1
|
Lukanov T, Mihaylova A, Al Hadra B, Lesichkova S, Georgieva A, Popov T, Krasteva Y, Mondeshki T, Naumova E. HLA-DQB1*05:03 is associated with an increased risk of COVID-19 progression in the Bulgarian population. Hum Immunol 2025; 86:111228. [PMID: 39755001 DOI: 10.1016/j.humimm.2024.111228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 12/21/2024] [Accepted: 12/23/2024] [Indexed: 01/06/2025]
Abstract
The SARS-CoV-2 outbreak represents a global health problem. The different infection rates are heavily influenced by host genetic factors, such as variability in the HLA region. The aim of our study was to investigate whether certain HLA alleles in the Bulgarian population contribute to COVID-19 progression and their role in anti-SARS-CoV-2 immunity. We evaluated 76 patients diagnosed with COVID-19 and classified them according to severity as mild, moderate, and severe. Data from a population cohort (n = 539), representative of the Bulgarian population, was used for comparisons. We found that the HLA-DQB1*05:03 (OR = 3.13, pc = 0.0008) allele was significantly associated with COVID-19 severity. Several other class I and class II alleles showed a promising association with a predisposition to disease severity or a protective role in its progression. This is the first study to assess the association between HLA and COVID-19 progression in the Bulgarian population. Despite some limitations, our results suggest that certain HLA alleles play a role in the severity of SARS-CoV-2 infection and it would be interesting to further trace their effect in the context of long COVID.
Collapse
Affiliation(s)
- Tsvetelin Lukanov
- Medical University - Sofia, Medical Faculty, Department of Clinical Immunology, Bulgaria; University Hospital Alexandrovska, Clinic of Clinical Immunology and Stem Cell Bank, Bulgaria.
| | - Anastasiya Mihaylova
- University Hospital Alexandrovska, Clinic of Clinical Immunology and Stem Cell Bank, Bulgaria
| | - Bushra Al Hadra
- University Hospital Alexandrovska, Clinic of Clinical Immunology and Stem Cell Bank, Bulgaria
| | - Spaska Lesichkova
- Medical University - Sofia, Medical Faculty, Department of Clinical Immunology, Bulgaria; University Hospital Alexandrovska, Clinic of Clinical Immunology and Stem Cell Bank, Bulgaria
| | - Atanaska Georgieva
- University Hospital Alexandrovska, Clinic of Clinical Immunology and Stem Cell Bank, Bulgaria
| | - Tsvetan Popov
- University Hospital Alexandrovska, Clinic of Surgery, Bulgaria; Medical University - Sofia, Medical Faculty, Department of Surgery, Bulgaria
| | - Yana Krasteva
- University Hospital Alexandrovska, Clinic of Clinical Immunology and Stem Cell Bank, Bulgaria
| | - Tsanko Mondeshki
- Medical University - Sofia, Medical Faculty, Department of Propaedeutic of Internal Medicine, Bulgaria; University Hospital Alexandrovska, Clinic of Propaedeutic of Internal Medicine, Bulgaria
| | - Elissaveta Naumova
- Medical University - Sofia, Medical Faculty, Department of Clinical Immunology, Bulgaria; University Hospital Alexandrovska, Clinic of Clinical Immunology and Stem Cell Bank, Bulgaria
| |
Collapse
|
2
|
Cacciottola L, Camboni A, Dolmans MM. Immune system regulation of physiological and pathological aspects of the ovarian follicle pool throughout the female reproductive lifespan. Hum Reprod 2025; 40:12-22. [PMID: 39607771 DOI: 10.1093/humrep/deae254] [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: 05/08/2024] [Revised: 10/14/2024] [Indexed: 11/30/2024] Open
Abstract
The immune system plays a major role in ovarian physiology by regulating the ovarian follicle pool through complex signaling of different growth factors, cytokines, and chemokines. These may promote follicle activation and further growth but could also trigger follicle atresia and clearance of aging or damaged cells within the ovarian cortex. Moreover, extraglandular steroidogenesis potentially occurring in different immune cells like macrophages and natural killer cells might be another way of modulating follicle growth. Ovarian macrophages have recently been found to contain two different populations, namely resident macrophages and monocyte-derived cells, with potentially different roles. The immune system also plays a role in the development of pathological conditions, including premature ovarian insufficiency (POI). Indeed, autoimmune activation against various ovarian antigen targets results in lymphocytic oophoritis mainly targeting early growing follicles, but later leading to complete follicle pool depletion. Immune-mediated ovarian damage may also be caused by viral infection or be the consequence of iatrogenic damage. Certain novel cancer immunotherapies like checkpoint inhibitors have recently been shown to induce ovarian reserve damage in a murine model. Studies are needed to corroborate these findings and further investigate the potential of newly developed immunotherapies to treat POI. Technological advances such as single-cell analyses of less represented cell populations like immune cells inside the ovary are now contributing to valuable new information, which will hopefully lead to the development of new therapeutic strategies for women with fertility issues.
Collapse
Affiliation(s)
- L Cacciottola
- Gynecology Research Unit, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - A Camboni
- Gynecology Research Unit, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
- Department of Anatomopathology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - M M Dolmans
- Gynecology Research Unit, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
- Gynecology Department, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| |
Collapse
|
3
|
Campbell AC, Kuonqui KG, Ashokan G, Rubin J, Shin J, Pollack BL, Roberts A, Sarker A, Park HJ, Kataru RP, Barrio AV, Mehrara BJ. Role of inducible nitric oxide (iNOS) and nitrosative stress in regulating sex differences in secondary lymphedema. Front Physiol 2024; 15:1510389. [PMID: 39691094 PMCID: PMC11649630 DOI: 10.3389/fphys.2024.1510389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 11/15/2024] [Indexed: 12/19/2024] Open
Abstract
Secondary lymphedema is a common complication following surgical treatment of solid tumors. Although more prevalent in women due to higher breast cancer rates, men also develop lymphedema, often with more severe manifestations. Despite these differences in clinical presentation, the cellular mechanisms underlying sex differences are poorly understood. Previous studies have shown that inducible nitric oxide synthase (iNOS) expression by inflammatory cells is an important regulator of lymphatic pumping and leakiness in lymphedema and that lymphatic endothelial cells are highly sensitive to nitrosative stress. Based on this rationale, we used a mouse tail model of lymphedema to study the role of nitric oxide in sex-related differences in disease severity. Consistent with clinical findings, we found that male mice have significantly worse tail edema and higher rates of tail necrosis compared with female mice following tail skin/lymphatic excision (p = 0.001). Our findings correlated with increased tissue infiltration of iNOS + inflammatory cells, increased iNOS protein expression, and increased nitrosative stress in male mouse lymphedematous skin tissues (p < 0.05). Importantly, transgenic male mice lacking the iNOS gene (iNOS-KO) displayed markedly reduced swelling, inflammation, and tissue necrosis rates, whereas no differences were observed between wild-type and iNOS-KO female mice. Overall, our results indicate that iNOS-mediated nitric oxide production contributes to sex-based differences in secondary lymphedema severity, emphasizing the need to consider sex as a biological variable in lymphedema research.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Raghu P. Kataru
- Plastic and Reconstructive Surgery, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | | | - Babak J. Mehrara
- Plastic and Reconstructive Surgery, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| |
Collapse
|
4
|
Nova A, Di Caprio G, Baldrighi GN, Galdiolo D, Bernardinelli L, Fazia T. Investigating the influence of oral contraceptive pill use on multiple sclerosis risk using UK Biobank data. Fertil Steril 2024; 122:1094-1104. [PMID: 39098539 DOI: 10.1016/j.fertnstert.2024.07.999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 07/31/2024] [Accepted: 07/31/2024] [Indexed: 08/06/2024]
Abstract
OBJECTIVE To investigate the association between oral contraceptive (OC) pill use and the risk of developing multiple sclerosis (MS), attempting to address the limitations present in previous studies that produced conflicting results. DESIGN A population-based cohort study using data from the UK Biobank. PATIENTS The study included 181,058 women of white ethnicity born in England between 1937 and 1970, among which 1,131 had an MS diagnosis. INTERVENTION Oral contraceptive use, considering the self-reported age of initiation and discontinuation. The exposures of interest include the following: ever-use, current use, duration of current use in years, and age and year at initiation. MAIN OUTCOME MEASURES Multiple sclerosis diagnosis (International Classification of Disease, 10th revision: G35) was used as an outcome of interest, and the associations with the exposures of interest were investigated using marginal structural models with a time-to-event approach. To adjust for confounding, we included in the models several variables, including MS polygenic risk score, education level, parity, smoking, fertility problems, obesity, and mononucleosis. We further aimed to evaluate the influence of parity using a mediation analysis. RESULTS The association of both ever and current OC use did not result in a statistically significant MS hazard increase (ever vs. never-users, hazard ratio [HR] = 1.30 [95% confidence interval {CI}: 0.93,1.82]; current vs. never-users, HR = 1.35 [95% CI: 0.81, 2.25]). However, we highlighted parity as an effect modifier for this association. In nulliparous women, ever and current use resulted in a significant twofold and threefold MS hazard increase (HR = 2.08 [95% CI: 1.04, 4.17] and HR = 3.15 [95% CI: 1.43, 6.9]). These associations were supported by significant MS hazard increases for a higher duration of current use and for an earlier age at initiation. We further highlighted genetic MS susceptibility as another effect modifier, as a stronger OC-MS hazard association was found in women with a low MS polygenic risk score. CONCLUSION Our findings highlighted how the association between OC use and MS varies on the basis of individual characteristics such as parity and genetic MS susceptibility. Importantly, current use in nulliparous women was found to be associated with a threefold increase in MS hazard. We acknowledge the need for cautious causal interpretation and further research to validate these findings across diverse populations and OC types.
Collapse
Affiliation(s)
- Andrea Nova
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy.
| | - Giovanni Di Caprio
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Giulia N Baldrighi
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Davide Galdiolo
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Luisa Bernardinelli
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Teresa Fazia
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| |
Collapse
|
5
|
Jarkas DA, Villeneuve AH, Daneshmend AZB, Villeneuve PJ, McQuaid RJ. Sex differences in the inflammation-depression link: A systematic review and meta-analysis. Brain Behav Immun 2024; 121:257-268. [PMID: 39089535 DOI: 10.1016/j.bbi.2024.07.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Revised: 07/25/2024] [Accepted: 07/28/2024] [Indexed: 08/04/2024] Open
Abstract
Major Depressive Disorder (MDD) is a heterogeneous disorder that affects twice as many women than men. Precluding advances in more tailored and efficacious treatments for depression is the lack of reliable biomarkers. While depression is linked to elevations in inflammatory immune system functioning, this relationship is not evident among all individuals with depression and may vary based on symptom subtypes and/or sex. This systematic review and meta-analysis examined whether inflammatory immune peripheral markers of depression are sex-specific. PRISMA guidelines were followed for the systematic review, and a comprehensive search strategy that identified studies from PubMed and PsycInfo was applied. Studies were included if they reported C-reactive protein (CRP), interleukin (IL)-6, tumor necrosis factor (TNF)-α and/or IL-1β for males and/or females among depressed and healthy adults. We identified 23 studies that satisfied these inclusion criteria. Random-effects meta-analysis models were fit, and measures of association were summarized between levels of circulating markers of inflammation in depressed and healthy males and females. Sex-based analyses revealed elevated levels of CRP among females with depression (Cohen's d = 0.19) relative to their healthy counterparts (p = 0.02), an effect not apparent among males (Cohen's d = -0.01). Similarly, levels of IL-6 were increased among females with depression compared to healthy controls (Cohen's d = 0.51; p = 0.04), but once again this was not found among males (Cohen's d = 0.16). While TNF-α levels were elevated among individuals with depression compared to controls (p = 0.01), no statistically significant sex differences were found. The meta-analysis for IL-1β resulted in only three articles, and thus, results are presented in the supplemental section. This meta-analysis advances our understanding of the unique involvement of inflammatory biomarkers in depression among men and women, which may help inform more tailored sex-specific treatment approaches in the future.
Collapse
Affiliation(s)
- Dana A Jarkas
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada; University of Ottawa Institute of Mental Health Research, Ottawa, Ontario, Canada.
| | - Ally H Villeneuve
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada; University of Ottawa Institute of Mental Health Research, Ottawa, Ontario, Canada
| | - Ayeila Z B Daneshmend
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada; University of Ottawa Institute of Mental Health Research, Ottawa, Ontario, Canada
| | - Paul J Villeneuve
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | - Robyn J McQuaid
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada; University of Ottawa Institute of Mental Health Research, Ottawa, Ontario, Canada
| |
Collapse
|
6
|
Zhu Z, Lin X, Wang C, Zhu S, Zhou X. Conditional associations of sex steroid hormones with C-reactive protein levels in American children and adolescents: evidence from NHANES 2015-2016. Front Endocrinol (Lausanne) 2024; 15:1431984. [PMID: 39381439 PMCID: PMC11458447 DOI: 10.3389/fendo.2024.1431984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 08/26/2024] [Indexed: 10/10/2024] Open
Abstract
Background The relationship between sex steroid hormones and high-sensitivity C-reactive protein(hs-CRP) levels in American children and adolescents is understudied. This research will examine this association. Methods The study conducted a data analysis from the National Health and Nutrition Examination Survey (NHANES) 2015-2016, adjusting multiple linear regression models with R 4.2.2 and EmpowerStats. A total of 1,768 children and adolescents were surveyed. Data collection involved measurements of serum levels of testosterone, estradiol, sex hormone-binding globulin (SHBG) and hs-CRP. Results With the increase in testosterone, a brief rise (β=0.082, P=0.047) followed by an overall decline (β=-0.028, P=0.023) in hs-CRP was observed in the Male Prepubertal population, while a continuous decline (β=-0.002, P<0.05) was seen in the Male Pubertal group. A positive correlation (β=0.047, P<0.05) was found between testosterone and hs-CRP in the Female Prepubertal population, whereas no significant association (β=0.002, P>0.05) was detected in the Female Pubertal group. A significant inverse correlation was observed between estradiol and hs-CRP solely in the Female Pubertal group (β=-0.002, P<0.05), while no association was found in other populations. An inverse relationship between SHBG and hs-CRP was consistently noted across all groups: Male Prepubertal, Male Pubertal, Female Prepubertal, and Female Pubertal. Conclusions The association between sex steroid hormones and high-sensitivity C-reactive protein (hs-CRP) levels among American children and adolescents is conditional and influenced by multiple factors.
Collapse
Affiliation(s)
- Zhisheng Zhu
- Plastic Surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Xingong Lin
- Plastic Surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Chaoyang Wang
- Plastic Surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Shize Zhu
- Plastic Surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Xianying Zhou
- Plastic Surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| |
Collapse
|
7
|
Delimitreva S, Boneva G, Chakarova I, Hadzhinesheva V, Zhivkova R, Markova M, Nikolova V, Kolarov A, Mladenov N, Bradyanova S, Prechl J, Mihaylova N, Tchorbanov A. Lupus progression deteriorates oogenesis quality in MRL/lpr mice. Immunol Res 2024; 72:811-827. [PMID: 38771487 DOI: 10.1007/s12026-024-09489-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 05/07/2024] [Indexed: 05/22/2024]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by the activation of the immune response against self antigens. Numerous reproductive complications, including reduced birth rate and complications for the mother and the fetus during pregnancy, have been observed in women with SLE. In the present study, we aimed to investigate the effect of SLE development on oocyte meiosis in lupus-prone mice. Lupus-prone MRL/lpr mice were used for the experiments: disease-free (4 weeks of age) and sick (20 weeks of age, virgin and postpartum). The immune response was monitored by flow cytometry, ELISpot, ELISA, and histology. Oocytes were analyzed by fluorescence microscopy based on chromatin, tubulin, and actin structures. The lupus-prone MRL/lpr mice developed age-dependent symptoms of SLE with increased levels of various autoantibodies, proteinuria, and renal infiltrates and a tendency for the immune response to worsen with changes in cell populations and the cytokine profile. The number and quality of oocytes were also affected, and the successful pregnancy rate of MRL/lpr mice was limited to only 60%. Isolated oocytes showed severe structural changes in all studied groups. Systemic alterations in immune homeostasis in SLE affect the quality of developing oocytes, which is evident from a young age. The data obtained is in line with the trend of reduced fertility in lupus-prone MRL/lpr mice. The phenomenon can be explained by changes in the microenvironment of the relevant organs and close connection between ovulation and inflammatory processes.
Collapse
Affiliation(s)
- Stefka Delimitreva
- Department of Biology, Medical University of Sofia, 2, Zdrave Str., 1431, Sofia, Bulgaria.
| | - Gabriela Boneva
- Department of Immunology, Stefan Angelov Institute of Microbiology, Bulgarian Academy of Sciences, Acad. G. Bonchev Street, Block 26, 1113, Sofia, Bulgaria
| | - Irina Chakarova
- Department of Biology, Medical University of Sofia, 2, Zdrave Str., 1431, Sofia, Bulgaria
| | | | - Ralitsa Zhivkova
- Department of Biology, Medical University of Sofia, 2, Zdrave Str., 1431, Sofia, Bulgaria
| | - Maya Markova
- Department of Biology, Medical University of Sofia, 2, Zdrave Str., 1431, Sofia, Bulgaria
| | - Venera Nikolova
- Department of Biology, Medical University of Sofia, 2, Zdrave Str., 1431, Sofia, Bulgaria
| | - Anton Kolarov
- Department of Biology, Medical University of Sofia, 2, Zdrave Str., 1431, Sofia, Bulgaria
| | - Nikola Mladenov
- Department of Biology, Medical University of Sofia, 2, Zdrave Str., 1431, Sofia, Bulgaria
| | - Silviya Bradyanova
- Department of Immunology, Stefan Angelov Institute of Microbiology, Bulgarian Academy of Sciences, Acad. G. Bonchev Street, Block 26, 1113, Sofia, Bulgaria
| | | | - Nikolina Mihaylova
- Department of Immunology, Stefan Angelov Institute of Microbiology, Bulgarian Academy of Sciences, Acad. G. Bonchev Street, Block 26, 1113, Sofia, Bulgaria
| | - Andrey Tchorbanov
- Department of Immunology, Stefan Angelov Institute of Microbiology, Bulgarian Academy of Sciences, Acad. G. Bonchev Street, Block 26, 1113, Sofia, Bulgaria.
- National Institute of Immunology, 1517, Sofia, Bulgaria.
| |
Collapse
|
8
|
Truffault F, Auger L, Dragin N, Vilquin JT, Fadel E, Thomas de Montpreville V, Mansuet-Lupo A, Regnard JF, Alifano M, Sharshar T, Behin A, Eymard B, Bolgert F, Demeret S, Berrih-Aknin S, Le Panse R. Comparison of juvenile and adult myasthenia gravis in a French cohort with focus on thymic histology. Sci Rep 2024; 14:13955. [PMID: 38886398 PMCID: PMC11183198 DOI: 10.1038/s41598-024-63162-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 05/26/2024] [Indexed: 06/20/2024] Open
Abstract
Myasthenia gravis (MG) is an autoimmune disease characterized by muscle fatigability due to acetylcholine receptor (AChR) autoantibodies. To better characterize juvenile MG (JMG), we analyzed 85 pre- and 132 post-pubescent JMG (with a cutoff age of 13) compared to 721 adult MG patients under 40 years old using a French database. Clinical data, anti-AChR antibody titers, thymectomy, and thymic histology were analyzed. The proportion of females was higher in each subgroup. No significant difference in the anti-AChR titers was observed. Interestingly, the proportion of AChR+ MG patients was notably lower among adult MG patients aged between 30 and 40 years, at 69.7%, compared to over 82.4% in the other subgroups. Thymic histological data were examined in patients who underwent thymectomy during the year of MG onset. Notably, in pre-JMG, the percentage of thymectomized patients was significantly lower (32.9% compared to more than 42.5% in other subgroups), and the delay to thymectomy was twice as long. We found a positive correlation between anti-AChR antibodies and germinal center grade across patient categories. Additionally, only females, particularly post-JMG patients, exhibited the highest rates of lymphofollicular hyperplasia (95% of cases) and germinal center grade. These findings reveal distinct patterns in JMG patients, particularly regarding thymic follicular hyperplasia, which appears to be exacerbated in females after puberty.
Collapse
Affiliation(s)
- Frédérique Truffault
- Center of Research in Myology, Institute of Myology, INSERM, Sorbonne University, 105, Boulevard de l'Hôpital, 75013, Paris, France
| | - Ludivine Auger
- Center of Research in Myology, Institute of Myology, INSERM, Sorbonne University, 105, Boulevard de l'Hôpital, 75013, Paris, France
| | - Nadine Dragin
- Center of Research in Myology, Institute of Myology, INSERM, Sorbonne University, 105, Boulevard de l'Hôpital, 75013, Paris, France
| | - Jean-Thomas Vilquin
- Center of Research in Myology, Institute of Myology, INSERM, Sorbonne University, 105, Boulevard de l'Hôpital, 75013, Paris, France
| | - Elie Fadel
- Marie Lannelongue Hospital, Paris Saclay University, Le Plessis-Robinson, France
| | | | - Audrey Mansuet-Lupo
- Department of Pathology, Cochin University Hospital Group, AP-HP, Paris-Descartes University, Paris, France
| | - Jean-François Regnard
- Department of Pathology, Cochin University Hospital Group, AP-HP, Paris-Descartes University, Paris, France
| | - Marco Alifano
- Department of Pathology, Cochin University Hospital Group, AP-HP, Paris-Descartes University, Paris, France
| | - Tarek Sharshar
- Anesthesia and Intensive Care Department, GHU Paris Psychiatrie et Neurosciences, Pole Neuro, Sainte‑Anne Hospital, Paris, Institute of Psychiatry and Neurosciences of Paris, INSERM U1266, Université Paris Cité, Paris, France
| | - Anthony Behin
- AP-HP, Referral Center for Neuromuscular Disorders, Institute of Myology, Pitié-Salpêtrière Hospital, AP-HP, Paris, France
| | - Bruno Eymard
- AP-HP, Referral Center for Neuromuscular Disorders, Institute of Myology, Pitié-Salpêtrière Hospital, AP-HP, Paris, France
| | - Francis Bolgert
- Neuro-Intensive Care Unit, Pitié-Salpêtrière Hospital, AP-HP, Sorbonne University, Paris, France
| | - Sophie Demeret
- Neuro-Intensive Care Unit, Pitié-Salpêtrière Hospital, AP-HP, Sorbonne University, Paris, France
| | - Sonia Berrih-Aknin
- Center of Research in Myology, Institute of Myology, INSERM, Sorbonne University, 105, Boulevard de l'Hôpital, 75013, Paris, France
| | - Rozen Le Panse
- Center of Research in Myology, Institute of Myology, INSERM, Sorbonne University, 105, Boulevard de l'Hôpital, 75013, Paris, France.
| |
Collapse
|
9
|
Gai D, Caviness PC, Lazarenko OP, Chen JF, Randolph CE, Zhang Z, Cheng Y, Sun F, Xu H, Blackburn ML, Tricot G, Shaughnessy JD, Chen JR, Zhan F. Cystatin M/E ameliorates bone resorption through increasing osteoclastic cell estrogen influx. RESEARCH SQUARE 2024:rs.3.rs-4313179. [PMID: 38766009 PMCID: PMC11100902 DOI: 10.21203/rs.3.rs-4313179/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
In multiple myeloma (MM), increased osteoclast differentiation leads to the formation of osteolytic lesions in most MM patients. Bisphosphonates, such as zoledronic acid (ZA), are used to ameliorate bone resorption, but due to risk of serious side effects as well as the lack of repair of existing lesions, novel anti-bone resorption agents are required. Previously, the absence of osteolytic lesions in MM was strongly associated with elevated levels of cystatin M/E (CST6), a cysteine protease inhibitor, secreted by MM cells. In this study, both MM- and ovariectomy (OVX)-induced osteoporotic mouse models were used to compare the effects of recombinant mouse CST6 (rmCst6) and ZA on preventing bone loss. μCT showed that rmCst6 and ZA had similar effects on improving percent bone volume, and inhibited differentiation of non-adherent bone marrow cells into mature osteoclasts. Single-cell RNA sequencing showed that rmCst6 and not ZA treatment reduced bone marrow macrophage percentage in the MM mouse model compared to controls. Protein and mRNA arrays showed that both rmCst6 and ZA significantly inhibit OVX-induced expression of inflammatory cytokines. For OVX mice, ERα protein expression in bone was brought to sham surgery level by only rmCst6 treatments. rmCst6 significantly increased mRNA and protein levels of ERα and significantly increased total intracellular estrogen concentrations for ex vivo osteoclast precursor cell cultures. Based on these results, we conclude that CST6 improves MM or OVX bone loss models by increasing the expression of estrogen receptors as well as the intracellular estrogen concentration in osteoclast precursors, inhibiting their maturation.
Collapse
Affiliation(s)
- Dongzheng Gai
- Myeloma Center, Winthrop P. Rockefeller Cancer Institute, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
- Department of Hematology, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, 046000, China
| | - Perry C. Caviness
- Arkansas Children’s Nutrition Center, Little Rock, AR 72205, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Oxana P. Lazarenko
- Arkansas Children’s Nutrition Center, Little Rock, AR 72205, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Jennifer F. Chen
- Undergraduate Pre-Medical Program, University of Arkansas at Fayetteville, Fayetteville, AR, 72701, USA
| | - Christopher E. Randolph
- Center for Translational Pediatric Research, Arkansas Children’s Research Institute, Little Rock, AR 72202, USA
| | - Zijun Zhang
- Myeloma Center, Winthrop P. Rockefeller Cancer Institute, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Yan Cheng
- Myeloma Center, Winthrop P. Rockefeller Cancer Institute, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Fumou Sun
- Myeloma Center, Winthrop P. Rockefeller Cancer Institute, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Hongwei Xu
- Myeloma Center, Winthrop P. Rockefeller Cancer Institute, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Michael L Blackburn
- Arkansas Children’s Nutrition Center, Little Rock, AR 72205, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Guido Tricot
- Myeloma Center, Winthrop P. Rockefeller Cancer Institute, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - John D Shaughnessy
- Myeloma Center, Winthrop P. Rockefeller Cancer Institute, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Jin-Ran Chen
- Arkansas Children’s Nutrition Center, Little Rock, AR 72205, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Fenghuang Zhan
- Myeloma Center, Winthrop P. Rockefeller Cancer Institute, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| |
Collapse
|
10
|
Kowsar R, Sadeghi K, Hashemzadeh F, Miyamoto A. Ovarian sex steroid and epithelial control of immune responses in the uterus and oviduct: human and animal models†. Biol Reprod 2024; 110:230-245. [PMID: 38038990 PMCID: PMC10873282 DOI: 10.1093/biolre/ioad166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 11/08/2023] [Accepted: 11/30/2023] [Indexed: 12/02/2023] Open
Abstract
The female reproductive tract (FRT), including the uterus and oviduct (Fallopian tube), is responsible for maintaining an optimal microenvironment for reproductive processes, such as gamete activation and transportation, sperm capacitation, fertilization, and early embryonic and fetal development. The mucosal surface of the FRT may be exposed to pathogens and sexually transmitted microorganisms due to the opening of the cervix during mating. Pathogens and endotoxins may also reach the oviduct through the peritoneal fluid. To maintain an optimum reproductive environment while recognizing and killing pathogenic bacterial and viral agents, the oviduct and uterus should be equipped with an efficient and rigorously controlled immune system. Ovarian sex steroids can affect epithelial cells and underlying stromal cells, which have been shown to mediate innate and adaptive immune responses. This, in turn, protects against potential infections while maintaining an optimal milieu for reproductive events, highlighting the homeostatic involvement of ovarian sex steroids and reproductive epithelial cells. This article will discuss how ovarian sex steroids affect the immune reactions elicited by the epithelial cells of the non-pregnant uterus and oviduct in the bovine, murine, and human species. Finally, we propose that there are regional and species-specific differences in the immune responses in FRT.
Collapse
Affiliation(s)
- Rasoul Kowsar
- Department of Animal Sciences, College of Agriculture, Isfahan University of Technology, Isfahan, Iran
| | | | - Farzad Hashemzadeh
- Department of Animal Sciences, College of Agriculture, Isfahan University of Technology, Isfahan, Iran
| | - Akio Miyamoto
- Global Agromedicine Research Center, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, Japan
| |
Collapse
|
11
|
Pandey SP, Bhaskar R, Han SS, Narayanan KB. Autoimmune Responses and Therapeutic Interventions for Systemic Lupus Erythematosus: A Comprehensive Review. Endocr Metab Immune Disord Drug Targets 2024; 24:499-518. [PMID: 37718519 DOI: 10.2174/1871530323666230915112642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 06/05/2023] [Accepted: 07/22/2023] [Indexed: 09/19/2023]
Abstract
Systemic Lupus Erythematosus (SLE) or Lupus is a multifactorial autoimmune disease of multiorgan malfunctioning of extremely heterogeneous and unclear etiology that affects multiple organs and physiological systems. Some racial groups and women of childbearing age are more susceptible to SLE pathogenesis. Impressive progress has been made towards a better understanding of different immune components contributing to SLE pathogenesis. Recent investigations have uncovered the detailed mechanisms of inflammatory responses and organ damage. Various environmental factors, pathogens, and toxicants, including ultraviolet light, drugs, viral pathogens, gut microbiome metabolites, and sex hormones trigger the onset of SLE pathogenesis in genetically susceptible individuals and result in the disruption of immune homeostasis of cytokines, macrophages, T cells, and B cells. Diagnosis and clinical investigations of SLE remain challenging due to its clinical heterogeneity and hitherto only a few approved antimalarials, glucocorticoids, immunosuppressants, and some nonsteroidal anti-inflammatory drugs (NSAIDs) are available for treatment. However, the adverse effects of renal and neuropsychiatric lupus and late diagnosis make therapy challenging. Additionally, SLE is also linked to an increased risk of cardiovascular diseases due to inflammatory responses and the risk of infection from immunosuppressive treatment. Due to the diversity of symptoms and treatment-resistant diseases, SLE management remains a challenging issue. Nevertheless, the use of next-generation therapeutics with stem cell and gene therapy may bring better outcomes to SLE treatment in the future. This review highlights the autoimmune responses as well as potential therapeutic interventions for SLE particularly focusing on the recent therapeutic advancements and challenges.
Collapse
Affiliation(s)
- Surya Prakash Pandey
- Aarogya Institute of Healthcare and Research, Jaipur, Rajasthan, 302033, India
- Department of Zoology, School of Science, IFTM University, Moradabad, Uttar Pradesh, 244102, India
| | - Rakesh Bhaskar
- School of Chemical Engineering, Yeungnam University, Gyeongsan, 38541, South Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, Gyeongbuk, 38541, South Korea
| | - Sung Soo Han
- School of Chemical Engineering, Yeungnam University, Gyeongsan, 38541, South Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, Gyeongbuk, 38541, South Korea
| | - Kannan Badri Narayanan
- School of Chemical Engineering, Yeungnam University, Gyeongsan, 38541, South Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, Gyeongbuk, 38541, South Korea
| |
Collapse
|
12
|
Lagou MK, Karagiannis GS. Obesity-induced thymic involution and cancer risk. Semin Cancer Biol 2023; 93:3-19. [PMID: 37088128 DOI: 10.1016/j.semcancer.2023.04.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 04/19/2023] [Accepted: 04/20/2023] [Indexed: 04/25/2023]
Abstract
Declining thymic functions associated either with old age (i.e., age-related thymic involution), or with acute involution as a result of stress, infectious disease, or cytoreductive therapies (e.g., chemotherapy/radiotherapy), have been associated with cancer development. A key mechanism underlying such increased cancer risk is the thymus-dependent debilitation of adaptive immunity, which is responsible for orchestrating immunoediting mechanisms and tumor immune surveillance. In the past few years, a blooming set of evidence has intriguingly linked obesity with cancer development and progression. The majority of such studies has focused on obesity-driven chronic inflammation, steroid/sex hormone and adipokine production, and hyperinsulinemia, as principal factors affecting the tumor microenvironment and driving the development of primary malignancy. However, experimental observations about the negative impact of obesity on T cell development and maturation have existed for more than half a century. Here, we critically discuss the molecular and cellular mechanisms of obesity-driven thymic involution as a previously underrepresented intermediary pathology leading to cancer development and progression. This knowledge could be especially relevant in the context of childhood obesity, because impaired thymic function in young individuals leads to immune system abnormalities, and predisposes to various pediatric cancers. A thorough understanding behind the molecular and cellular circuitries governing obesity-induced thymic involution could therefore help towards the rationalized development of targeted thymic regeneration strategies for obese individuals at high risk of cancer development.
Collapse
Affiliation(s)
- Maria K Lagou
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA; Tumor Microenvironment of Metastasis Program, Albert Einstein Cancer Center, Bronx, NY, USA
| | - George S Karagiannis
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA; Tumor Microenvironment of Metastasis Program, Albert Einstein Cancer Center, Bronx, NY, USA; Cancer Dormancy and Tumor Microenvironment Institute, Albert Einstein College of Medicine, Bronx, NY, USA; Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, USA; Integrated Imaging Program for Cancer Research, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
13
|
Szukiewicz D. Insight into the Potential Mechanisms of Endocrine Disruption by Dietary Phytoestrogens in the Context of the Etiopathogenesis of Endometriosis. Int J Mol Sci 2023; 24:12195. [PMID: 37569571 PMCID: PMC10418522 DOI: 10.3390/ijms241512195] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 07/25/2023] [Accepted: 07/27/2023] [Indexed: 08/13/2023] Open
Abstract
Phytoestrogens (PEs) are estrogen-like nonsteroidal compounds derived from plants (e.g., nuts, seeds, fruits, and vegetables) and fungi that are structurally similar to 17β-estradiol. PEs bind to all types of estrogen receptors, including ERα and ERβ receptors, nuclear receptors, and a membrane-bound estrogen receptor known as the G protein-coupled estrogen receptor (GPER). As endocrine-disrupting chemicals (EDCs) with pro- or antiestrogenic properties, PEs can potentially disrupt the hormonal regulation of homeostasis, resulting in developmental and reproductive abnormalities. However, a lack of PEs in the diet does not result in the development of deficiency symptoms. To properly assess the benefits and risks associated with the use of a PE-rich diet, it is necessary to distinguish between endocrine disruption (endocrine-mediated adverse effects) and nonspecific effects on the endocrine system. Endometriosis is an estrogen-dependent disease of unknown etiopathogenesis, in which tissue similar to the lining of the uterus (the endometrium) grows outside of the uterus with subsequent complications being manifested as a result of local inflammatory reactions. Endometriosis affects 10-15% of women of reproductive age and is associated with chronic pelvic pain, dysmenorrhea, dyspareunia, and infertility. In this review, the endocrine-disruptive actions of PEs are reviewed in the context of endometriosis to determine whether a PE-rich diet has a positive or negative effect on the risk and course of endometriosis.
Collapse
Affiliation(s)
- Dariusz Szukiewicz
- Department of Biophysics, Physiology & Pathophysiology, Faculty of Health Sciences, Medical University of Warsaw, 02-004 Warsaw, Poland
| |
Collapse
|
14
|
Kim DHJ, Iosif AM, Ramirez-Celis A, Ashwood P, Ames JL, Lyall K, Berger K, Croen LA, Van de Water J. Neonatal immune signatures differ by sex regardless of neurodevelopmental disorder status: Macrophage migration inhibitory factor (MIF) alone reveals a sex by diagnosis interaction effect. Brain Behav Immun 2023; 111:328-333. [PMID: 37164311 PMCID: PMC10796272 DOI: 10.1016/j.bbi.2023.05.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 04/10/2023] [Accepted: 05/05/2023] [Indexed: 05/12/2023] Open
Abstract
Immune dysregulation, including aberrant peripheral cytokine/chemokine levels, is implicated in neurodevelopmental disorders (NDD) such as autism spectrum disorder (ASD). While the diagnosis of ASD is more common in males compared to females, sex effects in immune dysregulation related to neurodevelopment remain understudied. The aim of this exploratory study was to determine whether there are sex-specific effects in neonatal immune dysregulation with respect to an ASD or delayed development (DD) diagnosis. We utilized the data from the Early Markers for Autism study, a population based case-control study of prenatal and neonatal biomarkers of ASD. The immune profile of newborns later diagnosed with ASD (n = 482, 91 females), DD (n = 140, 61 females) and sex-matched general population controls (GP; n = 378, 67 females) were analyzed using neonatal bloodspots (NBS) via 42-plex multiplex assay. Multiple linear regression analysis was performed to identify whether sex was associated with differences in cytokine/chemokine levels of children with ASD, DD, and GP. A sex by diagnosis interaction effect was observed only for the chemokine macrophage migration inhibitory factor (MIF), with males displaying higher levels of NBS MIF than females in the GP control group (p = 0.02), but not in ASD (p = 0.52) or DD (p = 0.29) groups. We found that regardless of child diagnosis, newborn bloodspot eluates from females had a significantly higher concentration than males with the same diagnosis of the chemokines granulocyte chemotactic protein 2 (GCP-2; p < 0.0001), macrophage inflammatory protein 2-alpha (GROβ; p = 0.002), interferon-inducible t-cell alpha chemoattractant (I-TAC; p < 0.0001), stromal cell-derived factor 1 alpha and beta (SDF-1α-β; p = 0.03), innate inflammatory chemokines interferon-gamma induced protein 10 (IP-10; p = 0.02), macrophage inflammatory protein 1-alpha (MIP-1α; p = 0.02), and Th1-related pro-inflammatory cytokine interleukin-12 active heterodimer (IL-12p70; p = 0.002). In contrast, males had a higher concentration than females of secondary lymphoid-tissue chemokine (6CKINE; p = 0.02), monocyte chemotactic protein 1 (MCP-1; p = 0.005) and myeloid progenitor inhibitory factor 1 (MPIF-1; p = 0.03). Results were similar when analyses were restricted to NBS from DD and ASD further classified as ASD with intellectual disability (ID), ASD without ID, and DD (GCP-2, p = 0.007; I-TAC, p = 0.001; IP-10, p = 0.005; IL-12p70, p = 0.03 higher in females; MPIF-1, p = 0.03 higher in male). This study is the first to examine sex differences in neonatal cytokine/chemokine concentrations, and whether these differences are associated with neurodevelopmental outcomes. Results highlight the importance of considering sex as a critical factor in understanding the immune system as it relates to child development.
Collapse
Affiliation(s)
- Danielle H J Kim
- Department of Internal Medicine, Division of Rheumatology, Allergy, and Clinical Immunology, University of California, Davis, CA, USA
| | - Ana-Maria Iosif
- Department of Public Health Sciences, University of California, Davis, CA, USA
| | - Alexandra Ramirez-Celis
- Department of Internal Medicine, Division of Rheumatology, Allergy, and Clinical Immunology, University of California, Davis, CA, USA
| | - Paul Ashwood
- MIND Institute, University of California, Davis, CA, USA
| | | | - Kristen Lyall
- AJ Drexel Autism Institute, Drexel University, Philadelphia, PA, USA
| | | | - Lisa A Croen
- Kaiser Permanente Northern California-Oakland, USA
| | - Judy Van de Water
- Department of Internal Medicine, Division of Rheumatology, Allergy, and Clinical Immunology, University of California, Davis, CA, USA; MIND Institute, University of California, Davis, CA, USA.
| |
Collapse
|
15
|
Vinciguerra C, Iacono S, Bevilacqua L, Landolfi A, Piscosquito G, Ginanneschi F, Schirò G, Di Stefano V, Brighina F, Barone P, Balistreri CR. Sex differences in neuromuscular disorders. Mech Ageing Dev 2023; 211:111793. [PMID: 36806604 DOI: 10.1016/j.mad.2023.111793] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/13/2023] [Accepted: 02/16/2023] [Indexed: 02/19/2023]
Abstract
The prevalence, onset, pathophysiology, and clinical course of many neuromuscular disorders (NMDs) may significantly differ between males and females. Some NMDs are more frequently observed in females, and characterized to show a higher grade of severity during or after the pregnancy. Meanwhile, others tend to have an earlier onset in males and exhibit a more variable progression. Prevalently, sex differences in NMDs have a familiar character given from genetic inheritance. However, they may also influence clinical presentation and disease severity of acquired NMD forms, and are represented by both hormonal and genetic factors. Consequently, to shed light on the distinctive role of biological factors in the different clinical phenotypes, we summarize in this review the sex related differences and their distinctive biological roles emerging from the current literature in both acquired and inherited NMDs.
Collapse
Affiliation(s)
- Claudia Vinciguerra
- Neurology Unit, Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, 84131 Salerno, Italy.
| | - Salvatore Iacono
- Neurology Unit, Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), University of Palermo, 90127 Palermo, Italy
| | - Liliana Bevilacqua
- Neurology Unit, Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, 84131 Salerno, Italy
| | - Annamaria Landolfi
- Neurology Unit, Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, 84131 Salerno, Italy
| | - Giuseppe Piscosquito
- Neurology Unit, Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, 84131 Salerno, Italy
| | - Federica Ginanneschi
- Department of Medical, Surgical and Neurological Sciences, University of Siena, 53100 Siena, Italy
| | - Giuseppe Schirò
- Neurology Unit, Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), University of Palermo, 90127 Palermo, Italy
| | - Vincenzo Di Stefano
- Neurology Unit, Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), University of Palermo, 90127 Palermo, Italy
| | - Filippo Brighina
- Neurology Unit, Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), University of Palermo, 90127 Palermo, Italy
| | - Paolo Barone
- Neurology Unit, Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, 84131 Salerno, Italy
| | - Carmela Rita Balistreri
- Cellular and Molecular Laboratory, Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), University of Palermo, 90134 Palermo
| |
Collapse
|
16
|
Edwards M, Lam S, Ranjan R, Pereira M, Babbitt C, Lacreuse A. Letrozole treatment alters hippocampal gene expression in common marmosets (Callithrix jacchus). Horm Behav 2023; 147:105281. [PMID: 36434852 PMCID: PMC9839488 DOI: 10.1016/j.yhbeh.2022.105281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 10/28/2022] [Accepted: 11/09/2022] [Indexed: 11/25/2022]
Abstract
Aromatase inhibitors (AIs) are a class of drugs commonly given to patients with estrogen receptor (ER)-dependent breast cancers to reduce estrogenic stimulation. However, AIs like Letrozole are associated with negative side effects such as cognitive deficits, sleep disturbances and hot flashes. We have previously shown that these negative effects can be recapitulated in common marmosets (Callithrix jacchus) treated with Letrozole (20 μg daily) for 4 weeks and that marmosets treated with Letrozole show increased levels of estradiol in the hippocampus (Gervais et al., 2019). In order to better understand the mechanisms through which AIs affect cognitive function and increase steroid levels in the hippocampus, we used bulk, paired-end RNA-sequencing to examine differentially expressed genes among Letrozole-treated (LET; n = 8) and vehicle-treated (VEH; n = 8) male and female animals. Gene ontology results show significant reduction across hundreds of categories, some of the most significant being inflammatory response, stress response, MHC Class II protein complex binding, T-cell activation, carbohydrate binding and signaling receptor binding in LET animals. GSEA results indicate that LET females, but not LET males, show enrichment for hormonal gene sets. Based on the transcriptional changes observed, we conclude that AIs may differentially affect the sexes in part due to processes mediated by the CYP-450 superfamily. Ongoing studies will further investigate the longitudinal effects of AIs on behavior and whether AIs increase the risk of stress-induced neurodegeneration.
Collapse
Affiliation(s)
- Mélise Edwards
- University of Massachusetts Amherst, Department of Psychological & Brain Sciences, Amherst, MA 01003, USA; Neuroscience and Behavior Graduate Program, University of Massachusetts, Amherst, MA 01003, USA.
| | - Sam Lam
- University of Massachusetts Amherst, Department of Psychological & Brain Sciences, Amherst, MA 01003, USA
| | - Ravi Ranjan
- University of Massachusetts Amherst, Department of Psychological & Brain Sciences, Amherst, MA 01003, USA; Genomics Resource Laboratory, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Mariana Pereira
- University of Massachusetts Amherst, Department of Psychological & Brain Sciences, Amherst, MA 01003, USA; Neuroscience and Behavior Graduate Program, University of Massachusetts, Amherst, MA 01003, USA
| | - Courtney Babbitt
- University of Massachusetts Amherst, Department of Psychological & Brain Sciences, Amherst, MA 01003, USA; University of Massachusetts Amherst, Department of Biology, Amherst, MA 01003, USA
| | - Agnès Lacreuse
- University of Massachusetts Amherst, Department of Psychological & Brain Sciences, Amherst, MA 01003, USA; Neuroscience and Behavior Graduate Program, University of Massachusetts, Amherst, MA 01003, USA
| |
Collapse
|
17
|
Kim J, Jeong J, Lee CM, Lee DW, Kang CK, Choe PG, Kim NJ, Oh MD, Lee CH, Park WB, Lee KH, Im SA. Prospective longitudinal analysis of antibody response after standard and booster doses of SARS-COV2 vaccination in patients with early breast cancer. Front Immunol 2022; 13:1028102. [DOI: 10.3389/fimmu.2022.1028102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 10/26/2022] [Indexed: 11/18/2022] Open
Abstract
IntroductionSevere acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and its variants brought waves of pandemics with breakthrough infections in vaccinated individuals. We analyzed the antibody responses after primary and booster vaccination in healthy controls (HC) and patients with early breast cancer (BC).MethodsIn this prospective longitudinal cohort study, the binding activity of serum antibody level against spike proteins and antigens of SARS-CoV-2 variants was measured within 21 days after each vaccination in the BC group and HC group.ResultsAll participants, 40 in the BC and 20 in the HC group, had increased antibody response after vaccination. BC group, however, had weaker humoral responses than the HC group (IgG: 1.5, 2.3, 2.5-folds in BC vs. 1.9, 3.6, 4.0-folds in HC after each dose; IgA: 2.1, 3.0, 3.6-folds in BC vs. 4.2, 10.4, 5.2-folds in HC after each dose, respectively). Those under concurrent cytotoxic chemotherapy had weaker antibody response than the non-cytotoxic treatment group and HC. Adjunct use of steroids and age were not significant risk factors. The levels of binding antibody against the Delta and the Omicron (BA1) variants were lower than the wild-type, especially in BC.ConclusionIn the waves of new sub-variants, our study suggests that an additional dose of vaccinations should be recommended according to the anti-cancer treatment modality in patients with BC who had received booster vaccination.
Collapse
|
18
|
Muacevic A, Adler JR, BHASKAR EMMANUEL, Marappa L. Bedside Scoring System for Predicting Adverse Outcomes Among Patients Suffering From SARS-CoV-2 Infection. Cureus 2022; 14:e32009. [PMID: 36589201 PMCID: PMC9798458 DOI: 10.7759/cureus.32009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/28/2022] [Indexed: 11/30/2022] Open
Abstract
Aim To develop a clinical risk score to predict adverse outcomes among diabetic hospitalized COVID-19 patients Methods The data was collected retrospectively from patients hospitalized with the SARS-CoV-2 virus at Sri Ramachandra Institute of Higher education and research. It integrated independent variables such as sex, age, glycemic status, socioeconomic status, and preexisting lung conditions. Each variable was assigned a value and the final score was calculated as a sum of all the variables. The final score was then compared with patient outcomes. The patients were scored from 0 to 8 and a score of 3 or more was considered as being at greater risk for developing complications. Number of mortalities in each group, any clinical deterioration requiring ICU admission, and the number of patients requiring a prolonged hospital stay of more than 10 days in each group were noted and the results compared. Results Higher blood glucose levels and preexisting lung conditions like chronic obstructive pulmonary disease (COPD), asthma, and pulmonary tuberculosis have been associated with a higher risk of developing complications related to SARS-CoV-2 illness. Of the 5023 patients enrolled in the study, 2402 had a score of 2 or below, and 2621 had a score of 3 or above. Among patients with a score of 2 or below 1.7% of the patients contracted a severe disease resulting in death. 2.9% were shifted to ICU, but recovered and 12.2% of patients had a prolonged hospital stay. Of those with a score of 3 or greater, 5.1% died, 7.36% were shifted to ICU, but recovered, and 19.5% required a prolonged hospital stay. The observed results were analyzed using the Chi-square test and were found to be significant at a p-level of 0.0001. Conclusion This clinical risk score has been built with routinely available data to help predict adverse outcomes in diabetic patients hospitalized with the SARS-CoV-2 virus. It is a good tool for resource-limited areas as it uses readily available data. It can also be used for other severe acute respiratory illnesses or influenza-like illnesses.
Collapse
|
19
|
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: 5] [Impact Index Per Article: 1.7] [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
|
20
|
Masama C, Jarkas DA, Thaw E, Daneshmend AZB, Franklyn SI, Beaurepaire C, McQuaid RJ. Hormone contraceptive use in young women: Altered mood states, neuroendocrine and inflammatory biomarkers. Horm Behav 2022; 144:105229. [PMID: 35779518 DOI: 10.1016/j.yhbeh.2022.105229] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 04/29/2022] [Accepted: 06/21/2022] [Indexed: 11/20/2022]
Abstract
Oral contraceptives are used by millions of women worldwide, yet there are questions regarding the psychological and biological consequences of these medications. Considering that sex steroid hormones can regulate neuroendocrine and behavioral responses to stress, the current study examined mood and stress symptomatologies, as well as circulating levels of cortisol and inflammatory biomarkers among young women (N = 388), of whom, 47.0 % (n = 182) were using a form of hormonal contraception. Women using hormone contraceptives displayed significantly higher depressive and stress scores compared to non-users, whereas no differences were found for anxiety symptoms. Moreover, contraceptive users had markedly elevated plasma cortisol and C-reactive protein levels in comparison to non-users. Upon assessing women at different phases of their menstrual cycle, hormone contraceptive users displayed higher levels of cortisol compared to women in the follicular and luteal phases, in addition to higher levels of CRP levels compared to women in the luteal phase. Together, these findings suggest that hormone contraceptive use is linked to exaggerated basal neuroendocrine and inflammatory profiles, which could potentially increase sensitivity to the impacts of stressors and mood disturbances.
Collapse
Affiliation(s)
- Coleka Masama
- Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario K1S 5B6, Canada
| | - Dana A Jarkas
- Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario K1S 5B6, Canada.
| | - Emily Thaw
- Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario K1S 5B6, Canada
| | - Ayeila Z B Daneshmend
- Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario K1S 5B6, Canada
| | - Sabina I Franklyn
- Department of Psychology, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario K1S 5B6, Canada
| | - Cecile Beaurepaire
- University of Ottawa Institute of Mental Health Research, 1145 Carling Avenue, K1Z 7K4 Ottawa, Ontario, Canada
| | - Robyn J McQuaid
- Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario K1S 5B6, Canada; University of Ottawa Institute of Mental Health Research, 1145 Carling Avenue, K1Z 7K4 Ottawa, Ontario, Canada
| |
Collapse
|
21
|
Wu H, Zeng L, Ou J, Wang T, Chen Y, Nandakumar KS. Estrogen Acts Through Estrogen Receptor-β to Promote Mannan-Induced Psoriasis-Like Skin Inflammation. Front Immunol 2022; 13:818173. [PMID: 35663991 PMCID: PMC9160234 DOI: 10.3389/fimmu.2022.818173] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 04/21/2022] [Indexed: 11/13/2022] Open
Abstract
Sex-bias is more obvious in several autoimmune disorders, but not in psoriasis. However, estrogen levels fluctuate during puberty, menstrual cycle, pregnancy, and menopause, which are related to variations in psoriasis symptoms observed in female patients. Estrogen has disease promoting or ameliorating functions based on the type of immune responses and tissues involved. To investigate the effects of estrogen on psoriasis, at first, we developed an innate immunity dependent mannan-induced psoriasis model, which showed a clear female preponderance in disease severity in several mouse strains. Next, we investigated the effects of endogenous and exogenous estrogen using ovariectomy and sham operated mice. 17-β-estradiol (E2) alone promoted the skin inflammation and it also significantly enhanced mannan-induced skin inflammation. We also observed a prominent estrogen receptor-β (ER-β) expression in the skin samples, especially on keratinocytes. Subsequently, we confirmed the effects of E2 on psoriasis using ER-β antagonist (PHTPP) and agonist (DPN). In addition, estrogen was found to affect the expression of certain genes (vgll3 and cebpb), microRNAs (miR146a and miR21), and immune cells (DCs and γδ T cells) as well as chemokines (CCL5 and CXCL10) and cytokines (TNF-α, IL-6, IL-22, IL-23, and IL-17 family), which promoted the skin inflammation. Thus, we demonstrate a pathogenic role for 17-β-estradiol in promoting skin inflammation, which should be considered while designing new treatment strategies for psoriasis patients.
Collapse
Affiliation(s)
- Huimei Wu
- Southern Medical University - Karolinska Institute United Medical Inflammation Center, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Longhui Zeng
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Jiaxin Ou
- Southern Medical University - Karolinska Institute United Medical Inflammation Center, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Tingting Wang
- Southern Medical University - Karolinska Institute United Medical Inflammation Center, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Yong Chen
- Department of Rheumatology and Immunology, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| | - Kutty Selva Nandakumar
- Southern Medical University - Karolinska Institute United Medical Inflammation Center, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| |
Collapse
|
22
|
Pan Z, Zhu T, Liu Y, Zhang N. Role of the CXCL13/CXCR5 Axis in Autoimmune Diseases. Front Immunol 2022; 13:850998. [PMID: 35309354 PMCID: PMC8931035 DOI: 10.3389/fimmu.2022.850998] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 02/16/2022] [Indexed: 12/12/2022] Open
Abstract
CXCL13 is a B-cell chemokine produced mainly by mesenchymal lymphoid tissue organizer cells, follicular dendritic cells, and human T follicular helper cells. By binding to its receptor, CXCR5, CXCL13 plays an important role in lymphoid neogenesis, lymphoid organization, and immune responses. Recent studies have found that CXCL13 and its receptor CXCR5 are implicated in the pathogenesis of several autoimmune diseases, such as rheumatoid arthritis, multiple sclerosis, systemic lupus erythematosus, primary Sjögren’s syndrome, myasthenia gravis, and inflammatory bowel disease. In this review, we discuss the biological features of CXCL13 and CXCR5 and the recent findings on the pathogenic roles of the CXCL13/CXCR5 axis in autoimmune diseases. Furthermore, we discuss the potential role of CXCL13 as a disease biomarker and therapeutic target in autoimmune diseases.
Collapse
Affiliation(s)
- Zijian Pan
- National Center for Birth Defect Monitoring, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, and State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, China
- West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Tong Zhu
- National Center for Birth Defect Monitoring, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, and State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, China
- West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yanjun Liu
- National Center for Birth Defect Monitoring, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, and State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, China
| | - Nannan Zhang
- National Center for Birth Defect Monitoring, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, and State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, China
- *Correspondence: Nannan Zhang,
| |
Collapse
|
23
|
Harding AT, Heaton NS. The Impact of Estrogens and Their Receptors on Immunity and Inflammation during Infection. Cancers (Basel) 2022; 14:cancers14040909. [PMID: 35205657 PMCID: PMC8870346 DOI: 10.3390/cancers14040909] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/31/2022] [Accepted: 02/04/2022] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Human health is significantly affected by microbial infections. One of the largest determinants of the outcomes of such infections is the host immune response. Too weak of a response can lead to enhanced spread by the pathogen, while an overstimulated response can lead to immune-induced tissue damage. Thus, to effectively treat infected individuals, it is critical to understand the regulators that control inflammatory responses. Recently, it has become widely accepted that estrogens, a class of sex hormones, are capable of dramatically altering the responses of host cells to microbes. In this review, we discuss how estrogens change the host immune response, as well as how these changes can alter the outcome of the infection for the individual. Abstract Sex hormones, such as estrogen and testosterone, are steroid compounds with well-characterized effects on the coordination and development of vertebrate reproductive systems. Since their discovery, however, it has become clear that these “sex hormones” also regulate/influence a broad range of biological functions. In this review, we will summarize some current findings on how estrogens interact with and regulate inflammation and immunity. Specifically, we will focus on describing the mechanisms by which estrogens alter immune pathway activation, the impact of these changes during infection and the development of long-term immunity, and how different types of estrogens and their respective concentrations mediate these outcomes.
Collapse
Affiliation(s)
- Alfred T. Harding
- Institute for Medical Engineering and Sciences, Massachusetts Institute of Technology, Cambridge, MA 02142, USA;
| | - Nicholas S. Heaton
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
- Correspondence: ; Tel.: +1-919-684-1351; Fax: +1-919-684-2790
| |
Collapse
|
24
|
Wu B, Zhao Q, Li Z, Min Z, Shi M, Nie X, He Q, Gui R. Environmental level bisphenol A accelerates alterations of the reno-cardiac axis by the MAPK cascades in male diabetic rats: An analysis based on transcriptomic profiling and bioinformatics. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 287:117671. [PMID: 34435562 DOI: 10.1016/j.envpol.2021.117671] [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] [Received: 02/23/2021] [Revised: 06/25/2021] [Accepted: 06/26/2021] [Indexed: 06/13/2023]
Abstract
In humans and animal models, the kidneys and cardiovascular systems are negatively affected by BPA from the environment. It is considered that BPA have some potential estrogen-like and non-hormone-like properties. In this study, RNA-sequencing and its-related bioinformatics was used as the basic strategy to clarify the characteristic mechanisms of kidney-heart axis remodeling and dysfunction in diabetic male rats under BPA exposure. We found that continuous BPA exposure in diabetic rats aggravated renal impairment, and caused hemodynamic disorders and dysfunctions. There were 655 and 125 differentially expressed genes in the kidney and heart, respectively. For the kidneys, functional annotation and enrichment, and gene set enrichment analyses identified bile acid secretion related to lipid synthesis and transport, and MAPK cascade pathways. For the heart, these bioinformatics analyses clearly pointed to MAPKs pathways. A total of 12 genes and another total of 6 genes were identified from the kidney tissue and heart tissue, respectively. Western blotting showed that exposure to BPA activated MAPK cascades in both organs. In this study, the exacerbated remodeling of diabetic kidney-heart axis under BPA exposure and diabetes might occur through hemodynamics, metabolism disorders, and the immune-inflammatory response, as well as continuous estrogen-like stimulation, with focus on the MAPK cascades.
Collapse
Affiliation(s)
- Bin Wu
- Department of Blood Transfusion, The Third Xiangya Hospital, Central South University, Changsha, China; Wuhan Hospital of Traditional Chinese and Western Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Physiology, Pathophysiology, Pharmacology and Toxicology (Laboratory of Physiological Science), Hubei University of Arts and Science, Xiangyang, China
| | - Qiangqiang Zhao
- Department of Blood Transfusion, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Zuoneng Li
- Institute of Environment Health and Food Safety, Wuhan Center for Diseases Control and Prevention, Wuhan, China
| | - Zhiteng Min
- Department of Occupational Health, Wuhan Center for Diseases Control and Prevention, Wuhan, China; Key Laboratory of Occupational Hazard Identification and Control of Hubei Province, Wuhan University of Science and Technology, Wuhan, China
| | - Mengdie Shi
- Institute of Environment Health and Food Safety, Wuhan Center for Diseases Control and Prevention, Wuhan, China
| | - Xinmin Nie
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Qingnan He
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Rong Gui
- Department of Blood Transfusion, The Third Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
25
|
Hazlett J, Niemi V, Aiderus A, Powell K, Wise L, Kemp R, Dunbier AK. Oestrogen deprivation induces chemokine production and immune cell recruitment in in vitro and in vivo models of oestrogen receptor-positive breast cancer. Breast Cancer Res 2021; 23:95. [PMID: 34602068 PMCID: PMC8489094 DOI: 10.1186/s13058-021-01472-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Accepted: 09/20/2021] [Indexed: 12/09/2022] Open
Abstract
Background Oestrogen receptor-positive (ER+) breast cancer is commonly treated using endocrine therapies such as aromatase inhibitors which block synthesis of oestradiol, but the influence of this therapy on the immune composition of breast tumours has not been fully explored. Previous findings suggest that tumour infiltrating lymphocytes and immune-related gene expression may be altered by treatment with aromatase inhibitors. However, whether these changes are a direct result of impacts on the host immune system or mediated through tumour cells is not known. We aimed to investigate the effect of oestrogen deprivation on the expression of chemokines and immune infiltration in vitro and in an ER+ immunocompetent mouse model. Methods RT-qPCR and a bead-based Bioplex system were used to investigate the expression of chemokines in MCF-7 breast cancer cells deprived of oestrogen. A migration assay and flow cytometry were used to measure the migration of human peripheral blood mononuclear cells (PBMCs) to MCF-7 cells grown without the main biologically active oestrogen, oestradiol. Using flow cytometry and immunohistochemistry, we examined the immune cell infiltrate into tumours created by injecting SSM3 ER+ breast cancer cells into wild-type, immunocompetent 129/SvEv mice. Results This study demonstrates that oestrogen deprivation increases breast cancer secretion of TNF, CCL5, IL-6, IL-8, and CCL22 and alters total human peripheral blood mononuclear cell migration in an in vitro assay. Oestrogen deprivation of breast cancer cells increases migration of CD4+ T cells and decreases migration of CD11c+ and CD14+ PBMC towards cancer cells. PBMC migration towards breast cancer cells can be reduced by treatment with the non-steroidal anti-inflammatory drugs, aspirin and celecoxib. Treatment with endocrine therapy using the aromatase inhibitor letrozole increases CD4+ T cell infiltration into ER+ breast cancer tumours in immune competent mice. Conclusions These results suggest that anti-oestrogen treatment of ER+ breast cancer cells can alter cytokine production and immune cells in the area surrounding the cancer cells. These findings may have implications for the combination and timing of anti-oestrogen therapies with other therapies. Supplementary Information The online version contains supplementary material available at 10.1186/s13058-021-01472-1.
Collapse
Affiliation(s)
- Jody Hazlett
- Department of Biochemistry, University of Otago, Dunedin, New Zealand.
| | - Virginia Niemi
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Aziz Aiderus
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
| | - Katelyn Powell
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
| | - Lyn Wise
- Department of Pharmacology and Toxicology, University of Otago, Dunedin, New Zealand
| | - Roslyn Kemp
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Anita K Dunbier
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
| |
Collapse
|
26
|
Chakraborty S, Pramanik J, Mahata B. Revisiting steroidogenesis and its role in immune regulation with the advanced tools and technologies. Genes Immun 2021; 22:125-140. [PMID: 34127827 PMCID: PMC8277576 DOI: 10.1038/s41435-021-00139-3] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 05/03/2021] [Accepted: 05/21/2021] [Indexed: 12/19/2022]
Abstract
Historically tools and technologies facilitated scientific discoveries. Steroid hormone research is not an exception. Unfortunately, the dramatic advancement of the field faded this research area and flagged it as a solved topic. However, it should have been the opposite. The area should glitter with its strong foundation and attract next-generation scientists. Over the past century, a myriad of new facts on biochemistry, molecular biology, cell biology, physiology and pathology of the steroid hormones was discovered. Several innovations were made and translated into life-saving treatment strategies such as synthetic steroids, and inhibitors of steroidogenesis and steroid signaling. Steroid molecules exhibit their diverse effects on cell metabolism, salt and water balance, development and function of the reproductive system, pregnancy, and immune-cell function. Despite vigorous research, the molecular basis of the immunomodulatory effect of steroids is still mysterious. The recent excitement on local extra-glandular steroidogenesis in regulating inflammation and immunity is revitalizing the topic with a new perspective. Therefore, here we review the role of steroidogenesis in regulating inflammation and immunity, discuss the unresolved questions, and how this area can bring another golden age of steroid hormone research with the development of new tools and technologies and advancement of the scientific methods.
Collapse
Affiliation(s)
| | - Jhuma Pramanik
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Bidesh Mahata
- Department of Pathology, University of Cambridge, Cambridge, UK.
| |
Collapse
|
27
|
Langton DJ, Bourke SC, Lie BA, Reiff G, Natu S, Darlay R, Burn J, Echevarria C. The influence of HLA genotype on the severity of COVID-19 infection. HLA 2021; 98:14-22. [PMID: 33896121 PMCID: PMC8251294 DOI: 10.1111/tan.14284] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 04/19/2021] [Indexed: 12/17/2022]
Abstract
The impact of COVID‐19 varies markedly, not only between individual patients but also between different populations. We hypothesised that differences in HLA genes might influence this variation. Using next generation sequencing, we analysed the class I and class II classical HLA genes of 147 individuals of European descent experiencing variable clinical outcomes following COVID‐19 infection. Forty‐nine of these patients were admitted to hospital with severe respiratory disease. They had no significant pre‐existing comorbidities. We compared the results to those obtained from a group of 69 asymptomatic hospital workers who evidence of COVID exposure based on blood antibody testing. Allele frequencies in both the severe and asymptomatic groups were compared to local and national healthy controls with adjustments made for age and sex. With the inclusion of hospital staff who had reported localised symptoms only (limited to loss of smell/taste, n = 13) or systemic symptoms not requiring hospital treatment (n = 16), we carried out ordinal logistic regression modelling to determine the relative influence of age, BMI, sex and the presence of specific HLA genes on symptomatology. We found a significant difference in the allele frequency of HLA‐DRB1*04:01 in the severe patient compared to the asymptomatic staff group (5.1% vs. 16.7%, P = .003 after adjustment for age and sex). There was a significantly lower frequency of the haplotype DQA1*01:01‐DQB1*05:01‐DRB1*01:01 in the asymptomatic group compared to the background population (P = .007). Ordinal logistic regression modelling confirmed the significant influence of DRB1*04:01 on the clinical severity of COVID‐19 observed in the cohorts. These alleles are found in greater frequencies in the North Western European population. This regional study provides evidence that HLA genotype influences clinical outcome in COVID‐19 infection. Validation studies must take account of the complex genetic architecture of the immune system across different geographies and ethnicities.
Collapse
Affiliation(s)
- David J Langton
- ExplantLab, The Biosphere, Newcastle Helix, Newcastle-upon-Tyne, UK
| | - Stephen C Bourke
- Northumbria Healthcare NHS Trust, North Tyneside General Hospital, North Shields, Tyne and Wear, UK
| | - Benedicte A Lie
- Department of Medical Genetics, University of Oslo, Oslo, Norway
| | | | | | - Rebecca Darlay
- Newcastle University Translational and Clinical Research Institute, International Centre for Life (for John Burn) and Population & Health Sciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom, for Rebecca Darlay, Newcastle-upon-Tyne, UK
| | - John Burn
- Newcastle University Translational and Clinical Research Institute, International Centre for Life (for John Burn) and Population & Health Sciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom, for Rebecca Darlay, Newcastle-upon-Tyne, UK
| | - Carlos Echevarria
- Translational and Clinical Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| |
Collapse
|
28
|
Serum Creatine Kinase Increases after Acute Strength Training in College Athletes with Menstrual Irregularities. WOMEN 2021. [DOI: 10.3390/women1020007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Chronic menstrual dysfunction and low female sex hormones adversely affect muscular performance in women but studies in college athletes are scarce. A cohort of 18 Japanese, female college athletes at the University of Tsukuba, Japan, were recruited and studied over 3 weeks under 2 conditions. One group had normal menstrual cycling (CYC, 9 athletes) while the other had irregular cycles (DYS, 9 athletes). Hormones and creatine kinase (CK) were measured from blood under both rest (RE) and exercise (EX) conditions. Biceps femoris tendon stiffness was measured by myometry. No differences in age, height, weight, menarche age, or one-repetition maximum weight existed between the groups. The DYS group had persistently low levels of estrogen and progesterone. In the CYC group, the CK level significantly increased at each point immediately post-exercise and 24 h post-exercise compared to pre-exercise in Weeks 1 and 2, and significantly increased at 24 h post-exercise compared to pre-exercise status in Week 3. The DYS group was significantly different only between pre-exercise and 24 h post-exercise over all 3 weeks. The DYS group also suffered from higher biceps femoris tendon stiffness at 24 h post-exercise. Chronic menstrual irregularities in Japanese college athletes increase muscle damage markers in the bloodstream and muscle stiffness after acute strength training.
Collapse
|
29
|
Roche P, Bouhour F. Myasthenia gravis and pregnancy. Rev Neurol (Paris) 2021; 177:215-219. [PMID: 33648779 DOI: 10.1016/j.neurol.2020.09.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 06/26/2020] [Accepted: 09/30/2020] [Indexed: 02/07/2023]
Abstract
Myasthenia gravis is an autoimmune disease characterised by fluctuating muscle weakness, which worsens during activity. It affects particularly scapular and pelvic girdles, axial and bulbar muscles. Myasthenia gravis is twice more frequent in women and symptoms often appear in the second and third decade of life. Thus, a growing number of women affected by this condition become pregnant. To minimise the effects of myasthenia gravis on pregnancy and the newborn, and to avoid myasthenia crisis in the post-partum, the pregnancy must be planned as far as possible. During pregnancy, treatment must be reviewed due to the threat of teratogenic effects (mycophenolate mofetil, rituximab), and the follow-up must be multidisciplinary.
Collapse
Affiliation(s)
- P Roche
- Service d'électroneuromyographie et pathologies neuromusculaires - Hôpital Pierre-Wertheimer - CHU de Lyon HCL - GH Est, 59, boulevard Pinel, Bron cedex, Lyon, France
| | - F Bouhour
- Service d'électroneuromyographie et pathologies neuromusculaires - Hôpital Pierre-Wertheimer - CHU de Lyon HCL - GH Est, 59, boulevard Pinel, Bron cedex, Lyon, France.
| |
Collapse
|
30
|
Kurosawa M, Shikama Y, Furukawa M, Arakaki R, Ishimaru N, Matsushita K. Chemokines Up-Regulated in Epithelial Cells Control Senescence-Associated T Cell Accumulation in Salivary Glands of Aged and Sjögren's Syndrome Model Mice. Int J Mol Sci 2021; 22:ijms22052302. [PMID: 33669065 PMCID: PMC7956724 DOI: 10.3390/ijms22052302] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 02/21/2021] [Accepted: 02/22/2021] [Indexed: 12/29/2022] Open
Abstract
Immunosenescence is characterized by age-associated changes in immunological functions. Although age- and autoimmune-related sialadenitis cause dry mouth (xerostomia), the roles of immunosenescence and cellular senescence in the pathogenesis of sialadenitis remain unknown. We demonstrated that acquired immune cells rather than innate immune cells infiltrated the salivary glands (SG) of aged mice. An analysis of isolated epithelial cells from SG revealed that the expression levels of the chemokine CXCL13 were elevated in aged mice. Senescence-associated T cells (SA-Ts), which secrete large amounts of atypical pro-inflammatory cytokines, are involved in the pathogenesis of metabolic disorders and autoimmune diseases. The present results showed that SA-Ts and B cells, which express the CXCL13 receptor CXCR5, accumulated in the SG of aged mice, particularly females. CD4+ T cells derived from aged mice exhibited stronger in vitro migratory activity toward CXCL13 than those from young mice. In a mouse model of Sjögren’s syndrome (SS), SA-Ts also accumulated in SG, presumably via CXCL12-CXCR4 signaling. Collectively, the present results indicate that SA-Ts accumulate in SG, contribute to the pathogenesis of age- and SS-related sialadenitis by up-regulating chemokines in epithelial cells, and have potential as therapeutic targets for the treatment of xerostomia caused by these types of sialadenitis.
Collapse
Affiliation(s)
- Mie Kurosawa
- Department of Oral Disease Research, National Center for Geriatrics and Gerontology, 7-430 Morioka-cho, Obu 474-8511, Japan; (M.K.); (M.F.); (K.M.)
| | - Yosuke Shikama
- Department of Oral Disease Research, National Center for Geriatrics and Gerontology, 7-430 Morioka-cho, Obu 474-8511, Japan; (M.K.); (M.F.); (K.M.)
- Correspondence: ; Tel.: +81-562-46-2311
| | - Masae Furukawa
- Department of Oral Disease Research, National Center for Geriatrics and Gerontology, 7-430 Morioka-cho, Obu 474-8511, Japan; (M.K.); (M.F.); (K.M.)
| | - Rieko Arakaki
- Department of Oral Molecular Pathology, Tokushima University Graduate School of Biomedical Sciences, 3-18-15 Kuramoto-cho, Tokushima 770-8504, Japan; (R.A.); (N.I.)
| | - Naozumi Ishimaru
- Department of Oral Molecular Pathology, Tokushima University Graduate School of Biomedical Sciences, 3-18-15 Kuramoto-cho, Tokushima 770-8504, Japan; (R.A.); (N.I.)
| | - Kenji Matsushita
- Department of Oral Disease Research, National Center for Geriatrics and Gerontology, 7-430 Morioka-cho, Obu 474-8511, Japan; (M.K.); (M.F.); (K.M.)
| |
Collapse
|
31
|
Peres LC, Townsend MK, Birmann BM, Conejo-Garcia JR, Kim Y, Kubzansky LD, Magpantay LI, Martinez-Maza O, Tworoger SS. Circulating Biomarkers of Inflammation and Ovarian Cancer Risk in the Nurses' Health Studies. Cancer Epidemiol Biomarkers Prev 2021; 30:710-718. [PMID: 33563649 DOI: 10.1158/1055-9965.epi-20-1390] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 12/01/2020] [Accepted: 01/29/2021] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Chronic inflammation is a well-established mechanism of ovarian carcinogenesis; however, the specific immunogenic processes influencing ovarian tumor development remain unclear. In a case-control study nested within the Nurses' Health Study (NHS) and the NHSII, we examined the association between six inflammatory chemokines and cytokines [B-cell activating factor (BAFF), C-X-C motif chemokine ligand 13 (CXCL13), IL8, soluble(s)IL2-receptor-α(Rα), sIL6Rα] and epithelial ovarian cancer risk. METHODS Among 299 epithelial ovarian cancer cases and 334 matched controls, six inflammatory biomarkers were measured in plasma collected 1-24 years before diagnosis or index date using two custom multiplex Luminex panels. ORs and 95% confidence intervals (CI) were estimated for the association between each biomarker and risk using multivariable conditional logistic regression with adjustment for relevant confounders. We additionally assessed heterogeneity in the risk associations by histotype [high-grade serous carcinoma (HGSC) vs. non-HGSC], body mass index, smoking status, menopausal status, and aspirin use. RESULTS Women with the highest versus lowest quartile (Q) levels of CXCL13 had a 72% increased ovarian cancer risk (OR = 1.72; 95% CI = 1.04-2.83; P trend = 0.007). The positive association with CXCL13 was stronger in magnitude for non-HGSC, overweight or obese women, and postmenopausal women, although only menopausal status demonstrated statistically significant heterogeneity (P interaction = 0.04). The remaining biomarkers were not associated with risk. CONCLUSIONS This first evidence that prediagnostic CXCL13, a B-cell chemoattractant, is associated with an increased risk of epithelial ovarian cancer expands current understanding of the role of inflammation in ovarian carcinogenesis. IMPACT CXCL13 may represent a novel biomarker for ovarian cancer.
Collapse
Affiliation(s)
- Lauren C Peres
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida.
| | - Mary K Townsend
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Brenda M Birmann
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Jose R Conejo-Garcia
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Yongjoo Kim
- Department of Social and Behavioral Sciences, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Laura D Kubzansky
- Department of Social and Behavioral Sciences, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Larry I Magpantay
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, California
| | - Otoniel Martinez-Maza
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, California.,Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, California
| | - Shelley S Tworoger
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| |
Collapse
|
32
|
Acheampong DO, Barffour IK, Boye A, Aninagyei E, Ocansey S, Morna MT. Male predisposition to severe COVID-19: Review of evidence and potential therapeutic prospects. Biomed Pharmacother 2020; 131:110748. [PMID: 33152916 PMCID: PMC7480230 DOI: 10.1016/j.biopha.2020.110748] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 09/04/2020] [Accepted: 09/07/2020] [Indexed: 02/07/2023] Open
Abstract
The severe form of COVID-19 has significant sex disparities, with high fatalities commonly reported among males than females. The incidence of COVID-19 has also been higher in males compared with their female counterparts. This trend could be attributed to a better responsive and robust immune system in females. Cytokine storm is one of the pathophysiological features of severe COVID-19, and it occurs as a result of over-activation of immune cells leading to severe inflammation and tissue damage. Nevertheless, it is well modulated in females compared to their male counterparts. Severe inflammation in males is reported to facilitate progression of mild to severe COVID-19. The sex hormones, estrogens and androgens which exist in varying functional levels respectively in females and males are cited as the underlying cause for the differential immune response to COVID-19. Evidence abounds that estrogen modulate the immune system to protect females from severe inflammation and for that matter severe COVID-19. On the contrary, androgen has been implicated in over-activation of immune cells, cytokine storm and the attendant severe inflammation, which perhaps predispose males to severe COVID-19. In this review efforts are made to expand understanding and explain the possible roles of the immune system, the sex hormones and the angiotensin-converting enzyme (ACE) systems in male bias to severe COVID-19. Also, this review explores possible therapeutic avenues including androgen deprivation therapy (ADT), estrogen-based therapy, and ACE inhibitors for consideration in the fight against COVID-19.
Collapse
MESH Headings
- Adolescent
- Adult
- Aged
- Aged, 80 and over
- Androgen Antagonists/pharmacology
- Androgen Antagonists/therapeutic use
- Angiotensin-Converting Enzyme 2
- Angiotensin-Converting Enzyme Inhibitors/pharmacology
- Angiotensin-Converting Enzyme Inhibitors/therapeutic use
- Animals
- Betacoronavirus/physiology
- COVID-19
- Child
- Child, Preschool
- Coronavirus Infections/complications
- Coronavirus Infections/drug therapy
- Coronavirus Infections/epidemiology
- Coronavirus Infections/immunology
- Coronavirus Infections/therapy
- Disease Susceptibility
- Female
- Gonadal Steroid Hormones/physiology
- Humans
- Immunity, Innate
- Infant
- Infant, Newborn
- Inflammation
- Male
- Mice
- Middle Aged
- Pandemics
- Peptidyl-Dipeptidase A/physiology
- Pneumonia, Viral/complications
- Pneumonia, Viral/epidemiology
- Pneumonia, Viral/immunology
- Pneumonia, Viral/therapy
- Prostatic Neoplasms/complications
- Prostatic Neoplasms/drug therapy
- Protein Disulfide-Isomerases/physiology
- Receptors, Cell Surface/physiology
- Receptors, Virus/physiology
- SARS-CoV-2
- Sex Distribution
- Smoking/adverse effects
- Young Adult
- COVID-19 Drug Treatment
Collapse
Affiliation(s)
- Desmond Omane Acheampong
- Department of Biomedical Sciences, School of Allied Health Sciences, College of Health and Allied Sciences, University of Cape Coast, Ghana.
| | - Isaac Kyei Barffour
- Department of Biomedical Sciences, School of Allied Health Sciences, College of Health and Allied Sciences, University of Cape Coast, Ghana
| | - Alex Boye
- Department of Medical Laboratory Science, School of Allied Health Sciences, College of Health and Allied Sciences, University of Cape Coast, Ghana
| | - Enoch Aninagyei
- Department of Biomedical Sciences, School of Basic and Biomedical Sciences, University of Allied Health Sciences, Ho, Ghana
| | - Stephen Ocansey
- Department of Optometry and Vision Science, School of Allied Health Sciences, College of Health and Allied Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Martin Tangnaa Morna
- Department of Surgery, School of Medical Sciences, College of Health and Allied Sciences, University of Cape Coast, Cape Coast, Ghana
| |
Collapse
|
33
|
Hases L, Archer A, Indukuri R, Birgersson M, Savva C, Korach-André M, Williams C. High-fat diet and estrogen impacts the colon and its transcriptome in a sex-dependent manner. Sci Rep 2020; 10:16160. [PMID: 32999402 PMCID: PMC7527340 DOI: 10.1038/s41598-020-73166-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 09/14/2020] [Indexed: 02/07/2023] Open
Abstract
There is a strong association between obesity and colorectal cancer (CRC), especially in men, whereas estrogen protects against both the metabolic syndrome and CRC. Colon is the first organ to respond to high-fat diet (HFD), and estrogen receptor beta (ERβ) can attenuate CRC development. How estrogen impacts the colon under HFD and related sex differences has, however, not been investigated. To dissect this, mice were fed control diet or HFD for 13 weeks and administered receptor-selective estrogenic ligands for the last three weeks. We recorded impact on metabolism, colon crypt proliferation, macrophage infiltration, and the colon transcriptome. We found clear sex differences in the colon transcriptome and in the impact by HFD and estrogens, including on clock genes. ERα-selective activation reduced body weight and generated systemic effects, whereas ERβ-selective activation had local effects in the colon, attenuating HFD-induced macrophage infiltration and epithelial cell proliferation. We here demonstrate how HFD and estrogens modulate the colon microenvironment in a sex- and ER-specific manner.
Collapse
Affiliation(s)
- L Hases
- Science for Life Laboratory, Department of Protein Science, KTH Royal Institute of Technology, Solna, Sweden.,Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - A Archer
- Science for Life Laboratory, Department of Protein Science, KTH Royal Institute of Technology, Solna, Sweden.,Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - R Indukuri
- Science for Life Laboratory, Department of Protein Science, KTH Royal Institute of Technology, Solna, Sweden.,Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - M Birgersson
- Science for Life Laboratory, Department of Protein Science, KTH Royal Institute of Technology, Solna, Sweden.,Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - C Savva
- Department of Medicine, Metabolism Unit and Integrated CardioMetabolic Center (ICMC), Karolinska Institutet and Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - M Korach-André
- Department of Medicine, Metabolism Unit and Integrated CardioMetabolic Center (ICMC), Karolinska Institutet and Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - C Williams
- Science for Life Laboratory, Department of Protein Science, KTH Royal Institute of Technology, Solna, Sweden. .,Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden.
| |
Collapse
|
34
|
Edwards MR, Dai R, Heid B, Cowan C, Werre SR, Cecere T, Ahmed SA. Low-dose 17α-ethinyl estradiol (EE) exposure exacerbates lupus renal disease and modulates immune responses to TLR7/9 agonists in genetically autoimmune-prone mice. Sci Rep 2020; 10:5210. [PMID: 32251357 PMCID: PMC7090002 DOI: 10.1038/s41598-020-62124-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 03/02/2020] [Indexed: 01/30/2023] Open
Abstract
Estrogens have been shown to regulate the immune system and modulate multiple autoimmune diseases. 17α-ethinyl estradiol (EE), a synthetic analog of 17β-estradiol, is prescribed commonly and found in oral contraceptives and hormone replacement therapies. Surprisingly, few studies have investigated the immunoregulatory effects of exposure to EE, especially in autoimmunity. In this study, we exposed autoimmune-prone female MRL/lpr mice to a human-relevant dose of EE through the oral route of exposure. Since lupus patients are prone to infections, groups of mice were injected with viral (Imiquimod, a TLR7 agonist) or bacterial (ODN 2395, a TLR9 agonist) surrogates. We then evaluated autoimmune disease parameters, kidney disease, and response to in vivo TLR7/9 pathogenic signals. EE-exposed mice had increased proteinuria as early as 7 weeks of age. Proteinuria, blood urea nitrogen, and glomerular immune complex deposition were also exacerbated when compared to controls. Production of cytokines by splenic leukocytes were altered in EE-exposed mice. Our study shows that oral exposure to EE, even at a very low dose, can exacerbate azotemia, increase clinical markers of renal disease, enhance glomerular immune complex deposition, and modulate TLR7/9 cytokine production in female MRL/lpr mice. This study may have implications for EE-exposure risk for genetically lupus-prone individuals.
Collapse
Affiliation(s)
- Michael R Edwards
- Infectious Disease Research Facility (IDRF), Department of Biomedical Sciences and Pathobiology, VA-MD College of Vet. Medicine, Virginia Tech, Blacksburg, VA, USA
| | - Rujuan Dai
- Infectious Disease Research Facility (IDRF), Department of Biomedical Sciences and Pathobiology, VA-MD College of Vet. Medicine, Virginia Tech, Blacksburg, VA, USA
| | - Bettina Heid
- Infectious Disease Research Facility (IDRF), Department of Biomedical Sciences and Pathobiology, VA-MD College of Vet. Medicine, Virginia Tech, Blacksburg, VA, USA
| | - Catharine Cowan
- Infectious Disease Research Facility (IDRF), Department of Biomedical Sciences and Pathobiology, VA-MD College of Vet. Medicine, Virginia Tech, Blacksburg, VA, USA
| | - Stephen R Werre
- Population Health Sciences, VA-MD College of Vet. Medicine, Virginia Tech, Blacksburg, VA, USA
| | - Thomas Cecere
- Infectious Disease Research Facility (IDRF), Department of Biomedical Sciences and Pathobiology, VA-MD College of Vet. Medicine, Virginia Tech, Blacksburg, VA, USA
| | - S Ansar Ahmed
- Infectious Disease Research Facility (IDRF), Department of Biomedical Sciences and Pathobiology, VA-MD College of Vet. Medicine, Virginia Tech, Blacksburg, VA, USA.
| |
Collapse
|
35
|
Merrheim J, Villegas J, Van Wassenhove J, Khansa R, Berrih-Aknin S, le Panse R, Dragin N. Estrogen, estrogen-like molecules and autoimmune diseases. Autoimmun Rev 2020; 19:102468. [PMID: 31927086 DOI: 10.1016/j.autrev.2020.102468] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 09/23/2019] [Indexed: 12/13/2022]
Abstract
In western countries, the slope of autoimmune disease (AD) incidence is increasing and affects 5-8% of the population. Mainly prevalent in women, these pathologies are due to thymic tolerance processes breakdown. The female sex hormone, estrogen, is involved in this AD female susceptibility. However, predisposition factors have to act in concert with unknown triggering environmental factors (virus, microbiota, pollution) to initiate AD. Individuals are exposed to various environmental compounds that display endocrine disruption abilities. The cellular effects of some of these molecules may be mediated through the aryl hydrocarbon receptor (AhR). Here, we review the effects of these molecules on the homeostasis of the thymic cells, the immune tolerance intrinsic factors (transcription factors, epigenetic marks) and on the immune tolerance extrinsic factors (microbiota, virus sensibility). This review highlights the contribution of estrogen and endocrine disruptors on the dysregulation of mechanisms sustaining AD development.
Collapse
Affiliation(s)
- Judith Merrheim
- Sorbonne Université, Paris, France; Inserm UMRS 974, Paris, France; AIM, Institute of Myology, Paris, France; Centre de Recherche en Myologie, Sorbonne Université, Inserm UMRS 974, Hôpital La Pitié- Salpêtrière, 105 Bd de l'hôpital, 75013 Paris, France
| | - José Villegas
- Sorbonne Université, Paris, France; Inserm UMRS 974, Paris, France; AIM, Institute of Myology, Paris, France; Centre de Recherche en Myologie, Sorbonne Université, Inserm UMRS 974, Hôpital La Pitié- Salpêtrière, 105 Bd de l'hôpital, 75013 Paris, France
| | - Jérôme Van Wassenhove
- Sorbonne Université, Paris, France; Inserm UMRS 974, Paris, France; AIM, Institute of Myology, Paris, France; Centre de Recherche en Myologie, Sorbonne Université, Inserm UMRS 974, Hôpital La Pitié- Salpêtrière, 105 Bd de l'hôpital, 75013 Paris, France
| | - Rémi Khansa
- Sorbonne Université, Paris, France; Inserm UMRS 974, Paris, France; AIM, Institute of Myology, Paris, France; Centre de Recherche en Myologie, Sorbonne Université, Inserm UMRS 974, Hôpital La Pitié- Salpêtrière, 105 Bd de l'hôpital, 75013 Paris, France
| | - Sonia Berrih-Aknin
- Sorbonne Université, Paris, France; Inserm UMRS 974, Paris, France; AIM, Institute of Myology, Paris, France; Centre de Recherche en Myologie, Sorbonne Université, Inserm UMRS 974, Hôpital La Pitié- Salpêtrière, 105 Bd de l'hôpital, 75013 Paris, France
| | - Rozen le Panse
- Sorbonne Université, Paris, France; Inserm UMRS 974, Paris, France; AIM, Institute of Myology, Paris, France; Centre de Recherche en Myologie, Sorbonne Université, Inserm UMRS 974, Hôpital La Pitié- Salpêtrière, 105 Bd de l'hôpital, 75013 Paris, France
| | - Nadine Dragin
- Sorbonne Université, Paris, France; Inserm UMRS 974, Paris, France; Inovarion, Paris, France; Centre de Recherche en Myologie, Sorbonne Université, Inserm UMRS 974, Hôpital La Pitié- Salpêtrière, 105 Bd de l'hôpital, 75013 Paris, France.
| |
Collapse
|
36
|
Zhong H, Zhao C, Luo S. HLA in myasthenia gravis: From superficial correlation to underlying mechanism. Autoimmun Rev 2019; 18:102349. [DOI: 10.1016/j.autrev.2019.102349] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 03/01/2019] [Indexed: 12/26/2022]
|
37
|
Villegas JA, Bayer AC, Ider K, Bismuth J, Truffault F, Roussin R, Santelmo N, Le Panse R, Berrih-Aknin S, Dragin N. Il-23/Th17 cell pathway: A promising target to alleviate thymic inflammation maintenance in myasthenia gravis. J Autoimmun 2019; 98:59-73. [PMID: 30578016 DOI: 10.1016/j.jaut.2018.11.005] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 11/29/2018] [Accepted: 11/30/2018] [Indexed: 12/27/2022]
Abstract
IL-23/Th17 pathway has been identified to sustain inflammatory condition in several autoimmune diseases and therefore being targeted in various therapeutic and effective approaches. Patients affected with autoimmune myasthenia gravis exhibit a disease effector tissue, the thymus, that harbors ectopic germinal centers that sustain production of auto-antibodies, targeting proteins located in the neuromuscular junction, cause of the organ-specific chronic autoimmune disease. The present study aims to investigate the IL-23/Th17 cell pathway in the thymic inflammatory and pathogenic events. We found that thymuses of MG patients displayed overexpression of Interleukin-17, signature cytokine of activated Th17 cells. This activation was sustained by a higher secretion of Interleukin-23 by TEC, in addition to the increased expression of cytokines involved in Th17 cell development. The overexpression of Interleukin-23 was due to a dysregulation of interferon type I pathway. Besides, Interleukin-17 secreted, and Th17 cells were localized around thymic ectopic germinal centers. These cells expressed podoplanin, a protein involved in B-cell maturation and antibody secretion. Finally, production of Interleukin-23 was also promoted by Interleukin-17 secreted itself by Th17 cells, highlighting a chronic loop of inflammation sustained by thymic cell interaction. Activation of the IL-23/Th17 pathway in the thymus of autoimmune myasthenia gravis patients creates an unstoppable loop of inflammation that may participate in ectopic germinal center maintenance. To alleviate the physio-pathological events in myasthenia gravis patients, this pathway may be considered as a new therapeutic target.
Collapse
Affiliation(s)
- José A Villegas
- Sorbonne Université, Centre de Recherche en Myologie, Paris, France; INSERM UMRS 974, Paris, France; AIM, Institute of Myology, Paris, France
| | - Alexandra C Bayer
- Sorbonne Université, Centre de Recherche en Myologie, Paris, France; INSERM UMRS 974, Paris, France; AIM, Institute of Myology, Paris, France
| | - Katia Ider
- Sorbonne Université, Centre de Recherche en Myologie, Paris, France; INSERM UMRS 974, Paris, France; AIM, Institute of Myology, Paris, France
| | - Jacky Bismuth
- Sorbonne Université, Centre de Recherche en Myologie, Paris, France; INSERM UMRS 974, Paris, France; AIM, Institute of Myology, Paris, France
| | - Frédérique Truffault
- Sorbonne Université, Centre de Recherche en Myologie, Paris, France; INSERM UMRS 974, Paris, France; AIM, Institute of Myology, Paris, France
| | | | | | - Rozen Le Panse
- Sorbonne Université, Centre de Recherche en Myologie, Paris, France; INSERM UMRS 974, Paris, France; AIM, Institute of Myology, Paris, France.
| | - Sonia Berrih-Aknin
- Sorbonne Université, Centre de Recherche en Myologie, Paris, France; INSERM UMRS 974, Paris, France; AIM, Institute of Myology, Paris, France
| | - Nadine Dragin
- Sorbonne Université, Centre de Recherche en Myologie, Paris, France; INSERM UMRS 974, Paris, France; AIM, Institute of Myology, Paris, France; Inovarion, Paris, France.
| |
Collapse
|
38
|
Balogh A, Karpati E, Schneider AE, Hetey S, Szilagyi A, Juhasz K, Laszlo G, Hupuczi P, Zavodszky P, Papp Z, Matko J, Than NG. Sex hormone-binding globulin provides a novel entry pathway for estradiol and influences subsequent signaling in lymphocytes via membrane receptor. Sci Rep 2019; 9:4. [PMID: 30626909 PMCID: PMC6327036 DOI: 10.1038/s41598-018-36882-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 11/22/2018] [Indexed: 02/07/2023] Open
Abstract
The complex effects of estradiol on non-reproductive tissues/cells, including lymphoid tissues and immunocytes, have increasingly been explored. However, the role of sex hormone binding globulin (SHBG) in the regulation of these genomic and non-genomic actions of estradiol is controversial. Moreover, the expression of SHBG and its internalization by potential receptors, as well as the influence of SHBG on estradiol uptake and signaling in lymphocytes has remained unexplored. Here, we found that human and mouse T cells expressed SHBG intrinsically. In addition, B lymphoid cell lines as well as both primary B and T lymphocytes bound and internalized external SHBG, and the amount of plasma membrane-bound SHBG decreased in B cells of pregnant compared to non-pregnant women. As potential mediators of this process, SHBG receptor candidates expressed by lymphocytes were identified in silico, including estrogen receptor (ER) alpha. Furthermore, cell surface-bound SHBG was detected in close proximity to membrane ERs while highly colocalizing with lipid rafts. The SHBG-membrane ER interaction was found functional since SHBG promoted estradiol uptake by lymphocytes and subsequently influenced Erk1/2 phosphorylation. In conclusion, the SHBG-SHBG receptor-membrane ER complex participates in the rapid estradiol signaling in lymphocytes, and this pathway may be altered in B cells in pregnant women.
Collapse
Affiliation(s)
- Andrea Balogh
- Department of Immunology, Eotvos Lorand University, Budapest, Hungary.,Systems Biology of Reproduction Lendulet Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Eva Karpati
- Department of Immunology, Eotvos Lorand University, Budapest, Hungary.,Systems Biology of Reproduction Lendulet Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | | | - Szabolcs Hetey
- Systems Biology of Reproduction Lendulet Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Andras Szilagyi
- Systems Biology of Reproduction Lendulet Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary.,Laboratory of Structural Biophysics, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Kata Juhasz
- Systems Biology of Reproduction Lendulet Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Gloria Laszlo
- Department of Immunology, Eotvos Lorand University, Budapest, Hungary
| | - Petronella Hupuczi
- Maternity Private Department, Kutvolgyi Clinical Block, Semmelweis University, Budapest, Hungary
| | - Peter Zavodszky
- Laboratory of Structural Biophysics, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Zoltan Papp
- Maternity Private Department, Kutvolgyi Clinical Block, Semmelweis University, Budapest, Hungary
| | - Janos Matko
- Department of Immunology, Eotvos Lorand University, Budapest, Hungary.
| | - Nandor Gabor Than
- Systems Biology of Reproduction Lendulet Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary. .,Maternity Private Department, Kutvolgyi Clinical Block, Semmelweis University, Budapest, Hungary. .,First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
39
|
Dai R, Edwards MR, Heid B, Ahmed SA. 17β-Estradiol and 17α-Ethinyl Estradiol Exhibit Immunologic and Epigenetic Regulatory Effects in NZB/WF1 Female Mice. Endocrinology 2019; 160:101-118. [PMID: 30418530 PMCID: PMC6305969 DOI: 10.1210/en.2018-00824] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Accepted: 11/06/2018] [Indexed: 02/07/2023]
Abstract
17α-Ethinyl estradiol (EE), a synthetic analog of natural estrogen 17β-estradiol (E2), is extensively used in hormonal contraceptives and estrogen replacement therapy, and it has also been found in sewage effluents. Given that E2 is a well-known immunomodulator, surprisingly there has been only limited information on the cellular and molecular immunologic consequences of exposure to EE. To address this fundamental gap, we directly compared the effects of EE with E2 on splenic leukocytes of New Zealand Black × New Zealand White F1 progeny (NZB/WF1) mice during the preautoimmune period. We found that EE and E2 have common, as well as distinctive, immunologic effects, with EE exposure resulting in more profound effects. Both EE and E2 increased numbers of splenic neutrophils, enhanced neutrophil serine proteases and myeloperoxidase expression, promoted the production of nitric oxide and monocyte chemoattractant protein-1, and altered adaptive immune T cell subsets. However, activation of splenic leukocytes through the T cell receptor or Toll-like receptor (TLR)4 revealed not only common (IL-10), but also hormone-specific alterations of cytokines (IFNγ, IL-1β, ΤΝFα, IL-2). Furthermore, in EE-exposed mice, TLR9 stimulation suppressed IFNα, in contrast to increased IFNα from E2-exposed mice. EE and E2 regulated common and hormone-specific expression of immune-related genes. Furthermore, EE exposure resulted in more marked alterations in miRNA expression levels than for E2. Only EE was able to reduce global DNA methylation significantly in splenic leukocytes. Taken together, our novel data revealed that EE and E2 exposure confers more similar effects in innate immune system-related cell development and responses, but has more differential regulatory effects in adaptive immune-related cell development and responses.
Collapse
Affiliation(s)
- Rujuan Dai
- Department of Biomedical Sciences and Pathobiology, Infectious Disease Research Facility (IDRF), Virginia-Maryland College of Veterinary Medicine, Virginia Tech/Virginia Polytechnic Institute and State University, Blacksburg, Virginia
| | - Michael R Edwards
- Department of Biomedical Sciences and Pathobiology, Infectious Disease Research Facility (IDRF), Virginia-Maryland College of Veterinary Medicine, Virginia Tech/Virginia Polytechnic Institute and State University, Blacksburg, Virginia
| | - Bettina Heid
- Department of Biomedical Sciences and Pathobiology, Infectious Disease Research Facility (IDRF), Virginia-Maryland College of Veterinary Medicine, Virginia Tech/Virginia Polytechnic Institute and State University, Blacksburg, Virginia
| | - S Ansar Ahmed
- Department of Biomedical Sciences and Pathobiology, Infectious Disease Research Facility (IDRF), Virginia-Maryland College of Veterinary Medicine, Virginia Tech/Virginia Polytechnic Institute and State University, Blacksburg, Virginia
| |
Collapse
|
40
|
Villegas JA, Gradolatto A, Truffault F, Roussin R, Berrih-Aknin S, Le Panse R, Dragin N. Cultured Human Thymic-Derived Cells Display Medullary Thymic Epithelial Cell Phenotype and Functionality. Front Immunol 2018; 9:1663. [PMID: 30083154 PMCID: PMC6064927 DOI: 10.3389/fimmu.2018.01663] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 07/04/2018] [Indexed: 12/13/2022] Open
Abstract
Thymic epithelial cells are one of the main components of the thymic microenvironment required for T-cell development. In this work, we describe an efficient method free of enzymatic and Facs-sorted methods to culture human medullary thymic epithelial cells without affecting the cell phenotypic, physiologic and functional features. Human medulla thymic epithelial cells (mTECs) are obtained by culturing thymic biopsies explants. After 7 days of primo-culture, mTECs keep their ability to express key molecules involved in immune tolerance processes such as autoimmune regulator, tissue-specific antigens, chemokines, and cytokines. In addition, the cells sensor their cultured environment and consequently adjust their gene expression network. Therefore, we describe and provide a human mTEC model that may be used to test the effect of various molecules on thymic epithelial cell homeostasis and physiology. This method should allow the investigations of the specificities and the knowledge of human mTECs in normal or pathological conditions and therefore discontinue the extrapolations done on the murine models.
Collapse
Affiliation(s)
- José A Villegas
- INSERM, AIM, Center of Research in Myology, UMRS974, Sorbonne University, Paris, France
| | - Angeline Gradolatto
- INSERM, AIM, Center of Research in Myology, UMRS974, Sorbonne University, Paris, France
| | - Frédérique Truffault
- INSERM, AIM, Center of Research in Myology, UMRS974, Sorbonne University, Paris, France
| | | | - Sonia Berrih-Aknin
- INSERM, AIM, Center of Research in Myology, UMRS974, Sorbonne University, Paris, France
| | - Rozen Le Panse
- INSERM, AIM, Center of Research in Myology, UMRS974, Sorbonne University, Paris, France
| | - Nadine Dragin
- INSERM, AIM, Center of Research in Myology, UMRS974, Sorbonne University, Paris, France.,Inovarion, Paris, France
| |
Collapse
|
41
|
Li QR, Ni WP, Lei NJ, Yang JY, Xuan XY, Liu PP, Gong GM, Yan F, Feng YS, Zhao R, Du Y. The overexpression of Fra1 disorders the inflammatory cytokine secretion by mTEC of myasthenia gravis thymus. Scand J Immunol 2018; 88:e12676. [PMID: 29807388 DOI: 10.1111/sji.12676] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 05/22/2018] [Indexed: 12/01/2022]
Abstract
The thymus of a myasthenia gravis (MG) patient is often accompanied by and effected with follicular hyperplasia. Inflammatory cytokines in thymus induce the formation of germinal centres (GC). MG thymic inflammatory cytokines are predominantly secreted by stromal cells. Our previous studies revealed that the expression level of the Fra1 protein, which is a Fos member of the activator protein 1 transcription factors (AP-1), was higher in the MG thymus compared with that of the normal thymus. Based on that, we demonstrated that Fra1 was mainly expressed in medulla thymic epithelial cells (mTECs) and that the rate of Fra1 positive mTECs in the MG thymus was higher than normal. In vitro, we found that the expression of CCL-5, CCL-19 and CCL-21 could be regulated by Fra1 in mTEC and that IL-1β, IL-6, IL-8 and ICAM1 were downregulated in the Fra1 overexpression group and upregulated in the Fra1 knock-down group. Meanwhile, we detected that the expression levels of suppressor of cytokine signalling 3 (SOCS3) were significantly upregulated along with the overexpression of Fra1. Hence, we considered that the overexpression of Fra1 disrupted inflammatory cytokine secretion by mTEC in the MG thymus and that STAT3 and SOCS3 were strongly involved in this process.
Collapse
Affiliation(s)
- Q-R Li
- Department of Immunology, School of Basic Medical, Zhengzhou University, Zhengzhou, China
| | - W-P Ni
- Department of Immunology, School of Basic Medical, Zhengzhou University, Zhengzhou, China
| | - N-J Lei
- Department of Immunology, School of Basic Medical, Zhengzhou University, Zhengzhou, China
| | - J-Y Yang
- College of Veterinary Medicine, North West Agriculture and Forestry University, Zhengzhou, China
| | - X-Y Xuan
- Department of Immunology, School of Basic Medical, Zhengzhou University, Zhengzhou, China
| | - P-P Liu
- Department of Immunology, School of Basic Medical, Zhengzhou University, Zhengzhou, China
| | - G-M Gong
- Department of Immunology, School of Basic Medical, Zhengzhou University, Zhengzhou, China
| | - F Yan
- Department of Immunology, School of Basic Medical, Zhengzhou University, Zhengzhou, China
| | - Y-S Feng
- Department of Immunology, School of Basic Medical, Zhengzhou University, Zhengzhou, China
| | - R Zhao
- Department of Immunology, School of Basic Medical, Zhengzhou University, Zhengzhou, China
| | - Y Du
- Department of Immunology, School of Basic Medical, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
42
|
Edwards M, Dai R, Ahmed SA. Our Environment Shapes Us: The Importance of Environment and Sex Differences in Regulation of Autoantibody Production. Front Immunol 2018; 9:478. [PMID: 29662485 PMCID: PMC5890161 DOI: 10.3389/fimmu.2018.00478] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 02/22/2018] [Indexed: 01/17/2023] Open
Abstract
Consequential differences exist between the male and female immune systems’ ability to respond to pathogens, environmental insults or self-antigens, and subsequent effects on immunoregulation. In general, females when compared with their male counterparts, respond to pathogenic stimuli and vaccines more robustly, with heightened production of antibodies, pro-inflammatory cytokines, and chemokines. While the precise reasons for sex differences in immune response to different stimuli are not yet well understood, females are more resistant to infectious diseases and much more likely to develop autoimmune diseases. Intrinsic (i.e., sex hormones, sex chromosomes, etc.) and extrinsic (microbiome composition, external triggers, and immune modulators) factors appear to impact the overall outcome of immune responses between sexes. Evidence suggests that interactions between environmental contaminants [e.g., endocrine disrupting chemicals (EDCs)] and host leukocytes affect the ability of the immune system to mount a response to exogenous and endogenous insults, and/or return to normal activity following clearance of the threat. Inherently, males and females have differential immune response to external triggers. In this review, we describe how environmental chemicals, including EDCs, may have sex differential influence on the outcome of immune responses through alterations in epigenetic status (such as modulation of microRNA expression, gene methylation, or histone modification status), direct and indirect activation of the estrogen receptors to drive hormonal effects, and differential modulation of microbial sensing and composition of host microbiota. Taken together, an intriguing question develops as to how an individual’s environment directly and indirectly contributes to an altered immune response, dysregulation of autoantibody production, and influence autoimmune disease development. Few studies exist utilizing well-controlled cohorts of both sexes to explore the sex differences in response to EDC exposure and the effects on autoimmune disease development. Translational studies incorporating multiple environmental factors in animal models of autoimmune disease are necessary to determine the interrelationships that occur between potential etiopathological factors. The presence or absence of autoantibodies is not a reliable predictor of disease. Therefore, future studies should incorporate all the susceptibility/influencing factors, coupled with individual genomics, epigenomics, and proteomics, to develop a model that better predicts, diagnoses, and treats autoimmune diseases in a personalized-medicine fashion.
Collapse
Affiliation(s)
- Michael Edwards
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| | - Rujuan Dai
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| | - S Ansar Ahmed
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| |
Collapse
|
43
|
Berrih-Aknin S, Panse RL, Dragin N. AIRE: a missing link to explain female susceptibility to autoimmune diseases. Ann N Y Acad Sci 2018; 1412:21-32. [PMID: 29291257 DOI: 10.1111/nyas.13529] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 09/20/2017] [Accepted: 09/25/2017] [Indexed: 12/13/2022]
Abstract
Women are more susceptible to autoimmune diseases than men. Autoimmunity results from tolerance breakdown toward self-components. Recently, three transcription modulators were identified in medullary thymic epithelial cells that orchestrate immune central tolerance processes: the autoimmune regulator (AIRE), FEZ family zinc finger 2 (FEZF2 or FEZ1), and PR domain zinc finger protein 1 (PRDM1). Interestingly, these three transcription modulators regulate nonredundant tissue-specific antigen subsets and thus cover broad antigen diversity. Recent data from different groups demonstrated that sex hormones (estrogen and testosterone) are involved in the regulation of thymic AIRE expression in humans and mice through direct transcriptional modulation and epigenetic changes. As a consequence, AIRE displays gender-biased thymic expression, with females showing a lower expression compared with males, a finding that could explain the female susceptibility to autoimmune diseases. So far, FEZF2 has not been related to an increased gender bias in autoimmune disease. PRDM1 expression has not been shown to display gender-differential thymic expression, but its expression level and its gene polymorphisms are associated with female-dependent autoimmune disease risk. Altogether, various studies have demonstrated that increased female susceptibility to autoimmune diseases is in part a consequence of hormone-driven reduced thymic AIRE expression.
Collapse
Affiliation(s)
- Sonia Berrih-Aknin
- UPMC Sorbonne Universities, Paris, France
- INSERM U974, Paris, France
- AIM, Institute of Myology, Paris, France
| | - Rozen Le Panse
- UPMC Sorbonne Universities, Paris, France
- INSERM U974, Paris, France
- AIM, Institute of Myology, Paris, France
| | - Nadine Dragin
- UPMC Sorbonne Universities, Paris, France
- INSERM U974, Paris, France
- AIM, Institute of Myology, Paris, France
- Inovarion, Paris, France
| |
Collapse
|