1
|
Miró L, Rosell-Cardona C, Amat C, Polo J, Moretó M, Pérez-Bosque A. Dietary supplementation with spray-dried animal plasma improves vaccine protection in aged mice. Front Nutr 2023; 10:1050961. [PMID: 37032769 PMCID: PMC10080719 DOI: 10.3389/fnut.2023.1050961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 02/27/2023] [Indexed: 04/11/2023] Open
Abstract
Background Senescence is characterized by an aggravated inflammatory state that reduces vaccine responsiveness. Dietary supplementation with spray-dried porcine plasma (SDP) exerts anti-inflammatory effects in different mucosal areas. We aimed to determine if the anti-inflammatory properties of SDP improve the efficiency of immunization in senescent animals. Methods Experiments were performed in 2-month-old and 6-month-old male SAMP8 mice fed control or SDP (8%) feeds for 4 months. The mice received nasal doses of 2.5 μg of Staphylococcus aureus enterotoxin B (SEB) or vehicle every 15 days (i.e., 3 times). Fifteen days after the last dose, a lethal shock was induced by intraperitoneal administration of SEB and LPS. Results Immunization increased anti-SEB IgA in intestinal and bronchoalveolar fluid (p < 0.05). After the lethal shock, all immunized aged mice that were supplemented with SDP survived, in contrast to only 66% of those fed the control feed (p < 0.05). Moreover, after the lethal challenge, aged mice showed higher expression levels of pro-inflammatory cytokines (Il-6, Tnf-α, Ifn-γ, and Il-1β) in jejunal and (Tnf-α, and Il-1β) in lung tissues (p < 0.05), which were reduced by SDP supplementation (p < 0.05). Furthermore, in senescent mice, SDP supplementation augmented Il-4 and Il-10 expression in both tissues (p < 0.05). Conclusion SDP reduces the mucosal inflammation associated with aging, improving vaccine protection in senescent mice.
Collapse
Affiliation(s)
- Lluïsa Miró
- Departament de Bioquímica i Fisiologia, Facultat de Farmàcia i Ciències de l’Alimentació and Institut de Recerca en Nutrició i Seguretat Alimentària (INSA-UB), Universitat de Barcelona (UB), Barcelona, Spain
| | - Cristina Rosell-Cardona
- Departament de Bioquímica i Fisiologia, Facultat de Farmàcia i Ciències de l’Alimentació and Institut de Recerca en Nutrició i Seguretat Alimentària (INSA-UB), Universitat de Barcelona (UB), Barcelona, Spain
| | - Concepció Amat
- Departament de Bioquímica i Fisiologia, Facultat de Farmàcia i Ciències de l’Alimentació and Institut de Recerca en Nutrició i Seguretat Alimentària (INSA-UB), Universitat de Barcelona (UB), Barcelona, Spain
| | | | - Miquel Moretó
- Departament de Bioquímica i Fisiologia, Facultat de Farmàcia i Ciències de l’Alimentació and Institut de Recerca en Nutrició i Seguretat Alimentària (INSA-UB), Universitat de Barcelona (UB), Barcelona, Spain
| | - Anna Pérez-Bosque
- Departament de Bioquímica i Fisiologia, Facultat de Farmàcia i Ciències de l’Alimentació and Institut de Recerca en Nutrició i Seguretat Alimentària (INSA-UB), Universitat de Barcelona (UB), Barcelona, Spain
| |
Collapse
|
2
|
Santos ACA, Sartori T, Borelli P, Fock RA. Prostaglandin F2α in vitro can affect basic inflammatory parameters of mesenchymal stem cells and slight modulating some of their immunomodulatory properties. Prostaglandins Leukot Essent Fatty Acids 2020; 163:102210. [PMID: 33242781 DOI: 10.1016/j.plefa.2020.102210] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 10/16/2020] [Accepted: 11/07/2020] [Indexed: 01/02/2023]
Abstract
In the last decade, mesenchymal stem cells (MSCs) have been gaining attention due their ability to influence the function of other cells as well as modulate the inflammatory response. This occurs via their immunomodulatory functions, acting through direct cell-cell interaction or by releasing a broad spectrum of bioactive factors such as cytokines and growth factors. In addition, prostaglandins are arachidonic acid metabolites that play a key role in the generation and modulation of the inflammatory response. Among the bioactive prostaglandins, PGF2α is able to stimulate cell proliferation as well as act to inhibit progenitor cell differentiation, but no information about this prostaglandin's action on the immunoregulatory function of MSCs has been reported. In this study we evaluate important aspects of the influence of PGF2α analog (17-phenyl-trinor PGF2α), which is a potent prostaglandin FP receptor agonist, on some mechanisms that control the main functions of MSCs. C3H10T1/2, a mesenchymal stem cell linage, was stimulated with PGF2α under inflammatory conditions trigged by LPS in order to investigate PGF2α inflammatory parameters as well as its ability to immunoregulate macrophages and lymphocytes. PGF2α has the ability to increase proliferation tax without altering the cell viability of LPS-stimulated MSCs, while also diminishing the phosphorylation of NFκB transcription factor leading to attenuation of IL-1β and GM-CSF production. Additionally, MSC-s conditioned media from cells stimulated with PGF2α was able to increase the lymphocytes' IL-10 production. Overall, this study implied that PGF2α are able to modify some properties of MSCs.
Collapse
Affiliation(s)
- Andressa Cristina Antunes Santos
- Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Talita Sartori
- Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Primavera Borelli
- Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Ricardo Ambrosio Fock
- Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
3
|
da Silva Lima F, da Rocha Romero AB, Hastreiter A, Nogueira-Pedro A, Makiyama E, Colli C, Fock RA. An insight into the role of magnesium in the immunomodulatory properties of mesenchymal stem cells. J Nutr Biochem 2018; 55:200-208. [PMID: 29554498 DOI: 10.1016/j.jnutbio.2018.02.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 01/04/2018] [Accepted: 02/05/2018] [Indexed: 12/14/2022]
Abstract
Magnesium (Mg2+) is a mineral with the ability to influence cell proliferation and to modulate inflammatory/immune responses, due to its anti-inflammatory properties. In addition, mesenchymal stem cells (MSCs) modulate the function of all major immune cell populations. Knowing that, the current work aimed to investigate the effects of Mg2+ enrichment, and its influence on the immunomodulatory capacity of MSCs. Murine C3H/10T1/2 MSCs were cultivated in media with different concentrations of Mg2+ (0, 1, 3 and 5 mM), in order to evaluate the effects of Mg2+ on MSC immunomodulatory properties, cell proliferation rates, expression of NFκB and STAT-3, production of IL-1β, IL-6, TGF-β, IL-10, PGE2 and NO, and TRPM7 expression. The results showed that TRPM7 is expressed in MSCs, but Mg2+, in the way that cells were cultivated, did not affect TRPM7 expression. Additionally, there was no difference in the intracellular concentration of Mg2+. Mg2+, especially at 5 mM, raised proliferation rates of MSCs, and modulated immune responses by decreasing levels of IL-1β and IL-6, and by increasing levels of IL-10 and PGE2 in cells stimulated with LPS or TNF-α. In addition, MSCs cultured in 5 mM Mg2+ expressed lower levels of pNFκB/NFκB and higher levels of pSTAT-3/STAT-3. Furthermore, conditioned media from MSCs reduced lymphocyte and macrophage proliferation, but Mg2+ did not affect this parameter. In addition, conditioned media from MSCs cultured at 5 mM of Mg2+ modulated the production profile of cytokines, especially of IL-1β and IL-6 in macrophages. In conclusion, Mg2+ is able to modulate some immunoregulatory properties of MSCs.
Collapse
Affiliation(s)
- Fabiana da Silva Lima
- Department of Food and Experimental Nutrition, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Amanda Batista da Rocha Romero
- Department of Food and Experimental Nutrition, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Araceli Hastreiter
- Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Amanda Nogueira-Pedro
- Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Edson Makiyama
- Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Célia Colli
- Department of Food and Experimental Nutrition, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Ricardo Ambrósio Fock
- Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
4
|
Anderson CF, Gerber JS, Mosser DM. Modulating macrophage function with IgG immune complexes. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/09680519020080060501] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Macrophages respond to bacterial products by releasing a large array of inflammatory mediators. We demonstrate that, in the presence of IgG immune complexes, macrophages produce high levels of IL-10 and virtually no IL-12, when they are exposed to bacterial products. The production of IL-10 by these cells can dampen innate inflammatory responses to microbial products, such as LPS. This alteration in macrophage cytokine production can also influence an adaptive immune response, preferentially inducing Th2-type immunity. Thus, immune complexes change the physiology of activated macrophages, converting them to anti-inflammatory cells that induce Th2-like immune responses. We have termed these cells type II activated macrophages.
Collapse
Affiliation(s)
- Charles F. Anderson
- Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, USA
| | - Jeffrey S. Gerber
- Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, USA
| | - David M. Mosser
- Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, USA,
| |
Collapse
|
5
|
Chapes S, Beharka A. Lipopolysaccharide is required for the lethal effects of enterotoxin B after D-galactosamine sensitization. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/096805199500200406] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
We tested the D-galactosamine sensitization model with staphylococcal enterotoxin B (SEB). LPS was required for the lethal effects of SEB in D-galactosamine sensitized mice. Only two (2/62) among the C3HeB/FeJ (H-2k), Balb/c (H-2d) and C57BL/6J (H-2b) mice died in response to SEB in the absence of LPS whereas injection of SEB and minimally lethal concentrations of LPS became highly toxic. Similar to LPS, the lethal effect of SEB was dependent on the mouse strain used. Mouse strains more sensitive to the effects of LPS (Balb/c and C57BL/6J) were also more sensitive to the effects of SEB in comparison to C3H mice when equivalent doses of LPS and SEB were used. Among Balb/c and C3HeB/FeJ but not C57BL/6J mice, SEB (20 μg) potentiated the lethal effects of LPS at low doses (0.1 μg LPS), but had an apparent protective effect at high doses (1 μg LPS). Lastly, there was an inverse correlation between pathogenesis and serum IL-2 concentrations and splenic T cell activation in C3H mice. However, macrophage mobilization did correlate with lethality. Therefore, some questions remain about the mechanisms involved in the D-galactosamine/SEB pathogenesis model. We conclude that when sensitizing mice with D-galactosamine and assessing the lethal effects of SEB, endotoxin contamination must be assessed.
Collapse
Affiliation(s)
- S.K. Chapes
- Division of Biology, Kansas State University, Manhattan, Kansas, USA
| | - A.A. Beharka
- Division of Biology, Kansas State University, Manhattan, Kansas, USA
| |
Collapse
|
6
|
Jia Y, Zhao J, Liu M, Li B, Song Y, Li Y, Wen A, Shi L. Brazilin exerts protective effects against renal ischemia-reperfusion injury by inhibiting the NF-κB signaling pathway. Int J Mol Med 2016; 38:210-6. [PMID: 27247107 PMCID: PMC4899020 DOI: 10.3892/ijmm.2016.2616] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 05/19/2016] [Indexed: 01/28/2023] Open
Abstract
Renal ischemia-reperfusion (I/R) injury is associated with high morbidity and mortality as there is currently no available effective therapeutic strategy with which to treat this injury. Thus, the aim of this study was to investigate the potential protective effects of brazilin, a major active component of the Chinese medicine Caesalpinia sappan L., against renal I/R injury in vitro and in vivo. Rats were subjected to removal of the right kidney and I/R injury to the left kidney (ischemia for 45 min followed by reperfusion for 24 h). Treatment with brazilin (30 mg/kg, administered intravenously at 30 min prior to ischemia) led to the reversal of I/R-induced changes in serum creatinine (Scr) and blood urea nitrogen (BUN) levels, and also attenuated the histopathological damage induced by I/R. Furthermore, TUNEL assay revealed that brazilin reduced cell necrosis, and significantly decreased the expression of tumor necrosis factor (TNF)-α and interleukin (IL)-1β in renal tissue. Moreover, HK-2 cells were used in order to elucidate the mechanisms responsible for the protective effects of brazilin. The levels of phosphorylated IκBα and the nuclear translocation of nuclear factor-κB (NF-κB) were all evidently decreased by brazilin. These findings suggested that pre-treatment with brazilin protects against I/R-induced renal damage and suppresses the inflammatory response by inhibiting the activation of the NF-κB signaling pathway.
Collapse
Affiliation(s)
- Yanyan Jia
- Department of Pharmacy, General Hospital of Guangzhou Military Command of PLA, Guangzhou, Guangdong 510010, P.R. China
| | - Jinyi Zhao
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Meiyou Liu
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Bingling Li
- Department of Pharmacy, General Hospital of Guangzhou Military Command of PLA, Guangzhou, Guangdong 510010, P.R. China
| | - Ying Song
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Yuwen Li
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Aidong Wen
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Lei Shi
- Department of Pharmacy, General Hospital of Guangzhou Military Command of PLA, Guangzhou, Guangdong 510010, P.R. China
| |
Collapse
|
7
|
Krakauer T, Pradhan K, Stiles BG. Staphylococcal Superantigens Spark Host-Mediated Danger Signals. Front Immunol 2016; 7:23. [PMID: 26870039 PMCID: PMC4735405 DOI: 10.3389/fimmu.2016.00023] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 01/18/2016] [Indexed: 12/19/2022] Open
Abstract
Staphylococcal enterotoxin B (SEB) of Staphylococcus aureus, and related superantigenic toxins produced by myriad microbes, are potent stimulators of the immune system causing a variety of human diseases from transient food poisoning to lethal toxic shock. These protein toxins bind directly to specific Vβ regions of T-cell receptors (TCR) and major histocompatibility complex (MHC) class II on antigen-presenting cells, resulting in hyperactivation of T lymphocytes and monocytes/macrophages. Activated host cells produce excessive amounts of proinflammatory cytokines and chemokines, especially tumor necrosis factor α, interleukin 1 (IL-1), IL-2, interferon γ (IFNγ), and macrophage chemoattractant protein 1 causing clinical symptoms of fever, hypotension, and shock. Because of superantigen-induced T cells skewed toward TH1 helper cells, and the induction of proinflammatory cytokines, superantigens can exacerbate autoimmune diseases. Upon TCR/MHC ligation, pathways induced by superantigens include the mitogen-activated protein kinase cascades and cytokine receptor signaling, resulting in activation of NFκB and the phosphoinositide 3-kinase/mammalian target of rapamycin pathways. Various mouse models exist to study SEB-induced shock including those with potentiating agents, transgenic mice and an “SEB-only” model. However, therapeutics to treat toxic shock remain elusive as host response genes central to pathogenesis of superantigens have only been identified recently. Gene profiling of a murine model for SEB-induced shock reveals novel molecules upregulated in multiple organs not previously associated with SEB-induced responses. The pivotal genes include intracellular DNA/RNA sensors, apoptosis/DNA damage-related molecules, immunoproteasome components, as well as antiviral and IFN-stimulated genes. The host-wide induction of these, and other, antimicrobial defense genes provide evidence that SEB elicits danger signals resulting in multi-organ damage and toxic shock. Ultimately, these discoveries might lead to novel therapeutics for various superantigen-based diseases.
Collapse
Affiliation(s)
- Teresa Krakauer
- Department of Immunology, Molecular Translational Sciences Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick , Frederick, MD , USA
| | - Kisha Pradhan
- Biology Department, Wilson College , Chambersburg, PA , USA
| | | |
Collapse
|
8
|
Immunomodulation and Disease Tolerance to Staphylococcus aureus. Pathogens 2015; 4:793-815. [PMID: 26580658 PMCID: PMC4693165 DOI: 10.3390/pathogens4040793] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 11/10/2015] [Indexed: 12/12/2022] Open
Abstract
The Gram-positive bacterium Staphylococcus aureus is one of the most frequent pathogens that causes severe morbidity and mortality throughout the world. S. aureus can infect skin and soft tissues or become invasive leading to diseases such as pneumonia, endocarditis, sepsis or toxic shock syndrome. In contrast, S. aureus is also a common commensal microbe and is often part of the human nasal microbiome without causing any apparent disease. In this review, we explore the immunomodulation and disease tolerance mechanisms that promote commensalism to S. aureus.
Collapse
|
9
|
Alfelali M, Khandaker G. Infectious causes of sudden infant death syndrome. Paediatr Respir Rev 2014; 15:307-11. [PMID: 25441371 DOI: 10.1016/j.prrv.2014.09.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 09/15/2014] [Indexed: 12/18/2022]
Abstract
Investigators have long suspected the role of infection in sudden infant death syndrome (SIDS). Evidence of infectious associations with SIDS is accentuated through the presence of markers of infection and inflammation on autopsy of SIDS infants and isolates of some bacteria and viruses. Several observational studies have looked into the relation between seasonality and incidence of SIDS, which often showed a winter peak. These all may suggest an infectious aetiology of SIDS. In this review we have summarised the current literature on infectious aetiologies of SIDS by looking at viral, bacterial, genetic and environmental factors which are believed to be associated with SIDS.
Collapse
Affiliation(s)
- Mohammad Alfelali
- National Centre for Immunisation Research and Surveillance (NCIRS), The Children's Hospital at Westmead, Sydney, NSW, Australia; Discipline of Paediatrics and Child Health, Sydney Medical School, University of Sydney, Sydney, NSW, Australia; Department of Family and Community Medicine, Faculty of Medicine in Rabigh, King Abdulaziz University, Saudi Arabia.
| | - Gulam Khandaker
- National Centre for Immunisation Research and Surveillance (NCIRS), The Children's Hospital at Westmead, Sydney, NSW, Australia; Discipline of Paediatrics and Child Health, Sydney Medical School, University of Sydney, Sydney, NSW, Australia; Centre for Perinatal Infection Research, The Children's Hospital at Westmead and The University of Sydney, Sydney, NSW, Australia; Marie Bashir Institute for Infectious Diseases and Biosecurity Institute (MBI), University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
10
|
Cheng YJ, Cheng SM, Teng YH, Shyu WC, Chen HL, Lee SD. Cordyceps sinensisPrevents Apoptosis in Mouse Liver with D-Galactosamine/Lipopolysaccharide-Induced Fulminant Hepatic Failure. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2014; 42:427-41. [DOI: 10.1142/s0192415x14500281] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Cordyceps sinensis (C. sinensis) has long been considered to be an herbal medicine and has been used in the treatment of various inflammatory diseases. The present study examined the cytoprotective properties of C. sinensis on D(+)-galactosamine (GalN)/lipopolysaccharide (LPS)-induced fulminant hepatic failure. Mice were randomly assigned into control, GalN/LPS, CS 20 mg and CS 40 mg groups (C. sinensis, oral gavage, five days/week, four weeks). After receiving saline or C. sinensis, mice were intraperitoneally given GalN (800 mg/kg)/LPS (10 μg/kg). The effects of C. sinensis on TNF-α, IL-10, AST, NO, SOD, and apoptoticrelated proteins after the onset of endotoxin intoxication were determined. Data demonstrated that GalN/LPS increased hepatocyte degeneration, circulating AST, TNF-α, IL-10, and hepatic apoptosis and caspase activity. C. sinensis pre-treatment reduced AST, TNF-α, and NO and increased IL-10 and SOD in GalN/LPS induced fulminant hepatic failure. C. sinensis attenuated the apoptosis of hepatocytes, as evidenced by the TUNEL and capase-3, 6 activity analyses. In summary, C. sinensis alleviates GalN/LPS-induced liver injury by modulating the cytokine response and inhibiting apoptosis. C. sinensis could be used as a potent antioxidant, anti-inflammatory and anti-apoptotic agent to reduce liver injury after the onset of sepsis.
Collapse
Affiliation(s)
- Yu-Jung Cheng
- Department of Physical Therapy and Graduate Institute of Rehabilitation Science, China Medical University, Taichung, Taiwan
| | - Shiu-Min Cheng
- Department of Psychology, Asia University, Taichung, Taiwan
| | - Yi-Hsien Teng
- Department of Physical Therapy and Graduate Institute of Rehabilitation Science, China Medical University, Taichung, Taiwan
| | - Woei-Cherng Shyu
- Center for Neuropsychiatry and Graduate Institute of Immunology, China Medical University and Hospital, Taichung, Taiwan
| | - Hsiu-Ling Chen
- Institute of Occupational Safety and Hazard Prevention, Hung Kuang University, Taichung, Taiwan
| | - Shin-Da Lee
- Department of Physical Therapy and Graduate Institute of Rehabilitation Science, China Medical University, Taichung, Taiwan
- Department of Healthcare Administration, Asia University, Taichung, Taiwan
| |
Collapse
|
11
|
Singh P, Castillo A, Majid DSA. Decrease in IL-10 and increase in TNF-α levels in renal tissues during systemic inhibition of nitric oxide in anesthetized mice. Physiol Rep 2014; 2:e00228. [PMID: 24744897 PMCID: PMC3966239 DOI: 10.1002/phy2.228] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Revised: 01/08/2014] [Accepted: 01/13/2014] [Indexed: 11/07/2022] Open
Abstract
Earlier, we demonstrated that the inhibition of nitric oxide synthase (NOS) by nitro‐l‐arginine methyl ester (l‐NAME) infusion increases the endogenous production of proinflammatory cytokine, tumor necrosis factor (TNF‐α). In the present study, we examined the hypothesis that inhibition of nitric oxide (NO) production leads to the suppression of interleukin (IL)‐10 (anti‐inflammatory cytokine) generation which facilitates the enhancement of TNF‐α production endogenously. Using appropriate enzyme‐linked immunosorbent assay kits and immunohistochemical staining, the levels of IL‐10 and TNF‐α in plasma (P) and in renal tissues (R) were measured in anesthetized mice (C57BL/6; ~10 weeks age; n = 6/group) infused with or without l‐NAME (200 μg/min/kg; i.v. for 2 h). Compared to vehicle‐treated control mice, l‐NAME‐treated mice had a lower level of IL‐10 (P, 0.3 ± 0.1 vs. 2.6 ± 0.6 ng/mL; R, 0.5 ± 0.1 vs. 3 ± 0.1 ng/mg protein) and a higher level of TNF‐α (P, 432 ± 82 vs. undetected pg/mL; R, 58 ± 7 vs. 6 ± 5 pg/mg protein). IL‐10 protein expression, present mostly in the distal nephron segments in control mice, was markedly downregulated in l‐NAME‐treated mice. Compared to control mice, TNF‐α expression increased 2.5‐fold in renal cortical sections (mostly in the distal nephron segments) in l‐NAME‐treated mice. Coinfusion of a NO donor, S‐nitroso‐N‐acetyl‐penicillamine (SNAP; 25 μg/min/kg) with l‐NAME in a separate group of mice prevented these changes in IL‐10 and TNF‐α induced by l‐NAME. IL‐10 infusion (0.075 ng/min/g) in l‐NAME‐treated mice markedly attenuated l‐NAME‐induced increments in TNF‐α. Thus, these results demonstrate that NOS inhibition decreases endogenous IL‐10 generation and thus, minimizes its immune downregulating action on the TNF‐α production in the kidney. In this study in mice, we examined the hypothesis that NOS inhibition reduces the endogenous production of anti‐inflammatory cytokine, IL‐10 which facilitates enhanced production of TNF‐α in the kidney. Using appropriate enzyme‐linked immunosorbent assay kits and immunohistochemical staining, levels of IL‐10 and TNF‐α in renal tissue were measured in anesthetized mice infused with or without l‐NAME (200 μg/min/kg) in the presence or absence of a NO donor, S‐nitroso‐N‐acetyl‐penicillamine (SNAP; 25 μg/min/kg) or IL‐10 infusion (0.075 ng/min/g). These results demonstrate that NOS inhibition decreases endogenous IL‐10 generation and thus, minimizes its immune downregulating action on the TNF‐α production in the kidney.
Collapse
Affiliation(s)
- Purnima Singh
- Department of Physiology, Hypertension & Renal Center of Excellence, Tulane University Health Sciences Center, New Orleans, 70112, Louisiana
| | - Alexander Castillo
- Department of Physiology, Hypertension & Renal Center of Excellence, Tulane University Health Sciences Center, New Orleans, 70112, Louisiana
| | - Dewan S A Majid
- Department of Physiology, Hypertension & Renal Center of Excellence, Tulane University Health Sciences Center, New Orleans, 70112, Louisiana
| |
Collapse
|
12
|
Zaga-Clavellina V, Flores-Espinosa P, Pineda-Torres M, Sosa-González I, Vega-Sánchez R, Estrada-Gutierrez G, Espejel-Núñez A, Flores-Pliego A, Maida-Claros R, Estrada-Juárez H, Chávez-Mendoza A. Tissue-specific IL-10 secretion profile from term human fetal membranes stimulated with pathogenic microorganisms associated with preterm labor in a two-compartment tissue culture system. J Matern Fetal Neonatal Med 2013; 27:1320-7. [PMID: 24138141 DOI: 10.3109/14767058.2013.857397] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Interleukin (IL)-10 is a cytokine with anti-inflammatory properties that plays pivotal roles in immune recognition and maintenance of pregnancy, limiting the harmful effects of pro-inflammatory modulators. The aim of this work was to characterize the contribution of amnion and choriodecidua regions of the human fetal membranes in the production of IL-10 after selective stimulation with Candida albicans, Gardnerella vaginalis and Streptococcus agalactiae. METHODS Pre-labor human fetal membranes were cultured in a two-compartment tissue culture system and stimulated with 1 × 10(6) CFU/ml of each pathogen added to either the amniotic or choriodecidual region or both. RESULTS Candida albicans and G. vaginalis were the pathogens most effective in inducing IL-10 secretion, increasing 20 and 10 times, respectively, the levels of this cytokine in the choriodecidual compartment. Stimulation with S. agalactiae was effective only in the choriodecidual region, increasing two times IL-10 concentration. CONCLUSIONS Synthesis and secretion of IL-10 in response to three different pathogens associated with intrauterine infection and preterm birth are differential and depend on the nature of the microorganism and initial contact region.
Collapse
|
13
|
Krakauer T. Update on staphylococcal superantigen-induced signaling pathways and therapeutic interventions. Toxins (Basel) 2013; 5:1629-54. [PMID: 24064719 PMCID: PMC3798877 DOI: 10.3390/toxins5091629] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Revised: 09/13/2013] [Accepted: 09/13/2013] [Indexed: 12/20/2022] Open
Abstract
Staphylococcal enterotoxin B (SEB) and related bacterial toxins cause diseases in humans and laboratory animals ranging from food poisoning, acute lung injury to toxic shock. These superantigens bind directly to the major histocompatibility complex class II molecules on antigen-presenting cells and specific Vβ regions of T-cell receptors (TCR), resulting in rapid hyper-activation of the host immune system. In addition to TCR and co-stimulatory signals, proinflammatory mediators activate signaling pathways culminating in cell-stress response, activation of NFκB and mammalian target of rapamycin (mTOR). This article presents a concise review of superantigen-activated signaling pathways and focuses on the therapeutic challenges against bacterial superantigens.
Collapse
Affiliation(s)
- Teresa Krakauer
- Department of Immunology, Integrated Toxicology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD 21702 5011, USA.
| |
Collapse
|
14
|
Interleukin 10 antioxidant effect decreases leukocytes/endothelial interaction induced by tumor necrosis factor α. Shock 2013; 39:83-8. [PMID: 23247124 DOI: 10.1097/shk.0b013e318278ae36] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Little is known about the endothelial mechanisms involved in the anti-inflammatory effects of interleukin 10 (IL-10). The goal of this study was to evaluate the effects of IL-10 on endothelial oxidative stress and endothelial inflammation induced by tumor necrosis factor α (TNF-α). Production of reactive oxygen species (ROS) in perfused human umbilical vein endothelial cells (HUVECs) was studied by fluorescent microscopy using dichlorodihydrofluorescein diacetate. Tumor necrosis factor α (1 ng/mL) was added to the perfusion medium in the absence and presence of IL-10 (1 ng/mL). The role of phosphatidylinositol 3-kinase (PI3-kinase) was assessed using wortmannin and LY 2940002 (inhibitors of PI3-kinase). Specific inhibition of p110 α and p110 γ/δ PI3-kinase subunits was studied using A66 and TG100-115. As well, levels of ceramide and intercellular adhesion molecule 1 (ICAM-1) expression were measured. Finally, the effect of IL-10 on TNF-α-induced leukocyte/endothelium interaction was examined using an ex vivo perfused vessel model. Interleukin 10 significantly reduced dichlorodihydrofluorescein diacetate fluorescence induced by TNF-α in HUVECs (12.5% ± 3.2% vs. 111.7% ± 21.6% at 60 min). Pretreatment by LY2940002 or wortmannin restored ROS production induced by TNF-α in the presence of IL-10. In HUVECs treated by TNF-α + IL-10, inhibition of p110 α PI3-kinase subunit significantly increased ROS production, whereas p110 γ/δ inhibition did not have a significant effect. Pretreatment with IL-10 significantly decreased TNF-α-induced increased levels of ceramide (TNF-α vs. TNF-α + IL-10: 6,278 ± 1,013 vs. 1,440 ± 130 pmol/mg prot), as well as ICAM-1 expression and leukocyte adhesion (TNF-α vs. TNF-α + IL-10: 26.8 ± 2.6 vs. 6.7 ± 0.4 adherent leukocytes/field at 15 min). Interleukin 10 decreases the level of inflammation induced by TNF-α in endothelial cells by reducing the TNF-α-induced ROS production, ICAM-1 expression, and leukocyte adhesion to the endothelium. The antioxidant effect of IL-10 is mediated through PI3-kinase and is paralleled by a decrease in ceramide synthesis induced by TNF-α.
Collapse
|
15
|
Krakauer T. PI3K/Akt/mTOR, a pathway less recognized for staphylococcal superantigen-induced toxicity. Toxins (Basel) 2012; 4:1343-66. [PMID: 23202320 PMCID: PMC3509712 DOI: 10.3390/toxins4111343] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2012] [Revised: 11/12/2012] [Accepted: 11/13/2012] [Indexed: 12/27/2022] Open
Abstract
Immunostimulating staphylococcal enterotoxin B (SEB) and related superantigenic toxins cause diseases in humans and laboratory animals by activating cells of the immune system. These toxins bind directly to the major histocompatibility complex (MHC) class II molecules on antigen-presenting cells and specific Vβ regions of T-cell receptors (TCR), resulting in hyperactivation of both T lymphocytes and monocytes/macrophages. Activated host cells produce excessive amounts of proinflammatory cytokines and chemokines, especially tumor necrosis factor α, interleukin 1 (IL-1), IL-2, interferon γ (IFNγ), and macrophage chemoattractant protein 1 causing clinical symptoms of fever, hypotension, and shock. The well-explored signal transduction pathways for SEB-induced toxicity downstream from TCR/MHC ligation and interaction of cell surface co-stimulatory molecules include the mitogen-activated protein kinase cascades and cytokine receptor signaling, culminating in NFκB activation. Independently, IL-2, IFNγ, and chemokines from activated T cells signal via the phosphoinositide 3-kinase (PI3K), the serine/threonine kinases, Akt and mammalian target of rapamycin (mTOR) pathways. This article reviews the signaling molecules induced by superantigens in the activation of PI3K/Akt/mTOR pathways leading to staphylococcal superantigen-induced toxicity and updates potential therapeutics against superantigens.
Collapse
Affiliation(s)
- Teresa Krakauer
- Department of Immunology, Integrated Toxicology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD 21702, USA.
| |
Collapse
|
16
|
Circulating Cytokine Levels in Acute Pancreatitis—Model of SIRS/CARS Can Help in the Clinical Assessment of Disease Severity. Inflammation 2011; 35:758-63. [DOI: 10.1007/s10753-011-9371-z] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
17
|
Hedrich CM, Bream JH. Cell type-specific regulation of IL-10 expression in inflammation and disease. Immunol Res 2010; 47:185-206. [PMID: 20087682 DOI: 10.1007/s12026-009-8150-5] [Citation(s) in RCA: 150] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
IL-10 plays an essential part in controlling inflammation and instructing adaptive immune responses. Consequently, dysregulation of IL-10 is linked with susceptibility to numerous infectious and autoimmune diseases in mouse models and in humans. It has become increasingly clear that appropriate temporal/spatial expression of IL-10 may be the key to how IL-10 contributes to the delicate balance between inflammation and immunoregulation. The mechanisms that govern the cell type- and receptor-specific induction of IL-10, however, remain unclear. This is due largely to the wide distribution of cellular sources that express IL-10 under diverse stimulation conditions and in a variety of tissue compartments. Further complicating the issue is the fact that human IL-10 expression patterns appear to be under genetic influence resulting in differential expression and disease susceptibility. In this review, we discuss the cellular sources of IL-10, their link to disease phenotypes and the molecular mechanisms implicated in IL-10 regulation.
Collapse
Affiliation(s)
- Christian M Hedrich
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Room E5624, Baltimore, MD 21205-1901, USA
| | | |
Collapse
|
18
|
Therapeutic down-modulators of staphylococcal superantigen-induced inflammation and toxic shock. Toxins (Basel) 2010; 2:1963-83. [PMID: 22069668 PMCID: PMC3153276 DOI: 10.3390/toxins2081963] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2010] [Revised: 07/16/2010] [Accepted: 07/28/2010] [Indexed: 12/21/2022] Open
Abstract
Staphylococcal enterotoxin B (SEB) and related superantigenic toxins are potent stimulators of the immune system and cause a variety of diseases in humans, ranging from food poisoning to toxic shock. These toxins bind directly to major histocompatibility complex (MHC) class II molecules on antigen-presenting cells and specific Vβ regions of T-cell receptors (TCR), resulting in hyperactivation of both monocytes/macrophages and T lymphocytes. Activated host cells produce massive amounts of proinflammatory cytokines and chemokines, activating inflammation and coagulation, causing clinical symptoms that include fever, hypotension, and shock. This review summarizes the in vitro and in vivo effects of staphylococcal superantigens, the role of pivotal mediators induced by these toxins in the pathogenic mechanisms of tissue injury, and the therapeutic agents to mitigate the toxic effects of superantigens.
Collapse
|
19
|
Palmatine attenuates d-galactosamine/lipopolysaccharide-induced fulminant hepatic failure in mice. Food Chem Toxicol 2010; 48:222-8. [DOI: 10.1016/j.fct.2009.10.004] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2009] [Revised: 09/08/2009] [Accepted: 10/01/2009] [Indexed: 11/20/2022]
|
20
|
Toll-like receptor 2 ligands on the staphylococcal cell wall downregulate superantigen-induced T cell activation and prevent toxic shock syndrome. Nat Med 2009; 15:641-8. [PMID: 19465927 DOI: 10.1038/nm.1965] [Citation(s) in RCA: 101] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2008] [Accepted: 04/07/2009] [Indexed: 01/08/2023]
Abstract
Staphylococcal superantigens are pyrogenic exotoxins that cause massive T cell activation leading to toxic shock syndrome and death. Despite the strong adaptive immune response induced by these toxins, infections by superantigen-producing staphylococci are very common clinical events. We hypothesized that this may be partly a result of staphylococcal strains having developed strategies that downregulate the T cell response to these toxins. Here we show that the human interleukin-2 response to staphylococcal superantigens is inhibited by the simultaneous presence of bacteria. Such a downregulatory effect is the result of peptidoglycan-embedded molecules binding to Toll-like receptor 2 and inducing interleukin-10 production and apoptosis of antigen-presenting cells. We corroborated these findings in vivo by showing substantial prevention of mortality after simultaneous administration of staphylococcal enterotoxin B with either heat-killed staphylococci or Staphylococcus aureus peptidoglycan in mouse models of superantigen-induced toxic shock syndrome.
Collapse
|
21
|
Rajagopalan G, Tilahun AY, Asmann YW, David CS. Early gene expression changes induced by the bacterial superantigen staphylococcal enterotoxin B and its modulation by a proteasome inhibitor. Physiol Genomics 2009; 37:279-93. [PMID: 19336531 DOI: 10.1152/physiolgenomics.90385.2008] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Toxic shock syndrome (TSS) is an acute, serious systemic illness caused by bacterial superantigens. Nonavailability of a suitable animal model until recently has hampered an in-depth understanding of the pathogenesis of TSS. In the current study, we characterized the early molecular events underlying TSS using our HLA-DR3 transgenic mouse model. Gene expression profiling using DNA microarrays identified a rapid and significant upregulation of several pro- as well as anti-inflammatory mediators, many of which have never been previously described in TSS. In vivo administration of staphylococcal enterotoxin B (SEB) led to an increase in the expression of Th0- (IL-2, 240-fold); Th1- (IFN-gamma, 360-fold; IL-12, 8-fold); Th2- (IL-4, 53-fold; IL-5, 4-fold) as well as Th17-type cytokines (IL-21, 19-fold; IL-17, 5-fold). The immunoregulatory cytokines (IL-6, 700-fold; IL-10, 18-fold); CC chemokines (such as CCL 2, 11, 3, 24, 17, 12, 7), CXC chemokines (such as CXCL 1, 2, 5, 11, 10, 19); and several proteases (matrix metalloproteinases 13, 8, 3, and 9) were also upregulated. Serum levels of several of these cytokines/chemokines were also significantly elevated. Pathway analyses revealed significant modulation in a variety of biochemical and cellular functions, providing molecular insights into the pathogenesis of TSS. Administration of bortezomib, a clinically approved proteasome inhibitor capable of blocking NF-kappaB pathway, was able to significantly modulate the expression of a variety of genes induced by SEB. Thus, our study showed that TSS is a complex process and emphasized the potential of use of bortezomib in the therapy of superantigen-induced TSS.
Collapse
|
22
|
Sponseller BA, de Macedo MMA, Clark SK, Gallup JM, Jones DE. Activation of peripheral blood monocytes results in more robust production of IL-10 in neonatal foals compared to adult horses. Vet Immunol Immunopathol 2008; 127:167-73. [PMID: 18976818 DOI: 10.1016/j.vetimm.2008.09.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2008] [Revised: 08/01/2008] [Accepted: 09/16/2008] [Indexed: 11/18/2022]
Abstract
Foals are particularly vulnerable to infection by Rhodococcus equi during the first 2 weeks of life whereas mature horses are not. While an innate immunodeficiency likely accounts for this clinically relevant vulnerability, the factors that contribute to infection by R. equi have not been fully elucidated. In this study, we demonstrate that cells of the monocyte lineage, including monocytes, macrophages, and dendritic cells, that have been activated with LPS and IFN-gamma, respond with a statistically significant, greater amount of cytokine mRNA production of IL-10, IL-12p35, and IL-12p40 than unstimulated control cells. Interestingly, activation of neonatal cells resulted in a twofold log increase in baseline cytokine mRNA expression of IL-10 compared with adult cells. In contrast, no significant differences in mean cytokine mRNA expression of IL-12p35 and IL-12p40 were detected, suggesting that the defect in chromosomal remodeling that prevents IL-12p35 gene transcription as a cause for decreased IL-12 synthesis in human neonates is not a likely occurrence in equine neonates. Collectively, these differences indicate that in vivo activation of equine cells of the monocyte lineage may result in different autocrine and paracrine cellular responses that vary according to age, with potential impact on regulation of adaptive and innate immune responses.
Collapse
Affiliation(s)
- B A Sponseller
- Department of Veterinary Microbiology and Preventive Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA.
| | | | | | | | | |
Collapse
|
23
|
Huzella LM, Buckley MJ, Alves DA, Stiles BG, Krakauer T. Central roles for IL-2 and MCP-1 following intranasal exposure to SEB: a new mouse model. Res Vet Sci 2008; 86:241-7. [PMID: 18793785 DOI: 10.1016/j.rvsc.2008.07.020] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2008] [Revised: 07/23/2008] [Accepted: 07/29/2008] [Indexed: 02/06/2023]
Abstract
Murine models for bacterial superantigens like staphylococcal enterotoxin B (SEB) have to date been rather cumbersome. The reasons include: (1) necessary use of potentiating agents such as actinomycin D, d-galactosamine, lipopolysaccharide (LPS), or viruses; (2) high toxin amounts required to elicit effects; and/or (3) generation of phenotypic-stable transgenic animals. Our study employed readily available C3H/HeJ (TLR4 negative, LPS-nonresponsive) mice with intranasal and intraperitoneal administration of low microgram quantities of SEB. These animals responded to SEB with severe lung inflammation and hypothermia, culminating in death. A survey of cytokines/chemokines in sera and lungs after lethal intoxication revealed that monocyte chemoattractant protein-1 and interleukin-2 were associated with effects in this model. In contrast, SEB had minimal effects upon congenic (TLR4 positive, LPS-responsive) C3H/OuJ mice. Lethality of SEB in C3H/HeJ mice was neutralized with SEB-specific antibodies, suggesting potential utility of this model for future therapeutic studies.
Collapse
Affiliation(s)
- Louis M Huzella
- Pathology Division, US Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702-5011, United States
| | | | | | | | | |
Collapse
|
24
|
|
25
|
Hayashida K, Chen Y, Bartlett AH, Park PW. Syndecan-1 is an in vivo suppressor of Gram-positive toxic shock. J Biol Chem 2008; 283:19895-903. [PMID: 18499671 DOI: 10.1074/jbc.m801614200] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
Heparan sulfate proteoglycans bind to and regulate many inflammatory mediators in vitro, suggesting that they serve an important role in influencing inflammatory responses in vivo. Here we evaluated the role of syndecan-1, a major heparan sulfate proteoglycan, in modulating inflammatory responses in Gram-positive toxic shock, a systemic disease that is a significant cause of morbidity and mortality. Syndecan-1-null and wild-type mice were injected intraperitoneally with staphylococcal enterotoxin B, a pyrogenic superantigen, and their inflammatory responses were assessed. Syndecan-1-null mice showed significantly increased liver injury, vascular permeability, and death in response to staphylococcal enterotoxin B challenge compared with wild-type mice. Although serum levels of systemic IL-2 and IFNgamma were similar between the two backgrounds, those of TNFalpha and IL-6 were significantly increased in syndecan-1-null mice undergoing Gram-positive toxic shock. Furthermore, syndecan-1-null mice challenged with staphylococcal enterotoxin B showed enhanced T cell accumulation in tissues, whereas immunodepletion of T cells protected syndecan-1-null mice from the magnified systemic cytokine storm, inflammatory tissue injury, and death. Importantly, syndecan-1 shedding was induced in wild-type mice injected with staphylococcal enterotoxin B, and the administration of heparan sulfate, but not syndecan-1 core protein, rescued syndecan-1-null mice from lethal toxic shock by suppressing the production of TNFalpha and IL-6, and attenuating inflammatory tissue injury. Altogether, these data suggest that syndecan-1 shedding is a key endogenous mechanism that protects the host from Gram-positive toxic shock by inhibiting the dysregulation and amplification of the inflammatory response.
Collapse
Affiliation(s)
- Kazutaka Hayashida
- Division of Respiratory Diseases, Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | |
Collapse
|
26
|
Ewaschuk J, Endersby R, Thiel D, Diaz H, Backer J, Ma M, Churchill T, Madsen K. Probiotic bacteria prevent hepatic damage and maintain colonic barrier function in a mouse model of sepsis. Hepatology 2007; 46:841-50. [PMID: 17659579 DOI: 10.1002/hep.21750] [Citation(s) in RCA: 144] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
UNLABELLED A breakdown in intestinal barrier function and increased bacterial translocation are key events in the pathogenesis of sepsis and liver disease. Altering gut microflora with noninvasive and immunomodulatory probiotic organisms has been proposed as an adjunctive therapy to reduce the level of bacterial translocation and prevent the onset of sepsis. The purpose of this study was to determine the efficacy of a probiotic compound in attenuating hepatic and intestinal injury in a mouse model of sepsis. Wild-type and interleukin-10 (IL-10) gene-deficient 129 Sv/Ev mice were fed the probiotic compound VSL#3 for 7 days. To induce sepsis, the mice were injected with lipopolysaccharide (LPS) and D-galactosamine (GalN) in the presence and absence of the peroxisome proliferator-activated receptor gamma (PPARgamma) inhibitor GW9662. The mice were killed after 6 hours, and their colons were removed for the measurement of the cytokine production and epithelial function. The functional permeability was assessed by the mannitol movement and cyclic adenosine monophosphate-dependent chloride secretion in tissue mounted in Ussing chambers. The livers were analyzed for bacterial translocation, cytokine production, histological injury, and PPARgamma levels. The tissue levels of tumor necrosis factor alpha, interferon gamma, IL-6, and IL-12p35 ribonucleic acid were measured by semiquantitative reverse transcription polymerase chain reaction. Mice injected with LPS/GalN demonstrated a breakdown in colonic barrier function, which correlated with enhanced proinflammatory cytokine secretion, bacterial translocation, and significant hepatic injury. A pretreatment with oral probiotics prevented the breakdown in intestinal barrier function, reduced bacterial translocation, and significantly attenuated liver injury. The inhibition of PPARgamma with GW9662 abrogated the protection induced by probiotics. CONCLUSION Orally administered probiotics prevented liver and intestinal damage in a mouse model of sepsis through a PPARgamma-dependent mechanism.
Collapse
Affiliation(s)
- Julia Ewaschuk
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Plaza R, Rodriguez-Sanchez JL, Juarez C. Staphylococcal enterotoxin B in vivo modulates both gamma interferon receptor expression and ligand-induced activation of signal transducer and activator of transcription 1 in T cells. Infect Immun 2007; 75:306-13. [PMID: 17074848 PMCID: PMC1828398 DOI: 10.1128/iai.01220-06] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2006] [Revised: 09/15/2006] [Accepted: 10/19/2006] [Indexed: 01/24/2023] Open
Abstract
Superantigens (SAg) are bacterial exotoxins that provoke extreme responses in the immune system; for example, the acute hyperactivation of SAg-reactive T cells that leads to toxic shock syndrome is followed within days by strong immunosuppression. The gamma interferon (IFN-gamma) response is deeply affected in both extremes. The implication of IFN-gamma in the pathophysiology of lethal shock induced in mice after a secondary challenge with the SAg staphylococcal enterotoxin B (SEB) prompted us to study the regulation of IFN-gamma secretion and the intracellular response. We demonstrate in this study that a rechallenge with SEB becomes lethal only when given inside a critical time window after SEB priming and is associated with an increase of IFN-gamma serum release 72 h after priming. However, at this time, a selective blockade of IFN-gamma/STAT1 signaling develops in spleen cells, correlating with a lack of expression of the IFN-gamma receptor beta subunit and STAT1 in the T-cell population. Selective blockade of the STAT1 signaling pathway--while simultaneously maintaining STAT3 signaling and expression--may be a protective mechanism that shortens IFN-gamma production during the Th1 effector response. This blockade may also have consequences on switching towards a suppressor phenotype with chronic exposure to the superantigen.
Collapse
Affiliation(s)
- R Plaza
- Department of Immunology, Hospital de la Santa Creu i Sant Pau, Avda. Sant Antoni Maria Claret 167, 08025 Barcelona, Spain
| | | | | |
Collapse
|
28
|
Johansson L, Rytkönen A, Wan H, Bergman P, Plant L, Agerberth B, Hökfelt T, Jonsson AB. Human-like immune responses in CD46 transgenic mice. THE JOURNAL OF IMMUNOLOGY 2005; 175:433-40. [PMID: 15972677 DOI: 10.4049/jimmunol.175.1.433] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Neisseria meningitidis is a major cause of sepsis and/or meningitis. These bacteria normally cause disease only in humans, however, mice expressing human CD46 are susceptible to meningococcal disease. To explain the sensitivity of CD46 transgenic mice to meningococci, we evaluated early immune responses. Stimulation of TNF, IL-6, and IL-10 was stronger in CD46 transgenic mice compared with nontransgenic mice, and resembled human responses. In CD46 transgenic mice, bacterial clearance in blood started at later time points, and neutrophil numbers in blood were lower compared with nontransgenic mice. Further, elevated levels of activated microglia cells and cyclooxygenase-2 were observed in brain of infected CD46 transgenic mice. Intraperitoneal administration of meningococci lead to increased levels of macrophages only in the i.p. cavity of CD46 transgenic mice. Most of the responses were impaired or absent using LPS-deficient meningococci, showing the importance of LPS in the early immune response to meningococcal infection. Taken together, these data demonstrate that responses in mice expressing human CD46 mimic human meningococcal disease in many aspects, and demonstrate novel important links between CD46 and the innate immune system.
Collapse
Affiliation(s)
- Linda Johansson
- Department of Infectious Diseases, Centre for Molecular Microbiology and Infection, Imperial College, London, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Chakraborty A, Yeung S, Pyszczynski NA, Jusko WJ. Pharmacodynamic interactions between recombinant mouse interleukin-10 and prednisolone using a mouse endotoxemia model. J Pharm Sci 2005; 94:590-603. [PMID: 15666292 PMCID: PMC4196336 DOI: 10.1002/jps.20257] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The pharmacodynamic interactions between recombinant mouse interleukin-10 (IL-10) and prednisolone were examined in lipopolysaccharide (LPS)-induced experimental endotoxemia in Balb/c mice. Treatment phases consists of single doses of IL-10 (10 microg/kg i.p.), prednisolone (25 (mg/kg i.p.), IL-10 (2.5 microg/kg i.p.) with prednisolone (6.25 mg/kg i.p.), or placebo (saline). Measurements included plasma steroid kinetics and IL-10 concentrations and responses to LPS including proinflammatory cytokines (TNF-alpha, IFN-gamma) and circulatory NO measured as plasma nitrate/nitrite concentrations. The intraperitoneal dosing of LPS produced large and transient elevations of plasma TNF-alpha, IFN-gamma, and NO concentrations. Noncompartmental and model fitting using extended indirect response models based on drug inhibition of multiphase stimulation of biomarkers by LPS were used to describe the in vivo pharmacodynamics and drug interactions. Dosing with prednisolone, IL-10, or their combinations produced strong inhibition of cytokine and NO production. The IC50 values of prednisolone ranged from 54 to 171 ng/mL, and IC50 values for IL-10 ranged from 0.06 to 0.69 ng/mL. The production of NO was described as a cascading consequence of the TNF-alpha and IFN-gamma plasma concentrations. The joint dosing of IL-10 with prednisolone produces moderately synergistic immunosuppressive effects in this system. Both drugs were sufficiently protective in suppressing the inflammatory mediators when administered prior to the LPS trigger, while such effects were modest when administered after the inflammatory stimulus was provoked. The integrated and complex pharmacokinetic/pharmacodynamic models well capture the in vivo processes, drug potencies, and interactions of IL-10 and prednisolone.
Collapse
Affiliation(s)
- Abhijit Chakraborty
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Buffalo, 565 Hochstetter Hall, Buffalo, New York 14260, USA
| | | | | | | |
Collapse
|
30
|
Moscovis SM, Gordon AE, Al Madani OM, Gleeson M, Scott RJ, Roberts-Thomson J, Hall ST, Weir DM, Busuttil A, Blackwell CC. Interleukin-10 and sudden infant death syndrome. ACTA ACUST UNITED AC 2004; 42:130-8. [PMID: 15325406 DOI: 10.1016/j.femsim.2004.06.020] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2004] [Accepted: 06/14/2004] [Indexed: 11/23/2022]
Abstract
Uncontrolled pro-inflammatory responses to infections or bacterial toxins have been suggested to play a role in triggering the physiological events leading to sudden infant death syndrome (SIDS). We tested the hypothesis that these uncontrolled responses might be due to interactions between the gene polymorphisms inducing low levels of IL-10 and exposure to cigarette smoke. In vitro, the IL-10 (G-1082A) polymorphism was associated with low IL-10 levels and the -1082G allele was associated with high levels. The first objective was to assess the distribution of this polymorphism among SIDS infants, parents of SIDS infants and controls, and two ethnic groups: Aboriginal Australians who have a high incidence of SIDS; and Bangladeshis who in Britain have a low incidence of SIDS compared with Europeans. The second objective was to assess effects of human recombinant IL-10 on interleukin-6 (IL-6) and tumour necrosis factor-alpha (TNF-alpha) responses of human leukocytes to staphylococcal toxins implicated in SIDS. The third objective was to assess IL-10 responses to endotoxin and toxic shock syndrome toxin (TSST) from leukocytes of smokers and non-smokers in relation to the IL-10 (G-1082A) polymorphism. There were major differences in the distributions of these polymorphisms between Europeans and Bangladeshis (p=0.00) and between Europeans and Aboriginal Australians (p=0.00); however, they were similar for the Bangladeshi and Aboriginal Australian subjects. There were no significant differences in the distribution of these polymorphisms among SIDS infants or parents of SIDS infants compared to control groups. IL-10 significantly reduced IL-6 and TNF-alpha responses to TSST and staphylococcal enterotoxins A and C. At 50 ng ml(-1), IL-10 significantly increased TNF-alpha but not IL-6 responses to TSST and enterotoxin A. Although IL-10 responses to endotoxin were lower from leukocytes of smokers who were homozygous for the G allele, the differences were not significant; however, significantly lower IL-10 responses were found for smokers who were homozygous for the A allele (p=0.01) and heterozygotes (p=0.04). The pooled data found smokers had significantly lower levels of IL-10 responses to TSST, but there were no significant differences for smokers compared with non-smokers for the three genotypes. The high incidence of SIDS and serious respiratory infections among Aboriginal Australian infants and the low incidence of these conditions among Bangladeshi infants might be explained in part by our findings of differences in IL-10 responses between smokers and non-smokers. The lowest levels of IL-10 responses were observed among smokers who were homozygous for the A allele which is most prevalent among the Aboriginal Australians (83%) and Bangladeshis (84%). The major difference between the risk factors for SIDS in these two groups is the level of exposure of infants to cigarette smoke associated with maternal smoking.
Collapse
Affiliation(s)
- Sophia M Moscovis
- School of Biomedical Sciences, Faculty of Health, University of Newcastle, and Hunter Medical Research Institute, Newcastle, NSW, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Blackwell CC, Moscovis SM, Gordon AE, Al Madani OM, Hall ST, Gleeson M, Scott RJ, Roberts-Thomson J, Weir DM, Busuttil A. Ethnicity, infection and sudden infant death syndrome. ACTA ACUST UNITED AC 2004; 42:53-65. [PMID: 15325398 DOI: 10.1016/j.femsim.2004.06.007] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2004] [Accepted: 06/14/2004] [Indexed: 11/24/2022]
Abstract
Epidemiological studies found the incidence of SIDS among Indigenous groups such as Aboriginal Australians, New Zealand Maoris and Native Americans were significantly higher than those for non-Indigenous groups within the same countries. Among other groups such as Asian families in Britain, the incidence of SIDS has been lower than among groups of European origin. Cultural and childrearing practices as well as socio-economic factors have been proposed to explain the greater risk of SIDS among Indigenous peoples; however, there are no definitive data to account for the differences observed. We addressed the differences among ethnic groups in relation to susceptibility to infection because there is evidence from studies of populations of European origin that infectious agents, particularly toxigenic bacteria might trigger the events leading to SIDS. The risk factors for SIDS parallel those for susceptibility to infections in infants, particularly respiratory tract infections which are also major health problems among Indigenous groups. Many of the risk factors identified in epidemiological studies of SIDS could affect three stages in the infectious process: (1) frequency or density of colonisation by the toxigenic species implicated in SIDS; (2) induction of temperature-sensitive toxins; (3) modulation of the inflammatory responses to infection or toxins. In this review we compare genetic, developmental and environmental risk factors for SIDS in ethnic groups with different incidences of SIDS: low (Asians in Britain); moderate (European/Caucasian); high (Aboriginal Australian). Our findings indicate: (1) the major difference was high levels of exposure to cigarette smoke among infants in the high risk groups; (2) cigarette smoke significantly reduced the anti-inflammatory cytokine interleukin-10 responses which control pro-inflammatory responses implicated in SIDS; (3) the most significant effect of cigarette smoke on reduction of IL-10 responses was observed for donors with a single nucleotide polymorphism for the IL-10 gene that is predominant among both Asian and Aboriginal populations. If genetic makeup were a major factor for susceptibility to SIDS, the incidence of these deaths should be similar for both populations. They are, however, significantly different and most likely reflect differences in maternal smoking which could affect frequency and density of colonisation of infants by potentially pathogenic bacteria and induction and control of inflammatory responses.
Collapse
Affiliation(s)
- C Caroline Blackwell
- Immunology and Microbiology, Faculty of Health, David Maddison Building, School of Biomedical Sciences, University of Newcastle, and Hunter Medical Research Institute, Newcastle, NSW 2300, Australia.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Zang Y, Dolan SM, Ni Choileain N, Kriynovich SJ, Murphy TJ, Sayles P, Mannick JA, Lederer JA. Burn Injury Initiates a Shift in Superantigen-Induced T Cell Responses and Host Survival. THE JOURNAL OF IMMUNOLOGY 2004; 172:4883-92. [PMID: 15067067 DOI: 10.4049/jimmunol.172.8.4883] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Severe injury induces a temporal shift in immune reactivity that can cause serious complications or even death. We previously reported that mice exposed to bacterial superantigen (SAg) early after injury undergo a strong SAg response with lethal consequences. This study compares the early and late effects of burn injury on SAg reactivity in vivo to establish how injury influences adaptive immune responses. We found that mice challenged with ordinarily sublethal doses of staphylococcal enterotoxin A or staphylococcal enterotoxin B at 1 day after burn injury exhibited high mortality, whereas no mortality occurred at 7 days after injury. This shift in mortality correlated with higher Th2-type cytokines (IL-4 and IL-10) being expressed by CD4(+) and CD8(+) T cells from burn as opposed to sham mice at 7 days after injury. Lymph node cells from burn-injured mice also produced higher levels of Th2-type cytokines at 7 days after injury. The results of cell-mixing studies using CD4(+) and CD8(+) T cells mixed with APCs from sham or burn mice suggested that changes in both T cells and APCs are involved in the altered SAg response. Finally, the biological significance of altered SAg reactivity following injury was shown by demonstrating that blocking IL-10 activity in vivo caused higher SAg-induced mortality at 7 days after injury. These findings support the idea that injury promotes a Th2-type shift in adaptive immune reactivity. Although prior studies link this counterinflammatory-type response to lowered resistance to infection, the present results suggest it may sometimes benefit the injured host.
Collapse
Affiliation(s)
- Yan Zang
- Department of Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Kastin AJ, Akerstrom V, Pan W. Interleukin-10 as a CNS therapeutic: the obstacle of the blood-brain/blood-spinal cord barrier. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 2003; 114:168-71. [PMID: 12829328 DOI: 10.1016/s0169-328x(03)00167-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Interleukin (IL)-10 exerts beneficial effects on the central nervous system (CNS) after peripheral administration, but its penetration across the blood-brain barrier (BBB) has not been quantified. We show that 125I-IL-10 is stable in circulating blood but does not cross the intact BBB after intravenous delivery. Thus, peripheral IL-10 probably can serve as a CNS therapeutic only when the BBB is disrupted.
Collapse
Affiliation(s)
- Abba J Kastin
- VA Medical Center and Tulane University School of Medicine, 1601 Perdido Street, New Orleans, LA 70112-1262, USA.
| | | | | |
Collapse
|
34
|
Kotake Y, Moore DR, Vasquez-Walden A, Tabatabaie T, Sang H. Antioxidant amplifies antibiotic protection in the cecal ligation and puncture model of microbial sepsis through interleukin-10 production. Shock 2003; 19:252-6. [PMID: 12630525 DOI: 10.1097/00024382-200303000-00009] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Preadministration of antioxidants such as pyrrolidine dithiocarbamate (PDTC) and phenyl N-tert-butyl nitrone (PBN) protects animals from lethality in sepsis models. However, the requirement of preadministration greatly diminishes the clinical significance of these studies. Although the synthetic antioxidant PBN has been shown to effectively protect rodents from lethality in endotoxemia (lipopolysaccharide [LPS] model), preliminary screening indicates that pre- or postadministration of PBN does not protect in the rat cecal ligation and puncture (CLP) model. We show in this report that in a rat CLP model, the administration of PBN (150 mg/kg, 30 min after CLP) followed by the antibiotic imipenem (IMP; 10 mg/kg, 1 h after CLP) significantly increased survival compared with other single treatment groups. Previously, we have shown that PBN's protection in a rat LPS model is mediated by the overproduction of the anti-inflammatory cytokine interleukin (IL)-10. We show in this study that the increase in survival found in the PBN + IMP-treated group was abrogated by immunoneutralization with anti-IL-10 antibody, indicating that endogenous IL-10 is an effective protective factor. Plasma LPS levels were shown to be elevated after imipenem treatment, and the increased LPS level could have assisted to overproduce endogenous IL-10, as in the case of the PBN-treated LPS model. Statistical analysis indicated that the increase of IL-10 in PBN + IMP-treated group at early time period has significant association to the improvement of survival.
Collapse
Affiliation(s)
- Yashige Kotake
- Free Radical Biology and Aging Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104, USA
| | | | | | | | | |
Collapse
|
35
|
Hong-Geller E, Gupta G. Therapeutic approaches to superantigen-based diseases: a review. J Mol Recognit 2003; 16:91-101. [PMID: 12720278 DOI: 10.1002/jmr.612] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Superantigens secreted by the bacterial pathogen Staphyloccocus aureus are extremely potent toxins that overstimulate the host immune system by binding to the MHC class II and T cell receptors and activating a large population of T cells. Superantigen infection has been shown to be the causative agents in acute diseases, food poisoning and toxic shock syndrome, and in more chronic conditions such as inflammatory skin diseases. In addition to the toll on public health, S. aureus superantigens also represent a potential biothreat to our national security. To address these risks, a number of different therapeutic strategies have been developed that target different aspects of the pathogenic mechanism of S. aureus and superantigen infection. These therapies, which encompass strategies as diverse as production of neutralizing antibodies, inhibitory peptide/receptor design and blockage of superantigen gene transcription, are being tested for treatment of established S. aureus infections in pre- and post-exposure scenarios. In this review, we will describe these different strategies and their efficacies in inhibition of superantigen-induced effects in the host, and present the future outlook for successfully producing therapies for superantigen-based disease.
Collapse
Affiliation(s)
- Elizabeth Hong-Geller
- Los Alamos National Laboratory, Biosciences Division, HRL-1, MS-M888, Los Alamos, NM 87545, USA
| | | |
Collapse
|
36
|
Yekebas EF, Strate T, Zolmajd S, Eisenberger CF, Erbersdobler A, Saalmüller A, Steffani K, Busch C, Elsner HA, Engelhardt M, Gillesen A, Meins J, The M, Knoefel WT, Izbicki JR. Impact of different modalities of continuous venovenous hemofiltration on sepsis-induced alterations in experimental pancreatitis. Kidney Int 2002; 62:1806-18. [PMID: 12371983 DOI: 10.1046/j.1523-1755.2002.00607.x] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Continuous venovenous hemofiltration (CVVH) is assumed to attenuate systemic complications in septic diseases. The impact of different treatment intensities of CVVH on immunologic and systemic alterations in experimental pancreatitis was evaluated. METHODS Eighty-four minipigs were allocated either to an untreated control group (group 1) or to one of six treatment groups (groups 2 to 7) that underwent CVVH in different modalities: (1): "late" CVVH, started after a decline of total peripheral resistance of 30% versus "prophylactic" CVVH started immediately after the induction of pancreatitis; (2) no change of hemofilters versus a periodic change of filters every 12 hours; (3) low-volume CVVH with a filtrate turnover of 20 mL/kg body weight (BW)/h versus high-volume CVVH (100 mL/kg/h). Pancreatitis was induced by intraductal injection of sodium-taurocholate (3%, 1 mL/kg BW) and enterokinase (2 U/kg BW). We focused on the occurrence of sepsis, serum cytokines, down-regulation of major histocompatibility complex II (MHC II) and the endotoxin receptor CD14 expression, bacterial translocation/endotoxemia, and pulmonary and renal histologic alterations. RESULTS CVVH delayed or definitively prevented the occurrence of sepsis. Pancreatitis was associated with a tremendous initial tumor necrosis factor-alpha (TNF-alpha) response prior to a return to near baseline levels in the late course of sepsis. Endotoxin hyporesponsiveness, suggested by the dissociation of decreasing TNF-alpha levels and increasing endotoxemia in end-stage sepsis, was favorably influenced by CVVH. Down-regulation of MHC II and CD14 expression was prevented in non-septic animals. CVVH-related sepsis-protection led to a significant attenuation of histological injury in lungs and kidneys. "Prophylactic" CVVH prevented histological changes more effectively than "late" CVVH. CONCLUSIONS CVVH offers a therapeutic option for supportive treatment in severe pancreatitis. The efficiency of CVVH is associated with the duration of filter use and cumulative plasma turnover. Since CVVH may lead to sepsis-protection and long-term survival, further evaluation in controlled, clinical trials is warranted.
Collapse
Affiliation(s)
- Emre F Yekebas
- Department of Surgery, University Hospital Eppendorf, Hamburg, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Zhou X, Meyer CU, Schmidtke P, Zepp F. Effect of cordycepin on interleukin-10 production of human peripheral blood mononuclear cells. Eur J Pharmacol 2002; 453:309-17. [PMID: 12398919 DOI: 10.1016/s0014-2999(02)02359-2] [Citation(s) in RCA: 79] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Therapeutic options for controlling autoimmune diseases are still very limited. Interleukin-10 has been reported to be a promising approach to therapeutic intervention. In the search for a drug which results in the selective upregulation of interleukin-10, we investigated the immunoregulative effects of cordycepin. We have measured interleukin-10 and interleukin-2 secretion of human peripheral blood mononuclear cells that were incubated with cordycepin and assessed the influence of cordycepin on the expression of interleukin-10 mRNA, the proliferative response and the expression of surface markers on T lymphocytes. In addition, the subsets of interleukin-10-secreting cells, the influence of anti-interleukin-10 neutralizing antibody and cytotoxicity of cordycepin were evaluated. Our results suggest that cordycepin has a significantly upregulative effect on interleukin-10 production and interleukin-10 mRNA expression. Interleukin-10-producing cells included in CD4+, CD8+, CD19+, CD56+ and CD14+ cells. At the same time, cordycepin inhibited phytohaemagglutinin-induced interleukin-2 production and proliferation of peripheral blood mononuclear cells. A restricted T lymphocyte activation was also reflected by a reduced expression of the surface markers CD25, CD45RO, CD54, CD71 and HLA DR. Anti-interleukin-10 neutralizing antibody could not completely block the suppressive effect of cordycepin on production of interleukin-2. Cordycepin in the effective concentration presented slight cytotoxicity but did not increase apoptosis. These results indicate that cordycepin exerts immunoregulative effects. Further research on it may provide an approach for the development of novel immunomodulatory drugs which directly alter the secretion of cytokines.
Collapse
Affiliation(s)
- Xiaoxia Zhou
- Pediatric Immunology and Infectious Diseases, Children's Hospital, Johannes Gutenberg, University of Mainz, Obere Zahlbacher Street 63, D-55131, Mainz, Germany.
| | | | | | | |
Collapse
|
38
|
Stewart D, Fulton WB, Wilson C, Monitto CL, Paidas CN, Reeves RH, De Maio A. Genetic contribution to the septic response in a mouse model. Shock 2002; 18:342-7. [PMID: 12392278 DOI: 10.1097/00024382-200210000-00009] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The response to injury is dependent on several factors, including the type and extent of the injury, genetics, and the environment. In the present study, the genetic contribution to sepsis was evaluated in a mouse model. Sepsis was induced in two inbred mouse strains, C57BL/6J (B6) and A/J, by cecal ligation and single puncture (CLP). Frequency of mortality was significantly higher in B6 than A/J mice from 36 to 132 h after CLP. Plasma TNF-alpha, IL-1beta, and IL-6 levels were similar in both strains after CLP. IL-10 plasma levels were significantly higher in B6 mice as opposed to A/J mice after 24 h of CLP. Similarly, hepatic myeloperoxidase activity, an index of polymorphonuclear leukocytes, was elevated in B6 mice as compared with A/J mice after 24 h of CLP. On the contrary, metallothionein mRNA levels were higher in A/J mice compared with B6 mice. Finally, leptin levels were also higher in A/J than B6 mice within 19 h of CLP. This study demonstrates a genetic contribution in the response to sepsis.
Collapse
Affiliation(s)
- Dylan Stewart
- Division of Pediatric Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | | | | | | | | | | | | |
Collapse
|
39
|
|
40
|
Collins LV, Eriksson K, Ulrich RG, Tarkowski A. Mucosal tolerance to a bacterial superantigen indicates a novel pathway to prevent toxic shock. Infect Immun 2002; 70:2282-7. [PMID: 11953361 PMCID: PMC127931 DOI: 10.1128/iai.70.5.2282-2287.2002] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Enterotoxins with superantigenic properties secreted during systemic Staphylococcus aureus infection are responsible for toxic shock. We show that intranasal administration of staphylococcal enterotoxin A (SEA), but not a recombinant SEA lacking superantigenic activity, protected mice against lethal systemic SEA challenge. Protection was superantigen specific since intranasal exposure to SEA would not protect against death caused by subsequent toxic shock syndrome toxin 1 systemic challenge. Protection was neither due to selective depletion of SEA-specific T-cell receptor Vbeta families nor due to production of neutralizing anti-SEA antibodies. Importantly, the production of interleukin 10 (IL-10) induced by "tolerization" (that is, by the induction of immunological tolerance) contributed to the observed protection against lethal superantigen-triggered disease. In support of this notion we found that (i) significantly increased levels of IL-10 in sera of "tolerized" animals (that is, animals rendered tolerant) and (ii) IL-10(-/-) mice could not be tolerized by mucosal SEA administration. Altogether, this is the first study to show that mucosal tolerance to a superantigen is readily triggered by means of immunodeviation.
Collapse
Affiliation(s)
- L Vincent Collins
- Department of Rheumatology, University of Göteborg, Göteborg, Sweden.
| | | | | | | |
Collapse
|
41
|
Kotake Y, Sang H, Tabatabaie T, Wallis GL, Moore DR, Stewart CA. Interleukin-10 overexpression mediates phenyl-N-tert-butyl nitrone protection from endotoxemia. Shock 2002; 17:210-6. [PMID: 11900340 DOI: 10.1097/00024382-200203000-00009] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The free radical trapping compound phenyl N-tert-butylnitrone (PBN) provides potent protection against lethal endotoxemia in rodents, but the mechanism of this protection is not well understood. The objective of this study was to show that PBN administration in lipopolysaccharide- (LPS) induced endotoxemia promotes enhanced production of endogenous interleukin 10 (IL-10), and the expressed IL-10 is a causal factor in the protection from endotoxemia. We show the amplified expression of IL-10 in liver and plasma in PBN- (150 mg/kg) plus LPS- (4 mg/kg) treated rats using ribonuclease protection assay (RPA) and ELISA. In situ hybridization was utilized to visualize the overexpression of the IL-10 gene, and ELISA was used to determine plasma IL-10 and TNFalpha levels. Plasma IL-10 showed a 3-fold increase in PBN/LPS- treated rats compared to those treated with LPS alone, and in contrast, TNFalpha level decreased by more than 90%. However, the administration of PBN alone induced no IL-10 production. Immunoneutralization of IL-10 through anti-IL-10 antibody administration to PBN/LPS-treated rats abrogated PBN's suppression of systemic nitric oxide (NO) formation, a surrogate marker for the severity of endotoxemia, indicating that IL-10 is a causal factor for the protection. In these experiments, systemic NO level was quantified using an in vivo electron paramagnetic resonance (EPR) NO-trapping technique. Gel-shift and immunohistochemical analyses indicated that the transcription factor NF-kappaB was deactivated after PBN treatment, suggesting that NF-kappaB deactivation is closely involved in IL-10 overexpression.
Collapse
Affiliation(s)
- Yashige Kotake
- Free Radical Biology and Aging Research Program, Oklahoma Medical Research Foundation, Oklahoma City 73104, USA
| | | | | | | | | | | |
Collapse
|
42
|
Lauw FN, Florquin S, Speelman P, van Deventer SJ, van der Poll T. Role of endogenous interleukin-12 in immune response to staphylococcal enterotoxin B in mice. Infect Immun 2001; 69:5949-52. [PMID: 11500480 PMCID: PMC98720 DOI: 10.1128/iai.69.9.5949-5952.2001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In the present study, the roles of interleukin 12 (IL-12) and IL-18 and their possible interaction during superantigen-induced responses were studied by injection of staphylococcal enterotoxin B (SEB) into mice. These data suggest that the role of IL-12 in SEB-induced responses is limited to sustaining gamma interferon release by an IL-18-independent mechanism.
Collapse
Affiliation(s)
- F N Lauw
- Laboratory of Experimental Internal Medicine, University of Amsterdam, Amsterdam, The Netherlands
| | | | | | | | | |
Collapse
|
43
|
Tan BB, Prewitt RL, Britt LD. Interleukin-10 prevents loss of tone of rat skeletal muscle arterioles exposed to endotoxin. J Surg Res 2001; 100:110-5. [PMID: 11516213 DOI: 10.1006/jsre.2001.6105] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
BACKGROUND The anti-inflammatory cytokine interleukin-10 (IL-10) is known to inhibit the development of septic shock in animal models. This study was conducted to investigate the effect of IL-10 on the loss of vascular tone during exposure to endotoxin. Unlike numerous proinflammatory cytokines, the effects of IL-10 at the level of the microvasculature have not been previously studied. MATERIALS AND METHODS First-order rat cremasteric arterioles (n = 27) were placed in an isolated vessel preparation and allowed to achieve spontaneous tone. An opened segment of thoracic aorta was then placed upstream from the arteriole in the superfusion line. The resistance arteriole of this in-series model, in contrast to the isolated arteriole alone, demonstrates a significant loss of tone when exposed to endotoxin. Following 1 h of equilibration in the presence or absence of IL-10 (20 ng/ml), the aorta and arteriole were then superfused with 2.5 microg/ml endotoxin or physiologic buffer for 60 min and serial arteriolar diameter measurements were recorded. Group 1 was exposed to endotoxin only, Group 2 was a time control, and Group 3 was pretreated with IL-10 prior to endotoxin exposure, while Group 4 was a control pretreated with IL-10 only. RESULTS After the 60-min equilibration period there were no differences among the four groups in arteriolar tone. At t = 120 min, the percentage of tone in the control group was 43.6 +/- 3% (mean +/- SEM) and this was not changed by treatment with IL-10 (47.0 +/- 7% tone). Endotoxin alone caused arteriolar tone to fall to 31.4 +/- 3% (P < 0.05). However, endotoxin applied to arterioles pretreated with IL-10 was without effect (47.5 +/- 2%). CONCLUSIONS Resistance arterioles pretreated with IL-10 maintain vascular tone during endotoxin exposure. We conclude that IL-10 pretreatment prevents loss of vascular tone of isolated arterioles exposed to endotoxin.
Collapse
Affiliation(s)
- B B Tan
- Department of Surgery, Eastern Virginia Medical School, Norfolk, Virginia 23507, USA
| | | | | |
Collapse
|
44
|
Gerber JS, Mosser DM. Reversing lipopolysaccharide toxicity by ligating the macrophage Fc gamma receptors. THE JOURNAL OF IMMUNOLOGY 2001; 166:6861-8. [PMID: 11359846 DOI: 10.4049/jimmunol.166.11.6861] [Citation(s) in RCA: 209] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Our laboratory has previously demonstrated that the ligation of phagocytic receptors on macrophages can influence cytokine production. In this study, we examine the cytokine responses to multiple inflammatory stimuli following FcgammaR ligation. Macrophages were stimulated in vitro with LPS, lipoteichoic acid, CD40 ligand, or low molecular mass hyaluronic acid. All of these stimuli were proinflammatory in character, inducing the production of high levels of IL-12, but only modest amounts of IL-10. The coligation of FcgammaR along with these stimuli resulted in an anti-inflammatory profile, abrogating IL-12 production and inducing high levels of IL-10. The modulation of these two cytokines occurred by two independent mechanisms. Whereas the abrogation of IL-12 biosynthesis was a property shared by several macrophage receptors, the induction of IL-10 was specific to the FcgammaR. The biological relevance of these observations was examined in murine models of endotoxemia, in which FcgammaR ligation induced the rapid production of IL-10 and prevented IL-12 synthesis. Mice could be passively immunized with Abs to LPS to reverse inflammatory cytokine production, and the transfer of macrophages whose FcgammaR had been ligated could rescue mice from lethal endotoxemia. Thus, the ligation of the macrophage FcgammaR can be exploited to prevent inappropriate inflammatory cytokine responses.
Collapse
Affiliation(s)
- J S Gerber
- Department of Microbiology and Immunology, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | | |
Collapse
|
45
|
Abstract
Interleukin-10 (IL-10), first recognized for its ability to inhibit activation and effector function of T cells, monocytes, and macrophages, is a multifunctional cytokine with diverse effects on most hemopoietic cell types. The principal routine function of IL-10 appears to be to limit and ultimately terminate inflammatory responses. In addition to these activities, IL-10 regulates growth and/or differentiation of B cells, NK cells, cytotoxic and helper T cells, mast cells, granulocytes, dendritic cells, keratinocytes, and endothelial cells. IL-10 plays a key role in differentiation and function of a newly appreciated type of T cell, the T regulatory cell, which may figure prominently in control of immune responses and tolerance in vivo. Uniquely among hemopoietic cytokines, IL-10 has closely related homologs in several virus genomes, which testify to its crucial role in regulating immune and inflammatory responses. This review highlights findings that have advanced our understanding of IL-10 and its receptor, as well as its in vivo function in health and disease.
Collapse
MESH Headings
- Animals
- Autoimmune Diseases/genetics
- Autoimmune Diseases/immunology
- Clinical Trials as Topic
- Clinical Trials, Phase II as Topic
- Dendritic Cells/immunology
- Diabetes Mellitus, Type 1/immunology
- Disease Models, Animal
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Gene Expression Regulation
- Herpesviridae/physiology
- Humans
- Infections
- Inflammation
- Interleukin-10/genetics
- Interleukin-10/physiology
- Interleukin-10/therapeutic use
- Lupus Erythematosus, Systemic/genetics
- Lupus Erythematosus, Systemic/immunology
- Lymphocyte Subsets/immunology
- Mice
- Mice, Inbred NOD
- Mice, Inbred NZB
- Mice, Knockout
- Neoplasms/immunology
- Neutrophils/immunology
- Primates
- Protein-Tyrosine Kinases/physiology
- Receptors, Interleukin/genetics
- Receptors, Interleukin/physiology
- Receptors, Interleukin-10
- Signal Transduction
- Transcription Factors/physiology
- Transcription, Genetic
- Viral Proteins/physiology
Collapse
Affiliation(s)
- K W Moore
- Department of Molecular Biology, DNAX Research Institute of Molecular and Cellular Biology Inc., Palo Alto, California 94304, USA.
| | | | | | | |
Collapse
|
46
|
Huang LY, Reis e Sousa C, Itoh Y, Inman J, Scott DE. IL-12 induction by a TH1-inducing adjuvant in vivo: dendritic cell subsets and regulation by IL-10. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 167:1423-30. [PMID: 11466361 DOI: 10.4049/jimmunol.167.3.1423] [Citation(s) in RCA: 86] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
IL-12 induction is critical for immune responses against many viruses and intracellular bacterial pathogens. Recent studies suggest that IL-12-secreting dendritic cells (DC) are potent Th1-inducing APC. However, controversy exists concerning the function of DC subsets. Murine studies have suggested that CD8(+) DC preferentially induce Th1 responses, whereas CD8(-) DC induce Th2 development; in this model, different DC subsets prime different responses. Alternatively, the propensity of DC subsets to prime a Th1 response could depend upon the type of initial stimulus. We used a prototypic Th1-inducing adjuvant, heat-killed Brucella abortus (HKBA) to assess stimulation of DC subsets, relationship between Ag burden and IL-12 production, and down-regulation of DC subset IL-12 production by IL-10. In this study, we show that DC were sole producers of IL-12, although most HKBA uptake was by splenic macrophages and granulocytes. More CD8(-) than CD8(+) DC produced IL-12 after HKBA challenge, whereas only CD8(+) DC produced IL-12 after injection of another Th1-promoting microbial substance, soluble Toxoplasma gondii Ags. Studies in IL-10-deficient mice revealed that IL-10 down-regulates frequency and duration of IL-12 production by both DC subsets. In the absence of IL-10, IL-12 expression is enabled in CD11c(low) cells, but not in macrophages or granulocytes. These findings support the concept of DC as the major IL-12 producers in spleens, but challenge the notion that CD8(+) and CD8(-) DC are destined to selectively induce Th1 or Th2 responses, respectively. Thus, the nature of the stimulating substance is important in determining which DC subsets are activated to produce IL-12.
Collapse
Affiliation(s)
- L Y Huang
- Laboratory of Plasma Derivatives, Division of Hematology, Center for Biologics Evaluation and Research, Food and Drug Administration, Bethesda, MD 20892, USA
| | | | | | | | | |
Collapse
|
47
|
Yekebas EF, Eisenberger CF, Ohnesorge H, Saalmüller A, Elsner HA, Engelhardt M, Gillesen A, Meins J, The M, Strate T, Busch C, Knoefel WT, Bloechle C, Izbicki JR. Attenuation of sepsis-related immunoparalysis by continuous veno-venous hemofiltration in experimental porcine pancreatitis. Crit Care Med 2001; 29:1423-30. [PMID: 11445702 DOI: 10.1097/00003246-200107000-00021] [Citation(s) in RCA: 90] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OBJECTIVES In light of evidence suggesting that hemofiltration favorably influences septic diseases by removing sepsis mediators, the impact of different modalities of continuous veno-venous hemofiltration (CVVH) on outcome and immunologic derangements in porcine pancreatogenic sepsis was evaluated. DESIGN Randomized, controlled intervention trial. SUBJECTS Forty-eight minipigs of either sex. INTERVENTIONS Pancreatitis was induced by intraductal injection of sodium taurocholate (4%, 1 mL/kg body weight [BW]) and enterokinase (2 U/kg BW). Animals were allocated either to untreated controls-group 1-or to one of three treatment groups-group 2: low-volume CVVH (20 mL/kg BW), no change of hemofilters; group 3: low-volume CVVH, filters changed every 12 hrs; and group 4: high-volume CVVH (100 mL/kg BW), filters changed every 12 hrs. Survival represented the major parameter of the study. Serum cytokine levels, sepsis-related down-regulation of major histocompatibility complex II and CD14 expression on leukocytes, bacterial translocation, and endotoxemia were further parameters evaluated in the study. MEASUREMENTS AND MAIN RESULTS High-volume CVVH combined with periodic filter change was significantly superior compared with less intensive treatment modalities (low-volume CVVH, no filter change) in sepsis protection. Long-term survival (>60 hrs) was found in 67% of group 4 and 33% of group 3 animals (p <.05), whereas in controls and group 2 no animal survived. CVVH ameliorated the initial serum tumor necrosis factor-alpha response and prevented sepsis-induced in vitro endotoxin hyporesponsiveness. Down-regulation of major histocompatibility complex II and CD14 expression on monocytes was significantly improved by CVVH. Improved oxidative burst and phagocytosis capacity in polymorphonuclear leukocytes suggested that leukocyte function was stabilized by CVVH. Also, CVVH significantly reduced bacterial translocation and endotoxemia. CONCLUSIONS Hemofiltration reversed sepsis-induced immunoparalysis in a porcine model of bile acid-induced pancreatitis. Implications for human pancreatitis must be validated in prospective, clinical protocols.
Collapse
Affiliation(s)
- E F Yekebas
- Department of Surgery, University Hospital Eppendorf, Hamburg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Arad G, Hillman D, Levy R, Kaempfer R. Superantigen antagonist blocks Th1 cytokine gene induction and lethal shock. J Leukoc Biol 2001. [DOI: 10.1189/jlb.69.6.921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Gila Arad
- Department of Molecular Virology, The Hebrew University‐Hadassah Medical School, Jerusalem, Israel
| | - Dalia Hillman
- Department of Molecular Virology, The Hebrew University‐Hadassah Medical School, Jerusalem, Israel
| | - Revital Levy
- Department of Molecular Virology, The Hebrew University‐Hadassah Medical School, Jerusalem, Israel
| | - Raymond Kaempfer
- Department of Molecular Virology, The Hebrew University‐Hadassah Medical School, Jerusalem, Israel
| |
Collapse
|
49
|
Komisar JL, Weng CF, Oyejide A, Hunt RE, Briscoe C, Tseng J. Cellular and cytokine responses in the circulation and tissue reactions in the lung of rhesus monkeys (Macaca mulatta) pretreated with cyclosporin A and challenged with staphylococcal enterotoxin B. Toxicol Pathol 2001; 29:369-78. [PMID: 11442023 DOI: 10.1080/019262301316905336] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cyclosporin A (CsA), an inhibitor of T cell cytokine production, protects mice against staphylococcal enterotoxin B (SEB) intoxication. To determine whether CsA treatment would work in a species closer to humans. 4 rhesus monkeys were given 50 mg/kg CsA followed by an intratracheal challenge with approximately 6 LD50 of SEB. The CsA was not protective: one of the monkeys died and the other three had to be euthanised when they became moribund. All monkeys made IL-2, TNF, and IFN-gamma in response to SEB. In addition, there was about a 10-fold increase in ACTH levels 2 hr after SEB challenge. CsA significantly suppressed in vitro proliferation of lymphocytes from treated monkeys. Both CsA-treated monkeys and monkeys that had been challenged in a previous experiment with a lethal dose of SEB but had received no cyclosporin had pathologic changes in several organs. The most prominent changes were marked edema and leukocytic infiltration of the bronchial and bronchiolar mucosa. The CsA treatment appeared to reduce the intensity of lung inflammation, but this effect was not sufficient to protect the monkeys. The results suggest that CsA alone may not be an effective therapeutic agent for humans suffering from SEB intoxication or gram-positive septic shock.
Collapse
Affiliation(s)
- J L Komisar
- Department of Experimental Pathology, Walter Reed Army Institute of Research, Silver Spring, MD 20910-7500, USA.
| | | | | | | | | | | |
Collapse
|
50
|
Abstract
Interleukin-10 (IL-10), first recognized for its ability to inhibit activation and effector function of T cells, monocytes, and macrophages, is a multifunctional cytokine with diverse effects on most hemopoietic cell types. The principal routine function of IL-10 appears to be to limit and ultimately terminate inflammatory responses. In addition to these activities, IL-10 regulates growth and/or differentiation of B cells, NK cells, cytotoxic and helper T cells, mast cells, granulocytes, dendritic cells, keratinocytes, and endothelial cells. IL-10 plays a key role in differentiation and function of a newly appreciated type of T cell, the T regulatory cell, which may figure prominently in control of immune responses and tolerance in vivo. Uniquely among hemopoietic cytokines, IL-10 has closely related homologs in several virus genomes, which testify to its crucial role in regulating immune and inflammatory responses. This review highlights findings that have advanced our understanding of IL-10 and its receptor, as well as its in vivo function in health and disease.
Collapse
Affiliation(s)
- Kevin W. Moore
- Departments of Molecular Biology, DNAX Research Institute of Molecular and Cellular Biology Inc., Palo Alto, California 94304
- Departments of Pharmacology, DNAX Research Institute of Molecular and Cellular Biology Inc., Palo Alto, California 94304
- Departments of Immunology, DNAX Research Institute of Molecular and Cellular Biology Inc., Palo Alto, California 94304;,
| | - Rene de Waal Malefyt
- Departments of Molecular Biology, DNAX Research Institute of Molecular and Cellular Biology Inc., Palo Alto, California 94304
- Departments of Pharmacology, DNAX Research Institute of Molecular and Cellular Biology Inc., Palo Alto, California 94304
- Departments of Immunology, DNAX Research Institute of Molecular and Cellular Biology Inc., Palo Alto, California 94304;,
| | - Robert L. Coffman
- Departments of Molecular Biology, DNAX Research Institute of Molecular and Cellular Biology Inc., Palo Alto, California 94304
- Departments of Pharmacology, DNAX Research Institute of Molecular and Cellular Biology Inc., Palo Alto, California 94304
- Departments of Immunology, DNAX Research Institute of Molecular and Cellular Biology Inc., Palo Alto, California 94304;,
| | - Anne O'Garra
- Departments of Molecular Biology, DNAX Research Institute of Molecular and Cellular Biology Inc., Palo Alto, California 94304
- Departments of Pharmacology, DNAX Research Institute of Molecular and Cellular Biology Inc., Palo Alto, California 94304
- Departments of Immunology, DNAX Research Institute of Molecular and Cellular Biology Inc., Palo Alto, California 94304;,
| |
Collapse
|