1
|
Hoffmann JP, Liu JA, Seddu K, Klein SL. Sex hormone signaling and regulation of immune function. Immunity 2023; 56:2472-2491. [PMID: 37967530 DOI: 10.1016/j.immuni.2023.10.008] [Citation(s) in RCA: 69] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 09/25/2023] [Accepted: 10/14/2023] [Indexed: 11/17/2023]
Abstract
Immune responses to antigens, including innocuous, self, tumor, microbial, and vaccine antigens, differ between males and females. The quest to uncover the mechanisms for biological sex differences in the immune system has intensified, with considerable literature pointing toward sex hormonal influences on immune cell function. Sex steroids, including estrogens, androgens, and progestins, have profound effects on immune function. As such, drastic changes in sex steroid concentrations that occur with aging (e.g., after puberty or during the menopause transition) or pregnancy impact immune responses and the pathogenesis of immune-related diseases. The effect of sex steroids on immunity involves both the concentration of the ligand and the density and distribution of genomic and nongenomic receptors that serve as transcriptional regulators of immune cellular responses to affect autoimmunity, allergy, infectious diseases, cancers, and responses to vaccines. The next frontier will be harnessing these effects of sex steroids to improve therapeutic outcomes.
Collapse
Affiliation(s)
- Joseph P Hoffmann
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Jennifer A Liu
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Kumba Seddu
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Sabra L Klein
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA; Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA; Department of Medicine, Division of Infectious Diseases, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
2
|
Yang SC, Park M, Hong KH, La H, Park C, Wang P, Li G, Chen Q, Choi Y, DeMayo FJ, Lydon JP, Skalnik DG, Lim HJ, Hong SH, Park SH, Kim YS, Kim HR, Song H. CFP1 governs uterine epigenetic landscapes to intervene in progesterone responses for uterine physiology and suppression of endometriosis. Nat Commun 2023; 14:3220. [PMID: 37270588 DOI: 10.1038/s41467-023-39008-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 05/24/2023] [Indexed: 06/05/2023] Open
Abstract
Progesterone (P4) is required for the preparation of the endometrium for a successful pregnancy. P4 resistance is a leading cause of the pathogenesis of endometrial disorders like endometriosis, often leading to infertility; however, the underlying epigenetic cause remains unclear. Here we demonstrate that CFP1, a regulator of H3K4me3, is required for maintaining epigenetic landscapes of P4-progesterone receptor (PGR) signaling networks in the mouse uterus. Cfp1f/f;Pgr-Cre (Cfp1d/d) mice showed impaired P4 responses, leading to complete failure of embryo implantation. mRNA and chromatin immunoprecipitation sequencing analyses showed that CFP1 regulates uterine mRNA profiles not only in H3K4me3-dependent but also in H3K4me3-independent manners. CFP1 directly regulates important P4 response genes, including Gata2, Sox17, and Ihh, which activate smoothened signaling pathway in the uterus. In a mouse model of endometriosis, Cfp1d/d ectopic lesions showed P4 resistance, which was rescued by a smoothened agonist. In human endometriosis, CFP1 was significantly downregulated, and expression levels between CFP1 and these P4 targets are positively related regardless of PGR levels. In brief, our study provides that CFP1 intervenes in the P4-epigenome-transcriptome networks for uterine receptivity for embryo implantation and the pathogenesis of endometriosis.
Collapse
Affiliation(s)
- Seung Chel Yang
- Department of Biomedical Science, CHA University, Seongnam, Gyeonggi, 13488, Korea
| | - Mira Park
- Department of Biomedical Science, CHA University, Seongnam, Gyeonggi, 13488, Korea
| | - Kwon-Ho Hong
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul, 05029, Korea
| | - Hyeonwoo La
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul, 05029, Korea
| | - Chanhyeok Park
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul, 05029, Korea
| | - Peike Wang
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Gaizhen Li
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Qionghua Chen
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Youngsok Choi
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul, 05029, Korea
| | - Francesco J DeMayo
- Department of Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, 12233, USA
| | - John P Lydon
- Department of Molecular and Cellular Biology and Center for Reproductive Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
| | - David G Skalnik
- Department of Biology, School of Science, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202, USA
| | - Hyunjung J Lim
- Department of Veterinary Science, Konkuk University, Seoul, 05029, Korea
| | - Seok-Ho Hong
- Department of Internal Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, 24431, Korea
- KW-Bio Co., Ltd, Wonju, 26493, Korea
| | - So Hee Park
- Department of Biomedical Science, CHA University, Seongnam, Gyeonggi, 13488, Korea
| | - Yeon Sun Kim
- Department of Biomedical Science, CHA University, Seongnam, Gyeonggi, 13488, Korea
| | - Hye-Ryun Kim
- Department of Biomedical Science, CHA University, Seongnam, Gyeonggi, 13488, Korea
| | - Haengseok Song
- Department of Biomedical Science, CHA University, Seongnam, Gyeonggi, 13488, Korea.
| |
Collapse
|
3
|
Zenere A, Hellberg S, Papapavlou Lingehed G, Svenvik M, Mellergård J, Dahle C, Vrethem M, Raffetseder J, Khademi M, Olsson T, Blomberg M, Jenmalm MC, Altafini C, Gustafsson M, Ernerudh J. Prominent epigenetic and transcriptomic changes in CD4 + and CD8 + T cells during and after pregnancy in women with multiple sclerosis and controls. J Neuroinflammation 2023; 20:98. [PMID: 37106402 PMCID: PMC10134602 DOI: 10.1186/s12974-023-02781-2] [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: 10/17/2022] [Accepted: 04/17/2023] [Indexed: 04/29/2023] Open
Abstract
BACKGROUND Multiple sclerosis (MS) is a neuroinflammatory disease in which pregnancy leads to a temporary amelioration in disease activity as indicated by the profound decrease in relapses rate during the 3rd trimester of pregnancy. CD4+ and CD8+ T cells are implicated in MS pathogenesis as being key regulators of inflammation and brain lesion formation. Although Tcells are prime candidates for the pregnancy-associated improvement of MS, the precise mechanisms are yet unclear, and in particular, a deep characterization of the epigenetic and transcriptomic events that occur in peripheral T cells during pregnancy in MS is lacking. METHODS Women with MS and healthy controls were longitudinally sampled before, during (1st, 2nd and 3rd trimesters) and after pregnancy. DNA methylation array and RNA sequencing were performed on paired CD4+ and CD8+ T cells samples. Differential analysis and network-based approaches were used to analyze the global dynamics of epigenetic and transcriptomic changes. RESULTS Both DNA methylation and RNA sequencing revealed a prominent regulation, mostly peaking in the 3rd trimester and reversing post-partum, thus mirroring the clinical course with improvement followed by a worsening in disease activity. This rebound pattern was found to represent a general adaptation of the maternal immune system, with only minor differences between MS and controls. By using a network-based approach, we highlighted several genes at the core of this pregnancy-induced regulation, which were found to be enriched for genes and pathways previously reported to be involved in MS. Moreover, these pathways were enriched for in vitro stimulated genes and pregnancy hormones targets. CONCLUSION This study represents, to our knowledge, the first in-depth investigation of the methylation and expression changes in peripheral CD4+ and CD8+ T cells during pregnancy in MS. Our findings indicate that pregnancy induces profound changes in peripheral T cells, in both MS and healthy controls, which are associated with the modulation of inflammation and MS activity.
Collapse
Affiliation(s)
- Alberto Zenere
- Division of Automatic Control, Department of Electrical Engineering, Linköping University, Linköping, Sweden
| | - Sandra Hellberg
- Bioinformatics, Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden.
| | - Georgia Papapavlou Lingehed
- Division of Inflammation and Infection, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Maria Svenvik
- Division of Inflammation and Infection, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
- Department of Obstetrics and Gynecology, Region Kalmar County, Kalmar, Sweden
| | - Johan Mellergård
- Department of Neurology, Linköping University, Linköping, Sweden
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Charlotte Dahle
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
- Department of Clinical Immunology and Transfusion Medicine, Linköping University, Linköping, Sweden
| | - Magnus Vrethem
- Department of Neurology, Linköping University, Linköping, Sweden
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Johanna Raffetseder
- Division of Inflammation and Infection, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Mohsen Khademi
- Neuroimmunology Unit, Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| | - Tomas Olsson
- Neuroimmunology Unit, Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| | - Marie Blomberg
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
- Department of Obstetrics and Gynecology, Linköping University, Linköping, Sweden
| | - Maria C Jenmalm
- Division of Inflammation and Infection, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Claudio Altafini
- Division of Automatic Control, Department of Electrical Engineering, Linköping University, Linköping, Sweden
| | - Mika Gustafsson
- Bioinformatics, Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden.
| | - Jan Ernerudh
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
- Department of Clinical Immunology and Transfusion Medicine, Linköping University, Linköping, Sweden
| |
Collapse
|
4
|
Lin HC, Chang HM, Hung YM, Chang R, Chen HH, Wei JCC. Hashimoto's thyroiditis increases the risk of new-onset systemic lupus erythematosus: a nationwide population-based cohort study. Arthritis Res Ther 2023; 25:20. [PMID: 36759862 PMCID: PMC9909872 DOI: 10.1186/s13075-023-02999-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Accepted: 01/27/2023] [Indexed: 02/11/2023] Open
Abstract
BACKGROUND Previous studies have shown systemic lupus erythematosus (SLE) patients had a significantly higher prevalence of thyroid diseases and hypothyroidism than matched controls, and some case reports showed SLE may occur after Hashimoto's thyroiditis (HT). OBJECTIVE This study aimed to investigate the subsequent risk of SLE in patients with HT. METHODS In this retrospective cohort study done by the Taiwan National Health Insurance Research Database, the HT group (exposure group) and the non-HT group (comparator group) were propensity score matched at a ratio of 1:2 by demographic data, comorbidities, medications, and the index date. We used Cox proportional hazards models to estimate hazard ratios (HRs) and 95% confidence intervals (CIs). Several sensitivity analyses were done for cross-validation of our findings. RESULTS We identified 15,512 HT patients and matched 31,024 individuals. The incidence rate ratio of SLE was 3.58 (95% CI, 2.43-5.28; p < 0.01). Several sensitivity analyses show adjusted hazard ratio (aHR) (CIs) of 4.35 (3.28-5.76), 4.39 (3.31-5.82), 5.11 (3.75-6.98), and 4.70 (3.46-6.38), consistent with the results of the main model. CONCLUSION Our study showed an increased risk of SLE in the HT group after adjustment for baseline characteristics, comorbidities, and medical confounders compared with the reference group.
Collapse
Affiliation(s)
- Hong-Ci Lin
- grid.411641.70000 0004 0532 2041School of Medicine, Department of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Hsu-Min Chang
- grid.415011.00000 0004 0572 9992Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Yao-Min Hung
- grid.415007.70000 0004 0477 6869Department of Internal Medicine, Kaohsiung Municipal United Hospital, Kaohsiung, Taiwan ,grid.419674.90000 0004 0572 7196College of Health and Nursing, Meiho University, Pingtung, Taiwan
| | - Renin Chang
- grid.415011.00000 0004 0572 9992Department of Emergency Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Hsin-Hua Chen
- Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, Taichung, Taiwan. .,Institute of Biomedical Science and Rong Hsing Research Centre for Translational Medicine, Chung Hsing University, Taichung, Taiwan. .,Department of Industrial Engineering and Enterprise Information, Tunghai University, Taichung, Taiwan.
| | - James Cheng-Chung Wei
- Division of Allergy, Immunology and Rheumatology, Chung Shan Medical University Hospital, Taichung, Taiwan. .,Institute of Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan. .,Graduate Institute of Integrated Medicine, China Medical University, Taichung, Taiwan.
| |
Collapse
|
5
|
Campe KNJ, Redlich A, Zenclussen AC, Busse M. An increased proportion of progesterone receptor A in peripheral B cells from women who ultimately underwent spontaneous preterm birth. J Reprod Immunol 2022; 154:103756. [PMID: 36356365 DOI: 10.1016/j.jri.2022.103756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 09/08/2022] [Accepted: 10/17/2022] [Indexed: 11/05/2022]
Abstract
The enormous challenge in unraveling the etiology of preterm birth (PTB) is to understand the complex interactions between gestational hormones, the immune system and reproductive tissues. PTB can be divided into spontaneous PTB (sPTB) and medically-indicated PTB, e.g. due to preeclampsia (PE) or HELLP syndrome. Progesterone (P4), important for establishment and maintenance of pregnancy, exerts anti-inflammatory effects. The impact of P4 on B cells and its support of maintaining maternal-fetal tolerance is widely unexplored. Therefore, we aimed to determine whether B cells express the progesterone receptor (PR) and to dissect a possible role of PR+ B cells in PTB. We found enhanced IL-6, IL-21 and TNF-α concentrations in maternal plasma in patients with sPTB and PE/HELLP compared to term delivery (TD), accompanied by enhanced PR-A expression by CD19+ B cells. In a second phase of the study, we recruited patients with imminent PTB (iPTB) and controls. Samples were collected at hospital admission and to a later time point, then divided into iPTB patients who delivered preterm and patients whose PTB was prevented. Within the group of iPTB patients, we observed very clear differences: enhanced levels of pro-inflammatory cytokines and increased percentages of PR-A+CD19+ B cells were found in iPTB patients that delivered preterm compared to patients who did not deliver preterm. We conclude that PTB is associated with the activation of an inflammatory pathway leading to the induction of PR-A by B cells. This might further trigger inflammation, result in the break of maternal-fetal tolerance and induce delivery.
Collapse
Affiliation(s)
- Kim-Norina Jutta Campe
- Experimental Obstetrics and Gynecology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Anke Redlich
- University hospital for Obstetrics and Gynecology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Ana Claudia Zenclussen
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research, Leipzig, Germany; Perinatal Research Group, Saxonian Incubator for Translation, Leipzig University, Leipzig, Germany
| | - Mandy Busse
- Experimental Obstetrics and Gynecology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany.
| |
Collapse
|
6
|
Bulletti C, Bulletti FM, Sciorio R, Guido M. Progesterone: The Key Factor of the Beginning of Life. Int J Mol Sci 2022; 23:ijms232214138. [PMID: 36430614 PMCID: PMC9692968 DOI: 10.3390/ijms232214138] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/03/2022] [Accepted: 11/05/2022] [Indexed: 11/17/2022] Open
Abstract
Progesterone is the ovarian steroid produced by the granulosa cells of follicles after the LH peak at mid-cycle. Its role is to sustain embryo endometrial implantation and ongoing pregnancy. Other biological effects of progesterone may exert a protective function in supporting pregnancy up to birth. Luteal phase support (LPS) with progesterone is the standard of care for assisted reproductive technology. Progesterone vaginal administration is currently the most widely used treatment for LPS. Physicians and patients have been reluctant to change an administration route that has proven to be effective. However, some questions remain open, namely the need for LPS in fresh and frozen embryo transfer, the route of administration, the optimal duration of LPS, dosage, and the benefit of combination therapies. The aim of this review is to provide an overview of the uterine and extra-uterine effects of progesterone that may play a role in embryo implantation and pregnancy, and to discuss the advantages of the use of progesterone for LPS in the context of Good Medical Practice.
Collapse
Affiliation(s)
- Carlo Bulletti
- Extra Omnes, Assisted Reproductive Technology, ART Center, Via Gallinelli, 8, 47841 Cattolica, Italy
- Department of Obstetrics, Gynecology, and Reproductive Science, Yale University, New Haven, CT 06510, USA
- Correspondence:
| | | | - Romualdo Sciorio
- Edinburgh Assisted Conception Programme, Royal Infirmary of Edinburgh, Edinburgh EH16 4SA, UK
| | - Maurizio Guido
- Obstetrics and Gynecology Unit, Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| |
Collapse
|
7
|
Sharma S, Zaher S, Rodrigues PRS, Davies LC, Edkins S, Strang A, Chakraborty M, Watkins WJ, Andrews R, Parkinson E, Angelopoulos N, Moet L, Shepherd F, Davies KMM, White D, Oram S, Siddall K, Keeping V, Simpson K, Faggian F, Bray M, Bertorelli C, Bell S, Collis RE, McLaren JE, Labeta M, O'Donnell VB, Ghazal P. mSep: investigating physiological and immune-metabolic biomarkers in septic and healthy pregnant women to predict feto-maternal immune health - a prospective observational cohort study protocol. BMJ Open 2022; 12:e066382. [PMID: 36115679 PMCID: PMC9486348 DOI: 10.1136/bmjopen-2022-066382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 08/24/2022] [Indexed: 11/25/2022] Open
Abstract
INTRODUCTION Maternal sepsis remains a leading cause of death in pregnancy. Physiological adaptations to pregnancy obscure early signs of sepsis and can result in delays in recognition and treatment. Identifying biomarkers that can reliably diagnose sepsis will reduce morbidity and mortality and antibiotic overuse. We have previously identified an immune-metabolic biomarker network comprising three pathways with a >99% accuracy for detecting bacterial neonatal sepsis. In this prospective study, we will describe physiological parameters and novel biomarkers in two cohorts-healthy pregnant women and pregnant women with suspected sepsis-with the aim of mapping pathophysiological drivers and evaluating predictive biomarkers for diagnosing maternal sepsis. METHODS AND ANALYSIS Women aged over 18 with an ultrasound-confirmed pregnancy will be recruited to a pilot and two main study cohorts. The pilot will involve blood sample collection from 30 pregnant women undergoing an elective caesarean section. Cohort A will follow 100 healthy pregnant women throughout their pregnancy journey, with collection of blood samples from participants at routine time points in their pregnancy: week 12 'booking', week 28 and during labour. Cohort B will follow 100 pregnant women who present with suspected sepsis in pregnancy or labour and will have at least two blood samples taken during their care pathway. Study blood samples will be collected during routine clinical blood sampling. Detailed medical history and physiological parameters at the time of blood sampling will be recorded, along with the results of routine biochemical tests, including C reactive protein, lactate and white blood cell count. In addition, study blood samples will be processed and analysed for transcriptomic, lipidomic and metabolomic analyses and both qualitative and functional immunophenotyping. ETHICS AND DISSEMINATION Ethical approval has been obtained from the Wales Research Ethics Committee 2 (SPON1752-19, 30 October 2019). TRIAL REGISTRATION NUMBER NCT05023954.
Collapse
Affiliation(s)
- Simran Sharma
- Department of Obstetrics and Gynaecology, University Hospital of Wales, Cardiff, UK
- Project Sepsis, Systems Immunity Research Institute, Cardiff University Cardiff Institute of Infection and Immunity, Cardiff, UK
| | - Summia Zaher
- Department of Obstetrics and Gynaecology, University Hospital of Wales, Cardiff, UK
- Project Sepsis, Systems Immunity Research Institute, Cardiff University Cardiff Institute of Infection and Immunity, Cardiff, UK
| | - Patrícia R S Rodrigues
- Project Sepsis, Systems Immunity Research Institute, Cardiff University Cardiff Institute of Infection and Immunity, Cardiff, UK
- Cardiff Division of Infection and Immunity, Cardiff University, Cardiff, UK
| | - Luke C Davies
- Project Sepsis, Systems Immunity Research Institute, Cardiff University Cardiff Institute of Infection and Immunity, Cardiff, UK
- Cardiff Division of Infection and Immunity, Cardiff University, Cardiff, UK
| | - Sarah Edkins
- Project Sepsis, Systems Immunity Research Institute, Cardiff University Cardiff Institute of Infection and Immunity, Cardiff, UK
- Cardiff Division of Infection and Immunity, Cardiff University, Cardiff, UK
| | - Angela Strang
- Project Sepsis, Systems Immunity Research Institute, Cardiff University Cardiff Institute of Infection and Immunity, Cardiff, UK
- Cardiff Division of Infection and Immunity, Cardiff University, Cardiff, UK
| | - Mallinath Chakraborty
- Regional Neonatal Intensive Care Unit, University Hospital of Wales Healthcare NHS Trust, Cardiff, UK
| | - W John Watkins
- Project Sepsis, Systems Immunity Research Institute, Cardiff University Cardiff Institute of Infection and Immunity, Cardiff, UK
| | - Robert Andrews
- Project Sepsis, Systems Immunity Research Institute, Cardiff University Cardiff Institute of Infection and Immunity, Cardiff, UK
- Cardiff Division of Infection and Immunity, Cardiff University, Cardiff, UK
| | - Edward Parkinson
- Project Sepsis, Systems Immunity Research Institute, Cardiff University Cardiff Institute of Infection and Immunity, Cardiff, UK
- Cardiff Division of Infection and Immunity, Cardiff University, Cardiff, UK
| | - Nicos Angelopoulos
- Project Sepsis, Systems Immunity Research Institute, Cardiff University Cardiff Institute of Infection and Immunity, Cardiff, UK
- Cardiff Division of Infection and Immunity, Cardiff University, Cardiff, UK
| | - Linda Moet
- Project Sepsis, Systems Immunity Research Institute, Cardiff University Cardiff Institute of Infection and Immunity, Cardiff, UK
- Cardiff Division of Infection and Immunity, Cardiff University, Cardiff, UK
| | - Freya Shepherd
- Project Sepsis, Systems Immunity Research Institute, Cardiff University Cardiff Institute of Infection and Immunity, Cardiff, UK
- Cardiff Division of Infection and Immunity, Cardiff University, Cardiff, UK
| | - Kate Megan Megan Davies
- Project Sepsis, Systems Immunity Research Institute, Cardiff University Cardiff Institute of Infection and Immunity, Cardiff, UK
- Cardiff Division of Infection and Immunity, Cardiff University, Cardiff, UK
| | - Daniel White
- Project Sepsis, Systems Immunity Research Institute, Cardiff University Cardiff Institute of Infection and Immunity, Cardiff, UK
- Cardiff Division of Infection and Immunity, Cardiff University, Cardiff, UK
| | - Shaun Oram
- Department of Anaesthesia and Critical Care, University Hospital of Wales, Cardiff, UK
| | - Kate Siddall
- Department of Obstetrics and Gynaecology, University Hospital of Wales, Cardiff, UK
| | - Vikki Keeping
- Department of Obstetrics and Gynaecology, University Hospital of Wales, Cardiff, UK
| | - Kathryn Simpson
- Department of Anaesthesia and Critical Care, University Hospital of Wales, Cardiff, UK
| | - Federica Faggian
- Department of Microbiology, University Hospital of Wales, Cardiff, UK
| | - Maryanne Bray
- Department of Obstetrics and Gynaecology, University Hospital of Wales, Cardiff, UK
| | - Claire Bertorelli
- Department of Obstetrics and Gynaecology, University Hospital of Wales, Cardiff, UK
| | - Sarah Bell
- Project Sepsis, Systems Immunity Research Institute, Cardiff University Cardiff Institute of Infection and Immunity, Cardiff, UK
- Department of Anaesthesia and Critical Care, University of Wales Cardiff, Cardiff, UK
| | - Rachel E Collis
- Project Sepsis, Systems Immunity Research Institute, Cardiff University Cardiff Institute of Infection and Immunity, Cardiff, UK
- Department of Anaesthetics, Intensive Care and Pain Medicine, Cardiff and Vale University Health Board, Cardiff, UK
| | - James E McLaren
- Project Sepsis, Systems Immunity Research Institute, Cardiff University Cardiff Institute of Infection and Immunity, Cardiff, UK
- Cardiff Division of Infection and Immunity, Cardiff University, Cardiff, UK
| | - Mario Labeta
- Cardiff Division of Infection and Immunity, Cardiff University, Cardiff, UK
| | - Valerie B O'Donnell
- Project Sepsis, Systems Immunity Research Institute, Cardiff University Cardiff Institute of Infection and Immunity, Cardiff, UK
- Cardiff Division of Infection and Immunity, Cardiff University, Cardiff, UK
| | - Peter Ghazal
- Project Sepsis, Systems Immunity Research Institute, Cardiff University Cardiff Institute of Infection and Immunity, Cardiff, UK
- Cardiff Division of Infection and Immunity, Cardiff University, Cardiff, UK
| |
Collapse
|
8
|
Taves MD, Ashwell JD. Effects of sex steroids on thymic epithelium and thymocyte development. Front Immunol 2022; 13:975858. [PMID: 36119041 PMCID: PMC9478935 DOI: 10.3389/fimmu.2022.975858] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 07/18/2022] [Indexed: 11/13/2022] Open
Abstract
Sex steroid hormones have major effects on the thymus. Age-related increases in androgens and estrogens and pregnancy-induced increases in progestins all cause dramatic thymic atrophy. Atrophy can also be induced by treatment with exogenous sex steroids and reversed by ablation of endogenous sex steroids. Although these observations are frequently touted as evidence of steroid lymphotoxicity, they are often driven by steroid signaling in thymic epithelial cells (TEC), which are highly steroid responsive. Here, we outline the effects of sex steroids on the thymus and T cell development. We focus on studies that have examined steroid signaling in vivo, aiming to emphasize the actions of endogenous steroids which, via TEC, have remarkable programming effects on the TCR repertoire. Due to the dramatic effects of steroids on TEC, especially thymic involution, the direct effects of sex steroid signaling in thymocytes are less well understood. We outline studies that could be important in addressing these possibilities, and highlight suggestive findings of sex steroid generation within the thymus itself.
Collapse
Affiliation(s)
- Matthew D. Taves
- Laboratory of Immune Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
- Department of Neurobiology and Behavior, Cornell University, Ithaca, NY, United States
| | - Jonathan D. Ashwell
- Laboratory of Immune Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
9
|
Moldenhauer LM, Jin M, Wilson JJ, Green ES, Sharkey DJ, Salkeld MD, Bristow TC, Hull ML, Dekker GA, Robertson SA. Regulatory T Cell Proportion and Phenotype Are Altered in Women Using Oral Contraception. Endocrinology 2022; 163:6628694. [PMID: 35786711 PMCID: PMC9354970 DOI: 10.1210/endocr/bqac098] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Indexed: 11/19/2022]
Abstract
Regulatory T (Treg) cells are a specialized CD4+ T cell subpopulation that are essential for immune homeostasis, immune tolerance, and protection against autoimmunity. There is evidence that sex-steroid hormones estrogen and progesterone modulate Treg cell abundance and phenotype in women. Since natural oscillations in these hormones are modified by hormonal contraceptives, we examined whether oral contraception (OC) use impacts Treg cells and related T cell populations. T cells were analyzed by multiparameter flow cytometry in peripheral blood collected across the menstrual cycle from healthy women either using OC or without hormonal contraception and from age-matched men. Compared to naturally cycling women, women using OC had fewer Treg cells and an altered Treg cell phenotype. Notably, Treg cells exhibiting a strongly suppressive phenotype, defined by high FOXP3, CD25, Helios, HLADR, CTLA4, and Ki67, comprised a lower proportion of total Treg cells, particularly in the early- and mid-cycle phases. The changes were moderate compared to more substantial differences in Treg cells between women and men, wherein women had fewer Treg cells-especially of the effector memory Treg cell subset-associated with more T helper type 1 (Th1) cells and CD8+ T cells and lower Treg:Th1 cell and Treg:CD8+ T cell ratios than men. These findings imply that OC can modulate the number and phenotype of peripheral blood Treg cells and raise the possibility that Treg cells contribute to the physiological changes and altered disease susceptibility linked with OC use.
Collapse
Affiliation(s)
| | | | - Jasmine J Wilson
- Robinson Research Institute and School of Biomedicine, University of Adelaide, Adelaide, Australia
| | - Ella S Green
- Robinson Research Institute and School of Biomedicine, University of Adelaide, Adelaide, Australia
| | - David J Sharkey
- Robinson Research Institute and School of Biomedicine, University of Adelaide, Adelaide, Australia
| | - Mark D Salkeld
- Robinson Research Institute and School of Biomedicine, University of Adelaide, Adelaide, Australia
| | - Thomas C Bristow
- Robinson Research Institute and School of Biomedicine, University of Adelaide, Adelaide, Australia
| | - M Louise Hull
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, Australia
| | - Gustaaf A Dekker
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, Australia
- Division of Women’s Health, Lyell McEwin Hospital, Elizabeth Vale, Australia
| | - Sarah A Robertson
- Correspondence: Sarah A. Robertson, PhD, Robinson Research Institute and the School of Biomedicine, University of Adelaide, Adelaide, SA 5005, Australia.
| |
Collapse
|
10
|
A Comprehensive Investigation into the Distribution of Circulating B Cell Subsets in the Third Trimester of Pregnancy. J Clin Med 2022; 11:jcm11113006. [PMID: 35683395 PMCID: PMC9181443 DOI: 10.3390/jcm11113006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/17/2022] [Accepted: 05/22/2022] [Indexed: 01/25/2023] Open
Abstract
Maternal B cells play a crucial role in the development and maintenance of pregnancy, due to their humoral activities and regulatory functions. In the study, we investigated the alterations in the distributions of naïve and memory B cell subsets, as well as regulatory B (Breg) cells, in the third trimester of pregnancy. Peripheral blood from 14 healthy pregnant women in the third trimester and 7 healthy non-pregnant women was collected and examined for the frequencies of B cell subsets, including IgD+CD27− naïve, IgD+CD27+ un-switched memory, IgD−CD27+ switched memory, CD38intCD24int mature–naïve, CD38−CD24hi primarily memory and CD38hiCD24hi transitional B cells by flow cytometry. Breg cell subsets were also characterized based on the expression of CD5, CD1d and IL-10. In pregnant women, the proportions of un-switched memory and transitional B cells were significantly decreased. Additionally, the frequencies of both CD5+CD1d+ Breg and IL-10-producing B10 cells were decreased in pregnancy. Changes in the distribution of transitional B cells as well as Breg cells may be crucial contributors for the development of altered maternal immune responses and tolerance needed for the maintenance of normal pregnancy in the third trimester.
Collapse
|
11
|
Hu M, Valicherla GR, Zhou T, Hillier SL, Rohan LC. Expression, Activity, and Regulation of Phosphorylating Enzymes in Tissues and Cells Relevant to HIV-1 Sexual Transmission. AIDS Res Hum Retroviruses 2022; 38:22-32. [PMID: 33567990 PMCID: PMC8785762 DOI: 10.1089/aid.2020.0250] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Phosphorylating enzymes (PEs) are responsible for activating nucleoside/nucleotide reverse transcriptase inhibitors (NRTIs) such as tenofovir (TFV) and are critical for their conversion to obtain intracellular antiviral activity. However, there are limited data available regarding the expression of PEs and their activity in the female genital tract. This work compared the messenger RNA (mRNA) expression levels of PEs in human female genital tissue, immune cells, and animal models that are commonly used in human immunodeficiency virus (HIV) research. Furthermore, the effect of contraceptive hormones and proinflammatory cytokines on tenofovir diphosphate (TFV-DP) formation and efficacy in human vaginal, epithelial, and immune cells was also evaluated. We found that human vaginal and ectocervical tissues had similar mRNA expression for seven PEs tested. Polymerase chain reaction results revealed that creatine kinase brain (CKB), mitochondrial creatine kinase 1 (CKMT1), mitochondrial creatine kinase 2 (CKMT2), adenylate kinase AK3L1 (AK4), and nucleoside diphosphate kinase 1 (NME1) exhibited a 10- to 10,000-fold higher expression level in a vaginal epithelial cell line, VK2, compared with CD4+ T cells (p < .05). Medroxyprogesterone acetate (MPA)/progesterone (P4) and IL-1β/IL-8 treatment resulted in altered TFV-DP levels in VK2 and PM1 cells. MPA and P4 at concentrations above 0.1 μM, as well as IL-1β and IL-8 at concentrations above 10 ng/mL, significantly decreased HIV-1BaL inhibition in PM1 cells when 1 μM TFV was added. However, this observed effect of hormones and cytokines was abrogated when TFV concentration was raised to 1 mM. These in vitro results elucidate the role of PEs in TFV metabolism and provide information regarding differences in PE tissue expression for animal models commonly used in HIV testing. This information can be applied to better understand and interpret data obtained using these models.
Collapse
Affiliation(s)
- Minlu Hu
- Magee-Womens Research Institute, Pittsburgh, Pennsylvania, USA.,Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Guru R. Valicherla
- Magee-Womens Research Institute, Pittsburgh, Pennsylvania, USA.,Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Tian Zhou
- Magee-Womens Research Institute, Pittsburgh, Pennsylvania, USA.,Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Sharon L. Hillier
- Magee-Womens Research Institute, Pittsburgh, Pennsylvania, USA.,Department of Obstetrics, Gynecology, and Reproductive Sciences, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Lisa C. Rohan
- Magee-Womens Research Institute, Pittsburgh, Pennsylvania, USA.,Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Obstetrics, Gynecology, and Reproductive Sciences, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Address correspondence to: Lisa C. Rohan, Magee-Womens Research Institute, 204 Craft Avenue, Pittsburgh, Pennsylvania 15213, USA
| |
Collapse
|
12
|
Xing E, Billi AC, Gudjonsson JE. Sex Bias and Autoimmune Diseases. J Invest Dermatol 2021; 142:857-866. [PMID: 34362556 DOI: 10.1016/j.jid.2021.06.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 06/03/2021] [Accepted: 06/15/2021] [Indexed: 02/08/2023]
Abstract
Sex bias in immune function has been well-described, and women have been shown to counter immunologically stimulating phenomena such as infection, malignancy, and trauma with more protective responses than men. Heightened immunity in women may also result in a predisposition for loss of self-tolerance and development of autoimmunity, reflected by the overwhelming female sex bias of patients with autoimmune diseases. In this review, we discuss the postulated evolutionary etiologies for sexual dimorphism in immunity. We also review the molecular mechanisms underlying divergent immune responses in men and women, including sex hormone effects, X chromosome dosage, and autosomal sex-biased genes. With increasing evidence that autoimmune disease susceptibility is influenced by numerous hormonal and genetic factors, a comprehensive understanding of these topics may facilitate the development of much-needed targeted therapeutics.
Collapse
Affiliation(s)
- Enze Xing
- Department of Dermatology, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Allison C Billi
- Department of Dermatology, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Johann E Gudjonsson
- Department of Dermatology, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA; A. Alfred Taubman Medical Research Institute, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA.
| |
Collapse
|
13
|
Hellberg S, Raffetseder J, Rundquist O, Magnusson R, Papapavlou G, Jenmalm MC, Ernerudh J, Gustafsson M. Progesterone Dampens Immune Responses in In Vitro Activated CD4 + T Cells and Affects Genes Associated With Autoimmune Diseases That Improve During Pregnancy. Front Immunol 2021; 12:672168. [PMID: 34054852 PMCID: PMC8149943 DOI: 10.3389/fimmu.2021.672168] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 04/26/2021] [Indexed: 12/13/2022] Open
Abstract
The changes in progesterone (P4) levels during and after pregnancy coincide with the temporary improvement and worsening of several autoimmune diseases like multiple sclerosis (MS) and rheumatoid arthritis (RA). Most likely immune-endocrine interactions play a major role in these pregnancy-induced effects. In this study, we used next generation sequencing to investigate the direct effects of P4 on CD4+ T cell activation, key event in pregnancy and disease. We report profound dampening effects of P4 on T cell activation, altering the gene and protein expression profile and reversing many of the changes induced during the activation. The transcriptomic changes induced by P4 were significantly enriched for genes associated with diseases known to be modulated during pregnancy such as MS, RA and psoriasis. STAT1 and STAT3 were significantly downregulated by P4 and their downstream targets were significantly enriched among the disease-associated genes. Several of these genes included well-known and disease-relevant cytokines, such as IL-12β, CXCL10 and OSM, which were further validated also at the protein level using proximity extension assay. Our results extend the previous knowledge of P4 as an immune regulatory hormone and support its importance during pregnancy for regulating potentially detrimental immune responses towards the semi-allogenic fetus. Further, our results also point toward a potential role for P4 in the pregnancy-induced disease immunomodulation and highlight the need for further studies evaluating P4 as a future treatment option.
Collapse
Affiliation(s)
- Sandra Hellberg
- Bioinformatics, Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden.,Division of Inflammation and Infection, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Johanna Raffetseder
- Division of Inflammation and Infection, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Olof Rundquist
- Bioinformatics, Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden
| | - Rasmus Magnusson
- Bioinformatics, Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden
| | - Georgia Papapavlou
- Division of Inflammation and Infection, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Maria C Jenmalm
- Division of Inflammation and Infection, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Jan Ernerudh
- Department of Clinical Immunology and Transfusion Medicine and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Mika Gustafsson
- Bioinformatics, Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden
| |
Collapse
|
14
|
Werner LR, Gibson KA, Goodman ML, Helm DE, Walter KR, Holloran SM, Trinca GM, Hastings RC, Yang HH, Hu Y, Wei J, Lei G, Yang XY, Madan R, Molinolo AA, Markiewicz MA, Chalise P, Axelrod ML, Balko JM, Hunter KW, Hartman ZC, Lange CA, Hagan CR. Progesterone promotes immunomodulation and tumor development in the murine mammary gland. J Immunother Cancer 2021; 9:e001710. [PMID: 33958486 PMCID: PMC8103939 DOI: 10.1136/jitc-2020-001710] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/18/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Clinical studies have linked usage of progestins (synthetic progesterone [P4]) to breast cancer risk. However, little is understood regarding the role of native P4, signaling through the progesterone receptor (PR), in breast tumor formation. Recently, we reported a link between PR and immune signaling pathways, showing that P4/PR can repress type I interferon signaling pathways. Given these findings, we sought to investigate whether P4/PR drive immunomodulation in the mammary gland and promote tumor formation. METHODS To determine the effect of P4 on immune cell populations in the murine mammary gland, mice were treated with P4 or placebo pellets for 21 days. Immune cell populations in the mammary gland, spleen, and inguinal lymph nodes were subsequently analyzed by flow cytometry. To assess the effect of PR overexpression on mammary gland tumor development as well as immune cell populations in the mammary gland, a transgenic mouse model was used in which PR was overexpressed throughout the entire mouse. Immune cell populations were assessed in the mammary glands, spleens, and inguinal lymph nodes of 6-month-old transgenic and control mice by flow cytometry. Transgenic mice were also monitored for mammary gland tumor development over a 2-year time span. Following development of mammary gland tumors, immune cell populations in the tumors and spleens of transgenic and control mice were analyzed by flow cytometry. RESULTS We found that mice treated with P4 exhibited changes in the mammary gland indicative of an inhibited immune response compared with placebo-treated mice. Furthermore, transgenic mice with PR overexpression demonstrated decreased numbers of immune cell populations in their mammary glands, lymph nodes, and spleens. On long-term monitoring, we determined that multiparous PR-overexpressing mice developed significantly more mammary gland tumors than control mice. Additionally, tumors from PR-overexpressing mice contained fewer infiltrating immune cells. Finally, RNA sequencing analysis of tumor samples revealed that immune-related gene signatures were lower in tumors from PR-overexpressing mice as compared with control mice. CONCLUSION Together, these findings offer a novel mechanism of P4-driven mammary gland tumor development and provide rationale in investigating the usage of antiprogestin therapies to promote immune-mediated elimination of mammary gland tumors.
Collapse
MESH Headings
- Adaptive Immunity/drug effects
- Animals
- Breast Neoplasms/chemically induced
- Breast Neoplasms/immunology
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Cell Line, Tumor
- Cell Transformation, Neoplastic/chemically induced
- Cell Transformation, Neoplastic/immunology
- Cell Transformation, Neoplastic/metabolism
- Cell Transformation, Neoplastic/pathology
- Drug Implants
- Female
- Galectin 4/genetics
- Galectin 4/metabolism
- Immunity, Innate/drug effects
- Lymphocytes, Tumor-Infiltrating/drug effects
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Mammary Glands, Animal/drug effects
- Mammary Glands, Animal/immunology
- Mammary Glands, Animal/metabolism
- Mammary Glands, Animal/pathology
- Mice, Transgenic
- Ovariectomy
- Progesterone/administration & dosage
- Receptors, Progesterone/agonists
- Receptors, Progesterone/genetics
- Receptors, Progesterone/metabolism
- Signal Transduction
- Time Factors
- Tumor Burden/drug effects
- Tumor Escape/drug effects
- Tumor Microenvironment/immunology
Collapse
Affiliation(s)
- Lauryn R Werner
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Katelin A Gibson
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Merit L Goodman
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Dominika E Helm
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Katherine R Walter
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Sean M Holloran
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Gloria M Trinca
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Richard C Hastings
- Flow Cytometry Core Laboratory, University of Kansas Medical Center, Kansas City, Kansas, USA
- Department of Microbiology, Molecular Genetics, and Immunology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Howard H Yang
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Ying Hu
- Center for Biomedical Informatics and Information Technology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Junping Wei
- Department of Surgery, Duke University, Durham, North Carolina, USA
| | - Gangjun Lei
- Department of Surgery, Duke University, Durham, North Carolina, USA
| | - Xiao-Yi Yang
- Department of Surgery, Duke University, Durham, North Carolina, USA
| | - Rashna Madan
- Division of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Alfredo A Molinolo
- Department of Pathology, University of California San Diego Moores Cancer Center, La Jolla, California, USA
| | - Mary A Markiewicz
- Department of Microbiology, Molecular Genetics, and Immunology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Prabhakar Chalise
- Department of Biostatistics and Data Science, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Margaret L Axelrod
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Justin M Balko
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Pathology Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Kent W Hunter
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | | | - Carol A Lange
- Department of Medicine (Hematology, Oncology, and Transplantation), University of Minnesota Cancer Center, Minneapolis, Minnesota, USA
- Department of Pharmacology, University of Minnesota Cancer Center, Minneapolis, Minnesota, USA
| | - Christy R Hagan
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, Kansas, USA
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
15
|
Kim YS, Yang SC, Park M, Choi Y, DeMayo FJ, Lydon JP, Kim H, Lim HJ, Song H. Different Cre systems induce differential microRNA landscapes and abnormalities in the female reproductive tracts of Dgcr8 conditional knockout mice. Cell Prolif 2021; 54:e12996. [PMID: 33496365 PMCID: PMC7941225 DOI: 10.1111/cpr.12996] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 01/05/2021] [Accepted: 01/07/2021] [Indexed: 12/25/2022] Open
Abstract
OBJECTIVES The female reproductive tract comprises several different cell types. Using three representative Cre systems, we comparatively analysed the phenotypes of Dgcr8 conditional knockout (cKO) mice to understand the function of Dgcr8, involved in canonical microRNA biogenesis, in the female reproductive tract. MATERIALS AND METHODS Dgcr8f/f mice were crossed with Ltficre/+ , Amhr2cre/+ or PRcre/+ mice to produce mice deficient in Dgcr8 in epithelial (Dgcr8ed/ed ), mesenchymal (Dgcr8md/md ) and all the compartments (Dgcr8td/td ) in the female reproductive tract. Reproductive phenotypes were evaluated in Dgcr8 cKO mice. Uteri and/or oviducts were used for small RNA-seq, mRNA-seq, real-time RT-PCR, and/or morphologic and histological analyses. RESULT Dgcr8ed/ed mice did not exhibit any distinct defects, whereas Dgcr8md/md mice showed sub-fertility and oviductal smooth muscle deformities. Dgcr8td/td mice were infertile due to anovulation and acute inflammation in the female reproductive tract and suffered from an atrophic uterus with myometrial defects. The microRNAs and mRNAs related to immune modulation and/or smooth muscle growth were systemically altered in the Dgcr8td/td uterus. Expression profiles of dysregulated microRNAs and mRNAs in the Dgcr8td/td uterus were different from those in other genotypes in a Cre-dependent manner. CONCLUSIONS Dgcr8 deficiency with different Cre systems induces overlapping but distinct phenotypes as well as the profiles of microRNAs and their target mRNAs in the female reproductive tract, suggesting the importance of selecting the appropriate Cre driver to investigate the genes of interest.
Collapse
Affiliation(s)
- Yeon Sun Kim
- Department of Biomedical ScienceCHA UniversitySeongnamKorea
- Present address:
Division of reproductive sciencesDepartment of PediatricsCincinnati Children’s HospitalOHUSA
| | | | - Mira Park
- Department of Biomedical ScienceCHA UniversitySeongnamKorea
| | - Youngsok Choi
- Department of Stem Cell and Regenerative BiotechnologyKonkuk UniversitySeoulKorea
| | - Francesco J. DeMayo
- Department of Reproductive and Developmental Biology LaboratoryNational Institute of Environmental Health SciencesResearch Triangle ParkNCUSA
| | - John P. Lydon
- Department of Molecular and Cellular Biology and Center for Reproductive MedicineBaylor College of MedicineHoustonTXUSA
| | - Hye‐Ryun Kim
- Department of Biomedical ScienceCHA UniversitySeongnamKorea
| | - Hyunjung Jade Lim
- Department of Veterinary Medicine, School of Veterinary MedicineKonkuk UniversitySeoulKorea
| | - Haengseok Song
- Department of Biomedical ScienceCHA UniversitySeongnamKorea
| |
Collapse
|
16
|
Boonyaratanakornkit V, McGowan EM, Márquez-Garbán DC, Burton LP, Hamilton N, Pateetin P, Pietras RJ. Progesterone Receptor Signaling in the Breast Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1329:443-474. [PMID: 34664251 DOI: 10.1007/978-3-030-73119-9_21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The tumor microenvironment (TME) is a complex infrastructure composed of stromal, epithelial, and immune cells embedded in a vasculature ECM. The microenvironment surrounding mammary epithelium plays a critical role during the development and differentiation of the mammary gland, enabling the coordination of the complex multihormones and growth factor signaling processes. Progesterone/progesterone receptor paracrine signaling interactions in the microenvironment play vital roles in stem/progenitor cell function during normal breast development. In breast cancer, the female sex hormones, estrogen and progesterone, and growth factor signals are altered in the TME. Progesterone signaling modulates not only breast tumors but also the breast TME, leading to the activation of a series of cross-communications that are implicated in the genesis of breast cancers. This chapter reviews the evidence that progesterone and PR signaling modulates not only breast epitheliums but also the breast TME. Furthermore, crosstalk between estrogen and progesterone signaling affecting different cell types within the TME is discussed. A better understanding of how PR and progesterone affect the TME of breast cancer may lead to novel drugs or a therapeutic approach for the treatment of breast cancer shortly.
Collapse
Affiliation(s)
- Viroj Boonyaratanakornkit
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand.
- Age-Related Inflammation and Degeneration Research Unit, Chulalongkorn University, Bangkok, Thailand.
- Graduate Program in Clinical Biochemistry and Molecular Medicine, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand.
| | - Eileen M McGowan
- School of Life Sciences, University of Technology Sydney, Sydney, NSW, Australia
- Central Laboratory, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Diana C Márquez-Garbán
- UCLA Jonsson Comprehensive Cancer Center and Department of Medicine, Division of Hematology-Oncology, UCLA David Geffen School of Medicine, Los Angeles, CA, USA
| | - L P Burton
- UCLA Jonsson Comprehensive Cancer Center and Department of Medicine, Division of Hematology-Oncology, UCLA David Geffen School of Medicine, Los Angeles, CA, USA
| | - Nalo Hamilton
- UCLA Jonsson Comprehensive Cancer Center and Department of Medicine, Division of Hematology-Oncology, UCLA David Geffen School of Medicine, Los Angeles, CA, USA
| | - Prangwan Pateetin
- Graduate Program in Clinical Biochemistry and Molecular Medicine, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Richard J Pietras
- UCLA Jonsson Comprehensive Cancer Center and Department of Medicine, Division of Hematology-Oncology, UCLA David Geffen School of Medicine, Los Angeles, CA, USA
| |
Collapse
|
17
|
Lundberg R, Toft MF, Metzdorff SB, Hansen CHF, Licht TR, Bahl MI, Hansen AK. Human microbiota-transplanted C57BL/6 mice and offspring display reduced establishment of key bacteria and reduced immune stimulation compared to mouse microbiota-transplantation. Sci Rep 2020; 10:7805. [PMID: 32385373 PMCID: PMC7211022 DOI: 10.1038/s41598-020-64703-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 04/20/2020] [Indexed: 02/07/2023] Open
Abstract
Transplantation of germ-free (GF) mice with microbiota from mice or humans stimulates the intestinal immune system in disparate ways. We transplanted a human microbiota into GF C57BL/6 mice and a murine C57BL/6 microbiota into GF C57BL/6 mice and Swiss-Webster (SW) mice. Mice were bred to produce an offspring generation. 56% of the Operational Taxonomic Units (OTUs) present in the human donor microbiota established in the recipient mice, whereas 81% of the C57BL/6 OTUs established in the recipient C57BL/6 and SW mice. Anti-inflammatory bacteria such as Faecalibacterium and Bifidobacterium from humans were not transferred to mice. Expression of immune-related intestinal genes was lower in human microbiota-mice and not different between parent and offspring generation. Expression of intestinal barrier-related genes was slightly higher in human microbiota-mice. Cytokines and chemokines measured in plasma were differentially present in human and mouse microbiota-mice. Minor differences in microbiota and gene expression were found between transplanted mice of different genetics. It is concluded that important immune-regulating bacteria are lost when transplanting microbiota from humans to C57BL/6 mice, and that the established human microbiota is a weak stimulator of the murine immune system. The results are important for study design considerations in microbiota transplantation studies involving immunological parameters.
Collapse
Affiliation(s)
- Randi Lundberg
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 1871, Frederiksberg C, Denmark.
- Internal Research and Development, Taconic Biosciences, 4623, Lille Skensved, Denmark.
- Chr. Hansen, 2970, Hoersholm, Denmark.
| | - Martin F Toft
- Internal Research and Development, Taconic Biosciences, 4623, Lille Skensved, Denmark
- QM Diagnostics, 6534, AT Nijmegen, The Netherlands
| | - Stine B Metzdorff
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 1871, Frederiksberg C, Denmark
| | - Camilla H F Hansen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 1871, Frederiksberg C, Denmark
| | - Tine R Licht
- National Food Institute, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Martin I Bahl
- National Food Institute, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Axel K Hansen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 1871, Frederiksberg C, Denmark
| |
Collapse
|
18
|
Rackaityte E, Halkias J. Mechanisms of Fetal T Cell Tolerance and Immune Regulation. Front Immunol 2020; 11:588. [PMID: 32328065 PMCID: PMC7160249 DOI: 10.3389/fimmu.2020.00588] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 03/13/2020] [Indexed: 12/19/2022] Open
Abstract
The developing human fetus generates both tolerogenic and protective immune responses in response to the unique requirements of gestation. Thus, a successful human pregnancy depends on a fine balance between two opposing immunological forces: the semi-allogeneic fetus learns to tolerate both self- and maternal- antigens and, in parallel, develops protective immunity in preparation for birth. This critical window of immune development bridges prenatal immune tolerance with the need for postnatal environmental protection, resulting in a vulnerable neonatal period with heightened risk of infection. The fetal immune system is highly specialized to mediate this transition and thus serves a different function from that of the adult. Adaptive immune memory is already evident in the fetal intestine. Fetal T cells with pro-inflammatory potential are born in a tolerogenic environment and are tightly controlled by both cell-intrinsic and -extrinsic mechanisms, suggesting that compartmentalization and specialization, rather than immaturity, define the fetal immune system. Dysregulation of fetal tolerance generates an inflammatory response with deleterious effects to the pregnancy. This review aims to discuss the recent advances in our understanding of the cellular and molecular composition of fetal adaptive immunity and the mechanisms that govern T cell development and function. We also discuss the tolerance promoting environment that impacts fetal immunity and the consequences of its breakdown. A greater understanding of fetal mechanisms of immune activation and regulation has the potential to uncover novel paradigms of immune balance which may be leveraged to develop therapies for transplantation, autoimmune disease, and birth-associated inflammatory pathologies.
Collapse
Affiliation(s)
- Elze Rackaityte
- Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA, United States
| | - Joanna Halkias
- Division of Neonatology, Department of Pediatrics, University of California, San Francisco, San Francisco, CA, United States
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, UCSF, San Francisco, CA, United States
| |
Collapse
|
19
|
Lower activation of CD4+ memory T cells in preeclampsia compared to healthy pregnancies persists postpartum. J Reprod Immunol 2019; 136:102613. [DOI: 10.1016/j.jri.2019.102613] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 08/19/2019] [Accepted: 09/17/2019] [Indexed: 11/23/2022]
|
20
|
Pan Q, Chen X, Liao S, Chen X, Zhao C, Xu YZ, Liu HF. Updated advances of linking psychosocial factors and sex hormones with systemic lupus erythematosus susceptibility and development. PeerJ 2019; 7:e7179. [PMID: 31275761 PMCID: PMC6598654 DOI: 10.7717/peerj.7179] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 05/22/2019] [Indexed: 12/17/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a systemic autoimmune disease that primarily affects women, especially those of reproductive age. Genetics, environment, and gene-environment interactions play key roles in the development of SLE. Despite the numerous susceptibility genes of SLE identified to date, gene therapy is far from a clinical reality. Thus, more attention should be paid to the risk factors and underlying mechanisms of SLE. Currently, it is reported that psychosocial factors and sex hormones play vital roles in patients with SLE, which still need further investigated. The purpose of this review is to update the roles and mechanisms of psychosocial factors and sex hormones in the susceptibility and development of SLE. Based on review articles and reports in reputable peer-reviewed journals and government websites, this paper summarized psychosocial factors (e.g., alexithymia, depression, anxiety, negative emotions, and perceived stress) and sex hormones (e.g., estrogens, progesterone, androgens, and prolactin) involved in SLE. We further explore the mechanisms linking these factors with SLE susceptibility and development, which can guide the establishment of practical measures to benefit SLE patients and offer new ideas for therapeutic strategies.
Collapse
Affiliation(s)
- Qingjun Pan
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Division of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Xiaoqun Chen
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Division of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Shuzhen Liao
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Division of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Xiaocui Chen
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Division of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Chunfei Zhao
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Division of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Yong-Zhi Xu
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Division of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Hua-Feng Liu
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Division of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
21
|
Robertson SA, Green ES, Care AS, Moldenhauer LM, Prins JR, Hull ML, Barry SC, Dekker G. Therapeutic Potential of Regulatory T Cells in Preeclampsia-Opportunities and Challenges. Front Immunol 2019; 10:478. [PMID: 30984163 PMCID: PMC6448013 DOI: 10.3389/fimmu.2019.00478] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 02/21/2019] [Indexed: 12/26/2022] Open
Abstract
Inflammation is a central feature and is implicated as a causal factor in preeclampsia and other hypertensive disorders of pregnancy. Inflammatory mediators and leukocytes, which are elevated in peripheral blood and gestational tissues, contribute to the uterine vascular anomalies and compromised placental function that characterize particularly the severe, early onset form of disease. Regulatory T (Treg) cells are central mediators of pregnancy tolerance and direct other immune cells to counteract inflammation and promote robust placentation. Treg cells are commonly perturbed in preeclampsia, and there is evidence Treg cell insufficiency predates onset of symptoms. A causal role is implied by mouse studies showing sufficient numbers of functionally competent Treg cells must be present in the uterus from conception, to support maternal vascular adaptation and prevent later placental inflammatory pathology. Treg cells may therefore provide a tractable target for both preventative strategies and treatment interventions in preeclampsia. Steps to boost Treg cell activity require investigation and could be incorporated into pregnancy planning and preconception care. Pharmacological interventions developed to target Treg cells in autoimmune conditions warrant consideration for evaluation, utilizing rigorous clinical trial methodology, and ensuring safety is paramount. Emerging cell therapy tools involving in vitro Treg cell generation and/or expansion may in time become relevant. The success of preventative and therapeutic approaches will depend on resolving several challenges including developing informative diagnostic tests for Treg cell activity applicable before conception or during early pregnancy, selection of relevant patient subgroups, and identification of appropriate windows of gestation for intervention.
Collapse
Affiliation(s)
- Sarah A. Robertson
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Ella S. Green
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Alison S. Care
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Lachlan M. Moldenhauer
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | | | - M. Louise Hull
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
- Women's and Children's Hospital, Adelaide, SA, Australia
| | - Simon C. Barry
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Gustaaf Dekker
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
22
|
Yeung HY, Dendrou CA. Pregnancy Immunogenetics and Genomics: Implications for Pregnancy-Related Complications and Autoimmune Disease. Annu Rev Genomics Hum Genet 2019; 20:73-97. [PMID: 30848957 DOI: 10.1146/annurev-genom-083118-014943] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Pregnancy presents a singular physiological scenario during which the maternal immune system must accommodate the semiallogeneic fetus. Fluctuations between pro- and anti-inflammatory states are required throughout gestation to facilitate uterine tissue remodeling, fetal growth and development, and finally birth. Tolerance for the fetus must be established and maintained without fundamentally compromising the maternal immune system function, so that both the mother and fetus are protected from foreign insults. Here, we review our current understanding of how genetic variation at both maternal and fetal loci affects implantation and placenta formation, thereby determining the likelihood of a successful pregnancy outcome or the development of pregnancy-related complications. We also consider the impact of pregnancy on both the maternal and fetal systemic immune systems and the related implications for modulating ongoing autoimmune diseases and triggering their development.
Collapse
Affiliation(s)
- Hing-Yuen Yeung
- Nuffield Department of Medicine, Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, United Kingdom;
| | - Calliope A Dendrou
- Nuffield Department of Medicine, Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, United Kingdom;
| |
Collapse
|
23
|
Eaddy Norton A, Broyles AD. Drug allergy in children and adults: Is it the double X chromosome? Ann Allergy Asthma Immunol 2018; 122:148-155. [PMID: 30465863 DOI: 10.1016/j.anai.2018.11.014] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 11/11/2018] [Accepted: 11/12/2018] [Indexed: 12/18/2022]
Abstract
OBJECTIVE This article reviews the latest science and epidemiologic studies related to drug allergy in children and adults to explore possible mechanisms related to female propensity for drug allergy. DATA SOURCES PubMed literature review, focusing primarily on the last 5 years. STUDY SELECTIONS Articles reviewing the science behind female predisposition to atopic and asthmatic conditions and epidemiologic studies reviewing drug allergy and drug-induced anaphylaxis. RESULTS Despite adult female predilection for atopic conditions, few laboratory studies explore sex-specific mechanisms in atopic/allergic diseases, and most are focused on autoimmunity and asthma. Drug allergy is more frequently reported in adult females compared with adult males. Adult females are also more likely to have drug-induced anaphylaxis (DIA), although no clear sex predominance has been reported in fatal or severe DIA. Studies in children suggest the reverse picture, with prepubertal males more likely to have drug allergy and DIA than prepubertal girls. CONCLUSION Possible explanations for female predisposition for drug allergy are multifactorial and include disproportionate utilization of health care with more exposure to antibiotics or medications, genetic factors related to the X chromosome, epigenetic changes, and discrepant hormonal interactions with immune cells.
Collapse
Affiliation(s)
- Allison Eaddy Norton
- Vanderbilt Children's Hospital, Division of Pediatric Pulmonary, Allergy and Immunology, School of Medicine, Nashville, Tennessee
| | - Ana Dioun Broyles
- Boston Children's Hospital, Division of Allergy and Immunology, Boston, Massachusetts; Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
24
|
Li J, Hong X, Mesiano S, Muglia LJ, Wang X, Snyder M, Stevenson DK, Shaw GM. Natural Selection Has Differentiated the Progesterone Receptor among Human Populations. Am J Hum Genet 2018; 103:45-57. [PMID: 29937092 PMCID: PMC6035283 DOI: 10.1016/j.ajhg.2018.05.009] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 05/21/2018] [Indexed: 11/23/2022] Open
Abstract
The progesterone receptor (PGR) plays a central role in maintaining pregnancy and is significantly associated with medical conditions such as preterm birth that affects 12.6% of all the births in U.S. PGR has been evolving rapidly since the common ancestor of human and chimpanzee, and we herein investigated evolutionary dynamics of PGR during recent human migration and population differentiation. Our study revealed substantial population differentiation at the PGR locus driven by natural selection, where very recent positive selection in East Asians has substantially decreased its genetic diversity by nearly fixing evolutionarily novel alleles. On the contrary, in European populations, the PGR locus has been promoted to a highly polymorphic state likely due to balancing selection. Integrating transcriptome data across multiple tissue types together with large-scale genome-wide association data for preterm birth, our study demonstrated the consequence of the selection event in East Asians on remodeling PGR expression specifically in the ovary and determined a significant association of early spontaneous preterm birth with the evolutionarily selected variants. To reconstruct its evolutionary trajectory on the human lineage, we observed substantial differentiation between modern and archaic humans at the PGR locus, including fixation of a deleterious missense allele in the Neanderthal genome that was later introgressed in modern human populations. Taken together, our study revealed substantial evolutionary innovation in PGR even during very recent human evolution, and its different forms among human populations likely result in differential susceptibility to progesterone-associated disease conditions including preterm birth.
Collapse
Affiliation(s)
- Jingjing Li
- The March of Dimes Prematurity Research Center, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA; Stanford Center for Genomics and Personalized Medicine, Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Xiumei Hong
- Center on the Early Life Origins of Disease, Department of Population, Family and Reproductive Health, Bloomberg School of Public Health, John Hopkins University, Baltimore, MD 21205, USA
| | - Sam Mesiano
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Louis J Muglia
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Departments of Pediatrics and Obstetrics and Gynecology, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Xiaobin Wang
- Center on the Early Life Origins of Disease, Department of Population, Family and Reproductive Health, Bloomberg School of Public Health, John Hopkins University, Baltimore, MD 21205, USA; Department of Pediatrics, John Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Michael Snyder
- Stanford Center for Genomics and Personalized Medicine, Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - David K Stevenson
- The March of Dimes Prematurity Research Center, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - Gary M Shaw
- The March of Dimes Prematurity Research Center, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
25
|
Recalde G, Moreno-Sosa T, Yúdica F, Quintero CA, Sánchez MB, Jahn GA, Kalergis AM, Mackern-Oberti JP. Contribution of sex steroids and prolactin to the modulation of T and B cells during autoimmunity. Autoimmun Rev 2018. [DOI: 10.1016/j.autrev.2018.03.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
26
|
Liu HX, Chen T, Wen X, Qu W, Liu S, Yan HY, Hou LF, Ping J. Maternal Glucocorticoid Elevation and Associated Fetal Thymocyte Apoptosis are Involved in Immune Disorders of Prenatal Caffeine Exposed Offspring Mice. Sci Rep 2017; 7:13746. [PMID: 29062003 PMCID: PMC5653827 DOI: 10.1038/s41598-017-14103-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 10/03/2017] [Indexed: 02/03/2023] Open
Abstract
Our previous study showed that prenatal caffeine exposure (PCE) could induce intrauterine growth retardation (IUGR) and glucocorticoid elevation in the fetus. Researchers suggested that IUGR is a risk factor for T helper cell (Th)1/Th2 deviation. However, whether PCE can induce these immune disorders and the underlying mechanisms of that induction remain unknown. This study aimed to observe the effects of PCE on the Th1/Th2 balance in offspring and further explore the developmental origin mechanisms from the perspective of glucocorticoid overexposure-induced thymocyte apoptosis. An IUGR model was established by caffeine administration from gestational day (GD) 9 to GD 18, and the offspring were immunized on postnatal day (PND) 42. The results show that maternal glucocorticoid overexposure increased fetal thymocyte apoptosis by activating both the Fas-mediated and the Bim-regulated apoptotic pathways. After birth, accelerated thymocyte apoptosis and Th1 suppression were also found in the PCE offspring at PND 14 and PND 49. Moreover, the PCE offspring showed immune disorders after immunization, manifesting as increased IgG1/IgG2a ratio and IL-4 production in the serum. In conclusion, PCE could induce fetal overexposure to maternal glucocorticoids and increase thymocyte apoptosis, which could persist into postnatal life and be implicated in Th1 inhibition and further immune disorders.
Collapse
Affiliation(s)
- Han-Xiao Liu
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China
| | - Ting Chen
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China
| | - Xiao Wen
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China
| | - Wen Qu
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China
| | - Sha Liu
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China
| | - Hui-Yi Yan
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China
| | - Li-Fang Hou
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China
| | - Jie Ping
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China.
| |
Collapse
|
27
|
Hall OJ, Klein SL. Progesterone-based compounds affect immune responses and susceptibility to infections at diverse mucosal sites. Mucosal Immunol 2017; 10:1097-1107. [PMID: 28401937 DOI: 10.1038/mi.2017.35] [Citation(s) in RCA: 109] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 03/04/2017] [Indexed: 02/04/2023]
Abstract
Over 100 million women worldwide are currently on progesterone-based contraceptives to improve their health outcomes through reduced maternal mortality and family planning. In addition to their role in reproduction, progesterone-based compounds modulate immune responses throughout the body, particularly at mucosal sites. By binding to receptors located in immune cells, including natural killer cells, macrophages, dendritic cells, and T cells, as well in non-immune cells, such as epithelial and endothelial cells, progesterone-based compounds alter cellular signaling and activity to affect the outcome of infections at diverse mucosal sites, including the genital, gastrointestinal, and respiratory tracts. As the use of progesterone-based compounds, in the form of contraceptives and hormone-based therapies, continue to increase worldwide, greater consideration should be given to how the immunomodulatory effects these compounds alter the outcome of diseases at mucosal sites beyond the reproductive tract, which has profound implications for women's health.
Collapse
Affiliation(s)
- Olivia J Hall
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland USA
| | - Sabra L Klein
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland USA
| |
Collapse
|
28
|
Monteiro C, Kasahara TM, Castro JR, Sacramento PM, Hygino J, Centurião N, Cassano T, Lopes LMF, Leite S, Silva VG, Gupta S, Bento CAM. Pregnancy favors the expansion of circulating functional follicular helper T Cells. J Reprod Immunol 2017; 121:1-10. [PMID: 28482188 DOI: 10.1016/j.jri.2017.04.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2017] [Accepted: 04/25/2017] [Indexed: 11/29/2022]
Abstract
Pregnancy favors antibody production, and some evidence has suggested a direct effect of estrogen on B cells. The impact of pregnancy on circulating follicular helper T (TFH) cells, typically identified by the expression of CD45RO and CXCR5, has not been previously investigated. Here, the percentage of TFH cells, co-expressing or not PD-1, ICOS, or CXCR3 markers was significantly higher in pregnant women (PW) as compared with non-pregnant ones (nPW). Furthermore, the percentage of CXCR3+ TFH cells able to produce IL-6, IL-21, and IL-10 was significantly higher in PW than nPW. Interestingly, anti-CMV and anti-HBs antibody titers were significantly higher in the plasma of PW and were directly correlated with IL-21-producing CXCR3+ TFH cells. Finally, peripheral estrogen levels, but not progesterone, were positively related to either PD-1+ CXCR3+ TFH cells or plasma anti-CMV and anti-HBs IgG antibodies. In summary, our data suggests a positive effect of pregnancy on the proportion of CD4+ T cell subset specialized in helping B cells. This phenomenon, which could be related to the high estrogen levels produced during pregnancy, may help to explain why pregnancy favor humoral immunity.
Collapse
Affiliation(s)
- Clarice Monteiro
- Department of Microbiology and Parasitology/Federal University of the State of Rio de Janeiro, Brazil; Department of Microbiology, Immunology and Parasitology, UERJ, Rio de Janeiro, Brazil
| | - Taissa M Kasahara
- Department of Microbiology and Parasitology/Federal University of the State of Rio de Janeiro, Brazil; Department of Microbiology, Immunology and Parasitology, UERJ, Rio de Janeiro, Brazil
| | - José Roberto Castro
- Department of Microbiology and Parasitology/Federal University of the State of Rio de Janeiro, Brazil
| | - Priscila M Sacramento
- Department of Microbiology and Parasitology/Federal University of the State of Rio de Janeiro, Brazil
| | - Joana Hygino
- Department of Microbiology and Parasitology/Federal University of the State of Rio de Janeiro, Brazil
| | - Newton Centurião
- Department of Microbiology and Parasitology/Federal University of the State of Rio de Janeiro, Brazil
| | - Tatiane Cassano
- Department of Microbiology and Parasitology/Federal University of the State of Rio de Janeiro, Brazil
| | - Lana M Ferreira Lopes
- Department of Microbiology and Parasitology/Federal University of the State of Rio de Janeiro, Brazil
| | - Simone Leite
- Fernandes Figueira Institute/IOC, Rio de Janeiro, Brazil
| | | | | | - Cleonice A M Bento
- Department of Microbiology and Parasitology/Federal University of the State of Rio de Janeiro, Brazil.
| |
Collapse
|
29
|
Sex and age as determinants of rat T-cell phenotypic characteristics: influence of peripubertal gonadectomy. Mol Cell Biochem 2017; 431:169-185. [PMID: 28281185 DOI: 10.1007/s11010-017-2989-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 02/24/2017] [Indexed: 12/19/2022]
Abstract
The study examined the influence of age, sex and peripubertal gonadectomy on a set of T-cell phenotypic parameters. Rats of both sexes were gonadectomised at the age of 1 month and peripheral blood and spleen T lymphocytes from non-gonadectomised and gonadectomised 3- and 11-month-old rats were examined for the expression of differentiation/activation (CD90/CD45RC) and immunoregulatory markers. Peripheral blood T lymphocytes from non-gonadectomised rats showed age-dependent sexual dimorphisms in (1) total count (lower in female than male 11-month-old rats); (2) CD4+:CD8 + cell ratio (higher in female than male rats of both ages); (3) the proportion of recent thymic emigrants in CD8 + T cells (lower in female than male 3-month-old rats) and (4) the proportions of mature naïve and memory/activated cells (irrespective of age, the proportion of naïve cells was higher, whereas that of memory/activated cells was lower in females). Gonadectomy influenced magnitudes or direction of these sex differences. Additionally, sex differences in peripheral blood T-lymphocyte parameters did not fully correspond to those observed in T-splenocyte parameters, suggesting the compartment-specific regulation of the major T-cell subpopulations' and their subsets' composition. Furthermore, there was no sexual dimorphism in the proportion of either CD25 + Foxp3 + cells among CD4 + or CD161+ (NKT) cells within CD8 + T lymphocytes. However, there was gonadal hormone-independent age-associated sexual dimorphism in the proportion of CD161 + cells (NKT cells) in CD8 + T splenocytes. Overall, the study revealed age-dependent variations in sexual dimorphisms in T-cell parameters relevant for immune response efficacy and showed that they are T-cell compartment-specific and partly gonadal hormone-related.
Collapse
|
30
|
Kieffer TE, Faas MM, Scherjon SA, Prins JR. Pregnancy persistently affects memory T cell populations. J Reprod Immunol 2017; 119:1-8. [DOI: 10.1016/j.jri.2016.11.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2016] [Revised: 10/28/2016] [Accepted: 11/10/2016] [Indexed: 11/28/2022]
|
31
|
Nair RR, Verma P, Singh K. Immune-endocrine crosstalk during pregnancy. Gen Comp Endocrinol 2017; 242:18-23. [PMID: 26965955 DOI: 10.1016/j.ygcen.2016.03.003] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2015] [Revised: 02/25/2016] [Accepted: 03/05/2016] [Indexed: 12/27/2022]
Abstract
The success of pregnancy depends mostly on a synchronized immune-endocrine crosstalk at the maternal-fetal interface. Hormones are important in terms of maintaining the suitable environment and sufficient nutrition for the developing fetus. They also play a major role during the process of parturition and lactation. Maternal immunomodulation is important for the tolerance of semiallogeneic fetus. This is achieved in concert with a variety of endocrine stimulation. Estrogen, progesterone, and Human Chorionic Gonadotropin play a major role in immune modulation during pregnancy. Hormones modulate B cells, dendritic cells, uterine natural killer cells, macrophages, neutrophils to adopt fetal friendly immune phenotypes. Recently the use of hormones in assisted reproductive technology has been found to improve the pregnancy outcome. The present review focuses on the pregnancy-related hormones, their role in immunomodulation for successful pregnancy outcome. This also shed light on the immune-endocrine crosstalk at maternal-fetal interface during pregnancy.
Collapse
Affiliation(s)
- Rohini R Nair
- Division of Genetics and Cell Biology, San Raffaele University and Institute, Milano, Italy
| | - Priyanka Verma
- Department of Molecular & Human Genetics, Banaras Hindu University, Varanasi 221005, India
| | - Kiran Singh
- Department of Molecular & Human Genetics, Banaras Hindu University, Varanasi 221005, India.
| |
Collapse
|
32
|
Rolf L, Damoiseaux J, Hupperts R, Huitinga I, Smolders J. Network of nuclear receptor ligands in multiple sclerosis: Common pathways and interactions of sex-steroids, corticosteroids and vitamin D3-derived molecules. Autoimmun Rev 2016; 15:900-10. [DOI: 10.1016/j.autrev.2016.07.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 06/08/2016] [Indexed: 01/12/2023]
|
33
|
Kim YS, Kim HR, Kim H, Yang SC, Park M, Yoon JA, Lim HJ, Hong SH, DeMayo FJ, Lydon JP, Choi Y, Lee DR, Song H. Deficiency in DGCR8-dependent canonical microRNAs causes infertility due to multiple abnormalities during uterine development in mice. Sci Rep 2016; 6:20242. [PMID: 26833131 PMCID: PMC4735737 DOI: 10.1038/srep20242] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 12/29/2015] [Indexed: 12/25/2022] Open
Abstract
DGCR8 is an RNA-binding protein that interacts with DROSHA to produce pre-microRNA in the nucleus, while DICER generates not only mature microRNA, but also endogenous small interfering RNAs in the cytoplasm. Here, we produced Dgcr8 conditional knock-out mice using progesterone receptor (PR)-Cre (Dgcr8d/d) and demonstrated that canonical microRNAs dependent on the DROSHA-DGCR8 complex are required for uterine development as well as female fertility in mice. Adult Dgcr8d/d females neither underwent regular reproductive cycles nor produced pups, whereas administration of exogenous gonadotropins induced normal ovulation in these mice. Interestingly, immune cells associated with acute inflammation aberrantly infiltrated into reproductive organs of pregnant Dgcr8d/d mice. Regarding uterine development, multiple uterine abnormalities were noticeable at 4 weeks of age when PR is significantly increased, and the severity of these deformities increased over time. Gland formation and myometrial layers were significantly reduced, and the stromal cell compartment did not expand and became atrophic during uterine development in these mice. These results were consistent with aberrantly reduced stromal cell proliferation and completely failed decidualization. Collectively, we suggest that DGCR8-dependent canonical microRNAs are essential for uterine development and physiological processes such as proper immune modulation, reproductive cycle, and steroid hormone responsiveness in mice.
Collapse
Affiliation(s)
- Yeon Sun Kim
- Department of Biomedical Science, CHA University, Seongnam, Gyeonggi, 463-400 Korea
| | - Hye-Ryun Kim
- Department of Biomedical Science, CHA University, Seongnam, Gyeonggi, 463-400 Korea
| | - Hyongbum Kim
- Department of Pharmacology and Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 120-752, Korea
| | - Seung Chel Yang
- Department of Biomedical Science, CHA University, Seongnam, Gyeonggi, 463-400 Korea
| | - Mira Park
- Department of Biomedical Science, CHA University, Seongnam, Gyeonggi, 463-400 Korea
| | - Jung Ah Yoon
- Fertility Center of CHA Gangnam Medical Center, CHA University, Seoul, Korea
| | - Hyunjung J Lim
- Department of Veterinary Medicine, College of Veterinary Medicine, Konkuk University, Seoul, 143-701 Korea
| | - Seok-Ho Hong
- Department of Internal Medicine, School of Medicine, Stem Cell Institute, Kangwon National University, Chuncheon, Kangwon 200-701, Korea
| | - Francesco J DeMayo
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, 77030 USA
| | - John P Lydon
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, 77030 USA
| | - Youngsok Choi
- Department of Biomedical Science, CHA University, Seongnam, Gyeonggi, 463-400 Korea
| | - Dong Ryul Lee
- Department of Biomedical Science, CHA University, Seongnam, Gyeonggi, 463-400 Korea.,Fertility Center of CHA Gangnam Medical Center, CHA University, Seoul, Korea
| | - Haengseok Song
- Department of Biomedical Science, CHA University, Seongnam, Gyeonggi, 463-400 Korea.,Fertility Center of CHA Gangnam Medical Center, CHA University, Seoul, Korea
| |
Collapse
|
34
|
Abstract
Pregnancy is an immunological paradox that implies that a semi-allogeneic fetus is not rejected by the maternal immune system, from implantation of the embryo to delivery. Progesterone (P4), estradiol (E2) and human chorionic gonadotropin (hCG), contribute to the transformation of immune cells in a transient tolerance state, necessary to the maintenance of pregnancy. The effects of pregnancy hormones depend probably of their maternal plasma level. hCG is dangerous at high concentrations because it can stimulate autoantibodies production, whereas in physiological concentrations, hCG, P4 and E2 upregulate immune response expanding regulatory T and B cells, allowing the fetus to grow within the maternal uterus in a protective environment. A second example of fetal-maternal relation found recently is the role of maternal nutrition on development of the fetal hypothalamic neurons. Experiments in mice fed on a high fat diet reveal a critical timing when altered maternal metabolism affect formation of hypothalamic neurocircuits of the offspring and predispose him to long-term metabolic disorders.
Collapse
|
35
|
Taves MD, Losie JA, Rahim T, Schmidt KL, Sandkam BA, Ma C, Silversides FG, Soma KK. Locally elevated cortisol in lymphoid organs of the developing zebra finch but not Japanese quail or chicken. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2016; 54:116-125. [PMID: 26366679 DOI: 10.1016/j.dci.2015.09.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Revised: 08/28/2015] [Accepted: 09/09/2015] [Indexed: 06/05/2023]
Abstract
Glucocorticoids are important for production of functional lymphocytes and immunity. In altricial neonates, adrenal glands are unresponsive and local glucocorticoid synthesis in lymphoid organs may be necessary to support lymphocyte development. Precocial neonates, in contrast, have fully responsive adrenal glucocorticoid production, and lymphoid glucocorticoid synthesis may not be necessary. Here, we found that in altricial zebra finch hatchlings, lymphoid organs had dramatically elevated endogenous glucocorticoid (and precursor) levels compared to levels in circulating blood. Furthermore, while avian adrenals produce corticosterone, finch lymphoid organs had much higher levels of cortisol, an unexpected glucocorticoid in birds. In contrast, precocial Japanese quail and chicken offspring did not have locally elevated lymphoid glucocorticoid levels, nor did their lymphoid organs contain high proportions of cortisol. These results show that lymphoid glucocorticoids differ in identity, concentration, and possibly source, in hatchlings of three different bird species. Locally-regulated glucocorticoids might have species-specific roles in immune development.
Collapse
Affiliation(s)
- Matthew D Taves
- Dept. of Psychology, University of British Columbia, Vancouver, BC, Canada; Dept. of Zoology, University of British Columbia, Vancouver, BC, Canada.
| | - Jennifer A Losie
- Dept. of Psychology, University of British Columbia, Vancouver, BC, Canada
| | - Titissa Rahim
- Dept. of Psychology, University of British Columbia, Vancouver, BC, Canada
| | - Kim L Schmidt
- Dept. of Biology, University of Western Ontario, London, ON, Canada; Advanced Facility for Avian Research, University of Western Ontario, London, ON, Canada
| | - Benjamin A Sandkam
- Department of Biological Sciences, Simon Fraser University, Vancouver, BC, Canada
| | - Chunqi Ma
- Dept. of Psychology, University of British Columbia, Vancouver, BC, Canada
| | | | - Kiran K Soma
- Dept. of Psychology, University of British Columbia, Vancouver, BC, Canada; Dept. of Zoology, University of British Columbia, Vancouver, BC, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
36
|
Wolfson ML, Schander JA, Bariani MV, Correa F, Franchi AM. Progesterone modulates the LPS-induced nitric oxide production by a progesterone-receptor independent mechanism. Eur J Pharmacol 2015; 769:110-6. [DOI: 10.1016/j.ejphar.2015.11.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Revised: 10/30/2015] [Accepted: 11/02/2015] [Indexed: 12/15/2022]
|
37
|
Jasienska G, Ellison PT, Galbarczyk A, Jasienski M, Kalemba-Drozdz M, Kapiszewska M, Nenko I, Thune I, Ziomkiewicz A. Apolipoprotein E (ApoE) polymorphism is related to differences in potential fertility in women: a case of antagonistic pleiotropy? Proc Biol Sci 2015; 282:20142395. [PMID: 25673673 DOI: 10.1098/rspb.2014.2395] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The alleles that are detrimental to health, especially in older age, are thought to persist in populations because they also confer some benefits for individuals (through antagonistic pleiotropy). The ApoE4 allele at the ApoE locus, encoding apolipoprotein E (ApoE), significantly increases risk of poor health, and yet it is present in many populations at relatively high frequencies. Why has it not been replaced by natural selection with the health-beneficial ApoE3 allele? ApoE is a major supplier of cholesterol precursor for the production of ovarian oestrogen and progesterone, thus ApoE has been suggested as the potential candidate gene that may cause variation in reproductive performance. Our results support this hypothesis showing that in 117 regularly menstruating women those with genotypes with at least one ApoE4 allele had significantly higher levels of mean luteal progesterone (144.21 pmol l(-1)) than women with genotypes without ApoE4 (120.49 pmol l(-1)), which indicates higher potential fertility. The hormonal profiles were based on daily data for entire menstrual cycles. We suggest that the finding of higher progesterone in women with ApoE4 allele could provide first strong evidence for an evolutionary mechanism of maintaining the ancestral and health-worsening ApoE4 allele in human populations.
Collapse
Affiliation(s)
- Grazyna Jasienska
- Department of Environmental Health, Jagiellonian University Medical College, Krakow, Poland
| | - Peter T Ellison
- Department of Human Evolutionary Biology, Harvard University, Cambridge, MA, USA
| | - Andrzej Galbarczyk
- Department of Environmental Health, Jagiellonian University Medical College, Krakow, Poland
| | - Michal Jasienski
- Center for Innovatics, Nowy Sacz Business School-National-Louis University, Zielona 27, 33-300 Nowy Sacz, Poland
| | | | | | - Ilona Nenko
- Department of Environmental Health, Jagiellonian University Medical College, Krakow, Poland
| | - Inger Thune
- Department of Community Medicine, Faculty of Health Sciences, University of Tromsø, Tromsø, Norway The Cancer Center, Oslo University Hospital, Oslo, Norway
| | - Anna Ziomkiewicz
- Polish Academy of Sciences, Unit of Anthropology in Wroclaw, Poland
| |
Collapse
|
38
|
Bishop CV, Xu F, Molskness TA, Stouffer RL, Hennebold JD. Dynamics of Immune Cell Types Within the Macaque Corpus Luteum During the Menstrual Cycle: Role of Progesterone. Biol Reprod 2015; 93:112. [PMID: 26400401 DOI: 10.1095/biolreprod.115.132753] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 09/18/2015] [Indexed: 12/20/2022] Open
Abstract
The goal of the current study was to characterize the immune cell types within the primate corpus luteum (CL). Luteal tissue was collected from rhesus females at discrete intervals during the luteal phase of the natural menstrual cycle. Dispersed cells were incubated with fluorescently labeled antibodies specific for the immune cell surface proteins CD11b (neutrophils and monocytes/macrophages), CD14 (monocytes/macrophages), CD16 (natural killer [NK] cells), CD20 (B-lymphocytes), and CD3epsilon (T-lymphocytes) for analysis by flow cytometry. Numbers of CD11b-positive (CD11b(+)) and CD14(+) cells increased significantly 3 to 4 days after serum progesterone (P4) concentrations declined below 0.3 ng/ml. CD16(+) cells were the most abundant immune cell type in CL during the mid and mid-late luteal phases and were 3-fold increased 3 to 4 days after serum P4 decreased to baseline levels. CD3epsilon(+) cells tended to increase 3 to 4 days after P4 decline. To determine whether immune cells were upregulated by the loss of luteotropic (LH) support or through loss of LH-dependent steroid milieu, monkeys were assigned to 4 groups: control (no treatment), the GnRH antagonist Antide, Antide plus synthetic progestin (R5020), or Antide plus the estrogen receptor agonists diarylpropionitrile (DPN)/propyl-pyrazole-triol (PPT) during the mid-late luteal phase. Antide treatment increased the numbers of CD11b(+) and CD14(+) cells, whereas progestin, but not estrogen, replacement suppressed the numbers of CD11b(+), CD14(+), and CD16(+) cells. Neither Antide nor steroid replacement altered numbers of CD3epsilon(+) cells. These data suggest that increased numbers of innate immune cells in primate CL after P4 synthesis declines play a role in onset of structural regression of primate CL.
Collapse
Affiliation(s)
- Cecily V Bishop
- Division of Reproductive and Developmental Sciences, Oregon Health & Science University, Portland, Oregon
| | - Fuhua Xu
- Division of Reproductive and Developmental Sciences, Oregon Health & Science University, Portland, Oregon
| | - Theodore A Molskness
- Division of Reproductive and Developmental Sciences, Oregon Health & Science University, Portland, Oregon
| | - Richard L Stouffer
- Division of Reproductive and Developmental Sciences, Oregon Health & Science University, Portland, Oregon Division of Obstetrics and Gynecology, Oregon Health & Science University, Portland, Oregon
| | - Jon D Hennebold
- Division of Reproductive and Developmental Sciences, Oregon Health & Science University, Portland, Oregon Division of Obstetrics and Gynecology, Oregon Health & Science University, Portland, Oregon
| |
Collapse
|
39
|
Lissauer D, Eldershaw SA, Inman CF, Coomarasamy A, Moss PAH, Kilby MD. Progesterone promotes maternal-fetal tolerance by reducing human maternal T-cell polyfunctionality and inducing a specific cytokine profile. Eur J Immunol 2015; 45:2858-72. [PMID: 26249148 PMCID: PMC4833190 DOI: 10.1002/eji.201445404] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Revised: 05/19/2015] [Accepted: 07/31/2015] [Indexed: 11/10/2022]
Abstract
Progesterone is a steroid hormone essential for the maintenance of human pregnancy, and its actions are thought to include promoting maternal immune tolerance of the semiallogenic fetus. We report that exposure of maternal T cells to progesterone at physiological doses induced a unique skewing of the cytokine production profile of CD4+ and CD8+ T cells, with reductions not only in potentially deleterious IFN‐γ and TNF‐α production but also in IL‐10 and IL‐5. Conversely, production of IL‐4 was increased. Maternal T cells also became less polyfunctional, focussing cytokine production toward profiles including IL‐4. This was accompanied by reduced T‐cell proliferation. Using fetal and viral antigen‐specific CD8+ T‐cell clones, we confirmed that this as a direct, nonantigen‐specific effect. Yet human T cells lacked conventional nuclear progesterone receptors, implicating a membrane progesterone receptor. CD4+ and CD8+ T cells responded to progesterone in a dose‐dependent manner, with subtle effects at concentrations comparable to those in maternal blood, but profound effects at concentrations similar to those at the maternal–fetal interface. This characterization of how progesterone modulates T‐cell function is important in understanding the normal biology of pregnancy and informing the rational use of progesterone therapy in pregnancies at risk of fetal loss.
Collapse
Affiliation(s)
- David Lissauer
- Centre for Women's and Children's Health, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Suzy A Eldershaw
- School of Cancer Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Charlotte F Inman
- School of Cancer Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Aravinthan Coomarasamy
- Centre for Women's and Children's Health, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Paul A H Moss
- School of Cancer Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Mark D Kilby
- Centre for Women's and Children's Health, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| |
Collapse
|
40
|
Zheng J, Zhang H, Bao K, Gao W, Xu C, Xia C. Preparation of Monoclonal Antibodies Against Bovine Progesterone. Monoclon Antib Immunodiagn Immunother 2015; 34:275-7. [PMID: 26301932 DOI: 10.1089/mab.2015.0001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Female, 8-week-old BALB/c mice were immunized with the progesterone complete antigen (P4-BSA). Two monoclonal antibodies (MAbs) against P4, named 8G6 and 11A11, were obtained by the lymphocyte hybridoma technique. The titers of the cell culture supernatant and ascitic fluid of MAb 8G6 were 1: 16,000 and 1:512,000, respectively, and the titers of the cell culture supernatant and ascitic fluid of MAb 11A11 were 1: 8000 and 1:256,000, respectively. The subtypes of the MAbs 8G6 and 11A11 were IgMκ. In specific analysis, both of the two MAbs did not react with estradiol. These data demonstrated that the MAbs 8G6 and 11A11 have a potential use for developing diagnostic reagents of progesterone.
Collapse
Affiliation(s)
- Jiasan Zheng
- 1 College of Animal Science and Veterinary Medicine, HeiLongjiang Bayi Agricultural University , Daqing, P.R. China
| | - Hongyou Zhang
- 1 College of Animal Science and Veterinary Medicine, HeiLongjiang Bayi Agricultural University , Daqing, P.R. China
| | - Kai Bao
- 1 College of Animal Science and Veterinary Medicine, HeiLongjiang Bayi Agricultural University , Daqing, P.R. China
| | - Weiming Gao
- 1 College of Animal Science and Veterinary Medicine, HeiLongjiang Bayi Agricultural University , Daqing, P.R. China
| | - Chuang Xu
- 1 College of Animal Science and Veterinary Medicine, HeiLongjiang Bayi Agricultural University , Daqing, P.R. China
| | - Cheng Xia
- 1 College of Animal Science and Veterinary Medicine, HeiLongjiang Bayi Agricultural University , Daqing, P.R. China .,2 Synergetic Innovation Center of Food Safety and Nutrition, Northeast Agricultural University , Harbin, P.R. China
| |
Collapse
|
41
|
Tan IJ, Peeva E, Zandman-Goddard G. Hormonal modulation of the immune system — A spotlight on the role of progestogens. Autoimmun Rev 2015; 14:536-42. [DOI: 10.1016/j.autrev.2015.02.004] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2014] [Accepted: 01/25/2015] [Indexed: 01/14/2023]
|
42
|
Guthrie BL, Introini A, Roxby AC, Choi RY, Bosire R, Lohman-Payne B, Hirbod T, Farquhar C, Broliden K. Depot Medroxyprogesterone Acetate Use Is Associated With Elevated Innate Immune Effector Molecules in Cervicovaginal Secretions of HIV-1-Uninfected Women. J Acquir Immune Defic Syndr 2015; 69:1-10. [PMID: 25622059 PMCID: PMC4424097 DOI: 10.1097/qai.0000000000000533] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
OBJECTIVE The effects of sex hormones on the immune defenses of the female genital mucosa and its susceptibility to infections are poorly understood. The injectable hormonal contraceptive depot medroxyprogesterone acetate (DMPA) may increase the risk for HIV-1 acquisition. We assessed the local concentration in the female genital mucosa of cationic polypeptides with reported antiviral activity in relation to DMPA use. METHODS HIV-1-uninfected women were recruited from among couples testing for HIV in Nairobi, Kenya. Cervicovaginal secretion samples were collected, and the concentrations of HNP1-3, LL-37, lactoferrin, HBD-2, and SLPI were measured by enzyme-linked immunosorbent assays. Levels of cationic polypeptides in cervicovaginal secretions were compared between women who were not using hormonal contraception and those using DMPA, oral, or implantable contraception. RESULTS Among 228 women, 165 (72%) reported not using hormonal contraception at enrollment, 41 (18%) used DMPA, 16 (7%) used an oral contraceptive, and 6 (3%) used a contraceptive implant. Compared with nonusers of hormonal contraception, DMPA users had significantly higher mean levels of HNP1-3 (2.38 vs. 2.04 log₁₀ ng/mL; P = 0.024), LL-37 (0.81 vs. 0.40 log10 ng/mL; P = 0.027), and lactoferrin (3.03 vs. 2.60 log₁₀ ng/mL; P = 0.002), whereas SLPI and HBD-2 were similar. CONCLUSIONS Although all analyzed cationic polypeptides have intrinsic antiviral capacity, their interaction and cumulative effect on female genital mucosa susceptibility to infections in vivo has yet to be unraveled. This study suggests a potential mechanism underlying the effect of DMPA on the innate immune defenses, providing a rationale to investigate its effect on HIV-1 acquisition risk.
Collapse
Affiliation(s)
- Brandon L. Guthrie
- Department of Global Health, University of Washington, Seattle, Washington, USA
- Department of Epidemiology, University of Washington, Seattle, Washington, USA
| | - Andrea Introini
- Department of Medicine Solna, Unit of Infectious Diseases, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Alison C. Roxby
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Robert Y. Choi
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Rose Bosire
- Center for Public Health Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Barbara Lohman-Payne
- Department of Global Health, University of Washington, Seattle, Washington, USA
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Taha Hirbod
- Department of Medicine Solna, Unit of Infectious Diseases, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Carey Farquhar
- Department of Global Health, University of Washington, Seattle, Washington, USA
- Department of Epidemiology, University of Washington, Seattle, Washington, USA
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Kristina Broliden
- Department of Medicine Solna, Unit of Infectious Diseases, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
43
|
Wong AH, Agrawal N, Hughes GC. Altered IgG autoantibody levels and CD4(+) T cell subsets in lupus-prone Nba2 mice lacking the nuclear progesterone receptor. Autoimmunity 2015; 48:389-401. [PMID: 25857203 DOI: 10.3109/08916934.2015.1030613] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Important interactions between female reproduction and autoimmunity are suggested by the female-predominance of systemic lupus erythematosus (SLE) and other autoimmune diseases and the amelioration of certain autoimmune diseases during pregnancy. Sexually dimorphic risk of developing SLE involves modulation of genetic risk by environmental factors, sex hormones and non-hormonal factors encoded on the sex chromosomes. In some lupus models, estrogen, via estrogen receptor alpha (ER-α), enhances production of highly pathogenic IgG2a/c autoantibodies (autoAbs). Some studies indicate that treatment with progesterone, a chief female reproductive steroid, can suppress IgG2a/2c autoAb production. Little is known about how endogenous progesterone impacts lupus autoimmunity. To investigate this, we introduced a disruptive progesterone receptor (PR) gene mutation into lupus-prone mice and tracked the development of spontaneous IgG autoAbs. Here, we present evidence that PR can suppress the emergence of class-switched IgG2c autoAbs, suggesting that PR and ER-α counter-regulate a critical step in lupus autoimmunity. PR's control of IgG2c autoAb production correlates with alterations in the relative abundance of splenic T follicular helper (TFH) cells and non-TFH CD4(+) T cells, especially regulatory T cells (TREGS). Surprisingly, PR also appears to help to maintain sexually dimorphic abundance of splenic leukocytes, a feature common to many mouse models of SLE. Together our results identify a novel molecular link between female reproduction and lupus autoimmunity. Further investigation into the immunomodulatory functions of PR promises to inform reproductive health care in women and offers mechanistic insight into important immunologic phenomena of pregnancy.
Collapse
Affiliation(s)
- Alan H Wong
- a Division of Rheumatology, Department of Medicine , University of Washington , Seattle , WA , USA
| | | | | |
Collapse
|
44
|
Solano ME, Kowal MK, O'Rourke GE, Horst AK, Modest K, Plösch T, Barikbin R, Remus CC, Berger RG, Jago C, Ho H, Sass G, Parker VJ, Lydon JP, DeMayo FJ, Hecher K, Karimi K, Arck PC. Progesterone and HMOX-1 promote fetal growth by CD8+ T cell modulation. J Clin Invest 2015; 125:1726-38. [PMID: 25774501 DOI: 10.1172/jci68140] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 01/29/2015] [Indexed: 12/20/2022] Open
Abstract
Intrauterine growth restriction (IUGR) affects up to 10% of pregnancies in Western societies. IUGR is a strong predictor of reduced short-term neonatal survival and impairs long-term health in children. Placental insufficiency is often associated with IUGR; however, the molecular mechanisms involved in the pathogenesis of placental insufficiency and IUGR are largely unknown. Here, we developed a mouse model of fetal-growth restriction and placental insufficiency that is induced by a midgestational stress challenge. Compared with control animals, pregnant dams subjected to gestational stress exhibited reduced progesterone levels and placental heme oxygenase 1 (Hmox1) expression and increased methylation at distinct regions of the placental Hmox1 promoter. These stress-triggered changes were accompanied by an altered CD8+ T cell response, as evidenced by a reduction of tolerogenic CD8+CD122+ T cells and an increase of cytotoxic CD8+ T cells. Using progesterone receptor- or Hmox1-deficient mice, we identified progesterone as an upstream modulator of placental Hmox1 expression. Supplementation of progesterone or depletion of CD8+ T cells revealed that progesterone suppresses CD8+ T cell cytotoxicity, whereas the generation of CD8+CD122+ T cells is supported by Hmox1 and ameliorates fetal-growth restriction in Hmox1 deficiency. These observations in mice could promote the identification of pregnancies at risk for IUGR and the generation of clinical interventional strategies.
Collapse
|
45
|
Taves MD, Plumb AW, Sandkam BA, Ma C, Van Der Gugten JG, Holmes DT, Close DA, Abraham N, Soma KK. Steroid profiling reveals widespread local regulation of glucocorticoid levels during mouse development. Endocrinology 2015; 156:511-22. [PMID: 25406014 DOI: 10.1210/en.2013-1606] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Glucocorticoids (GCs) are produced by the adrenal glands and circulate in the blood to coordinate organismal physiology. In addition, different tissues may independently regulate their local GC levels via local GC synthesis. Here, we find that in the mouse, endogenous GCs show tissue-specific developmental patterns, rather than mirroring GCs in the blood. Using solid-phase extraction, HPLC, and specific immunoassays, we quantified endogenous steroids and found that in tissues of female and male mice, (1) local GC levels can be much higher than systemic GC levels, (2) local GCs follow age-related patterns different from those of systemic GCs, and (3) local GCs have identities different from those of systemic GCs. For example, whereas corticosterone is the predominant circulating adrenal GC in mice, high concentrations of cortisol were measured in neonatal thymus, bone marrow, and heart. The presence of cortisol was confirmed with liquid chromatography-tandem mass spectrometry. In addition, gene expression of steroidogenic enzymes was detected across multiple tissues, consistent with local GC production. Our results demonstrate that local GCs can differ from GCs in circulating blood. This finding suggests that steroids are widely used as local (paracrine or autocrine) signals, in addition to their classic role as systemic (endocrine) signals. Local GC regulation may even be the norm, rather than the exception, especially during development.
Collapse
Affiliation(s)
- Matthew D Taves
- Departments of Psychology (M.D.T., C.M., K.K.S.), Zoology (M.D.T., D.A.C., N.A., K.K.S.), Microbiology and Immunology (A.W.P., N.A.), and Fisheries (D.A.C.) and Brain Research Centre (K.K.S.), University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada; Department of Biological Sciences (B.A.S.), Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada; and Department of Pathology and Laboratory Medicine (J.G.V.D.G., D.T.H.), St. Paul's Hospital, Vancouver, British Columbia V6Z 1Y6, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Furman D. Sexual dimorphism in immunity: improving our understanding of vaccine immune responses in men. Expert Rev Vaccines 2014; 14:461-71. [PMID: 25278153 DOI: 10.1586/14760584.2015.966694] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Weaker immune responses are often observed in males compared to females. Since female hormones have proinflammatory properties and androgens have potent immunomodulatory effects, this sexual dimorphism in the immune response seems to be hormone dependent. Despite our current knowledge about the effect of sex hormones on immune cells, definition of the factors driving the sex differences in immunoclinical outcomes, such as the diminished response to infection and vaccination observed in men or the higher rates of autoimmunity observed in females, remains elusive. Recently, systems approaches to immune function have started to suggest a way toward establishing this connection. Such studies promise to improve our understanding of the mechanisms underlying the sexual dimorphism observed in the human immune system.
Collapse
Affiliation(s)
- David Furman
- Institute for Immunity, Transplantation and Infection, Stanford University, 279 Campus Drive, B240 Beckman Center, Stanford, CA 94305-5124, USA
| |
Collapse
|
47
|
Hsu P, Nanan R. Foetal immune programming: hormones, cytokines, microbes and regulatory T cells. J Reprod Immunol 2014; 104-105:2-7. [DOI: 10.1016/j.jri.2014.02.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Revised: 02/05/2014] [Accepted: 02/10/2014] [Indexed: 01/19/2023]
|
48
|
|
49
|
Jones HE. Time to focus on improving the contraceptive method mix in high HIV prevalence settings and let go of unanswerable questions. Contraception 2014; 90:357-9. [PMID: 24993486 DOI: 10.1016/j.contraception.2014.05.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 05/12/2014] [Accepted: 05/27/2014] [Indexed: 10/25/2022]
Affiliation(s)
- Heidi E Jones
- City University of New York (CUNY) School of Public Health, Hunter College.
| |
Collapse
|
50
|
Schumacher A, Costa SD, Zenclussen AC. Endocrine factors modulating immune responses in pregnancy. Front Immunol 2014; 5:196. [PMID: 24847324 PMCID: PMC4021116 DOI: 10.3389/fimmu.2014.00196] [Citation(s) in RCA: 157] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 04/22/2014] [Indexed: 12/16/2022] Open
Abstract
How the semi-allogeneic fetus is tolerated by the maternal immune system remains a fascinating phenomenon. Despite extensive research activity in this field, the mechanisms underlying fetal tolerance are still not well understood. However, there are growing evidences that immune–immune interactions as well as immune–endocrine interactions build up a complex network of immune regulation that ensures fetal survival within the maternal uterus. In the present review, we aim to summarize emerging research data from our and other laboratories on immune modulating properties of pregnancy hormones with a special focus on progesterone, estradiol, and human chorionic gonadotropin. These pregnancy hormones are critically involved in the successful establishment, maintenance, and termination of pregnancy. They suppress detrimental maternal alloresponses while promoting tolerance pathways. This includes the reduction of the antigen-presenting capacity of dendritic cells (DCs), monocytes, and macrophages as well as the blockage of natural killer cells, T and B cells. Pregnancy hormones also support the proliferation of pregnancy supporting uterine killer cells, retain tolerogenic DCs, and efficiently induce regulatory T (Treg) cells. Furthermore, they are involved in the recruitment of mast cells and Treg cells into the fetal–maternal interface contributing to a local accumulation of pregnancy-protective cells. These findings highlight the importance of endocrine factors for the tolerance induction during pregnancy and encourage further research in the field.
Collapse
Affiliation(s)
- Anne Schumacher
- Department of Experimental Obstetrics and Gynecology, Medical Faculty, Otto-von-Guericke University , Magdeburg , Germany
| | - Serban-Dan Costa
- University Women's Clinic, Otto-von-Guericke University , Magdeburg , Germany
| | - Ana Claudia Zenclussen
- Department of Experimental Obstetrics and Gynecology, Medical Faculty, Otto-von-Guericke University , Magdeburg , Germany
| |
Collapse
|