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










Publication year range
1.
J Crohns Colitis ; 17(11): 1817-1832, 2023 Nov 24.
Article in English | MEDLINE | ID: mdl-37208197

ABSTRACT

BACKGROUND AND AIMS: The anti-MAdCAM-1 antibody ontamalimab demonstrated efficacy in a phase II trial in ulcerative colitis and results of early terminated phase III trials are pending, but its precise mechanisms of action are still unclear. Thus, we explored the mechanisms of action of ontamalimab and compared it to the anti-α4ß7 antibody vedolizumab. METHODS: We studied MAdCAM-1 expression with RNA sequencing and immunohistochemistry. The mechanisms of action of ontamalimab were assessed with fluorescence microscopy, dynamic adhesion and rolling assays. We performed in vivo cell trafficking studies in mice and compared ontamalimab and vedolizumab surrogate [-s] antibodies in experimental models of colitis and wound healing. We analysed immune cell infiltration under anti-MAdCAM-1 and anti-α4ß7 treatment by single-cell transcriptomics and studied compensatory trafficking pathways. RESULTS: MAdCAM-1 expression was increased in active inflammatory bowel disease. Binding of ontamalimab to MAdCAM-1 induced the internalization of the complex. Functionally, ontamalimab blocked T cell adhesion similar to vedolizumab, but also inhibited L-selectin-dependent rolling of innate and adaptive immune cells. Despite conserved mechanisms in mice, the impact of ontamalimab-s and vedolizumab-s on experimental colitis and wound healing was similar. Single-cell RNA sequencing demonstrated enrichment of ontamalimab-s-treated lamina propria cells in specific clusters, and in vitro experiments indicated that redundant adhesion pathways are active in these cells. CONCLUSIONS: Ontamalimab has unique and broader mechanisms of action compared to vedolizumab. However, this seems to be compensated for by redundant cell trafficking circuits and leads to similar preclinical efficacy of anti-α4ß7 and anti-MAdCAM-1 treatment. These results will be important for the interpretation of pending phase III data.


Subject(s)
Colitis, Ulcerative , Inflammatory Bowel Diseases , Animals , Mice , Gastrointestinal Agents/pharmacology , Gastrointestinal Agents/therapeutic use , Inflammatory Bowel Diseases/drug therapy , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/therapeutic use , Colitis, Ulcerative/drug therapy , Inflammation/drug therapy , Integrins
2.
Mol Ther ; 24(1): 146-55, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26412590

ABSTRACT

Due to their ability to knock down the expression of any gene, siRNAs have been heralded as ideal candidates for treating a wide variety of diseases, including those involving "undruggable" targets. However, the therapeutic potential of siRNAs remains severely limited by a lack of effective delivery vehicles. Recently, lipid nanoparticles (LNPs) containing ionizable cationic lipids have been developed for hepatic siRNA delivery. However, their suitability for delivery to other cell types has not been determined. We have modified LNPs for preferential targeting to dendritic cells (DCs), central regulators of immune responses. To achieve directed delivery, we coated LNPs with a single-chain antibody (scFv; DEC-LNPs), specific to murine DEC205, which is highly expressed on distinct DC subsets. Here we show that injection of siRNAs encapsulated in DEC-LNPs are preferentially delivered to DEC205(+) DCs. Gene knockdown following uptake of DEC-LNPs containing siRNAs specific for the costimulatory molecules CD40, CD80, and CD86 dramatically decreases gene expression levels. We demonstrate the functionality of this knockdown with a mixed lymphocyte response (MLR). Overall, we report that injection of LNPs modified to restrict their uptake to a distinct cell population can confer profound gene knockdown, sufficient to inhibit powerful immune responses like the MLR.


Subject(s)
B7-1 Antigen/metabolism , B7-2 Antigen/metabolism , CD40 Antigens/metabolism , Dendritic Cells/immunology , Lipids/chemistry , RNA, Small Interfering/administration & dosage , Animals , Gene Expression Regulation , Injections , Liver/metabolism , Mice , Molecular Targeted Therapy , Nanoparticles/administration & dosage , Nanoparticles/chemistry
3.
Cell Cycle ; 14(7): 964-72, 2015.
Article in English | MEDLINE | ID: mdl-25830414

ABSTRACT

Necrotic cell death triggers a range of biological responses including a strong adaptive immune response, yet we know little about the cellular pathways that control necrotic cell death. Inhibitor studies suggest that proteases, and in particular cathepsins, drive necrotic cell death. The cathepsin B-selective inhibitor CA-074-Me blocks all forms of programmed necrosis by an unknown mechanism. We found that cathepsin B deficiency does not prevent induction of pyroptosis and lysosome-mediated necrosis suggesting that CA-074-Me blocks necrotic cell death by targeting cathepsins other than cathepsin B. A single cathepsin, cathepsin C, drives necrotic cell death mediated by the lysosome-destabilizing agent Leu-Leu-OMe (LLOMe). Here we present evidence that cathepsin C-deficiency and CA-074-Me block LLOMe killing in a distinct and cell type-specific fashion. Cathepsin C-deficiency and CA-074-Me block LLOMe killing of all myeloid cells, except for neutrophils. Cathepsin C-deficiency, but not CA-074-Me, blocks LLOMe killing of neutrophils suggesting that CA-074-Me does not target cathepsin C directly, consistent with inhibitor studies using recombinant cathepsin C. Unlike other cathepsins, cathepsin C lacks endoproteolytic activity, and requires activation by other lysosomal proteases, such as cathepsin D. Consistent with this theory, we found that lysosomotropic agents and cathepsin D downregulation by siRNA block LLOMe-mediated necrosis. Our findings indicate that a proteolytic cascade, involving cathepsins C and D, controls LLOMe-mediated necrosis. In contrast, cathepsins C and D were not required for pyroptotic cell death suggesting that distinct cathepsins control pyroptosis and lysosome-mediated necrosis.


Subject(s)
Cathepsin C/physiology , Cathepsin D/physiology , Lysosomes/enzymology , Animals , Apoptosis , Cathepsin B/antagonists & inhibitors , Cathepsin B/physiology , Dipeptides/pharmacology , Lysosomes/drug effects , Mice, Inbred BALB C , Mice, Inbred C57BL , Necrosis
4.
Genome Med ; 6(9): 68, 2014.
Article in English | MEDLINE | ID: mdl-25302077

ABSTRACT

BACKGROUND: The evolutionarily conserved DNA mismatch repair (MMR) system corrects base-substitution and insertion-deletion mutations generated during erroneous replication. The mutation or inactivation of many MMR factors strongly predisposes to cancer, where the resulting tumors often display resistance to standard chemotherapeutics. A new direction to develop targeted therapies is the harnessing of synthetic genetic interactions, where the simultaneous loss of two otherwise non-essential factors leads to reduced cell fitness or death. High-throughput screening in human cells to directly identify such interactors for disease-relevant genes is now widespread, but often requires extensive case-by-case optimization. Here we asked if conserved genetic interactors (CGIs) with MMR genes from two evolutionary distant yeast species (Saccharomyces cerevisiae and Schizosaccharomyzes pombe) can predict orthologous genetic relationships in higher eukaryotes. METHODS: High-throughput screening was used to identify genetic interaction profiles for the MutSα and MutSß heterodimer subunits (msh2Δ, msh3Δ, msh6Δ) of fission yeast. Selected negative interactors with MutSß (msh2Δ/msh3Δ) were directly analyzed in budding yeast, and the CGI with SUMO-protease Ulp2 further examined after RNA interference/drug treatment in MSH2-deficient and -proficient human cells. RESULTS: This study identified distinct genetic profiles for MutSα and MutSß, and supports a role for the latter in recombinatorial DNA repair. Approximately 28% of orthologous genetic interactions with msh2Δ/msh3Δ are conserved in both yeasts, a degree consistent with global trends across these species. Further, the CGI between budding/fission yeast msh2 and SUMO-protease Ulp2 is maintained in human cells (MSH2/SENP6), and enhanced by Olaparib, a PARP inhibitor that induces the accumulation of single-strand DNA breaks. This identifies SENP6 as a promising new target for the treatment of MMR-deficient cancers. CONCLUSION: Our findings demonstrate the utility of employing evolutionary distance in tractable lower eukaryotes to predict orthologous genetic relationships in higher eukaryotes. Moreover, we provide novel insights into the genome maintenance functions of a critical DNA repair complex and propose a promising targeted treatment for MMR deficient tumors.

5.
Immunity ; 40(6): 974-88, 2014 Jun 19.
Article in English | MEDLINE | ID: mdl-24931122

ABSTRACT

Cells of the innate immune system are essential for host defenses against primary microbial pathogen infections, yet their involvement in effective memory responses of vaccinated individuals has been poorly investigated. Here we show that memory T cells instruct innate cells to become potent effector cells in a systemic and a mucosal model of infection. Memory T cells controlled phagocyte, dendritic cell, and NK or NK T cell mobilization and induction of a strong program of differentiation, which included their expression of effector cytokines and microbicidal pathways, all of which were delayed in nonvaccinated hosts. Disruption of IFN-γ signaling in Ly6C+ monocytes, dendritic cells, and macrophages impaired these processes and the control of pathogen growth. These results reveal how memory T cells, through rapid secretion of IFN-γ, orchestrate extensive modifications of host innate immune responses that are essential for effective protection of vaccinated hosts.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Immunologic Memory , Interferon-gamma/immunology , Lymphocyte Activation/immunology , Adoptive Transfer , Animals , Antigens, Ly , Cell Differentiation/immunology , Cytokines/immunology , Dendritic Cells/immunology , Female , Herpesvirus 2, Human/immunology , Immunity, Innate , Immunization , Killer Cells, Natural/immunology , Listeria monocytogenes/immunology , Listeriosis/immunology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/immunology , Natural Killer T-Cells/immunology , Phagocytosis/immunology , Receptors, Interferon/genetics , Interferon gamma Receptor
6.
Mol Ther ; 22(7): 1375-1387, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24682172

ABSTRACT

Effective therapeutic vaccines often require activation of T cell-mediated immunity. Robust T cell activation, including CD8 T cell responses, can be achieved using antibodies or antibody fragments to direct antigens of interest to professional antigen presenting cells. This approach represents an important advance in enhancing vaccine efficacy. Nucleic acid aptamers present a promising alternative to protein-based targeting approaches. We have selected aptamers that specifically bind the murine receptor, DEC205, a C-type lectin expressed predominantly on the surface of CD8α(+) dendritic cells (DCs) that has been shown to be efficient at facilitating antigen crosspresentation and subsequent CD8(+) T cell activation. Using a minimized aptamer conjugated to the model antigen ovalbumin (OVA), DEC205-targeted antigen crosspresentation was verified in vitro and in vivo by proliferation and cytokine production by primary murine CD8(+) T cells expressing a T cell receptor specific for the major histocompatibility complex (MHC) I-restricted OVA257-264 peptide SIINFEKL. Compared with a nonspecific ribonucleic acid (RNA) of similar length, DEC205 aptamer-OVA-mediated antigen delivery stimulated strong proliferation and production of interferon (IFN)-γ and interleukin (IL)-2. The immune responses elicited by aptamer-OVA conjugates were sufficient to inhibit the growth of established OVA-expressing B16 tumor cells. Our results demonstrate a new application of aptamer technology for the development of effective T cell-mediated vaccines.


Subject(s)
Antigen Presentation/immunology , Antigens/immunology , Aptamers, Nucleotide/administration & dosage , Aptamers, Nucleotide/genetics , Animals , Antigens/administration & dosage , CD8-Positive T-Lymphocytes/immunology , CHO Cells , Cricetinae , Cricetulus , Dendritic Cells/metabolism , Immunity, Cellular , Interferon-gamma/metabolism , Mice , Mice, Inbred C57BL
7.
Mol Ther ; 20(3): 616-24, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22233578

ABSTRACT

We have developed a selection scheme to generate nucleic acid sequences that recognize and directly internalize into mammalian cells without the aid of conventional delivery methods. To demonstrate the generality of the technology, two independent selections with different starting pools were performed against distinct target cells. Each selection yielded a single highly functional sequence, both of which folded into a common core structure. This internalization signal can be adapted for use as a general purpose reagent for transfection into a wide variety of cell types including primary cells.


Subject(s)
RNA/chemistry , Transfection , Animals , Base Sequence , Cell Line , Endocytosis , Female , Flow Cytometry , Gene Library , Gene Transfer Techniques , Humans , Male , Mice , Mice, Inbred BALB C , Mucous Membrane/metabolism , Nucleic Acid Conformation , Nucleotide Motifs , Vagina/metabolism
8.
Discov Med ; 11(57): 124-32, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21356167

ABSTRACT

RNA interference (RNAi) describes a highly conserved pathway, present in eukaryotic cells, for regulating gene expression. Small stretches of double-stranded RNA, termed small interfering RNAs (siRNAs), utilize this pathway to bind homologous mRNA, resulting in site-specific mRNA cleavage and subsequent protein degradation. The ubiquitous presence of the RNAi machinery, combined with its specificity and efficacy, makes it an attractive mechanism for reducing aberrant gene expression in therapeutic settings. However, a major obstacle to utilizing RNAi in the clinic is siRNA delivery. Administered siRNAs must make contact with the appropriate cell types and, following internalization, gain access to the cytosol where the RNAi machinery resides. This must be achieved so that silencing is maximized, whilst minimizing any undesirable off-target effects. Recently, the utility of siRNAs as a microbicide, usually applied to the genital mucosa for preventing transmission of sexually transmitted diseases including HIV-1 and HSV-2, has been investigated. In this review we will describe these studies and discuss potential strategies for improving gene silencing.


Subject(s)
Gene Transfer Techniques , Genitalia/metabolism , Mucous Membrane/metabolism , RNA, Small Interfering/metabolism , Animals , Anti-Infective Agents/pharmacology , Genitalia/drug effects , Humans , Mucous Membrane/drug effects
9.
Curr Opin Mol Ther ; 12(2): 192-202, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20373263

ABSTRACT

Sexually transmitted infections (STIs) are a major cause of morbidity and mortality worldwide. Although a vaccine is available for HPV, no effective vaccines exist for the HIV-1 and HSV-2 viral pathogens, and there are no cures for these infections. Furthermore, recent setbacks in clinical trials, such as the failure of the STEP trial to prevent HIV-1 infection, have emphasized the need to develop alternative approaches to interrupt the transmission of these pathogens. One alternative strategy is represented by the use of topically applied microbicides, and such agents are being developed against various viruses. RNAi-based microbicides have recently been demonstrated to prevent HSV-2 transmission, and may be useful for targeting multiple STIs. In this review, microbicides that are under development for the prevention of STIs are described, with a focus on topically applied microbicidal siRNAs.


Subject(s)
Anti-Infective Agents , RNA, Small Interfering , Sexually Transmitted Diseases/drug therapy , Anti-Infective Agents/chemistry , Anti-Infective Agents/therapeutic use , Disease Transmission, Infectious/prevention & control , HIV Infections/drug therapy , HIV Infections/prevention & control , HIV Infections/transmission , Herpes Genitalis/drug therapy , Herpes Genitalis/prevention & control , Herpes Genitalis/transmission , Humans , Papillomavirus Infections/drug therapy , Papillomavirus Infections/prevention & control , Papillomavirus Infections/transmission , RNA Interference , RNA, Small Interfering/chemistry , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Sexually Transmitted Diseases/mortality , Sexually Transmitted Diseases/prevention & control
10.
Cell Host Microbe ; 5(1): 84-94, 2009 Jan 22.
Article in English | MEDLINE | ID: mdl-19154990

ABSTRACT

A vaginal microbicide should prevent pathogen transmission without disrupting tissue barriers to infection. Ideally, it would not need to be applied immediately before sexual intercourse, when compliance is a problem. Intravaginal administration of small interfering RNA (siRNA) lipoplexes targeting Herpes Simplex Virus Type 2 (HSV-2) genes protects mice from HSV-2. However, protection is short-lived, and the transfection lipid on its own unacceptably enhances transmission. Here, we show that cholesterol-conjugated (chol)-siRNAs without lipid silence gene expression in the vagina without causing inflammation or inducing interferons. A viral siRNA prevents transmission within a day of challenge, whereas an siRNA targeting the HSV-2 receptor nectin-1 protects for a week, but protection is delayed for a few days until the receptor is downmodulated. Combining siRNAs targeting a viral and host gene protects mice from HSV-2 for a week, irrespective of the time of challenge. Therefore, intravaginal siRNAs could provide sustained protection against viral transmission.


Subject(s)
Antiviral Agents/administration & dosage , Disease Transmission, Infectious/prevention & control , Herpes Genitalis/prevention & control , Herpes Genitalis/transmission , Herpesvirus 2, Human/genetics , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Administration, Intravaginal , Animals , Antiviral Agents/chemistry , Cell Adhesion Molecules/antagonists & inhibitors , Female , Gene Silencing , Mice , Mice, Inbred BALB C , Nectins , RNA, Small Interfering/chemistry , Survival Analysis , Vagina/pathology , Vagina/virology
11.
Nature ; 439(7072): 89-94, 2006 Jan 05.
Article in English | MEDLINE | ID: mdl-16306938

ABSTRACT

Herpes simplex virus 2 (HSV-2) infection causes significant morbidity and is an important cofactor for the transmission of HIV infection. A microbicide to prevent sexual transmission of HSV-2 would contribute substantially to controlling the spread of HIV and other infections. Because RNA interference (RNAi) provides effective antiviral defence in plants and other organisms, several studies have focused on harnessing RNAi to inhibit viral infection. Here we show that vaginal instillation of small interfering RNAs (siRNAs) targeting HSV-2 protects mice from lethal infection. siRNAs mixed with lipid are efficiently taken up by epithelial and lamina propria cells and silence gene expression in the mouse vagina and ectocervix for at least nine days. Intravaginal application of siRNAs targeting the HSV-2 UL27 and UL29 genes (which encode an envelope glycoprotein and a DNA binding protein, respectively) was well tolerated, did not induce interferon-responsive genes or cause inflammation, and protected mice when administered before and/or after lethal HSV-2 challenge. These results suggest that siRNAs are attractive candidates for the active component of a microbicide designed to prevent viral infection or transmission.


Subject(s)
Herpes Genitalis/prevention & control , Herpesvirus 2, Human/genetics , Herpesvirus 2, Human/pathogenicity , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Administration, Intravaginal , Animals , Cell Line , Cervix Uteri/virology , Female , Gene Silencing , Genes, Essential/genetics , Genes, Viral/genetics , Herpes Genitalis/complications , Herpesvirus 2, Human/physiology , Inflammation , Interferons/physiology , Liposomes/administration & dosage , Mice , Mice, Inbred BALB C , RNA, Small Interfering/administration & dosage , Time Factors , Vagina/virology , Viral Proteins/genetics , Virus Replication
12.
J Immunol ; 175(1): 421-32, 2005 Jul 01.
Article in English | MEDLINE | ID: mdl-15972676

ABSTRACT

The intracellular bacterium Listeria monocytogenes infects dendritic cells (DC) and other APCs and induces potent cell-mediated protective immunity. However, heat-killed bacteria fail to do so. This study explored whether DC differentially respond to live and killed Listeria and how this affects T cell activation. To control for bacterial number, a replication-deficient strain, Lmdd, defective in D-alanine biosynthesis, was used. We found that DC internalize both live and heat-killed Lmdd and similarly up-regulate the expression of costimulatory molecules, a necessary step for T cell activation. However, only live Lmdd-infected DC stimulate T cells to express the early activation marker CD69 and enhance T cell activation upon TCR engagement. Infection with live, but not heat-killed, Lmdd induces myeloid DC to secrete copious amounts of IFN-beta, which requires bacterial cytosolic invasion. Exposure to high concentrations of IFN-beta sensitizes naive T cells for Ag-dependent activation.


Subject(s)
Dendritic Cells/immunology , Interferon-beta/biosynthesis , Listeria monocytogenes/immunology , Listeria monocytogenes/pathogenicity , Myeloid Cells/immunology , T-Lymphocytes/immunology , Animals , Antigens, CD/metabolism , Antigens, Differentiation, T-Lymphocyte/metabolism , CD3 Complex/immunology , Hot Temperature , In Vitro Techniques , Interferon-beta/genetics , Lectins, C-Type , Listeria monocytogenes/genetics , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mutation , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Antigen, T-Cell/metabolism
13.
Nat Biotechnol ; 23(6): 709-17, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15908939

ABSTRACT

Delivery of small interfering RNAs (siRNAs) into cells is a key obstacle to their therapeutic application. We designed a protamine-antibody fusion protein to deliver siRNA to HIV-infected or envelope-transfected cells. The fusion protein (F105-P) was designed with the protamine coding sequence linked to the C terminus of the heavy chain Fab fragment of an HIV-1 envelope antibody. siRNAs bound to F105-P induced silencing only in cells expressing HIV-1 envelope. Additionally, siRNAs targeted against the HIV-1 capsid gene gag, inhibited HIV replication in hard-to-transfect, HIV-infected primary T cells. Intratumoral or intravenous injection of F105-P-complexed siRNAs into mice targeted HIV envelope-expressing B16 melanoma cells, but not normal tissue or envelope-negative B16 cells; injection of F105-P with siRNAs targeting c-myc, MDM2 and VEGF inhibited envelope-expressing subcutaneous B16 tumors. Furthermore, an ErbB2 single-chain antibody fused with protamine delivered siRNAs specifically into ErbB2-expressing cancer cells. This study demonstrates the potential for systemic, cell-type specific, antibody-mediated siRNA delivery.


Subject(s)
Drug Delivery Systems , RNA, Small Interfering/administration & dosage , Receptors, Cell Surface/physiology , Animals , Antibodies/chemistry , COS Cells , HIV-1 , Humans , Interferons/metabolism , Melanoma, Experimental/therapy , Mice , Receptors, HIV/physiology , Recombinant Fusion Proteins
14.
J Immunol ; 174(4): 1879-87, 2005 Feb 15.
Article in English | MEDLINE | ID: mdl-15699114

ABSTRACT

Soluble heat shock fusion proteins (Hsfp) stimulate mice to produce CD8+ CTL, indicating that these proteins are cross-presented by dendritic cells (DC) to naive CD8 T cells. We report that cross-presentation of these proteins depends upon their binding to DC receptors, likely belonging to the scavenger receptor superfamily. Hsfp entered DC by receptor-mediated endocytosis that was either inhibitable by cytochalasin D or not inhibitable, depending upon aggregation state and time. Most endocytosed Hsfp was transported to lysosomes, but not the small cross-presented fraction that exited early from the endocytic pathway and required access to proteasomes and TAP. Naive CD8 T cell (2C and OT-I) responses to DC incubated with Hsfp at 1 microM were matched by incubating DC with cognate octapeptides at 1-10 pM, indicating that display of very few class I MHC-peptide complexes per DC can be sufficient for cross-presentation. With an Hsfp (heat shock protein-OVA) having peptide sequences for both CD4+ (OT-II) and CD8+ (OT-I) cells, the CD4 cells responded far more vigorously than the CD8 cells and many more class II MHC-peptide than class I MHC-peptide complexes were displayed.


Subject(s)
Bacterial Proteins/immunology , Chaperonins/immunology , Cross-Priming/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Intracellular Fluid/immunology , Intracellular Fluid/metabolism , Recombinant Fusion Proteins/immunology , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/physiology , Amino Acid Sequence , Animals , Antigen Presentation/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cells, Cultured , Chaperonin 60 , Chaperonins/genetics , Chaperonins/metabolism , Cross-Priming/genetics , Dendritic Cells/enzymology , Endocytosis/immunology , Endosomes/immunology , Endosomes/metabolism , Lysosomes/immunology , Lysosomes/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Molecular Sequence Data , Nucleocapsid Proteins , Nucleoproteins/genetics , Nucleoproteins/immunology , Nucleoproteins/metabolism , Ovalbumin/immunology , Ovalbumin/metabolism , Phagocytosis/immunology , Proteasome Endopeptidase Complex/metabolism , Protein Binding/immunology , Protein Transport/immunology , RNA-Binding Proteins/genetics , RNA-Binding Proteins/immunology , RNA-Binding Proteins/metabolism , Receptors, Immunologic/metabolism , Receptors, Scavenger , Recombinant Fusion Proteins/metabolism , Signal Transduction/immunology , Solubility , Viral Core Proteins/genetics , Viral Core Proteins/immunology , Viral Core Proteins/metabolism
15.
J Immunol ; 172(6): 3415-21, 2004 Mar 15.
Article in English | MEDLINE | ID: mdl-15004140

ABSTRACT

We describe a role for myeloid differentiation factor 88 (MyD88) in the induction of functional CTLs in vivo, in response to exogenously administered Ag, using a heat shock fusion protein, hsp65-P1, as a model Ag. CD8 T cells transferred into MyD88-deficient animals produce normal numbers of CD8 effector cells that have normal activation marker profiles after immunization with hsp65-P1. However, these CD8 T cells produced significantly less IFN-gamma and showed reduced killing activity. This reduction in activation of functional CTLs appears to be unrelated to Toll-like receptor 4 function, because in vitro hsp65-P1-experienced Toll-like receptor 4-deficient dendritic cells (DCs), but not MyD88-deficient DCs, activated CD8 T cells to a similar extent to wild-type DCs. We identify a cross-presentation defect in MyD88-deficient DCs that, when treated with hsp65-P1 fusion protein, results in surface display of fewer SIYRYYGL/class I MHC complexes. Thus, MyD88 plays a role in the developmental maturation of DCs that allows them to prime CD8 T cells through cross-presentation.


Subject(s)
Antigen Presentation , Antigens, Differentiation/physiology , Cytotoxicity, Immunologic , Immunization , Receptors, Immunologic/physiology , T-Lymphocytes, Cytotoxic/immunology , Adaptor Proteins, Signal Transducing , Animals , Antigen Presentation/genetics , Antigens, Bacterial/administration & dosage , Antigens, Bacterial/metabolism , Antigens, Differentiation/genetics , Bacterial Proteins/administration & dosage , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Bacterial Proteins/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , Cells, Cultured , Chaperonin 60 , Chaperonins/administration & dosage , Chaperonins/genetics , Chaperonins/immunology , Chaperonins/metabolism , Cytotoxicity, Immunologic/genetics , Dendritic Cells/cytology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Immunization/methods , Injections, Subcutaneous , Lymphocyte Activation/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mycobacterium tuberculosis/immunology , Myeloid Differentiation Factor 88 , Peptide Fragments/immunology , Peptide Fragments/metabolism , Receptors, Immunologic/deficiency , Receptors, Immunologic/genetics , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/transplantation
16.
J Immunol ; 172(5): 2885-93, 2004 Mar 01.
Article in English | MEDLINE | ID: mdl-14978090

ABSTRACT

Recombinant heat shock fusion proteins (Hsfp) injected into mice without added adjuvants can stimulate production of CD8 cytolytic T cells. Because initiation of productive immune responses generally requires dendritic cell (DC) activation, the question arises as to whether the Hsfp can activate DC independently of contaminating LPS. Using microarray analyses of DC from LPS-insensitive mice having a point mutation in Toll-like receptor 4 (Tlr4) (C3H/HeJ), or lacking Tlr4 (B10/ScNCr), we show here that unlike a LPS standard, Hsfp activated DC from HeJ mice almost as well as DC from wild-type mice. Consistent with the microarray analysis, the Hsfp's ability to activate DC was not eliminated by polymyxin B but was destroyed by proteinase K. The Hsfp did not, however, stimulate DC from mice lacking Tlr4. In vivo the CD8 T cell response to the Hsfp in mice lacking Tlr4 was impaired: the responding CD8 cells initially proliferated vigorously but their development into cytolytic effector cells was diminished. Overall, the results indicate that this Hsfp can activate DC independently of LPS but still requires Tlr4 for an optimal CD8 T cell response.


Subject(s)
Bacterial Proteins/physiology , Chaperonins/physiology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Membrane Glycoproteins/physiology , Receptors, Cell Surface/physiology , Recombinant Fusion Proteins/physiology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , Animals , Bacterial Proteins/administration & dosage , Cell Differentiation/immunology , Chaperonin 60 , Chaperonins/administration & dosage , Cytotoxicity, Immunologic/genetics , Dendritic Cells/drug effects , Dose-Response Relationship, Immunologic , Endopeptidase K/pharmacology , Gene Expression Profiling , Lipopolysaccharides/administration & dosage , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Polymyxin B/pharmacology , Receptors, Cell Surface/deficiency , Receptors, Cell Surface/genetics , Recombinant Fusion Proteins/administration & dosage , T-Lymphocytes, Cytotoxic/cytology , Toll-Like Receptor 4 , Toll-Like Receptors , Up-Regulation/genetics , Up-Regulation/immunology
17.
J Immunol ; 172(5): 2994-3002, 2004 Mar 01.
Article in English | MEDLINE | ID: mdl-14978103

ABSTRACT

We identify and consider some characteristics of a peptide antagonist for the Ag-specific receptor on 2C cells (the 2C TCR). The peptide, GNYSFYAL (called GNY), binds to H-2K(b), and a very high-resolution crystal structure of the GNY-K(b) complex at 1.35 A is described. Although the GNY peptide does not bind to L(d), the potency of GNY-K(b) as an antagonist is evident from its ability to specifically inhibit 2C TCR-mediated reactions to an allogenic agonist complex (QLSPFPFDL-L(d)), as well as to a syngeneic agonist complex (SIYRYYGL-K(b)). The crystal structure and the activities of alanine-substituted peptide variants point to the properties of the peptide P4 side chain and the conformation of the Tyr-P6 side chain as the structural determinants of GNYSFYAL antagonist activity.


Subject(s)
Isoantigens/physiology , Oligopeptides/chemistry , Oligopeptides/physiology , Receptors, Antigen, T-Cell/antagonists & inhibitors , Receptors, Antigen, T-Cell/physiology , Alanine/metabolism , Amino Acid Sequence , Amino Acid Substitution/immunology , Animals , Arginine/metabolism , Cell Line , Cell Line, Tumor , Clone Cells , Crystallography, X-Ray , Cytotoxicity Tests, Immunologic , Female , H-2 Antigens/metabolism , Histocompatibility Antigen H-2D , Lysine/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oligopeptides/metabolism , Protein Binding/immunology , Receptors, Antigen, T-Cell/agonists , Serine/metabolism , Structure-Activity Relationship
18.
RNA ; 9(4): 493-501, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12649500

ABSTRACT

Genome-wide genetic approaches have proven useful for examining pathways of biological significance in model organisms such as Saccharomyces cerevisiae, Drosophila melanogastor, and Caenorhabditis elegans, but similar techniques have proven difficult to apply to mammalian systems. Although manipulation of the murine genome has led to identification of genes and their function, this approach is laborious, expensive, and often leads to lethal phenotypes. RNA interference (RNAi) is an evolutionarily conserved process of gene silencing that has become a powerful tool for investigating gene function by reverse genetics. Here we describe the delivery of cassettes expressing hairpin RNA targeting green fluorescent protein (GFP) using Moloney leukemia virus-based and lentivirus-based retroviral vectors. Both transformed cell lines and primary dendritic cells, normally refractory to transfection-based gene transfer, demonstrated stable silencing of targeted genes, including the tumor suppressor gene TP53 in normal human fibroblasts. This report demonstrates that both Moloney leukemia virus and lentivirus vector-mediated expression of RNAi can achieve effective, stable gene silencing in diverse biological systems and will assist in elucidating gene functions in numerous cell types including primary cells.


Subject(s)
Lentivirus/genetics , Lentivirus/physiology , RNA Interference/physiology , Dendritic Cells , Genes, Reporter , HeLa Cells , Humans , In Vitro Techniques
19.
Proc Natl Acad Sci U S A ; 99(5): 2983-8, 2002 Mar 05.
Article in English | MEDLINE | ID: mdl-11854473

ABSTRACT

Naive T cells do not proliferate in normal individuals in the absence of antigen stimulation, but they proliferate spontaneously when T cells are severely depleted. We show here that coculture of syngeneic dendritic cells (DC) with naive T cells expressing a single T cell receptor also results in T cell proliferation in the absence of foreign antigen. As in lymphopenic mice, where T cell proliferation depends upon DC, this response in the coculture system requires interaction of the T cells' T cell receptor with self-peptide-MHCs on DC. This in vitro proliferation also requires soluble factors, including IL-15 secreted by DC, and can be inhibited potently by cell-cell contact with CD4+CD25+ regulatory T cells. The coculture system described may illuminate mechanisms that maintain stable numbers of T cells in normal individuals.


Subject(s)
Dendritic Cells/metabolism , T-Lymphocytes/cytology , Animals , CD4-Positive T-Lymphocytes/metabolism , Cell Division , Coculture Techniques , Cytokines/metabolism , Female , Homeodomain Proteins/genetics , Homeostasis , Interleukin-15/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Receptors, Interleukin-2 , Solubility
20.
Int Immunol ; 14(1): 23-30, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11751748

ABSTRACT

Many murine T cell clones grow continuously in culture despite weekly ligation of their TCR by antigen. To learn how the cultured cells avoid or minimize antigen-induced cell death (AICD), we compared Fas and tumor necrosis factor (TNF) receptors (TNFR) on several long-term cultured CD8(+) T cell clones with those on naive and activated naive cells expressing the same TCR (2C). In contrast to the naive cells, Fas was absent on the cultured clones and the TNFR-II receptor, present initially at high levels on the cultured cells, was rapidly down-modulated in response to TCR ligation and had virtually disappeared by 2 h, when only approximately 10% of the cloned cells had been induced to express TNF-alpha. The extent of AICD of the cultured clones in response to cognate peptide-MHC on the presenting cells used for routine stimulation of the cultures was also considerably less than the massive cell death of the clones following exposure to anti-CD3 antibody plate-bound at high density.


Subject(s)
CD8-Positive T-Lymphocytes/physiology , Receptors, Antigen, T-Cell/physiology , Animals , Apoptosis , Cell Culture Techniques , Cell Survival , Clone Cells , Mice , Receptors, Tumor Necrosis Factor/metabolism , T-Lymphocytes, Cytotoxic/physiology , fas Receptor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...