Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters











Publication year range
1.
Cancer Immunol Immunother ; 73(10): 209, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39112670

ABSTRACT

BACKGROUND: Cancer immunotherapy approaches that elicit immune cell responses, including T and NK cells, have revolutionized the field of oncology. However, immunosuppressive mechanisms restrain immune cell activation within solid tumors so additional strategies to augment activity are required. METHODS: We identified the co-stimulatory receptor NKG2D as a target based on its expression on a large proportion of CD8+ tumor infiltrating lymphocytes (TILs) from breast cancer patient samples. Human and murine surrogate NKG2D co-stimulatory receptor-bispecifics (CRB) that bind NKG2D on NK and CD8+ T cells as well as HER2 on breast cancer cells (HER2-CRB) were developed as a proof of concept for targeting this signaling axis in vitro and in vivo. RESULTS: HER2-CRB enhanced NK cell activation and cytokine production when co-cultured with HER2 expressing breast cancer cell lines. HER2-CRB when combined with a T cell-dependent-bispecific (TDB) antibody that synthetically activates T cells by crosslinking CD3 to HER2 (HER2-TDB), enhanced T cell cytotoxicity, cytokine production and in vivo antitumor activity. A mouse surrogate HER2-CRB (mHER2-CRB) improved in vivo efficacy of HER2-TDB and augmented NK as well as T cell activation, cytokine production and effector CD8+ T cell differentiation. CONCLUSION: We demonstrate that targeting NKG2D with bispecific antibodies (BsAbs) is an effective approach to augment NK and CD8+ T cell antitumor immune responses. Given the large number of ongoing clinical trials leveraging NK and T cells for cancer immunotherapy, NKG2D-bispecifics have broad combinatorial potential.


Subject(s)
Breast Neoplasms , CD8-Positive T-Lymphocytes , Killer Cells, Natural , NK Cell Lectin-Like Receptor Subfamily K , Humans , Animals , NK Cell Lectin-Like Receptor Subfamily K/metabolism , NK Cell Lectin-Like Receptor Subfamily K/immunology , Mice , CD8-Positive T-Lymphocytes/immunology , Killer Cells, Natural/immunology , Female , Breast Neoplasms/immunology , Breast Neoplasms/therapy , Receptor, ErbB-2/immunology , Cell Line, Tumor , Immunotherapy/methods , Lymphocyte Activation/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism
2.
Cancer Immunol Res ; 8(6): 806-818, 2020 06.
Article in English | MEDLINE | ID: mdl-32238381

ABSTRACT

Antiangiogenic therapies that target the VEGF pathway have been used clinically to combat cancer for over a decade. Beyond having a direct impact on blood vessel development and tumor perfusion, accumulating evidence indicates that these agents also affect antitumor immune responses. Numerous clinical trials combining antiangiogenic drugs with immunotherapies for the treatment of cancer are ongoing, but a mechanistic understanding of how disruption of tumor angiogenesis may impact immunity is not fully discerned. Here, we reveal that blockade of VEGF-A with a mAb to VEGF augments activation of CD8+ T cells within tumors and potentiates their capacity to produce cytokines. We demonstrate that this phenomenon relies on the disruption of VEGFR2 signaling in the tumor microenvironment but does not affect CD8+ T cells directly. Instead, the augmented functional capacity of CD8+ T cells stems from increased tumor hypoxia that initiates a hypoxia-inducible factor-1α program within CD8+ T cells that directly enhances cytokine production. Finally, combinatorial administration of anti-VEGF with an immunotherapeutic antibody, anti-OX40, improved antitumor activity over single-agent treatments. Our findings illustrate that anti-VEGF treatment enhances CD8+ T-cell effector function and provides a mechanistic rationale for combining antiangiogenic and immunotherapeutic drugs for cancer treatment.


Subject(s)
Bevacizumab/pharmacology , CD8-Positive T-Lymphocytes/immunology , Colonic Neoplasms/therapy , Hypoxia/pathology , Lymphocyte Activation/immunology , Melanoma, Experimental/therapy , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Angiogenesis Inhibitors/pharmacology , Animals , Apoptosis , CD8-Positive T-Lymphocytes/drug effects , Cell Proliferation , Colonic Neoplasms/immunology , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Cytotoxicity, Immunologic/immunology , Female , Humans , Hypoxia/immunology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunotherapy , Lymphocyte Activation/drug effects , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Tumor Cells, Cultured , Tumor Microenvironment , Vascular Endothelial Growth Factor A/immunology , Xenograft Model Antitumor Assays
3.
Sci Signal ; 11(547)2018 09 11.
Article in English | MEDLINE | ID: mdl-30206136

ABSTRACT

The Hippo signaling pathway regulates organ size and plays critical roles in maintaining tissue growth, homeostasis, and regeneration. Dysregulated in a wide spectrum of cancers, in mammals, this pathway is regulated by two key effectors, YAP and TAZ, that may functionally overlap. We found that TAZ promoted liver inflammation and tumor development. The expression of TAZ, but not YAP, in human liver tumors positively correlated with the expression of proinflammatory cytokines. Hyperactivated TAZ induced substantial myeloid cell infiltration into the liver and the secretion of proinflammatory cytokines through a TEAD-dependent mechanism. Furthermore, tumors with hyperactivated YAP and TAZ had distinct transcriptional signatures, which included the increased expression of inflammatory cytokines in TAZ-driven tumors. Our study elucidated a previously uncharacterized link between TAZ activity and inflammatory responses that influence tumor development in the liver.


Subject(s)
DNA-Binding Proteins/genetics , Inflammation/genetics , Intracellular Signaling Peptides and Proteins/genetics , Liver Neoplasms/genetics , Liver/metabolism , Nuclear Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Transcription Factors/genetics , Animals , Cell Cycle Proteins , Cytokines/genetics , Cytokines/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Profiling/methods , Hippo Signaling Pathway , Humans , Inflammation/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Liver/pathology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Mice, Inbred C57BL , Mutation , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/genetics , TEA Domain Transcription Factors , Trans-Activators , Transcription Factors/metabolism , Transcriptional Coactivator with PDZ-Binding Motif Proteins , Transplantation, Heterologous
4.
Clin Cancer Res ; 24(24): 6447-6458, 2018 12 15.
Article in English | MEDLINE | ID: mdl-29950350

ABSTRACT

PURPOSE: The response to cancer immune therapy is dependent on endogenous tumor-reactive T cells. To bypass this requirement, CD3-bispecific antibodies have been developed to induce a polyclonal T-cell response against the tumor. Anti-HER2/CD3 T-cell-dependent bispecific (TDB) antibody is highly efficacious in the treatment of HER2-overexpressing tumors in mice. Efficacy and immunologic effects of anti-HER2/CD3 TDB were investigated in mammary tumor model with very few T cells prior treatment. We further describe the mechanism for TDB-induced T-cell recruitment to tumors. EXPERIMENTAL DESIGN: The immunologic effects and the mechanism of CD3-bispecific antibody-induced T-cell recruitment into spontaneous HER2-overexpressing mammary tumors was studied using human HER2 transgenic, immunocompetent mouse models. RESULTS: Anti-HER2/CD3 TDB treatment induced an inflammatory response in tumors converting them from poorly infiltrated to an inflamed, T-cell abundant, phenotype. Multiple mechanisms accounted for the TDB-induced increase in T cells within tumors. TDB treatment induced CD8+ T-cell proliferation. T cells were also actively recruited post-TDB treatment by IFNγ-dependent T-cell chemokines mediated via CXCR3. This active T-cell recruitment by TDB-induced chemokine signaling was the dominant mechanism and necessary for the therapeutic activity of anti-HER2/CD3 TDB. CONCLUSIONS: In summary, we demonstrate that the activity of anti-HER2/CD3 TDB was not dependent on high-level baseline T-cell infiltration. Our results suggest that anti-HER2/CD3 TDB may be efficacious in patients and indications that respond poorly to checkpoint inhibitors. An active T-cell recruitment mediated by TDB-induced chemokine signaling was the major mechanism for T-cell recruitment.


Subject(s)
Antibodies, Bispecific/pharmacology , CD3 Complex/antagonists & inhibitors , Chemokines/metabolism , Interferon-gamma/metabolism , Neoplasms/metabolism , Receptor, ErbB-2/antagonists & inhibitors , Receptors, CXCR3/metabolism , T-Lymphocytes/metabolism , Adoptive Transfer , Animals , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Cytokines/metabolism , Disease Models, Animal , Female , Humans , Inflammation Mediators/metabolism , Lymphocyte Activation/immunology , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Mice , Neoplasms/drug therapy , Neoplasms/etiology , Neoplasms/pathology , Signal Transduction , T-Lymphocytes/immunology , Xenograft Model Antitumor Assays
5.
Proc Natl Acad Sci U S A ; 111(24): 8925-30, 2014 Jun 17.
Article in English | MEDLINE | ID: mdl-24889626

ABSTRACT

The outcome of a viral infection reflects the balance between virus virulence and host susceptibility. The clone 13 (Cl13) variant of lymphocytic choriomeningitis virus--a prototype of Old World arenaviruses closely related to Lassa fever virus--elicits in C57BL/6 and BALB/c mice abundant negative immunoregulatory molecules, associated with T-cell exhaustion, negligible T-cell-mediated injury, and high virus titers that persist. Conversely, here we report that in NZB mice, despite the efficient induction of immunoregulatory molecules and high viremia, Cl13 generated a robust cytotoxic T-cell response, resulting in thrombocytopenia, pulmonary endothelial cell loss, vascular leakage, and death within 6-8 d. These pathogenic events required type I IFN (IFN-I) signaling on nonhematopoietic cells and were completely abrogated by IFN-I receptor blockade. Thus, IFN-I may play a prominent role in hemorrhagic fevers and other acute virus infections associated with severe vascular pathology, and targeting IFN-I or downstream effector molecules may be an effective therapeutic approach.


Subject(s)
Interferon Type I/metabolism , Lassa Fever/virology , Vascular Diseases/virology , Animals , Bronchoalveolar Lavage , Cell Line , Cricetinae , Cytokines/metabolism , Female , Lassa virus , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred NZB , Mice, Transgenic , Signal Transduction , Stem Cells/chemistry , T-Lymphocytes, Cytotoxic/virology , Virus Activation
6.
J Virol ; 88(11): 6281-93, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24672024

ABSTRACT

UNLABELLED: The cytokine storm is an intensified, dysregulated, tissue-injurious inflammatory response driven by cytokine and immune cell components. The cytokine storm during influenza virus infection, whereby the amplified innate immune response is primarily responsible for pulmonary damage, has been well characterized. Now we describe a novel event where virus-specific T cells induce a cytokine storm. The paramyxovirus pneumonia virus of mice (PVM) is a model of human respiratory syncytial virus (hRSV). Unexpectedly, when C57BL/6 mice were infected with PVM, the innate inflammatory response was undetectable until day 5 postinfection, at which time CD8(+) T cells infiltrated into the lung, initiating a cytokine storm by their production of gamma interferon (IFN-γ) and tumor necrosis factor alpha (TNF-α). Administration of an immunomodulatory sphingosine-1-phosphate (S1P) receptor 1 (S1P1R) agonist significantly inhibited PVM-elicited cytokine storm by blunting the PVM-specific CD8(+) T cell response, resulting in diminished pulmonary disease and enhanced survival. IMPORTANCE: A dysregulated overly exuberant immune response, termed a "cytokine storm," accompanies virus-induced acute respiratory diseases (VARV), is primarily responsible for the accompanying high morbidity and mortality, and can be controlled therapeutically in influenza virus infection of mice and ferrets by administration of sphingosine-1-phosphate 1 receptor (S1P1R) agonists. Here, two novel findings are recorded. First, in contrast to influenza infection, where the cytokine storm is initiated early by the innate immune system, for pneumonia virus of mice (PVM), a model of RSV, the cytokine storm is initiated late in infection by the adaptive immune response: specifically, by virus-specific CD8 T cells via their release of IFN-γ and TNF-α. Blockading these cytokines with neutralizing antibodies blunts the cytokine storm and protects the host. Second, PVM infection is controlled by administration of an S1P1R agonist.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cytokines/immunology , Disease Models, Animal , Murine pneumonia virus , Pneumovirus Infections/immunology , Respiratory Syncytial Viruses , Animals , Antibodies/administration & dosage , Cytokines/metabolism , Flow Cytometry , Immunoglobulin G , Interferon-gamma/immunology , Lung/immunology , Mice , Mice, Inbred C57BL , Receptors, Lysosphingolipid/agonists , Sphingosine-1-Phosphate Receptors , Tumor Necrosis Factor-alpha/immunology
7.
Virology ; 452-453: 152-7, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24606692

ABSTRACT

Influenza infection of humans remains an important public health problem. Vaccine strategies result in a significant but only partial control (65-85%) of infection. Thus, chemotherapeutic approaches are needed to provide a solution both for vaccine failures and to limit infection in the unvaccinated population. Previously (Walsh et al., 2011; Teijaro et al., 2011) documented that sphingosine-1-phosphate 1 receptor (S1P1R) agonists significantly protected mice against pathogenic H1N1 influenza virus by limiting immunopathologic damage while allowing host control of the infection. Here we extend that observation by documenting S1P1R agonist can control pathogenic H1N1 influenza infection in ferrets. S1P1R agonist was more effective in reducing pulmonary injury than the antiviral drug oseltamivir but, importantly, combined therapy was significantly more effective than either therapy alone.


Subject(s)
Antibodies, Viral/immunology , Antiviral Agents/administration & dosage , Influenza A Virus, H1N1 Subtype/drug effects , Influenza, Human/drug therapy , Lung/pathology , Receptors, Lysosphingolipid/agonists , Animals , Disease Models, Animal , Ferrets , Humans , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/physiology , Influenza, Human/immunology , Influenza, Human/pathology , Influenza, Human/virology , Lung/immunology , Lung/virology , Male , Sphingosine-1-Phosphate Receptors
8.
Proc Natl Acad Sci U S A ; 111(10): 3799-804, 2014 Mar 11.
Article in English | MEDLINE | ID: mdl-24572573

ABSTRACT

During pathogenic influenza virus infection, robust cytokine production (cytokine storm), excessive inflammatory infiltrates, and virus-induced tissue destruction all contribute to morbidity and mortality. Earlier we reported that modulation of sphingosine-1-phosphate-1 receptor (S1P1R) signaling provided a chemically tractable approach for the effective blunting of cytokine storm, leading to the improvement of clinical and survival outcomes. Here, we show that S1P1R agonist treatment suppresses global cytokine amplification. Importantly, S1P1R agonist treatment was able to blunt cytokine/chemokine production and innate immune cell recruitment in the lung independently of endosomal and cytosolic innate sensing pathways. S1P1R signaling suppression of cytokine amplification was independent of multiple innate signaling adaptor pathways for myeloid differentiation primary response gene 88 (MyD88) and IFN-ß promoter stimulator-1 signaling, indicating a common pathway inhibition of cytokine storm. We identify the MyD88 adaptor molecule as responsible for the majority of cytokine amplification observed following influenza virus challenge.


Subject(s)
Cytokines/immunology , Immunity, Innate/immunology , Orthomyxoviridae Infections/immunology , Signal Transduction/immunology , Animals , Bone Marrow Transplantation , Dogs , Flow Cytometry , Madin Darby Canine Kidney Cells , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88 , Orthomyxoviridae Infections/physiopathology , Receptors, Lysosphingolipid/metabolism
9.
J Virol ; 87(17): 9949-52, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23824814

ABSTRACT

The paramyxovirus pneumonia virus of mice (PVM) is a rodent model of human respiratory syncytial virus (hRSV) pathogenesis. Here we characterized the PVM-specific CD8(+) T-cell repertoire in susceptible C57BL/6 mice. In total, 15 PVM-specific CD8(+) T-cell epitopes restricted by H-2D(b) and/or H-2K(b) were identified. These data open the door for using widely profiled, genetically manipulated C57BL/6 mice to study the contribution of epitope-specific CD8(+) T cells to PVM pathogenesis.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/virology , Murine pneumonia virus/immunology , Amino Acid Sequence , Animals , Antigens, Viral/genetics , Epitope Mapping , Epitopes, T-Lymphocyte/genetics , H-2 Antigens/metabolism , Histocompatibility Antigen H-2D/metabolism , Humans , Interferon-gamma/biosynthesis , Mice , Mice, Inbred C57BL , Murine pneumonia virus/genetics , Murine pneumonia virus/pathogenicity , Pneumovirus Infections/immunology , Pneumovirus Infections/virology , Viral Proteins/genetics , Viral Proteins/immunology
10.
PLoS One ; 8(3): e58033, 2013.
Article in English | MEDLINE | ID: mdl-23533579

ABSTRACT

Influenza-induced lung edema and inflammation are exacerbated by a positive feedback loop of cytokine and chemokine production termed a 'cytokine storm', a hallmark of increased influenza-related morbidity and mortality. Upon infection, an immune response is rapidly initiated in the lungs and draining lymph node, leading to expansion of virus-specific effector cells. Using two-photon microscopy, we imaged the dynamics of dendritic cells (DC) and virus-specific eGFP(+)CD8(+) T cells in the lungs and draining mediastinal lymph nodes during the first two weeks following influenza infection. Three distinct phases of T cell and CD11c(+) DC behavior were revealed: 1) Priming, facilitated by the arrival of lung DCs in the lymph node and characterized by antigen recognition and expansion of antigen-specific CD8(+) T cells; asymmetric T cell division in contact with DCs was frequently observed. 2) Clearance, during which DCs re-populate the lung and T cells leave the draining lymph node and re-enter the lung tissue where enlarged, motile T cells come into contact with DCs and form long-lived interactions. 3) Maintenance, characterized by T-cell scanning of the lung tissue and dissociation from local antigen presenting cells; the T cells spend less time in association with DCs and migrate rapidly on collagen. A single dose of a sphingosine-1-phosphate receptor agonist, AAL-R, sufficient to suppress influenza-induced cytokine-storm, altered T cell and DC behavior during influenza clearance, delaying T cell division, cellular infiltration in the lung, and suppressing T-DC interactions in the lung. Our results provide a detailed description of T cell and DC choreography and dynamics in the lymph node and the lung during influenza infection. In addition, we suggest that phase lags in T cell and DC dynamics induced by targeting S1P receptors in vivo may attenuate the intensity of the immune response and can be manipulated for therapeutic benefit.


Subject(s)
Antiviral Agents/therapeutic use , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/physiology , Influenza A Virus, H1N1 Subtype/pathogenicity , Lung/immunology , Lung/virology , Lysophospholipids/agonists , Orthomyxoviridae Infections/drug therapy , Orthomyxoviridae Infections/immunology , Sphingosine/analogs & derivatives , Animals , CD11c Antigen/metabolism , Cells, Cultured , Dendritic Cells/metabolism , Dendritic Cells/physiology , Flow Cytometry , Lung/metabolism , Mice , Orthomyxoviridae Infections/metabolism , Sphingosine/agonists
11.
Virology ; 435(1): 92-101, 2013 Jan 05.
Article in English | MEDLINE | ID: mdl-23217619

ABSTRACT

The cytokine storm is an aggressive immune response characterized by the recruitment of inflammatory leukocytes and exaggerated levels of cytokines and chemokines at the site of infection. Here we review evidence that cytokine storm directly contributes to the morbidity and mortality resulting from influenza virus infection and that sphingosine-1-phosphate (S1P) receptor agonists can abort cytokine storms providing significant protection against pathogenic human influenza viral infections. In experiments using murine models and the human pathogenic 2009 influenza viruses, S1P1 receptor agonist alone reduced deaths from influenza virus by over 80% as compared to lesser protection (50%) offered by the antiviral neuraminidase inhibitor oseltamivir. Optimal protection of 96% was achieved by combined therapy with the S1P1 receptor agonist and oseltamivir. The functional mechanism of S1P receptor agonist(s) action and the predominant role played by pulmonary endothelial cells as amplifiers of cytokine storm during influenza infection are described.


Subject(s)
Enzyme Inhibitors/pharmacology , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza, Human/drug therapy , Mycotoxins/pharmacology , Oseltamivir/pharmacology , Receptors, Lysosphingolipid/agonists , Animals , Cytokines/antagonists & inhibitors , Cytokines/biosynthesis , Drug Synergism , Drug Therapy, Combination , Endothelial Cells/drug effects , Endothelial Cells/pathology , Endothelial Cells/virology , Enzyme Inhibitors/therapeutic use , Humans , Influenza A Virus, H1N1 Subtype/physiology , Influenza, Human/mortality , Influenza, Human/pathology , Influenza, Human/virology , Lung/drug effects , Lung/pathology , Lung/virology , Mice , Mycotoxins/therapeutic use , Orthomyxoviridae Infections/drug therapy , Orthomyxoviridae Infections/mortality , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/virology , Oseltamivir/therapeutic use , Receptors, Lysosphingolipid/genetics , Survival Rate
12.
Cell Host Microbe ; 11(6): 643-53, 2012 Jun 14.
Article in English | MEDLINE | ID: mdl-22704624

ABSTRACT

TLR7 is an innate signaling receptor that recognizes single-stranded viral RNA and is activated by viruses that cause persistent infections. We show that TLR7 signaling dictates either clearance or establishment of life-long chronic infection by lymphocytic choriomeningitis virus (LCMV) Cl 13 but does not affect clearance of the acute LCMV Armstrong 53b strain. TLR7(-/-) mice infected with LCMV Cl 13 remained viremic throughout life from defects in the adaptive antiviral immune response-notably, diminished T cell function, exacerbated T cell exhaustion, decreased plasma cell maturation, and negligible antiviral antibody production. Adoptive transfer of TLR7(+/+) LCMV immune memory cells that enhanced clearance of persistent LCMV Cl 13 infection in TLR7(+/+) mice failed to purge LCMV Cl 13 infection in TLR7(-/-) mice, demonstrating that a TLR7-deficient environment renders antiviral responses ineffective. Therefore, methods that promote TLR7 signaling are promising treatment strategies for chronic viral infections.


Subject(s)
Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Membrane Glycoproteins/immunology , Toll-Like Receptor 7/immunology , Adoptive Transfer , Animals , Antibodies, Viral/blood , Disease Models, Animal , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Plasma Cells/immunology , T-Lymphocytes/immunology
13.
Immunol Res ; 51(1): 15-25, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21901448

ABSTRACT

Initial and early tissue injury associated with severe influenza virus infection is the result of both virus-mediated lysis of infected pulmonary cells coupled with an exuberant immune response generated against the virus. The excessive host immune response associated with influenza virus infection has been termed "cytokine storm." Therapies that target virus replication are available; however, the selective pressure by such antiviral drugs on the virus often results in mutation and the escape of virus progeny now resistant to the antiviral regimen, thereby rendering such treatments ineffective. This event highlights the necessity for developing novel methods to combat morbidity and mortality caused by influenza virus infection. One potential method is restricting the host's immune response. However, prior treatment regimens employing drugs like corticosteroids that globally suppress the host's immune response were found unsatisfactory in large part because they disrupted the host's ability to control virus replication. Here, we discuss a novel therapy that utilizes sphingosine-1-phosphate (S1P) receptor signaling that has the ability to significantly limit immunopathologic injury caused by the host's innate and adaptive immune response, thereby significantly aborting morbidity and mortality associated with influenza virus infection. Moreover, S1P analog therapy allows for sufficient anti-influenza T cell and antibody formation to control infection. We review the anti-inflammatory effects of S1P signaling pathways and how modulation of these pathways during influenza virus infection restricts immunopathology. Finally, we discuss that combinatorial administration of S1P simultaneously with a current antiviral enhances the treatment efficacy for virulent influenza virus infections above that of either drug treatment alone. Interestingly, the scope of S1P receptor therapy reported here is likely to extend beyond influenza virus infection and could prove useful for the treatment of multiple maladies like other viral infections and autoimmune diseases where the host's inflammatory response is a major component in the disease process.


Subject(s)
Antiviral Agents/therapeutic use , Cytokines/immunology , Orthomyxoviridae Infections/drug therapy , Orthomyxoviridae Infections/immunology , Orthomyxoviridae/immunology , Receptors, Lysosphingolipid/immunology , Animals , Antibodies, Viral/immunology , Drug Resistance, Viral/drug effects , Drug Resistance, Viral/genetics , Drug Resistance, Viral/immunology , Humans , Lung/immunology , Lung/virology , Mutation , Orthomyxoviridae/genetics , Orthomyxoviridae Infections/genetics , Receptors, Lysosphingolipid/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/immunology , T-Lymphocytes/immunology , T-Lymphocytes/virology
14.
Cell ; 146(6): 980-91, 2011 Sep 16.
Article in English | MEDLINE | ID: mdl-21925319

ABSTRACT

Cytokine storm during viral infection is a prospective predictor of morbidity and mortality, yet the cellular sources remain undefined. Here, using genetic and chemical tools to probe functions of the S1P(1) receptor, we elucidate cellular and signaling mechanisms that are important in initiating cytokine storm. Whereas S1P(1) receptor is expressed on endothelial cells and lymphocytes within lung tissue, S1P(1) agonism suppresses cytokines and innate immune cell recruitment in wild-type and lymphocyte-deficient mice, identifying endothelial cells as central regulators of cytokine storm. Furthermore, our data reveal immune cell infiltration and cytokine production as distinct events that are both orchestrated by endothelial cells. Moreover, we demonstrate that suppression of early innate immune responses through S1P(1) signaling results in reduced mortality during infection with a human pathogenic strain of influenza virus. Modulation of endothelium with a specific agonist suggests that diseases in which amplification of cytokine storm is a significant pathological component could be chemically tractable.


Subject(s)
Cytokines/immunology , Endothelial Cells/immunology , Influenza A Virus, H1N1 Subtype/physiology , Influenza, Human/immunology , Orthomyxoviridae Infections/immunology , Animals , Disease Models, Animal , Humans , Interferons/immunology , Lung/cytology , Lung/immunology , Lung/virology , Lymphocytes/immunology , Mice , Mice, Inbred C57BL , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/virology , Receptors, Lysosphingolipid/agonists , Signal Transduction
15.
Proc Natl Acad Sci U S A ; 108(29): 12018-23, 2011 Jul 19.
Article in English | MEDLINE | ID: mdl-21715659

ABSTRACT

Human pandemic H1N1 2009 influenza virus rapidly infected millions worldwide and was associated with significant mortality. Antiviral drugs that inhibit influenza virus replication are the primary therapy used to diminish disease; however, there are two significant limitations to their effective use: (i) antiviral drugs exert selective pressure on the virus, resulting in the generation of more fit viral progeny that are resistant to treatment; and (ii) antiviral drugs do not directly inhibit immune-mediated pulmonary injury that is a significant component of disease. Here we show that dampening the host's immune response against influenza virus using an immunomodulatory drug, AAL-R, provides significant protection from mortality (82%) over that of the neuraminidase inhibitor oseltamivir alone (50%). AAL-R combined with oseltamivir provided maximum protection against a lethal challenge of influenza virus (96%). Mechanistically, AAL-R inhibits cellular and cytokine/chemokine responses to limit immunopathologic damage, while maintaining host control of virus replication. With cytokine storm playing a role in the pathogenesis of a wide assortment of viral, bacterial, and immunologic diseases, a therapeutic approach using sphingosine analogs is of particular interest.


Subject(s)
Cytokines/immunology , Immunomodulation/immunology , Influenza A virus/immunology , Orthomyxoviridae Infections/drug therapy , Oseltamivir/pharmacology , Sphingosine/pharmacology , Alternaria/chemistry , Animals , Bronchoalveolar Lavage Fluid/chemistry , Cell Line , Cytokines/metabolism , Dogs , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Male , Mice , Mice, Inbred C57BL , Neutralization Tests , Orthomyxoviridae Infections/immunology , Oseltamivir/metabolism , Oseltamivir/therapeutic use , Sphingosine/metabolism , Sphingosine/therapeutic use
16.
PLoS One ; 5(3): e9915, 2010 Mar 29.
Article in English | MEDLINE | ID: mdl-20369005

ABSTRACT

BACKGROUND: Alpha-dystroglycan (alpha-DG) is a cell surface receptor providing a molecular link between the extracellular matrix (ECM) and the actin-based cytoskeleton. During its biosynthesis, alpha-DG undergoes specific and unusual O-glycosylation crucial for its function as a high-affinity cellular receptor for ECM proteins. METHODOLOGY/PRINCIPAL FINDINGS: We report that expression of functionally glycosylated alpha-DG during thymic development is tightly regulated in developing T cells and largely confined to CD4(-)CD8(-) double negative (DN) thymocytes. Ablation of DG in T cells had no effect on proliferation, migration or effector function but did reduce the size of the thymus due to a significant loss in absolute numbers of thymocytes. While numbers of DN thymocytes appeared normal, a marked reduction in CD4(+)CD8(+) double positive (DP) thymocytes occurred. In the periphery mature naïve T cells deficient in DG showed both normal proliferation in response to allogeneic cells and normal migration, effector and memory T cell function when tested in acute infection of mice with either lymphocytic choriomeningitis virus (LCMV) or influenza virus. CONCLUSIONS/SIGNIFICANCE: Our study demonstrates that DG function is modulated by glycosylation during T cell development in vivo and that DG is essential for normal development and differentiation of T cells.


Subject(s)
Dystroglycans/chemistry , Dystroglycans/metabolism , Glycosylation , Thymus Gland/cytology , Actins/chemistry , Animals , Cell Cycle , Cell Membrane/metabolism , Cytoskeleton/metabolism , Extracellular Matrix/metabolism , Flow Cytometry/methods , Humans , Lymphocytic choriomeningitis virus/metabolism , Mice , Orthomyxoviridae/metabolism , T-Lymphocytes/cytology
17.
Proc Natl Acad Sci U S A ; 107(7): 3018-23, 2010 Feb 16.
Article in English | MEDLINE | ID: mdl-20133700

ABSTRACT

Mounting effective T cell responses is critical for eliciting long-lasting immunity following viral infection and vaccination. A multitude of inhibitory and stimulatory factors are induced following infection, and it is the compilation of these signals that quantitatively and qualitatively program the ensuing effector and memory T cell response. In response to lymphocytic choriomeningitis virus (LCMV) infection, the immunosuppressive cytokine IL-10 is rapidly up-regulated; however, how IL-10 is regulating what is often considered an "optimal" immune response is unclear. We demonstrate that IL-10 directly inhibits effector and memory CD4 T cell responses following an acutely resolved viral infection. Blockade of IL-10 enhanced the magnitude and the functional capacity of effector CD4 T cells that translated into increased and more effective memory responses. On the other hand, lack of IL-10 signaling did not impact memory CD8 T cell development. We propose that blockade of IL-10 may be an effective adjuvant to specifically enhance CD4 T cell immunity and protection following vaccination.


Subject(s)
Arenaviridae Infections/immunology , CD4-Positive T-Lymphocytes/immunology , Gene Expression Regulation/immunology , Immunologic Memory/immunology , Interleukin-10/immunology , Lymphocytic choriomeningitis virus/immunology , Animals , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Interleukin-10/metabolism , Mice , Mice, Inbred C57BL , Statistics, Nonparametric
18.
Virology ; 397(2): 260-9, 2010 Feb 20.
Article in English | MEDLINE | ID: mdl-19962171

ABSTRACT

There is no known antiviral drug treatment that routinely terminates persistent virus infections. A recent provocative report indicated that low dosage of the sphingosine analog FTY720 caused lymphopenia in mice persistently infected with lymphocytic choriomeningitis virus (LCMV)-clone 13 (Cl 13) and induced viral clearance within 30 days post-treatment (Premenko-Lanier et al., 2008). However, we find that low dosage of FTY720 fails to purge LCMV-Cl 13 infection and does not induce lymphopenia in LCMV-Cl 13-infected mice. In fact, infection with non-persistent LCMV-Arm53b or with persistent LCMV-Cl 13 induces an equivalent lymphopenia, demonstrating that the quantity of circulating cells has little bearing on viral persistence. In addition, treatment with FTY720 or the sphingosine-1-phosphate receptor 1 (S1P1)-specific agonist, AUY954, does not alleviate T cell exhaustion and exacerbates disruption of the CD8(+) T cells response following LCMV-Cl 13 infection. Therefore, treatment with a sphingosine analog does not ameliorate persistent LCMV-Cl 13 infection.


Subject(s)
Antiviral Agents/therapeutic use , Lymphocytic Choriomeningitis/drug therapy , Propylene Glycols/therapeutic use , Sphingosine/analogs & derivatives , Animals , Fingolimod Hydrochloride , Lymphocytic choriomeningitis virus/isolation & purification , Lymphopenia/virology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Sphingosine/therapeutic use , Thiophenes/therapeutic use , Treatment Outcome , beta-Alanine/analogs & derivatives , beta-Alanine/therapeutic use
19.
Proc Natl Acad Sci U S A ; 106(5): 1560-5, 2009 Feb 03.
Article in English | MEDLINE | ID: mdl-19164548

ABSTRACT

Pulmonary tissue damage resulting from influenza virus infection is caused by both the cytolytic activity of the virus and the host immune response. Immune-mediated injury results from T cell-mediated destruction of virus-infected cells and by release of cytokines and chemokines that attract polymorphonuclear leukocytes (PML) and macrophages to the infected site. The cytokines/chemokines potentiate dendritic cell (DC) activation and T cell expansion, which further enhances local damage. Here we report that immune modulation by local administration to the respiratory tract of sphingosine analog AAL-R significantly dampens the release of cytokines and chemokines while maintaining protective neutralizing antibody and cytotoxic T cell responses. As a result there was a marked reduction of infiltrating PML and macrophages into the lung and resultant pulmonary tissue injury. DC maturation was suppressed, which limited proliferation of specific antiviral T cells in the lung and draining lymph nodes. Further, AAL-R was effective in controlling CD8(+) T cell accumulation in the lungs even when given 4 days after initiation of influenza virus infection. These data indicate that sphingosine analogs display useful potential for controlling the immunopathology caused by influenza virus.


Subject(s)
Cytokines/biosynthesis , Influenza, Human/physiopathology , Sphingosine/pharmacology , Amino Acid Sequence , Animals , Antibodies, Viral/biosynthesis , Antibodies, Viral/immunology , Antigen-Presenting Cells/immunology , Base Sequence , DNA, Viral , Disease Models, Animal , Humans , Influenza, Human/immunology , Influenza, Human/metabolism , Lung/immunology , Mice , Molecular Sequence Data , Neutralization Tests , T-Lymphocytes, Cytotoxic/immunology
20.
Mol Pharmacol ; 74(3): 896-903, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18577684

ABSTRACT

The mechanism by which locally delivered sphingosine analogs regulate host response to localized viral infection has never been addressed. In this report, we show that intratracheal delivery of the chiral sphingosine analog (R)-2-amino-4-(4-heptyloxyphenyl)-2-methylbutanol (AAL-R) or its phosphate ester inhibits the T-cell response to influenza virus infection. In contrast, neither intraperitoneal delivery of AAL-R nor intratracheal instillation of the non-phosphorylatable stereoisomer AAL-S suppressed virus-specific T-cell response, indicating that in vivo phosphorylation of AAL-R and sphingosine 1-phosphate (S1P) receptor modulation in lungs is essential for immunomodulation. Intratracheal delivery of water-soluble S1P(1) receptor agonist at doses sufficient to induce systemic lymphopenia did not inhibit virus-specific T-cell response, indicating that S1P(1) is not involved in the immunosuppressive activities of AAL-R and that immunosuppression acts independently of naive lymphocyte recirculation. Accumulation of dendritic cells (DCs) in draining lymph nodes was inhibited by intratracheal but not intraperitoneal delivery of AAL-R. Direct modulation of DCs is demonstrated by the impaired ability of virus-infected bone marrow-derived DCs treated in vitro with AAL-R to trigger in vivo T-cell response after adoptive transfer to the airways. Thus, our results suggest that locally delivered sphingosine analogs induce immunosuppression by modulating S1P receptors other than S1P(1) or S1P(2) on dendritic cells in the lungs after influenza virus infection.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/virology , Lung/immunology , Lung/virology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Receptors, Lysosphingolipid/immunology , Animals , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/virology , Cytokines/metabolism , Dendritic Cells/drug effects , Immune Tolerance/drug effects , Influenza A virus/drug effects , Lung/metabolism , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/drug effects , Male , Mice , Mice, Inbred C57BL , Phosphorylation/drug effects , Sphingosine/administration & dosage , Sphingosine/analogs & derivatives , Sphingosine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL