Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
J Clin Invest ; 128(5): 1873-1887, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29611821

ABSTRACT

Uncontrolled secretion of type I IFN, as the result of endosomal TLR (i.e., TLR7 and TLR9) signaling in plasmacytoid DCs (pDCs), and abnormal production of autoantibodies by B cells are critical for systemic lupus erythematosus (SLE) pathogenesis. The importance of galectin-9 (Gal-9) in regulating various autoimmune diseases, including lupus, has been demonstrated. However, the precise mechanism by which Gal-9 mediates this effect remains unclear. Here, using spontaneous murine models of lupus (i.e., BXSB/MpJ and NZB/W F1 mice), we demonstrate that administration of Gal-9 results in reduced TLR7-mediated autoimmune manifestations. While investigating the mechanism underlying this phenomenon, we observed that Gal-9 inhibits the phenotypic maturation of pDCs and B cells and abrogates their ability to mount cytokine responses to TLR7/TLR9 ligands. Importantly, immunocomplex-mediated (IC-mediated) and neutrophil extracellular trap-mediated (NET-mediated) pDC activation was inhibited by Gal-9. Additionally, the mTOR/p70S6K pathway, which is recruited by both pDCs and B cells for TLR-mediated IFN secretion and autoantibody generation, respectively, was attenuated. Gal-9 was found to exert its inhibitory effect on both the cells by interacting with CD44.


Subject(s)
Autoantibodies/immunology , B-Lymphocytes/immunology , Dendritic Cells/immunology , Galectins/immunology , Lupus Erythematosus, Systemic/immunology , Membrane Glycoproteins/immunology , Toll-Like Receptor 7/immunology , Animals , B-Lymphocytes/pathology , Dendritic Cells/pathology , Disease Models, Animal , Female , Humans , Lupus Erythematosus, Systemic/pathology , Male , Mice , Ribosomal Protein S6 Kinases, 70-kDa/immunology , Signal Transduction/immunology , TOR Serine-Threonine Kinases/immunology , Toll-Like Receptor 9/immunology
2.
J Allergy Clin Immunol ; 137(4): 1216-1225.e5, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26560039

ABSTRACT

BACKGROUND: Food-mediated allergic reactions have emerged as a major health problem. The underlying mechanisms that promote uncontrolled type 2 immune responses to dietary allergens in the gastrointestinal tract remain elusive. OBJECTIVE: We investigated whether altering IL-25 signaling enhances or attenuates allergic responses to food allergens. METHODS: Mice of an IL-25 transgenic mouse line (iIL-25Tg mice), which constitutively overexpress intestinal IL-25, and Il17rb(-/-) mice, in which Il17rb gene expression is disrupted, were sensitized and gavage fed with ovalbumin (OVA). We assessed symptomatic characteristics of experimental food allergy, including incidence of diarrhea, incidence of hypothermia, intestinal TH2 immune response, and serum OVA-specific IgE and mast cell protease 1 production. RESULTS: Rapid induction of Il25 expression in the intestinal epithelium preceded onset of the anaphylactic response to ingested OVA antigen. iIL-25Tg mice were more prone and Il17rb(-/-) mice were more resistant to experimental food allergy. Resident intestinal type 2 innate lymphoid cells (ILC2s) were identified as the major producers of IL-5 and IL-13 in response to IL-25. Reconstituting irradiated wild-type mice with Rora(-/-) or Il17rb(-/-) bone marrow resulted in a deficiency or dysfunction of the ILC2 compartment, respectively, and resistance to experimental food allergy. Repeated intragastric antigen challenge induced a significant increase in numbers of CD4(+) TH2 cells, which enhance IL-25-stimulated IL-13 production by ILC2s ex vivo and in vivo. Finally, reconstituted IL-13-deficient ILC2s had reduced capability to promote allergic inflammation, resulting in increased resistance to experimental food allergy. CONCLUSION: IL-25 and CD4(+) TH2 cells induced by ingested antigens enhance ILC2-derived IL-13 production, thereby promoting IgE-mediated experimental food allergy.


Subject(s)
Egg Hypersensitivity/immunology , Immunoglobulin E/immunology , Interleukin-13/immunology , Interleukins/immunology , Ovalbumin/immunology , Th2 Cells/immunology , Animals , Biomarkers/metabolism , Mice , Mice, Transgenic
3.
J Immunol ; 191(7): 3641-50, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-24014877

ABSTRACT

Current cancer vaccines induce tumor-specific T cell responses without sustained tumor regression because immunosuppressive elements within the tumor induce exhaustion of effector T cells and infiltration of immune-suppressive regulatory T cells (Tregs). Therefore, much effort has been made to generate agonistic Abs targeting members of the TNFR superfamily, such as OX40, 4-1BB, and GITR, expressed on effector T cells and Tregs, to reinvigorate T cell effector function and block Treg-suppressive function. In this article, we describe the development of a panel of anti-human OX40 agonistic mouse mAbs that could promote effector CD4(+) and CD8(+) T cell proliferation, inhibit the induction of CD4(+) IL-10 -producing type 1 regulatory T cells, inhibit the expansion of ICOS(+)IL-10(+) Tregs, inhibit TGF-ß-induced FOXP3 expression on naive CD4(+) T cells, and block natural Treg-suppressive function. We humanized two anti-human OX40 mAb clones, and they retained the potency of their parental clones. These Abs should provide broad opportunities for potential combination therapy to treat a wide realm of cancers and preventative vaccines against infectious diseases.


Subject(s)
Antibodies, Monoclonal/pharmacology , Receptors, OX40/antagonists & inhibitors , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Animals , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal, Humanized/metabolism , Antibodies, Monoclonal, Humanized/pharmacology , Cell Line , Cell Proliferation/drug effects , Female , Humans , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Macaca mulatta , Mice , Protein Binding , Receptors, OX40/metabolism
4.
J Immunol ; 189(2): 786-92, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22706086

ABSTRACT

The human plasmacytoid dendritic cell (pDC) receptor BDCA2 forms a complex with the adaptor FcεR1γ to activate an ITAM-signaling cascade. BDCA2 receptor signaling negatively regulates the TLR7/9-mediated type 1 IFN responses in pDCs, which may play a key role in controlling self-DNA/RNA-induced autoimmunity. We report in this article that CD2-associated adaptor protein (CD2AP), which is highly expressed in human pDCs, positively regulates BDCA2/FcεR1γ receptor signaling. By immunoprecipitation and mass spectrometry analyses, we found that CD2AP bound to SHIP1. Knockdown of CD2AP or SHIP1 reduced the BDCA2/FcεR1γ-mediated ITAM signaling and blocked its inhibition of TLR9-mediated type 1 IFN production. Knockdown of CD2AP or SHIP1 also enhanced the ubiquitination and degradation of Syk and FcεR1γ that was mediated by the E3 ubiquitin ligase Cbl. This led us to discover that, upon BDCA2 cross-linking, the CD2AP/SHIP1 complex associated with Cbl and inhibited its E3 ubiquitin ligase activity. In human primary pDCs, cross-linking of the BDCA2/FcεR1γ complex induced the recruitment of the CD2AP/SHIP1/Cbl complex to the plasma membrane of pDCs, where it colocalized with the BDCA2/FcεR1γ complex. Therefore, CD2AP positively regulates BDCA2/FcεR1γ signaling by forming a complex with SHIP1 to inhibit the E3 ubiquitin ligase Cbl.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Cytoskeletal Proteins/physiology , Dendritic Cells/immunology , Multiprotein Complexes/physiology , Phosphoric Monoester Hydrolases/physiology , Proto-Oncogene Proteins c-cbl/antagonists & inhibitors , Proto-Oncogene Proteins c-cbl/metabolism , Signal Transduction/immunology , Up-Regulation/immunology , Cells, Cultured , Cross-Linking Reagents/metabolism , Dendritic Cells/enzymology , Dendritic Cells/metabolism , HEK293 Cells , Humans , Inositol Polyphosphate 5-Phosphatases , Lectins, C-Type/physiology , Membrane Glycoproteins/physiology , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases , Receptors, Immunologic/physiology
5.
Cell Res ; 22(7): 1129-39, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22453241

ABSTRACT

Toll-like receptor 9 (TLR9) senses microbial DNA in the endosomes of plasmacytoid dendritic cells (pDCs) and triggers MyD88-dependent type I interferon (IFN) responses. To better understand TLR9 biology in pDCs, we established a yeast two-hybrid library for the identification of TLR9-interacting proteins. Here, we report that an IFN-inducible protein, phospholipid scramblase 1 (PLSCR1), interacts with TLR9 in pDCs. Knockdown of PLSCR1 expression by siRNA in human pDC cell line led to a 60-70% reduction of IFN-α responses following CpG-ODN (oligodeoxynucleotide) stimulation. Primary pDCs from PLSCR1-deficient mice produced lower amount of type 1 IFN than pDCs from the wild-type mice in response to CpG-ODN, herpes simplex virus and influenza A virus. Following CpG-A stimulation, there were much lower amounts of TLR9 in the early endosomes together with CpG-A in pDCs from PLSCR1-deficient mice. Our study demonstrates that PLSCR1 is a TLR9-interacting protein that plays an important role in pDC's type 1 IFN responses by regulating TLR9 trafficking to the endosomal compartment.


Subject(s)
Dendritic Cells/metabolism , Interferon Type I/metabolism , Phospholipid Transfer Proteins/metabolism , Toll-Like Receptor 9/metabolism , Animals , Cell Line , Cells, Cultured , Dendritic Cells/drug effects , Fluorescent Antibody Technique , Humans , Immunoblotting , Interferon Regulatory Factor-7/metabolism , Interferon-alpha/metabolism , Mice , Oligodeoxyribonucleotides/pharmacology , Phospholipid Transfer Proteins/genetics , Toll-Like Receptor 9/genetics , Two-Hybrid System Techniques
6.
EMBO J ; 31(5): 1095-108, 2012 Mar 07.
Article in English | MEDLINE | ID: mdl-22343943

ABSTRACT

The lysosome plays a key role in cellular homeostasis by controlling both cellular clearance and energy production to respond to environmental cues. However, the mechanisms mediating lysosomal adaptation are largely unknown. Here, we show that the Transcription Factor EB (TFEB), a master regulator of lysosomal biogenesis, colocalizes with master growth regulator mTOR complex 1 (mTORC1) on the lysosomal membrane. When nutrients are present, phosphorylation of TFEB by mTORC1 inhibits TFEB activity. Conversely, pharmacological inhibition of mTORC1, as well as starvation and lysosomal disruption, activates TFEB by promoting its nuclear translocation. In addition, the transcriptional response of lysosomal and autophagic genes to either lysosomal dysfunction or pharmacological inhibition of mTORC1 is suppressed in TFEB-/- cells. Interestingly, the Rag GTPase complex, which senses lysosomal amino acids and activates mTORC1, is both necessary and sufficient to regulate starvation- and stress-induced nuclear translocation of TFEB. These data indicate that the lysosome senses its content and regulates its own biogenesis by a lysosome-to-nucleus signalling mechanism that involves TFEB and mTOR.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cell Nucleus/physiology , Lysosomes/physiology , Proteins/metabolism , Signal Transduction , Animals , Cell Line , Humans , Immunoprecipitation , Lysosomal Membrane Proteins/metabolism , Lysosomes/metabolism , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Knockout , Microscopy, Fluorescence , Models, Biological , Multiprotein Complexes , Protein Binding , Protein Interaction Mapping , TOR Serine-Threonine Kinases
7.
J Immunol ; 187(9): 4501-8, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-21957149

ABSTRACT

The innate immune system is equipped with many molecular sensors for microbial DNA/RNA to quickly mount antimicrobial host immune responses. In this paper, we identified DHX9, a DExDc helicase family member, as an important viral dsRNA sensor in myeloid dendritic cells (mDCs). Knockdown of DHX9 expression by small heteroduplex RNA dramatically blocked the ability of mDCs to produce IFN-α/ß and proinflammatory cytokines in response to polyinosine-polycytidylic acid, influenza A, and reovirus. DHX9 could specifically bind polyinosine-polycytidylic acid via its double-strand RNA binding motifs. DHX9 interacted with IPS-1 via the HelicC-HA2-DUF and CARD domains of DHX9 and IPS-1, respectively. Knockdown of DHX9 expression in mDCs blocked the activation of NF-κB and IFN regulatory factor 3 by dsRNA. Collectively, these results suggest that DHX9 is an important RNA sensor that is dependent on IPS-1 to sense pathogenic RNA.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , DEAD-box RNA Helicases/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Myeloid Cells/immunology , Myeloid Cells/metabolism , Neoplasm Proteins/metabolism , RNA, Double-Stranded/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Line , Cells, Cultured , Cytokines/antagonists & inhibitors , Cytokines/biosynthesis , DEAD-box RNA Helicases/antagonists & inhibitors , DEAD-box RNA Helicases/genetics , Dendritic Cells/virology , HEK293 Cells , Humans , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/metabolism , Influenza A virus/immunology , Interferon Type I/antagonists & inhibitors , Interferon Type I/biosynthesis , Mice , Myeloid Cells/virology , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Nucleic Acid Heteroduplexes/pharmacology , Poly I-C/metabolism , Protein Binding/genetics , Protein Binding/immunology , RNA, Double-Stranded/genetics , RNA, Small Interfering/antagonists & inhibitors , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , RNA, Viral/antagonists & inhibitors , RNA, Viral/genetics , RNA, Viral/metabolism
8.
Immunity ; 34(6): 866-78, 2011 Jun 24.
Article in English | MEDLINE | ID: mdl-21703541

ABSTRACT

The innate immune system detects viral infection predominantly by sensing viral nucleic acids. We report the identification of a viral sensor, consisting of RNA helicases DDX1, DDX21, and DHX36, and the adaptor molecule TRIF, by isolation and sequencing of poly I:C-binding proteins in myeloid dendritic cells (mDCs). Knockdown of each helicase or TRIF by shRNA blocked the ability of mDCs to mount type I interferon (IFN) and cytokine responses to poly I:C, influenza A virus, and reovirus. Although DDX1 bound poly I:C via its Helicase A domain, DHX36 and DDX21 bound the TIR domain of TRIF via their HA2-DUF and PRK domains, respectively. This sensor was localized within the cytosol, independent of the endosomes. Thus, the DDX1-DDX21-DHX36 complex represents a dsRNA sensor that uses the TRIF pathway to activate type I IFN responses in the cytosol of mDCs.


Subject(s)
Adaptor Proteins, Vesicular Transport/immunology , DEAD-box RNA Helicases/immunology , Dendritic Cells/immunology , RNA, Double-Stranded/genetics , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Cell Line , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Dendritic Cells/metabolism , Humans , Mice , Protein Binding , Signal Transduction
9.
Sci Transl Med ; 3(73): 73ra19, 2011 Mar 09.
Article in English | MEDLINE | ID: mdl-21389263

ABSTRACT

Systemic lupus erythematosus (SLE) is a severe and incurable autoimmune disease characterized by chronic activation of plasmacytoid dendritic cells (pDCs) and production of autoantibodies against nuclear self-antigens by hyperreactive B cells. Neutrophils are also implicated in disease pathogenesis; however, the mechanisms involved are unknown. Here, we identified in the sera of SLE patients immunogenic complexes composed of neutrophil-derived antimicrobial peptides and self-DNA. These complexes were produced by activated neutrophils in the form of web-like structures known as neutrophil extracellular traps (NETs) and efficiently triggered innate pDC activation via Toll-like receptor 9 (TLR9). SLE patients were found to develop autoantibodies to both the self-DNA and antimicrobial peptides in NETs, indicating that these complexes could also serve as autoantigens to trigger B cell activation. Circulating neutrophils from SLE patients released more NETs than those from healthy donors; this was further stimulated by the antimicrobial autoantibodies, suggesting a mechanism for the chronic release of immunogenic complexes in SLE. Our data establish a link between neutrophils, pDC activation, and autoimmunity in SLE, providing new potential targets for the treatment of this devastating disease.


Subject(s)
Dendritic Cells/immunology , Lupus Erythematosus, Systemic/immunology , Neutrophils/immunology , Antibodies, Antinuclear/blood , Antigen-Antibody Complex/blood , Antimicrobial Cationic Peptides , Autoantigens/blood , B-Lymphocytes/immunology , Case-Control Studies , Cathelicidins/immunology , DNA/blood , DNA/immunology , Humans , Lymphocyte Activation , Peptides/blood , Peptides/immunology , Toll-Like Receptor 9/metabolism
10.
EMBO J ; 29(23): 3939-51, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-21045808

ABSTRACT

The mechanisms that couple translation and protein processing are poorly understood in higher eukaryotes. Although mammalian target of rapamycin (mTOR) complex 1 (mTORC1) controls translation initiation, the function of mTORC2 in protein synthesis remains to be defined. In this study, we find that mTORC2 can colocalize with actively translating ribosomes and can stably interact with rpL23a, a large ribosomal subunit protein present at the tunnel exit. Exclusively during translation of Akt, mTORC2 mediates phosphorylation of the nascent polypeptide at the turn motif (TM) site, Thr450, to avoid cotranslational Akt ubiquitination. Constitutive TM phosphorylation occurs because the TM site is accessible, whereas the hydrophobic motif (Ser473) site is concealed in the ribosomal tunnel. Thus, mTORC2 can function cotranslationally by phosphorylating residues in nascent chains that are critical to attain proper conformation. Our findings reveal that mTOR links protein production with quality control.


Subject(s)
Proto-Oncogene Proteins c-akt/metabolism , Ribosomes/metabolism , TOR Serine-Threonine Kinases/metabolism , Amino Acid Sequence , Animals , Cell Line , Humans , Mice , Molecular Sequence Data , Multiprotein Complexes/metabolism , Phosphorylation , Protein Biosynthesis , Ribosomal Proteins/metabolism , Ubiquitination
11.
Proc Natl Acad Sci U S A ; 107(34): 15181-6, 2010 Aug 24.
Article in English | MEDLINE | ID: mdl-20696886

ABSTRACT

Toll-like receptor 9 (TLR9) senses microbial DNA and triggers type I IFN responses in plasmacytoid dendritic cells (pDCs). Previous studies suggest the presence of myeloid differentiation primary response gene 88 (MyD88)-dependent DNA sensors other than TLR9 in pDCs. Using MS, we investigated C-phosphate-G (CpG)-binding proteins from human pDCs, pDC-cell lines, and interferon regulatory factor 7 (IRF7)-expressing B-cell lines. CpG-A selectively bound the aspartate-glutamate-any amino acid-aspartate/histidine (DExD/H)-box helicase 36 (DHX36), whereas CpG-B selectively bound DExD/H-box helicase 9 (DHX9). Although the aspartate-glutamate-alanine-histidine box motif (DEAH) domain of DHX36 was essential for CpG-A binding, the domain of unknown function 1605 (DUF1605 domain) of DHX9 was required for CpG-B binding. DHX36 is associated with IFN-alpha production and IRF7 nuclear translocation in response to CpG-A, but DHX9 is important for TNF-alpha and IL-6 production and NF-kappaB activation in response to CpG-B. Knocking down DHX9 or DHX36 significantly reduced the cytokine responses of pDCs to a DNA virus but had no effect on the cytokine responses to an RNA virus. We further showed that both DHX9 and DHX36 are localized within the cytosol and are directly bound to the Toll-interleukin receptor domain of MyD88 via their helicase-associated domain 2 and DUF domains. This study demonstrates that DHX9/DHX36 represent the MyD88-dependent DNA sensors in the cytosol of pDCs and suggests a much broader role for DHX helicases in viral sensing.


Subject(s)
DEAD-box RNA Helicases/metabolism , DNA, Viral/metabolism , Dendritic Cells/immunology , Dendritic Cells/virology , Neoplasm Proteins/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , B-Lymphocytes/virology , Base Sequence , Binding Sites , Cell Line , CpG Islands , DEAD-box RNA Helicases/antagonists & inhibitors , DEAD-box RNA Helicases/chemistry , DEAD-box RNA Helicases/genetics , Dendritic Cells/metabolism , Humans , Immunity, Innate , Interferon Regulatory Factor-7/metabolism , Myeloid Differentiation Factor 88/metabolism , NF-kappa B p50 Subunit/metabolism , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Phylogeny , Protein Structure, Tertiary , RNA, Small Interfering/genetics , Receptors, Transferrin/metabolism , Signal Transduction
12.
Mol Cell ; 39(3): 433-43, 2010 Aug 13.
Article in English | MEDLINE | ID: mdl-20705244

ABSTRACT

Mammalian target of rapamycin (mTOR) is an important mediator of phosphoinositol-3-kinase (PI3K) signaling. PI3K signaling regulates B cell development, homeostasis, and immune responses. However, the function and molecular mechanism of mTOR-mediated PI3K signaling in B cells has not been fully elucidated. Here we show that Sin1, an essential component of mTOR complex 2 (mTORC2), regulates B cell development. Sin1 deficiency results in increased IL-7 receptor (il7r) and RAG recombinase (rag1 and rag2) gene expression, leading to enhanced pro-B cell survival and augmented V(D)J recombinase activity. We further show that Akt2 specifically mediates the Sin1-mTORC2 dependent suppression of il7r and rag gene expression in B cells by regulating FoxO1 phosphorylation. Finally, we demonstrate that the mTOR inhibitor rapamycin induces rag expression and promotes V(D)J recombination in B cells. Our study reveals that the Sin1/mTORC2-Akt2 signaling axis is a key regulator of FoxO1 transcriptional activity in B cells.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , B-Lymphocytes/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation/physiology , Homeodomain Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Interleukin-7/metabolism , TOR Serine-Threonine Kinases/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , B-Lymphocytes/cytology , Cell Line, Transformed , DNA-Binding Proteins/genetics , Forkhead Box Protein O1 , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Rearrangement, B-Lymphocyte/physiology , Homeodomain Proteins/genetics , Mice , Mice, Knockout , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Receptors, Interleukin-7/genetics , Signal Transduction/physiology , Transcription Factors
13.
Cancer Res ; 70(4): 1334-43, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-20124484

ABSTRACT

The role of vasculogenesis as opposed to angiogenesis in tumor formation has been little explored genetically. Endothelial cells that lack the MEK kinase MEKK3 cannot form vessels. In this study, we employed mice with hematopoietic deletions of the Mekk3 gene to evaluate the importance of vasculogenesis in the formation of Ewing's sarcoma tumors. Bone marrow cells (BM) from LacZ(+) Mekk3-deficient conditional knockout mice (Mekk3(Deltaflox/-) mice) were transplanted into irradiated nude mice before injection of Ewing's sarcoma cells. Because the grafted Mekk3(Deltaflox/-) BM cells cannot contribute to vessel development in the same way as the host Mekk3(+/+) endothelial cells, angiogenesis is normal in the model whereas vasculogenesis is impaired. Four weeks after BM transplant, Ewing's sarcoma TC71 or A4573 cells were injected, and tumor growth and vessel density were compared. Strikingly, chimeric mice transplanted with Mekk3(Deltaflox/-) BM exhibited a reduction in tumor growth and vessel density compared with mice transplanted with Mekk3(Deltaflox/+) BM cells. Mekk3(Deltaflox/-) cells that were LacZ positive were visualized within the tumor; however, few of the LacZ(+) cells colocalized with either CD31(+) endothelial cells or desmin(+) pericytes. Quantification of double-positive LacZ(+) and CD31(+) endothelial cells or LacZ(+) and desmin(+) pericytes confirmed that chimeric mice transplanted with Mekk3(Deltaflox/-) BM were impaired for tumor vessel formation. In contrast, siRNA-mediated knockdown of Mekk3 in TC71 Ewing's sarcoma cells had no effect on tumor growth or vessel density. Our findings indicate that vasculogenesis is critical in the expansion of the tumor vascular network.


Subject(s)
Bone Marrow Cells/physiology , Bone Neoplasms/blood supply , Bone Neoplasms/pathology , Neovascularization, Pathologic/pathology , Sarcoma, Ewing/blood supply , Sarcoma, Ewing/pathology , Animals , Bone Marrow Cells/metabolism , Bone Marrow Transplantation/adverse effects , Bone Marrow Transplantation/physiology , Cell Proliferation , Embryo, Mammalian , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/physiology , Humans , MAP Kinase Kinase Kinase 3/antagonists & inhibitors , MAP Kinase Kinase Kinase 3/genetics , MAP Kinase Kinase Kinase 3/metabolism , MAP Kinase Kinase Kinase 3/physiology , Male , Mice , Mice, Nude , Mice, Transgenic , Neovascularization, Pathologic/etiology , Neovascularization, Pathologic/genetics , Neovascularization, Physiologic/genetics , Neovascularization, Physiologic/physiology , RNA, Small Interfering/pharmacology , Tumor Burden/genetics , Tumor Cells, Cultured
14.
J Exp Med ; 206(9): 1983-94, 2009 Aug 31.
Article in English | MEDLINE | ID: mdl-19703986

ABSTRACT

Dendritic cell (DC) responses to extracellular self-DNA and self-RNA are prevented by the endosomal seclusion of nucleic acid-recognizing Toll-like receptors (TLRs). In psoriasis, however, plasmacytoid DCs (pDCs) sense self-DNA that is transported to endosomal TLR9 upon forming a complex with the antimicrobial peptide LL37. Whether LL37 also interacts with extracellular self-RNA and how this may contribute to DC activation in psoriasis is not known. Here, we report that LL37 can bind self-RNA released by dying cells, protect it from extracellular degradation, and transport it into endosomal compartments of DCs. In pDC, self-RNA-LL37 complexes activate TLR7 and, like self-DNA-LL37 complexes, trigger the secretion of IFN-alpha without inducing maturation or the production of IL-6 and TNF-alpha. In contrast to self-DNA-LL37 complexes, self-RNA-LL37 complexes also trigger the activation of classical myeloid DCs (mDCs). This occurs through TLR8 and leads to the production of TNF-alpha and IL-6, and the differentiation of mDCs into mature DCs. We also found that self-RNA-LL37 complexes are present in psoriatic skin lesions and are associated with mature mDCs in vivo. Our results demonstrate that the cationic antimicrobial peptide LL37 converts self-RNA into a trigger of TLR7 and TLR8 in human DCs, and provide new insights into the mechanism that drives the auto-inflammatory responses in psoriasis.


Subject(s)
Antimicrobial Cationic Peptides/metabolism , Dendritic Cells/immunology , Macromolecular Substances/metabolism , Psoriasis/immunology , RNA/metabolism , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 8/metabolism , Amino Acid Sequence , Antimicrobial Cationic Peptides/genetics , Antimicrobial Cationic Peptides/immunology , Cathelicidins , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Luciferases , Macromolecular Substances/immunology , Molecular Sequence Data , RNA/immunology , Toll-Like Receptor 7/immunology , Toll-Like Receptor 8/immunology
15.
J Immunol ; 183(6): 3831-8, 2009 Sep 15.
Article in English | MEDLINE | ID: mdl-19710465

ABSTRACT

MEKK1-dependent signaling regulates HECT E3 ligase Itch, resulting in elevated catalytic activity. After TCR costimulation, MEKK1 predominantly induces JNK1 activation, whereas the related kinase MEKK2 regulates ERK5 activation. MEKK1 becomes phosphorylated on multiple sites and polyubiquitinated following TCR costimulation. E3 ligase Itch is recruited to activated MEKK1, but not MEKK2, and this novel scaffolding interaction is dependent on MEKK1 Thr(1381) phosphorylation within the kinase domain and an intact MEKK1 RING finger motif. MEKK1 phosphorylation on Thr(1381) is observed during Th2 differentiation, but not under Th1 differentiation. Both Itch and the MEKK1 kinase domain are important for Il4 and Il6 cytokine gene expression under Th2 conditions.


Subject(s)
Cytokines/genetics , Gene Expression Regulation , MAP Kinase Kinase Kinase 1/metabolism , Th2 Cells , Ubiquitin-Protein Ligases/metabolism , Animals , Binding Sites , MAP Kinase Kinase 4/metabolism , Mice , Mice, Knockout , Phosphorylation , Protein Binding , RING Finger Domains , Receptors, Antigen, T-Cell/metabolism
16.
J Immunol ; 182(6): 3597-608, 2009 Mar 15.
Article in English | MEDLINE | ID: mdl-19265138

ABSTRACT

T cell homeostasis is crucial for maintaining an efficient and balanced T cell immunity. The interaction between TCR and self peptide (sp) MHC ligands is known to be the key driving force in this process, and it is believed to be functionally and mechanistically different from that initiated by the antigenic TCR stimulation. Yet, very little is known about the downstream signaling events triggered by this TCR-spMHC interaction and how they differ from those triggered by antigenic TCR stimulation. In this study, we show that T cell conditional ablation of MEKK3, a Ser/Thr kinase in the MAPK cascade, causes a significant reduction in peripheral T cell numbers in the conditional knockout mice, but does not perturb thymic T cell development and maturation. Using an adoptive mixed transfer method, we show that MEKK3-deficient T cells are severely impaired in lymphopenia-induced cell proliferation and survival. Interestingly, the Ag-induced T cell proliferation proceeds normally in the absence of MEKK3. Finally, we found that the activity of ERK1/2, but not p38 MAPK, was attenuated during the lymphopenia-driven response in MEKK3-deficient T cells. Together, these data suggest that MEKK3 may play a crucial selective role for spMHC-mediated T cell homeostasis.


Subject(s)
CD4-Positive T-Lymphocytes/enzymology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/enzymology , CD8-Positive T-Lymphocytes/immunology , Cell Proliferation , Lymphopenia/enzymology , Lymphopenia/immunology , MAP Kinase Kinase Kinase 3/physiology , Animals , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/pathology , Cell Differentiation/immunology , Cell Survival/genetics , Cell Survival/immunology , Cells, Cultured , Gene Knock-In Techniques , Homeostasis/genetics , Homeostasis/immunology , Lymphopenia/genetics , Lymphopenia/pathology , MAP Kinase Kinase Kinase 3/deficiency , MAP Kinase Kinase Kinase 3/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/metabolism , Thymus Gland/cytology , Thymus Gland/enzymology , Thymus Gland/immunology
17.
EMBO J ; 27(14): 1932-43, 2008 Jul 23.
Article in English | MEDLINE | ID: mdl-18566586

ABSTRACT

The target of rapamycin (TOR), as part of the rapamycin-sensitive TOR complex 1 (TORC1), regulates various aspects of protein synthesis. Whether TOR functions in this process as part of TORC2 remains to be elucidated. Here, we demonstrate that mTOR, SIN1 and rictor, components of mammalian (m)TORC2, are required for phosphorylation of Akt and conventional protein kinase C (PKC) at the turn motif (TM) site. This TORC2 function is growth factor independent and conserved from yeast to mammals. TM site phosphorylation facilitates carboxyl-terminal folding and stabilizes newly synthesized Akt and PKC by interacting with conserved basic residues in the kinase domain. Without TM site phosphorylation, Akt becomes protected by the molecular chaperone Hsp90 from ubiquitination-mediated proteasome degradation. Finally, we demonstrate that mTORC2 independently controls the Akt TM and HM sites in vivo and can directly phosphorylate both sites in vitro. Our studies uncover a novel function of the TOR pathway in regulating protein folding and stability, processes that are most likely linked to the functions of TOR in protein synthesis.


Subject(s)
Protein Kinase C/metabolism , Protein Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Motifs , Animals , Carrier Proteins/metabolism , Cell Line , Embryo, Mammalian/cytology , Fibroblasts/metabolism , Humans , Mice , Models, Molecular , Multiprotein Complexes/metabolism , Protein Folding , Protein Kinase C/chemistry , Proto-Oncogene Proteins c-akt/chemistry , TOR Serine-Threonine Kinases
18.
Nature ; 449(7162): 564-9, 2007 Oct 04.
Article in English | MEDLINE | ID: mdl-17873860

ABSTRACT

Plasmacytoid dendritic cells (pDCs) sense viral and microbial DNA through endosomal Toll-like receptors to produce type 1 interferons. pDCs do not normally respond to self-DNA, but this restriction seems to break down in human autoimmune disease by an as yet poorly understood mechanism. Here we identify the antimicrobial peptide LL37 (also known as CAMP) as the key factor that mediates pDC activation in psoriasis, a common autoimmune disease of the skin. LL37 converts inert self-DNA into a potent trigger of interferon production by binding the DNA to form aggregated and condensed structures that are delivered to and retained within early endocytic compartments in pDCs to trigger Toll-like receptor 9. Thus, our data uncover a fundamental role of an endogenous antimicrobial peptide in breaking innate tolerance to self-DNA and suggest that this pathway may drive autoimmunity in psoriasis.


Subject(s)
Antimicrobial Cationic Peptides/metabolism , Autoantigens/metabolism , Autoimmune Diseases/metabolism , Autoimmunity , DNA/metabolism , Dendritic Cells/metabolism , Psoriasis/metabolism , Autoantigens/immunology , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Cathelicidins , DNA/immunology , Dendritic Cells/immunology , Endocytosis , Endosomes/immunology , Endosomes/metabolism , Humans , Psoriasis/immunology , Psoriasis/pathology , Toll-Like Receptor 9/immunology , Toll-Like Receptor 9/metabolism
19.
Cell ; 127(1): 125-37, 2006 Oct 06.
Article in English | MEDLINE | ID: mdl-16962653

ABSTRACT

Mammalian target of rapamycin (mTOR) controls cell growth and proliferation via the raptor-mTOR (TORC1) and rictor-mTOR (TORC2) protein complexes. Recent biochemical studies suggested that TORC2 is the elusive PDK2 for Akt/PKB Ser473 phosphorylation in the hydrophobic motif. Phosphorylation at Ser473, along with Thr308 of its activation loop, is deemed necessary for Akt function, although the regulatory mechanisms and physiological importance of each phosphorylation site remain to be fully understood. Here, we report that SIN1/MIP1 is an essential TORC2/PDK2 subunit. Genetic ablation of sin1 abolished Akt-Ser473 phosphorylation and disrupted rictor-mTOR interaction but maintained Thr308 phosphorylation. Surprisingly, defective Ser473 phosphorylation affected only a subset of Akt targets in vivo, including FoxO1/3a, while other Akt targets, TSC2 and GSK3, and the TORC1 effectors, S6K and 4E-BP1, were unaffected. Our findings reveal that the SIN1-rictor-mTOR function in Akt-Ser473 phosphorylation is required for TORC2 function in cell survival but is dispensable for TORC1 function.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Carrier Proteins/metabolism , Protein Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Carrier Proteins/genetics , Cells, Cultured , Fibroblasts/cytology , Fibroblasts/metabolism , Forkhead Box Protein O1 , Forkhead Box Protein O3 , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Silencing , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiprotein Complexes , Phosphoproteins/genetics , Phosphoproteins/metabolism , Phosphorylation , Protein Kinases/genetics , Proteins/genetics , Proteins/metabolism , Proto-Oncogene Proteins c-akt/genetics , Rapamycin-Insensitive Companion of mTOR Protein , Regulatory-Associated Protein of mTOR , Signal Transduction/physiology , Substrate Specificity , TOR Serine-Threonine Kinases , Threonine/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
20.
Clin Cancer Res ; 12(16): 4949-57, 2006 Aug 15.
Article in English | MEDLINE | ID: mdl-16914584

ABSTRACT

PURPOSE: Renal cell carcinoma (RCC) is considered immunogenic; nonetheless, rare tumor-associated antigens have been identified or are expressed in RCC. Peptidome (i.e., the total content of natural peptides of whole cells) from other tumors, such as melanoma, has proved to be immunogenic. The aims of this study were to determine whether peptidome from RCC is immunogenic and whether it contains tumor peptides shared among allogenic RCCs. EXPERIMENTAL DESIGN: Autologous dendritic cells pulsed with RCC peptidome were used to activate in vitro CD4(+) T cells from healthy donors and a metastatic RCC patient. CD4(+) T-cell polyclonal lines and clones were characterized for tumor cell recognition by proliferation assay, killing activity, and cytokine secretion. RESULTS: CD4(+) T-cell lines and clones recognized HLA-DR-matched allogenic RCC and, for the patient, the autologous tumor. RCC-reactive CD4(+) T cells showed a heterogeneous Th1 or Th0/Th2 pattern of cytokine secretion. Moreover, RCC-reactive CD4(+) T cells recognized also melanoma, colon carcinoma, cervical carcinoma, pancreas carcinoma, lung carcinoma, gastric carcinoma, and lymphoma cells but not autologous T-cell blasts. CONCLUSIONS: Our results show that (a) the RCC peptidome contain antigens recognized by CD4(+) T cells and (b) shared among tumors of different histology and (c) it induces both Th1-type and Th2/Th0-type immune responses. These data support the use of the peptidome from allogenic RCC for specific immunotherapy in RCC and possibly in other neoplastic diseases. Moreover, the CD4(+) T-cell clones generated here are useful tools for tumor antigen identification.


Subject(s)
Adenocarcinoma, Clear Cell/immunology , Antigens, Neoplasm/immunology , CD4-Positive T-Lymphocytes/immunology , Carcinoma, Renal Cell/immunology , Epitopes, T-Lymphocyte/immunology , Kidney Neoplasms/immunology , Dendritic Cells/immunology , HLA-DR Antigens/immunology , Humans , Immunotherapy/methods , Lymphocyte Activation , Peptides/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...