Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
2.
J Neonatal Perinatal Med ; 13(4): 469-476, 2020.
Article in English | MEDLINE | ID: mdl-32444566

ABSTRACT

BACKGROUND: Hypothermia with xenon gas has been used to reduce brain injury and disability rate after perinatal hypoxia-ischemia. We evaluated xenon gas therapy effects in an in vitro model with or without hypothermia on cultured human airway epithelial cells (Calu-3). METHODS: Calu-3 monolayers were grown at an air-liquid interface and exposed to one of the following conditions: 1) 21% FiO2 at 37°C (control); 2) 45% FiO2 and 50% xenon at 37°C; 3) 21% FiO2 and 50% xenon at 32°C; 4) 45% FiO2 and 50% xenon at 32°C for 24 hours. Transepithelial resistance (TER) measurements were performed and apical surface fluids were collected and assayed for total protein, IL-6, and IL-8. Three monolayers were used for immunofluorescence localization of zonula occludens-1 (ZO-1). The data were analyzed by one-way ANOVA. RESULTS: TER decreased at 24 hours in all treatment groups. Xenon with hyperoxia and hypothermia resulted in greatest decrease in TER compared with other groups. Immunofluorescence localization of ZO-1 (XY) showed reduced density of ZO-1 rings and incomplete ring-like staining in the 45% FiO2- 50% xenon group at 32°C compared with other groups. Secretion of total protein was not different among groups. Secretion of IL-6 in 21% FiO2 with xenon group at 32°C was less than that of the control group. The secretion of IL-8 in 45% FiO2 with xenon at 32°C was greater than that of other groups. CONCLUSION: Hyperoxia and hypothermia result in detrimental epithelial cell function and inflammation over 24-hour exposure. Xenon gas did not affect cell function or reduce inflammation.


Subject(s)
Hyperoxia/immunology , Hypothermia/immunology , Hypoxia-Ischemia, Brain , Interleukin-6/immunology , Interleukin-8/immunology , Xenon/pharmacology , Anesthetics, Inhalation/pharmacology , Cells, Cultured , Humans , Hypoxia-Ischemia, Brain/immunology , Hypoxia-Ischemia, Brain/therapy , Inflammation , Inflammation Mediators/immunology , Respiratory Mucosa/drug effects , Respiratory Mucosa/physiology , Tight Junctions/physiology , Treatment Outcome
3.
Trends Endocrinol Metab ; 30(12): 875-878, 2019 12.
Article in English | MEDLINE | ID: mdl-31668960

ABSTRACT

Host defense relies not only on microbicidal mechanisms (resistance), but also on management of collateral damage (tolerance). Here, we discuss how this immunology concept converges with a physiology-born theory on the dichotomy of thermometabolic responses in infection (fever versus hypothermia), yielding a model of immunity that transcends discipline barriers.


Subject(s)
Immunity/physiology , Inflammation/immunology , Inflammation/physiopathology , Animals , Fever/immunology , Fever/metabolism , Fever/physiopathology , Humans , Hypothermia/immunology , Hypothermia/metabolism , Hypothermia/physiopathology , Inflammation/metabolism
4.
Mol Pharm ; 16(4): 1467-1476, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30821985

ABSTRACT

Peptides are generally needed as T-helper epitopes in nicotine vaccines to induce effective antibody responses, but the highly polymorphic property of major histocompatibility complex (MHC) molecules may limit opportunities of B cell to receive CD4+ T-cell help. Invariant natural killer T (iNKT) cells recognize lipid antigens presented by the nonpolymorphic CD1d molecule that is conserved in mammals to a great extent. iNKT cells also display some similar functions to conventional CD4+ T-helper cells, especially they license dendritic cells stimulate antibody isotype switching by B cells. Herein, α-galactosylceramide (αGalCer), a classical iNKT cell agonist, serves as an adjuvant in synthetic nicotine vaccine candidates absent of peptide or protein. Our study reveals that αGalCer displays better adjuvant activity than Pam3CSK4 (a commonly used lipopeptide TLR agonist). Remarkably, the covalent linker between the nicotine hapten and αGalCer is not critical. Self-assembly of the lipid-tailed nicotine and αGalCer into the liposome represents a structurally simple but immunologically effective way to develop nicotine vaccines. This is the first time to introduce the iNKT cell agonist as an adjuvant to an antidrug vaccine. This discovery may contribute to improving the efficacy of clinical candidate nicotine vaccines in the future.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Analgesics/administration & dosage , Antibodies, Monoclonal/immunology , Galactosylceramides/immunology , Hypothermia/drug therapy , Nicotine/administration & dosage , Vaccines, Synthetic/administration & dosage , Animals , Female , Galactosylceramides/metabolism , Hypothermia/immunology , Hypothermia/metabolism , Immunization , Lipopeptides/administration & dosage , Mice , Mice, Inbred BALB C , Natural Killer T-Cells/immunology , Natural Killer T-Cells/metabolism , Nicotine/immunology
5.
Ned Tijdschr Geneeskd ; 1632019 01 14.
Article in Dutch | MEDLINE | ID: mdl-30676705

ABSTRACT

BACKGROUND: Multiple sclerosis (MS) can be accompanied by paroxysmal symptoms. These are diverse in nature and often not well known. CASE DESCRIPTION: In the emergency department, we repeatedly saw a 49-year-old man with secondary progressive MS with recurrent episodes of hypothermia, loss of consciousness, thrombocytopenia and worsening of pre-existing neurological deficits. In some of these episodes we identified an underlying urinary tract infection, which we treated. After normalization of the body temperature, we observed complete recovery of the clinical signs and platelet count. CONCLUSION: Paroxysmal hypothermia can be a symptom of MS and is accompanied by a change in level of consciousness, increase of neurological deficits and haematological disturbances. Usually, there is full recovery after normalization of the body temperature and treatment of any underlying infections. Early recognition of these recurrent symptoms is important to prevent unnecessary diagnostics and overtreatment.


Subject(s)
Hypothermia/immunology , Multiple Sclerosis/complications , Body Temperature , Humans , Male , Middle Aged , Multiple Sclerosis/immunology , Recurrence , Urinary Tract Infections/immunology
6.
Pharm Res ; 35(9): 169, 2018 Jun 27.
Article in English | MEDLINE | ID: mdl-29951887

ABSTRACT

PURPOSE: Administration of therapeutic monoclonal antibodies (mAbs) is frequently accompanied by severe first infusion reactions (FIR). The mechanism driving FIR is still unclear. This study aimed to investigate the cellular and molecular mechanisms causing FIR in humanized mouse models and their potential for evaluating FIR risk in patients. METHODS: Mice humanized for Fc gamma receptors (FcγRs) were generated by recombination-mediated genomic replacement. Body temperature, cytokine release and reactive oxygen species (ROS) were measured to assess FIR to mAbs. RESULTS: Infusion of human mAb specific for mouse transferrin receptor (HamTfR) into FcγR-humanized mice, produced marked transient hypothermia accompanied by an increase in inflammatory cytokines KC and MIP-2, and ROS. FIR were dependent on administration route and Fc-triggered effector functions mediated by neutrophils. Human neutrophils also induced FIR in wild type mice infused with HamTfR. Specific knock-in mice demonstrated that human FcγRIIIb on neutrophils was both necessary and sufficient to cause FIR. FcγRIIIb-mediated FIR was abolished by depleting neutrophils or blocking FcγRIIIb with CD11b antibodies. CONCLUSIONS: Human FcγRIIIb and neutrophils are primarily responsible for triggering FIR. Clinical strategies to prevent FIR in patients should focus on this pathway and may include transient depletion of neutrophils or blocking FcγRIIIb with specific mAbs.


Subject(s)
Antibodies, Monoclonal/adverse effects , Hypothermia/chemically induced , Inflammation/chemically induced , Neutrophils/immunology , Receptors, IgG/immunology , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/immunology , GPI-Linked Proteins/genetics , GPI-Linked Proteins/immunology , Humans , Hypothermia/immunology , Inflammation/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neutrophils/drug effects , Receptors, IgG/genetics , Receptors, Transferrin/immunology
7.
PLoS One ; 13(5): e0197487, 2018.
Article in English | MEDLINE | ID: mdl-29791477

ABSTRACT

BACKGROUND: Oxazolone-induced colitis has been frequently used in literature as a model of IBD, but insights into the underlying immune response and pathological features are surprisingly still very limited. Vagus nerve stimulation (VNS) has proven to be effective in innate and Th1/Th17 predominant inflammatory models, including pre-clinical models of colitis, however to what extent VNS is also effective in ameliorating Th2-driven colitis remains to be studied. In the present study, we therefore further characterized the immune response in oxazolone-induced colitis and investigated the potential therapeutic effect of VNS. METHODS: Colitis was induced in Balb/c mice by cutaneous sensitization with 3% oxazolone followed by intracolonic administration of 1% oxazolone 7 days later. To evaluate the effect of VNS on the development of Th2-driven colitis, VNS and sham-treated mice were challenged with 1% oxazolone. RESULTS: Intracolonic oxazolone administration resulted in a severe destruction of the colonic mucosa and a rapid drop in body temperature leading to a 65% mortality rate at day 5. Severe infiltration of neutrophils and monocytes was detected 6h after oxazolone administration which was associated with a Th2-type inflammatory response. VNS significantly improved survival rate which correlated with decreased levels of HMGB1 and reduced colonic (il6 and cxcl1 mRNA) and serum cytokine levels (IL-6, TNFα and CXCL1) compared to sham treated mice. CONCLUSIONS: Oxazolone-induced colitis rather represents a model of sepsis and, at best, may resemble a severe type of ulcerative colitis, associated with early and severe mucosal damage and a high mortality rate. VNS reduces colonic inflammation and improves survival in this model, supporting the anti-inflammatory properties of VNS, even in an aggressive model as oxazolone-induced colitis.


Subject(s)
Colitis/physiopathology , Colitis/therapy , Vagus Nerve Stimulation , Animals , Colitis/chemically induced , Colitis/immunology , Cytokines/metabolism , Disease Models, Animal , Female , Hypothermia/complications , Hypothermia/immunology , Hypothermia/pathology , Hypothermia/physiopathology , Inflammation/complications , Inflammation/pathology , Intestinal Mucosa/pathology , Mice, Inbred BALB C , Natural Killer T-Cells/immunology , Oxazolone , Survival Analysis
8.
J Allergy Clin Immunol ; 139(2): 584-596.e10, 2017 02.
Article in English | MEDLINE | ID: mdl-27555460

ABSTRACT

BACKGROUND: Conflicting results have been obtained regarding the roles of Fc receptors and effector cells in models of active systemic anaphylaxis (ASA). In part, this might reflect the choice of adjuvant used during sensitization because various adjuvants might differentially influence the production of particular antibody isotypes. OBJECTIVE: We developed an "adjuvant-free" mouse model of ASA and assessed the contributions of components of the "classical" and "alternative" pathways in this model. METHODS: Mice were sensitized intraperitoneally with ovalbumin at weekly intervals for 6 weeks and challenged intraperitoneally with ovalbumin 2 weeks later. RESULTS: Wild-type animals had immediate hypothermia and late-phase intraperitoneal inflammation in this model. These features were reduced in mice lacking the IgE receptor FcεRI, the IgG receptor FcγRIII or the common γ-chain FcRγ. FcγRIV blockade resulted in a partial reduction of inflammation without any effect on hypothermia. Depletion of monocytes/macrophages with clodronate liposomes significantly reduced the hypothermia response. By contrast, depletion of neutrophils or basophils had no significant effects in this ASA model. Both the hypothermia and inflammation were dependent on platelet-activating factor and histamine and were reduced in 2 types of mast cell (MC)-deficient mice. Finally, engraftment of MC-deficient mice with bone marrow-derived cultured MCs significantly exacerbated the hypothermia response and restored inflammation to levels similar to those observed in wild-type mice. CONCLUSION: Components of the classical and alternative pathways contribute to anaphylaxis in this adjuvant-free model, with key roles for MCs and monocytes/macrophages.


Subject(s)
Anaphylaxis/immunology , Cell Movement , Hypothermia/immunology , Leukocytes/immunology , Macrophages/immunology , Mast Cells/immunology , Adjuvants, Immunologic , Animals , Cells, Cultured , Complement Pathway, Alternative , Complement Pathway, Classical , Disease Models, Animal , Humans , Immunization , Mast Cells/transplantation , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, IgE/genetics , Receptors, IgE/metabolism , Receptors, IgG/genetics , Receptors, IgG/metabolism
9.
Crit Care ; 20(1): 328, 2016 10 14.
Article in English | MEDLINE | ID: mdl-27737683

ABSTRACT

BACKGROUND: Hypothermia is associated with adverse outcome in patients with sepsis. The objective of this study was to characterize the host immune response in patients with hypothermic sepsis in order to determine if an excessive anti-inflammatory response could explain immunosuppression and adverse outcome. Markers of endothelial activation and integrity were also measured to explore potential alternative mechanisms of hypothermia. Finally we studied risk factors for hypothermia in an attempt to find new clues to the etiology of hypothermia in sepsis. METHODS: Consecutive patients diagnosed with sepsis within 24 hours after admission to ICUs in two tertiary hospitals in the Netherlands were included in the study (n = 525). Hypothermia was defined as body temperature below 36 °C in the first 24 h of ICU admission. RESULTS: Hypothermia was identified in 186 patients and was independently associated with mortality. Levels of proinflammatory and anti-inflammatory cytokines were not different between groups. Hypothermia was also not associated with an altered response to ex vivo stimulation with lipopolysaccharide in a subset of 15 patients. Risk factors for hypothermia included low body mass index, hypertension and chronic cardiovascular insufficiency. Levels of the endothelial activation marker fractalkine were increased during the first 4 days of ICU stay. CONCLUSIONS: Hypothermia during sepsis is independently associated with mortality, which cannot be attributed to alterations in the host immune responses that were measured in this study. Given that risk factors for hypothermic sepsis are mainly cardiovascular and that the endothelial activation marker fractalkine increased in hypothermia, these findings may suggest that vascular dysfunction plays a role in hypothermic sepsis.


Subject(s)
Hospital Mortality/trends , Hypothermia/immunology , Hypothermia/mortality , Immunity, Cellular/immunology , Sepsis/immunology , Sepsis/mortality , Aged , Body Temperature/immunology , Female , Humans , Hypothermia/diagnosis , Inflammation Mediators/immunology , Intensive Care Units/trends , Male , Middle Aged , Netherlands/epidemiology , Risk Factors , Sepsis/diagnosis , Treatment Outcome
10.
Endocr Regul ; 50(3): 137-44, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27560796

ABSTRACT

OBJECTIVE: Continuous exposure to cold leads to an activation of adaptive thermogenesis in the brown adipose tissue and induction of brown/beige cell phenotype in the white adipose tissue. Thermogenic response is associated with alternatively activated macrophages producing catecholamines, which subsequently activate the uncoupling protein 1 (UCP-1). The aim of this work was to elucidate the effect of cold exposure on catecholamine and immune responses associated with adipocyte browning in the mesenteric adipose tissue (mWAT) of rat. METHODS: The rats were exposed to continuous cold (4 °C) for 1 or 7 days. Catecholamines production and gene expressions of inflammatory and other factors, related to adipocyte "browning", were analyzed in the homogenized mWAT samples using 2-CAT ELISA kits. RESULTS: Cold exposure induced a sympathetic response in the mWAT, evidenced by the tyrosine hydroxylase (TH) protein level rise. Induction of non-sympathetical catecholamine production was observed 7 days after cold exposure by elevated TH and phenylethanolamine-N-methyltransferase (PNMT) expression, leading to an increased epinephrine levels. Cold exposure for 7 days stimulated the infiltration of macrophages, evaluated by F4/80 and CD68 expressions, and expression of anti-inflammatory mediators, while pro-inflammatory cytokines were inhibited. Anti- inflammatory response, accompanied by de novo catecholamine production and up-regulation of ß3-adrenergic receptors, led to the stimulation of UCP-1 and PGC1α expression, suggesting a cold-induced "browning" of the mWAT, mediated by alternatively activated macrophages. CONCLUSIONS: The present data indicate that prolonged cold exposure may induce anti-inflammatory response in mWAT associated with induction of UCP-1 expression. Although functional thermogenesis in the mWAT is most likely redundant, a highly efficient dissipation of energy by UCP1 may affect the energy homeostasis in this visceral fat.


Subject(s)
Adipose Tissue, Beige/metabolism , Catecholamines/metabolism , Cold Temperature , Cytokines/metabolism , Hypothermia/metabolism , Inflammation Mediators/metabolism , Inflammation/prevention & control , Intra-Abdominal Fat/metabolism , Thermogenesis , Uncoupling Protein 1/metabolism , Adaptation, Physiological , Adipose Tissue, Beige/immunology , Adipose Tissue, Beige/physiopathology , Animals , Cytokines/genetics , Cytokines/immunology , Disease Models, Animal , Energy Metabolism , Gene Expression Regulation , Hypothermia/immunology , Hypothermia/physiopathology , Inflammation/immunology , Inflammation/metabolism , Inflammation/physiopathology , Inflammation Mediators/immunology , Intra-Abdominal Fat/immunology , Intra-Abdominal Fat/physiopathology , Male , Mesentery , Phenotype , Rats, Sprague-Dawley , Signal Transduction , Time Factors , Uncoupling Protein 1/genetics
11.
Q Rev Biol ; 91(1): 25-46, 2016 Mar.
Article in English | MEDLINE | ID: mdl-27192778

ABSTRACT

Fever, the rise in body temperature set point in response to infection or injury, is a highly conserved trait among vertebrates, and documented in many arthropods. Fever is known to reduce illness duration and mortality. These observations present an evolutionary puzzle: why has fever continued to be an effective response to fast-evolving pathogenic microbes across diverse phyla, and probably over countless millions of years? Framing fever as part of a more general thermal manipulation strategy that we term defensive hyperthermia, we hypothesize that the solution lies in the independent contributions to pathogen fitness played by virulence and infectivity. A host organism deploying defensive hyperthermia alters the ecological environment of an invading pathogen. To the extent that the pathogen evolves to be able to function effectively at elevated temperatures, it disadvantages itself at infecting the next (thermonormative) host, becoming more likely to be thwarted by that host's immune system and outcompeted by wild ecotype conspecifics (a genetically distinct strain adapted to specific environmental conditions) that, although more vulnerable to elevated temperatures, operate more effectively at the host's normal temperature. We evaluate this hypothesis in light of existing evidence concerning pathogen thermal specialization, and discuss theoretical and translational implications of this model.


Subject(s)
Bacterial Infections/immunology , Biological Evolution , Fever/genetics , Animals , Fever/immunology , Fever/metabolism , Humans , Hypothermia/immunology , Immunity, Innate
12.
J Allergy Clin Immunol ; 137(4): 1197-1205, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26581914

ABSTRACT

BACKGROUND: Mast cells are indispensable for LPS-induced septic hypothermia, in which TNF-α plays an essential role to initiate septic responses. ITK and BTK regulate mast cell responses to allergens, but their roles in mast cell responses in LPS-induced sepsis are unclear. OBJECTIVE: We sought to investigate the roles of ITK and BTK in mast cell responses during LPS-induced septic inflammation. METHODS: Mice (genetically modified or bone marrow-derived mast cell-reconstituted Sash) were given LPS to induce septic hypothermia in the presence or absence of indicated inhibitors. Flow cytometry was used to determine LPS-induced cell influx and TNF-α production in peritoneal cells. Microarray was used for genomewide gene expression analysis on bone marrow-derived mast cells. Quantitative PCR and multiplex were used to determine transcribed and secreted proinflammatory cytokines. Microscopy and Western blotting were used to determine activation of signal transduction pathways. RESULTS: The absence of ITK and BTK leads to exacerbation of LPS-induced septic hypothermia and neutrophil influx. Itk(-/-)Btk(-/-) mast cells exhibit hyperactive preformed and LPS-induced TNF-α production, and lead to more severe LPS-induced septic hypothermia when reconstituted into mast cell-deficient Sash mice. LPS-induced nuclear factor kappa B, Akt, and p38 activation is enhanced in Itk(-/-)Btk(-/-) mast cells, and blockage of phosphatidylinositol-4,5-bisphosphate 3-kinase, Akt, or p38 downstream mitogen-activated protein kinase interacting serine/threonine kinase 1 activation significantly suppresses TNF-α hyperproduction and attenuates septic hypothermia. CONCLUSIONS: ITK and BTK regulate thermal homeostasis during septic response through mast cell function in mice. They share regulatory function downstream of Toll-like receptor 4/LPS in mast cells, through regulating the activation of canonical nuclear factor kappa B, phosphatidylinositol-4,5-bisphosphate 3-kinase/Akt, and p38 signaling pathways.


Subject(s)
Hypothermia/immunology , Lipopolysaccharides/immunology , Mast Cells/immunology , Protein-Tyrosine Kinases/immunology , Sepsis/immunology , Agammaglobulinaemia Tyrosine Kinase , Animals , Biomarkers/metabolism , Blotting, Western , Cytokines/metabolism , Hypothermia/etiology , Mast Cells/metabolism , Mice , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Protein-Tyrosine Kinases/metabolism , Sepsis/complications
13.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 31(11): 1479-82, 1487, 2015 Nov.
Article in Chinese | MEDLINE | ID: mdl-26522355

ABSTRACT

OBJECTIVE: To investigate the impact of mild hypothermia on the lipopolysaccharide (LPS)-induced expressions of Toll-like receptor 4 (TLR4), nuclear factor κB (NF-κB) and downstream inflammatory cytokines in BV-2 microglias. METHODS: BV-2 cells cultured in vitro were divided into 33 Degrees Celsius-PBS, 33 Degrees Celsius-LPS, 37 Degrees Celsius-PBS and 37 Degrees Celsius-LPS groups. Real-time quantitative PCR was performed to detect the mRNA expressions of TLR4 and NF-κB in BV-2 cells, Western blotting was performed to detect the protein expressions of TLR4 and NF-κB in BV-2 cells, and ELISA to detect the levels of tumor necrosis factor α (TNF-α) and interleukin-1ß (IL-1ß) in the culture medium. RESULTS: After LPS stimulated BV-2 cells, TLR4 pathway protein exhibited the trend of increasing firstly and decreasing afterwards, while the expression of NF-κB protein in the pathway downstream continued to increase, and the release of inflammatory cytokines TNF-α and IL-1ß were significantly promoted. Mild hypothermia could significantly inhibit the transcription and expressions of TLR4 and NF-κB as well as the release of inflammatory cytokines. CONCLUSION: The mild hypothermia could inhibit the LPS/TLR4 pathway in BV-2 cells, and depress the expressions of TNF-α and IL-1ß.


Subject(s)
Cytokines/analysis , Hypothermia/immunology , Lipopolysaccharides/pharmacology , Microglia/immunology , Toll-Like Receptor 4/physiology , Animals , Cells, Cultured , Cytokines/genetics , Interleukin-1beta/analysis , Mice , NF-kappa B/genetics , Toll-Like Receptor 4/genetics , Tumor Necrosis Factor-alpha/analysis
14.
Infect Immun ; 83(12): 4693-700, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26416909

ABSTRACT

Although Leptospira can infect a wide range of mammalian species, most studies have been conducted in golden Syrian hamsters, a species particularly sensitive to acute disease. Chronic disease has been well characterized in the rat, one of the natural reservoir hosts. Studies in another asymptomatic reservoir host, the mouse, have occasionally been done and have limited infection to mice younger than 6 weeks of age. We analyzed the outcome of sublethal infection of C3H/HeJ mice older than age 10 weeks with Leptospira interrogans serovar Copenhageni. Infection led to bloodstream dissemination of Leptospira, which was followed by urinary shedding, body weight loss, hypothermia, and colonization of the kidney by live spirochetes 2 weeks after infection. In addition, Leptospira dissemination triggered inflammation in the kidney but not in the liver or lung, as determined by increased levels of mRNA transcripts for the keratinocyte-derived chemokine, RANTES, macrophage inflammatory protein 2, tumor necrosis factor alpha, interleukin-1ß, inducible nitric oxide synthase, interleukin-6, and gamma interferon in kidney tissue. The acquired humoral response to Leptospira infection led to the production of IgG mainly of the IgG1 subtype. Flow cytometric analysis of splenocytes from infected mice revealed that cellular expansion was primarily due to an increase in the levels of CD4(+) and double-negative T cells (not CD8(+) cells) and that CD4(+) T cells acquired a CD44(high) CD62L(low) effector phenotype not accompanied by increases in memory T cells. A mouse model for sublethal Leptospira infection allows understanding of the bacterial and host factors that lead to immune evasion, which can result in acute or chronic disease or resistance to infection (protection).


Subject(s)
Bacteriuria/immunology , Disease Models, Animal , Kidney/immunology , Leptospira interrogans/immunology , Leptospirosis/immunology , Mice/immunology , Animals , Bacteremia/genetics , Bacteremia/immunology , Bacteremia/microbiology , Bacteremia/pathology , Bacteriuria/genetics , Bacteriuria/microbiology , Bacteriuria/pathology , CD4-Positive T-Lymphocytes , Chemokine CCL5/genetics , Chemokine CCL5/immunology , Chemokine CXCL2/genetics , Chemokine CXCL2/immunology , Chemokines/genetics , Chemokines/immunology , Chronic Disease , Female , Gene Expression Regulation , Host-Pathogen Interactions , Hypothermia/genetics , Hypothermia/immunology , Hypothermia/microbiology , Hypothermia/pathology , Immunoglobulin G/biosynthesis , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Kidney/microbiology , Kidney/pathology , Leptospirosis/genetics , Leptospirosis/microbiology , Leptospirosis/pathology , Mice/genetics , Mice/microbiology , Mice, Inbred C3H , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/immunology , Signal Transduction , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Weight Loss/immunology
15.
Am J Emerg Med ; 33(10): 1430-5, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26275630

ABSTRACT

PURPOSE: This study aimed to investigate the influence of mild hypothermia on the number of CD11b+ Gr-1+ myeloid-derived suppressor cells (MDSCs) induced by lipopolysaccharide (LPS) injection in mice with sepsis. METHODS: BALB/c mice were administered LPS to establish a mouse model of sepsis. Then, these mice were randomly divided into 3 groups: the mild hypothermia plus LPS group, the normothermia plus LPS group, and the LPS group. The normal control group was injected the same amount of 0.9% sodium chloride solution. The ratio of CD11b+ Gr-1+ MDSCs in the mouse spleen and bone marrow was determined at 6, 12, 24, 48, and 72 hours after LPS injection and after injected 0.9% sodium chloride solution. RESULTS: Compared with the control group, the number of MDSCs in the spleen in the sepsis group increased gradually, and the difference was significant at 12 hours after injection (P<.01). Moreover, the number of MDSCs was the lowest in the mild hypothermia group, and there was a significant difference than the other 2 groups at 48 hours (P<.01). The number of MDSCs in the bone marrow increased gradually, and the difference between the sepsis and control groups was significant at 24 hours (P<.01). The number of MDSCs in the mild hypothermia group was the lowest, and there was a statistically significant difference than the other 2 groups (P<.05). CONCLUSION: Mild hypothermia inhibited the production and accumulation of MDSCs induced by LPS administration in septic mice.


Subject(s)
Cell Proliferation/drug effects , Hypothermia/immunology , Lipopolysaccharides/immunology , Myeloid Cells/drug effects , Sepsis/immunology , Animals , Bone Marrow Cells/drug effects , CD11b Antigen/analysis , China , Disease Models, Animal , Escherichia coli , Flow Cytometry , Lipopolysaccharides/administration & dosage , Lipopolysaccharides/adverse effects , Male , Mice , Mice, Inbred BALB C , Receptors, Chemokine/analysis , Spleen/cytology , Spleen/drug effects , Survival Rate
16.
Surgery ; 158(3): 646-54, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26054320

ABSTRACT

INTRODUCTION: Hypothermia is a well-known risk factor for postoperative complications because it prolongs the monocyte inflammatory response. The purpose of this study was to investigate whether temperature-activated ion channels (transient receptor protein channels [TRP] A1 and V1) mediate the effects of temperature on monocytes. METHODS: Primary human monocytes were isolated and stimulated with lipopolysaccharide at 32°C or 39°C. RNA was isolated for analysis of microRNA (miR)-155 expression, and cytokines in the supernatant were measured with an enzyme-linked immunosorbent assay. Specific inhibitors of TRPA1 (HC- 030031) and a specific activator of TRPV1 (capsaicin) were used to block or activate TRPA1 and TRPV1, respectively. Statistical analysis was performed using the Wilcoxon signed-rank test. RESULTS: TRPM8 mRNA was not expressed in primary human monocytes, whereas TRPA1 and TRPV1 were expressed. TRPV1 mRNA expression was suppressed at 32°C but not at 39°C. TRPA1 was induced strongly at 32°C and 39°C. Immunofluorescence microscopy confirmed that monocytes express TRPA1 and TRPV1 on their cell surface. Interleukin-10 secretion was increased by blocking TRPA1 (77.8 ± 3 2.8 pg/mL) and activating TRPA1 (79.4 ± 16.1 pg/mL) after 24 hours at 32°C (control 37.4 ± 17.1 pg/mL, P < .05). At 36 hours, tumor necrosis factor secretion was decreased after TRPA1 blockade (2,321 ± 439 pg/mL) and TRPV1 activation (2,137 ± 411 pg/mL) compared with control (2,567 ± 495 pg/mL, P < .05). Furthermore, miR-155 expression also was suppressed at 24 hours by TRPA1 blockade and TRPV1 activation (both P < .05). Silencing of TRPA1 normalized monocyte IL-10 secretion at 32°C. CONCLUSION: These results demonstrate that hypothermia mediates its effects on monocytes through TRPA1. Blockade of TRPA1 or activation of TRPV1 may be used to modify the effects of hypothermia on the monocyte inflammatory response.


Subject(s)
Calcium Channels/metabolism , Cold Temperature/adverse effects , Hypothermia/immunology , Monocytes/metabolism , Nerve Tissue Proteins/metabolism , TRPV Cation Channels/metabolism , Transient Receptor Potential Channels/metabolism , Biomarkers/metabolism , Blotting, Western , Cells, Cultured , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Humans , Hypothermia/metabolism , Microscopy, Fluorescence , Nerve Tissue Proteins/antagonists & inhibitors , TRPA1 Cation Channel , TRPV Cation Channels/antagonists & inhibitors , Transient Receptor Potential Channels/antagonists & inhibitors
17.
J Neuroimmune Pharmacol ; 10(2): 364-70, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25715681

ABSTRACT

Cannabinoid receptor agonists, such as Δ(9)-THC, the primary active constituent of Cannabis sativa, have anti-pyrogenic effects in a variety of assays. Recently, attention has turned to the endogenous cannabinoid system and how endocannabinoids, including 2-arachidonoylglycerol (2-AG) and anandamide, regulate multiple homeostatic processes, including thermoregulation. Inhibiting endocannabinoid catabolic enzymes, monoacylglycerol lipase (MAGL) or fatty acid amide hydrolase (FAAH), elevates levels of 2-AG or anandamide in vivo, respectively. The purpose of this experiment was to test the hypothesis that endocannabinoid catabolic enzymes function to maintain thermal homeostasis in response to hypothermic challenge. In separate experiments, male C57BL/6J mice were administered a MAGL or FAAH inhibitor, and then challenged with the bacterial endotoxin lipopolysaccharide (LPS; 2 mg/kg ip) or a cold (4 °C) ambient environment. Systemic LPS administration caused a significant decrease in core body temperature after 6 h, and this hypothermia persisted for at least 12 h. Similarly, cold environment induced mild hypothermia that resolved within 30 min. JZL184 exacerbated hypothermia induced by either LPS or cold challenge, both of which effects were blocked by rimonabant, but not SR144528, indicating a CB1 cannabinoid receptor mechanism of action. In contrast, the FAAH inhibitor, PF-3845, had no effect on either LPS-induced or cold-induced hypothermia. These data indicate that unlike direct acting cannabinoid receptor agonists, which elicit profound hypothermic responses on their own, neither MAGL nor FAAH inhibitors affect normal body temperature. However, these endocannabinoid catabolic enzymes play distinct roles in thermoregulation following hypothermic challenges.


Subject(s)
Amidohydrolases/physiology , Body Temperature Regulation/immunology , Endocannabinoids/immunology , Environment , Homeostasis/immunology , Monoacylglycerol Lipases/physiology , Amidohydrolases/antagonists & inhibitors , Animals , Body Temperature Regulation/drug effects , Endocannabinoids/metabolism , Enzyme Inhibitors/pharmacology , Homeostasis/drug effects , Hypothermia/chemically induced , Hypothermia/enzymology , Hypothermia/immunology , Lipopolysaccharides/toxicity , Male , Metabolism/drug effects , Metabolism/immunology , Mice , Mice, Inbred C57BL , Monoacylglycerol Lipases/antagonists & inhibitors
18.
J Allergy Clin Immunol ; 135(6): 1625-35.e5, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25556996

ABSTRACT

BACKGROUND: Remodeling of quiescent vessels with increases in permeability, vasodilatation, and edema are hallmarks of inflammatory disorders. Factors involved in this type of remodeling represent potential therapeutic targets. OBJECTIVES: We investigated whether the nuclear hormone receptor peroxisome proliferator-activated receptor (PPAR) ß/δ, a regulator of metabolism, fibrosis, and skin homeostasis, is involved in regulation of this type of remodeling. METHODS: Wild-type and various Pparb/d mutant mice were used to monitor dermal acute vascular hyperpermeability (AVH) and passive systemic anaphylaxis-induced hypothermia and edema. PPARß/δ-dependent kinase activation and remodeling of endothelial cell-cell junctions were addressed by using human endothelial cells. RESULTS: AVH and dilatation of dermal microvessels stimulated by vascular endothelial growth factor A, histamine, and thrombin are severely compromised in PPARß/δ-deficient mice. Selective deletion of the Pparb/d-encoding gene in endothelial cells in vivo similarly limits dermal AVH and vasodilatation, providing evidence that endothelial PPARß/δ is the major player in regulating acute dermal microvessel remodeling. Furthermore, endothelial PPARß/δ regulatory functions are not restricted to the skin vasculature because its deletion in the endothelium, but not in smooth muscle cells, also leads to reduced systemic anaphylaxis, the most severe form of allergic reaction, in which an acute vascular response plays a key role. PPARß/δ-dependent AVH activation likely involves the activation of mitogen-activated protein kinase and Akt pathways and leads to downstream destabilization of endothelial cell-cell junctions. CONCLUSION: These results unveil not only a novel function of PPARß/δ as a direct regulator of acute vessel permeability and dilatation but also provide evidence that antagonizing PPARß/δ represents an important strategy to consider for moderating diseases with altered endothelial integrity, such as acute inflammatory and allergic disorders.


Subject(s)
Anaphylaxis/immunology , Capillary Permeability/immunology , Endothelial Cells/immunology , PPAR delta/immunology , PPAR-beta/immunology , Skin/immunology , Anaphylaxis/genetics , Anaphylaxis/pathology , Animals , Capillary Permeability/drug effects , Edema/genetics , Edema/immunology , Edema/pathology , Endothelial Cells/drug effects , Endothelial Cells/pathology , Female , Gene Expression Regulation , Histamine/pharmacology , Hypothermia/genetics , Hypothermia/immunology , Hypothermia/pathology , Intercellular Junctions/drug effects , Intercellular Junctions/immunology , Intercellular Junctions/pathology , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/immunology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/immunology , Myocytes, Smooth Muscle/pathology , PPAR delta/deficiency , PPAR delta/genetics , PPAR-beta/deficiency , PPAR-beta/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/immunology , Signal Transduction , Skin/blood supply , Skin/drug effects , Skin/pathology , Thrombin/pharmacology , Vascular Endothelial Growth Factor A/pharmacology
19.
Exp Hematol ; 42(10): 919-23.e1, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25201754

ABSTRACT

MicroRNAs (miRNAs) are small, noncoding RNAs that have been shown to play a critical role in normal physiology and disease, such as hematopoietic development and cancer. However, their role in mast-cell function and development is poorly understood. The major objective of this study was to determine how global miRNA expression affects mast-cell physiology. The RNase III endonuclease, Dicer, is required for the processing of pre-miRNAs into mature miRNAs. To investigate the effect of global miRNA depletion on mast cells in vivo, we generated a mast-cell-specific knock out of Dicer in mice. Transgenic mice (Mcpt5-Cre) that express Cre selectively in connective tissue mast cells were crossed with mice carrying the floxed conditional Dicer allele (Dicer fl/fl). Mcpt5-Cre × Dicer fl/fl mice with homozygous Dicer gene deletion in mast cells were found to have a profound mast-cell deficiency with near complete loss of peritoneal, gastrointestinal, and skin mast cells. We examined the in vivo functional consequence of mast-cell-specific Dicer deletion using an immunoglobulin-E-dependent passive systemic anaphylaxis murine model. Immunoglobulin-E-sensitized wild type Mcpt5-Cre × Dicer +/+ and heterozygous Mcpt5-Cre × Dicer fl/+ mice show marked hypothermia with antigen; however, homozygous Mcpt5-Cre × Dicer fl/fl mice were completely unresponsive to antigen challenge. These studies suggest a critical role for Dicer and miRNA expression for establishment of tissue compartments of functional mast cells in vivo.


Subject(s)
DEAD-box RNA Helicases/physiology , Mast Cells/cytology , MicroRNAs/genetics , Ribonuclease III/physiology , Anaphylaxis/immunology , Animals , Cell Count , Crosses, Genetic , DEAD-box RNA Helicases/deficiency , DEAD-box RNA Helicases/genetics , Gene Expression Regulation , Genotype , Humans , Hypothermia/immunology , Immunization , Mast Cells/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Organ Specificity , Ovalbumin/immunology , Ovalbumin/toxicity , Peritoneum/immunology , Peritoneum/pathology , Ribonuclease III/deficiency , Ribonuclease III/genetics , Serum Albumin/immunology , Skin/immunology , Skin/pathology , Stomach/immunology , Stomach/pathology
20.
Cell Immunol ; 289(1-2): 97-105, 2014.
Article in English | MEDLINE | ID: mdl-24751884

ABSTRACT

IgG-induced passive systemic anaphylaxis (PSA), a serious adverse effect of passive immune therapy using therapeutic monoclonal antibodies, has been greatly emphasized. However, controversy exists regarding the type of effector cells involved in IgG-induced anaphylaxis as a result of the induction of PSA by different IgG subtypes or the use of mice with varying genetic backgrounds. To clarify the effector cells for PSA, the PSA model with serious hypothermia was established by IgG monoclonal antibody (mAb) against natural protein or complete antigen, not hapten conjugate, in BALB/c and C57BL/6 mice. The results indicated that PSA could be remarkably inhibited by the depletion of macrophages but not by the depletion of whole leukocytes, basophils, neutrophils or monocytes. We further confirmed that macrophages are indispensable for the PSA induced by all six IgG-natural antigen complexes in both strains of mice. Additionally, platelet-activating factor (PAF) was found to be the major effector mediator for IgG-induced anaphylaxis. Moreover, gene knock-out of the third component of complement (C3) did not affect PSA-related hypothermia in C57BL/6 mice. These results indicate that macrophages and PAF act as dominant effector cells and mediator molecules, respectively, and are indispensable components in the induction of IgG-mediated PSA induced by IgG mAb and natural protein antigen. Based on the above results, we hypothesize that inconsistencies in effector cells for PSA may be associated with different features of the mAb-antigen system that might affect the magnitude of FcγRs cross-linking on effector cells.


Subject(s)
Anaphylaxis/immunology , Immunoglobulin G/immunology , Macrophages/immunology , Platelet Activating Factor/immunology , Animals , Antibodies, Monoclonal/immunology , Basophils/immunology , Complement C3/genetics , Female , Hypothermia/immunology , Immunization, Passive/adverse effects , Leukocytes, Mononuclear/immunology , Mast Cells/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/immunology , Platelet-Derived Growth Factor/immunology , Receptors, IgG/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...