Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Dis Model Mech ; 16(10)2023 10 01.
Article in English | MEDLINE | ID: mdl-37718909

ABSTRACT

Sezary syndrome (SS) is a rare, aggressive leukemic variant of cutaneous T-cell lymphoma (CTCL) that lacks adequate therapeutic options and representative small-animal models. Here, we demonstrate that IL-15 is a critical CTCL growth factor. Importantly, an immunodeficient knock-in mouse model genetically engineered to express human IL-15 uniquely supported the growth of SS patient samples relative to conventional immunodeficient mouse strains. SS patient-derived xenograft (PDX) models recapacitated key pathological features of the human disease, including skin infiltration and spread of leukemic cells to the periphery, and maintained the dependence on human IL-15 upon serial in vivo passaging. Detailed molecular characterization of the engrafted cells by single-cell transcriptomic analysis revealed congruent neoplastic gene expression signatures but distinct clonal engraftment patterns. Overall, we document an important dependence of Sezary cell survival and proliferation on IL-15 signaling and the utility of immunodeficient humanized IL-15 mice as hosts for SS - and potentially other T and NK cell-derived hematologic malignancies - PDX model generation. Furthermore, these studies advocate the thorough molecular understanding of the resultant PDX models to maximize their translational impact.


Subject(s)
Lymphoma, T-Cell, Cutaneous , Sezary Syndrome , Skin Neoplasms , Humans , Animals , Mice , Skin Neoplasms/metabolism , Interleukin-15 , Lymphoma, T-Cell, Cutaneous/pathology , Sezary Syndrome/metabolism , Sezary Syndrome/pathology , Lymphocytes/metabolism , Tumor Microenvironment
2.
Commun Biol ; 6(1): 447, 2023 04 25.
Article in English | MEDLINE | ID: mdl-37185301

ABSTRACT

Efficacy of immune checkpoint inhibitors in cancers can be limited by CD8 T cell dysfunction or HLA-I down-regulation. Tumor control mechanisms independent of CD8/HLA-I axis would overcome these limitations. Here, we report potent CD4 T cell-mediated tumor regression and memory responses in humanized immune system (HIS) mice implanted with HT-29 colorectal tumors. The regressing tumors showed increased CD4 cytotoxic T lymphocyte (CTL) infiltration and enhanced tumor HLA-II expression compared to progressing tumors. The intratumoral CD4 T cell subset associated with tumor regression expressed multiple cytotoxic markers and exhibited clonal expansion. Notably, tumor control was abrogated by depletion of CD4 but not CD8 T cells. CD4 T cells derived from tumor-regressing mice exhibited HLA-II-dependent and tumor-specific killing ex vivo. Taken together, our study demonstrates a critical role of human CD4 CTLs in mediating tumor clearance independent of CD8 T cells and provides a platform to study human anti-tumor immunity in vivo.


Subject(s)
Neoplasms , T-Lymphocytes, Cytotoxic , Humans , Mice , Animals , CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , Neoplasms/metabolism
3.
Commun Biol ; 6(1): 444, 2023 04 22.
Article in English | MEDLINE | ID: mdl-37087494

ABSTRACT

Immunodeficient mice reconstituted with a human immune system (HIS mice) give rise to human T cells, which make them an attractive system to study human immune responses to tumors. However, such HIS mice typically exhibit sub-optimal responses to immune challenges as well as fail to develop antigen-specific B or T cell memory. Here we report HIS mice mediate spontaneous regression of human B cell lymphoma Raji. Tumor regression was dependent on CD4+ and CD8+ T cell responses and resulted in T cell memory. The T cell memory elicited was mainly Raji-specific, however some level of cross-protection was also elicited to a related B cell lymphoma cell line Ramos. Single-cell RNAseq analysis indicated activation of CD8+ T cells in regressing Raji tumors as well as clonal expansion of specific T cell receptors (TCRs). Cloning of TCRs from Raji-infiltrating T cells into a Jurkat reporter cell line showed reactivity specific for Raji tumor cells. Overall, we report a platform for studying in vivo human T cell tumor immunity by highlighting spontaneous Raji tumor regression, clonal TCR expansion, and T cell memory in HIS mice.


Subject(s)
CD8-Positive T-Lymphocytes , Lymphoma, B-Cell , Humans , Mice , Animals , Receptors, Antigen, T-Cell/metabolism , Jurkat Cells , Lymphoma, B-Cell/metabolism
4.
Sci Adv ; 9(15): eadf4490, 2023 04 14.
Article in English | MEDLINE | ID: mdl-37058568

ABSTRACT

Liver steatosis is an increasing health issue with few therapeutic options, partly because of a paucity of experimental models. In humanized liver rodent models, abnormal lipid accumulation in transplanted human hepatocytes occurs spontaneously. Here, we demonstrate that this abnormality is associated with compromised interleukin-6 (IL-6)-glycoprotein 130 (GP130) signaling in human hepatocytes because of incompatibility between host rodent IL-6 and human IL-6 receptor (IL-6R) on donor hepatocytes. Restoration of hepatic IL-6-GP130 signaling, through ectopic expression of rodent IL-6R, constitutive activation of GP130 in human hepatocytes, or humanization of an Il6 allele in recipient mice, substantially reduced hepatosteatosis. Notably, providing human Kupffer cells via hematopoietic stem cell engraftment in humanized liver mice also corrected the abnormality. Our observations suggest an important role of IL-6-GP130 pathway in regulating lipid accumulation in hepatocytes and not only provide a method to improve humanized liver models but also suggest therapeutic potential for manipulating GP130 signaling in human liver steatosis.


Subject(s)
Fatty Liver , Interleukin-6 , Humans , Mice , Animals , Interleukin-6/metabolism , Cytokine Receptor gp130/metabolism , Lipid Droplets/metabolism , Hepatocytes/metabolism , Glycoproteins , Lipids
5.
Sci Transl Med ; 14(670): eabn1082, 2022 11 09.
Article in English | MEDLINE | ID: mdl-36350988

ABSTRACT

Although many patients with diffuse large B cell lymphoma (DLBCL) may achieve a complete response to frontline chemoimmunotherapy, patients with relapsed/refractory disease typically have poor outcomes. Odronextamab, a CD20xCD3 bispecific antibody that provides "signal 1" through the activation of the T cell receptor/CD3 complex, has exhibited early, promising activity for patients with highly refractory DLBCL in phase 1 trials. However, not all patients achieve complete responses, and many relapse, thus representing a high unmet medical need. Here, we investigated whether adding a costimulatory "signal 2" by engaging CD28 receptors on T cells could augment odronextamab activity. We demonstrate that REGN5837, a bispecific antibody that cross-links CD22-expressing tumor cells with CD28-expressing T cells, enhances odronextamab by potentiating T cell activation and cytolytic function. In preclinical DLBCL studies using human immune system-reconstituted animals, REGN5837 promotes the antitumor activity of odronextamab and induces intratumoral expansion of reprogrammable T cells while skewing away from a dysfunctional state. Although REGN5837 monotherapy shows limited activity and no toxicity in primate studies, it augments T cell activation when dosed in combination with odronextamab. In addition, analysis of non-Hodgkin lymphoma clinical samples reveals an increase in CD28+CD8+ T cells after odronextamab treatment, demonstrating the presence of a population that could potentially be targeted by REGN5837. Collectively, our data demonstrate that REGN5837 can markedly enhance the antitumor activity of odronextamab in preclinical NHL models, and the combination of these two bispecific antibodies may provide a chemotherapy-free approach for the treatment of DLBCL.


Subject(s)
Antibodies, Bispecific , Antineoplastic Agents , Lymphoma, Large B-Cell, Diffuse , Lymphoma, Non-Hodgkin , Animals , Humans , Antibodies, Bispecific/pharmacology , Antibodies, Bispecific/therapeutic use , CD28 Antigens , CD8-Positive T-Lymphocytes , Antigens, CD19 , Neoplasm Recurrence, Local/drug therapy , Lymphoma, Non-Hodgkin/drug therapy , Lymphoma, Large B-Cell, Diffuse/drug therapy , Antineoplastic Agents/pharmacology , Sialic Acid Binding Ig-like Lectin 2/therapeutic use
6.
Proc Natl Acad Sci U S A ; 114(45): E9626-E9634, 2017 11 07.
Article in English | MEDLINE | ID: mdl-29078283

ABSTRACT

Immunodeficient mice reconstituted with a human immune system represent a promising tool for translational research as they may allow modeling and therapy of human diseases in vivo. However, insufficient development and function of human natural killer (NK) cells and T cell subsets limit the applicability of humanized mice for studying cancer biology and therapy. Here, we describe a human interleukin 15 (IL15) and human signal regulatory protein alpha (SIRPA) knock-in mouse on a Rag2-/- Il2rg-/- background (SRG-15). Transplantation of human hematopoietic stem and progenitor cells into SRG-15 mice dramatically improved the development and functional maturation of circulating and tissue-resident human NK and CD8+ T cells and promoted the development of tissue-resident innate lymphoid cell (ILC) subsets. Profiling of human NK cell subsets by mass cytometry revealed a highly similar expression pattern of killer inhibitory receptors and other candidate molecules in NK cell subpopulations between SRG-15 mice and humans. In contrast to nonobese diabetic severe combined immunodeficient Il2rg-/- (NSG) mice, human NK cells in SRG-15 mice did not require preactivation but infiltrated a Burkitt's lymphoma xenograft and efficiently inhibited tumor growth following treatment with the therapeutic antibody rituximab. Our humanized mouse model may thus be useful for preclinical testing of novel human NK cell-targeted and combinatory cancer immunotherapies and for studying how they elicit human antitumor immune responses in vivo.


Subject(s)
Killer Cells, Natural/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Disease Models, Animal , Humans , Immunity, Innate/immunology , Interleukin Receptor Common gamma Subunit/immunology , Interleukin-15/immunology , Lymphocytes/immunology , Mice , Mice, SCID , Receptors, Immunologic/immunology , Rituximab/immunology
7.
Blood ; 129(8): 959-969, 2017 02 23.
Article in English | MEDLINE | ID: mdl-28077418

ABSTRACT

Humanized mice are a powerful tool for the study of human hematopoiesis and immune function in vivo. However, the existing models cannot support robust adaptive immune responses, especially the generation of class-switched, antigen-specific antibody responses. Here we describe a new mouse strain, in which human interleukin 6 (IL-6) gene encoding the cytokine that is important for B- and T-cell differentiation was knocked into its respective mouse locus. The provision of human IL-6 not only enhanced thymopoiesis and periphery T-cell engraftment, but also significantly increased class switched memory B cells and serum immunoglobulin G (IgG). In addition, immunization with ovalbumin (OVA) induced OVA-specific B cells only in human IL-6 knock-in mice. These OVA-specific antibodies displayed the highest frequency of somatic mutation, further suggesting that human IL-6 is important for efficient B-cell activation and selection. We conclude that human IL-6 knock-in mice represent a novel and improved model for human adaptive immunity without relying on complex surgery to transplant human fetal thymus and liver. These mice can therefore be used to exploit or evaluate immunization regimes that would be unethical or untenable in humans.


Subject(s)
Adaptive Immunity , Antibody Formation , Gene Knock-In Techniques , Immunoglobulin Class Switching , Interleukin-6/genetics , Animals , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Chickens , Gene Expression , Gene Knock-In Techniques/methods , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/immunology , Humans , Immunization , Immunoglobulin G/immunology , Interleukin-6/immunology , Mice , Ovalbumin/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology
8.
Trends Immunol ; 35(3): 114-22, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24246474

ABSTRACT

Although the major targets of HIV infection are CD4⁺ T cells, dendritic cells (DCs) represent a crucial subset in HIV infection because they influence viral transmission and target cell infection and presentation of HIV antigens. DCs are potent antigen-presenting cells that can modulate antiviral immune responses. Through secretion of inflammatory cytokines and interferons (IFNs), DCs also alter T cell proliferation and differentiation, participating in the immune dysregulation characteristic of chronic HIV infection. Their wide distribution in close proximity with the mucosal epithelia makes them one of the first cell types to encounter HIV during sexual transmission. We discuss here the multiple roles that DCs play at different stages of HIV infection, emphasizing their relevance to HIV pathology and progression.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/virology , HIV Infections/immunology , Antigen Presentation/immunology , Disease Progression , Humans , Lymphocyte Activation/immunology , T-Lymphocytes/immunology , T-Lymphocytes/virology
9.
Cancer Immunol Immunother ; 62(4): 811-22, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23306863

ABSTRACT

PURPOSE: Dendritic cells (DCs) can induce strong tumor-specific T-cell immune responses. Constitutive upregulation of the mitogen-activated protein kinase (MAPK) pathway by a BRAF(V600) mutation, which is present in about 50 % of metastatic melanomas, may be linked to compromised function of DCs in the tumor microenvironment. Targeting both MEK and BRAF has shown efficacy in BRAF(V600) mutant melanoma. METHODS: We co-cultured monocyte-derived human DCs with melanoma cell lines pretreated with the MEK inhibitor U0126 or the BRAF inhibitor vemurafenib. Cytokine production (IL-12 and TNF-α) and surface marker expression (CD80, CD83, and CD86) in DCs matured with the Toll-like receptor 3/Melanoma Differentiation-Associated protein 5 agonist polyI:C was examined. Additionally, DC function, viability, and T-cell priming capacity were assessed upon direct exposure to U0126 and vemurafenib. RESULTS: Cytokine production and co-stimulation marker expression were suppressed in polyI:C-matured DCs exposed to melanoma cells in co-cultures. This suppression was reversed by MAPK blockade with U0126 and/or vemurafenib only in melanoma cell lines carrying a BRAF(V600E) mutation. Furthermore, when testing the effect of U0126 directly on DCs, marked inhibition of function, viability, and DC priming capacity was observed. In contrast, vemurafenib had no effect on DC function across a wide range of dose concentrations. CONCLUSIONS: BRAF(V600E) mutant melanoma cells modulate DC through the MAPK pathway as its blockade can reverse suppression of DC function. MEK inhibition negatively impacts DC function and viability if applied directly. In contrast, vemurafenib does not have detrimental effects on important functions of DCs and may therefore be a superior candidate for combination immunotherapy approaches in melanoma patients.


Subject(s)
Dendritic Cells/immunology , MAP Kinase Kinase Kinases/antagonists & inhibitors , Melanoma/enzymology , Melanoma/immunology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Antigens, CD/biosynthesis , Antigens, CD/immunology , B7-1 Antigen/biosynthesis , B7-1 Antigen/immunology , Butadienes/pharmacology , CD40 Antigens/biosynthesis , CD40 Antigens/immunology , Cell Line, Tumor , Coculture Techniques , Cytokines/biosynthesis , Cytokines/immunology , Humans , Immunoglobulins/biosynthesis , Immunoglobulins/immunology , Indoles/pharmacology , MAP Kinase Kinase Kinases/immunology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/immunology , Melanoma/genetics , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/immunology , Mutation , Nitriles/pharmacology , Poly I-C/immunology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/immunology , Sulfonamides/pharmacology , T-Lymphocytes/immunology , Vemurafenib , CD83 Antigen
10.
J Clin Invest ; 122(12): 4685-97, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23160198

ABSTRACT

Acute HIV-1 infection results in dysregulated immunity, which contributes to poor control of viral infection. DCs are key regulators of both adaptive and innate immune responses needed for controlling HIV-1, and we surmised that factors elicited during acute HIV-1 infection might impede DC function. We derived immature DCs from healthy donor peripheral blood monocytes and treated them with plasma from uninfected control donors and donors with acute HIV-1 infections. We found that the plasma from patients with HIV specifically inhibited DC function. This suppression was mediated by elevated apoptotic microparticles derived from dying cells during acute HIV-1 infection. Apoptotic microparticles bound to and inhibited DCs through the hyaluronate receptor CD44. These data suggest that targeting this CD44-mediated inhibition by apoptotic microparticles could be a novel strategy to potentiate DC activation of HIV-specific immunity.


Subject(s)
Apoptosis , Cell-Derived Microparticles/immunology , Dendritic Cells/immunology , HIV Infections/immunology , HIV-1/immunology , Hyaluronan Receptors/metabolism , Cell-Derived Microparticles/virology , Dendritic Cells/physiology , Dendritic Cells/virology , HIV Infections/blood , HIV-1/physiology , Humans , Hyaluronan Receptors/physiology , Immunity, Innate , Toll-Like Receptors/metabolism , Viremia/virology
11.
J Acquir Immune Defic Syndr ; 61(5): 535-44, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-22902724

ABSTRACT

OBJECTIVE: Myeloid dendritic cell (mDC) dysfunction during HIV infection may hinder the formation of both innate and adaptive immune responses and contribute to pathogenesis. Our objective was to determine whether circulating factors during chronic HIV infection impair mDC function with respect to secretion of IL-12, a pro-Th1 cytokine, and T-cell stimulatory capacity. Particular focus was placed on the effect of combination antiretroviral therapy (cART) and the role of HIV itself on mDC function. METHODS: Monocyte-derived DC (moDC) from uninfected donors were exposed to plasma from HIV-infected individuals before Toll-like receptor (TLR) stimulation. Cytokine secretion was measured via cytokine bead arrays, and T-cell proliferation and IFNγ secretion was evaluated after coculture with naive CD4 T cells. Expression of genes central to TLR-mediated signal transduction was analyzed via quantitative reverse transcriptase-polymerase chain reaction (qRT-PCR) arrays and western blot. RESULTS: Exposure of monocyte-derived DC to plasma from untreated HIV-infected donors suppressed secretion of IL-12, and impaired Th1-skewing of CD4 T cells. The suppressive effect was less by plasma donors receiving cART. Removal of virus from plasma did not relieve suppression nor was IL-12 secretion decreased on addition of HIV to control plasma. On a transcriptional level, decreased expression of IKKß, a key regulator in the TLR/NF-kappaB signaling pathway, corresponded to suppressed cytokine secretion. CONCLUSIONS: Plasma factors during chronic HIV infection impair mDC function in a manner that likely impacts the formation of immune responses to HIV, opportunistic pathogens, and vaccines. Despite partial alleviation by cART, this suppression was not directly mediated by HIV.


Subject(s)
Dendritic Cells/immunology , HIV Infections/blood , HIV Infections/immunology , HIV-1 , Interleukin-12/biosynthesis , Adult , CD4-Positive T-Lymphocytes/immunology , Coculture Techniques , Cross-Sectional Studies , Female , Gene Expression , HIV Infections/drug therapy , HIV Infections/genetics , HIV-1/immunology , Humans , I-kappa B Kinase/genetics , Immune Tolerance , Male , Middle Aged , Signal Transduction , Th1 Cells/immunology , Viral Load/immunology , Young Adult
12.
Nat Rev Immunol ; 11(3): 176-86, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21350578

ABSTRACT

Dendritic cells (DCs) and natural killer (NK) cells have central roles in antiviral immunity by shaping the quality of the adaptive immune response to viruses and by mediating direct antiviral activity. HIV-1 infection is characterized by a severe dysregulation of the antiviral immune response that starts during early infection. This Review describes recent insights into how HIV-1 infection affects DC and NK cell function, and the roles of these innate immune cells in HIV-1 pathogenesis. The importance of understanding DC and NK cell crosstalk during HIV infection for the development of effective antiviral strategies is also discussed.


Subject(s)
Dendritic Cells/immunology , HIV Infections/immunology , HIV-1/immunology , Killer Cells, Natural/immunology , AIDS Vaccines/immunology , Adaptive Immunity/immunology , Animals , Antigen Presentation , Antigens, CD/metabolism , Autophagy , Cell Adhesion Molecules/metabolism , Cell Communication , Dendritic Cells/virology , Female , HIV Antigens/immunology , HIV Envelope Protein gp120/metabolism , Haplorhini , Humans , Lectins, C-Type/metabolism , Male , Mannose-Binding Lectins/metabolism , Receptors, Cell Surface/metabolism , Receptors, HIV/immunology , Receptors, HIV/physiology , Receptors, KIR/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/immunology , Toll-Like Receptor 8/metabolism , Vaccination
13.
J Immunol ; 182(5): 2766-76, 2009 Mar 01.
Article in English | MEDLINE | ID: mdl-19234171

ABSTRACT

During viral infection, dendritic cells (DCs) capture infected cells and present viral Ags to CD8(+) T cells. However, activated DCs might potentially present cell-associated Ags derived from captured dead cells. In this study, we find that human DCs that captured dead cells containing the TLR3 agonist poly(I:C) produced cytokines and underwent maturation, but failed to elicit autologous CD8(+) T cell responses against Ags of dead cells. Accordingly, DCs that captured dead cells containing poly(I:C), or influenza virus, are unable to activate CD8(+) T cell clones specific to cell-associated Ags of captured dead cells. CD4(+) T cells are expanded with DCs that have captured poly(I:C)-containing dead cells, indicating the inhibition is specific for MHC class I-restricted cross-presentation. Furthermore, these DCs can expand naive allogeneic CD8(+) T cells. Finally, soluble or targeted Ag is presented when coloaded onto DCs that have captured poly(I:C)-containing dead cells, indicating the inhibition is specific for dead cell cargo that is accompanied by viral or poly(I:C) stimulus. Thus, DCs have a mechanism that prevents MHC class I-restricted cross-presentation of cell-associated Ag when they have captured dead infected cells.


Subject(s)
Cross-Priming/immunology , Dendritic Cells/virology , Growth Inhibitors/immunology , HLA-A2 Antigen/immunology , Immunosuppression Therapy , Influenza A virus/immunology , Melanoma/virology , Poly I-C/immunology , Antigens, Neoplasm/immunology , Antigens, Neoplasm/metabolism , Cell Line, Tumor , Cells, Cultured , Coculture Techniques , Dendritic Cells/immunology , Dendritic Cells/pathology , HLA-A2 Antigen/metabolism , Humans , Immunosuppression Therapy/methods , Lymphocyte Activation/immunology , Melanoma/immunology , Melanoma/pathology , Necrosis , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/pathology , T-Lymphocytes, Cytotoxic/virology
14.
Hematol Oncol Clin North Am ; 20(3): 689-710, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16762730

ABSTRACT

Vaccination against infectious agents represents a success of immunology, although many infectious diseases still evade the immune system, including chronic infections, such as tuberculosis, malaria, and HIV. Further progress is expected through rational design based on increased understanding of how the immune system works, and how the induction of protective immunity is regulated. The same principle applies to cancer vaccines, particularly because cancer is a chronic disease. Owing to their capacity to regulate cellular and humoral immunity, dendritic cells are increasingly used as vaccines; the immunogenicity of antigens delivered on dendritic cells has been shown in cancer patients. A better understanding of how dendritic cells regulate immune responses would allow clinicians to exploit them better to induce effective immunity against cancer.


Subject(s)
Cancer Vaccines , Dendritic Cells/transplantation , Immunotherapy, Adoptive , Antigen Presentation , Antigens, Neoplasm/therapeutic use , Dendritic Cells/immunology , Dendritic Cells/physiology , Humans
15.
J Leukoc Biol ; 74(6): 1064-73, 2003 Dec.
Article in English | MEDLINE | ID: mdl-12960264

ABSTRACT

Toll-like receptors (TLRs) recognize pathogen-associated molecular patterns, which are non-self macromolecular components of pathogens that allow the innate-immune system to recognize infection. TLRs are expressed on macrophages and dendritic cells (DC). TLR stimulation or CD40 agonists can induce inflammatory cytokine secretion from macrophages and DC, and promote DC maturation. The regulation of TLR expression by inflammation has begun to be explored. Our studies have focused on the regulation of TLR4 surface expression on DC. TLR4, along with the adaptor molecule MD2, is involved in the recognition of lipopolysaccharide (LPS). CD40 stimulation via cross-linked anti-CD40 monoclonal antibody (mAb) up-regulates TLR4-MD2 surface expression on a DC cell line (DC2.4) and on ex vivo-cultured splenic DC. LPS treatment down-regulated surface TLR4-MD2 on DC2.4 cells, but if combined with anti-CD40 mAb, increased TLR4-MD2 expression was observed. The increased TLR4-MD2 surface expression by any treatment did not correlate with TLR4 mRNA levels. The functional consequence of increased TLR4-MD2 expression following LPS and anti-CD40 treatment was examined. Although CD40 prestimulation did slightly enhance interleukin-12p70 secretion after LPS restimulation, simultaneous anti-CD40 mAb and LPS treatment, which up-regulates TLR4-MD2 complex, does not restore DC responsiveness to subsequent LPS.


Subject(s)
Antigens, Ly/metabolism , CD40 Antigens/metabolism , Dendritic Cells/metabolism , Membrane Glycoproteins/metabolism , Receptors, Cell Surface/metabolism , Adjuvants, Immunologic , Animals , Antigens, Ly/genetics , Enzyme-Linked Immunosorbent Assay , Female , Genes, RAG-1/physiology , Interleukin-12/metabolism , Lipopolysaccharides/metabolism , Lymphocyte Antigen 96 , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Receptors, Cell Surface/genetics , Reverse Transcriptase Polymerase Chain Reaction , Spleen/immunology , Spleen/metabolism , Toll-Like Receptor 4 , Toll-Like Receptors , Up-Regulation
16.
J Immunother ; 26(1): 72-84, 2003.
Article in English | MEDLINE | ID: mdl-12514431

ABSTRACT

Dendritic cells (DCs) can be matured by CD40 stimulation to upregulate their MHC class II/peptide complexes and costimulatory molecule surface expression to become adept at presenting antigen to and activating naive T lymphocytes. The use of anti-CD40 antibodies as adjuvants for DC-based therapy has been advanced. Little is known as to how DC biology in response to CD40 ligation differs between in vitro versus in vivo ligation. Therefore, the authors analyzed the expression kinetics of MHC class II (I-Ak)/HEL peptide "complex," total MHC class II, CD80, and CD86 on in vitro or in vivo CD40-stimulated DCs over a period of 5 days. MHC class II, "complex," and costimulatory molecule expression was elevated at 1 day in vitro and stayed high for the culture period, whereas in vivo expression of the cohort of molecules peaked earlier and then declined. When purified DCs were co-cultured in vitro with antigen-specific T cell hybridomas, the DCs had lower expression of total MHC class II and "complex," but did not reduce their CD80 and CD86 expression. The lower expression was dependent on cognate interaction as a non-antigen-specific T cell hybridoma was without effect. Blocking antigen-specific MHC class II/peptide-T cell receptor (TcR) complex interaction with antibody inhibited the reduction of MHC class II expression on CD40-stimulated DCs in vitro. Overall, their studies suggest distinct response of DCs to typical conditions that feature anti-CD40 monoclonal antibody (mAb)-activated DCs in vitro or in vivo.


Subject(s)
CD40 Antigens/immunology , Dendritic Cells/immunology , Genes, MHC Class II/genetics , Animals , Antibodies, Monoclonal/pharmacology , Antigen Presentation , Cells, Cultured , Flow Cytometry , Gene Expression , In Vitro Techniques , Lymphocyte Activation , Mice , Mice, Inbred C3H , Models, Animal , Probability , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...