1
|
Zhao R, Zhang X, Geng Y, Lu D, Wang Y, Xie H, Zhang X, Xu S, Cao Y. SPRY1 regulates macrophage M1 polarization in skin aging and melanoma prognosis. Transl Oncol 2025; 54:102331. [PMID: 40023001 PMCID: PMC11915026 DOI: 10.1016/j.tranon.2025.102331] [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: 11/28/2024] [Revised: 01/28/2025] [Accepted: 02/10/2025] [Indexed: 03/04/2025] Open
Abstract
INTRODUCTION Skin aging is a complex, multifactorial process involving cellular damage, inflammation, and increased susceptibility to diseases. Despite its importance, the role of SPRY1 in skin aging remains poorly understood. This study aims to investigate the function of SPRY1 in skin aging, particularly its impact on macrophage M1 polarization, and explore its potential as a therapeutic target for mitigating skin aging and melanoma. METHODS Bioinformatics analyses were performed using datasets from the GTEx and GEO databases, alongside in vitro cellular experiments. These included Weighted Gene Co-expression Network Analysis (WGCNA), single-cell sequencing, and various cellular assays in RAW264.7 murine monocyte/macrophage leukemia cells and NIH/3T3 mouse skin fibroblasts. The assays comprised gene transfection, Cell Counting Kit-8 (CCK-8) assays, quantitative real-time PCR (qRT-PCR), and measurements of reactive oxygen species (ROS) and superoxide dismutase (SOD) activity. RESULTS SPRY1 was identified as a key gene within modules linked to skin aging. Single-cell sequencing revealed its enrichment in macrophages and keratinocytes. Knockdown of SPRY1 in RAW264.7 cells resulted in a shift from M1 to M2 macrophage polarization, reduced oxidative stress, and decreased expression of inflammatory markers. In NIH/3T3 cells, SPRY1 knockdown reduced cell viability and lowered the expression of inflammatory genes. Additionally, SPRY1 expression was downregulated in melanoma, and its reduced levels were associated with poorer survival outcomes. CONCLUSIONS SPRY1 accelerates skin aging by promoting macrophage M1 polarization and may serve as a promising therapeutic target. Future research should focus on in vivo validation and further exploration of its regulatory networks to develop novel treatments.
Collapse
Affiliation(s)
- Rongxin Zhao
- Department of Dermatology, Pudong New Area People's Hospital, 490 Chuanhuang South Road, Pudong New Area, Shanghai, China
| | - Xun Zhang
- Digestive Endoscopy Center, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, 1111 XianXia Road, Shanghai 200336, China
| | - Yingnan Geng
- Department of Burns and Plastic Surgery, Second Affiliated Hospital of Naval Medical University, 415 Fengyang Road, Huangpu District, Shanghai 200003, China
| | - Dan Lu
- Department of Dermatology, Pudong New Area People's Hospital, 490 Chuanhuang South Road, Pudong New Area, Shanghai, China
| | - Yuqing Wang
- Department of Dermatology, Xuzhou Huamei Cosmetology Hospital, Jiangsu, West Huaihai Road, Quanshan District, Xuzhou, Jiangsu, China
| | - Han Xie
- The Fifth People's Hospital of Shanghai, Fudan University, No. 128, Ruili Road, Minhang District, Shanghai, China
| | - Xiaofei Zhang
- Shanghai Xinmei Medical Beauty Outpatient Department, 202A, No.285, Jianguo West Road, Xuhui District, Shanghai, China.
| | - Shunming Xu
- Department of Dermatology, Pudong New Area People's Hospital, 490 Chuanhuang South Road, Pudong New Area, Shanghai, China.
| | - Yanyun Cao
- Department of Dermatology, Pudong New Area People's Hospital, 490 Chuanhuang South Road, Pudong New Area, Shanghai, China.
| |
Collapse
|
2
|
Pereiro P, Tur R, García M, Figueras A, Novoa B. Unravelling turbot ( Scophthalmus maximus) resistance to Aeromonas salmonicida: transcriptomic insights from two full-sibling families with divergent susceptibility. Front Immunol 2024; 15:1522666. [PMID: 39712009 PMCID: PMC11659141 DOI: 10.3389/fimmu.2024.1522666] [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: 11/04/2024] [Accepted: 11/22/2024] [Indexed: 12/24/2024] Open
Abstract
Introduction Furunculosis, caused by the gram-negative bacterium Aeromonas salmonicida subsp. salmonicida, remains a significant threat to turbot (Scophthalmus maximus) aquaculture. Identifying genetic backgrounds with enhanced disease resistance is critical for improving aquaculture health management, reducing antibiotic dependency, and mitigating economic losses. Methods In this study, five full-sibling turbot families were challenged with A. salmonicida, which revealed one family with significantly greater resistance. Transcriptomic analyses (RNA-Seq) were performed on resistant and susceptible families, examining both naïve and 24-h postinfection (hpi) samples from head kidney and liver tissues. Results In the absence of infection, differentially expressed genes (DEGs) were identified predominantly in the liver. Following infection, a marked increase in DEGs was observed in the head kidney, with many genes linked to immune functions. Interestingly, the resistant family displayed a more controlled inflammatory response and upregulation of genes related to antigen presentation and T-cell activity in the head kidney at early infection stages, which may have contributed to its increased survival rate. In the liver, transcriptomic differences between the families were associated mainly with cytoskeletal organization, cell cycle regulation, and metabolic processes, including insulin signalling and lipid metabolism, regardless of infection status. Additionally, many DEGs overlapped with previously identified quantitative trait loci (QTLs) associated with resistance to A. salmonicida, providing further insights into the genetic basis of disease resistance. Discussion This study represents the first RNA-Seq analysis comparing resistant and susceptible turbot families and contributes valuable knowledge for the development of selective breeding programs targeting disease resistance in turbot and other aquaculture species susceptible to A. salmonicida.
Collapse
Affiliation(s)
- Patricia Pereiro
- Instituto de Investigaciones Marinas (IIM), Consejo Superior de Investigaciones Científicas (CSIC), Vigo, Spain
| | - Ricardo Tur
- Nueva Pescanova Biomarine Center, S.L., O Grove, Spain
| | - Miguel García
- Nueva Pescanova Biomarine Center, S.L., O Grove, Spain
| | - Antonio Figueras
- Instituto de Investigaciones Marinas (IIM), Consejo Superior de Investigaciones Científicas (CSIC), Vigo, Spain
| | - Beatriz Novoa
- Instituto de Investigaciones Marinas (IIM), Consejo Superior de Investigaciones Científicas (CSIC), Vigo, Spain
| |
Collapse
|
3
|
Specktor P, Hadar D, Cohen H. Glucocorticoid treatment during COVID-19 infection: does it affect the incidence of long COVID? Inflammopharmacology 2024; 32:3707-3715. [PMID: 39361178 DOI: 10.1007/s10787-024-01576-y] [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: 09/06/2024] [Accepted: 09/16/2024] [Indexed: 11/10/2024]
Abstract
BACKGROUND Long COVID (LC) is a frequent complication of COVID infection. It usually results in cognitive impairment, myalgia, headache and fatigue. No effective treatment has been found yet. We aimed to explore the effect of glucocorticoid (GC) treatment during COVID-19 infection on the later development of LC. METHODS We examined electronic health records from Clalit Health Services for documentation of COVID-19, GC treatment, and LC frequency. Background diagnoses, demographic data, hospitalization rates, and the use of anti-COVID drugs were recorded. RESULTS 1,322,599 cases of COVID-19 infection met the inclusion criteria; 13,530 patients (1.02%) received GC treatment. 149,272 patients, 11.29% of COVID-19 patients were diagnosed with LC. Age and female gender were prognostic risk factors for LC (OR 1.06 for age, OR 1.4 for female gender; p value < 0.0001). Background psychiatric diagnoses, migraine, backache and irritable bowel syndrome were predisposing conditions for LC (OR 2.7, p value < .0001). Higher BMI was associated with a greater probability of LC (OR of 1.25 for obese population). COVID patients who received GC were diagnosed with LC more frequently: 2294 cases (16.95%) compared to 146,978 cases (11.23%) in the non-GC group; (adjusted OR of 1.28 ± 0.07, 95% CI, p < 0.0001). CONCLUSIONS GC treatment during COVID-19 is correlated with the development of LC. In vivo and animal models may be used to explore the mechanism of this correlation. Future directions include prospective studies as well.
Collapse
Affiliation(s)
- Polina Specktor
- Department of Neurology, Carmel Medical Center, Mikhal St 7, 3436212, Haifa, Israel.
| | - Dana Hadar
- Department of Neurology, Carmel Medical Center, Mikhal St 7, 3436212, Haifa, Israel
| | - Hilla Cohen
- Department of Neurology, Carmel Medical Center, Mikhal St 7, 3436212, Haifa, Israel
| |
Collapse
|
4
|
Severa M, Ricci D, Etna MP, Facchini M, Puzelli S, Fedele G, Iorio E, Cairo G, Castrechini S, Ungari V, Iannetta M, Leone P, Chirico M, Pisanu ME, Bottazzi B, Benedetti L, Sali M, Bartolomucci R, Balducci S, Garlanda C, Stefanelli P, Spadea A, Palamara AT, Coccia EM. A Serum Multi-Parametric Analysis Identifies an Early Innate Immune Signature Associated to Increased Vaccine-Specific Antibody Production and Seroconversion in Simultaneous COVID-19 mRNA and Cell-Based Quadrivalent Influenza Vaccination. Vaccines (Basel) 2024; 12:1050. [PMID: 39340080 PMCID: PMC11436141 DOI: 10.3390/vaccines12091050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 08/28/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024] Open
Abstract
In this pilot study, a multi-parametric analysis comparing immune responses in sera of adult healthy subjects (HS) or people with type 2 diabetes mellitus (T2D) undergoing the single or simultaneous administration of mRNA-based COVID-19 and cellular quadrivalent inactivated influenza vaccines was conducted. While SARS-CoV-2 antibodies remains comparable, influenza antibody titers and seroconversion were significantly higher upon simultaneous vaccination. Magnitude of anti-influenza humoral response closely correlated with an early innate immune signature, previously described for the COVID-19 vaccine, composed of IL-15, IL-6, TNF-α, IFN-γ, CXCL-10 and here extended also to acute-phase protein Pentraxin 3. People with T2D receiving simultaneous vaccination showed a protective response comparable to HS correlating with the early induction of IFN-γ/CXCL10 and a significant reduction of the circulating glucose level due to increased oxidation of glucose digestion and consumption. These data, although preliminary and in-need of validation in larger cohorts, might be exploited to optimize future vaccination in people with chronic disorders, including diabetes.
Collapse
Affiliation(s)
- Martina Severa
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy; (D.R.); (M.P.E.); (M.F.); (S.P.); (G.F.); (G.C.); (P.L.); (P.S.); (A.T.P.)
| | - Daniela Ricci
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy; (D.R.); (M.P.E.); (M.F.); (S.P.); (G.F.); (G.C.); (P.L.); (P.S.); (A.T.P.)
| | - Marilena Paola Etna
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy; (D.R.); (M.P.E.); (M.F.); (S.P.); (G.F.); (G.C.); (P.L.); (P.S.); (A.T.P.)
| | - Marzia Facchini
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy; (D.R.); (M.P.E.); (M.F.); (S.P.); (G.F.); (G.C.); (P.L.); (P.S.); (A.T.P.)
| | - Simona Puzelli
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy; (D.R.); (M.P.E.); (M.F.); (S.P.); (G.F.); (G.C.); (P.L.); (P.S.); (A.T.P.)
| | - Giorgio Fedele
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy; (D.R.); (M.P.E.); (M.F.); (S.P.); (G.F.); (G.C.); (P.L.); (P.S.); (A.T.P.)
| | - Egidio Iorio
- High Resolution NMR Unit, Core Facilities, Istituto Superiore di Sanità, 00161 Rome, Italy; (E.I.); (M.C.); (M.E.P.)
| | - Giada Cairo
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy; (D.R.); (M.P.E.); (M.F.); (S.P.); (G.F.); (G.C.); (P.L.); (P.S.); (A.T.P.)
| | - Sara Castrechini
- ASL ROMA 1, Regione Lazio, 00145 Rome, Italy; (S.C.); (V.U.); (R.B.); (A.S.)
| | - Valentina Ungari
- ASL ROMA 1, Regione Lazio, 00145 Rome, Italy; (S.C.); (V.U.); (R.B.); (A.S.)
| | - Marco Iannetta
- Infectious Disease Clinic, Tor Vergata University Hospital, 00133 Rome, Italy; (M.I.); (L.B.)
| | - Pasqualina Leone
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy; (D.R.); (M.P.E.); (M.F.); (S.P.); (G.F.); (G.C.); (P.L.); (P.S.); (A.T.P.)
| | - Mattea Chirico
- High Resolution NMR Unit, Core Facilities, Istituto Superiore di Sanità, 00161 Rome, Italy; (E.I.); (M.C.); (M.E.P.)
| | - Maria Elena Pisanu
- High Resolution NMR Unit, Core Facilities, Istituto Superiore di Sanità, 00161 Rome, Italy; (E.I.); (M.C.); (M.E.P.)
| | - Barbara Bottazzi
- Department of Inflammation and Immunology, Humanitas Clinical and Research Centre—IRCCS, 20019 Milan, Italy; (B.B.); (C.G.)
| | - Livia Benedetti
- Infectious Disease Clinic, Tor Vergata University Hospital, 00133 Rome, Italy; (M.I.); (L.B.)
| | - Michela Sali
- Department of Laboratory and Infectious Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy;
- Department of Basic Biotechnological Sciences, Intensive and Perioperative Clinics, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Remo Bartolomucci
- ASL ROMA 1, Regione Lazio, 00145 Rome, Italy; (S.C.); (V.U.); (R.B.); (A.S.)
| | | | - Cecilia Garlanda
- Department of Inflammation and Immunology, Humanitas Clinical and Research Centre—IRCCS, 20019 Milan, Italy; (B.B.); (C.G.)
- Department of Biomedical Sciences, Humanitas University, 20090 Milan, Italy
| | - Paola Stefanelli
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy; (D.R.); (M.P.E.); (M.F.); (S.P.); (G.F.); (G.C.); (P.L.); (P.S.); (A.T.P.)
| | - Antonietta Spadea
- ASL ROMA 1, Regione Lazio, 00145 Rome, Italy; (S.C.); (V.U.); (R.B.); (A.S.)
| | - Anna Teresa Palamara
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy; (D.R.); (M.P.E.); (M.F.); (S.P.); (G.F.); (G.C.); (P.L.); (P.S.); (A.T.P.)
| | - Eliana Marina Coccia
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy; (D.R.); (M.P.E.); (M.F.); (S.P.); (G.F.); (G.C.); (P.L.); (P.S.); (A.T.P.)
| |
Collapse
|
5
|
Wensveen FM, Šestan M, Polić B. The immunology of sickness metabolism. Cell Mol Immunol 2024; 21:1051-1065. [PMID: 39107476 PMCID: PMC11364700 DOI: 10.1038/s41423-024-01192-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 05/29/2024] [Indexed: 09/01/2024] Open
Abstract
Everyone knows that an infection can make you feel sick. Although we perceive infection-induced changes in metabolism as a pathology, they are a part of a carefully regulated process that depends on tissue-specific interactions between the immune system and organs involved in the regulation of systemic homeostasis. Immune-mediated changes in homeostatic parameters lead to altered production and uptake of nutrients in circulation, which modifies the metabolic rate of key organs. This is what we experience as being sick. The purpose of sickness metabolism is to generate a metabolic environment in which the body is optimally able to fight infection while denying vital nutrients for the replication of pathogens. Sickness metabolism depends on tissue-specific immune cells, which mediate responses tailored to the nature and magnitude of the threat. As an infection increases in severity, so do the number and type of immune cells involved and the level to which organs are affected, which dictates the degree to which we feel sick. Interestingly, many alterations associated with metabolic disease appear to overlap with immune-mediated changes observed following infection. Targeting processes involving tissue-specific interactions between activated immune cells and metabolic organs therefore holds great potential for treating both people with severe infection and those with metabolic disease. In this review, we will discuss how the immune system communicates in situ with organs involved in the regulation of homeostasis and how this communication is impacted by infection.
Collapse
Affiliation(s)
| | - Marko Šestan
- University of Rijeka Faculty of Medicine, Rijeka, Croatia
| | - Bojan Polić
- University of Rijeka Faculty of Medicine, Rijeka, Croatia
| |
Collapse
|
6
|
Šestan M, Mikašinović S, Benić A, Wueest S, Dimitropoulos C, Mladenić K, Krapić M, Hiršl L, Glantzspiegel Y, Rasteiro A, Aliseychik M, Cekinović Grbeša Đ, Turk Wensveen T, Babić M, Gat-Viks I, Veiga-Fernandes H, Konrad D, Wensveen FM, Polić B. An IFNγ-dependent immune-endocrine circuit lowers blood glucose to potentiate the innate antiviral immune response. Nat Immunol 2024; 25:981-993. [PMID: 38811816 DOI: 10.1038/s41590-024-01848-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 04/18/2024] [Indexed: 05/31/2024]
Abstract
Viral infection makes us feel sick as the immune system alters systemic metabolism to better fight the pathogen. The extent of these changes is relative to the severity of disease. Whether blood glucose is subject to infection-induced modulation is mostly unknown. Here we show that strong, nonlethal infection restricts systemic glucose availability, which promotes the antiviral type I interferon (IFN-I) response. Following viral infection, we find that IFNγ produced by γδ T cells stimulates pancreatic β cells to increase glucose-induced insulin release. Subsequently, hyperinsulinemia lessens hepatic glucose output. Glucose restriction enhances IFN-I production by curtailing lactate-mediated inhibition of IRF3 and NF-κB signaling. Induced hyperglycemia constrained IFN-I production and increased mortality upon infection. Our findings identify glucose restriction as a physiological mechanism to bring the body into a heightened state of responsiveness to viral pathogens. This immune-endocrine circuit is disrupted in hyperglycemia, possibly explaining why patients with diabetes are more susceptible to viral infection.
Collapse
Affiliation(s)
- Marko Šestan
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
- Champalimaud Research, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Sanja Mikašinović
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Ante Benić
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Stephan Wueest
- Division of Pediatric Endocrinology and Diabetology and Children's Research Centre, University Children's Hospital, University of Zurich, Zurich, Switzerland
| | | | - Karlo Mladenić
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Mia Krapić
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Lea Hiršl
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Yossef Glantzspiegel
- School of Molecular Cell Biology and Biotechnology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Ana Rasteiro
- Champalimaud Research, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Maria Aliseychik
- Champalimaud Research, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | | | - Tamara Turk Wensveen
- Center for Diabetes, Endocrinology and Cardiometabolism, Thallassotherapia, Opatija, Croatia
- Department of Internal Medicine, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Marina Babić
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
- Innate Immunity, German Rheumatism Research Centre, Leibniz Institute, Berlin, Germany
| | - Irit Gat-Viks
- School of Molecular Cell Biology and Biotechnology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | | | - Daniel Konrad
- Division of Pediatric Endocrinology and Diabetology and Children's Research Centre, University Children's Hospital, University of Zurich, Zurich, Switzerland
| | - Felix M Wensveen
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Bojan Polić
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia.
| |
Collapse
|
7
|
Maddalon A, Pierzchalski A, Krause JL, Bauer M, Finckh S, Brack W, Zenclussen AC, Marinovich M, Corsini E, Krauss M, Herberth G. Impact of chemical mixtures from wastewater treatment plant effluents on human immune cell activation: An effect-based analysis. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 906:167495. [PMID: 37804965 DOI: 10.1016/j.scitotenv.2023.167495] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 09/27/2023] [Accepted: 09/28/2023] [Indexed: 10/09/2023]
Abstract
BACKGROUND Humans are exposed to many different chemicals on a daily basis, mostly as chemical mixtures, usually from food, consumer products and the environment. Wastewater treatment plant effluent contains mixtures of chemicals that have been discarded or excreted by humans and not removed by water treatment. These effluents contribute directly to water pollution, they are used in agriculture and may affect human health. The possible effect of such chemical mixtures on the immune system has not been characterized. OBJECTIVE The aim of this study was to investigate the effect of extracts obtained from four European wastewater treatment plant effluents on human primary immune cell activation. METHODS Immune cells were exposed to the effluent extracts and modulation of cell activation was performed by multi-parameter flow cytometry. Messenger-RNA (mRNA) expression of genes related to immune system and hormone receptors was measured by RT-PCR. RESULTS The exposure of immune cells to these extracts, containing 339 detected chemicals, significantly reduced the activation of human lymphocytes, mainly affecting T helper and mucosal-associated invariant T cells. In addition, basophil activation was also altered upon mixture exposure. Concerning mRNA expression, we observed that 12 transcripts were down-regulated by at least one extract while 11 were up-regulated. Correlation analyses between the analyzed immune parameters and the concentration of chemicals in the WWTP extracts, highlighted the most immunomodulatory chemicals. DISCUSSION Our results suggest that the mixture of chemicals present in the effluents of wastewater treatment plants could be considered as immunosuppressive, due to their ability to interfere with the activation of immune cells, a process of utmost importance for the functionality of the immune system. The combined approach of immune effect-based analysis and chemical content analysis used in our study provides a useful tool for investigating the effect of environmental mixtures on the human immune response.
Collapse
Affiliation(s)
- Ambra Maddalon
- Laboratory of Toxicology, Department of Pharmacological and Biomolecular Sciences 'Rodolfo Paoletti', Università degli Studi di Milano, Via Balzaretti 9, 20133 Milan, Italy
| | - Arkadiusz Pierzchalski
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research - UFZ, Leipzig, Germany
| | - Jannike Lea Krause
- Schwiete Laboratory for Microbiota and Inflammation, German Rheumatism Research (DRFZ), Centre-a Leibniz Institute, Berlin, Germany
| | - Mario Bauer
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research - UFZ, Leipzig, Germany
| | - Saskia Finckh
- Department of Effect-Directed Analysis, Helmholtz Centre for Environmental Research - UFZ, Leipzig, Germany
| | - Werner Brack
- Department of Effect-Directed Analysis, Helmholtz Centre for Environmental Research - UFZ, Leipzig, Germany; Department of Evolutionary Ecology and Environmental Toxicology, Faculty of Biological Sciences, Goethe University Frankfurt, Frankfurt/Main, Germany
| | - Ana C Zenclussen
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research - UFZ, Leipzig, Germany; Perinatal Immunology Research Group, Medical Faculty, Saxonian Incubator for Clinical Translation (SIKT), University of Leipzig, Germany
| | - Marina Marinovich
- Laboratory of Toxicology, Department of Pharmacological and Biomolecular Sciences 'Rodolfo Paoletti', Università degli Studi di Milano, Via Balzaretti 9, 20133 Milan, Italy
| | - Emanuela Corsini
- Laboratory of Toxicology, Department of Pharmacological and Biomolecular Sciences 'Rodolfo Paoletti', Università degli Studi di Milano, Via Balzaretti 9, 20133 Milan, Italy
| | - Martin Krauss
- Department of Effect-Directed Analysis, Helmholtz Centre for Environmental Research - UFZ, Leipzig, Germany
| | - Gunda Herberth
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research - UFZ, Leipzig, Germany.
| |
Collapse
|
8
|
Kumar V, Stewart JH. Immune Homeostasis: A Novel Example of Teamwork. Methods Mol Biol 2024; 2782:1-24. [PMID: 38622389 DOI: 10.1007/978-1-0716-3754-8_1] [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] [Indexed: 04/17/2024]
Abstract
All living organisms must maintain homeostasis to survive, reproduce, and pass their traits on to the next generation. If homeostasis is not maintained, it can result in various diseases and ultimately lead to death. Physiologists have coined the term "homeostasis" to describe this process. With the emergence of immunology as a separate branch of medicine, the concept of immune homeostasis has been introduced. Maintaining immune homeostasis is crucial to support overall homeostasis through different immunological and non-immunological routes. Any changes in the immune system can lead to chronic inflammatory or autoimmune diseases, immunodeficiency diseases, frequent infections, and cancers. Ongoing scientific advances are exploring new avenues in immunology and immune homeostasis maintenance. This chapter introduces the concept of immune homeostasis and its maintenance through different mechanisms.
Collapse
Affiliation(s)
- Vijay Kumar
- Department of Surgery, Laboratory of Tumor Immunology and Immunotherapy, Medical Education Building-C, Morehouse School of Medicine, Atlanta, GA, USA
| | - John H Stewart
- Department of Surgery, Laboratory of Tumor Immunology and Immunotherapy, Medical Education Building-C, Morehouse School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
9
|
Danil K, Colegrove KM, Delaney MA, Mena A, Stedman N, Wurster E. Systemic Erysipelas Outbreak among Free-Ranging Bottlenose Dolphins, San Diego, California, USA, 2022. Emerg Infect Dis 2023; 29:2561-2563. [PMID: 37987589 PMCID: PMC10683814 DOI: 10.3201/eid2912.230811] [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] [Indexed: 11/22/2023] Open
Abstract
We diagnosed fatal Erysipelothrix rhusiopathiae sepsis in 3 stranded bottlenose dolphins (Tursiops truncatus) during summer 2022, in San Diego, California, USA. The previously undetected disease in this relatively small, regional population of dolphins most likely indicates an environmental or biological change in the coastal ocean or organisms.
Collapse
|
10
|
Devalloir Q, Fritsch C, Alchammas Y, Raoul F, Driget V, Amiot C, Ozaki S, van den Brink N, Scheifler R. Environmental pollution and nutritional quality modulate immune response of the wood mouse (Apodemus sylvaticus) through hormonal disturbances. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 337:122100. [PMID: 37392867 DOI: 10.1016/j.envpol.2023.122100] [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: 02/06/2023] [Revised: 05/16/2023] [Accepted: 06/22/2023] [Indexed: 07/03/2023]
Abstract
Cadmium (Cd) and lead (Pb) are known to enhance immune cell damages and to decrease cellular immunity, promoting higher susceptibility to infectious diseases. Selenium (Se) is an essential element involved in immunity and reactive oxygen species scavenging. This study aimed at evaluating how Cd and Pb and low nutritional (Se) quality modulate immune response to a bacterial lipopolysaccharide (LPS) challenge in wood mice (Apodemus sylvaticus). Mice were trapped near a former smelter in northern France in sites of High or Low contamination. Individuals were challenged immediately after capture or after five days of captivity, fed a standard or a Se-deficient diet. Immune response was measured with leukocyte count and plasma concentration of TNF-α, a pro-inflammatory cytokine. Faecal and plasma corticosterone (CORT), a stress-hormone involved in anti-inflammatory processes, was measured to assess potential endocrine mechanisms. Higher hepatic Se and lower faecal CORT were measured in free-ranging wood mice from High site. LPS-challenged individuals from High site showed steeper decrease of circulating leukocytes of all types, higher TNF-α concentrations, and a significant increase of CORT, compared to individuals from Low site. Challenged captive animals fed standard food exhibited similar patterns (decrease of leukocytes, increase of CORT, and detectable levels of TNF-α), with individuals from lowly contaminated site having higher immune responses than their counterparts from highly polluted site. Animals fed Se-deficient food exhibited lymphocytes decrease, no CORT variation, and average levels of TNF-α. These results suggest (i) a higher inflammatory response to immune challenge in free-ranging animals highly exposed to Cd and Pb, (ii) a faster recovery of inflammatory response in animals lowly exposed to pollution when fed standard food than more exposed individuals, and (iii) a functional role of Se in the inflammatory response. The role of Se and mechanisms underlying the relationship between glucocorticoid and cytokine remain to be elucidated.
Collapse
Affiliation(s)
- Quentin Devalloir
- UMR 6249 Chrono-environnement, CNRS / Université Bourgogne Franche-Comté, Université de Franche-Comté, 16 Route de Gray, 25030, Besançon Cedex, France.
| | - Clémentine Fritsch
- UMR 6249 Chrono-environnement, CNRS / Université Bourgogne Franche-Comté, Université de Franche-Comté, 16 Route de Gray, 25030, Besançon Cedex, France
| | - Yara Alchammas
- UMR 6249 Chrono-environnement, CNRS / Université Bourgogne Franche-Comté, Université de Franche-Comté, 16 Route de Gray, 25030, Besançon Cedex, France
| | - Francis Raoul
- UMR 6249 Chrono-environnement, CNRS / Université Bourgogne Franche-Comté, Université de Franche-Comté, 16 Route de Gray, 25030, Besançon Cedex, France
| | - Vincent Driget
- UMR 6249 Chrono-environnement, CNRS / Université Bourgogne Franche-Comté, Université de Franche-Comté, 16 Route de Gray, 25030, Besançon Cedex, France
| | - Caroline Amiot
- UMR 6249 Chrono-environnement, CNRS / Université Bourgogne Franche-Comté, Université de Franche-Comté, 16 Route de Gray, 25030, Besançon Cedex, France
| | - Shinji Ozaki
- UMR 6249 Chrono-environnement, CNRS / Université Bourgogne Franche-Comté, Université de Franche-Comté, 16 Route de Gray, 25030, Besançon Cedex, France; UK Centre for Ecology & Hydrology, Lancaster Environment Centre, Library Avenue, Bailrigg, Lancaster, LA1 4AP, UK
| | - Nico van den Brink
- Division of Toxicology, Wageningen University, 6700 EA, Wageningen, the Netherlands
| | - Renaud Scheifler
- UMR 6249 Chrono-environnement, CNRS / Université Bourgogne Franche-Comté, Université de Franche-Comté, 16 Route de Gray, 25030, Besançon Cedex, France
| |
Collapse
|
11
|
Lindsay CV, Potter JA, Grimshaw AA, Abrahams VM, Tong M. Endometrial responses to bacterial and viral infection: a scoping review. Hum Reprod Update 2023; 29:675-693. [PMID: 37290428 PMCID: PMC10477945 DOI: 10.1093/humupd/dmad013] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 04/07/2023] [Indexed: 06/10/2023] Open
Abstract
BACKGROUND The endometrium is a highly dynamic tissue that undergoes dramatic proliferation and differentiation monthly in order to prepare the uterus for implantation and pregnancy. Intrauterine infection and inflammation are being increasingly recognized as potential causes of implantation failure and miscarriage, as well as obstetric complications later in gestation. However, the mechanisms by which the cells of the endometrium respond to infection remain understudied and recent progress is slowed in part owing to similar overlapping studies being performed in different species. OBJECTIVE AND RATIONALE The aim of this scoping review is to systematically summarize all published studies in humans and laboratory animals that have investigated the innate immune sensing and response of the endometrium to bacteria and viruses, and the signaling mechanisms involved. This will enable gaps in our knowledge to be identified to inform future studies. SEARCH METHODS The Cochrane Library, Ovid Embase/Medline, PubMed, Scopus, Google Scholar, and Web of Science databases were searched using a combination of controlled and free text terms for uterus/endometrium, infections, and fertility to March 2022. All primary research papers that have reported on endometrial responses to bacterial and viral infections in the context of reproduction were included. To focus the scope of the current review, studies in domesticated animals, included bovine, porcine, caprine, feline, and canine species were excluded. OUTCOMES This search identified 42 728 studies for screening and 766 full-text studies were assessed for eligibility. Data was extracted from 76 studies. The majority of studies focused on endometrial responses to Escherichia coli and Chlamydia trachomatis, with some studies of Neisseria gonorrhea, Staphylococcus aureus, and the Streptococcus family. Endometrial responses have only been studied in response to three groups of viruses thus far: HIV, Zika virus, and the herpesvirus family. For most infections, both cellular and animal models have been utilized in vitro and in vivo, focusing on endometrial production of cytokines, chemokines, and antiviral/antimicrobial factors, and the expression of innate immune signaling pathway mediators after infection. This review has identified gaps for future research in the field as well as highlighted some recent developments in organoid systems and immune cell co-cultures that offer new avenues for studying endometrial responses to infection in more physiologically relevant models that could accelerate future findings in this area. WIDER IMPLICATIONS This scoping review provides an overarching summary and benchmark of the current state of research on endometrial innate immune responses to bacterial and viral infection. This review also highlights some exciting recent developments that enable future studies to be designed to deepen our understanding of the mechanisms utilized by the endometrium to respond to infection and their downstream effects on uterine function.
Collapse
Affiliation(s)
- Christina V Lindsay
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Julie A Potter
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Alyssa A Grimshaw
- Harvey Cushing/John Hay Whitney Medical Library, Yale School of Medicine, New Haven, CT, USA
| | - Vikki M Abrahams
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Mancy Tong
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
12
|
Zheng Z, Liu S, Lin Z, Aweya JJ, Zheng Z, Zhao Y, Chen X, Li S, Zhang Y. Kruppel homolog 1 modulates ROS production and antimicrobial peptides expression in shrimp hemocytes during infection by the Vibrio parahaemolyticus strain that causes AHPND. Front Immunol 2023; 14:1246181. [PMID: 37711612 PMCID: PMC10497957 DOI: 10.3389/fimmu.2023.1246181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 08/09/2023] [Indexed: 09/16/2023] Open
Abstract
Shrimp aquaculture has been seriously affected by acute hepatopancreatic necrosis disease (AHPND), caused by a strain of Vibrio parahaemolyticus that carries the Pir toxin plasmids (V. parahaemolyticus (AHPND)). In this study, the transcription factor, Kruppel homolog 1-like of Peneaus vannamei (PvKr-h1), was significantly induced in shrimp hemocytes after V. parahaemolyticus (AHPND) challenge, suggesting that PvKr-h1 is involved in shrimp immune response. Knockdown of PvKr-h1 followed by V. parahaemolyticus (AHPND) challenge increased bacterial abundance in shrimp hemolymph coupled with high shrimp mortality. Moreover, transcriptome and immunofluorescence analyses revealed that PvKr-h1 silencing followed by V. parahaemolyticus (AHPND) challenge dysregulated the expression of several antioxidant-related enzyme genes, such as Cu-Zu SOD, GPX, and GST, and antimicrobial peptide genes, i.e., CRUs and PENs, and reduced ROS activity and nuclear translocation of Relish. These data reveal that PvKr-h1 regulates shrimps' immune response to V. parahaemolyticus (AHPND) infection by suppressing antioxidant-related enzymes, enhancing ROS production and promoting nuclei import of PvRelish to stimulate antimicrobial peptide genes expression.
Collapse
Affiliation(s)
- Zhou Zheng
- Institute of Marine Sciences and Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou, China
- Department of Medical Laboratory and Department of Reproductive Medicine, Luohu Clinical College of Shantou University Medical College, Shantou University, Shantou, China
| | - Shangjie Liu
- Institute of Marine Sciences and Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou, China
- Department of Medical Laboratory and Department of Reproductive Medicine, Luohu Clinical College of Shantou University Medical College, Shantou University, Shantou, China
| | - Zhongyang Lin
- Institute of Marine Sciences and Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou, China
| | - Jude Juventus Aweya
- Institute of Marine Sciences and Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou, China
- College of Ocean Food and Biological Engineering, Fujian Provincial Key Laboratory of Food Microbiology and Enzyme Engineering, Jimei University, Xiamen, Fujian, China
| | - Zhihong Zheng
- Institute of Marine Sciences and Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou, China
| | - Yongzhen Zhao
- Guangxi Academy of Fishery Sciences, Guangxi Key Laboratory of Aquatic Genetic Breeding and Healthy Aquaculture, Nanning, China
| | - Xiuli Chen
- Guangxi Academy of Fishery Sciences, Guangxi Key Laboratory of Aquatic Genetic Breeding and Healthy Aquaculture, Nanning, China
| | - Shengkang Li
- Institute of Marine Sciences and Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou, China
| | - Yueling Zhang
- Institute of Marine Sciences and Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou, China
| |
Collapse
|
13
|
Parker J. Pathophysiological Effects of Contemporary Lifestyle on Evolutionary-Conserved Survival Mechanisms in Polycystic Ovary Syndrome. Life (Basel) 2023; 13:life13041056. [PMID: 37109585 PMCID: PMC10145572 DOI: 10.3390/life13041056] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 04/15/2023] [Accepted: 04/18/2023] [Indexed: 04/29/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) is increasingly being characterized as an evolutionary mismatch disorder that presents with a complex mixture of metabolic and endocrine symptoms. The Evolutionary Model proposes that PCOS arises from a collection of inherited polymorphisms that have been consistently demonstrated in a variety of ethnic groups and races. In utero developmental programming of susceptible genomic variants are thought to predispose the offspring to develop PCOS. Postnatal exposure to lifestyle and environmental risk factors results in epigenetic activation of developmentally programmed genes and disturbance of the hallmarks of health. The resulting pathophysiological changes represent the consequences of poor-quality diet, sedentary behaviour, endocrine disrupting chemicals, stress, circadian disruption, and other lifestyle factors. Emerging evidence suggests that lifestyle-induced gastrointestinal dysbiosis plays a central role in the pathogenesis of PCOS. Lifestyle and environmental exposures initiate changes that result in disturbance of the gastrointestinal microbiome (dysbiosis), immune dysregulation (chronic inflammation), altered metabolism (insulin resistance), endocrine and reproductive imbalance (hyperandrogenism), and central nervous system dysfunction (neuroendocrine and autonomic nervous system). PCOS can be a progressive metabolic condition that leads to obesity, gestational diabetes, type two diabetes, metabolic-associated fatty liver disease, metabolic syndrome, cardiovascular disease, and cancer. This review explores the mechanisms that underpin the evolutionary mismatch between ancient survival pathways and contemporary lifestyle factors involved in the pathogenesis and pathophysiology of PCOS.
Collapse
Affiliation(s)
- Jim Parker
- School of Medicine, University of Wollongong, Wollongong, NSW 2522, Australia
| |
Collapse
|
14
|
Nutritional feed additives reduce the adverse effects of transport stress in the immune system of Tambaqui (Colossoma macropomum). FISH AND SHELLFISH IMMUNOLOGY REPORTS 2022; 3:100051. [DOI: 10.1016/j.fsirep.2022.100051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 01/05/2022] [Accepted: 01/05/2022] [Indexed: 11/18/2022] Open
|
15
|
Thomas N, Gurvich C, Huang K, Gooley PR, Armstrong CW. The underlying sex differences in neuroendocrine adaptations relevant to Myalgic Encephalomyelitis Chronic Fatigue Syndrome. Front Neuroendocrinol 2022; 66:100995. [PMID: 35421511 DOI: 10.1016/j.yfrne.2022.100995] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 02/27/2022] [Accepted: 03/29/2022] [Indexed: 11/16/2022]
Abstract
INTRODUCTION Myalgic Encephalomyelitis/ Chronic Fatigue Syndrome (ME/CFS) is a complex multisystem disease characterised by severe and disabling new-onset symptoms of post-exertional malaise (PEM), fatigue, brain fog, and sleep dysfunction that lasts for at least six months. Accumulating evidence suggests that sex and endocrine events have a significant influence on symptom onset and moderation of ME/CFS, with female sex being one of the most consistent and credible predictive risk factors associated with diagnosis. Such sex differences suggest sex chromosomes and sex steroids may play a part in the development of the condition or moderation of symptoms, although this has yet to be explored in detail. METHODS/AIMS This narrative review outlines sex differences in ME/CFS in terms of vulnerability factors and clinical phenotype and explores the known sex differences in neuroendocrine systems affected in ME/CFS and how this may relate to disease risk, onset, pathophysiology, and potential treatment avenues. CONCLUSIONS There is clear evidence of a sex dimorphism with regards to prevalence (3:1 female preponderance), clinical phenotypes, and aetiological triggers prior to symptom onset of ME/CFS. Endocrinological events, particularly those throughout the female lifespan, are associated with ME/CFS and include reproductive menstrual cycle fluctuations, pregnancy, post-partum and perimenopause. Further, there is evidence for gonadal sex, adrenal stress and renal neuroendocrine systems as implicated in ME/CFS, including changes in estrogen, progesterone compounds, aldosterone, and cortisol levels, of which there are established sex differences. The broad effects of steroid hormones on the physiological systems may also speak to the diversity of ME/CFS symptomatology observed in patients. Further attention must be paid to sex, age, and steroid biology in ME/CFS.
Collapse
Affiliation(s)
- Natalie Thomas
- Department of Biochemistry & Pharmacology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Australia.
| | - Caroline Gurvich
- Department of Psychiatry, Faculty of Medicine, Nursing and Health Sciences, Monash University, Australia
| | - Katherine Huang
- Department of Biochemistry & Pharmacology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Australia
| | - Paul R Gooley
- Department of Biochemistry & Pharmacology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Australia
| | - Christopher W Armstrong
- Department of Biochemistry & Pharmacology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Australia
| |
Collapse
|
16
|
Kavazović I, Krapić M, Beumer-Chuwonpad A, Polić B, Turk Wensveen T, Lemmermann NA, van Gisbergen KPJM, Wensveen FM. Hyperglycemia and Not Hyperinsulinemia Mediates Diabetes-Induced Memory CD8 T-Cell Dysfunction. Diabetes 2022; 71:706-721. [PMID: 35044446 DOI: 10.2337/db21-0209] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 01/06/2022] [Indexed: 11/13/2022]
Abstract
Type 2 diabetes (T2D) causes an increased risk of morbidity and mortality in response to viral infection. T2D is characterized by hyperglycemia and is typically associated with insulin resistance and compensatory hyperinsulinemia. CD8 T cells express the insulin receptor, and previously, we have shown that insulin is able to directly modulate effector CD8 T-cell function. We therefore hypothesized that memory CD8 T-cell responsiveness in the context of T2D is negatively impacted by hyperinsulinemia or hyperglycemia. Using a mouse model for T2D, we could show that memory CD8 T-cell function was significantly reduced in response to rechallenge by viral infection or with melanoma cells. Basal insulin injection of mice increased GLUT-1 expression and glucose uptake in memory CD8 T-cell precursors early after infection, which was prevented when these cells were deficient for the insulin receptor. However, neither insulin injection nor insulin receptor deficiency resulted in a difference in metabolism, memory formation, cytokine production, or recall responses of memory CD8 T cells compared with controls. Importantly, in context of obesity, insulin receptor deficiency on CD8 T cells did not affect the functional capacity of memory CD8 T cells. In contrast, we could show in vitro and in vivo that hyperglycemia significantly impairs the antiviral capacity of memory CD8 T cells. Our findings indicate that obesity impairs the memory CD8 T-cell response against viral infection and cancer through the detrimental effects of hyperglycemia rather than hyperinsulinemia.
Collapse
Affiliation(s)
- Inga Kavazović
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Mia Krapić
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Ammarina Beumer-Chuwonpad
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, the Netherlands
| | - Bojan Polić
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Tamara Turk Wensveen
- Center for Diabetes, Endocrinology and Cardiometabolism, Thalassotherapia, Opatija, Croatia
- Department of Internal Medicine, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Niels A Lemmermann
- Institute for Virology and Research Center for Immunotherapy (FZI) at the University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Klaas P J M van Gisbergen
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, the Netherlands
| | - Felix M Wensveen
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| |
Collapse
|
17
|
Liu M, Yang X, Zeng C, Zhao H, Li J, Hou Z, Wen H. Transcriptional Signatures of Immune, Neural, and Endocrine Functions in the Brain and Kidney of Rainbow Trout (Oncorhynchus mykiss) in Response to Aeromonas salmonicida Infection. Int J Mol Sci 2022; 23:ijms23031340. [PMID: 35163263 PMCID: PMC8835788 DOI: 10.3390/ijms23031340] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/21/2022] [Accepted: 01/21/2022] [Indexed: 02/01/2023] Open
Abstract
Rainbow trout (Oncorhynchus mykiss) serves as one of the most important commercial fish with an annual production of around 800,000 tonnes. However, infectious diseases, such as furunculosis caused by Aeromonas salmonicida infection, results in great economic loss in trout culture. The brain and kidney are two important organs associated with “sickness behaviors” and immunomodulation in response to disease. Therefore, we worked with 60 trout and investigated transcriptional responses and enrichment pathways between healthy and infected trout. We observed that furunculosis resulted in the activation of toll-like receptors with neuroinflammation and neural dysfunction in the brain, which might cause the “sickness behaviors” of infected trout including anorexia and lethargy. We also showed the salmonid-specific whole genome duplication contributed to duplicated colony stimulating factor 1 (csf-1) paralogs, which play an important role in modulating brain immunomodulation. Enrichment analyses of kidneys showed up-regulated immunomodulation and down-regulated neural functions, suggesting an immune-neural interaction between the brain and kidney. Moreover, the kidney endocrine network was activated in response to A. salmonicida infection, further convincing the communications between endocrine and immune systems in regulating internal homeostasis. Our study provided a foundation for pathophysiological responses of the brain and kidney in response to furunculosis and potentially offered a reference for generating disease-resistant trout strains.
Collapse
Affiliation(s)
| | | | | | | | | | - Zhishuai Hou
- Correspondence: (Z.H.); (H.W.); Tel.: +86-133-4524-7715 (Z.H.); +86-532-8203-1825 (H.W.)
| | - Haishen Wen
- Correspondence: (Z.H.); (H.W.); Tel.: +86-133-4524-7715 (Z.H.); +86-532-8203-1825 (H.W.)
| |
Collapse
|
18
|
Krapić M, Kavazović I, Wensveen FM. Immunological Mechanisms of Sickness Behavior in Viral Infection. Viruses 2021; 13:v13112245. [PMID: 34835051 PMCID: PMC8624889 DOI: 10.3390/v13112245] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 11/04/2021] [Indexed: 12/11/2022] Open
Abstract
Sickness behavior is the common denominator for a plethora of changes in normal behavioral routines and systemic metabolism during an infection. Typical symptoms include temperature, muscle weakness, and loss of appetite. Whereas we experience these changes as a pathology, in fact they are a carefully orchestrated response mediated by the immune system. Its purpose is to optimize immune cell functionality against pathogens whilst minimizing viral replication in infected cells. Sickness behavior is controlled at several levels, most notably by the central nervous system, but also by other organs that mediate systemic homeostasis, such as the liver and adipose tissue. Nevertheless, the changes mediated by these organs are ultimately initiated by immune cells, usually through local or systemic secretion of cytokines. The nature of infection determines which cytokine profile is induced by immune cells and therefore which sickness behavior ensues. In context of infection, sickness behavior is typically beneficial. However, inappropriate activation of the immune system may induce adverse aspects of sickness behavior. For example, tissue stress caused by obesity may result in chronic activation of the immune system, leading to lasting changes in systemic metabolism. Concurrently, metabolic disease prevents induction of appropriate sickness behavior following viral infection, thus impairing the normal immune response. In this article, we will revisit recent literature that elucidates both the benefits and the negative aspects of sickness behavior in context of viral infection.
Collapse
|
19
|
Abstract
The immune and endocrine systems collectively control homeostasis in the body. The endocrine system ensures that values of essential factors and nutrients such as glucose, electrolytes and vitamins are maintained within threshold values. The immune system resolves local disruptions in tissue homeostasis, caused by pathogens or malfunctioning cells. The immediate goals of these two systems do not always align. The immune system benefits from optimal access to nutrients for itself and restriction of nutrient availability to all other organs to limit pathogen replication. The endocrine system aims to ensure optimal nutrient access for all organs, limited only by the nutrients stores that the body has available. The actual state of homeostatic parameters such as blood glucose levels represents a careful balance based on regulatory signals from the immune and endocrine systems. This state is not static but continuously adjusted in response to changes in the current metabolic needs of the body, the amount of resources it has available and the level of threats it encounters. This balance is maintained by the ability of the immune and endocrine systems to interact and co-regulate systemic metabolism. In context of metabolic disease, this system is disrupted, which impairs functionality of both systems. The failure of the endocrine system to retain levels of nutrients such as glucose within threshold values impairs functionality of the immune system. In addition, metabolic stress of organs in context of obesity is perceived by the immune system as a disruption in local homeostasis, which it tries to resolve by the excretion of factors which further disrupt normal metabolic control. In this chapter, we will discuss how the immune and endocrine systems interact under homeostatic conditions and during infection with a focus on blood glucose regulation. In addition, we will discuss how this system fails in the context of metabolic disease.
Collapse
|
20
|
Gvozdeva OV, Achasova KM, Litvinova NA, Kozhevnikova EN, Litvinova EA. Female Scent Activated Expression of Arginase1 and Inducible NO-Synthetase in Lung of BALB/c Male Mice. Animals (Basel) 2021; 11:1756. [PMID: 34208376 PMCID: PMC8231181 DOI: 10.3390/ani11061756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 06/09/2021] [Accepted: 06/10/2021] [Indexed: 11/16/2022] Open
Abstract
Scent signals play an important role in the life of rodents. The scent of the opposite sex can modulate immunity. In mice populations with natural specific pathogens, in males, the scent of a female leads to a redistribution of leukocytes between the lung and the blood, resistance to the influenza virus, and a decrease in antibody production, but not in the development of inflammation induced by bacterial endotoxins. This study demonstrates the effect of the scent of soiled bedding of specific pathogen-free (SPF) status female mice on the percentage of different types of leukocytes in the blood, the expression of Nos2, Arg1, and Foxp3 genes, and the presence of M1/M2 macrophages in the lungs of male BALB/c mice. The scent of the female SPF mice caused a redistribution between T- and B-cells in the blood, the increase in the expression of Nos2, Arg1 genes, and the percentage of M1 type macrophages in the lung, but did not affect the different types of T-cells in the periphery or the lungs. Activation of macrophages in the lung is part of mucosal immunity, which is necessary for males as an adaptive mechanism to prevent potential infection during the search for a sexual partner.
Collapse
Affiliation(s)
- Olga V. Gvozdeva
- Siberian Federal Scientific Centre of Agro-BioTechnologies of the Russian Academy of Sciences, P.O. Box 267, 630501 Krasnoobsk, Russia; (O.V.G.); (K.M.A.); (E.N.K.)
| | - Kseniya M. Achasova
- Siberian Federal Scientific Centre of Agro-BioTechnologies of the Russian Academy of Sciences, P.O. Box 267, 630501 Krasnoobsk, Russia; (O.V.G.); (K.M.A.); (E.N.K.)
| | - Nadezhda A. Litvinova
- Faculty of General Medicine, Kemerovo State Medical University, 650001 Kemerovo, Russia;
| | - Elena N. Kozhevnikova
- Siberian Federal Scientific Centre of Agro-BioTechnologies of the Russian Academy of Sciences, P.O. Box 267, 630501 Krasnoobsk, Russia; (O.V.G.); (K.M.A.); (E.N.K.)
- Scientific Research Institute of Neurosciences and Medicine, 630117 Novosibirsk, Russia
| | - Ekaterina A. Litvinova
- Siberian Federal Scientific Centre of Agro-BioTechnologies of the Russian Academy of Sciences, P.O. Box 267, 630501 Krasnoobsk, Russia; (O.V.G.); (K.M.A.); (E.N.K.)
- Scientific Research Institute of Neurosciences and Medicine, 630117 Novosibirsk, Russia
| |
Collapse
|
21
|
Turk Wensveen T, Gašparini D, Rahelić D, Wensveen FM. Type 2 diabetes and viral infection; cause and effect of disease. Diabetes Res Clin Pract 2021; 172:108637. [PMID: 33352263 PMCID: PMC8050380 DOI: 10.1016/j.diabres.2020.108637] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 11/27/2020] [Accepted: 12/16/2020] [Indexed: 02/06/2023]
Abstract
The recent pandemic of COVID-19 has made abundantly clear that Type 2 diabetes (T2D) increases the risk of more frequent and more severe viral infections. At the same time, pro-inflammatory cytokines of an anti-viral Type-I profile promote insulin resistance and form a risk factor for development of T2D. What this illustrates is that there is a reciprocal, detrimental interaction between the immune and endocrine system in the context of T2D. Why these two systems would interact at all long remained unclear. Recent findings indicate that transient changes in systemic metabolism are induced by the immune system as a strategy against viral infection. In people with T2D, this system fails, thereby negatively impacting the antiviral immune response. In addition, immune-mediated changes in systemic metabolism upon infection may aggravate glycemic control in T2D. In this review, we will discuss recent literature that sheds more light on how T2D impairs immune responses to viral infection and how virus-induced activation of the immune system increases risk of development of T2D.
Collapse
Affiliation(s)
- Tamara Turk Wensveen
- Center for Diabetes, Endocrinology and Cardiometabolism, Thallassotherapia, Opatija, Croatia; Department of Endocrinology, Diabetes and Metabolic Disorders, Clinical Hospital Centre, Rijeka, Croatia; Department of Internal Medicine, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Dora Gašparini
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Dario Rahelić
- Vuk Vrhovac University Clinic for Diabetes, Endocrinology and Metabolic Diseases, Merkur University Hospital, Zagreb, Croatia; School of Medicine, University of Zagreb, Zagreb, Croatia; School of Medicine, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia
| | - Felix M Wensveen
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia.
| |
Collapse
|
22
|
Klein JR. Dynamic Interactions Between the Immune System and the Neuroendocrine System in Health and Disease. Front Endocrinol (Lausanne) 2021; 12:655982. [PMID: 33828532 PMCID: PMC8020567 DOI: 10.3389/fendo.2021.655982] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 03/05/2021] [Indexed: 01/21/2023] Open
Abstract
The immune system and the neuroendocrine system share many common features. Both consist of diverse components consisting of receptors and networks that are widely distributed throughout the body, and both sense and react to external stimuli which, on the one hand control mechanisms of immunity, and on the other hand control and regulate growth, development, and metabolism. It is thus not surprising, therefore, that the immune system and the neuroendocrine system communicate extensively. This article will focus on bi-directional immune-endocrine interactions with particular emphasis on the hormones of the hypothalamus-pituitary-thyroid (HPT) axis. New findings will be discussed demonstrating the direct process through which the immune system-derived thyroid stimulating hormone (TSH) controls thyroid hormone synthesis and bone metamorphosis, particularly in the context of a novel splice variant of TSHβ made by peripheral blood leukocytes (PBL). Also presented are the ways whereby the TSHβ splice variant may be a contributing factor in the development and/or perpetuation of autoimmune thyroid disease (AIT), and how systemic infection may elicit immune-endocrine responses. The relationship between non-HPT hormones, in particular adipose hormones, and immunity is discussed.
Collapse
|
23
|
Mukherjee S, Banerjee O, Singh S, Maji BK. COVID 19 could trigger global diabetes burden - A hypothesis. Diabetes Metab Syndr 2020; 14:963-964. [PMID: 32604015 PMCID: PMC7319643 DOI: 10.1016/j.dsx.2020.06.049] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 06/21/2020] [Accepted: 06/24/2020] [Indexed: 01/19/2023]
Affiliation(s)
- Sandip Mukherjee
- Department of Physiology, Serampore College, 9 William Carey Road, Serampore, Hooghly, 712201, India.
| | - Oly Banerjee
- Department of Physiology, Serampore College, 9 William Carey Road, Serampore, Hooghly, 712201, India
| | - Siddhartha Singh
- Department of Physiology, Serampore College, 9 William Carey Road, Serampore, Hooghly, 712201, India
| | - Bithin Kumar Maji
- Department of Physiology, Serampore College, 9 William Carey Road, Serampore, Hooghly, 712201, India
| |
Collapse
|
24
|
Galbadage T, Peterson BM, Awada J, Buck AS, Ramirez DA, Wilson J, Gunasekera RS. Systematic Review and Meta-Analysis of Sex-Specific COVID-19 Clinical Outcomes. Front Med (Lausanne) 2020; 7:348. [PMID: 32671082 PMCID: PMC7331754 DOI: 10.3389/fmed.2020.00348] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 06/10/2020] [Indexed: 01/08/2023] Open
Abstract
To successfully mitigate the extraordinary devastation caused by the Coronavirus disease 2019 (COVID-19) pandemic, it is crucial to identify important risk factors for this disease. One such neglected health determinant is the sex of the patient. This is an essential clinical characteristic, as it can factor into a patient's clinical management and preventative measures. Some clinical studies have shown disparities in the proportion between males and females that have more severe clinical outcomes or, subsequently, die from this disease. However, this association has not been unequivocally established. Thus, the purpose of this investigation was to examine the association between male sex and COVID-19 severity. We systematically reviewed the literature, identified studies that matched predetermined selection criteria, and performed a meta-analysis to evaluate the proportion of males among four disease severity categories. Appropriate assessment strategies were implemented to assess and minimize potential biases. The results of this meta-analysis indicated that males constituted a significantly higher proportion of those who had adverse clinical outcomes and died from COVID-19. As the coronavirus spread from the East to the West, male sex remained a consistent risk factor. Our results support the establishment of the male sex as an important risk factor for this disease. Early identification and appropriate medical care for males with lab-confirmed COVID-19 may substantially change the course of clinical prognosis, resulting in greater numbers of lives saved.
Collapse
Affiliation(s)
- Thushara Galbadage
- Department of Kinesiology and Health Science, Biola University, La Mirada, CA, United States
| | - Brent M Peterson
- Department of Kinesiology and Health Science, Biola University, La Mirada, CA, United States
| | - Joseph Awada
- Department of Chemistry, Physics, and Engineering, Biola University, La Mirada, CA, United States
| | - Alison S Buck
- Department of Chemistry, Physics, and Engineering, Biola University, La Mirada, CA, United States
| | - Danny A Ramirez
- Department of Chemistry, Physics, and Engineering, Biola University, La Mirada, CA, United States
| | - Jason Wilson
- Department of Mathematics and Computer Science, Biola University, La Mirada, CA, United States
| | - Richard S Gunasekera
- Department of Chemistry, Physics, and Engineering, Biola University, La Mirada, CA, United States
| |
Collapse
|
25
|
Manipulation of Metabolic Pathways and Its Consequences for Anti-Tumor Immunity: A Clinical Perspective. Int J Mol Sci 2020; 21:ijms21114030. [PMID: 32512898 PMCID: PMC7312891 DOI: 10.3390/ijms21114030] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 05/28/2020] [Accepted: 06/01/2020] [Indexed: 02/06/2023] Open
Abstract
In the relatively short history of anti-tumor treatment, numerous medications have been developed against a variety of targets. Intriguingly, although many anti-tumor strategies have failed in their clinical trials, metformin, an anti-diabetic medication, demonstrated anti-tumor effects in observational studies and even showed its synergistic potential with immune checkpoint inhibitors (ICIs) in subsequent clinical studies. Looking back from bedside-to-bench, it may not be surprising that the anti-tumor effect of metformin derives largely from its ability to rewire aberrant metabolic pathways within the tumor microenvironment. As one of the most promising breakthroughs in oncology, ICIs were also found to exert their immune-stimulatory effects at least partly via rewiring metabolic pathways. These findings underscore the importance of correcting metabolic pathways to achieve sufficient anti-tumor immunity. Herein, we start by introducing the tumor microenvironment, and then we review the implications of metabolic syndrome and treatments for targeting metabolic pathways in anti-tumor therapies. We further summarize the close associations of certain aberrant metabolic pathways with impaired anti-tumor immunity and introduce the therapeutic effects of targeting these routes. Lastly, we go through the metabolic effects of ICIs and conclude an overall direction to manipulate metabolic pathways in favor of anti-tumor responses.
Collapse
|