvelund J, Hjorth M, Berg G, Matthiesen L, Jenmalm MC, Ernerudh J. Regulatory T-cell Subpopulations in Severe or Early-onset Preeclampsia. Am J Reprod Immunol 2015; 74:368-78. [DOI: 10.1111/aji.12410] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 05/26/2015] [Indexed: 01/10/2023] Open
Affiliation(s)
- Roland Boij
- Department of Clinical and Experimental Medicine; Linköping University; Linköping Sweden
- Department of Obstetrics and Gynecology; County Hospital Ryhov; Jönköping Sweden
| | - Jenny Mjösberg
- Center for Infectious Medicine; Department of Medicine Huddinge; Karolinska Institutet; Stockholm Sweden
| | | | - Maria Hjorth
- Department of Clinical Immunology and Transfusion Medicine; Linköping University; Linköping Sweden
| | - Göran Berg
- Department of Clinical and Experimental Medicine; Linköping University; Linköping Sweden
| | - Leif Matthiesen
- Department of Clinical and Experimental Medicine; Linköping University; Linköping Sweden
- Department of Obstetrics and Gynecology; Helsingborg Hospital; Helsingborg Sweden
| | - Maria C. Jenmalm
- Department of Clinical and Experimental Medicine; Linköping University; Linköping Sweden
| | - Jan Ernerudh
- Department of Clinical and Experimental Medicine; Linköping University; Linköping Sweden
- Department of Clinical Immunology and Transfusion Medicine; Linköping University; Linköping Sweden
| |
Collapse
|
41
|
Pregnancy Associated with Systemic Lupus Erythematosus: Immune Tolerance in Pregnancy and Its Deficiency in Systemic Lupus Erythematosus--An Immunological Dilemma. J Immunol Res 2015; 2015:241547. [PMID: 26090485 PMCID: PMC4451247 DOI: 10.1155/2015/241547] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Revised: 02/05/2015] [Accepted: 02/06/2015] [Indexed: 01/04/2023] Open
Abstract
Pregnancy is a physiological condition that requires immune tolerance to the product of conception. Systemic lupus erythematosus (SLE) is a disease with well-represented immune mechanisms that disturb immune tolerance. The association of pregnancy with systemic lupus erythematosus creates a particular immune environment in which the immune tolerance specific of pregnancy is required to coexist with alterations of the immune system caused by SLE. The main role is played by T regulatory (Treg) cells, which attempt to regulate and adapt the immune system of the mother to the new conditions of pregnancy. Other components of the immune system also participate to maintain maternal-fetal immune tolerance. If the immune system of pregnant women with SLE is not able to maintain maternal immune tolerance to the fetus, pregnancy complications (miscarriage, fetal hypotrophy, and preterm birth) or maternal complications (preeclampsia or activation of SLE, especially in conditions of lupus nephritis) may occur. In certain situations this can be responsible for neonatal lupus. At the same time, it must be noted that during pregnancy, the immune system is able to achieve immune tolerance while maintaining the anti-infectious immune capacity of the mother. Immunological monitoring of pregnancy during SLE, as well as of the mother's disease, is required. It is important to understand immune tolerance to grafts in transplant pathology.
Collapse
|
42
|
Cordero-Coma M, Salazar-Méndez R, Yilmaz T. Treatment of severe non-infectious uveitis in high-risk conditions (Part I): pregnancy and malignancies, management and safety issues. Expert Opin Drug Saf 2015; 14:1071-86. [DOI: 10.1517/14740338.2015.1044969] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
43
|
Nancy P, Erlebacher A. T cell behavior at the maternal-fetal interface. THE INTERNATIONAL JOURNAL OF DEVELOPMENTAL BIOLOGY 2015; 58:189-98. [PMID: 25023685 DOI: 10.1387/ijdb.140054ae] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Understanding the function of T cells at the maternal-fetal interface remains one of the most difficult problems in reproductive immunology. A great deal of work over the last two decades has led to the view that the T cells that populate the decidua have important roles in both normal and pathological pregnancies, but the exact nature of these roles has remained unclear. Indeed, the old assumption that decidual T cells are uniformly threatening to fetal survival because the placenta is fundamentally an 'allograft' has given way to the idea that different T cell subsets contribute in different ways to pregnancy success or failure. Accordingly, some T cells are thought to protect the placenta from immune rejection and facilitate embryo implantation, while others are thought to contribute to pregnancy pathologies such as preeclampsia and spontaneous abortion. Here, we review the current state of information on the behavior of decidual T cells with a focus on both mouse and human studies, and with an emphasis on the many unresolved areas within this overall emerging framework.
Collapse
Affiliation(s)
- Patrice Nancy
- Department of Pathology, NYU School of Medicine, New York, USA.
| | | |
Collapse
|
44
|
Decidual cytokines and pregnancy complications: focus on spontaneous miscarriage. J Reprod Immunol 2015; 108:83-9. [PMID: 25771398 DOI: 10.1016/j.jri.2015.02.003] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Revised: 02/08/2015] [Accepted: 02/17/2015] [Indexed: 11/20/2022]
Abstract
The establishment of pregnancy requires the co-ordinated implantation of the embryo into the receptive decidua, placentation, trophoblast invasion of the maternal decidua and myometrium in addition to remodelling of the uterine spiral arteries. Failure of any of these steps can lead to a range of pregnancy complications, including miscarriage, pre-eclampsia, fetal growth restriction, placenta accreta and pre-term birth. Cytokines are small multifunctional proteins often derived from leucocytes and have primarily been described through their immunomodulatory actions. The maternal-fetal interface is considered to be immunosuppressed to allow development of the semi-allogeneic placental fetal unit. However, cytokine profiles of the decidua and different decidual cell types suggest that the in vivo situation might be more complex. Data suggest that decidual-derived cytokines not only play roles in immunosuppression, but also in other aspects of the establishment of pregnancy, including the regulation of trophoblast invasion and spiral artery remodelling. This review focuses on the potential role of decidua-derived cytokines in the aetiology of unexplained spontaneous miscarriage.
Collapse
|
45
|
Areia A, Vale-Pereira S, Alves V, Rodrigues-Santos P, Moura P, Mota-Pinto A. Membrane progesterone receptors in human regulatory T cells: a reality in pregnancy. BJOG 2015; 122:1544-50. [PMID: 25639501 DOI: 10.1111/1471-0528.13294] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/06/2014] [Indexed: 12/01/2022]
Abstract
OBJECTIVE To provide evidence of the existence of membrane progesterone receptor alpha (mPRα) on regulatory T cells (Treg) in peripheral blood during pregnancy, postulating a possible explanation for the effect of progesterone on preterm birth. DESIGN Cross-sectional study. SETTING Tertiary Obstetric Department in a University Hospital. POPULATION Healthy pregnant women. METHODS Treg cells from peripheral blood samples were studied by flow cytometry using multiple monoclonal antibody expression. MAIN OUTCOME MEASURES Evaluate the number and percentage of CD4(+) CD25(high) CD127(low) , the number and percentage of Treg cells among the total CD4(+) T cells, and the percentage and mean fluorescence intensity (MFI) of mPRα in that population, using several gating strategies. RESULTS 43 peripheral blood samples were collected from healthy women during pregnancy, whose median gestational age was 28.7 ± 7.1 (16-40) weeks. The percentage of CD4(+) in the total lymphocytes was 43% (32-51) and the percentage of CD4(+) CD25(high) CD127(low) was 4.8% (1.6-5.9), with only 45% (16-72) of those cells expressing the intracellular marker FoxP3 (Treg cell pool). We confirmed the existence of mPRα in that specific population because 8.0% (2.02-33) of the Treg cells were marked with the specific monoclonal antibody, with an mPRα(+) MFI of 719 (590-1471). CONCLUSIONS This research shows that Treg cells express mPRα during pregnancy, which might play an important role in immune modulation by progesterone.
Collapse
Affiliation(s)
- A Areia
- Faculty of Medicine, University of Coimbra and Obstetric Unit, Coimbra University Hospital Centre, Coimbra, Portugal
| | - S Vale-Pereira
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - V Alves
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | | | - P Moura
- Faculty of Medicine, University of Coimbra and Obstetric Unit, Coimbra University Hospital Centre, Coimbra, Portugal
| | - A Mota-Pinto
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
46
|
Svensson-Arvelund J, Ernerudh J. The Role of Macrophages in Promoting and Maintaining Homeostasis at the Fetal-Maternal Interface. Am J Reprod Immunol 2015; 74:100-9. [DOI: 10.1111/aji.12357] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Accepted: 02/24/2014] [Indexed: 12/25/2022] Open
Affiliation(s)
- Judit Svensson-Arvelund
- Clinical Immunology; Department of Clinical and Experimental Medicine; Faculty of Health Sciences; Linköping University; Linköping Sweden
| | - Jan Ernerudh
- Clinical Immunology; Department of Clinical and Experimental Medicine; Faculty of Health Sciences; Linköping University; Linköping Sweden
| |
Collapse
|
47
|
Svensson-Arvelund J, Mehta RB, Lindau R, Mirrasekhian E, Rodriguez-Martinez H, Berg G, Lash GE, Jenmalm MC, Ernerudh J. The Human Fetal Placenta Promotes Tolerance against the Semiallogeneic Fetus by Inducing Regulatory T Cells and Homeostatic M2 Macrophages. THE JOURNAL OF IMMUNOLOGY 2015; 194:1534-44. [DOI: 10.4049/jimmunol.1401536] [Citation(s) in RCA: 181] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
48
|
Abstract
Uveitides can be due to non-infectious and infectious etiologies. It has been observed that there is a gender difference with a greater preponderance of non-infectious uveitis in women than in men. This review will describe both non-infectious and infectious uveitides and describes some of the current autoimmune mechanisms thought to be underlying the gender difference. It will specifically look at non-infectious uveitides with systemic involvement including juvenile idiopathic arthritis, spondyloarthopathies, sarcoidosis, Behçet’s disease, and Vogt-Koyanagi-Harada disease and at uveitides without systemic involvement including sympathetic ophthalmia, birdshot chorioretinitis, and the white dot syndromes. Infectious uveitides like acute retinal necrosis, progressive outer retinal necrosis, and cytomegalovirus mediated uveitis will be mentioned. Different uveitides with female- or male- predominance are presented and discussed.
Collapse
|
49
|
Rosenzwajg M, Churlaud G, Hartemann A, Klatzmann D. Interleukin 2 in the pathogenesis and therapy of type 1 diabetes. Curr Diab Rep 2014; 14:553. [PMID: 25344788 DOI: 10.1007/s11892-014-0553-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Regulatory T cells (Tregs) play a major role in controlling effector T cells (Teffs) responding to self-antigens, which cause autoimmune diseases. An improper Treg/Teff balance contributes to most autoimmune diseases, including type 1 diabetes (T1D). To restore a proper balance, blocking Teffs with immunosuppressants has been the only option, which was partly effective and too toxic. It now appears that expanding/activating Tregs with low-dose interleukin-2 (IL-2) could provide immunoregulation without immunosuppression. This is particularly interesting in T1D as Tregs from T1D patients are reported as dysfunctional and a relative deficiency in IL-2 production and/or IL-2-mediated signaling could contribute to this phenotype. A clinical study of low-dose IL-2 showed a very good safety profile and good Treg expansion/activation in T1D patients. This opens the way for efficacy trials to test low-dose IL-2 in prevention and treatment of T1D and to establish in which condition restoration of a proper Treg/Teff balance would be beneficial in the field of autoimmune and inflammatory diseases.
Collapse
Affiliation(s)
- Michelle Rosenzwajg
- Biotherapy (CIC-BTi) and Inflammation-Immunopathology-Biotherapy Department (i2B), AP-HP, Hôpital Pitié-Salpêtrière, 83 Boulevard de l'Hôpital, 75651, Paris, France,
| | | | | | | |
Collapse
|
50
|
Faas MM, Spaans F, De Vos P. Monocytes and macrophages in pregnancy and pre-eclampsia. Front Immunol 2014; 5:298. [PMID: 25071761 PMCID: PMC4074993 DOI: 10.3389/fimmu.2014.00298] [Citation(s) in RCA: 136] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Accepted: 06/12/2014] [Indexed: 02/06/2023] Open
Abstract
Preeclampsia is an important complication in pregnancy, characterized by hypertension and proteinuria in the second half of pregnancy. Generalized activation of the inflammatory response is thought to play a role in the pathogenesis of pre-eclampsia. Monocytes may play a central role in this inflammatory response. Monocytes are short lived cells that mature in the circulation and invade into tissues upon an inflammatory stimulus and develop into macrophages. Macrophages are abundantly present in the endometrium and play a role in implantation and placentation in normal pregnancy. In pre-eclampsia, these macrophages appear to be present in larger numbers and are also activated. In the present review, we focused on the role of monocytes and macrophages in the pathophysiology of pre-eclampsia.
Collapse
Affiliation(s)
- Marijke M Faas
- Immunoendocrinology, Department of Pathology and Medical Biology, Division of Medical Biology, University Medical Center Groningen, University of Groningen , Groningen , Netherlands
| | - Floor Spaans
- Immunoendocrinology, Department of Pathology and Medical Biology, Division of Medical Biology, University Medical Center Groningen, University of Groningen , Groningen , Netherlands
| | - Paul De Vos
- Immunoendocrinology, Department of Pathology and Medical Biology, Division of Medical Biology, University Medical Center Groningen, University of Groningen , Groningen , Netherlands
| |
Collapse
|