Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 12 de 12
1.
Shock ; 49(4): 466-473, 2018 04.
Article En | MEDLINE | ID: mdl-28682939

BACKGROUND: A complete understanding of the role of the liver in burn-induced hypermetabolism is lacking. We investigated the acute effect of severe burn trauma on liver mitochondrial respiratory capacity and coupling control as well as the signaling events underlying these alterations. METHODS: Male BALB/c mice (8-12 weeks) received full-thickness scald burns on ∼30% of the body surface. Liver tissue was harvested 24 h postinjury. Mitochondrial respiration was determined by high-resolution respirometry. Citrate synthase activity was determined as a proxy of mitochondrial density. Male Sprague-Dawley rats received full-thickness scald burns to ∼60% of the body surface. Serum was collected 24 h postinjury. HepG2 cells were cultured with serum-enriched media from either sham- or burn-treated rats. Protein levels were analyzed via western blot. RESULTS: Mass-specific (P = 0.01) and mitochondrial-specific (P = 0.01) respiration coupled to ATP production significantly increased in the liver after burn. The respiratory control ratio for ADP (P = 0.04) and the mitochondrial flux control ratio (P = 0.03) were elevated in the liver of burned animals. Complex III and Complex IV protein abundance in the liver increased after burn by 17% and 14%, respectively. Exposure of HepG2 cells to serum from burned rats increased the pAMPKα:AMPKα ratio (P < 0.001) and levels of SIRT1 (P = 0.01), Nrf2 (P < 0.001), and PGC1α (P = 0.02). CONCLUSIONS: Severe burn trauma augments respiratory capacity and function of liver mitochondria, adaptations that augment ATP production. This response may be mediated by systemic factors that activate signaling proteins responsible for regulating cellular energy metabolism and mitochondrial biogenesis.


Burns/metabolism , Mitochondria, Liver/metabolism , Mitochondria/metabolism , Animals , Citrate (si)-Synthase/metabolism , Electron Transport/physiology , Hep G2 Cells , Hepatocytes/metabolism , Humans , Male , Mice , Mice, Inbred BALB C , Rats , Rats, Sprague-Dawley
2.
Crit Care ; 18(3): R113, 2014 Jun 02.
Article En | MEDLINE | ID: mdl-24890566

INTRODUCTION: The chemokine CXCL10 is produced during infection and inflammation to activate the chemokine receptor CXCR3, an important regulator of lymphocyte trafficking and activation. The goal of this study was to assess the contributions of CXCL10 to the pathogenesis of experimental septic shock in mice. METHODS: Septic shock was induced by cecal ligation and puncture (CLP) in mice resuscitated with lactated Ringer's solution and, in some cases, the broad spectrum antibiotic Primaxin. Studies were performed in CXCL10 knockout mice and mice treated with anti-CXCL10 immunoglobulin G (IgG). Endpoints included leukocyte trafficking and activation, core body temperature, plasma cytokine concentrations, bacterial clearance and survival. RESULTS: CXCL10 was present at high concentrations in plasma and peritoneal cavity during CLP-induced septic shock. Survival was significantly improved in CXCL10 knockout (CXCL10KO) mice and mice treated with anti-CXCL10 IgG compared to controls. CXCL10KO mice and mice treated with anti-CXCL10 IgG showed attenuated hypothermia, lower concentrations of interleukin-6 (IL-6) and macrophage inhibitory protein-2 (MIP-2) in plasma and lessened natural killer (NK) cell activation compared to control mice. Compared to control mice, bacterial burden in blood and lungs was lower in CXCL10-deficient mice but not in mice treated with anti-CXCL10 IgG. Treatment of mice with anti-CXCL10 IgG plus fluids and Primaxin at 2 or 6 hours after CLP significantly improved survival compared to mice treated with non-specific IgG under the same conditions. CONCLUSIONS: CXCL10 plays a role in the pathogenesis of CLP-induced septic shock and could serve as a therapeutic target during the acute phase of septic shock.


Chemokine CXCL10/metabolism , Shock, Septic/immunology , Animals , Body Temperature , Chemokine CXCL10/antagonists & inhibitors , Chemokine CXCL10/blood , Cytokines/blood , Female , Immunoglobulin G/immunology , Killer Cells, Natural/immunology , Lymphocyte Activation , Male , Mice, Inbred C57BL , Mice, Knockout , Peritoneal Cavity/physiology , Shock, Septic/microbiology
3.
Crit Care ; 16(5): R168, 2012 Sep 19.
Article En | MEDLINE | ID: mdl-22992408

INTRODUCTION: In our previous studies we demonstrated that CXC chemokine receptor 3 (CXCR3) participates in the regulation of lymphocyte trafficking during cecal ligation and puncture (CLP)-induced sepsis. In this study, we evaluated the effects of treatment with anti-CXCR3 immunoglobulin (IgG) and antibiotics on outcome during septic shock caused by CLP. METHODS: C57BL/6J mice were treated with neutralizing IgG against CXCR3 plus Primaxin either 24 hours prior to, 2 hours after or 6 hours after CLP. Control mice received nonspecific IgG plus Primaxin in the same regimen. Survival, core body temperature, bacterial clearance and systemic cytokine production were evaluated. RESULTS: Our results show that treatment with anti-CXCR3 IgG plus Primaxin significantly improved survival when administered 24 hours prior to CLP (50% vs. 10%), 2 hours after CLP (55% vs. 10%) or 6 hours after CLP (55% vs. 25%) compared with mice receiving nonspecific IgG plus Primaxin. Treatment with anti-CXCR3 plus Primaxin 24 hours prior to CLP attenuated hypothermia and IL-6 and macrophage inflammatory protein 2 (MIP-2) production but did not alter bacterial clearance. Treatment with anti-CXCR3 IgG and Primaxin 2 hours after CLP did not improve bacterial clearance and systemic cytokine production compared with mice treated with IgG and Primaxin, whereas 6 hours after CLP the bacterial clearance and IL-6 and MIP-2 concentrations, both in plasma and peritoneal lavage fluid, were significantly improved in mice receiving anti-CXCR3 IgG and Primaxin compared with mice that only received nonspecific IgG and Primaxin. CONCLUSION: The results from this study indicate that neutralization of CXCR3 prior to, 2 hours after or 6 hours after the initiation of CLP-induced septic shock improves survival and attenuates CLP-induced inflammation and physiologic dysfunction.


Antibodies, Anti-Idiotypic/therapeutic use , Disease Models, Animal , Immunoglobulin G , Receptors, CXCR3/antagonists & inhibitors , Sepsis/drug therapy , Animals , Antibodies, Anti-Idiotypic/pharmacology , Mice , Mice, Inbred C57BL , Receptors, CXCR3/metabolism , Sepsis/metabolism , Treatment Outcome
4.
Shock ; 38(4): 395-402, 2012 Oct.
Article En | MEDLINE | ID: mdl-22777121

STAT1 (signal transducer and activator of transcription 1) is a member of the JAK-STAT signaling family and plays a key role in facilitating gene transcription in response to activation of the types I and II interferon (IFN) receptors. TYK2 is essential for type I, but not type II, IFN-induced STAT1 activation. Previous studies show that STAT1-deficient mice are resistant to endotoxin-induced shock. The goal of the present study was to assess the response of STAT1- and TYK2-deficient mice to septic shock caused by cecal ligation and puncture (CLP). End points included survival, core temperature, organ injury, systemic cytokine production, and bacterial clearance. Results showed that survival rates were significantly higher in STAT1 knockout (STAT1KO) mice compared with wild-type controls (80% vs. 10%). The improved survival of STAT1KO mice was associated with less hypothermia, metabolic acidosis, hypoglycemia, and hepatocellular injury. Plasma interleukin 6, MIP-2, CXCL10, and IFN-α concentrations were significantly lower in STAT1KO mice than in wild-type mice. In the absence of antibiotic treatment, blood and lung bacterial counts were significantly lower in STAT1KO mice than in controls. However, treatment with antibiotics ablated that difference. A survival advantage was not observed in TYK2-deficient mice compared with control. However, CLP-induced hypothermia and systemic interleukin 6 and CXCL10 production were significantly attenuated in TYK2-deficient mice. These results indicate that STAT1 activation is an important factor in the pathogenesis of CLP-induced septic shock and is associated with the development of systemic inflammation and organ injury. TYK2 activation also appears to contribute to CLP-induced inflammation, but to a lesser extent than STAT1.


STAT1 Transcription Factor/metabolism , Shock, Septic/metabolism , Animals , Anti-Bacterial Agents/pharmacology , Bacteremia/drug therapy , Bacteremia/genetics , Bacteremia/metabolism , Bacteremia/pathology , Cecum , Cytokines/blood , Disease Models, Animal , Hypothermia/genetics , Hypothermia/metabolism , Inflammation/drug therapy , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Ligation , Mice , Mice, Knockout , STAT1 Transcription Factor/genetics , Shock, Septic/drug therapy , Shock, Septic/genetics , Shock, Septic/pathology , TYK2 Kinase/genetics , TYK2 Kinase/metabolism
5.
Am J Respir Crit Care Med ; 185(3): 291-300, 2012 Feb 01.
Article En | MEDLINE | ID: mdl-22135342

RATIONALE: Lymphocytes have been shown to facilitate systemic inflammation and physiologic dysfunction in experimental models of severe sepsis. Our previous studies show that natural killer (NK) cells migrate into the peritoneal cavity during intraabdominal sepsis, but the trafficking of NKT and T lymphocytes has not been determined. The factors that regulate lymphocyte trafficking during sepsis are currently unknown. OBJECTIVES: To ascertain the importance of CXC chemokine receptor 3 (CXCR3) as a regulator of lymphocyte trafficking during sepsis and determine the contribution of CXCR3-mediated lymphocyte trafficking to the pathogenesis of septic shock. METHODS: Lymphocyte trafficking was evaluated in control and CXCR3-deficient mice using flow cytometry during sepsis caused by cecal ligation and puncture (CLP). Survival, core temperature, cytokine production, and bacterial clearance were measured as pathobiological endpoints. MEASUREMENTS AND MAIN RESULTS: This study shows that concentrations of the CXCR3 ligands CXCL9 (monokine induced by interferon γ, MIG) and CXCL10 (interferon γ-induced protein 10, IP-10) increase in plasma and the peritoneal cavity after CLP, peak at 8 hours after infection, and are higher in the peritoneal cavity than in plasma. The numbers of CXCR3(+) NK cells progressively decreased in spleen after CLP with a concomitant increase within the peritoneal cavity, a pattern that was ablated in CXCR3-deficient mice. CXCR3-dependent recruitment of T cells was also evident at 16 hours after CLP. Treatment of mice with anti-CXCR3 significantly attenuated CLP-induced hypothermia, decreased systemic cytokine production, and improved survival. CONCLUSIONS: CXCR3 regulates NK- and T-cell trafficking during sepsis and blockade of CXCR3 attenuates the pathogenesis of septic shock.


Intraabdominal Infections/immunology , Killer Cells, Natural/metabolism , Receptors, CXCR3/metabolism , Shock, Septic/immunology , T-Lymphocytes/metabolism , Animals , Cell Movement/physiology , Chemokine CXCL10/metabolism , Chemokine CXCL9/metabolism , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Intraabdominal Infections/mortality , Mice , Mice, Inbred C57BL , Natural Killer T-Cells/metabolism , Receptors, CXCR3/antagonists & inhibitors , Shock, Septic/mortality
6.
Crit Care Med ; 38(1): 202-8, 2010 Jan.
Article En | MEDLINE | ID: mdl-19770742

OBJECTIVE: This study was designed to determine the ability of insulin to improve outcome following a Pseudomonas aeruginosa wound infection in a rodent model of severe burn injury. BACKGROUND: Severe burn injury predisposes patients to burn wound infections that can disseminate, lead to uncontrolled inflammation, and induce septic shock. Whereas insulin administration has been extensively discussed to improve morbidity and mortality in critically ill patients, the ability of insulin to improve outcomes of severely burned patients with infected burn wounds is not known. DESIGN: Sprague-Dawley rats. SETTING: University setting. INTERVENTION: Burn-injured Sprague Dawley rats were randomized into treatment groups that received either saline or insulin. Burn wounds were topically inoculated with a lethal dose of Pseudomonas aeruginosa 6 days after injury. MEASUREMENTS AND MAIN RESULTS: Survival, systemic dissemination of bacteria, systemic inflammation, and immune activation were examined. Insulin decreased the early inflammatory response to a severe burn injury. Treatment with low doses of insulin following burn injury improved the outcome of rats in response to a lethal burn wound infection. Specifically, survival was improved and systemic dissemination of bacteria from the wound was decreased. Systemic inflammation, indicated by serum interleukin-6 levels, was significantly decreased by insulin treatments after injury. Additionally, insulin treatments were associated with alterations in B and T lymphocyte responses to wound infection. CONCLUSIONS: Although the mechanisms by which insulin improves outcome following a lethal burn wound infection are not known, the data suggest that immunologic responses to infection may be altered by postburn insulin treatments.


Bacteremia/drug therapy , Burns/drug therapy , Insulin/pharmacology , Pseudomonas Infections/drug therapy , Wound Infection/drug therapy , Animals , Bacteremia/mortality , Bacteremia/prevention & control , Burns/complications , Burns/mortality , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Administration Schedule , Flow Cytometry , Inflammation/prevention & control , Lymphocyte Activation/drug effects , Multivariate Analysis , Probability , Pseudomonas Infections/complications , Pseudomonas Infections/mortality , Random Allocation , Rats , Rats, Sprague-Dawley , Risk Factors , Statistics, Nonparametric , Survival Rate , Wound Healing/drug effects , Wound Healing/physiology , Wound Infection/complications , Wound Infection/mortality
7.
J Immunol ; 180(9): 6334-45, 2008 May 01.
Article En | MEDLINE | ID: mdl-18424757

Evidence suggests that NK and NKT cells contribute to inflammation and mortality during septic shock caused by cecal ligation and puncture (CLP). However, the specific contributions of these cell types to the pathogenesis of CLP-induced septic shock have not been fully defined. The goal of the present study was to determine the mechanisms by which NK and NKT cells mediate the host response to CLP. Control, NK cell-deficient, and NKT cell-deficient mice underwent CLP. Survival, cytokine production, and bacterial clearance were measured. NK cell trafficking and interaction with myeloid cells was also studied. Results show that mice treated with anti-asialoGM1 (NK cell deficient) or anti-NK1.1 (NK/NKT cell deficient) show less systemic inflammation and have improved survival compared with IgG-treated controls. CD1 knockout mice (NKT cell deficient) did not demonstrate decreased cytokine production or improved survival compared with wild type mice. Trafficking studies show migration of NK cells from blood and spleen into the inflamed peritoneal cavity where they appear to facilitate the activation of peritoneal macrophages (F4-80(+)GR-1(-)) and F4-80(+)Gr-1(+) myeloid cells. These findings indicate that NK but not CD1-restricted NKT cells contribute to acute CLP-induced inflammation. NK cells appear to mediate their proinflammatory functions during septic shock, in part, by migration into the peritoneal cavity and amplification of the proinflammatory activities of specific myeloid cell populations. These findings provide new insights into the mechanisms used by NK cells to facilitate acute inflammation during septic shock.


Antigens, CD1/immunology , Cell Movement/immunology , Killer Cells, Natural/immunology , Shock, Septic/immunology , T-Lymphocytes/immunology , Abdomen/pathology , Animals , Antigens, Ly , Antigens, Surface/immunology , Cell Movement/drug effects , G(M1) Ganglioside/antagonists & inhibitors , G(M1) Ganglioside/immunology , Immunoglobulin G/immunology , Immunoglobulin G/pharmacology , Inflammation/immunology , Inflammation/pathology , Killer Cells, Natural/pathology , Lectins, C-Type/antagonists & inhibitors , Lectins, C-Type/immunology , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/pathology , Mice , Mice, Inbred BALB C , Mice, Knockout , NK Cell Lectin-Like Receptor Subfamily B , Shock, Septic/pathology , T-Lymphocytes/pathology
8.
Shock ; 23(3): 224-32, 2005 Mar.
Article En | MEDLINE | ID: mdl-15718919

These studies evaluated the effects treatment with glucan phosphate, a soluble polysaccharide immunomodulator, on the inflammatory response induced by burn injury and on resistance to Pseudomonas aeruginosa burn wound infection. Mice were exposed to 35% total body surface area burns and were resuscitated with lactated Ringer's (LR) solution alone or LR supplemented with glucan phosphate (40 mg/kg). Glucan phosphate treatment attenuated burn-induced expression of interleukin (IL)-1beta, IL-6, and IL-10 mRNAs in spleen, lung, and heart. Plasma concentrations of IL-1beta, IL-6, macrophage inflammatory protein (MIP)-2, and IL-10 were also decreased in burned mice treated with glucan phosphate compared with vehicle-treated controls. Early postburn mortality was not significantly different between control (20%) and glucan phosphate-treated (10%) mice, but there was a small improvement in acid-base balance in the glucan phosphate-treated group. Mice received a second injection of glucan phosphate or LR on day 4 postburn and were infected by topical application of P. aeruginosa to the burn wound on day 5. Glucan phosphate treatment significantly improved survival in mice exposed to P. aeruginosa burn wound infection. The improved survival correlated with lower bacterial burden in the burn wound, attenuated production of proinflammatory cytokines, and enhanced production of Th1 cytokines. These studies show that glucan phosphate treatment attenuates burn-induced inflammation and increases resistance to P. aeruginosa burn wound infection in an experimental model of burn injury.


Burns/drug therapy , Glucans/therapeutic use , Inflammation/prevention & control , Pseudomonas Infections/prevention & control , Wound Infection/prevention & control , Acid-Base Equilibrium/drug effects , Animals , Burns/complications , Burns/genetics , Burns/immunology , Chemokine CXCL2 , Chemokines/blood , Female , Gene Expression/drug effects , Histocompatibility Antigens Class II/metabolism , Interferon-gamma/biosynthesis , Interleukin-1/biosynthesis , Interleukin-1/genetics , Interleukin-10/biosynthesis , Interleukin-10/genetics , Interleukin-12/biosynthesis , Interleukin-6/biosynthesis , Interleukin-6/genetics , Mice , Mice, Inbred C57BL , RNA, Messenger/genetics , RNA, Messenger/metabolism
9.
J Immunol ; 174(1): 404-10, 2005 Jan 01.
Article En | MEDLINE | ID: mdl-15611264

Fms-like tyrosine kinase-3 ligand (Flt3L) is a hemopoietic cytokine that stimulates the production of dendritic cells. This study evaluated the ability of Flt3L-enhanced dendritic cell production to increase the resistance of mice to a burn wound infection with Pseudomonas aeruginosa, a common source of infections in burn patients that have impaired immunity and are susceptible to opportunistic microorganisms. Treatment of mice with Flt3L for 5 days caused a significant increase in dendritic cell numbers in the spleen and significantly increased survival upon a subsequent burn wound infection. Improved survival in Flt3L-treated mice was associated with limited bacterial growth and spread within the burn wounds and a decrease in systemic dissemination of P. aeruginosa. Resistance to burn wound infection could also be conferred to recipient mice by the adoptive transfer of dendritic cells that had been isolated from spleens of Flt3L-treated mice. Adoptive transfer of the same number of splenic dendritic cells from nontreated mice did not confer resistance to burn wound infection. These data indicate that Flt3L can increase the resistance of mice to a P. aeruginosa burn wound infection through both stimulation of dendritic cell production and enhancement of dendritic cell function.


Burns/complications , Dendritic Cells/cytology , Pseudomonas Infections/prevention & control , Wound Infection/prevention & control , Adoptive Transfer , Animals , Cell Count , Dendritic Cells/immunology , Flow Cytometry , Humans , Ligands , Mice , Proto-Oncogene Proteins/metabolism , Pseudomonas Infections/etiology , Receptor Protein-Tyrosine Kinases/metabolism , Spleen/cytology , Wound Infection/etiology , fms-Like Tyrosine Kinase 3
10.
Infect Immun ; 71(6): 3058-67, 2003 Jun.
Article En | MEDLINE | ID: mdl-12761083

Patients with large burn injuries are susceptible to opportunistic infections due to impaired functions of multiple effector cells of innate immunity and acquired immunity, including macrophages, dendritic cells (DC), natural killer (NK) cells, and T cells. The ability of a host to produce Th1 cytokines, such as gamma interferon (IFN-gamma) and interleukin-12 (IL-12), upon infectious challenge is also impaired after burn injury. Stimulation of hematopoiesis, to regenerate new immune cells, may be an effective strategy for improving resistance to infections after severe burn trauma. Fms-like tyrosine kinase 3 ligand (Flt3L) is a hematopoietic cytokine that stimulates the expansion and differentiation of NK cells and DC. Using a mouse model, we tested the hypothesis that Flt3L treatments after burn injury stimulate the production of functional effector cells of innate immunity and restore appropriate Th1 cytokine responses to Pseudomonas aeruginosa, a common source of pneumonia and wound infections in burn victims. Flt3L increased splenic cellularity in sham (uninjured) and burned mice and increased the numbers of NK cells (DX5(+)) and DC (CD11c(+)). In response to P. aeruginosa, significant increases in the serum IFN-gamma levels and the numbers of splenic IFN-gamma-producing DC, NK cells, and T cells were observed in Flt3L-treated burned mice compared to the values obtained for untreated burned mice. The splenic levels of IL-12 and IL-15 mRNAs and the IL-12 and IL-15 receptors were also increased. In addition, Flt3L treatment restored the ability of splenic cultures prepared from burned mice to produce IFN-gamma and IL-12 after in vitro challenge with P. aeruginosa. Flt3L may have potential for restoring NK cell and DC functions and improving immunity after burn injury.


Burns/immunology , Cytokines/biosynthesis , Hematopoiesis/drug effects , Membrane Proteins/pharmacology , Pseudomonas aeruginosa/immunology , Th1 Cells/immunology , Animals , Dendritic Cells/physiology , Interferon-gamma/biosynthesis , Interleukin-12/biosynthesis , Interleukin-15/biosynthesis , Killer Cells, Natural/physiology , Mice , Mice, Inbred BALB C
11.
Shock ; 18(4): 322-30, 2002 Oct.
Article En | MEDLINE | ID: mdl-12392275

Thermal injury to 40% or more of the total body surface area poses a significant risk for the development of opportunistic infections that increase complications and mortality. Altered cytokine induction profiles, including suppression of the Th1 cytokines IFN-gamma and IL-12 and elevations in the anti-inflammatory cytokine IL-10, are believed to contribute to burn-associated immunosuppression and the development of sepsis. The specific changes that lead to altered cytokine production following major burns are not known. We examined the effects of burn injuries to 40% of the mouse body surface on IFN-gamma induction in the major IFN-gamma-producing cell types of the spleen. Additionally, effects on key IFN-gamma-regulatory cytokines were examined after bacterial challenge. We report that in vivo induction of IFN-gamma in natural killer lymphocytes is suppressed in burned mice. Splenic IFN-gamma was suppressed at both the mRNA and protein levels. Early suppression was associated with impairments in both the macrophage/dendritic cell and lymphocyte populations, whereas persistent suppression was associated with impaired lymphocyte function and decreased responsiveness to IFN-gamma-inducing factors. IFN-gamma production could be restored by neutralization of the upregulated cytokine IL-10. Induction of the IFN-gamma-inducers IL-15, IL-12, and IL-2 was also impaired after burn injury, whereas IL-18 levels remained unaffected. Exogenous application of these suppressed cytokines to isolated splenocytes did not restore IFN-gamma to sham levels, indicating a loss of responsiveness to these factors. Expression of the IL-2, IL-12, and IL-15 receptors was suppressed after thermal injury. We conclude that burn-associated suppression of IFN-gamma is due to deficient production of inducing factors and their receptors, leading to severe impairments in cellular IFN-gamma induction pathways.


Burns/metabolism , Cytokines/biosynthesis , Interferon-gamma/biosynthesis , Lymphocytes/metabolism , Receptors, Cytokine/metabolism , Animals , Cells, Cultured , Coculture Techniques , Cytokines/genetics , Female , Interleukin-10/metabolism , Interleukin-12/metabolism , Killer Cells, Natural/metabolism , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Pseudomonas aeruginosa/pathogenicity , Spleen/cytology , Spleen/metabolism
12.
Clin Diagn Lab Immunol ; 9(3): 530-43, 2002 May.
Article En | MEDLINE | ID: mdl-11986256

Gamma interferon (IFN-gamma) is an important mediator of endotoxin (lipopolysaccharide [LPS])-induced immune responses. However, the specific cell types that produce IFN-gamma in response to LPS and the cellular factors that regulate LPS-induced IFN-gamma production have not been fully determined. The present studies were undertaken to characterize the cell populations that produce IFN-gamma after LPS challenge in the spleens of mice and to determine the regulatory factors that modulate LPS-induced production of IFN-gamma. Our studies show that the levels of splenic IFN-gamma mRNA and protein production peak at 6 and 8 h, respectively, after systemic LPS challenge. Approximately 60% of IFN-gamma-producing cells are natural killer (NK) cells (CD3(-)DX5(+)) and 25% are NKT cells (CD3(+)DX5(+)). Most of the remaining IFN-gamma-producing cells are T cells (CD3(+)DX5(-)), macrophages, and dendritic cells. Functionally, interleukin-12 (IL-12) is the major IFN-gamma-stimulating factor after LPS challenge, with costimulation provided by IL-15, IL-18, and B7 proteins. IL-10 is a major inhibitor of LPS-induced IFN-gamma production. Unlike intact heat-killed gram-negative and gram-positive bacteria, the class II major histocompatibility complex did not play a functional role in LPS-induced IFN-gamma production. LPS is a potent stimulus for splenic IL-10, IL-12 p40, and IL-15 mRNA expression, whereas IL-12 p35 and IL-18 mRNAs, as well as B7 proteins, are constitutively expressed in the mouse spleen. Of the factors studied, IL-18 serves as the most potent costimulus with IL-12 for IFN-gamma production, followed by IL-15 and B7 proteins. These data demonstrate that NK cells and NKT cells are the most abundant IFN-gamma-producing cells in the mouse spleen after LPS challenge and that IL-10 and IL-12 are key functional regulators of LPS-induced IFN-gamma production.


B7-1 Antigen/immunology , Interferon-gamma/biosynthesis , Interleukin-12/immunology , Interleukin-15/immunology , Interleukin-18/immunology , Signal Transduction/immunology , Animals , CD28 Antigens/immunology , Cells, Cultured , Dendritic Cells/drug effects , Dendritic Cells/immunology , Female , Gene Expression Profiling , Histocompatibility Antigens Class II/immunology , Interferon-gamma/genetics , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Lipopolysaccharides/immunology , Lipopolysaccharides/pharmacology , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/immunology , Mice , Mice, Inbred BALB C , RNA, Messenger , Spleen/cytology , Spleen/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
...