Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
PLoS One ; 14(3): e0212670, 2019.
Article in English | MEDLINE | ID: mdl-30913212

ABSTRACT

Immunotherapy has fundamentally changed the landscape of cancer treatment. Despite the encouraging results with the checkpoint modulators, response rates vary widely across tumor types, with a majority of patients exhibiting either primary resistance without a significant initial response to treatment or acquired resistance with subsequent disease progression. Hematopoietic progenitor kinase 1 (HPK1) is predominantly expressed in hematopoietic cell linages and serves as a negative regulator in T cells and dendritic cells (DC). While HPK1 gene knockout (KO) studies suggest its role in anti-tumor immune responses, the involvement of kinase activity and thereof its therapeutic potential remain unknown. To investigate the potential of pharmacological intervention using inhibitors of HPK1, we generated HPK1 kinase dead (KD) mice which carry a single loss-of-function point mutation in the kinase domain and interrogated the role of kinase activity in immune cells in the context of suppressive factors or the tumor microenvironment (TME). Our data provide novel findings that HKP1 kinase activity is critical in conferring suppressive functions of HPK1 in a wide range of immune cells including CD4+, CD8+, DC, NK to Tregs, and inactivation of kinase domain was sufficient to elicit robust anti-tumor immune responses. These data support the concept that an HPK1 small molecule kinase inhibitor could serve as a novel agent to provide additional benefit in combination with existing immunotherapies, particularly to overcome resistance to current treatment regimens.


Subject(s)
Immunity, Cellular , Immunologic Surveillance , Lymphocytes/immunology , Neoplasms, Experimental/immunology , Protein Serine-Threonine Kinases/immunology , Tumor Microenvironment/immunology , Animals , Cell Line, Tumor , Lymphocytes/pathology , Mice , Mice, Mutant Strains , Neoplasms, Experimental/genetics , Point Mutation , Protein Serine-Threonine Kinases/genetics , Tumor Microenvironment/genetics
2.
J Leukoc Biol ; 102(5): 1271-1280, 2017 11.
Article in English | MEDLINE | ID: mdl-28899907

ABSTRACT

IFN-γ-inducible protein 10 (CXCL10), a chemokine that is abundantly secreted in response to inflammatory stimuli, has been implicated in the pathogenesis of multiple inflammatory diseases, such as inflammatory bowel disease. Whereas CXCL10 is traditionally recognized for recruiting pathogenic T cells to inflamed sites, its nonchemotactic role during inflammation remains poorly defined. In this report, we identified a novel function of CXCL10 in the regulation of the inflammatory potential of human monocytes to produce cytokines. We found that CXCL10 was necessary and sufficient for IFN-γ-primed human monocytes to induce a robust production of proinflammatory cytokines, such as IL-12 and IL-23. CXCL10-induced monocyte production of these cytokines depended on CXCR3 receptor engagement as well as on the Iκ B kinase and p38 MAPK signaling pathways. By using an innate-mediated murine colitis model, we demonstrated that anti-CXCL10 Ab treatment robustly suppressed the local production of myeloid-derived inflammatory cytokines and intestinal tissue damage. Together, our data unravel a previously unappreciated role of CXCL10 in the amplification of myeloid cell-mediated inflammatory responses. Targeting CXCL10 is therefore an attractive approach to treating inflammatory diseases that are driven by innate and adaptive immunity.


Subject(s)
Adaptive Immunity , Chemokine CXCL10/immunology , Colitis, Ulcerative/immunology , Crohn Disease/immunology , Immunity, Innate , Monocytes/immunology , Animals , Antibodies, Neutralizing/administration & dosage , CD40 Antigens/antagonists & inhibitors , Chemokine CXCL10/antagonists & inhibitors , Chemokine CXCL10/genetics , Colitis/chemically induced , Colitis/genetics , Colitis/immunology , Colitis/pathology , Colitis, Ulcerative/genetics , Colitis, Ulcerative/pathology , Crohn Disease/genetics , Crohn Disease/pathology , Female , Gene Expression Regulation , Humans , I-kappa B Kinase/genetics , I-kappa B Kinase/immunology , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-12/genetics , Interleukin-12/immunology , Interleukin-23/genetics , Interleukin-23/immunology , Male , Mice , Mice, Inbred BALB C , Monocytes/cytology , Primary Cell Culture , Receptors, CXCR3/genetics , Receptors, CXCR3/immunology , Signal Transduction , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/immunology
3.
JCI Insight ; 2(2): e88257, 2017 01 26.
Article in English | MEDLINE | ID: mdl-28138553

ABSTRACT

Loss of function or overexpression of methyl-CpG-binding protein 2 (MeCP2) results in the severe neurodevelopmental disorders Rett syndrome and MeCP2 duplication syndrome, respectively. MeCP2 plays a critical role in neuronal function and the function of cells throughout the body. It has been previously demonstrated that MeCP2 regulates T cell function and macrophage response to multiple stimuli, and that immune-mediated rescue imparts significant benefit in Mecp2-null mice. Unlike Rett syndrome, MeCP2 duplication syndrome results in chronic, severe respiratory infections, which represent a significant cause of patient morbidity and mortality. Here, we demonstrate that MeCP2Tg3 mice, which overexpress MeCP2 at levels 3- to 5-fold higher than normal, are hypersensitive to influenza A/PR/8/34 infection. Prior to death, MeCP2Tg3 mice experienced a host of complications during infection, including neutrophilia, increased cytokine production, excessive corticosterone levels, defective adaptive immunity, and vascular pathology characterized by impaired perfusion and pulmonary hemorrhage. Importantly, we found that radioresistant cells are essential to infection-related death after bone marrow transplantation. In all, these results demonstrate that influenza A infection in MeCP2Tg3 mice results in pathology affecting both immune and nonhematopoietic cells, suggesting that failure to effectively respond and clear viral respiratory infection has a complex, multicompartment etiology in the context of MeCP2 overexpression.


Subject(s)
Methyl-CpG-Binding Protein 2/genetics , Orthomyxoviridae Infections/genetics , Adaptive Immunity/immunology , Animals , Corticosterone/metabolism , Cytokines/immunology , Genetic Predisposition to Disease , Hemorrhage/etiology , Influenza A virus , Interferon-gamma/immunology , Lung Diseases/etiology , Mental Retardation, X-Linked/genetics , Mental Retardation, X-Linked/immunology , Methyl-CpG-Binding Protein 2/immunology , Mice , Neutrophils/immunology , Orthomyxoviridae Infections/complications , Orthomyxoviridae Infections/immunology , Radiation Tolerance , Vascular Diseases/etiology
4.
PLoS Pathog ; 13(1): e1006140, 2017 01.
Article in English | MEDLINE | ID: mdl-28085958

ABSTRACT

The Influenza A virus (IAV) is a major human pathogen that produces significant morbidity and mortality. To explore the contribution of alveolar macrophages (AlvMΦs) in regulating the severity of IAV infection we employed a murine model in which the Core Binding Factor Beta gene is conditionally disrupted in myeloid cells. These mice exhibit a selective deficiency in AlvMΦs. Following IAV infection these AlvMΦ deficient mice developed severe diffuse alveolar damage, lethal respiratory compromise, and consequent lethality. Lethal injury in these mice resulted from increased infection of their Type-1 Alveolar Epithelial Cells (T1AECs) and the subsequent elimination of the infected T1AECs by the adaptive immune T cell response. Further analysis indicated AlvMΦ-mediated suppression of the cysteinyl leukotriene (cysLT) pathway genes in T1AECs in vivo and in vitro. Inhibition of the cysLT pathway enzymes in a T1AECs cell line reduced the susceptibility of T1AECs to IAV infection, suggesting that AlvMΦ-mediated suppression of this pathway contributes to the resistance of T1AECs to IAV infection. Furthermore, inhibition of the cysLT pathway enzymes, as well as blockade of the cysteinyl leukotriene receptors in the AlvMΦ deficient mice reduced the susceptibility of their T1AECs to IAV infection and protected these mice from lethal infection. These results suggest that AlvMΦs may utilize a previously unappreciated mechanism to protect T1AECs against IAV infection, and thereby reduce the severity of infection. The findings further suggest that the cysLT pathway and the receptors for cysLT metabolites represent potential therapeutic targets in severe IAV infection.


Subject(s)
Alveolar Epithelial Cells/immunology , Cysteine/metabolism , Influenza A virus/immunology , Influenza, Human/immunology , Leukotrienes/metabolism , Macrophages, Alveolar/immunology , Pneumonia, Viral/immunology , Adaptive Immunity , Alveolar Epithelial Cells/virology , Animals , Disease Models, Animal , Humans , Influenza, Human/virology , Lung/immunology , Lung/pathology , Mice , Mutation , Myeloid Cells/immunology , Pneumonia, Viral/virology , Specific Pathogen-Free Organisms
5.
Phys Chem Chem Phys ; 18(36): 24880-9, 2016 Sep 28.
Article in English | MEDLINE | ID: mdl-27337993

ABSTRACT

The alkyl oxonium ion, which is a protonated alcohol, has long been proposed as a key reaction intermediate in alcohol dehydration. Nonetheless, the dynamics and structure of this simple but important intermediate species have not been adequately examined due to the transient nature of the oxonium ion. Here, we devised a model system for the key step in the alcohol dehydration reaction, in which a photoacid transfers a proton to alcohols of different basicity in the acetonitrile solvent. Using time-resolved spectroscopy and computation, we have found that the linkage of at least two alcohol molecules via hydrogen bonding is critical for their enhanced reactivity and extraction of the proton from the acid. This finding addresses the cooperative role of the simplest organic protic compounds, namely alcohols, in nonaqueous acid-base reactions.

6.
Methods Appl Fluoresc ; 4(2): 024004, 2016 05 04.
Article in English | MEDLINE | ID: mdl-28809169

ABSTRACT

The excited-state proton transfer (ESPT) of the strong photoacid, N-methyl-7-hydroxyquinolinium, was studied in the presence of different weak bases such as methanol, ethanol, and dimethyl sulfoxide in an aprotic solvent of acetonitrile. Here, we present chemical kinetics analysis of the ESPT mechanism to explain biphasic fluorescence decay of the parent photoacid and the sign reversal of the rise and decay of the resulting conjugate-base fluorescence. The ESPT of the free photoacid showed a molecularity of 2 with reacting alcohol molecules. In the ground state, it was found that a fraction of the photoacid formed 1 : 2 hydrogen-bonded complexes with the residual water present in the aprotic solvent or 1 : 1 complexes with the additive alcohols. In the excited state, these adducts underwent proton transfer when complexed further with diffusing alcohol molecules.

7.
J Clin Invest ; 125(10): 3965-80, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26389678

ABSTRACT

Erythropoiesis is an important response to certain types of stress, including hypoxia, hemorrhage, bone marrow suppression, and anemia, that result in inadequate tissue oxygenation. This stress-induced erythropoiesis is distinct from basal red blood cell generation; however, neither the cellular nor the molecular factors that regulate this process are fully understood. Here, we report that type 1 conventional dendritic cells (cDC1s), which are defined by expression of CD8α in the mouse and XCR1 and CLEC9 in humans, are critical for induction of erythropoiesis in response to stress. Specifically, using murine models, we determined that engagement of a stress sensor, CD24, on cDC1s upregulates expression of the Kit ligand stem cell factor on these cells. The increased expression of stem cell factor resulted in Kit-mediated proliferative expansion of early erythroid progenitors and, ultimately, transient reticulocytosis in the circulation. Moreover, this stress response was triggered in part by alarmin recognition and was blunted in CD24 sensor- and CD8α+ DC-deficient animals. The contribution of the cDC1 subset to the initiation of stress erythropoiesis was distinct from the well-recognized role of macrophages in supporting late erythroid maturation. Together, these findings offer insight into the mechanism of stress erythropoiesis and into disorders of erythrocyte generation associated with stress.


Subject(s)
Dendritic Cells/physiology , Erythropoiesis/physiology , Stress, Physiological/physiology , Alarmins/physiology , Animals , CD24 Antigen/physiology , CD8 Antigens/analysis , Cisplatin/toxicity , Colony-Forming Units Assay , Dendritic Cells/classification , Erythroid Precursor Cells/physiology , Female , Gene Expression Profiling , HMGB1 Protein/toxicity , Hematopoietic Stem Cell Transplantation , Heterografts , Humans , Hypoxia/physiopathology , Imatinib Mesylate/toxicity , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Phlebotomy/adverse effects , Radiation Chimera , Recombinant Proteins/toxicity , Splenectomy/adverse effects , Stem Cell Factor/biosynthesis , Stem Cell Factor/genetics
8.
PLoS One ; 10(4): e0120169, 2015.
Article in English | MEDLINE | ID: mdl-25849970

ABSTRACT

Influenza A virus (IAV) infection of the respiratory tract elicits a robust immune response, which is required for efficient virus clearance but at the same time can contribute to lung damage and enhanced morbidity. IL-21 is a member of the type I cytokine family and has many different immune-modulatory functions during acute and chronic virus infections, although its role in IAV infection has not been fully evaluated. In this report we evaluated the contributions of IL-21/IL-21 receptor (IL-21R) signaling to host defense in a mouse model of primary IAV infection using IL-21R knock out (KO) mice. We found that lack of IL-21R signaling had no significant impact on virus clearance, adaptive T cell responses, or myeloid cell accumulations in the respiratory tract. However, a subset of inflammatory cytokines were elevated in the bronchoalveolar lavage fluid of IL-21R KO mice, including IL-17. Although there was only a small increase in Th17 cells in the lungs of IL-21R KO mice, we observed a dramatic increase in gamma delta (γδ) T cells capable of producing IL-17 both after IAV infection and at steady state in the respiratory tract. Finally, we found that IL-21R signaling suppressed the accumulation of IL-17+ γδ T cells in the respiratory tract intrinsically. Thus, our study reveals a previously unrecognized role of IL-21R signaling in regulating IL-17 production by γδ T cells.


Subject(s)
Cytokines/metabolism , Influenza A virus/pathogenicity , Interleukin-17/metabolism , Lung/immunology , Orthomyxoviridae Infections/immunology , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Receptors, Interleukin-21/physiology , Th17 Cells/immunology , Animals , Bronchoalveolar Lavage Fluid/chemistry , Cells, Cultured , Cytokines/genetics , Female , Flow Cytometry , Influenza A virus/immunology , Interleukin-17/genetics , Lung/pathology , Lung/virology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Orthomyxoviridae Infections/virology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptors, Antigen, T-Cell, gamma-delta/genetics , Reverse Transcriptase Polymerase Chain Reaction
9.
J Immunol ; 194(7): 3213-22, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25712214

ABSTRACT

The liver maintains a tolerogenic environment to avoid unwarranted activation of its resident immune cells upon continuous exposure to food and bacterially derived Ags. However, in response to hepatotropic viral infection, the liver's ability to switch from a hyporesponsive to a proinflammatory environment is mediated by select sentinels within the parenchyma. To determine the contribution of hepatic dendritic cells (DCs) in the activation of naive CD8(+) T cells, we first characterized resident DC subsets in the murine liver. Liver DCs exhibit unique properties, including the expression of CD8α (traditionally lymphoid tissue specific), CD11b, and CD103 markers. In both the steady-state and following viral infection, liver CD103(+) DCs express high levels of MHC class II, CD80, and CD86 and contribute to the high number of activated CD8(+) T cells. Importantly, viral infection in the Batf3(-/-) mouse, which lacks CD8α(+) and CD103(+) DCs in the liver, results in a 3-fold reduction in the proliferative response of Ag-specific CD8(+) T cells. Limiting DC migration out of the liver does not significantly alter CD8(+) T cell responsiveness, indicating that CD103(+) DCs initiate the induction of CD8(+) T cell responses in situ. Collectively, these data suggest that liver-resident CD103(+) DCs are highly immunogenic in response to hepatotropic viral infection and serve as a major APC to support the local CD8(+) T cell response. It also implies that CD103(+) DCs present a promising cellular target for vaccination strategies to resolve chronic liver infections.


Subject(s)
Antigens, CD/metabolism , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Integrin alpha Chains/metabolism , Liver/immunology , Lymphocyte Activation/immunology , Adenoviridae/immunology , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Antigens, Surface/metabolism , CD11b Antigen/metabolism , Cell Movement , Female , Immunophenotyping , Liver/pathology , Liver/virology , Male , Mice , Mice, Transgenic , Phenotype , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Viruses/immunology
10.
Curr Top Microbiol Immunol ; 386: 423-55, 2015.
Article in English | MEDLINE | ID: mdl-25033753

ABSTRACT

Influenza virus infection induces a potent initial innate immune response, which serves to limit the extent of viral replication and virus spread. However, efficient (and eventual) viral clearance within the respiratory tract requires the subsequent activation, rapid proliferation, recruitment, and expression of effector activities by the adaptive immune system, consisting of antibody producing B cells and influenza-specific T lymphocytes with diverse functions. The ensuing effector activities of these T lymphocytes ultimately determine (along with antibodies) the capacity of the host to eliminate the viruses and the extent of tissue damage. In this review, we describe this effector T cell response to influenza virus infection. Based on information largely obtained in experimental settings (i.e., murine models), we will illustrate the factors regulating the induction of adaptive immune T cell responses to influenza, the effector activities displayed by these activated T cells, the mechanisms underlying the expression of these effector mechanisms, and the control of the activation/differentiation of these T cells, in situ, in the infected lungs.


Subject(s)
Influenza, Human/immunology , T-Lymphocytes/immunology , Adaptive Immunity , Animals , Antigen Presentation , Dendritic Cells/immunology , Exocytosis , Humans , Immunity, Innate , Lung/immunology , Lymphocyte Activation
11.
PLoS One ; 9(8): e105576, 2014.
Article in English | MEDLINE | ID: mdl-25153088

ABSTRACT

T cell development and activation are highly regulated processes, and their proper execution is important for a competent immune system. Shc SH2-domain binding protein-1 (Shcbp1) is an evolutionarily conserved protein that binds to the adaptor protein ShcA. Studies in Drosophila and in cell lines have strongly linked Shcbp1 to cell proliferation, embryonic development, growth factor signaling, and tumorigenesis. Here we show that Shcbp1 expression is strikingly upregulated during the ß-selection checkpoint in thymocytes, and that its expression tightly correlates with proliferative stages of T cell development. To evaluate the role for Shcbp1 during thymic selection and T cell function in vivo, we generated mice with global and conditional deletion of Shcbp1. Surprisingly, the loss of Shcbp1 expression did not have an obvious effect during T cell development. However, in a mouse model of experimental autoimmune encephalomyelitis (EAE), which depends on CD4(+) T cell function and mimics multiple features of the human disease multiple sclerosis, Shcbp1 deficient mice had reduced disease severity and improved survival, and this effect was T cell intrinsic. These data suggest that despite the striking upregulation of Shcbp1 during T cell proliferation, loss of Shcbp1 does not directly affect T cell development, but regulates CD4(+) T cell effector function in vivo.


Subject(s)
Cell Proliferation/genetics , Shc Signaling Adaptor Proteins/genetics , T-Lymphocytes/metabolism , Animals , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/metabolism , Mice , Mice, Knockout , Phenotype , Shc Signaling Adaptor Proteins/metabolism , Signal Transduction/genetics , T-Lymphocytes/cytology , Thymus Gland/cytology , Thymus Gland/metabolism
13.
Immunity ; 40(3): 400-13, 2014 Mar 20.
Article in English | MEDLINE | ID: mdl-24631155

ABSTRACT

The contribution of different DC subsets to effector and memory CD8(+) T cell generation during infection and the mechanism by which DCs controls these fate decisions is unclear. Here we demonstrated that the CD103(+) and CD11b(hi) migratory respiratory DC (RDC) subsets after influenza virus infection activated naive virus-specific CD8(+) T cells differentially. CD103(+) RDCs supported the generation of CD8(+) T effector (Teff) cells, which migrate from lymph nodes to the infected lungs. In contrast, migrant CD11b(hi) RDCs activated CD8(+) T cells characteristic of central memory CD8(+) T (CD8(+) Tcm) cells including retention within the draining lymph nodes. CD103(+) RDCs expressed CD24 at an elevated level, contributing to the propensity of this DC subpopulation to support CD8(+) Teff cell differentiation. Mechanistically, CD24 was shown to regulate CD8(+) T cell activation through HMGB1-mediated engagement of T cell RAGE. Thus, there is distribution of labor among DC subsets in regulating CD8(+) T cell differentiation.


Subject(s)
CD24 Antigen/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Immunologic Memory , Animals , Antigens, CD/metabolism , CD11b Antigen/metabolism , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/virology , Female , Immunophenotyping , Integrin alpha Chains/metabolism , Lung/immunology , Lung/metabolism , Lung/virology , Lymph Nodes/immunology , Lymph Nodes/metabolism , Lymph Nodes/virology , Lymphocyte Activation/immunology , Mice , Mice, Knockout , Phenotype , Protein Binding , Receptor for Advanced Glycation End Products , Receptors, Immunologic/metabolism , Virus Release/immunology
14.
J Allergy Clin Immunol ; 132(6): 1263-76; quiz 1277, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23915713

ABSTRACT

Because of its essential role in gas exchange and oxygen delivery, the lung has evolved a variety of strategies to control inflammation and maintain homeostasis. Invasion of the lung by pathogens (and in some instances exposure to certain noninfectious particulates) disrupts this equilibrium and triggers a cascade of events aimed at preventing or limiting colonization (and more importantly infection) by pathogenic microorganisms. In this review we focus on viral infection of the lung and summarize recent advances in our understanding of the triggering of innate and adaptive immune responses to viral respiratory tract infection, mechanisms of viral clearance, and the well-recognized consequences of acute viral infection complicating underlying lung diseases, such as asthma.


Subject(s)
Asthma/immunology , Asthma/physiopathology , Host-Pathogen Interactions , Lung/immunology , Pneumonia, Viral/immunology , Adaptive Immunity , Animals , Humans , Immunity, Innate , Lung/virology
15.
Nat Rev Immunol ; 12(4): 295-305, 2012 Mar 09.
Article in English | MEDLINE | ID: mdl-22402670

ABSTRACT

Recent years have seen several advances in our understanding of immunity to virus infection of the lower respiratory tract, including to influenza virus infection. Here, we review the cellular targets of viruses and the features of the host immune response that are unique to the lungs. We describe the interplay between innate and adaptive immune cells in the induction, expression and control of antiviral immunity, and discuss the impact of the infected lung milieu on moulding the response of antiviral effector T cells. Recent findings on the mechanisms that underlie the increased frequency of severe pulmonary bacterial infections following respiratory virus infection are also discussed.


Subject(s)
Adaptive Immunity/immunology , Respiratory Tract Infections/immunology , Respiratory Tract Infections/virology , Animals , Antigen-Presenting Cells/immunology , Humans , Respiratory Tract Infections/microbiology , Superinfection/immunology , Superinfection/virology , T-Lymphocytes/immunology
16.
Korean J Hepatobiliary Pancreat Surg ; 16(4): 147-53, 2012 Nov.
Article in English | MEDLINE | ID: mdl-26388926

ABSTRACT

BACKGROUNDS/AIMS: In the treatment of complicated cholecystitis, laparoscopic cholecystectomy (LC) has limited efficacy due to its substantial post-operative complications. In addition, the clinical characteristics of complicated cholecystitis (CC) patients were suspected as advanced age with highly risky comorbidity. Percutaneous transhepatic gall bladder (PTGBD) drainage could be an alternative option for successful LC. Hence, this study evaluated the outcome of PTGBD for CC within and after 5 days. METHODS: The medical records of 109 consecutive CC patients who had undergone an LC between January 2007 and December 2011 were retrospectively reviewed and compared with the medical records of CC patients who had undergone an LC within 72 hours of (group I, n=63) or 5 days after PTGBD (group II, n=40). In addition, group I was divided into group Ia (n=46) and group Ib (n=17), according to the patients' development of open-conversion or post-operative complications. The clinical outcomes of the four groups were analyzed. RESULTS: There was a significantly higher reference to age, the ASA score grading, and predominant comorbidities in group II than in group I. The peri-operative results of group II showed lower blood loss and relatively shorter operating times than those of group I. In the cases of early LC within 72 hours (group Ia vs. group Ib), the difference was statistically insignificant. CONCLUSIONS: The delayed LC after PTGBD for complicated cholecystitis with high clinical risk had better results in this study, although it prolonged the patient's hospital stay.

17.
J Clin Invest ; 121(12): 4636-9, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22105176

ABSTRACT

Decline in immune function with age has been attributed to defects or alterations in both the innate and the adaptive immune system. In this issue of the JCI, Zhao and coworkers provide evidence for a novel mechanism of immune dysfunction in aging mice. They show that migration of respiratory DCs from the site of virus replication to the draining lymph nodes in response to infection with several different respiratory viruses is markedly diminished with increasing age. The impaired DC migration was a result of increased levels of the lipid mediator prostaglandin D(2) (PGD(2)) in the respiratory tract with age and could be partially reversed by blockade of PGD(2) synthesis or action.


Subject(s)
Aging/metabolism , Coronavirus Infections/immunology , Dendritic Cells/pathology , Orthomyxoviridae Infections/immunology , Prostaglandin D2/physiology , Respiratory Syncytial Virus Infections/immunology , T-Lymphocyte Subsets/immunology , Animals
18.
Trends Immunol ; 32(5): 225-31, 2011 May.
Article in English | MEDLINE | ID: mdl-21435950

ABSTRACT

The 2009 influenza pandemic highlighted the threat that type A influenza poses to human health. Thus, there is an urgency to understand the pathobiology of influenza infection and the contribution of the host immune response to virus elimination and the development of lung injury. This review focuses on the T cell arm of the adaptive host immune response to influenza. We assess recent developments in the understanding of how primary influenza virus-specific T cell responses are induced by antigen-presenting cells, the interaction of activated effector T cells with antigen-bearing cells in the infected lungs. Also examined is the contribution of influenza-specific effector T cells to the development and control of lung injury and inflammation during infection.


Subject(s)
Influenza A virus/immunology , Influenza, Human/immunology , Lymphocyte Activation/immunology , T-Lymphocytes/immunology , Adaptive Immunity , Animals , Antigen-Presenting Cells/immunology , Dendritic Cells/immunology , Humans , Inflammation/immunology , Influenza, Human/virology , Lung/immunology , Lung/virology , Mice
19.
J Exp Med ; 208(1): 167-80, 2011 Jan 17.
Article in English | MEDLINE | ID: mdl-21187318

ABSTRACT

Cytotoxic T lymphocytes (CTLs) play a prominent role in the resolution of viral infections through their capacity both to mediate contact-dependent lysis of infected cells and to release soluble proinflammatory cytokines and chemokines. The factors controlling these antiviral effector activities in vivo at infection sites are ill defined. Using a mouse model of influenza infection, we observed that the expression of CTL effector activity in the infected lungs is dictated by the target cell type encountered. CD45(+) lung infiltrating inflammatory mononuclear cells, particularly CD11c(hi) dendritic cells, trigger both CTL cytotoxicity and release of inflammatory mediators, whereas CD45(-) influenza-infected respiratory epithelial cells stimulate only CTL cytotoxicity. CTL proinflammatory mediator release is modulated by co-stimulatory ligands (CD80 and CD86) expressed by the CD45(+) inflammatory cells. These findings suggest novel mechanisms of control of CTL effector activity and have potentially important implications for the control of excess pulmonary inflammation and immunopathology while preserving optimal viral clearance during respiratory virus infections.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Leukocyte Common Antigens/immunology , Animals , CD11 Antigens/immunology , Coculture Techniques , Female , Histocompatibility Antigens Class I/immunology , Influenza A virus/immunology , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Kinetics , Mice
20.
PLoS One ; 5(11): e15423, 2010 Nov 08.
Article in English | MEDLINE | ID: mdl-21079741

ABSTRACT

A hallmark of cells comprising the mammalian adaptive immune system is the requirement for these rare naïve T (and B) lymphocytes directed to a specific microorganism to undergo proliferative expansion upon first encounter with this antigen. In the case of naïve CD8(+) T cells the ability of these rare quiescent lymphocytes to rapidly activate and expand into effector T cells in numbers sufficient to control viral and certain bacterial infections can be essential for survival. In this report we examined the activation, cell cycle time and initial proliferative response of naïve murine CD8(+) T cells responding in vivo to Influenza and Vaccinia virus infection or vaccination with viral antigens. Remarkably, we observed that CD8(+) T cells could divide and proliferate with an initial cell division time of as short as 2 hours. The initial cell cycle time of responding CD8(+) T cells is not fixed but is controlled by the antigenic stimulus provided by the APC in vivo. Initial cell cycle time influences the rate of T cell expansion and the numbers of effector T cells subsequently accumulating at the site of infection. The T cell cycle time varies with duration of the G(1) phase of the cell cycle. The duration of G(1) is inversely correlated with the phosphorylation state of the retinoblastoma (Rb) protein in the responding T cells. The implication of these findings for the development of adaptive immune responses and the regulation of cell cycle in higher eukaryotic cells is discussed.


Subject(s)
Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Cycle/immunology , Lymphocyte Activation/immunology , Animals , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/virology , Cell Cycle/genetics , Cell Division/genetics , Cell Division/immunology , Cell Proliferation , Cells, Cultured , DNA/genetics , DNA/metabolism , Flow Cytometry , Gene Expression Profiling , Lymph Nodes/immunology , Mice , Mice, Inbred BALB C , Oligonucleotide Array Sequence Analysis , Orthomyxoviridae/immunology , Time Factors , Vaccinia virus/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...