Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
PLoS Pathog ; 13(4): e1006305, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28380049

ABSTRACT

Antiviral responses must rapidly defend against infection while minimizing inflammatory damage, but the mechanisms that regulate the magnitude of response within an infected cell are not well understood. miRNAs are small non-coding RNAs that suppress protein levels by binding target sequences on their cognate mRNA. Here, we identify miR-144 as a negative regulator of the host antiviral response. Ectopic expression of miR-144 resulted in increased replication of three RNA viruses in primary mouse lung epithelial cells: influenza virus, EMCV, and VSV. We identified the transcriptional network regulated by miR-144 and demonstrate that miR-144 post-transcriptionally suppresses TRAF6 levels. In vivo ablation of miR-144 reduced influenza virus replication in the lung and disease severity. These data suggest that miR-144 reduces the antiviral response by attenuating the TRAF6-IRF7 pathway to alter the cellular antiviral transcriptional landscape.


Subject(s)
Influenza, Human/immunology , MicroRNAs/metabolism , Orthomyxoviridae/genetics , Signal Transduction , TNF Receptor-Associated Factor 6/genetics , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/genetics , Animals , Cell Line , Epithelial Cells/virology , Gene Expression Profiling , Genes, Reporter , Humans , Influenza, Human/virology , Lung/virology , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Orthomyxoviridae/immunology , Orthomyxoviridae/physiology , RNA Processing, Post-Transcriptional , RNA, Messenger/genetics , RNA, Messenger/metabolism , TNF Receptor-Associated Factor 6/metabolism , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/metabolism , Viral Load , Virus Replication
2.
J Gen Virol ; 95(Pt 2): 350-362, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24243730

ABSTRACT

Type I alveolar epithelial cells are a replicative niche for influenza in vivo, yet their response to infection is not fully understood. To better characterize their cellular responses, we have created an immortalized murine lung epithelial type I cell line (LET1). These cells support spreading influenza virus infection in the absence of exogenous protease and thus permit simultaneous analysis of viral replication dynamics and host cell responses. LET1 cells can be productively infected with human, swine and mouse-adapted strains of influenza virus and exhibit expression of an antiviral transcriptional programme and robust cytokine secretion. We characterized influenza virus replication dynamics and host responses of lung type I epithelial cells and identified the capacity of epithelial cell-derived type I IFN to regulate specific modules of antiviral effectors to establish an effective antiviral state. Together, our results indicate that the type I epithelial cell can play a major role in restricting influenza virus infection without contribution from the haematopoietic compartment.


Subject(s)
Epithelial Cells/immunology , Epithelial Cells/virology , Immunity, Innate , Influenza A virus/immunology , Influenza A virus/physiology , Virus Replication , Animals , Cell Line , Interferon Type I/immunology , Interferon Type I/metabolism , Mice , Mice, Inbred C57BL
3.
J Immunol ; 189(1): 23-7, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22661094

ABSTRACT

Cellular fusion of macrophages into multinucleated giant cells is a distinguishing feature of the granulomatous response to inflammation, infection, and foreign bodies (Kawai and Akira. 2011. Immunity 34: 637-650). We observed a marked increase in fusion of macrophages genetically deficient in Dicer, an enzyme required for canonical microRNA (miRNA) biogenesis. Gene expression profiling of miRNA-deficient macrophages revealed an upregulation of the IL-4-responsive fusion protein Tm7sf4, and analyses identified miR-7a-1 as a negative regulator of macrophage fusion, functioning by directly targeting Tm7sf4 mRNA. miR-7a-1 is itself an IL-4-responsive gene in macrophages, suggesting feedback control of cellular fusion. Collectively, these data indicate that miR-7a-1 functions to regulate IL-4-directed multinucleated giant cell formation.


Subject(s)
Cell Differentiation/immunology , Giant Cells, Langhans/immunology , Macrophages/cytology , Macrophages/immunology , MicroRNAs/physiology , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Cell Differentiation/genetics , Cell Fusion/methods , Cells, Cultured , DEAD-box RNA Helicases/deficiency , DEAD-box RNA Helicases/genetics , Giant Cells, Langhans/cytology , Giant Cells, Langhans/metabolism , HEK293 Cells , Humans , Interleukin-4/physiology , Macrophages/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , MicroRNAs/genetics , Ribonuclease III/deficiency , Ribonuclease III/genetics , Transcription, Genetic/immunology
4.
Proc Natl Acad Sci U S A ; 108(28): 11536-41, 2011 Jul 12.
Article in English | MEDLINE | ID: mdl-21709223

ABSTRACT

Precise control of the innate immune response is essential to ensure host defense against infection while avoiding inflammatory disease. Systems-level analyses of Toll-like receptor (TLR)-stimulated macrophages suggested that SHANK-associated RH domain-interacting protein (SHARPIN) might play a role in the TLR pathway. This hypothesis was supported by the observation that macrophages derived from chronic proliferative dermatitis mutation (cpdm) mice, which harbor a spontaneous null mutation in the Sharpin gene, exhibited impaired IL-12 production in response to TLR activation. Systems biology approaches were used to define the SHARPIN-regulated networks. Promoter analysis identified NF-κB and AP-1 as candidate transcription factors downstream of SHARPIN, and network analysis suggested selective attenuation of these pathways. We found that the effects of SHARPIN deficiency on the TLR2-induced transcriptome were strikingly correlated with the effects of the recently described hypomorphic L153P/panr2 point mutation in Ikbkg [NF-κB Essential Modulator (NEMO)], suggesting that SHARPIN and NEMO interact. We confirmed this interaction by co-immunoprecipitation analysis and furthermore found it to be abrogated by panr2. NEMO-dependent signaling was affected by SHARPIN deficiency in a manner similar to the panr2 mutation, including impaired p105 and ERK phosphorylation and p65 nuclear localization. Interestingly, SHARPIN deficiency had no effect on IκBα degradation and on p38 and JNK phosphorylation. Taken together, these results demonstrate that SHARPIN is an essential adaptor downstream of the branch point defined by the panr2 mutation in NEMO.


Subject(s)
Carrier Proteins/immunology , Carrier Proteins/metabolism , Toll-Like Receptor 2/immunology , Toll-Like Receptor 2/metabolism , Animals , Base Sequence , Carrier Proteins/genetics , DNA Primers/genetics , Immunity, Innate/genetics , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/immunology , Intracellular Signaling Peptides and Proteins/metabolism , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , NF-kappa B/metabolism , Protein Interaction Mapping , Signal Transduction , Systems Analysis , Systems Biology , Toll-Like Receptor 2/genetics , Transcription Factor AP-1/metabolism
5.
Nat Med ; 8(12): 1433-7, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12426560

ABSTRACT

Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) is a member of the TNF superfamily that exhibits specific tumoricidal activity against a variety of tumors. It is expressed on different cells of the immune system and plays a role in natural killer cell-mediated tumor surveillance. In allogeneic hematopoietic-cell transplantation, the reactivity of the donor T cell against malignant cells is essential for the graft-versus-tumor (GVT) effect. Cytolytic activity of T cells is primarily mediated through the Fas-Fas ligand and perforin-granzyme pathways. However, T cells deficient for both Fas ligand and perforin can still exert GVT activity in vivo in mouse models. To uncover a potential role for TRAIL in donor T cell-mediated GVT activity, we compared donor T cells from TRAIL-deficient and wild-type mice in clinically relevant mouse bone-marrow transplantation models. We found that alloreactive T cells can express TRAIL, but the absence of TRAIL had no effect on their proliferative and cytokine response to alloantigens. TRAIL-deficient T cells showed significantly lower GVT activity than did TRAIL-expressing T cells, but no important differences in graft-versus-host disease, a major complication of allogeneic hematopoietic cell transplantation, were observed. These data suggest that strategies to enhance TRAIL-mediated GVT activity could decrease relapse rates of malignancies after hematopoietic cell transplantation without exacerbation of graft-versus-host disease.


Subject(s)
Graft vs Tumor Effect/immunology , Membrane Glycoproteins/physiology , T-Lymphocytes/immunology , Tumor Necrosis Factor-alpha/physiology , Animals , Apoptosis Regulatory Proteins , Graft vs Host Disease/etiology , Hematopoietic Stem Cell Transplantation , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , TNF-Related Apoptosis-Inducing Ligand , Transplantation, Homologous , Tumor Cells, Cultured
6.
J Exp Med ; 198(10): 1495-506, 2003 Nov 17.
Article in English | MEDLINE | ID: mdl-14610045

ABSTRACT

Extensive studies of mice deficient in one or several cytokine receptors have failed to support an indispensable role of cytokines in development of multiple blood cell lineages. Whereas B1 B cells and Igs are sustained at normal levels throughout life of mice deficient in IL-7, IL-7Ralpha, common cytokine receptor gamma chain, or flt3 ligand (FL), we report here that adult mice double deficient in IL-7Ralpha and FL completely lack visible LNs, conventional IgM+ B cells, IgA+ plasma cells, and B1 cells, and consequently produce no Igs. All stages of committed B cell progenitors are undetectable in FL-/- x IL-7Ralpha-/- BM that also lacks expression of the B cell commitment factor Pax5 and its direct target genes. Furthermore, in contrast to IL-7Ralpha-/- mice, FL-/- x IL-7Ralpha-/- mice also lack mature B cells and detectable committed B cell progenitors during fetal development. Thus, signaling through the cytokine tyrosine kinase receptor flt3 and IL-7Ralpha are indispensable for fetal and adult B cell development.


Subject(s)
B-Lymphocytes/physiology , Cell Differentiation/physiology , Membrane Proteins/metabolism , Receptors, Interleukin-7/metabolism , Animals , B-Lymphocytes/cytology , Membrane Proteins/genetics , Membrane Proteins/immunology , Mice , Mice, Knockout , Peyer's Patches/metabolism , Receptors, Interleukin-7/genetics , Receptors, Interleukin-7/immunology , Signal Transduction/immunology , Signal Transduction/physiology
7.
J Immunol ; 181(11): 7473-9, 2008 Dec 01.
Article in English | MEDLINE | ID: mdl-19017936

ABSTRACT

Th17 cells, a subset of T cells involved in autoimmunity and host defense against extracellular Gram-negative infection, express both IL-17A and IL-17F. Both IL-17A and IL-17F can signal via the IL-17RA; however, IL-17F does so at a 1- to 2-log higher concentration than IL-17A. In this study, we show that the IL-17F homodimer via IL-17RA is a negative regulator of IL-17 production in T cells and suggest a mechanism whereby IL-17RA on T cells serves as an autocrine/paracrine regulator of IL-17 synthesis in T cells.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Chemical and Drug Induced Liver Injury/immunology , Concanavalin A/toxicity , Interleukin-17/immunology , Mitogens/toxicity , Receptors, Interleukin-17/immunology , Animals , Autocrine Communication/genetics , Autocrine Communication/immunology , Chemical and Drug Induced Liver Injury/genetics , Gram-Negative Bacterial Infections/genetics , Gram-Negative Bacterial Infections/immunology , Interleukin-17/genetics , Mice , Mice, Knockout , Paracrine Communication/genetics , Paracrine Communication/immunology , Receptors, Interleukin-17/genetics , Signal Transduction/genetics , Signal Transduction/immunology
8.
Nature ; 424(6947): 398-405, 2003 Jul 24.
Article in English | MEDLINE | ID: mdl-12879062

ABSTRACT

Striking parallels exist between immune and nervous system cellular signalling mechanisms. Molecules originally shown to be critical for immune responses also serve neuronal functions, and similarly neural guidance cues can modulate immune function. We show here that semaphorin 7A (Sema7A), a membrane-anchored member of the semaphorin family of guidance proteins previously known for its immunomodulatory effects, can also mediate neuronal functions. Unlike many other semaphorins, which act as repulsive guidance cues, Sema7A enhances central and peripheral axon growth and is required for proper axon tract formation during embryonic development. Unexpectedly, Sema7A enhancement of axon outgrowth requires integrin receptors and activation of MAPK signalling pathways. These findings define a previously unknown biological function for semaphorins, identify an unexpected role for integrins and integrin-dependent intracellular signalling in mediating semaphorin responses, and provide a framework for understanding and interfering with Sema7A function in both immune and nervous systems.


Subject(s)
Antigens, CD/metabolism , Axons/metabolism , Integrins/metabolism , Mitogen-Activated Protein Kinases/metabolism , Semaphorins/metabolism , Animals , Antigens, CD/genetics , Cell Line , Enzyme Activation , Female , Focal Adhesion Kinase 1 , Focal Adhesion Protein-Tyrosine Kinases , GPI-Linked Proteins , Gene Deletion , Humans , Integrins/chemistry , MAP Kinase Signaling System , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuroglia/cytology , Neuroglia/metabolism , Protein Subunits , Protein-Tyrosine Kinases/metabolism , Rats , Receptors, Virus/genetics , Receptors, Virus/metabolism , Semaphorins/genetics
9.
J Clin Invest ; 109(8): 1031-40, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11956240

ABSTRACT

Endothelial CD39 metabolizes ADP released from activated platelets. Recombinant soluble human CD39 (solCD39) potently inhibited ex vivo platelet aggregation in response to ADP and reduced cerebral infarct volumes in mice following transient middle cerebral artery occlusion, even when given 3 hours after stroke. Postischemic platelet and fibrin deposition were decreased and perfusion increased without increasing intracerebral hemorrhage. In contrast, aspirin did not increase postischemic blood flow or reduce infarction volume, but did increase intracerebral hemorrhage. Mice lacking the enzymatically active extracellular portion of the CD39 molecule were generated by replacement of exons 4-6 (apyrase-conserved regions 2-4) with a PGKneo cassette. Although CD39 mRNA 3' of the neomycin cassette insertion site was detected, brains from these mice lacked both apyrase activity and CD39 immunoreactivity. Although their baseline phenotype, hematological profiles, and bleeding times were normal, cd39(-/-) mice exhibited increased cerebral infarct volumes and reduced postischemic perfusion. solCD39 reconstituted these mice, restoring postischemic cerebral perfusion and rescuing them from cerebral injury. These data demonstrate that CD39 exerts a protective thromboregulatory function in stroke.


Subject(s)
Adenosine Triphosphatases/physiology , Antigens, CD/physiology , Apyrase/physiology , Brain Ischemia/blood , Adenosine Triphosphatases/deficiency , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/pharmacology , Animals , Antigens, CD/genetics , Antigens, CD/pharmacology , Apyrase/deficiency , Apyrase/genetics , Apyrase/pharmacology , Aspirin/pharmacology , Brain Ischemia/physiopathology , Disease Models, Animal , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Platelet Aggregation/drug effects , Stroke/blood , Stroke/physiopathology , Stroke/prevention & control , Thrombosis/blood , Thrombosis/physiopathology , Thrombosis/prevention & control
10.
Mol Immunol ; 43(10): 1694-9, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16256200

ABSTRACT

Protein kinase C-associated kinase (PKK; DIK/RIP4) is an ankyrin-repeat containing serine/threonine receptor-interacting protein (RIP)-family kinase that can activate NFkappaB, and is required for keratinocyte development. In earlier studies, the expression of a catalytically inactive mutant of PKK in the B cell lineage resulted in a marked decrease in peripheral B cells in the spleen and a severe reduction of B-1 B cells. Here we explore the consequences of a null mutation in PKK with respect to the generation of peripheral B cell lineages and the activation of NFkappaB. We show that PKK is not required for the production of B cells in the bone marrow or for the development and maintenance of all mature B lymphocyte populations. We also show that PKK is not required for the activation of NFkappaB downstream of the BCR, CD40, or TLR-4 in B cells. Taken together, these data demonstrate that the loss of this RIP-family kinase does not compromise B lymphocyte development and maintenance, but leaves open the possibility that PKK may have a redundant role in these processes.


Subject(s)
B-Lymphocytes/cytology , B-Lymphocytes/enzymology , Protein Kinases/physiology , Animals , Bone Marrow/immunology , CD40 Antigens/metabolism , Cell Lineage , Enzyme Activation , Mice , Mice, Mutant Strains , Mutation , Protein Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Receptors, Antigen, B-Cell/metabolism , Toll-Like Receptor 4/metabolism , NF-kappaB-Inducing Kinase
11.
FEBS Lett ; 553(3): 257-61, 2003 Oct 23.
Article in English | MEDLINE | ID: mdl-14572634

ABSTRACT

ADAMs (A Disintegrin And Metalloprotease domain) are metalloprotease-disintegrin proteins that have been implicated in cell adhesion, protein ectodomain shedding, matrix protein degradation and cell fusion. Since such events are critical for bone resorption and osteoclast recruitment, we investigated whether they require ADAMs. We report here which ADAMs we have identified in bone cells, as well as our analysis of the generation, migration and resorptive activity of osteoclasts in developing metatarsals of mouse embryos lacking catalytically active ADAM 17 [TNFalpha converting enzyme (TACE)]. The absence of TACE activity still allowed the generation of cells showing an osteoclastic phenotype, but prevented their migration into the core of the diaphysis and the subsequent formation of marrow cavity. This suggests a role of TACE in the recruitment of osteoclasts to future resorption sites.


Subject(s)
Bone Development/physiology , Bone Marrow/metabolism , Metalloendopeptidases/metabolism , Metatarsal Bones/physiology , Osteoclasts/physiology , ADAM Proteins , ADAM17 Protein , Animals , Bone Marrow/enzymology , Bone Resorption/metabolism , Carrier Proteins/metabolism , Cell Movement/physiology , DNA Primers/genetics , Diaphyses/cytology , Diaphyses/growth & development , Disintegrins/chemistry , Immunohistochemistry , Membrane Glycoproteins/metabolism , Metalloendopeptidases/chemistry , Metalloendopeptidases/genetics , Metatarsal Bones/growth & development , Mice , Mice, Inbred DBA , Osteoclasts/cytology , Osteoclasts/enzymology , Phenotype , Protein Structure, Tertiary , RANK Ligand , Rabbits , Receptor Activator of Nuclear Factor-kappa B , Tumor Necrosis Factor-alpha
12.
Biochem Soc Symp ; (70): 39-52, 2003.
Article in English | MEDLINE | ID: mdl-14587281

ABSTRACT

Tumour necrosis factor alpha (TNF alpha)-converting enzyme (TACE/ADAM-17, where ADAM stands for a disintegrin and metalloproteinase) releases from the cell surface the extracellular domains of TNF and several other proteins. Previous studies have found that, while purified TACE preferentially cleaves peptides representing the processing sites in TNF and transforming growth factor alpha, the cellular enzyme nonetheless also sheds proteins with divergent cleavage sites very efficiently. More recent work, identifying the cleavage site in the p75 TNF receptor, quantifying the susceptibility of additional peptides to cleavage by TACE and identifying additional protein substrates, underlines the complexity of TACE-substrate interactions. In addition to substrate specificity, the mechanism underlying the increased rate of shedding caused by agents that activate cells remains poorly understood. Recent work in this area, utilizing a peptide substrate as a probe for cellular TACE activity, indicates that the intrinsic activity of the enzyme is somehow increased.


Subject(s)
Alanine/metabolism , Metalloendopeptidases/metabolism , Valine/metabolism , ADAM Proteins , ADAM17 Protein , Enzyme Induction , Metalloendopeptidases/biosynthesis , Metalloendopeptidases/chemistry , Substrate Specificity
13.
J Invest Dermatol ; 130(1): 102-12, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19626033

ABSTRACT

The receptor-interacting protein (RIP) family kinase RIP4 interacts with protein kinase C (PKC) isoforms and is implicated in PKC-dependent signaling pathways. RIP4(-/-) mice die at birth with epidermal differentiation defects, causing fusions of all external orifices and loss of the esophageal lumen. To further understand RIP4 function in the skin, we generated transgenic mice with epidermal-specific expression of RIP4 using the human keratin-14 promoter (K14-RIP4). The K14-RIP4 transgene rescued the epidermal phenotype of RIP4(-/-) mice, showing that RIP4 acts autonomously in the epidermis to regulate differentiation. Although RIP4(-/-) mice share many phenotypic similarities with inhibitor kappaB kinase (IKK)alpha(-/-) mice and stratifin repeated epilation (Sfn(Er/Er)) mice, the K14-RIP4 transgene failed to promote epidermal differentiation in these mutant backgrounds. Unexpectedly, topical treatment of K14-RIP4 mice with 12-O-tetradecanoylphorbol-13-acetate (TPA) induced dramatic, neutrophilic inflammation, an effect that was independent of tumor necrosis factor type 1 receptor (TNFR1/p55) function. Despite their enhanced sensitivity to TPA, K14-RIP4 mice did not have an altered frequency of tumor formation in TPA-promoted skin cancer initiated with 7,12-dimethylbenz[a]anthracene (DMBA). These data suggest that RIP4 functions in the epidermis through PKC-specific signaling pathways to regulate differentiation and inflammation.


Subject(s)
Dermatitis, Contact/immunology , Dermatitis, Contact/physiopathology , Epidermis/immunology , Epidermis/pathology , Protein Kinases , 14-3-3 Proteins/genetics , 14-3-3 Proteins/metabolism , 9,10-Dimethyl-1,2-benzanthracene/toxicity , Animals , Carcinogens/toxicity , Cell Differentiation/physiology , Dermatitis, Contact/pathology , Female , Humans , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , Keratin-14/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pregnancy , Promoter Regions, Genetic/physiology , Protein Kinase C/metabolism , Protein Kinases/genetics , Protein Kinases/immunology , Protein Kinases/metabolism , Protein Serine-Threonine Kinases , Receptors, Tumor Necrosis Factor, Type I/metabolism , Skin Neoplasms/chemically induced , Skin Neoplasms/immunology , Tetradecanoylphorbol Acetate/toxicity
14.
J Exp Med ; 204(11): 2603-14, 2007 Oct 29.
Article in English | MEDLINE | ID: mdl-17908936

ABSTRACT

The interleukin (IL)-1 family members IL-1alpha, -1beta, and -18 are potent inflammatory cytokines whose activities are dependent on heterodimeric receptors of the IL-1R superfamily, and which are regulated by soluble antagonists. Recently, several new IL-1 family members have been identified. To determine the role of one of these family members in the skin, transgenic mice expressing IL1F6 in basal keratinocytes were generated. IL1F6 transgenic mice exhibit skin abnormalities that are dependent on IL-1Rrp2 and IL-1RAcP, which are two members of the IL-1R family. The skin phenotype is characterized by acanthosis, hyperkeratosis, the presence of a mixed inflammatory cell infiltrate, and increased cytokine and chemokine expression. Strikingly, the combination of the IL-1F6 transgene with an IL1F5 deficiency results in exacerbation of the skin phenotype, demonstrating that IL-1F5 has antagonistic activity in vivo. Skin from IL1F6 transgenic, IL1F5(-/-) pups contains intracorneal and intraepithelial pustules, nucleated corneocytes, and dilated superficial dermal blood vessels. Additionally, expression of IL1RL2, -1F5, and -1F6 is increased in human psoriatic skin. In summary, dysregulated expression of novel agonistic and antagonistic IL-1 family member ligands can promote cutaneous inflammation, revealing potential novel targets for the treatment of inflammatory skin disorders.


Subject(s)
Inflammation/physiopathology , Interleukin-1/physiology , Skin Diseases/physiopathology , Animals , Bacterial Capsules , Humans , Interleukin-1/genetics , Interleukin-1/immunology , Ligands , Mice , Mice, Transgenic , Polysaccharides, Bacterial/genetics , Promoter Regions, Genetic , Skin/pathology
15.
J Immunol ; 175(5): 3360-8, 2005 Sep 01.
Article in English | MEDLINE | ID: mdl-16116229

ABSTRACT

IL-17 is a proinflammatory cytokine suspected to be involved in inflammatory and autoimmune diseases such as rheumatoid arthritis. In the present study, we report that IL-17R signaling is required in radiation-resistant cells in the joint for full progression of chronic synovitis and bone erosion. Repeated injections of Gram-positive bacterial cell wall fragments (streptococcal cell wall) directly into the knee joint of naive IL-17R-deficient (IL-17R-/-) mice had no effect on the acute phase of arthritis but prevented progression to chronic destructive synovitis as was noted in wild-type (wt) mice. Microarray analysis revealed significant down-regulation of leukocyte-specific chemokines, selectins, cytokines, and collagenase-3 in the synovium of IL-17R-/- mice. Bone marrow (BM) chimeric mice revealed the need for IL-17R expression on radiation-resistant joint cells for destructive inflammation. Chimeric mice of host wt and donor IL-17R-/- BM cells developed destructive synovitis in this chronic reactivated streptococcal cell wall arthritis model similar to wt-->wt chimeras. In contrast, chimeric mice of host IL-17R-/- and donor wt BM cells were protected from chronic destructive arthritis similar as IL-17R-/- -->IL-17R-/- chimeras. These data strongly indicate that IL-17R signaling in radiation-resistant cells in the joint is required for turning an acute macrophage-mediated inflammation into a chronic destructive synovitis.


Subject(s)
Knee Joint/metabolism , Radiation Tolerance , Receptors, Interleukin/physiology , Signal Transduction/physiology , Synovitis/etiology , Animals , Chemokines/genetics , Cytokines/genetics , Disease Progression , Female , Interleukin-1/physiology , Male , Mice , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , Receptors, Interleukin-17 , Selectins/genetics , Tumor Necrosis Factor-alpha/physiology
16.
J Immunol ; 168(3): 1356-61, 2002 Feb 01.
Article in English | MEDLINE | ID: mdl-11801676

ABSTRACT

We have previously implicated TNF-related apoptosis-inducing ligand (TRAIL) in innate immune surveillance against tumor development. In this study, we describe the use of TRAIL gene-targeted mice to demonstrate the key role of TRAIL in suppressing tumor initiation and metastasis. Liver and spleen mononuclear cells from TRAIL gene-targeted mice were devoid of TRAIL expression and TRAIL-mediated cytotoxicity. TRAIL gene-targeted mice were more susceptible to experimental and spontaneous tumor metastasis, and the immunotherapeutic value of alpha-galactosylceramide was diminished in TRAIL gene-targeted mice. TRAIL gene-targeted mice were also more sensitive to the chemical carcinogen methylcholanthrene. These results substantiated TRAIL as an important natural effector molecule used in the host defense against transformed cells.


Subject(s)
Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/immunology , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Neoplasm Metastasis/genetics , Neoplasm Metastasis/immunology , Tumor Necrosis Factor-alpha/deficiency , Tumor Necrosis Factor-alpha/genetics , Adenocarcinoma/genetics , Adenocarcinoma/immunology , Adenocarcinoma/secondary , Animals , Apoptosis Regulatory Proteins , Cell Division/genetics , Cell Division/immunology , Cytotoxicity, Immunologic/genetics , Disease Susceptibility/immunology , Female , Fibrosarcoma/chemically induced , Fibrosarcoma/genetics , Fibrosarcoma/immunology , Fibrosarcoma/pathology , Gene Targeting , Genetic Predisposition to Disease , Kidney Neoplasms/genetics , Kidney Neoplasms/immunology , Killer Cells, Natural/immunology , Ligands , Liver Neoplasms/genetics , Liver Neoplasms/immunology , Liver Neoplasms/prevention & control , Liver Neoplasms/secondary , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/pathology , Mammary Neoplasms, Experimental/prevention & control , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/physiology , Methylcholanthrene/toxicity , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Transplantation , TNF-Related Apoptosis-Inducing Ligand , Tumor Cells, Cultured/transplantation , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/physiology
17.
Immunity ; 18(1): 1-6, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12530970

ABSTRACT

The TNF-related apoptosis-inducing ligand (TRAIL) offers great promise as a cancer therapeutic. Initially, soluble recombinant versions of the TRAIL molecule have exhibited specific tumoricidal activity against a variety of tumors alone, or in combination with other cancer treatments, and much anticipation awaits the outcomes from early clinical trials. More recently, the natural role of TRAIL has been explored in tumor and allogeneic bone marrow transplantation models in the mouse. Strikingly, the TRAIL effector pathway appears a vital component of immunosurveillance of spontaneous or resident tumor cells by both T cells and NK cells, stimulating more hope that manipulating TRAIL activity is a natural path to improved cancer immunotherapy.


Subject(s)
Immunotherapy , Membrane Glycoproteins/therapeutic use , Neoplasms/therapy , Tumor Necrosis Factor-alpha/therapeutic use , Animals , Apoptosis Regulatory Proteins , Gene Expression , Graft vs Tumor Effect , Humans , Killer Cells, Natural/immunology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/physiology , Models, Immunological , Monitoring, Immunologic , Neoplasms/genetics , Neoplasms/immunology , Recombinant Proteins/therapeutic use , TNF-Related Apoptosis-Inducing Ligand , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/physiology
18.
J Biol Chem ; 279(7): 5612-20, 2004 Feb 13.
Article in English | MEDLINE | ID: mdl-14625290

ABSTRACT

Transmembrane metalloproteinases of the disintegrin and metalloproteinase (ADAM) family control cell signaling interactions via hydrolysis of protein extracellular domains. Prior work has shown that the receptor tyrosine kinase, c-Kit (CD117), is essential for mast cell survival and that serum levels of c-Kit increase in proliferative mast cell disorders, suggesting the existence of c-Kit shedding pathways in mast cells. In the present work, we report that tumor necrosis factor alpha-converting enzyme (TACE; ADAM-17) mediates shedding of c-Kit. Stimulation of transfected cells with phorbol 12-myristate 13-acetate (PMA) induced metalloproteinase-mediated release of c-Kit ectodomain, which increased further upon TACE overexpression. By contrast, TACE-deficient fibroblasts did not demonstrate inducible release, thus identifying TACE as the metalloproteinase primarily responsible for PMA-induced c-Kit shedding. Surface expression of c-Kit by the human mast cell-1 line decreased upon phorbol-induced shedding, which involved metalloproteinase activity susceptible to inhibition by tissue inhibitor of metalloproteinase (TIMP)-3. To further explore the role of TACE in shedding of c-Kit from mast cells, we compared the behavior of mast cells derived from murine embryonic stem cells. In these studies, PMA decreased surface c-Kit levels on mast cells expressing wild-type (+/+) TACE but not on those expressing an inactive mutant (DeltaZn/DeltaZn), confirming the role of TACE in PMA-induced c-Kit shedding. Compared with TACE(+/+) cells, TACE(DeltaZn/DeltaZn) mast cells also demonstrated decreased constitutive shedding and increased basal surface expression of c-Kit, with diminished apoptosis in response to c-Kit ligand deprivation. These data suggest that TACE controls mast cell survival by regulating shedding and surface expression of c-Kit.


Subject(s)
Embryo, Mammalian/cytology , Mast Cells/metabolism , Metalloendopeptidases/metabolism , Proto-Oncogene Proteins c-kit/metabolism , Stem Cells/metabolism , ADAM Proteins , ADAM17 Protein , Animals , Apoptosis , Cell Differentiation , Cell Division , Cell Line , Cell Separation , Cell Survival , Cytoplasm/metabolism , Detergents/pharmacology , Dose-Response Relationship, Drug , Fibroblasts/metabolism , Flow Cytometry , Humans , Immunoblotting , Ligands , Mice , Mice, Transgenic , Mutagenesis, Site-Directed , Octoxynol , Polyethylene Glycols/pharmacology , Precipitin Tests , Protein Structure, Tertiary , Proto-Oncogene Proteins c-kit/biosynthesis , Stem Cell Factor/metabolism , Transfection
19.
Mol Cell Proteomics ; 1(1): 30-6, 2002 Jan.
Article in English | MEDLINE | ID: mdl-12096138

ABSTRACT

Proteolytic cleavage (shedding) of extracellular domains of many membrane proteins by metalloproteases is an important regulatory mechanism used by mammalian cells in response to environmental and physiological changes. Here we describe a proteomic system for analyzing cell surface shedding. The method utilized short-term culture supernatants from induced cells as starting material, followed by lectin-affinity purification, deglycosylation, and polyacrylamide gel electrophoresis separation. Relative quantitation of proteins was achieved via isotope dilution. In this study, a number of proteins already known to be shed were identified from activated monocytes and endothelial cells, thereby validating the method. In addition, a group of proteins were newly identified as being shed. The method provides an unbiased means to screen for shed proteins.


Subject(s)
Cell Membrane/metabolism , Membrane Proteins/analysis , Metalloendopeptidases/metabolism , Adult , Alkylation , Animals , Carcinogens/pharmacology , Cell Line , Chromatography, Affinity , Dithiothreitol/metabolism , Electrophoresis, Gel, Two-Dimensional , Endothelium, Vascular/metabolism , Glycoproteins/analysis , Glycosylation , Homozygote , Humans , Lectins/chemistry , Lectins/metabolism , Mass Spectrometry , Mice , Mice, Knockout , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Proteome , Skin/metabolism , Tetradecanoylphorbol Acetate/pharmacology
20.
J Immunol ; 169(11): 6193-201, 2002 Dec 01.
Article in English | MEDLINE | ID: mdl-12444124

ABSTRACT

Suppression of bone marrow myeloid and erythroid progenitor cells occurs after infection with a variety of different viruses. In this study, we characterize the alterations in bone marrow (BM) lymphocytes after influenza virus infection in mice. We found a severe loss of BM B cells, particularly CD43(low/-)B220(+) pre-B and immature B cells, in influenza virus-infected mice. Depletion of BM B lineage cells resulted primarily from cell cycle arrest and most likely apoptosis within the BM environment, rather than from increased trafficking of BM emigrants to peripheral lymphoid tissues. Use of gene-knockout mice indicates that depletion of BM B cells is dependent on TNF-alpha, lymphotoxin-alpha, and both TNF receptors, TNFR1-p55 and TNFR2-p75. Thus, TNF-alpha and lymphotoxin-alpha are required for loss of BM B lineage cells during respiratory infection with influenza virus.


Subject(s)
Apoptosis/immunology , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Lymphotoxin-alpha/metabolism , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/pathology , Tumor Necrosis Factor-alpha/metabolism , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Base Sequence , Bone Marrow Cells/immunology , Bone Marrow Cells/pathology , Cell Cycle , Cell Differentiation , Killer Cells, Natural/immunology , Kinetics , Lymphotoxin-alpha/deficiency , Lymphotoxin-alpha/genetics , Macrophages, Alveolar/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Orthomyxoviridae/pathogenicity , Orthomyxoviridae/physiology , Orthomyxoviridae Infections/virology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Tumor Necrosis Factor/deficiency , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/metabolism , Receptors, Tumor Necrosis Factor, Type I , Receptors, Tumor Necrosis Factor, Type II , T-Lymphocyte Subsets/immunology , Tumor Necrosis Factor-alpha/deficiency , Tumor Necrosis Factor-alpha/genetics , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL