1
|
Henze L, Will N, Lee D, Haas V, Casar C, Meyer J, Stein S, Mangler F, Steinmann S, Poch T, Krause J, Fuss J, Schröder J, Kulle AE, Holterhus PM, Bonn S, Altfeld M, Huber S, Lohse AW, Schwinge D, Schramm C. Testosterone affects female CD4+ T cells in healthy individuals and autoimmune liver diseases. JCI Insight 2025; 10:e184544. [PMID: 40260919 PMCID: PMC12016935 DOI: 10.1172/jci.insight.184544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 03/04/2025] [Indexed: 04/24/2025] Open
Abstract
Autoimmune hepatitis (AIH) and primary biliary cholangitis (PBC) are autoimmune liver diseases with strong female predominance. They are caused by T cell-mediated injury of hepatic parenchymal cells, but the mechanisms underlying this sex bias are unknown. Here, we investigated whether testosterone contributes to T cell activation in women with PBC. Compared with sex- and age-matched healthy controls (n = 23), cisgender (cis) women with PBC (n = 24) demonstrated decreased testosterone serum levels and proinflammatory CD4+ T cell profile in peripheral blood. Testosterone suppressed the expression of TNF and IFN-γ by human CD4+ T cells in vitro. In trans men receiving gender-affirming hormone therapy (GAHT) (n = 25), testosterone affected CD4+ T cell function by inhibiting Th1 and Th17 differentiation and by supporting the differentiation into regulatory Treg. Mechanistically, we provide evidence for a direct effect of testosterone on T cells using mice with T cell-specific deletion of the cytosolic androgen receptor. Supporting a role for testosterone in autoimmune liver disease, we observed an improved disease course and profound changes in T cell states in a trans man with AIH/primary sclerosing cholangitis (PSC) variant syndrome receiving GAHT. We here report a direct effect of testosterone on CD4+ T cells that may contribute to future personalized treatment strategies.
Collapse
Affiliation(s)
| | | | | | | | - Christian Casar
- 1st Department of Medicine, and
- Bioinformatics Core, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | | | | | | | | | | | - Johannes Fuss
- Institute of Forensic Psychiatry and Sex Research, Center for Translational Neuro and Behavioral Sciences, University of Duisburg-Essen, Essen, Germany
| | - Johanna Schröder
- Institute for Clinical Psychology and Psychotherapy, Department for Psychology, Medical School Hamburg, Hamburg, Germany
| | - Alexandra E. Kulle
- Division of Pediatric Endocrinology and Diabetes, Department of Children and Adolescent Medicine, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Paul-Martin Holterhus
- Division of Pediatric Endocrinology and Diabetes, Department of Children and Adolescent Medicine, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Stefan Bonn
- Institute of Medical Systems Biology and
- Center for Biomedical AI, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marcus Altfeld
- Research Department Virus Immunology, Leibniz Institute of Virology, Hamburg, Germany
| | - Samuel Huber
- 1st Department of Medicine, and
- Hamburg Center for Translational Immunology and
| | - Ansgar W. Lohse
- 1st Department of Medicine, and
- Hamburg Center for Translational Immunology and
| | | | - Christoph Schramm
- 1st Department of Medicine, and
- Hamburg Center for Translational Immunology and
- Martin Zeitz Center for Rare Diseases and, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
2
|
Pinzon-Hoyos N, Li Y, McGee M, Poolos NP, Marchi N, Brewster AL. Drug-resistant epilepsy associated with peripheral complement decreases and sex-specific cytokine imbalances: a pilot study. Sci Rep 2025; 15:5096. [PMID: 39934240 PMCID: PMC11814292 DOI: 10.1038/s41598-025-88654-5] [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: 11/18/2024] [Accepted: 01/29/2025] [Indexed: 02/13/2025] Open
Abstract
Drug-resistant epilepsy (DRE) presents significant challenges in treatment and management. While seizure-related alterations in peripheral immune players are increasingly recognized, the involvement of the immune complement system remains insufficiently explored in DRE. We studied complement components and their relationship to cytokine profiles in serum samples from 46 DRE patients and 45 matched healthy controls. We examined relationships between these molecules and clinical outcomes, including epilepsy duration, intelligence scores, and age. We identified DRE-associated complement decreases, including reduced levels of C1q, Factor H, C4, C4b, C3, and C3b/iC3b, as well as elevated bFGF. DRE females showed dysregulation of the classical complement pathway and lower TNFα and interleukin-8 compared to healthy females. DRE males exhibited dysregulation of the classical, lectin, and terminal complement pathways, with trends of increased CCL2 and CCL5 compared to healthy males. Specific complement and inflammatory markers (C2, IL-8, and IL-9) correlated with full-scale IQ scores in DRE patients. Our study reveals significantly lower levels of circulating complement components in DRE and sex-specific complement dysregulation and cytokine imbalances. These findings suggest an underlying peripheral immune system vulnerability that may be sex-dependent and warrants further investigation in DRE.
Collapse
Affiliation(s)
- Nicole Pinzon-Hoyos
- Department of Biological Sciences, Dedman College of Humanities & Science, Southern Methodist University, 6501 Airline Rd, Dallas, TX, 75205, USA
| | - Yibo Li
- Department of Biological Sciences, Dedman College of Humanities & Science, Southern Methodist University, 6501 Airline Rd, Dallas, TX, 75205, USA
| | - Monnie McGee
- Department of Statistics and Data Science, Southern Methodist University, Dallas, TX, USA
| | - Nicholas P Poolos
- Department of Neurology and Regional Epilepsy Center, University of Washington, Seattle, WA, USA
| | - Nicola Marchi
- University of Montpellier, CNRS, INSERM, Institut de Génomique Fonctionnelle, Montpellier, France
| | - Amy L Brewster
- Department of Biological Sciences, Dedman College of Humanities & Science, Southern Methodist University, 6501 Airline Rd, Dallas, TX, 75205, USA.
| |
Collapse
|
3
|
Roerink AMC, Nelissen RGHH, Holder C, Graves SE, Dunbar M, Bohm E, Grimberg AW, Steinbrück A, Dale H, Fenstad AM, Blom AW, Lenguerrand E, Frampton C, Willems T, Victor J, Espallargues M, Arias-de la Torre J, Ciminello E, Torre M, Pijls BG. Sex-based differences in risk of revision for infection after hip, knee, shoulder, and ankle arthroplasty in osteoarthritis patients: a multinational registry study of 4,800,000 implants. Acta Orthop 2024; 95:730-736. [PMID: 39656214 PMCID: PMC11632196 DOI: 10.2340/17453674.2024.42183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 10/02/2024] [Indexed: 12/13/2024] Open
Abstract
BACKGROUND AND PURPOSE We aimed to determine sex differences for periprosthetic joint infections after primary arthroplasty of the hip, knee, ankle, and shoulder in osteoarthritis patients in an international perspective. METHODS This is a multinational combined arthroplasty registry study. Each arthroplasty registry performed Cox-regression analysis of their data and reported the crude and adjusted hazard ratios (HR) with an a priori designed data form. A random-effects model was used to pool these HRs to estimate an overall HR with 95% confidence interval (CI). Adjustment was undertaken for patient age, BMI, ASA grade, type of fixation, and type of implant. 9 arthroplasty registries participated. Patients who received primary total joint arthroplasty for primary osteoarthritis were considered: 2,134,313 hip arthroplasties, 2,658,237 knee arthroplasties, 57,889 shoulder arthroplasties, and 8,445 ankle arthroplasties. We calculated hazard ratios (HR) for the overall risk of complete revision due to infection for each implant type and follow-up. RESULTS The pooled HR for revision due to infection for men compared with women at 1-year follow-up was 1.60 (95% confidence interval [CI] 1.42-1.80) for hip arthroplasties; 2.06 (CI 1.90-2.46) for knee arthroplasties; 4.51 (CI 2.99-6.80) for shoulder arthroplasties; and 0.87 (CI 0.46-1.62) for ankle arthroplasties. These results remained consistent over time and were identified in both unadjusted and adjusted models. CONCLUSION Men have a higher risk of revision due to infection than women after primary hip, knee, and shoulder arthroplasty. No evidence of difference was found for ankle arthroplasty. These elevated relative risks persist in the fully adjusted investigations and over the 10-year postoperative period studied.
Collapse
MESH Headings
- Humans
- Male
- Female
- Registries
- Reoperation/statistics & numerical data
- Prosthesis-Related Infections/epidemiology
- Prosthesis-Related Infections/etiology
- Aged
- Middle Aged
- Sex Factors
- Risk Factors
- Osteoarthritis/surgery
- Arthroplasty, Replacement, Hip/adverse effects
- Arthroplasty, Replacement, Ankle/adverse effects
- Arthroplasty, Replacement, Shoulder/adverse effects
- Arthroplasty, Replacement, Knee/adverse effects
- Proportional Hazards Models
- Osteoarthritis, Hip/surgery
- Arthroplasty, Replacement/adverse effects
Collapse
Affiliation(s)
- Anne M C Roerink
- Department of Orthopaedics, Leiden University Medical Center, Leiden, The Netherlands
| | - Rob G H H Nelissen
- Department of Orthopaedics, Leiden University Medical Center, Leiden, The Netherlands
| | - Carl Holder
- South Australian Health and Medical Research Institute (SAHMRI), Adelaide, Australia
| | - Stephen E Graves
- Australian Orthopaedic Association National Joint Replacement Registry (AOANJRR), Adelaide; Clinical and Health Sciences, University of South Australia, Adelaide, Australia
| | - Michael Dunbar
- Division of Orthopaedics, Dalhousie University, Halifax, Nova Scotia; Canadian Joint Replacement Registry, Canada
| | - Eric Bohm
- Canadian Joint Replacement Registry; Concordia Joint Replacement Group, University of Manitoba, Winnipeg, Canada
| | - Alexander W Grimberg
- German Arthroplasty Registry (EPRD Deutsche Endoprothesenregister gGmbH), Berlin, Germany
| | - Arnd Steinbrück
- German Arthroplasty Registry (EPRD Deutsche Endoprothesenregister gGmbH), Berlin, Germany
| | - Håvard Dale
- The Norwegian Arthroplasty Register, Department of Orthopedic Surgery, Haukeland University Hospital, Bergen; Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Anne Marie Fenstad
- The Norwegian Arthroplasty Register, Department of Orthopedic Surgery, Haukeland University Hospital, Bergen, Norway
| | - Ashley W Blom
- Bristol Medical School, University of Bristol, Bristol, UK
| | | | | | - Tine Willems
- Department of Rehabilitation Sciences, Ghent University, Ghent, Belgium
| | - Jan Victor
- Department of Orthopedics and Traumatology; Ghent University, Ghent University Hospital, Ghent, Belgium
| | - Mireia Espallargues
- gència de Qualitat i Avaluació Sanitàries de Catalunya (AQuAS), Barcelona, Spain
| | - Jorge Arias-de la Torre
- Agència de Qualitat i Avaluació Sanitàries de Catalunya (AQuAS), Barcelona, Spain; 17 Care in Long Term Conditions Research Division, King's College London, London, UK; CIBER Epidemiology and Public Health (CIBERESP), Madrid; Institute of Biomedicine (IBIOMED). Universidad de León, León, Spain
| | - Enrico Ciminello
- Italian Arthroplasty Registry (RIAP), Rome; Italian National Institute of Health, Rome, Italy
| | - Marina Torre
- Italian Arthroplasty Registry (RIAP), Rome; Italian National Institute of Health, Rome, Italy
| | - Bart G Pijls
- Department of Orthopaedics, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
4
|
Pinzon-Hoyos N, Li Y, McGee M, Poolos NP, Marchi N, Brewster AL. Sex-Specific Complement and Cytokine Imbalances in Drug-Resistant Epilepsy: Biomarkers of Immune Vulnerability. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.16.612934. [PMID: 39345496 PMCID: PMC11429787 DOI: 10.1101/2024.09.16.612934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Objective Drug-resistant epilepsy (DRE) poses significant challenges in treatment and management. While seizure-related alterations in peripheral immune players are increasingly recognized, the involvement of the complement system, central to immune function, remains insufficiently explored in DRE. This study aimed to investigate the levels of complement system components and their association with cytokine profiles in patients with DRE. Methods We analyzed serum samples from DRE patients (n = 46) and age- and sex-matched healthy controls (n = 45). Complement components and cytokines were quantified using Multi- and Single-plex ELISA. Statistical analyses examined relationships between complement molecules, cytokines, and clinical outcomes including epilepsy duration, Full-Scale Intelligence Quotient (FSIQ) scores, and age. Results We found common alterations in all DRE cases, including significant complement deficiencies (C1q, Factor H, C4, C4b, C3, and C3b/iC3b) and detectable bFGF levels. DRE females showed significantly lower levels of TNFα and IL-8 compared to healthy females. We observed a trend towards elevated CCL2 and CCL5 levels in DRE males compared to healthy males. These findings suggest potential sex dimorphism in immune profiles. Our analysis also indicated associations between specific complement and inflammatory markers (C2, IL-8, and IL-9) and Full-Scale Intelligence Quotient (FSIQ) scores in DRE patients. Interpretation Our study reveals sex-specific peripheral complement deficiencies and cytokine dysregulation in DRE patients, indicating an underlying immune system vulnerability. These findings provide new insights into DRE mechanisms, potentially guiding future research on complement and cytokine signaling toward personalized treatments for DRE patients.
Collapse
Affiliation(s)
- Nicole Pinzon-Hoyos
- Department of Biological Sciences, Southern Methodist University, Dallas, TX, USA
| | - Yibo Li
- Department of Biological Sciences, Southern Methodist University, Dallas, TX, USA
| | - Monnie McGee
- Department of Statistics and Data Science, Southern Methodist University, Dallas, TX, USA
| | - Nicholas P. Poolos
- Department of Neurology and Regional Epilepsy Center, University of Washington, Seattle, WA, USA
| | - Nicola Marchi
- Institut de Génomique Fonctionnelle, University of Montpellier, CNRS, INSERM; Cerebrovascular and Glia Research, Montpellier, France
| | - Amy L. Brewster
- Department of Biological Sciences, Southern Methodist University, Dallas, TX, USA
| |
Collapse
|
5
|
Salimans L, Liberman K, Cools W, Njemini R, Debacq-Chainiaux F, Forti LN, De Dobbeleer L, Kooijman R, Beyer I, Bautmans I. Inflammation-Related Genes Are Differentially Expressed in Lipopolysaccharide-Stimulated Peripheral Blood Mononuclear Cells after 3 Months of Resistance Training in Older Women. Cells 2024; 13:1416. [PMID: 39272988 PMCID: PMC11394400 DOI: 10.3390/cells13171416] [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: 06/30/2024] [Revised: 08/11/2024] [Accepted: 08/14/2024] [Indexed: 09/15/2024] Open
Abstract
Recently, we showed that three months of resistance exercise significantly alters 18 canonical pathways related to chronic inflammation in PBMCs of older adults. In this exploratory sub-study, the aim is to explore whether resistance exercise enhances the PBMCs stress response by mimicking an acute infection through in vitro LPS stimulation. Women (≥65 years) were randomly divided into intensive strength training (IST), strength endurance training (SET), or flexibility training (as control group, CON) groups. PBMCs were isolated and cultured with and without LPS for 24 h. Their RNA was analyzed via targeted RNA sequencing of 407 inflammation-related genes, with relevant fold-changes defined as ≤0.67 or ≥1.5 (3 months vs. baseline). A pathway analysis using ingenuity pathway analyses identified significant pathways among 407 genes with p < 0.05 and z-scores of ≤-2 or ≥2. Fourteen women were included in the analyses. A total of 151 genes with a significant fold-change were identified. In the CON group, a less-pronounced effect was observed. Strength training altered 23 pathways in the LPS-stimulated PBMCs, none of which overlapped between the IST and SET groups. A balanced exercise program that includes both IST and SET could beneficially adapt the immune responses in older adults by inducing alterations in the inflammatory stress response of PBMCs through different genes and pathways.
Collapse
Affiliation(s)
- Lene Salimans
- Frailty & Resilience in Ageing Research Unit (FRIA), Vitality Research Group, Vrije Universiteit Brussel (VUB), B-1090 Brussels, Belgium
- Gerontology Department, Vrije Universiteit Brussel (VUB), B-1090 Brussels, Belgium
| | - Keliane Liberman
- Frailty & Resilience in Ageing Research Unit (FRIA), Vitality Research Group, Vrije Universiteit Brussel (VUB), B-1090 Brussels, Belgium
| | - Wilfried Cools
- Biostatistics and Medical Informatics Department, Vrije Universiteit Brussel (VUB), B-1090 Brussels, Belgium
| | - Rose Njemini
- Frailty & Resilience in Ageing Research Unit (FRIA), Vitality Research Group, Vrije Universiteit Brussel (VUB), B-1090 Brussels, Belgium
- Gerontology Department, Vrije Universiteit Brussel (VUB), B-1090 Brussels, Belgium
| | - Florence Debacq-Chainiaux
- URBC, NAmur Research Institute for LIfe Science (NARILIS), University of Namur, B-5000 Namur, Belgium
| | - Louis Nuvagah Forti
- Frailty & Resilience in Ageing Research Unit (FRIA), Vitality Research Group, Vrije Universiteit Brussel (VUB), B-1090 Brussels, Belgium
| | - Liza De Dobbeleer
- Frailty & Resilience in Ageing Research Unit (FRIA), Vitality Research Group, Vrije Universiteit Brussel (VUB), B-1090 Brussels, Belgium
- Gerontology Department, Vrije Universiteit Brussel (VUB), B-1090 Brussels, Belgium
| | - Ron Kooijman
- Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), B-1090 Brussels, Belgium
| | - Ingo Beyer
- Frailty & Resilience in Ageing Research Unit (FRIA), Vitality Research Group, Vrije Universiteit Brussel (VUB), B-1090 Brussels, Belgium
- Geriatrics Department, Universitair Ziekenhuis Brussel (UZB), B-1090 Brussels, Belgium
| | - Ivan Bautmans
- Frailty & Resilience in Ageing Research Unit (FRIA), Vitality Research Group, Vrije Universiteit Brussel (VUB), B-1090 Brussels, Belgium
- Gerontology Department, Vrije Universiteit Brussel (VUB), B-1090 Brussels, Belgium
- Geriatrics Department, Universitair Ziekenhuis Brussel (UZB), B-1090 Brussels, Belgium
- Geriatric Physiotherapy Department, SOMT University of Physiotherapy, 3821 BN Amersfoort, The Netherlands
| |
Collapse
|
6
|
Popotas A, Casimir GJ, Corazza F, Lefèvre N. Sex-related immunity: could Toll-like receptors be the answer in acute inflammatory response? Front Immunol 2024; 15:1379754. [PMID: 38835761 PMCID: PMC11148260 DOI: 10.3389/fimmu.2024.1379754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 05/06/2024] [Indexed: 06/06/2024] Open
Abstract
An increasing number of studies have highlighted the existence of a sex-specific immune response, wherein men experience a worse prognosis in cases of acute inflammatory diseases. Initially, this sex-dependent inflammatory response was attributed to the influence of sex hormones. However, a growing body of evidence has shifted the focus toward the influence of chromosomes rather than sex hormones in shaping these inflammatory sex disparities. Notably, certain pattern recognition receptors, such as Toll-like receptors (TLRs), and their associated immune pathways have been implicated in driving the sex-specific immune response. These receptors are encoded by genes located on the X chromosome. TLRs are pivotal components of the innate immune system, playing crucial roles in responding to infectious diseases, including bacterial and viral pathogens, as well as trauma-related conditions. Importantly, the TLR-mediated inflammatory responses, as indicated by the production of specific proteins and cytokines, exhibit discernible sex-dependent patterns. In this review, we delve into the subject of sex bias in TLR activation and explore its clinical implications relatively to both the X chromosome and the hormonal environment. The overarching objective is to enhance our understanding of the fundamental mechanisms underlying these sex differences.
Collapse
Affiliation(s)
- Alexandros Popotas
- Laboratory of Pediatrics, Université Libre de Bruxelles, Brussels, Belgium
- Laboratory of Translational Research, Université Libre de Bruxelles, Brussels, Belgium
| | - Georges Jacques Casimir
- Laboratory of Pediatrics, Université Libre de Bruxelles, Brussels, Belgium
- Department of Pulmonology, Allergology and Cystic Fibrosis, Queen Fabiola Childrens University Hospital (Hôpital Universitaire des Enfants Reine Fabiola) – University Hospital of Brussels (Hôpital Universitaire de Bruxelles), Brussels, Belgium
| | - Francis Corazza
- Laboratory of Translational Research, Université Libre de Bruxelles, Brussels, Belgium
- Laboratory of Immunology, Centre Hospitalier Universitaire (CHU) Brugmann, Université Libre de Bruxelles, Brussels, Belgium
| | - Nicolas Lefèvre
- Laboratory of Pediatrics, Université Libre de Bruxelles, Brussels, Belgium
- Department of Pulmonology, Allergology and Cystic Fibrosis, Queen Fabiola Childrens University Hospital (Hôpital Universitaire des Enfants Reine Fabiola) – University Hospital of Brussels (Hôpital Universitaire de Bruxelles), Brussels, Belgium
| |
Collapse
|
7
|
Saarenpää M, Roslund MI, Nurminen N, Puhakka R, Kummola L, Laitinen OH, Hyöty H, Sinkkonen A. Urban indoor gardening enhances immune regulation and diversifies skin microbiota - A placebo-controlled double-blinded intervention study. ENVIRONMENT INTERNATIONAL 2024; 187:108705. [PMID: 38688234 DOI: 10.1016/j.envint.2024.108705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 03/26/2024] [Accepted: 04/25/2024] [Indexed: 05/02/2024]
Abstract
According to the hygiene and biodiversity hypotheses, frequent exposure to environmental microbiota, especially through soil contact, diversifies commensal microbiota, enhances immune modulation, and ultimately lowers the risk of immune-mediated diseases. Here we test the underlying assumption of the hygiene and biodiversity hypotheses by instructing volunteers to grow edible plants indoors during the winter season when natural exposure to environmental microbiota is low. The one-month randomized, placebo-controlled double-blind trial consisted of two treatments: participants received either microbially diverse growing medium or visually similar but microbially poor growing medium. Skin microbiota and a panel of seven immune markers were analyzed in the beginning of the trial and after one month. The diversity of five bacterial phyla (Bacteroidetes, Planctomycetes, Proteobacteria, Cyanobacteria, and Verrucomicrobia) and one class (Bacteroidia) increased on the skin of participants in the intervention group while no changes were observed in the placebo group. The number of nodes and edges in the co-occurrence networks of the skin bacteria increased on average three times more in the intervention group than in the placebo group. The plasma levels of the immunomodulatory cytokine interleukin 10 (IL-10) increased in the intervention group when compared with the placebo group. A similar trend was observed in the interleukin 17A (IL-17A) levels and in the IL-10:IL-17A ratios. Participants in both groups reported high satisfaction and adherence to the trial. The current study provides evidence in support of the core assumption of the hygiene and biodiversity hypotheses of immune-mediated diseases. Indoor urban gardening offers a meaningful and convenient approach for increasing year-round exposure to environmental microbiota, paving the way for other prophylactic practices that might help prevent immune-mediated diseases.
Collapse
Affiliation(s)
- Mika Saarenpää
- Ecosystems and Environment Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Niemenkatu 73, 15140 Lahti, Finland; Natural Resources Institute Finland, Turku and Helsinki, Finland.
| | - Marja I Roslund
- Natural Resources Institute Finland, Turku and Helsinki, Finland.
| | - Noora Nurminen
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön Katu 34, 33520 Tampere, Finland.
| | - Riikka Puhakka
- Ecosystems and Environment Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Niemenkatu 73, 15140 Lahti, Finland.
| | - Laura Kummola
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön Katu 34, 33520 Tampere, Finland.
| | - Olli H Laitinen
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön Katu 34, 33520 Tampere, Finland.
| | - Heikki Hyöty
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön Katu 34, 33520 Tampere, Finland.
| | - Aki Sinkkonen
- Natural Resources Institute Finland, Turku and Helsinki, Finland.
| |
Collapse
|
8
|
Lu C, Donners MMPC, de Baaij JBJ, Jin H, Otten JJT, Manca M, van Zonneveld AJ, Jukema JW, Kraaijeveld A, Kuiper J, Pasterkamp G, Mees B, Sluimer JC, Cavill R, Karel JMH, Goossens P, Biessen EAL. Identification of a gene network driving the attenuated response to lipopolysaccharide of monocytes from hypertensive coronary artery disease patients. Front Immunol 2024; 15:1286382. [PMID: 38410507 PMCID: PMC10894924 DOI: 10.3389/fimmu.2024.1286382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 01/24/2024] [Indexed: 02/28/2024] Open
Abstract
Introduction The impact of cardiovascular disease (CVD) risk factors, encompassing various biological determinants and unhealthy lifestyles, on the functional dynamics of circulating monocytes-a pivotal cell type in CVD pathophysiology remains elusive. In this study, we aimed to elucidate the influence of CVD risk factors on monocyte transcriptional responses to an infectious stimulus. Methods We conducted a comparative analysis of monocyte gene expression profiles from the CTMM - CIRCULATING CELLS Cohort of coronary artery disease (CAD) patients, at baseline and after lipopolysaccharide (LPS) stimulation. Gene co-expression analysis was used to identify gene modules and their correlations with CVD risk factors, while pivotal transcription factors controlling the hub genes in these modules were identified by regulatory network analyses. The identified gene module was subjected to a drug repurposing screen, utilizing the LINCS L1000 database. Results Monocyte responsiveness to LPS showed a highly significant, negative correlation with blood pressure levels (ρ< -0.4; P<10-80). We identified a ZNF12/ZBTB43-driven gene module closely linked to diastolic blood pressure, suggesting that monocyte responses to infectious stimuli, such as LPS, are attenuated in CAD patients with elevated diastolic blood pressure. This attenuation appears associated with a dampening of the LPS-induced suppression of oxidative phosphorylation. Finally, we identified the serine-threonine inhibitor MW-STK33-97 as a drug candidate capable of reversing this aberrant LPS response. Conclusions Monocyte responses to infectious stimuli may be hampered in CAD patients with high diastolic blood pressure and this attenuated inflammatory response may be reversed by the serine-threonine inhibitor MW-STK33-97. Whether the identified gene module is a mere indicator of, or causal factor in diastolic blood pressure and the associated dampened LPS responses remains to be determined.
Collapse
Affiliation(s)
- Chang Lu
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, Netherlands
- Institute for Computational Biomedicine, Faculty of Medicine, Heidelberg University and Heidelberg University Hospital, Heidelberg, Germany
| | - Marjo M P C Donners
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, Netherlands
| | - Julius B J de Baaij
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, Netherlands
| | - Han Jin
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, Netherlands
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - Jeroen J T Otten
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, Netherlands
| | | | - Anton Jan van Zonneveld
- Department of Internal Medicine (Nephrology), Leiden University Medical Center, Leiden, Netherlands
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, Netherlands
- Netherlands Heart Institute, Utrecht, Netherlands
| | - Adriaan Kraaijeveld
- Department of Cardiology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Johan Kuiper
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands
| | - Gerard Pasterkamp
- Circulatory Health Research Center, University Medical Center Utrecht, Utrecht, Netherlands
| | - Barend Mees
- Department of Vascular Surgery, Maastricht University Medical Center, Maastricht, Netherlands
| | - Judith C Sluimer
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, Netherlands
- Centre for Cardiovascular Science (CVS), University of Edinburgh, Edinburgh, United Kingdom
| | - Rachel Cavill
- Department of Advanced Computing Sciences, Maastricht University, Maastricht, Netherlands
| | - Joël M H Karel
- Department of Advanced Computing Sciences, Maastricht University, Maastricht, Netherlands
| | - Pieter Goossens
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, Netherlands
| | - Erik A L Biessen
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, Netherlands
- Institute for Molecular Cardiovascular Research, Klinikum RWTH Aachen, Aachen, Germany
| |
Collapse
|
9
|
Ligotti ME, Accardi G, Aiello A, Aprile S, Calabrò A, Caldarella R, Caruso C, Ciaccio M, Corsale AM, Dieli F, Di Simone M, Giammanco GM, Mascarella C, Akbar AN, Meraviglia S, Candore G. Sicilian semi- and supercentenarians: identification of age-related T-cell immunophenotype to define longevity trait. Clin Exp Immunol 2023; 214:61-78. [PMID: 37395602 PMCID: PMC10711357 DOI: 10.1093/cei/uxad074] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 05/12/2023] [Accepted: 06/30/2023] [Indexed: 07/04/2023] Open
Abstract
The immunophenotype of oldest centenarians, i.e. semi- and supercentenarians, could provide important information about their ability to adapt to factors associated with immune changes, including ageing per se and chronic Cytomegalovirus infection. We investigated, by flow cytometry, variations in percentages and absolute numbers of immune cell subsets, focusing on T cells, and pro-inflammatory parameters in a cohort of 28 women and 26 men (age range 19-110 years). We observed variability in hallmarks of immunosenescence related to age and Cytomegalovirus serological status. The eight oldest centenarians showed the lowest percentages of naïve T cells, due to their age, and the highest percentages of T-effector memory cells re-expressing CD45RA (TEMRA), according to their cytomegalovirus status, and high levels of serum pro-inflammatory parameters, although their means were lower than that of remaining 90+ donors. Some of them showed CD8 naïve and TEMRA percentages, and exhaustion/pro-inflammatory markers comparable to the younger ones. Our study supports the suggestion that immune ageing, especially of oldest centenarians, exhibits great variability that is not only attributable to a single contributor but should also be the full result of a combination of several factors. Everyone ages differently because he/she is unique in genetics and experience of life and this applies even more to the immune system; everybody has had a different immunological history. Furthermore, our findings on inflammatory markers, TEMRA and CMV seropositivity in centenarians, discussed in the light of the most recent literature, suggest that these changes might be not unfavourable for centenarians, and in particular for the oldest ones.
Collapse
Affiliation(s)
- Mattia Emanuela Ligotti
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Giulia Accardi
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Anna Aiello
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Stefano Aprile
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
- Unit of Transfusion Medicine, San Giovanni di Dio Hospital, Agrigento, Italy
| | - Anna Calabrò
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Rosalia Caldarella
- Department of Laboratory Medicine, University Hospital “P. Giaccone”, Palermo, Italy
| | - Calogero Caruso
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Marcello Ciaccio
- Department of Laboratory Medicine, University Hospital “P. Giaccone”, Palermo, Italy
- Section of Clinical Biochemistry, Clinical Molecular Medicine and Clinical Laboratory Medicine Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Anna Maria Corsale
- Central Laboratory of Advanced Diagnosis and Biomedical Research, University Hospital “P. Giaccone”, Palermo, Italy
- Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties “G. D’Alessandro”, University of Palermo, Palermo, Italy
| | - Francesco Dieli
- Central Laboratory of Advanced Diagnosis and Biomedical Research, University Hospital “P. Giaccone”, Palermo, Italy
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Marta Di Simone
- Central Laboratory of Advanced Diagnosis and Biomedical Research, University Hospital “P. Giaccone”, Palermo, Italy
- Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties “G. D’Alessandro”, University of Palermo, Palermo, Italy
| | - Giovanni Maurizio Giammanco
- Section of Microbiology, Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties “G. D’Alessandro”, University of Palermo, Palermo, Italy
| | - Chiara Mascarella
- Section of Microbiology, Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties “G. D’Alessandro”, University of Palermo, Palermo, Italy
| | - Arne N Akbar
- Division of Medicine, Experimental and Therapeutic Medicine, University College London, London, UK
| | - Serena Meraviglia
- Central Laboratory of Advanced Diagnosis and Biomedical Research, University Hospital “P. Giaccone”, Palermo, Italy
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Giuseppina Candore
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| |
Collapse
|
10
|
Zhang L, Altemus J, Ding L, Cherepanova O, Byzova TV, Podrez EA. Enhanced Akt3 kinase activity reduces atherosclerosis in hyperlipidemic mice in a gender-dependent manner. J Biol Chem 2023; 299:105425. [PMID: 37926285 PMCID: PMC10716582 DOI: 10.1016/j.jbc.2023.105425] [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: 02/13/2023] [Revised: 10/10/2023] [Accepted: 10/13/2023] [Indexed: 11/07/2023] Open
Abstract
Akt3 is one of the three members of the serine/threonine protein kinase B (AKT) family, which regulates multiple cellular processes. We have previously demonstrated that global knockout of Akt3 in mice promotes atherogenesis in a macrophage-dependent manner. Whether enhanced Akt3 kinase activity affects atherogenesis is not known. In this study, we crossed atherosclerosis-prone ApoE-/- mice with a mouse strain that has enhanced Akt3 kinase activity (Akt3nmf350) and assessed atherosclerotic lesion formation and the role of macrophages in atherogenesis. Significant reduction in atherosclerotic lesion area and macrophage accumulation in lesions were observed in ApoE-/-/Akt3nmf350 mice fed a Western-type diet. Experiments using chimeric ApoE-/- mice with either ApoE-/-/Akt3nmf350 bone marrow or ApoE-/- bone marrow cells showed that enhanced Akt3 activity specifically in bone marrow-derived cells is atheroprotective. The atheroprotective effect of Akt3nmf350 was more pronounced in male mice. In line with this result, the release of the pro-inflammatory cytokines IL-6, MCP1, TNF-α, and MIP-1α was reduced by macrophages from male but not female ApoE-/-/Akt3nmf350 mice. Levels of IL-6 and TNF-α were also reduced in atherosclerotic lesions of ApoE-/-/Akt3nmf350 male mice compared to ApoE-/- mice. Macrophages from male ApoE-/-/Akt3nmf350 mice were also more resistant to apoptosis in vitro and in vivo and tended to have more pronounced M2 polarization in vitro. These findings demonstrated that enhanced Akt3 kinase activity in macrophages protects mice from atherosclerosis in hyperlipidemic mice in a gender-dependent manner.
Collapse
Affiliation(s)
- Lifang Zhang
- Department of Inflammation & Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA; Department of Chemistry, Cleveland State University, Cleveland, Ohio, USA
| | - Jessica Altemus
- Department of Inflammation & Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Liang Ding
- Department of Inflammation & Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Olga Cherepanova
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Tatiana V Byzova
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Eugene A Podrez
- Department of Inflammation & Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA.
| |
Collapse
|
11
|
Sim J, O'Guin E, Monahan K, Sugimoto C, McLean SA, Albertorio-Sáez L, Zhao Y, Laumet S, Dagenais A, Bernard MP, Folger JK, Robison AJ, Linnstaedt SD, Laumet G. Interleukin-10-producing monocytes contribute to sex differences in pain resolution in mice and humans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.03.565129. [PMID: 37961295 PMCID: PMC10635095 DOI: 10.1101/2023.11.03.565129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Pain is closely associated with the immune system, which exhibits sexual dimorphism. For these reasons, neuro-immune interactions are suggested to drive sex differences in pain pathophysiology. However, our understanding of peripheral neuro-immune interactions on sex differences in pain resolution remains limited. Here, we have shown, in both a mouse model of inflammatory pain and in humans following traumatic pain, that males had higher levels of interleukin (IL)-10 than females, which were correlated with faster pain resolution. Following injury, we identified monocytes (CD11b+ Ly6C+ Ly6G-F4/80 mid ) as the primary source of IL-10, with IL-10-producing monocytes being more abundant in males than females. In a mouse model, neutralizing IL-10 signaling through antibodies, genetically ablating IL-10R1 in sensory neurons, or depleting monocytes with clodronate all impaired the resolution of pain hypersensitivity in both sexes. Furthermore, manipulating androgen levels in mice reversed the sexual dimorphism of pain resolution and the levels of IL-10-producing monocytes. These results highlight a novel role for androgen-driven peripheral IL-10-producing monocytes in the sexual dimorphism of pain resolution. These findings add to the growing concept that immune cells play a critical role in resolving pain and preventing the transition into chronic pain. Graphical abstract
Collapse
|
12
|
Lu C, Donners MMPC, Karel J, de Boer H, van Zonneveld AJ, den Ruijter H, Jukema JW, Kraaijeveld A, Kuiper J, Pasterkamp G, Cavill R, Perales-Patón J, Ferrannini E, Goossens P, Biessen EAL. Sex-specific differences in cytokine signaling pathways in circulating monocytes of cardiovascular disease patients. Atherosclerosis 2023; 384:117123. [PMID: 37127497 DOI: 10.1016/j.atherosclerosis.2023.04.005] [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: 11/10/2022] [Revised: 03/14/2023] [Accepted: 04/14/2023] [Indexed: 05/03/2023]
Abstract
BACKGROUND AND AIMS This study aims to identify sex-specific transcriptional differences and signaling pathways in circulating monocytes contributing to cardiovascular disease. METHODS AND RESULTS We generated sex-biased gene expression signatures by comparing male versus female monocytes of coronary artery disease (CAD) patients (n = 450) from the Center for Translational Molecular Medicine-Circulating Cells Cohort. Gene set enrichment analysis demonstrated that monocytes from female CAD patients carry stronger chemotaxis and migratory signature than those from males. We then inferred cytokine signaling activities based on CytoSig database of 51 cytokine and growth factor regulation profiles. Monocytes from females feature a higher activation level of EGF, IFN1, VEGF, GM-CSF, and CD40L pathways, whereas IL-4, INS, and HMGB1 signaling was seen to be more activated in males. These sex differences were not observed in healthy subjects, as shown for an independent monocyte cohort of healthy subjects (GSE56034, n = 485). More pronounced GM-CSF signaling in monocytes of female CAD patients was confirmed by the significant enrichment of GM-CSF-activated monocyte signature in females. As we show these effects were not due to increased plasma levels of the corresponding ligands, sex-intrinsic differences in monocyte signaling regulation are suggested. Consistently, regulatory network analysis revealed jun-B as a shared transcription factor activated in all female-specific pathways except IFN1 but suppressed in male-activated IL-4. CONCLUSIONS We observed overt CAD-specific sex differences in monocyte transcriptional profiles and cytokine- or growth factor-induced responses, which provide insights into underlying mechanisms of sex differences in CVD.
Collapse
Affiliation(s)
- Chang Lu
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht UMC+, Maastricht University, Maastricht, the Netherlands
| | - Marjo M P C Donners
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht UMC+, Maastricht University, Maastricht, the Netherlands.
| | - Joël Karel
- Department of Advanced Computing Sciences, Maastricht University, Maastricht, the Netherlands
| | - Hetty de Boer
- Department of Internal Medicine (Nephrology), Leiden UMC, Leiden, the Netherlands
| | | | - Hester den Ruijter
- Laboratory for Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, the Netherlands; Netherlands Heart Institute, Utrecht, the Netherlands
| | - Adriaan Kraaijeveld
- Department of Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Johan Kuiper
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands
| | | | - Rachel Cavill
- Department of Advanced Computing Sciences, Maastricht University, Maastricht, the Netherlands
| | - Javier Perales-Patón
- Institute for Computational Biomedicine, Faculty of Medicine, Heidelberg University and Heidelberg University Hospital, Heidelberg, Germany; Institute of Experimental Medicine and Systems Biology, RWTH Aachen University, Aachen, Germany; Joint Research Centre for Computational Biomedicine (JRC COMBINE), Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Ele Ferrannini
- Consiglio Nazionale Delle Ricerche (CNR) Institute of Clinical Physiology, Pisa, Italy
| | - Pieter Goossens
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht UMC+, Maastricht University, Maastricht, the Netherlands
| | - Erik A L Biessen
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht UMC+, Maastricht University, Maastricht, the Netherlands; Institute for Molecular Cardiovascular Research, RWTH Aachen University, Aachen, 52074, Germany
| |
Collapse
|
13
|
Li Y, Li L, Wu G, Xie G, Yi L, Zhu J, Liang S, Huang Y, Chen J, Ji S, Sun F, Liu R. The unique interplay of mitochondrial oxidative phosphorylation (OXPHOS) and immunity and its potential implication for the sex- and age-related morbidity of severe COVID-19 patients. MedComm (Beijing) 2023; 4:e371. [PMID: 37750090 PMCID: PMC10518039 DOI: 10.1002/mco2.371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 08/11/2023] [Accepted: 08/19/2023] [Indexed: 09/27/2023] Open
Abstract
Aged male patients are more vulnerable to severe or critical symptoms of COVID-19, but the underlying mechanism remains elusive. In this study, we analyzed previously published scRNA-seq data from a large cohort of COVID-19 patients, castrated and regenerated mice, and bulk RNA-seq of a RNAi library of 400 genes, and revealed that both immunity and OXPHOS displayed cell-type-, sex-, and age-related variation in the severe or critical COVID-19 patients during disease progression, with a more prominent increase in immunity and decrease in OXPHOS in myeloid cells in the males relative to the females (60-69 years old). Male severe or critical patients above 70 years old were an exception in that the compromised negative correlation between OXPHOS and immunity in these patients was associated with its disordered transcriptional regulation. Finally, the expression levels of OXPHOS and androgens were revealed to be positively correlated, and the responses of macrophages to android fluctuation were more striking than other types of detected immune cells in the castrated mice model. Therefore, the interplay of OXPHOS and immunity displayed a cell-type-specific, age-related, and sex-biased pattern, and the underlying potential regulatory role of the hormonal milieu should not be neglected.
Collapse
Affiliation(s)
- Yinchuan Li
- Institute of Reproductive MedicineMedical School of Nantong UniversityNantongJiangsuP. R. China
| | - Lei Li
- National Clinical Research Center for Obstetric & Gynecologic DiseasesDepartment of Obstetrics and GynecologyPeking Union Medical College HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingP. R. China
| | - Guanghao Wu
- School of Materials Science and EngineeringBeijing Institute of TechnologyBeijingP. R. China
| | - Gangcai Xie
- Institute of Reproductive MedicineMedical School of Nantong UniversityNantongJiangsuP. R. China
| | - Lirong Yi
- Institute of Reproductive MedicineMedical School of Nantong UniversityNantongJiangsuP. R. China
| | - Jie Zhu
- National Key Laboratory of Biochemical EngineeringInstitute of Process EngineeringChinese Academy of SciencesBeijingP. R. China
- University of Chinese Academy of SciencesBeijingP. R. China
| | - ShiYu Liang
- National Key Laboratory of Biochemical EngineeringInstitute of Process EngineeringChinese Academy of SciencesBeijingP. R. China
| | - Ya‐ru Huang
- National Key Laboratory of Biochemical EngineeringInstitute of Process EngineeringChinese Academy of SciencesBeijingP. R. China
| | - Juan Chen
- National Clinical Research Center for Obstetric & Gynecologic DiseasesDepartment of Obstetrics and GynecologyPeking Union Medical College HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingP. R. China
| | - Shaoyang Ji
- National Key Laboratory of Biochemical EngineeringInstitute of Process EngineeringChinese Academy of SciencesBeijingP. R. China
| | - Fei Sun
- Institute of Reproductive MedicineMedical School of Nantong UniversityNantongJiangsuP. R. China
| | - Rui‐tian Liu
- National Key Laboratory of Biochemical EngineeringInstitute of Process EngineeringChinese Academy of SciencesBeijingP. R. China
| |
Collapse
|
14
|
Prout AJ, Banks RK, Reeder RW, Zimmerman JJ, Meert KL. Association of Sex and Age with Mortality and Health-Related Quality of Life in Children with Septic Shock: A Secondary Analysis of the Life After Pediatric Sepsis Evaluation. J Intensive Care Med 2023; 39:8850666231190270. [PMID: 37529851 DOI: 10.1177/08850666231190270] [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] [Indexed: 08/03/2023]
Abstract
Introduction: Sepsis is more common in males than females, but whether outcomes differ by sex in various pediatric age groups is unclear. The Life After Pediatric Sepsis Evaluation (LAPSE) was a multicenter prospective cohort study that evaluated health-related quality of life (HRQL) in children after community-acquired septic shock. In this secondary analysis, we evaluated whether male children are at increased risk of mortality or long-term decline in HRQL than female children by age group. Methods: Children (1 month-18 years) with community-acquired septic shock were recruited from 12 pediatric intensive care units in the U.S. Data included sex, age group (<1 year, 1-<13 years, 13-18 years), acute illness severity (acute organ dysfunction and inflammation), and longitudinal assessments of HRQL and mortality. Persistent decline in HRQL was defined as a 10% decrease in HRQL comparing baseline to 3 months following admission. Male and female children were stratified by age group and compared to evaluate the difference in the composite outcome of death or persistent decline in HRQL using the Cochran-Mantel-Haenszel test. Results: Of 389 children, 54.2% (n = 211) were male. Overall, 10% (21/211) of males and 12% (22/178) of females died by 3 months (p = 0.454). Among children with follow-up data, 41% (57/138) of males and 44% (48/108) of females died or had persistent decline in HRQL at 3 months (p = 0.636), with no observed difference by sex when stratified by age group. There was no significant difference in acute illness severity between males and females overall or stratified by age group. Conclusions: In this secondary analysis of the LAPSE cohort, HRQL, and mortality were not different between male and female children when stratified by age group. There were no significant differences by sex across multiple measures of illness severity or treatment intensity.
Collapse
Affiliation(s)
- Andrew J Prout
- Section of Pediatric Critical Care, Department of Pediatrics, Children's Hospital of Michigan, Detroit, MI, USA
- Department of Pediatrics, Central Michigan University School of Medicine, Mt. Pleasant, MI, USA
| | - Russell K Banks
- Department of Pediatrics, University of Utah, Salt Lake City, UT, USA
| | - Ron W Reeder
- Department of Pediatrics, University of Utah, Salt Lake City, UT, USA
| | - Jerry J Zimmerman
- Section of Pediatric Critical Care, Department of Pediatrics, Seattle Children's Hospital, Seattle Children's Research Institute, University of Washington School of Medicine, Seattle, WA, USA
| | - Kathleen L Meert
- Section of Pediatric Critical Care, Department of Pediatrics, Children's Hospital of Michigan, Detroit, MI, USA
- Department of Pediatrics, Central Michigan University School of Medicine, Mt. Pleasant, MI, USA
| |
Collapse
|
15
|
Wang S, Singh M, Yang H, Morrell CN, Mohamad LA, Xu JJ, Nguyen T, Ture S, Tyrell A, Maggirwar SB, Schifitto G, Pang J. Monocyte-derived Dll4 is a novel contributor to persistent systemic inflammation in HIV patients. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.18.537330. [PMID: 37131726 PMCID: PMC10153122 DOI: 10.1101/2023.04.18.537330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Background In people living with HIV (PLWH) on combination antiretroviral therapy (cART), persistent systemic inflammation is a driving force for the progression of comorbidities, such as cardiovascular and cerebrovascular diseases. In this context, monocyte- and macrophage-related inflammation rather than T cell activation is a major cause of chronic inflammation. However, the underlying mechanism of how monocytes cause persistent systemic inflammation in PLWH is elusive. Methods and Results In vitro, we demonstrated that lipopolysaccharides (LPS) or tumor necrosis factor alpha (TNFα), induced a robust increase of Delta-like ligand 4 (Dll4) mRNA and protein expression in human monocytes and Dll4 secretion (extracellular Dll4, exDll4) from monocytes. Enhanced membrane-bound Dll4 (mDll4) expression in monocytes triggered Notch1 activation to promote pro-inflammatory factors expression. Dll4 silencing and inhibition of Nocth1 activation diminished the LPS or TNFα -induced inflammation. exDll4 releases in response to cytokines occurred in monocytes but not endothelial cells or T cells. In clinical specimens, we found that PLWH, both male and female, on cART, showed a significant increase in mDll4 expression, activation of Dll4-Notch1 signaling, and inflammatory markers in monocytes. Although there was no sex effect on mDII4 in PLWH, plasma exDll4 was significantly elevated in males but not females compared to HIV uninfected individuals. Furthermore, exDll4 plasma levels paralleled with monocytes mDll4 in male PLWH. Circulating exDll4 was also positively associated with pro-inflammatory monocytes phenotype and negatively associated with classic monocytes phenotype in male PLWH. Conclusion Pro-inflammatory stimuli increase Dll4 expression and Dll4-Notch1 signaling activation in monocytes and enhance monocyte proinflammatory phenotype, contributing to persistent systemic inflammation in male and female PLWH. Therefore, monocyte mDll4 could be a potential biomarker and therapeutic target of systemic inflammation. Plasma exDll4 may also play an additional role in systemic inflammation but primarily in men.
Collapse
|
16
|
Yerly A, van der Vorst EPC, Baumgartner I, Bernhard SM, Schindewolf M, Döring Y. Sex-specific and hormone-related differences in vascular remodelling in atherosclerosis. Eur J Clin Invest 2023; 53:e13885. [PMID: 36219492 DOI: 10.1111/eci.13885] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 10/04/2022] [Indexed: 12/24/2022]
Abstract
Atherosclerosis, a lipid-driven inflammatory disease, is the main underlying cause of cardiovascular diseases (CVDs) both in men and women. Sex-related dimorphisms regarding CVDs and atherosclerosis were observed since more than a decade ago. Inflammatory mediators such as cytokines, but also endothelial dysfunction, vascular smooth muscle cell migration and proliferation lead to vascular remodelling but are differentially affected by sex. Each year a greater number of men die of CVDs compared with women and are also affected by CVDs at an earlier age (40-70 years old) while women develop atherosclerosis-related complications mainly after menopause (60+ years). The exact biological reasons behind this discrepancy are still not well-understood. From the numerous animal studies on atherosclerosis, only a few include both sexes and even less investigate and highlight the sex-specific differences that may arise. Endogenous sex hormones such as testosterone and oestrogen modulate the atherosclerotic plaque composition and the frequency of such plaques. In men, testosterone seems to act like a double-edged sword as its decrease with ageing correlates with an increased risk of atherosclerotic CVDs, while testosterone is also reported to promote inflammatory immune cell recruitment into the atherosclerotic plaque. In premenopausal women, oestrogen exerts anti-atherosclerotic effects, which decline together with its level after menopause resulting in increased CVD risk in ageing women. However, the interplay of sex hormones, sex-specific immune responses and other sex-related factors is still incompletely understood. This review highlights reported sex differences in atherosclerotic vascular remodelling and the role of endogenous sex hormones in this process.
Collapse
Affiliation(s)
- Anaïs Yerly
- Division of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.,Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland.,Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Emiel P C van der Vorst
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich (LMU), Munich, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany.,Institute for Molecular Cardiovascular Research (IMCAR) and Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, Aachen, Germany
| | - Iris Baumgartner
- Division of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.,Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Sarah Maike Bernhard
- Division of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.,Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Marc Schindewolf
- Division of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.,Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Yvonne Döring
- Division of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.,Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland.,Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich (LMU), Munich, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| |
Collapse
|
17
|
Abstract
There is a growing awareness of the importance of sex and gender in medicine and research. Women typically have stronger immune responses to self and foreign antigens than men, resulting in sex-based differences in autoimmunity and infectious diseases. In both animals and humans, males are generally more susceptible than females to bacterial infections. At the same time, gender differences in health-seeking behavior, quality of health care, and adherence to treatment recommendations have been reported. This review explores our current understanding of differences between males and females in bacterial diseases. We describe how genetic, immunological, hormonal, and anatomical factors interact to influence sex-based differences in pathophysiology, epidemiology, clinical presentation, disease severity, and prognosis, and how gender roles affect the behavior of patients and providers in the health care system.
Collapse
|
18
|
Coales I, Tsartsalis S, Fancy N, Weinert M, Clode D, Owen D, Matthews PM. Alzheimer's disease-related transcriptional sex differences in myeloid cells. J Neuroinflammation 2022; 19:247. [PMID: 36199077 PMCID: PMC9535846 DOI: 10.1186/s12974-022-02604-w] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Accepted: 09/21/2022] [Indexed: 11/30/2022] Open
Abstract
Sex differences have been identified in many diseases associated with dysregulated immune responses, including Alzheimer's disease (AD), for which approximately two-thirds of patients are women. An accumulating body of research indicates that microglia may play a causal role in the pathogenesis of this disease. We hypothesised that sex differences in the transcriptome of human myeloid cells may contribute to the sex difference observed in AD prevalence. To explore this, we assessed bulk and single-nuclear RNA sequencing data sets generated from four human derived myeloid cell populations: post-mortem microglial nuclei, peripheral monocytes, monocyte-derived macrophages (MDMs) and induced pluripotent stem cell derived microglial-like cells (MGLs). We found that expression of AD risk genes, gene signatures associated with the inflammatory response in AD, and genes related to proinflammatory immune responses were enriched in microglial nuclei isolated from aged female donors without ante-mortem neurological disease, relative to those from males. In addition, these inflammation-associated gene sets were found to be enriched in peripheral monocytes isolated from postmenopausal women and in MDMs obtained from premenopausal individuals relative to age-matched males. Expression of these gene sets did not differ in MDMs derived from women whose blood was sampled across the menstrual cycle or in MGLs cultured with 17β-oestradiol. This suggests that the observed gene set enrichments in myeloid cells from women were not being driven by acute hormonal influences. Together, these data support the hypothesis that the increased prevalence of AD in women may be partly explained by a myeloid cell phenotype biased towards expression of biological processes relevant to AD.
Collapse
Affiliation(s)
- Isabelle Coales
- Department of Brain Sciences, Imperial College London, London, UK
- Centre for Host Microbiome Interactions, King's College London, London, SE1 9RT, UK
| | - Stergios Tsartsalis
- Department of Brain Sciences, Imperial College London, London, UK
- Department of Psychiatry, University of Geneva, Geneva, Switzerland
| | - Nurun Fancy
- Department of Brain Sciences, Imperial College London, London, UK
- UK Dementia Research Centre at Imperial College London, London, UK
| | - Maria Weinert
- Department of Brain Sciences, Imperial College London, London, UK
| | - Daniel Clode
- UK Dementia Research Centre at Imperial College London, London, UK
| | - David Owen
- Department of Brain Sciences, Imperial College London, London, UK.
- Clinical Research Facility, Hammersmith Hospital, ICTM Building, DuCane Road, London, W12 0NN, UK.
| | - Paul M Matthews
- Department of Brain Sciences, Imperial College London, London, UK.
- UK Dementia Research Centre at Imperial College London, London, UK.
- Hammersmith Hospital, E502, Burlington Danes Building, DuCane Road, London, W12 0NN, UK.
| |
Collapse
|
19
|
Flood TR, Kuennen MR, Blacker SD, Myers SD, Walker EF, Lee BJ. The effect of sex, menstrual cycle phase and oral contraceptive use on intestinal permeability and ex-vivo monocyte TNFα release following treatment with lipopolysaccharide and hyperthermia. Cytokine 2022; 158:155991. [PMID: 35944412 DOI: 10.1016/j.cyto.2022.155991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 07/22/2022] [Accepted: 08/01/2022] [Indexed: 11/15/2022]
Abstract
PURPOSE Investigate the impact of sex, menstrual cycle phase and oral contraceptive use on intestinal permeability and ex-vivo tumour necrosis factor alpha (TNFα) release following treatment with lipopolysaccharide (LPS) and hyperthermia. METHODS Twenty-seven participants (9 men, 9 eumenorrheic women (MC) and 9 women taking an oral contraceptive pill (OC)) completed three trials. Men were tested on 3 occasions over 6 weeks; MC during early-follicular, ovulation, and mid-luteal phases; OC during the pill and pill-free phase. Intestinal permeability was assessed following a 4-hour dual sugar absorption test (lactulose: rhamnose). Venous blood was collected each trial and stimulated with 100 μg·mL-1 LPS before incubation at 37 °C and 40 °C and analysed for TNFα via ELISA. RESULTS L:R ratio was higher in OC than MC (+0.003, p = 0.061) and men (+0.005, p = 0.007). Men had higher TNFα responses than both MC (+53 %, p = 0.004) and OC (+61 %, p = 0.003). TNFα release was greater at 40 °C than 37 °C (+23 %, p < 0.001). CONCLUSIONS Men present with lower resting intestinal barrier permeability relative to women regardless of OC use and displayed greater monocyte TNFα release following whole blood treatment with LPS and hyperthermia. Oral contraceptive users had highest intestinal permeability however, neither permeability or TNFα release were impacted by the pill cycle. Although no statistical effect was seen in the menstrual cycle, intestinal permeability and TNFα release were more variable across the phases.
Collapse
Affiliation(s)
- Tessa R Flood
- Occupational Performance Research Group, University of Chichester UK
| | - Matthew R Kuennen
- Department of Exercise Science, High Point University, High Point, NC, USA
| | - Sam D Blacker
- Occupational Performance Research Group, University of Chichester UK
| | - Stephen D Myers
- Occupational Performance Research Group, University of Chichester UK
| | | | - Ben J Lee
- Occupational and Environmental Physiology Group, Coventry University, UK.
| |
Collapse
|
20
|
Vajavaara H, Leivonen S, Jørgensen J, Holte H, Leppä S. Low lymphocyte-to-monocyte ratio predicts poor outcome in high-risk aggressive large B-cell lymphoma. EJHAEM 2022; 3:681-687. [PMID: 36051040 PMCID: PMC9421995 DOI: 10.1002/jha2.409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 02/17/2022] [Accepted: 02/21/2022] [Indexed: 11/12/2022]
Abstract
Low lymphocyte-to-monocyte-ratio (LMR) has been associated with unfavorable survival in patients with diffuse large B-cell lymphoma (DLBCL). To date, however, the impact of LMR on survival has not been examined in a uniformly treated cohort of patients with high-risk aggressive large B-cell lymphoma. We collected peripheral blood absolute lymphocyte counts (ALCs) and absolute monocyte counts (AMC) prior to treatment and calculated LMR from 112 adult patients, who were less than 65 years of age, had age-adjusted International Prognostic Index 2-3, or site-specific risk factors for central nervous system (CNS) recurrence, and were treated in a Nordic Lymphoma Group LBC-05 trial with dose-dense immunochemotherapy and early systemic CNS prophylaxis (www.ClinicalTrials.gov, number NCT01325194). Median pretreatment ALC was 1.40 × 109/l (range, 0.20-4.95), AMC 0.68 × 109/l (range, 0.10-2.62), and LMR 2.08 (range, 0.10-12.00). ALC did not correlate with tumor-infiltrating lymphocytes, AMC did not correlate with tumor-associated macrophages, and neither ALC nor AMC correlated with survival. However, low LMR (<1.72) translated to unfavourable progression-free survival (PFS) (5-year PFS 70% vs. 92%, p = 0.002) and overall survival (OS) (5-year OS, 77% vs. 92%, p = 0.020). In the patients with low LMR, relative risk of progression was 4.4-fold (95% confidence interval [CI] 1.60-12.14, p = 0.004), and relative risk of death was 3.3-fold (95% CI 1.18-9.50, p = 0.024) in comparison to the patients with high LMR. We conclude that low LMR is an adverse prognostic factor in uniformly treated young patients with high-risk aggressive large B-cell lymphoma.
Collapse
Affiliation(s)
- Heli Vajavaara
- Research Program UnitApplied Tumor GenomicsFaculty of MedicineUniversity of HelsinkiHelsinkiFinland
- Department of OncologyHelsinki University Hospital Comprehensive Cancer CenterHelsinkiFinland
- iCAN Digital Precision Cancer Medicine FlagshipHelsinkiFinland
| | - Suvi‐Katri Leivonen
- Research Program UnitApplied Tumor GenomicsFaculty of MedicineUniversity of HelsinkiHelsinkiFinland
- Department of OncologyHelsinki University Hospital Comprehensive Cancer CenterHelsinkiFinland
- iCAN Digital Precision Cancer Medicine FlagshipHelsinkiFinland
| | - Judit Jørgensen
- Department of HematologyAarhus University HospitalAarhusDenmark
| | - Harald Holte
- Department of OncologyKG Jebsen Center for B‐Cell MalignanciesOslo University HospitalOsloNorway
| | - Sirpa Leppä
- Research Program UnitApplied Tumor GenomicsFaculty of MedicineUniversity of HelsinkiHelsinkiFinland
- Department of OncologyHelsinki University Hospital Comprehensive Cancer CenterHelsinkiFinland
- iCAN Digital Precision Cancer Medicine FlagshipHelsinkiFinland
| |
Collapse
|
21
|
Islam H, Jackson GS, Yoon JSJ, Cabral-Santos C, Lira FS, Mui AL, Little JP. Sex differences in IL-10's anti-inflammatory function: Greater STAT3 activation and stronger inhibition of TNF-α production in male blood leukocytes ex vivo. Am J Physiol Cell Physiol 2022; 322:C1095-C1104. [PMID: 35508192 DOI: 10.1152/ajpcell.00091.2022] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Interleukin-10 (IL-10) inhibits pro-inflammatory cytokine production in blood leukocytes - an effect mediated by signal transducer and activator of transcription 3 (STAT3) activation. To examine potential sex-based differences in IL-10's anti-inflammatory function, we treated whole blood from healthy males and females (n=16 each; age: 28±6 years; body mass index: 23.5±2.3 kg/m2) with increasing concentrations of IL-10 (1-100 ng/mL) and quantified changes in phosphorylated STAT3 (pSTAT3) in CD14+ monocytes and CD4+ lymphocytes via flow cytometry. In parallel, liposaccharide (LPS)-stimulated whole-blood cultures were used to assess sex-based differences in IL-10's ability to inhibit tumour necrosis factor (TNF)-α production. IL-10 concentration-dependently increased pSTAT3 mean fluorescent intensity (MFI) in CD14+ and CD4+ cells (main effects of concentration, P<0.01) with males exhibiting larger changes in pSTAT3 MFI in both cell types (main effects of sex, P<0.01). Accordingly, IL-10-mediated inhibition of TNF-α production was more pronounced in males (main effect of sex, P<0.01) with changes in other monocyte-derived cytokines (IL-1b, IL-1RA, IL-15) also supporting a sexual dimorphism in IL-10 action (P<0.05). These sex-based differences were not explained by differences in circulating plasma IL-10 concentrations, basal IL-10 receptor expression in unstimulated CD14+ and CD4+ cells, nor the basal expression of IL-10 signaling proteins (STAT3, SHIP1, p38 MAPK) in unstimulated peripheral blood mononuclear cells. We conclude that IL-10's anti-inflammatory function differs between male and female blood leukocytes ex vivo. This sexual dimorphism should be considered in future work investigating IL-10's anti-inflammatory action in humans as it may represent a mechanism contributing to sex differences in overall immune function.
Collapse
Affiliation(s)
- Hashim Islam
- School of Health and Exercise Sciences, University of British Columbia Okanagan, Kelowna, BC, Canada
| | - Garett S Jackson
- School of Health and Exercise Sciences, University of British Columbia Okanagan, Kelowna, BC, Canada
| | - Jeff S J Yoon
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada.,Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | - Carolina Cabral-Santos
- Exercise and Immunometabolism Research Group, Department of Physical Education, Post-Graduation Program in Movement Sciences, State University of São Paulo (UNESP), Presidente Prudente, São Paulo, Brazil
| | - Fabio S Lira
- Exercise and Immunometabolism Research Group, Department of Physical Education, Post-Graduation Program in Movement Sciences, State University of São Paulo (UNESP), Presidente Prudente, São Paulo, Brazil
| | - Alice L Mui
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada.,Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | - Jonathan P Little
- School of Health and Exercise Sciences, University of British Columbia Okanagan, Kelowna, BC, Canada
| |
Collapse
|
22
|
Collier F, Chau C, Mansell T, Faye-Chauhan K, Vuillermin P, Ponsonby AL, Saffery R, Tang MLK, O'Hely M, Carlin J, Gray LEK, Bekkering S, Burgner D. Innate Immune Activation and Circulating Inflammatory Markers in Preschool Children. Front Immunol 2022; 12:830049. [PMID: 35211111 PMCID: PMC8860896 DOI: 10.3389/fimmu.2021.830049] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 12/28/2021] [Indexed: 12/17/2022] Open
Abstract
Early childhood is characterised by repeated infectious exposures that result in inflammatory responses by the innate immune system. In addition, this inflammatory response to infection is thought to contribute to the epidemiological evidence linking childhood infection and adult non-communicable diseases. Consequently, the relationship between innate immune responses and inflammation during early life may inform prevention of NCDs later in life. In adults, non-genetic host factors such as age, sex, and obesity, strongly impact cytokine production and circulating mediators, but data in children are lacking. Here, we assessed cytokine responses and inflammatory markers in a population of healthy preschool children (mean age 4.2 years). We studied associations between cytokines, plasma inflammatory markers and non-genetic host factors, such as sex, age, adiposity, season, and immune cell composition. Similar to adults, boys had a higher inflammatory response than girls, with IL-12p70 and IL-10 upregulated following TLR stimulation. Adiposity and winter season were associated with increased circulating inflammatory markers but not cytokine production. The inflammatory markers GlycA and hsCRP were positively associated with production of a number of cytokines and may therefore reflect innate immune function and inflammatory potential. This dataset will be informative for future prospective studies relating immune parameters to preclinical childhood NCD phenotypes.
Collapse
Affiliation(s)
- Fiona Collier
- School of Medicine, Deakin University, Geelong, VIC, Australia.,Child Health Research Unit, Barwon Health, Geelong, VIC, Australia
| | - Cerys Chau
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, VIC, Australia
| | - Toby Mansell
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, VIC, Australia
| | | | - Peter Vuillermin
- School of Medicine, Deakin University, Geelong, VIC, Australia.,Child Health Research Unit, Barwon Health, Geelong, VIC, Australia.,Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, VIC, Australia
| | - Anne-Louise Ponsonby
- School of Medicine, Deakin University, Geelong, VIC, Australia.,Department of Neuroepidemiology, The Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
| | - Richard Saffery
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, VIC, Australia.,Department of Neuroepidemiology, The Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
| | - Mimi L K Tang
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, VIC, Australia.,Department of Paediatrics, Melbourne University, Parkville, VIC, Australia
| | - Martin O'Hely
- School of Medicine, Deakin University, Geelong, VIC, Australia.,Child Health Research Unit, Barwon Health, Geelong, VIC, Australia.,Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, VIC, Australia
| | - John Carlin
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, VIC, Australia.,Department of Paediatrics, Melbourne University, Parkville, VIC, Australia
| | | | - Siroon Bekkering
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, VIC, Australia.,Department of Internal Medicine and Radboud Institute for Molecular Life Science (RIMLS), Radboud University Medical Center, Nijmegen, Netherlands
| | - David Burgner
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, VIC, Australia.,Department of Paediatrics, Melbourne University, Parkville, VIC, Australia.,Department of Paediatrics, Monash University, Clayton, VIC, Australia
| | | |
Collapse
|
23
|
Rodas L, Martínez S, Riera-Sampol A, Moir HJ, Tauler P. Blood Cell In Vitro Cytokine Production in Response to Lipopolysaccharide Stimulation in a Healthy Population: Effects of Age, Sex, and Smoking. Cells 2021; 11:cells11010103. [PMID: 35011664 PMCID: PMC8750398 DOI: 10.3390/cells11010103] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 12/27/2021] [Accepted: 12/27/2021] [Indexed: 01/03/2023] Open
Abstract
Immune system functionality has been commonly assessed by a whole-blood or isolated-cell stimulation assay. The aim of this study was to determine whether cytokine production in whole-blood-stimulated samples is influenced by age, sex, and smoking. A descriptive cross-sectional study in 253 healthy participants aged 18-55 years was conducted. Whole blood samples were stimulated for 24 h with LPS and concentrations of IL-6, IL-10, and TNF-α were determined in the culture media. Among parameters considered, statistical regression analysis indicated that smoking (change in R2 = 0.064, p < 0.001) and sex (change in R2 = 0.070, p < 0.001) were the main predictors for IL-10 production, with higher values for women and non-smokers. Age was also found to be a significant predictor (change in R2 = 0.021, p < 0.001), with higher values for younger ages. Age (change in R2 = 0.089, p = 0.013) and smoking (change in R2 = 0.037, p = 0.002) were found to be negative predictors for IL-6 production. Regarding TNF-α-stimulated production, age (change in R2 = 0.029, p = 0.009) and smoking (change in R2 = 0.022, p = 0.022) were found to be negative predictors. Furthermore, sex (change in R2 = 0.016, p = 0.045) was found to be a significant predictor, with lower values for women. In conclusion, sex, age, and smoking were found to be independent determinants of stimulated cytokine production. While female sex is associated with higher IL-10 and lower TNF-α production, aging and smoking are associated with lower IL-6, IL-10, and TNF-α production.
Collapse
Affiliation(s)
- Lluis Rodas
- Research Group on Evidence, Lifestyles and Health, Department of Fundamental Biology and Health Sciences, Research Institute of Health Sciences (IUNICS), University of the Balearic Islands, 07122 Palma, Spain;
| | - Sonia Martínez
- Research Group on Evidence, Lifestyles and Health, Department of Nursing and Physiotherapy, Research Institute of Health Sciences (IUNICS), University of the Balearic Islands, 07122 Palma, Spain;
- Health Research Institute of the Balearic Islands (IdISBa), 07120 Palma, Spain
- Correspondence: (S.M.); (P.T.); Tel.: +34-971-172858 (P.T.)
| | - Aina Riera-Sampol
- Research Group on Evidence, Lifestyles and Health, Department of Nursing and Physiotherapy, Research Institute of Health Sciences (IUNICS), University of the Balearic Islands, 07122 Palma, Spain;
- Health Research Institute of the Balearic Islands (IdISBa), 07120 Palma, Spain
| | - Hannah J. Moir
- School of Life Sciences, Pharmacy and Chemistry, Faculty of Science Engineering and Computing, Kingston University London, Penrhyn Road, Kingston upon Thames KT1 2EE, UK;
| | - Pedro Tauler
- Research Group on Evidence, Lifestyles and Health, Department of Fundamental Biology and Health Sciences, Research Institute of Health Sciences (IUNICS), University of the Balearic Islands, 07122 Palma, Spain;
- Health Research Institute of the Balearic Islands (IdISBa), 07120 Palma, Spain
- Correspondence: (S.M.); (P.T.); Tel.: +34-971-172858 (P.T.)
| |
Collapse
|
24
|
Nguyen KT, Gates CA, Hassell JE, Foxx CL, Salazar SN, Luthens AK, Arnold AL, Elam BL, Elsayed AI, Leblanc M, Adams SC, Lowry CA, Reuter JD. Evaluation of the effects of altitude on biological signatures of inflammation and anxiety- and depressive-like behavioral responses. Prog Neuropsychopharmacol Biol Psychiatry 2021; 111:110331. [PMID: 33891978 DOI: 10.1016/j.pnpbp.2021.110331] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 04/15/2021] [Accepted: 04/15/2021] [Indexed: 12/11/2022]
Abstract
Over sixteen million people suffer from a depressive episode annually in the United States, with females affected at twice the rate of males. Little is known about the effects of exposure to high altitude on the risk of development of major depressive disorder, despite reports of higher suicide rates at higher altitudes. We hypothesize that exposure to hypobaric hypoxia at high altitude increases endophenotypes of self-directed suicidal violence, including biological signatures of chronic inflammation and vulnerability to anxiety-like and depressive-like behavioral responses in a sex-specific manner. Biological signatures of inflammation, including granulocyte:lymphocyte ratios, monocyte cell counts, and monocyte:lymphocyte ratios were assessed using complete blood count data, anhedonia, and anxiety- and depressive-like behavioral responses were evaluated. We assessed biological signatures of inflammation and behavioral responses in the open-field test, sucrose preference test, and modified Porsolt forced swim test in young adult male and female Long-Evans and Sprague Dawley rats. All tests were conducted near sea level (374 ft [114 m] elevation) and at moderate-high altitude (5430 ft [1655 m] elevation) during acclimation periods of one, two, three, four, and five weeks following shipment from a sea level animal breeding facility (N = 320, n = 8 per group). Exposure to moderate-high altitude induced a biological signature of increased inflammation, as evidenced by main effects of altitude for: 1) increased granulocyte:lymphocyte ratio; 2) increased count and relative abundance of circulating monocytes; and 3) increased monocyte:lymphocyte ratios. Exposure to moderate-high altitude also increased anhedonia as assessed in the sucrose preference test in both male and female rats, when data were collapsed across strain and time. Among male and female Long Evans rats, exposure to moderate-high altitude increased immobility in the forced swim test, without changing anxiety-like behaviors in the open-field test. Finally, granulocyte:lymphocyte ratios were correlated with anhedonia in the sucrose preference test. These data are consistent with the hypothesis that hypobaric hypoxia at moderate-high altitude induces persistent endophenotypes of self-directed suicidal violence including biological signatures of inflammation, anhedonia, and depressive-like behavioral responses.
Collapse
Affiliation(s)
- Kadi T Nguyen
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA.
| | - Chloé A Gates
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA.
| | - James E Hassell
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA; Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA.
| | - Christine L Foxx
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA; Center for Microbial Exploration, University of Colorado Boulder, Boulder, CO 80309, USA.
| | - Stephanie N Salazar
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA.
| | - Amalia K Luthens
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA.
| | - Andrea L Arnold
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA.
| | - Brooke L Elam
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA.
| | - Ahmed I Elsayed
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA; Center for Microbial Exploration, University of Colorado Boulder, Boulder, CO 80309, USA.
| | - Mathias Leblanc
- Animal Resources Department, Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA.
| | - Sean C Adams
- Animal Resources Department, Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA.
| | - Christopher A Lowry
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA; Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA; Center for Microbial Exploration, University of Colorado Boulder, Boulder, CO 80309, USA; Department of Physical Medicine & Rehabilitation and Center for Neuroscience, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Veterans Health Administration, Rocky Mountain Mental Illness Research Education and Clinical Center (MIRECC), Rocky Mountain Regional Veterans Affairs Medical Center (RMRVAMC), Aurora, CO 80045, USA; Military and Veteran Microbiome: Consortium for Research and Education (MVM-CoRE), Aurora, CO 80045, USA.
| | - Jon D Reuter
- Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA; Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA; Office of Animal Resources, University of Colorado Boulder, Boulder, CO 80309, USA.
| |
Collapse
|
25
|
Antequera A, Stallings E, Henry RS, Lopez-Alcalde J, Runnels V, Tudiver S, Tugwell P, Welch V. Sex and Gender Appraisal Tool-Systematic Reviews-2 and Participation-To-Prevalence Ratio assessed to whom the evidence applies in sepsis reviews. J Clin Epidemiol 2021; 142:119-132. [PMID: 34763038 DOI: 10.1016/j.jclinepi.2021.11.006] [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: 04/07/2021] [Revised: 10/08/2021] [Accepted: 11/03/2021] [Indexed: 12/22/2022]
Abstract
OBJECTIVES To revise a sex and gender appraisal tool for systematic reviews (SGAT-SR) and apply it to Cochrane sepsis reviews. STUDY DESIGN AND SETTING The revision process was informed by existing literature on sex, gender, intersectionality, and feedback from an expert advisory board. We revised the items to consider additional factors associated with health inequities and appraised sex and gender considerations using the SGAT-SR-2 and female Participation-to-Prevalence Ratio (PPR) in Cochrane sepsis reviews. RESULTS SGAT-SR-2 consists of 19 questions appraising the review's sections and use of the terms sex and gender. amongst 71 SRs assessed, 50.7% included at least one tool item, the most frequent being the number of participants by sex or gender at included study-level (24/71 reviews). Only four reviews provided disaggregated data for the full set of included trials, while two considered other equity-related factors. Reviews rarely appraised possible similarities and differences across sex and gender. In half of a subset of reviews, female participants were under-represented relative to their share of the sepsis population (PPR<0.8). CONCLUSION The SGAT-SR-2 tool and the PPR can support the design and appraisal of systematic reviews to assess sex and gender considerations, address to whom evidence applies, and determine future research needs.
Collapse
Affiliation(s)
- A Antequera
- Biomedical Research Institute Sant Pau, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.
| | - E Stallings
- Clinical Biostatistics Unit, Instituto Ramón y Cajal de Investigación Sanitaria, Madrid, Spain. CIBER Epidemiología y Salud Pública (CIBERESP), Spain
| | - R S Henry
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada; Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| | - J Lopez-Alcalde
- Clinical Biostatistics Unit, Instituto Ramón y Cajal de Investigación Sanitaria, Madrid, Spain. CIBER Epidemiología y Salud Pública (CIBERESP), Spain; Institute for Complementary and Integrative Medicine, University Hospital Zurich and University of Zurich, Zurich, Switzerland; Universidad Francisco de Vitoria, Pozuelo de Alarcón, Spain
| | - V Runnels
- University of Ottawa, Ottawa, Ontario, Canada
| | - S Tudiver
- Researcher/Consultant - Gender and Health, Ottawa, Ontario, Canada
| | - P Tugwell
- Department of Medicine, and School of Epidemiology and Public Health, University of Ottawa, Ottawa, Ontario, Canada; Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada; WHO Collaborating Centre for Knowledge Translation and Health Technology Assessment in Health Equity, Bruyère Research Institute, Ottawa, Ontario, Canada
| | - V Welch
- Bruyère Research Institute, University of Ottawa, Ottawa, Ontario, Canada; School of Epidemiology and Public Health, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
26
|
Egesten A, Herwald H. Some Like It Hot. J Innate Immun 2021; 13:321-322. [PMID: 34724673 DOI: 10.1159/000520270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 10/14/2021] [Indexed: 11/19/2022] Open
|
27
|
Ozturk M, Chia JE, Hazra R, Saqib M, Maine R, Guler R, Suzuki H, Mishra BB, Brombacher F, Parihar SP. Evaluation of Berberine as an Adjunct to TB Treatment. Front Immunol 2021; 12:656419. [PMID: 34745081 PMCID: PMC8563784 DOI: 10.3389/fimmu.2021.656419] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 09/30/2021] [Indexed: 01/23/2023] Open
Abstract
Tuberculosis (TB) is the global health problem with the second highest number of deaths from a communicable disease after COVID-19. Although TB is curable, poor health infrastructure, long and grueling TB treatments have led to the spread of TB pandemic with alarmingly increasing multidrug-resistant (MDR)-TB prevalence. Alternative host modulating therapies can be employed to improve TB drug efficacies or dampen the exaggerated inflammatory responses to improve lung function. Here, we investigated the adjunct therapy of natural immune-modulatory compound berberine in C57BL/6 mouse model of pulmonary TB. Berberine treatment did not affect Mtb growth in axenic cultures; however, it showed increased bacterial killing in primary murine bone marrow-derived macrophages and human monocyte-derived macrophages. Ad libitum berberine administration was beneficial to the host in combination with rifampicin and isoniazid. Berberine adjunctive treatment resulted in decreased lung pathology with no additive or synergistic effects on bacterial burdens in mice. Lung immune cell flow cytometry analysis showed that adjunctive berberine treatment decreased neutrophil, CD11b+ dendritic cell and recruited interstitial macrophage numbers. Late onset of adjunctive berberine treatment resulted in a similar phenotype with consistently reduced numbers of neutrophils both in lungs and the spleen. Together, our results suggest that berberine can be supplemented as an immunomodulatory agent depending on the disease stage and inflammatory status of the host.
Collapse
Affiliation(s)
- Mumin Ozturk
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town, South Africa
- Division of Immunology and South African Medical Research Council (SAMRC) Immunology of Infectious Diseases, Institute of Infectious Diseases and Molecular Medicine (IDM), Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Julius E. Chia
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town, South Africa
- Division of Immunology and South African Medical Research Council (SAMRC) Immunology of Infectious Diseases, Institute of Infectious Diseases and Molecular Medicine (IDM), Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Rudranil Hazra
- Wellcome Centre for Infectious Diseases Research in Africa (CIDRI-Africa) and Institute of Infectious Diseases and Molecular Medicine (IDM), Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Division of Medical Microbiology, Institute of Infectious Diseases and Molecular Medicine (IDM), Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Mohd Saqib
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, United States
| | - Rebeng A. Maine
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town, South Africa
- Division of Immunology and South African Medical Research Council (SAMRC) Immunology of Infectious Diseases, Institute of Infectious Diseases and Molecular Medicine (IDM), Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Department of Molecular and Cell Biology, University of Cape Town, Cape Town, South Africa
| | - Reto Guler
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town, South Africa
- Division of Immunology and South African Medical Research Council (SAMRC) Immunology of Infectious Diseases, Institute of Infectious Diseases and Molecular Medicine (IDM), Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Wellcome Centre for Infectious Diseases Research in Africa (CIDRI-Africa) and Institute of Infectious Diseases and Molecular Medicine (IDM), Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Harukazu Suzuki
- RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Bibhuti B. Mishra
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, United States
| | - Frank Brombacher
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town, South Africa
- Division of Immunology and South African Medical Research Council (SAMRC) Immunology of Infectious Diseases, Institute of Infectious Diseases and Molecular Medicine (IDM), Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Wellcome Centre for Infectious Diseases Research in Africa (CIDRI-Africa) and Institute of Infectious Diseases and Molecular Medicine (IDM), Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Suraj P. Parihar
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town, South Africa
- Division of Immunology and South African Medical Research Council (SAMRC) Immunology of Infectious Diseases, Institute of Infectious Diseases and Molecular Medicine (IDM), Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Wellcome Centre for Infectious Diseases Research in Africa (CIDRI-Africa) and Institute of Infectious Diseases and Molecular Medicine (IDM), Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Division of Medical Microbiology, Institute of Infectious Diseases and Molecular Medicine (IDM), Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
28
|
Antequera A, Lopez-Alcalde J, Stallings E, Muriel A, Fernández Félix B, Del Campo R, Ponce-Alonso M, Fidalgo P, Halperin AV, Madrid-Pascual O, Álvarez-Díaz N, Solà I, Gordo F, Urrutia G, Zamora J. Sex as a prognostic factor for mortality in critically ill adults with sepsis: a systematic review and meta-analysis. BMJ Open 2021; 11:e048982. [PMID: 34551945 PMCID: PMC8461281 DOI: 10.1136/bmjopen-2021-048982] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
OBJECTIVE To assess the role of sex as an independent prognostic factor for mortality in patients with sepsis admitted to intensive care units (ICUs). DESIGN Systematic review and meta-analysis. DATA SOURCES MEDLINE, Embase, Web of Science, ClinicalTrials.gov and the WHO Clinical Trials Registry from inception to 17 July 2020. STUDY SELECTION Studies evaluating independent associations between sex and mortality in critically ill adults with sepsis controlling for at least one of five core covariate domains prespecified following a literature search and consensus among experts. DATA EXTRACTION AND SYNTHESIS Two authors independently extracted and assessed the risk of bias using Quality In Prognosis Studies tool. Meta-analysis was performed by pooling adjusted estimates. The Grades of Recommendations, Assessment, Development and Evaluation approach was used to rate the certainty of evidence. RESULTS From 14 304 records, 13 studies (80 520 participants) were included. Meta-analysis did not find sex-based differences in all-cause hospital mortality (OR 1.02, 95% CI 0.79 to 1.32; very low-certainty evidence) and all-cause ICU mortality (OR 1.19, 95% CI 0.79 to 1.78; very low-certainty evidence). However, females presented higher 28-day all-cause mortality (OR 1.18, 95% CI 1.05 to 1.32; very low-certainty evidence) and lower 1-year all-cause mortality (OR 0.83, 95% CI 0.68 to 0.98; low-certainty evidence). There was a moderate risk of bias in the domain adjustment for other prognostic factors in six studies, and the certainty of evidence was further affected by inconsistency and imprecision. CONCLUSION The prognostic independent effect of sex on all-cause hospital mortality, 28-day all-cause mortality and all-cause ICU mortality for critically ill adults with sepsis was uncertain. Female sex may be associated with decreased 1-year all-cause mortality. PROSPERO REGISTRATION NUMBER CRD42019145054.
Collapse
Affiliation(s)
- Alba Antequera
- Institut d'Investigació Biomèdica Sant Pau IIB Sant Pau, Barcelona, Spain
| | - Jesus Lopez-Alcalde
- Faculty of Health Sciences, Universidad Francisco de Vitoria, Pozuelo de Alarcón, Spain
- Clinical Biostatistics Unit, Instituto Ramon y Cajal de Investigacion Sanitaria, Madrid, Spain
- Institute for Complementary and Integrative Medicine, University Hospital Zurich and University Zurich, Zurich, Switzerland
- CIBERESP, Madrid, Spain
| | - Elena Stallings
- Clinical Biostatistics Unit, Instituto Ramon y Cajal de Investigacion Sanitaria, Madrid, Spain
- CIBERESP, Madrid, Spain
| | - Alfonso Muriel
- Clinical Biostatistics Unit, Instituto Ramon y Cajal de Investigacion Sanitaria, Madrid, Spain
- CIBERESP, Madrid, Spain
- Department of Nursing and Physiotherapy, Universidad de Alcala de Henares, Alcala de Henares, Spain
| | - Borja Fernández Félix
- Clinical Biostatistics Unit, Instituto Ramon y Cajal de Investigacion Sanitaria, Madrid, Spain
- CIBERESP, Madrid, Spain
| | - Rosa Del Campo
- Department of Microbiology, Hospital Universitario Ramon y Cajal, Madrid, Spain
| | - Manuel Ponce-Alonso
- Department of Microbiology, Hospital Universitario Ramon y Cajal, Madrid, Spain
| | - Pilar Fidalgo
- Faculty of Health Sciences, Universidad Francisco de Vitoria, Pozuelo de Alarcón, Spain
- Department of Internal Medicine, Hospital Universitario del Henares, Coslada, Spain
| | | | | | | | - Ivan Solà
- Iberoamerican Cochrane Centre, Institut d'Investigació Biomèdica Sant Pau IIB Sant Pau, Barcelona, Spain
- CIBERESP, Barcelona, Spain
| | - Federico Gordo
- Faculty of Health Sciences, Universidad Francisco de Vitoria, Pozuelo de Alarcón, Spain
- Department of Intensive Care, Hospital Universitario del Henares, Coslada, Spain
| | - Gerard Urrutia
- Iberoamerican Cochrane Centre, Institut d'Investigació Biomèdica Sant Pau IIB Sant Pau, Barcelona, Spain
- CIBERESP, Barcelona, Spain
| | - Javier Zamora
- Clinical Biostatistics Unit, Instituto Ramon y Cajal de Investigacion Sanitaria, Madrid, Spain
- CIBERESP, Madrid, Spain
- Institute of metabolism and systems research, University of Birmingham, Birmingham, UK
| |
Collapse
|
29
|
Age and Sex: Impact on adipose tissue metabolism and inflammation. Mech Ageing Dev 2021; 199:111563. [PMID: 34474078 DOI: 10.1016/j.mad.2021.111563] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 08/19/2021] [Accepted: 08/26/2021] [Indexed: 02/08/2023]
Abstract
Age associated chronic inflammation is a major contributor to diseases with advancing age. Adipose tissue function is at the nexus of processes contributing to age-related metabolic disease and mediating longevity. Hormonal fluctuations in aging potentially regulate age-associated visceral adiposity and metabolic dysfunction. Visceral adiposity in aging is linked to aberrant adipogenesis, insulin resistance, lipotoxicity and altered adipokine secretion. Age-related inflammatory phenomena depict sex differences in macrophage polarization, changes in T and B cell numbers, and types of dendritic cells. Sex differences are also observed in adipose tissue remodeling and cellular senescence suggesting a role for sex steroid hormones in the regulation of the adipose tissue microenvironment. It is crucial to investigate sex differences in aging clinical outcomes to identify and better understand physiology in at-risk individuals. Early interventions aimed at targets involved in adipose tissue adipogenesis, remodeling and inflammation in aging could facilitate a profound impact on health span and overcome age-related functional decline.
Collapse
|
30
|
Herwald H, Egesten A. C-Reactive Protein: More than a Biomarker. J Innate Immun 2021; 13:257-258. [PMID: 34469890 DOI: 10.1159/000519091] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 08/12/2021] [Indexed: 12/23/2022] Open
|
31
|
Association between high-sensitivity C-reactive protein levels and depression: Moderation by age, sex, obesity, and aerobic physical activity. J Affect Disord 2021; 291:375-383. [PMID: 34091325 DOI: 10.1016/j.jad.2021.05.040] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 05/08/2021] [Accepted: 05/23/2021] [Indexed: 11/24/2022]
Abstract
BACKGROUND Low-grade systemic inflammation evidenced by elevated serum high-sensitivity C-reactive protein (hsCRP) levels can be a biomarker for depression. This study aimed to investigate the association between serum hsCRP levels and depressive symptoms and to explore the potential moderating effects of age, sex, body mass index (BMI), and aerobic physical activity on the association. METHODS Data of 10,702 adults (≥ 19 years) were obtained from the nationwide cross-sectional Korea National Health and Nutrition Examination Surveys of 2016 and 2018. Significant depressive symptoms were defined as ≥ 10 on the Patient Health Questionnaire-9, and high hsCRP level was defined as > 3.0 mg/L. RESULTS Adults with high hsCRP levels were more likely to have depressive symptoms (odds ratio [OR]: 1.41, 95% confidence interval [CI]: 1.07-1.84) and suicidal ideation (OR: 1.39, 95% CI: 1.07-1.80) than those with low hsCRP levels. In the age- and sex-stratified analysis, high hsCRP levels were associated with depressive symptoms in the non-geriatric population (age ≤ 64 years) alone, with a higher OR in males than females. In subgroup analyses, the association between them was observed only among obese adults and adults without aerobic physical activity. LIMITATIONS Causal interpretation is limited due to the cross-sectional design. CONCLUSIONS Our results replicate previous findings of an association between high hsCRP levels and depressive symptoms in adults using a large nationally representative sample. The association between them was more prominent in the non-geriatric population, males, obese adults, and those without aerobic physical activity.
Collapse
|
32
|
Gusdon AM, Savarraj JPJ, Shihabeddin E, Paz A, Assing A, Ko SB, McCullough LD, Choi HA. Time Course of Peripheral Leukocytosis and Clinical Outcomes After Aneurysmal Subarachnoid Hemorrhage. Front Neurol 2021; 12:694996. [PMID: 34381415 PMCID: PMC8350167 DOI: 10.3389/fneur.2021.694996] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 06/24/2021] [Indexed: 11/29/2022] Open
Abstract
Objective: Systemic inflammation after subarachnoid hemorrhage (SAH) is implicated in delayed cerebral ischemia (DCI) and adverse clinical outcomes. We hypothesize that early changes in peripheral leukocytes will be associated with outcomes after SAH. Methods: SAH patients admitted between January 2009 and December 2016 were enrolled into a prospective observational study and were assessed for Hunt Hess Scale (HHS) at admission, DCI, and modified Ranked Scale (mRS) at discharge. Total white blood cell (WBC) counts and each component of the differential cell count were determined on the day of admission (day 0) to 8 days after bleed (day 8). Global cerebral edema (GCE) was assessed on admission CT, and presence of any infection was determined. Statistical tests included student's t-test, Chi-square test, and multivariate logistic regression (MLR) models. Results: A total of 451 subjects were analyzed. Total WBCs and neutrophils decreased initially reaching a minimum at day 4–5 after SAH. Monocyte count increased gradually after SAH and peaked between day 6–8, while basophils and lymphocytes decreased initially from day 0 to 1 and steadily increased thereafter. Neutrophil to lymphocyte ratio (NLR) reached a peak on day 1 and decreased thereafter. WBCs, neutrophils, monocytes, and NLR were higher in patients with DCI and poor functional outcomes. WBCs, neutrophils, and NLR were higher in subjects who developed infections. In MLR models, neutrophils and monocytes were associated with DCI and worse functional outcomes, while NLR was only associated with worse functional outcomes. Occurrence of infection was associated with poor outcome. Neutrophils and NLR were associated with infection, while monocytes were not. Monocytes were higher in males, and ROC curve analysis revealed improved ability of monocytes to predict DCI and poor functional outcomes in male subjects. Conclusions: Monocytosis was associated with DCI and poor functional outcomes after SAH. The association between neutrophils and NLR and infection may impact outcomes. Early elevation in monocytes had an improved ability to predict DCI and poor functional outcomes in males, which was independent of the occurrence of infection.
Collapse
Affiliation(s)
- Aaron M Gusdon
- Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center, Houston, TX, United States
| | - Jude P J Savarraj
- Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center, Houston, TX, United States
| | - Eyad Shihabeddin
- Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center, Houston, TX, United States
| | - Atzhiry Paz
- Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center, Houston, TX, United States
| | - Andres Assing
- Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center, Houston, TX, United States
| | - Sang-Bae Ko
- Department of Neurology, Seoul National University College of Medicine, Seoul, South Korea
| | - Louise D McCullough
- Department of Neurology, McGovern Medical School, University of Texas Health Science Center, Houston, TX, United States
| | - Huimahn Alex Choi
- Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center, Houston, TX, United States
| |
Collapse
|
33
|
Campesi I, Montella A, Franconi F. Human monocytes respond to lipopolysaccharide (LPS) stimulation in a sex-dependent manner. J Cell Physiol 2021; 237:580-588. [PMID: 34252202 PMCID: PMC9292909 DOI: 10.1002/jcp.30503] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 06/28/2021] [Accepted: 06/29/2021] [Indexed: 01/08/2023]
Abstract
Monocytes play a critical role in inflammation and immune response, their activity being sex‐dependent. However, the basis of sex differences is not well understood. Therefore, we investigated the lipopolysaccharide (LPS) effects on tumor necrosis factor‐α (TNF‐α) release, autophagy, and chemotaxis in freshly isolated monocytes from healthy young men and women. In basal conditions, male and female monocytes had similar TNF‐α release, chemotaxis, and estrogen receptors (ER‐α) and ER‐β expression, while the LC3II/I ratio was significantly higher in males. LPS treatment induced qualitative and quantitative sex differences. It reduced autophagy and increased TNF‐α release only in male monocytes, while, chemotaxis was significantly influenced only in female cells. Moreover, it reduced the expression of ER‐α only in female cells, while ER‐β expression was reduced in both sexes, but more markedly in female cells. Finally, the interplay between LPS treatment and 17‐β‐estradiol (E2) was present only in female cells. Globally, these findings expand the concept that sex plays a role in regulating monocytes' functions, being sex differences cell‐ and parameter‐specific.
Collapse
Affiliation(s)
- Ilaria Campesi
- Dipartimento di Scienze Biomediche, Università Degli Studi di Sassari, Sassari, Italy.,Laboratorio Nazionale di Medicina e Farmacologia di Genere, Istituto Nazionale Biostrutture Biosistemi, Sassari, Italy
| | - Andrea Montella
- Dipartimento di Scienze Biomediche, Università Degli Studi di Sassari, Sassari, Italy
| | - Flavia Franconi
- Laboratorio Nazionale di Medicina e Farmacologia di Genere, Istituto Nazionale Biostrutture Biosistemi, Sassari, Italy
| |
Collapse
|
34
|
Albracht CD, Hreha TN, Hunstad DA. Sex effects in pyelonephritis. Pediatr Nephrol 2021; 36:507-515. [PMID: 32040629 PMCID: PMC7415591 DOI: 10.1007/s00467-020-04492-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 12/30/2019] [Accepted: 01/27/2020] [Indexed: 02/07/2023]
Abstract
Urinary tract infections (UTIs) are generally considered a disease of women. However, UTIs affect females throughout the lifespan, and certain male populations (including infants and elderly men) are also susceptible. Epidemiologically, pyelonephritis is more common in women but carries increased morbidity when it does occur in men. Among children, high-grade vesicoureteral reflux is a primary risk factor for upper-tract UTI in both sexes. However, among young infants with UTI, girls are outnumbered by boys; risk factors include posterior urethral valves and lack of circumcision. Recent advances in mouse models of UTI reveal sex differences in innate responses to UTI, which vary somewhat depending on the system used. Moreover, male mice and androgenized female mice suffer worse outcomes in experimental pyelonephritis; evidence suggests that androgen exposure may suppress innate control of infection in the urinary tract, but additional androgen effects, as well as non-hormonal sex effects, may yet be specified. Among other intriguing directions, recent experiments raise the hypothesis that the postnatal testosterone surge that occurs in male infants may represent an additional factor driving the higher incidence of UTI in males under 6 months of age. Ongoing work in contemporary models will further illuminate sex- and sex-hormone-specific effects on UTI pathogenesis and immune responses.
Collapse
Affiliation(s)
- Clayton D Albracht
- Department of Pediatrics, Washington University School of Medicine, 660 S. Euclid Ave., Campus Box 8208, St. Louis, MO, 63110, USA
| | - Teri N Hreha
- Department of Pediatrics, Washington University School of Medicine, 660 S. Euclid Ave., Campus Box 8208, St. Louis, MO, 63110, USA
| | - David A Hunstad
- Department of Pediatrics, Washington University School of Medicine, 660 S. Euclid Ave., Campus Box 8208, St. Louis, MO, 63110, USA.
- Department of Molecular Microbiology, Washington University School of Medicine, 660 S. Euclid Ave., Campus Box 8230, St. Louis, MO, 63110, USA.
| |
Collapse
|
35
|
Pijls BG, Jolani S, Atherley A, Derckx RT, Dijkstra JIR, Franssen GHL, Hendriks S, Richters A, Venemans-Jellema A, Zalpuri S, Zeegers MP. Demographic risk factors for COVID-19 infection, severity, ICU admission and death: a meta-analysis of 59 studies. BMJ Open 2021; 11:e044640. [PMID: 33431495 PMCID: PMC7802392 DOI: 10.1136/bmjopen-2020-044640] [Citation(s) in RCA: 334] [Impact Index Per Article: 83.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
OBJECTIVE We aimed to describe the associations of age and sex with the risk of COVID-19 in different severity stages ranging from infection to death. DESIGN Systematic review and meta-analysis. DATA SOURCES PubMed and Embase through 4 May 2020. STUDY SELECTION We considered cohort and case-control studies that evaluated differences in age and sex on the risk of COVID-19 infection, disease severity, intensive care unit (ICU) admission and death. DATA EXTRACTION AND SYNTHESIS We screened and included studies using standardised electronic data extraction forms and we pooled data from published studies and data acquired by contacting authors using random effects meta-analysis. We assessed the risk of bias using the Newcastle-Ottawa Scale. RESULTS We screened 11.550 titles and included 59 studies comprising 36.470 patients in the analyses. The methodological quality of the included papers was high (8.2 out of 9). Men had a higher risk for infection with COVID-19 than women (relative risk (RR) 1.08, 95% CI 1.03 to 1.12). When infected, they also had a higher risk for severe COVID-19 disease (RR 1.18, 95% CI 1.10 to 1.27), a higher need for intensive care (RR 1.38, 95% CI 1.09 to 1.74) and a higher risk of death (RR 1.50, 95% CI 1.18 to 1.91). The analyses also showed that patients aged 70 years and above have a higher infection risk (RR 1.65, 95% CI 1.50 to 1.81), a higher risk for severe COVID-19 disease (RR 2.05, 95% CI 1.27 to 3.32), a higher need for intensive care (RR 2.70, 95% CI 1.59 to 4.60) and a higher risk of death once infected (RR 3.61, 95% CI 2.70 to 4.84) compared with patients younger than 70 years. CONCLUSIONS Meta-analyses on 59 studies comprising 36.470 patients showed that men and patients aged 70 and above have a higher risk for COVID-19 infection, severe disease, ICU admission and death. PROSPERO REGISTRATION NUMBER CRD42020180085.
Collapse
Affiliation(s)
- Bart G Pijls
- Department of Orthopaedics, Leiden University Medical Center, Leiden, The Netherlands
| | - Shahab Jolani
- Department of Methodology and Statistics, Care and Public Health Research Institute (CAPHRI), Maastricht University, Maastricht, The Netherlands
| | - Anique Atherley
- Department of Educational Research and Development, School of Health Professions Education, Maastricht University, Maastricht, The Netherlands
| | - Raissa T Derckx
- Department of General Practice, Care and Public Health Research Institute (CAPHRI), Maastricht University, Maastricht, The Netherlands
| | - Janna I R Dijkstra
- Amsterdam University Medical Center, location VUmc, Amsterdam, The Netherlands
| | - Gregor H L Franssen
- Maastricht University Library, Maastricht University, Maastricht, The Netherlands
| | - Stevie Hendriks
- School of Mental Health and Neuroscience (MHeNS), Maastricht University, Maastricht, The Netherlands
| | - Anke Richters
- Department of Research and Development, The Netherlands Comprehensive Cancer Organisation, Utrecht, The Netherlands
| | | | | | - Maurice P Zeegers
- NUTRIM School of Translational Research in Metabolism, Care and Public Health Research Institute (CAPHRI), Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
36
|
De Maeyer RPH, Chambers ES. The impact of ageing on monocytes and macrophages. Immunol Lett 2020; 230:1-10. [PMID: 33309673 DOI: 10.1016/j.imlet.2020.12.003] [Citation(s) in RCA: 141] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 12/03/2020] [Accepted: 12/07/2020] [Indexed: 12/12/2022]
Abstract
Ageing is a global burden. Increasing age is associated with increased incidence of infections and cancer and decreased vaccine efficacy. This increased morbidity observed with age, is believed to be due in part to a decline in adaptive immunity, termed immunosenescence. However not all aspects of immunity decrease with age as ageing presents with systemic low grade chronic inflammation, characterised by elevated concentrations of mediators such as IL-6, TNFα and C Reactive protein (CRP). Inflammation is a strong predictor of morbidity and mortality, and chronic inflammation is known to be detrimental to a functioning immune system. Although the source of the inflammation is much discussed, the key cells which are believed to facilitate the inflammageing phenomenon are the monocytes and macrophages. In this review we detail how macrophage and monocyte phenotype and function change with age. The impact of ageing on macrophages includes decreased phagocytosis and immune resolution, increased senescent-associated markers, increased inflammatory cytokine production, reduced autophagy, and a decrease in TLR expression. With monocytes there is an increase in circulating CD16+ monocytes, decreased type I IFN production, and decreased efferocytosis. In conclusion, we believe that monocytes and macrophages contribute to immunosenescence and inflammageing and as a result have an important role in defective immunity with age.
Collapse
Affiliation(s)
| | - Emma S Chambers
- Centre of Immunobiology, Blizard Institute, Queen Mary University of London, London, UK.
| |
Collapse
|
37
|
Regional, cellular and species difference of two key neuroinflammatory genes implicated in schizophrenia. Brain Behav Immun 2020; 88:826-839. [PMID: 32450195 DOI: 10.1016/j.bbi.2020.05.055] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 05/20/2020] [Accepted: 05/20/2020] [Indexed: 02/07/2023] Open
Abstract
The transcription factor nuclear factor kappa B (NF-κB) regulates the expression of many inflammatory genes that are overexpressed in a subset of people with schizophrenia. Transcriptional reduction in one NF-κB inhibitor, Human Immunodeficiency Virus Enhancer Binding Protein 2 (HIVEP2), is found in the brain of patients, aligning with evidence of NF-κB over-activity. Cellular co-expression of HIVEP2 and cytokine transcripts is a prerequisite for a direct effect of HIVEP2 on pro-inflammatory transcription, and we do not know if changes in HIVEP2 and markers of neuroinflammation are occurring in the same brain cell type. We performed in situ hybridisation on postmortem dorsolateral prefrontal cortex tissue to map and compare the expression of HIVEP2 and Serpin Family A Member 3 (SERPINA3), one of the most consistently increased inflammatory genes in schizophrenia, between schizophrenia patients and controls. We find that HIVEP2 expression is neuronal and is decreased in almost all grey matter cortical layers in schizophrenia patients with neuroinflammation, and that SERPINA3 is increased in the dorsolateral prefrontal cortex grey matter and white matter in the same group of patients. We are the first to map the upregulation of SERPINA3 to astrocytes and to some neurons, and find evidence to suggest that blood vessel-associated astrocytes are the main cellular source of SERPINA3 in the schizophrenia cortex. We show that a lack of HIVEP2 in mice does not cause astrocytic upregulation of Serpina3n but does induce its transcription in neurons. We speculate that HIVEP2 downregulation is not a direct cause of astrocytic pro-inflammatory cytokine synthesis in schizophrenia but may contribute to neuronally-mediated neuroinflammation.
Collapse
|
38
|
Gilli F, DiSano KD, Pachner AR. SeXX Matters in Multiple Sclerosis. Front Neurol 2020; 11:616. [PMID: 32719651 PMCID: PMC7347971 DOI: 10.3389/fneur.2020.00616] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 05/27/2020] [Indexed: 12/12/2022] Open
Abstract
Multiple sclerosis (MS) is the most common chronic inflammatory and neurodegenerative disease of the central nervous system (CNS). An interesting feature that this debilitating disease shares with many other inflammatory disorders is that susceptibility is higher in females than in males, with the risk of MS being three times higher in women compared to men. Nonetheless, while men have a decreased risk of developing MS, many studies suggest that males have a worse clinical outcome. MS exhibits an apparent sexual dimorphism in both the immune response and the pathophysiology of the CNS damage, ultimately affecting disease susceptibility and progression differently. Overall, women are predisposed to higher rates of inflammatory relapses than men, but men are more likely to manifest signs of disease progression and worse CNS damage. The observed sexual dimorphism in MS may be due to sex hormones and sex chromosomes, acting in parallel or combination. In this review, we outline current knowledge on the sexual dimorphism in MS and discuss the interplay of sex chromosomes, sex hormones, and the immune system in driving MS disease susceptibility and progression.
Collapse
Affiliation(s)
- Francesca Gilli
- Department of Neurology, Dartmouth Hitchcock Medical Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
| | | | | |
Collapse
|
39
|
Bernardi S, Toffoli B, Tonon F, Francica M, Campagnolo E, Ferretti T, Comar S, Giudici F, Stenner E, Fabris B. Sex Differences in Proatherogenic Cytokine Levels. Int J Mol Sci 2020; 21:ijms21113861. [PMID: 32485823 PMCID: PMC7311959 DOI: 10.3390/ijms21113861] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/22/2020] [Accepted: 05/27/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND It has been shown that sex affects immunity, including cytokine production. Given that atherosclerosis is an inflammatory disease promoted by specific cytokines, such as interleukin (IL)-1β, IL-6, and tumor necrosis factor (TNF)-α, we aimed at evaluating whether sex could affect the levels of these proatherogenic cytokines in a group of healthy adults. In this analysis, we also included other cytokines and peptides that have been implicated in atherosclerosis development and progression. METHODS A total of 104 healthy adults were recruited; we measured circulating levels of IL-1β, IL-6, TNF-α, angiotensins and angiotensin-converting enzyme-2 (ACE2), as well as osteoprotegerin and receptor activator of nuclear factor κB ligand (RANKL). RESULTS IL-1β, IL-6, and TNF-α were significantly higher in men as compared to women. They were all associated with testosterone and the testosterone/estradiol ratio. They remained significantly associated with sex (but not with hormones) after being tested for potential confounders. CONCLUSIONS Sex seems to influence the levels of proatherogenic cytokines. This is consistent not only with sex differences in vulnerability to infections but also with the higher cardiovascular risk exhibited by the male gender as compared to the female gender. Nevertheless, this association is only partly explained by hormone levels.
Collapse
Affiliation(s)
- Stella Bernardi
- Department of Medical, Surgical, and Health Sciences, University of Trieste, Cattinara Teaching Hospital UCO Medicina Clinica, 34100 Trieste, Italy; (F.T.); (M.F.); (E.C.); (T.F.); (S.C.); (F.G.); (B.F.)
- ASUGI Azienda Sanitaria Universitaria Integrata di Trieste, Cattinara Teaching Hospital, UCO Medicina Clinica, 34100 Trieste, Italy
- Correspondence: ; Tel.: +39-040-399-4318
| | - Barbara Toffoli
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, 34100 Trieste, Italy;
| | - Federica Tonon
- Department of Medical, Surgical, and Health Sciences, University of Trieste, Cattinara Teaching Hospital UCO Medicina Clinica, 34100 Trieste, Italy; (F.T.); (M.F.); (E.C.); (T.F.); (S.C.); (F.G.); (B.F.)
| | - Morena Francica
- Department of Medical, Surgical, and Health Sciences, University of Trieste, Cattinara Teaching Hospital UCO Medicina Clinica, 34100 Trieste, Italy; (F.T.); (M.F.); (E.C.); (T.F.); (S.C.); (F.G.); (B.F.)
| | - Elena Campagnolo
- Department of Medical, Surgical, and Health Sciences, University of Trieste, Cattinara Teaching Hospital UCO Medicina Clinica, 34100 Trieste, Italy; (F.T.); (M.F.); (E.C.); (T.F.); (S.C.); (F.G.); (B.F.)
| | - Tommaso Ferretti
- Department of Medical, Surgical, and Health Sciences, University of Trieste, Cattinara Teaching Hospital UCO Medicina Clinica, 34100 Trieste, Italy; (F.T.); (M.F.); (E.C.); (T.F.); (S.C.); (F.G.); (B.F.)
| | - Sarah Comar
- Department of Medical, Surgical, and Health Sciences, University of Trieste, Cattinara Teaching Hospital UCO Medicina Clinica, 34100 Trieste, Italy; (F.T.); (M.F.); (E.C.); (T.F.); (S.C.); (F.G.); (B.F.)
- ASUGI Azienda Sanitaria Universitaria Integrata di Trieste, Cattinara Teaching Hospital, UCO Medicina Clinica, 34100 Trieste, Italy
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, 34100 Trieste, Italy;
- Department of Life Sciences, University of Trieste, 34100 Trieste, Italy
| | - Fabiola Giudici
- Department of Medical, Surgical, and Health Sciences, University of Trieste, Cattinara Teaching Hospital UCO Medicina Clinica, 34100 Trieste, Italy; (F.T.); (M.F.); (E.C.); (T.F.); (S.C.); (F.G.); (B.F.)
- ASUGI Azienda Sanitaria Universitaria Integrata di Trieste, Cattinara Teaching Hospital, UCO Medicina Clinica, 34100 Trieste, Italy
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, 34100 Trieste, Italy;
- Department of Life Sciences, University of Trieste, 34100 Trieste, Italy
- Unit of Biostatistics, Epidemiology and Public Health, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padua, 35100 Padova, Italy
| | - Elisabetta Stenner
- Department of Diagnostics, Azienda USL Toscana Nordovest, 57100 Livorno, Italy;
| | - Bruno Fabris
- Department of Medical, Surgical, and Health Sciences, University of Trieste, Cattinara Teaching Hospital UCO Medicina Clinica, 34100 Trieste, Italy; (F.T.); (M.F.); (E.C.); (T.F.); (S.C.); (F.G.); (B.F.)
- ASUGI Azienda Sanitaria Universitaria Integrata di Trieste, Cattinara Teaching Hospital, UCO Medicina Clinica, 34100 Trieste, Italy
| |
Collapse
|
40
|
Herwald H, Egesten A. Serious, Severe, Sepsis. J Innate Immun 2020; 12:129-130. [PMID: 32007993 DOI: 10.1159/000505623] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Accepted: 12/21/2019] [Indexed: 11/19/2022] Open
|