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










Publication year range
1.
J Immunol ; 194(5): 2289-99, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25624454

ABSTRACT

Despite the emergence of the programmed cell death 1 (PD-1):PD-1 ligand (PD-L) regulatory axis as a promising target for treating multiple human diseases, remarkably little is known about how this pathway regulates responses to extracellular bacterial infections. We found that PD-1(-/-) mice, as well as wild-type mice treated with a PD-1 blocking Ab, exhibited significantly increased survival against lethal Streptococcus pneumoniae infection following either priming with low-dose pneumococcal respiratory infection or S. pneumoniae-capsular polysaccharide immunization. Enhanced survival in mice with disrupted PD-1:PD-L interactions was explained by significantly increased proliferation, isotype switching, and IgG production by pneumococcal capsule-specific B cells. Both PD-L, B7-H1 and B7-DC, contributed to PD-1-mediated suppression of protective capsule-specific IgG. Importantly, PD-1 was induced on capsule-specific B cells and suppressed IgG production and protection against pneumococcal infection in a B cell-intrinsic manner. To our knowledge, these results provide the first demonstration of a physiologic role for B cell-intrinsic PD-1 expression in vivo. In summary, our study reveals that B cell-expressed PD-1 plays a central role in regulating protection against S. pneumoniae, and thereby represents a promising target for bolstering immunity to encapsulated bacteria.


Subject(s)
Antibodies, Bacterial/biosynthesis , B-Lymphocytes/immunology , B7-H1 Antigen/immunology , Pneumococcal Infections/prevention & control , Pneumococcal Vaccines/administration & dosage , Programmed Cell Death 1 Receptor/immunology , Animals , B-Lymphocytes/microbiology , B7-H1 Antigen/genetics , Gene Expression Regulation , Immunity, Humoral/drug effects , Immunization , Immunoglobulin G/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Knockout , Pneumococcal Infections/genetics , Pneumococcal Infections/immunology , Pneumococcal Infections/mortality , Polysaccharides, Bacterial/administration & dosage , Programmed Cell Death 1 Ligand 2 Protein/genetics , Programmed Cell Death 1 Ligand 2 Protein/immunology , Programmed Cell Death 1 Receptor/deficiency , Programmed Cell Death 1 Receptor/genetics , Signal Transduction , Streptococcus pneumoniae/immunology , Survival Analysis
2.
Cancer Res ; 72(8): 2089-99, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22496463

ABSTRACT

Although cancer tends to affect the elderly, most preclinical studies are carried out in young subjects. In this study, we developed a melanoma-specific cancer immunotherapy that shows efficacy in aged but not young hosts by mitigating age-specific tumor-associated immune dysfunction. Both young and aged CD4(+)CD25(hi) regulatory T cells (Treg) exhibited equivalent in vitro T-cell suppression and tumor-associated augmentation in numbers. However, denileukin diftitox (DT)-mediated Treg depletion improved tumor-specific immunity and was clinically effective only in young mice. DT-mediated Treg depletion significantly increased myeloid-derived suppressor cell (MDSC) numbers in aged but not young mice, and MDSC depletion improved tumor-specific immunity and reduced tumor growth in aged mice. Combining Treg depletion with anti-Gr-1 antibody was immunologically and clinically more efficacious than anti-Gr-1 antibody alone in aged B16-bearing mice, similar to Treg depletion alone in young mice. In contrast, DT increased MDSCs in young and aged mice following MC-38 tumor challenge, although effects were greater in aged mice. Anti-Gr-1 boosted DT effects in young but not aged mice. Aged antitumor immune effector cells are therefore competent to combat tumor when underlying tumor-associated immune dysfunction is appropriately mitigated, but this dysfunction varies with tumor, thus also varying responses to immunotherapy. By tailoring immunotherapy to account for age-related tumor-associated immune dysfunctions, cancer immunotherapy for aged patients with specific tumors can be remarkably improved.


Subject(s)
Aging/immunology , Immunotherapy/methods , Neoplasms, Experimental/immunology , Neoplasms, Experimental/therapy , Receptors, Cell Surface/antagonists & inhibitors , Animals , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Diphtheria Toxin/therapeutic use , Disease Models, Animal , Flow Cytometry , Interleukin-2/therapeutic use , Lymphocyte Depletion/methods , Mice , Mice, Inbred C57BL , Recombinant Fusion Proteins/therapeutic use , T-Lymphocytes, Regulatory/immunology
3.
Aging Cell ; 11(3): 509-19, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22372596

ABSTRACT

Regulatory T cells (Tregs) are specialized CD4(+) T lymphocytes helping defend against autoimmunity and inflammation. Although age is associated with increased inflammation and autoimmunity, few reports address age effects of immune regulation or auto-aggressive T cells. We show here that young and aged naïve CD4(+) T cells are equivalently auto-aggressive in vivo in T cell-driven autoimmune colitis. Young and aged CD4(+) Tregs equally suppressed age-matched T cell proliferation in vitro and controlled clinical and pathologic T cell-driven autoimmune colitis, suggesting equivalent regulatory function. However, whereas young and aged CD4(+) Tregs suppressed interferon (IFN)-γ(+) T cells equivalently in this model, aged CD4(+) Tregs unexpectedly failed to restrain interleukin (IL)-17(+) T cells. Nonetheless, young and aged CD4(+) Tregs equally restrained IL-17(+) T cells in vivo during acute inflammation, suggesting a chronic inflammation-related defect in aged CD4(+) Tregs. In support, aged Tregs expressed reduced STAT3 activation, a defect associated with poor IL-17-producing T cell restraint. Aged naïve mice had markedly increased programmed death (PD)-1(+) T cells, but these exhibited no significant auto-aggressive or regulatory functions in T cell-driven colitis. Young CD8(+) CD122(-) T cells induce autoimmune bone marrow failure, but we show that aged CD8(+) CD122(-) T cells do not. These data demonstrate no apparent age-related increase in auto-aggressive T cell behavior, but disclose previously unrecognized functional defects in aged CD4(+) Tregs during chronic inflammation. IL-17 can be inflammatory and contributes to certain autoimmune disorders. Reduced aged Treg function during chronic inflammation and reduced IL-17 restraint could contribute to age-related inflammation or autoimmunity.


Subject(s)
Autoimmune Diseases/immunology , Inflammation/immunology , Interleukin-17/biosynthesis , STAT3 Transcription Factor/immunology , T-Lymphocytes, Regulatory/immunology , Age Factors , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/pathology , Cell Growth Processes/physiology , Cellular Senescence/physiology , Disease Models, Animal , Inflammation/pathology , Interleukin-17/immunology , Mice , Mice, Inbred C57BL , STAT3 Transcription Factor/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/metabolism
4.
Aging Cell ; 11(1): 128-38, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22044484

ABSTRACT

Because most patients with cancer are aged and because immunological functions are altered during aging, it is important to account for aging-associated immunological alterations in the design of new cancer immunotherapies. We thus compared immune populations in young and aged mice and found that B7-DC(+) (PD-L2/CD273) B cells, a minor population in young mice, were significantly increased in aged mice. Induction of both Th1 and Th17 cells was significantly augmented by B7-DC(+) B cells from aged mice, and this effect was blocked with anti-B7-DC antibodies in vitro and in vivo. Moreover, retardation of tumor growth in aged mice was largely B7-DC dependent. Tumor growth in young mice was significantly inhibited by immunization with B7-DC(+) B cells from aged mice owing to increased induction of tumor antigen-specific cytotoxic T lymphocytes. These data indicate that B7-DC(+) B cells could play an important role in aging-associated cancer immunopathology as well as in other aging-associated diseases and further suggest that B7-DC(+) B cells have potential for future cancer immunotherapy.


Subject(s)
Aging/immunology , B-Lymphocytes/immunology , Melanoma, Experimental/immunology , Programmed Cell Death 1 Ligand 2 Protein/immunology , Spleen/immunology , Th1 Cells/immunology , Th17 Cells/immunology , Animals , Antibodies, Neutralizing/administration & dosage , Antigens, Neoplasm/immunology , B-Lymphocytes/cytology , Cell Proliferation , Cytokines/biosynthesis , Cytokines/immunology , Flow Cytometry , Humans , Melanoma, Experimental/pathology , Melanoma, Experimental/therapy , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Programmed Cell Death 1 Ligand 2 Protein/biosynthesis , Spleen/pathology , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/immunology , Th1 Cells/cytology , Th17 Cells/cytology , Xenograft Model Antitumor Assays
5.
J Clin Invest ; 121(6): 2371-82, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21537079

ABSTRACT

CD73 is overexpressed in many types of human and mouse cancers and is implicated in the control of tumor progression. However, the specific contribution from tumor or host CD73 expression to tumor growth remains unknown to date. Here, we show that host CD73 promotes tumor growth in a T cell-dependent manner and that the optimal antitumor effect of CD73 blockade requires inhibiting both tumor and host CD73. Notably, enzymatic activity of CD73 on nonhematopoietic cells limited tumor-infiltrating T cells by controlling antitumor T cell homing to tumors in multiple mouse tumor models. In contrast, CD73 on hematopoietic cells (including CD4⁺CD25⁺ Tregs) inhibited systemic antitumor T cell expansion and effector functions. Thus, CD73 on hematopoietic and nonhematopoietic cells has distinct adenosinergic effects in regulating systemic and local antitumor T cell responses. Importantly, pharmacological blockade of CD73 using its selective inhibitor or an anti-CD73 mAb inhibited tumor growth and completely restored efficacy of adoptive T cell therapy in mice. These findings suggest that both tumor and host CD73 cooperatively protect tumors from incoming antitumor T cells and show the potential of targeting CD73 as a cancer immunotherapy strategy.


Subject(s)
5'-Nucleotidase/physiology , Lymphoma, Non-Hodgkin/pathology , Melanoma, Experimental/pathology , Ovarian Neoplasms/pathology , T-Lymphocytes/immunology , 5'-Nucleotidase/antagonists & inhibitors , 5'-Nucleotidase/deficiency , 5'-Nucleotidase/immunology , Adenosine/physiology , Adenosine Diphosphate/analogs & derivatives , Adenosine Diphosphate/pharmacology , Adenosine Diphosphate/therapeutic use , Animals , Chemotaxis, Leukocyte , Disease Progression , Female , Immunotherapy, Adoptive , Lymphocytes, Tumor-Infiltrating/immunology , Lymphoma, Non-Hodgkin/immunology , Lymphoma, Non-Hodgkin/therapy , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Molecular Targeted Therapy , Ovarian Neoplasms/immunology , Ovarian Neoplasms/therapy , Peritoneal Neoplasms/immunology , Peritoneal Neoplasms/secondary , Peritoneal Neoplasms/therapy , Radiation Chimera , Second Messenger Systems/physiology , T-Lymphocytes, Regulatory/immunology , Tumor Burden
6.
J Immunol ; 185(5): 2747-53, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20686128

ABSTRACT

CD4(+)CD25(+)Foxp3(+) regulatory T cells (Tregs) are immunopathogenic in cancers by impeding tumor-specific immunity. B7-homologue 1 (B7-H1) (CD274) is a cosignaling molecule with pleiotropic effects, including hindering antitumor immunity. In this study, we demonstrate sex-dependent, B7-H1-dependent differences in tumor immunity and response to immunotherapy in a hormone-independent cancer, murine B16 melanoma. Antitumor immunity was better in B7-H1(-/-) females versus males as a result of reduced regulatory T cell function in the B7-H1(-/-) females, and clinical response following B7-H1 blockade as tumor immunotherapy was significantly better in wild-type females than in males, owing to greater B7-H1 blockade-mediated reduction of Treg function in females. Wild-type female Tregs expressed significantly lower B7-H1 versus males but were insensitive to estrogen in vitro. Female B7-H1(-/-) Tregs were exquisitely sensitive to estrogen-mediated functional reduction in vitro, suggesting that B7-H1 effects occur before terminal Treg differentiation. Immune differences were independent of known B7-H1 ligands. Sex-dependent immune differences are seldom considered in designing immune therapy or interpreting immunotherapy treatment results. Our data demonstrate that sex is an important variable in tumor immunopathogenesis and immunotherapy responses through differential Treg function and B7-H1 signaling.


Subject(s)
B7-1 Antigen/physiology , Immunotherapy, Adoptive/methods , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Membrane Glycoproteins/physiology , Peptides/physiology , Sex Characteristics , Animals , Antigens, Differentiation/physiology , B7-1 Antigen/genetics , B7-H1 Antigen , Cell Line, Tumor , Female , Immunity, Innate/genetics , Male , Melanoma, Experimental/physiopathology , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Ovalbumin/biosynthesis , Ovalbumin/genetics , Ovalbumin/immunology , Peptides/deficiency , Peptides/genetics , Programmed Cell Death 1 Receptor , Signal Transduction/genetics , Signal Transduction/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/pathology
7.
Blood ; 116(13): 2266-76, 2010 Sep 30.
Article in English | MEDLINE | ID: mdl-20574047

ABSTRACT

Oral tolerance is a key feature of intestinal immunity, generating systemic tolerance to fed antigens. However, the molecular mechanism mediating oral tolerance remains unclear. In this study, we examined the role of the B7 family members of costimulatory molecules in the establishment of oral tolerance. Deficiencies of B7-H1 and B7-DC abrogated the oral tolerance, accompanied by enhanced antigen-specific CD4(+) T-cell response and IgG(1) production. Mesenteric lymph node (MLN) dendritic cells (DCs) displayed higher levels of B7-H1 and B7-DC than systemic DCs, whereas they showed similar levels of CD80, CD86, and B7-H2. MLN DCs enhanced the antigen-specific generation of CD4(+)Foxp3(+) inducible regulatory T cells (iT(regs)) from CD4(+)Foxp3(-) T cells rather than CD4(+) effector T cells (T(eff)) relative to systemic DCs, owing to the dominant expression of B7-H1 and B7-DC. Furthermore, the antigen-specific conversion of CD4(+)Foxp3(-) T cells into CD4(+)Foxp3(+) iT(regs) occurred in MLNs greater than in peripheral organs during oral tolerance under steady-state conditions, and such conversion required B7-H1 and B7-DC more than other B7 family members, whereas it was severely impaired under inflammatory conditions. In conclusion, our findings suggest that B7-H1 and B7-DC expressed on MLN DCs are essential for establishing oral tolerance through the de novo generation of antigen-specific CD4(+)Foxp3(+) iT(regs).


Subject(s)
B7-1 Antigen/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Immune Tolerance , Membrane Glycoproteins/metabolism , Peptides/metabolism , T-Lymphocytes, Regulatory/immunology , Administration, Oral , Animals , Antigen Presentation , Antigens/administration & dosage , B7-1 Antigen/genetics , B7-H1 Antigen , Forkhead Transcription Factors/metabolism , Immunoglobulin G/biosynthesis , In Vitro Techniques , Lymph Nodes/cytology , Lymph Nodes/immunology , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Mesentery/cytology , Mesentery/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Ovalbumin/administration & dosage , Ovalbumin/immunology , Peptides/deficiency , Peptides/genetics , Programmed Cell Death 1 Ligand 2 Protein , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , T-Lymphocytes, Regulatory/metabolism
9.
J Immunol ; 184(11): 6151-60, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20427766

ABSTRACT

Both innate and adaptive immune systems are considered important for cancer prevention, immunosurveillance, and control of cancer progression. It is known that, although both systems initially eliminate emerging tumor cells efficiently, tumors eventually escape immune attack by a variety of mechanisms, including differentiation and recruitment of immunosuppressive CD11b(+)Gr-1(+) myeloid suppressor cells into the tumor microenvironment. However, we show that CD11b(+)Gr-1(+) cells found in ascites of epithelial ovarian cancer-bearing mice at advanced stages of disease are immunostimulatory rather than being immunosuppressive. These cells consist of a homogenous population of cells that morphologically resemble neutrophils. Moreover, like dendritic cells, immunostimulatory CD11b(+)Gr-1(+) cells can strongly cross-prime, augmenting the proliferation of functional CTLs via signaling through the expression of costimulatory molecule CD80. Adoptive transfer of these immunostimulatory CD11b(+)Gr-1(+) cells from ascites of ovarian cancer-bearing mice results in the significant regression of s.c. tumors even without being pulsed with exogenous tumor Ag prior to adoptive transfer. We now show for the first time that adaptive immune responses against cancer can be augmented by these cancer-induced granulocyte-like immunostimulatory myeloid (CD11b(+)Gr-1(+)) cells, thereby mediating highly effective antitumor immunity in an adoptive transfer model of immunity.


Subject(s)
CD11b Antigen/immunology , Cross-Priming/immunology , Myeloid Cells/immunology , Ovarian Neoplasms/immunology , Receptors, Chemokine/immunology , Adoptive Transfer , Animals , Antigens/immunology , Cell Separation , Female , Flow Cytometry , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , T-Lymphocytes/immunology
10.
Cancer Res ; 70(6): 2245-55, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-20179192

ABSTRACT

CD73, originally defined as a lymphocyte differentiation antigen, is thought to function as a cosignaling molecule on T lymphocytes and an adhesion molecule that is required for lymphocyte binding to endothelium. We show here that CD73 is widely expressed on many tumor cell lines and is upregulated in cancerous tissues. Because the ecto-5'-nucleotidase activity of CD73 catalyzes AMP breakdown to immunosuppressive adenosine, we hypothesized that CD73-generated adenosine prevents tumor destruction by inhibiting antitumor immunity. We confirmed this hypothesis by showing that combining tumor CD73 knockdown and tumor-specific T-cell transfer cured all tumor-bearing mice. In striking contrast, there was no therapeutic benefit of adoptive T-cell immunotherapy in mice bearing tumors without CD73 knockdown. Moreover, blockade of the A2A adenosine receptor with a selective antagonist also augmented the efficacy of adoptive T-cell therapy. These findings identify a potential mechanism for CD73-mediated tumor immune evasion and point to a novel cancer immunotherapy strategy by targeting the enzymatic activity of tumor CD73.


Subject(s)
5'-Nucleotidase/immunology , Ovarian Neoplasms/immunology , T-Lymphocytes/immunology , 5'-Nucleotidase/biosynthesis , 5'-Nucleotidase/genetics , Adenosine/antagonists & inhibitors , Adenosine/immunology , Adenosine A2 Receptor Antagonists , Animals , Apoptosis/immunology , Female , Flow Cytometry , Immunotherapy, Adoptive/methods , Mice , Mice, Inbred C57BL , Mice, Knockout , Ovarian Neoplasms/pathology , Ovarian Neoplasms/therapy , Pyrimidines/pharmacology , RNA, Small Interfering/genetics , Receptor, Adenosine A2A/immunology , Triazoles/pharmacology
11.
J Immunol ; 184(4): 2086-94, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-20065112

ABSTRACT

The caliber and magnitude of T cell responses are regulated by costimulatory molecules following the engagement of TCRs and MHC molecules. B7-DC has the highest homology with B7-H1 in the B7 family, and both of them bind an immunoregulatory molecule, programmed death 1. Previous studies have demonstrated that B7-DC stimulates T cell proliferation and CTL generation, which sharply contrasts the inhibitory role of B7-H1. Th2 cytokines prompt B7-DC expression, which in turn enhances Th1 responses. In this study, we used an intestinal nematode, Nippostrongylus brasiliensis, to induce strong Th2 responses and to evaluate B7-DC function under Th2-polarizing conditions in vivo. By either blocking B7-DC expression during N. brasiliensis infection or by examining N. brasiliensis-infected B7-DC knockout mice, we observed enhanced eosinophilia, the overproduction of serum IgE, and increased Th2 cytokine production along with decreased Th1 cytokine production (particularly IFN-gamma production), indicating that B7-DC inhibits Th2 responses. Our results further demonstrate that the inhibition of Th2 responses by B7-DC occurs independently of programmed death 1 but conceivably acts through an as yet unknown alternative receptor that enhances Th1 responses. Although the deficiency of B7-DC expression that enhanced the production of IL-13 paradoxically resulted in better protection against N. brasiliensis infection, our results show that B7-DC plays an important role in bolstering a robust Th1 response that is required for effective antiviral and anticancer immunity, even under a strong Th2-polarizing environment induced by N. brasiliensis infection.


Subject(s)
B7-1 Antigen/physiology , Nippostrongylus/immunology , Strongylida Infections/immunology , Th2 Cells/immunology , Animals , B7-1 Antigen/genetics , Cells, Cultured , Eosinophilia/immunology , Eosinophilia/parasitology , Feedback, Physiological , Immunoglobulin E/biosynthesis , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Transgenic , Programmed Cell Death 1 Ligand 2 Protein , Th2 Cells/metabolism , Th2 Cells/parasitology
12.
Eur J Immunol ; 38(10): 2706-17, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18825752

ABSTRACT

Disease progression in experimental autoimmune encephalomyelitis (EAE) is regulated by programmed death receptor 1 (PD-1) and its ligands, B7-H1 (programmed death ligand 1 (PD-L1)) and B7-DC (PD-L2). B7-H1 and B7-DC have negative regulatory effects upon binding PD-1 on activated T cells and B7-H1 deficiency increases severity of both diabetes and EAE. However, the role of PD-L expression on different APC in the CNS in regulating local T-cell function during relapsing EAE has not been examined. Our data show that the majority of CNS CD4+ T cells isolated during acute EAE are PD-1+, and T cells specific for relapse-associated epitopes express PD-1 upon antigen stimulation in the CNS. B7-H1 and B7-DC are differentially expressed on discrete APC populations in the inflamed CNS. B7-H1 and PD-1 have mainly inhibitory functions on CNS T cells. B7-H1 negatively regulates the stimulation of activated PD-1+ T(H) cells, in co-cultures with microglia and different CNS-infiltrating APC presenting endogenously processed peptides. The preponderance of IFN-gamma+ versus IL-17+ T cells in the CNS of B7-H1(-/-) mice suggests that B7-H1 more selectively suppresses T(H)-1 than T(H)-17 responses in vivo. In contrast, blockade of B7-DC has less pronounced regulatory effects. Overall, the results demonstrate that B7-H1 expressed by CNS myeloid APC negatively regulates T-cell activation during acute relapsing EAE.


Subject(s)
Antigen-Presenting Cells/immunology , Antigens, Differentiation/immunology , B7-1 Antigen/immunology , Central Nervous System/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Membrane Glycoproteins/immunology , Peptides/immunology , T-Lymphocyte Subsets/immunology , Animals , Antigen-Presenting Cells/metabolism , Antigens, Differentiation/metabolism , B7-1 Antigen/metabolism , B7-H1 Antigen , Central Nervous System/metabolism , Female , Gene Knockdown Techniques , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-17/immunology , Interleukin-17/metabolism , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Peptides/metabolism , Programmed Cell Death 1 Ligand 2 Protein , Programmed Cell Death 1 Receptor , T-Lymphocyte Subsets/metabolism
13.
Circulation ; 117(5): 660-9, 2008 Feb 05.
Article in English | MEDLINE | ID: mdl-18212277

ABSTRACT

BACKGROUND: Allograft vasculopathy is a major limiting factor in the long-term success of cardiac transplantation. T cells play a critical role in initiation of cardiac allograft rejection and allograft vasculopathy. The negative T-cell costimulatory pathway PD-1:PDL1/PDL2 (programmed death-1:programmed death ligand-1/2) plays an important role in regulating alloimmune responses. We investigated the role of recipient versus donor PD-1 ligands in the pathogenesis of allograft rejection with emphasis on the role of tissue expression in regulating this alloimmune response in vivo. METHODS AND RESULTS: We used established major histocompatibility complex class II- and class I-mismatched models of vascularized cardiac allograft rejection, blocking anti-PDL1 and anti-PDL2 antibodies, and PDL1- and PDL2-deficient mice (as donors or recipients) to study the role of the PD-1:PDL1/PDL2 pathway in chronic rejection. We also used PDL1-deficient and wild-type mice and bone marrow transplantation to generate chimeric animals that express PDL1 exclusively on either hematopoietic or parenchymal cells. PDL1 but not PDL2 blockade significantly accelerated cardiac allograft rejection in the bm12-into-B6 and B6-into-bm12 models. Although wild-type cardiac allografts survived long term, PDL1-/- donor hearts transplanted into wild-type bm12 mice exhibited accelerated rejection and vasculopathy associated with enhanced recipient T-cell alloreactivity. Interestingly, PDL1-/- recipients did not exhibit an accelerated tempo of cardiac allograft rejection. Using chimeric animals as donors, we show that PDL1 expression on cardiac tissue alone significantly prolonged graft survival compared with full PDL1-/- donor grafts in transplanted wild-type recipients. CONCLUSIONS: This is the first report to demonstrate that expression of the negative costimulatory molecule PDL1 on donor cardiac tissue regulates recipient alloimmune responses, allograft rejection, and vasculopathy.


Subject(s)
Antigens, Surface/genetics , Apoptosis Regulatory Proteins/genetics , Gene Expression Regulation , Graft Rejection/genetics , Heart Transplantation/pathology , Animals , Apoptosis Regulatory Proteins/deficiency , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Heart Transplantation/immunology , Histocompatibility Antigens Class II/immunology , Histocompatibility Testing , Mice , Mice, Inbred C57BL , Mice, Knockout , Programmed Cell Death 1 Receptor , Tissue Donors , Transplantation, Homologous
14.
Biochem Biophys Res Commun ; 365(1): 170-5, 2008 Jan 04.
Article in English | MEDLINE | ID: mdl-17981145

ABSTRACT

B7-DC is a costimulatory molecule belonging to the B7 family. We previously found that treatment with anti-B7-DC mAb during the effector phase enhances asthma phenotypes in mice. We investigated the mechanisms of B7-DC induction and how B7-DC regulates asthma phenotypes. In allergen-challenged IFN-gamma-deficient mice, anti-B7-DC mAb failed to enhance the asthma phenotypes although the induction of B7-DC on dendritic cells of the mice was comparable with that on dendritic cells of wild-type mice. B7-DC on dendritic cells was up-regulated by IL-13 in vitro. The induction of B7-DC on dendritic cells after allergen challenge was attenuated by blockade of IL-13 in vivo. The asthma phenotypes were enhanced in B7-DC-deficient mice, more than in wild-type mice. The enhancement was concurrent with the down-regulation of IFN-gamma and up-regulation of IL-13. These results suggest that B7-DC induced by IL-13 works as a feedback regulator by up-regulating IFN-gamma production during the effector phase of allergic asthma.


Subject(s)
Asthma/immunology , B7-1 Antigen/immunology , Hypersensitivity/immunology , Interleukin-13/metabolism , Animals , Asthma/metabolism , B7-1 Antigen/metabolism , Dendritic Cells/metabolism , Down-Regulation , Feedback, Physiological , Interferon-gamma/genetics , Interferon-gamma/immunology , Mice , Mice, Inbred BALB C , Mice, Transgenic , Phenotype , Programmed Cell Death 1 Ligand 2 Protein , Up-Regulation
15.
J Immunol ; 179(8): 5204-10, 2007 Oct 15.
Article in English | MEDLINE | ID: mdl-17911605

ABSTRACT

The PD-1:PDL pathway plays an important role in regulating alloimmune responses but its role in transplantation tolerance is unknown. We investigated the role of PD-1:PDL costimulatory pathway in peripheral and a well established model of central transplantation tolerance. Early as well as delayed blockade of PDL1 but not PDL2 abrogated tolerance induced by CTLA4Ig in a fully MHC-mismatched cardiac allograft model. Accelerated rejection was associated with a significant increase in the frequency of IFN-gamma-producing alloreactive T cells and expansion of effector CD8(+) T cells in the periphery, and a decline in the percentage of Foxp3(+) graft infiltrating cells. Similarly, studies using PDL1/L2-deficient recipients confirmed the results with Ab blockade. Interestingly, while PDL1-deficient donor allografts were accepted by wild-type recipients treated with CTLA4Ig, the grafts developed severe chronic rejection and vasculopathy when compared with wild-type grafts. Finally, in a model of central tolerance induced by mixed allogeneic chimerism, engraftment was not abrogated by PDL1/L2 blockade. These novel data demonstrate the critical role of PDL1 for induction and maintenance of peripheral transplantation tolerance by its ability to alter the balance between pathogenic and regulatory T cells. Expression of PDL1 in donor tissue is critical for prevention of in situ graft pathology and chronic rejection.


Subject(s)
B7-1 Antigen/physiology , Graft Rejection/immunology , Membrane Glycoproteins/physiology , Peptides/physiology , Transplantation Tolerance/immunology , Abatacept , Animals , B7-1 Antigen/genetics , B7-H1 Antigen , Chronic Disease , Graft Rejection/pathology , Graft Rejection/prevention & control , H-2 Antigens/genetics , H-2 Antigens/immunology , Heart Transplantation/immunology , Heart Transplantation/pathology , Immunoconjugates/therapeutic use , Lymphocyte Culture Test, Mixed , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Peptides/antagonists & inhibitors , Peptides/deficiency , Peptides/genetics , Programmed Cell Death 1 Ligand 2 Protein , Radiation Chimera , Skin Transplantation/immunology , Transplantation Tolerance/genetics
16.
Clin Cancer Res ; 13(15 Pt 1): 4538-46, 2007 Aug 01.
Article in English | MEDLINE | ID: mdl-17671140

ABSTRACT

PURPOSE: High-dose interleukin 2 (IL-2) is a Food and Drug Administration-approved regimen for patients with metastatic renal cell carcinoma. However, the toxicity and limited clinical benefit associated with IL-2 has hampered its use. Histone deacetylase (HDAC) inhibitors have been shown to have antitumor activity in different tumor models including renal cell carcinoma, and to have immunomodulatory properties. In our study, we tested the effectiveness of combination therapy of IL-2 with the HDAC inhibitor MS-275 in a murine renal cell carcinoma (RENCA) model. EXPERIMENTAL DESIGN: RENCA luciferase-expressing cells were implanted in the left kidney of BALB/C mice. Animals were randomly divided into four groups and treated with either vehicle, 150,000 IU of IL-2 twice daily by i.p. injections (twice weekly), 5 mg/kg of MS-275 daily by oral gavage (5 d/wk), or its combination. Treatment was started either 3 or 9 days following tumor cell injection. RESULTS: Weekly luciferase images and tumor weight after 2 weeks of treatment showed significant tumor inhibition (>80%) in the combination treatment as compared with the IL-2 (no significant inhibition) or MS-275 (approximately 40% inhibition) treatment groups. Spontaneous lung metastases were also inhibited in the combination treatment (>90% inhibition) as compared with the single treatment group. Kaplan-Meier analyses showed statistically significant increased survival in the combination group as compared with controls and single agents. Splenocytes from mice treated with combination treatment showed greater lysis of RENCA cells than splenocytes from mice treated with single agents. The percentage of CD4(+)CD25(+) T cells and Foxp3(+) T cells (T regulatory cells) was increased or reduced, respectively, in lymph nodes from tumor-bearing animals treated with the combination of MS-275 and IL-2 as compared with control and single agents. Depletion of CD8(+) T cells abrogated the survival benefit from MS-275 + IL-2 combination. CONCLUSIONS: These results show that the combination of IL-2 and MS-275 has a synergistic antitumor effect in vivo in an immunocompetent murine model of renal cell carcinoma. The antitumor effect was associated with the decreased number of T regulatory cells and the increased antitumor cytotoxicity by splenocytes. In conclusion, these preclinical data provide the rationale for clinical testing of the combination of IL-2 and HDAC inhibitors in the treatment of patients with renal cell carcinoma.


Subject(s)
Antineoplastic Agents/therapeutic use , Benzamides/therapeutic use , Carcinoma, Renal Cell/drug therapy , Histone Deacetylase Inhibitors , Interleukin-2/therapeutic use , Kidney Neoplasms/drug therapy , Pyridines/therapeutic use , Animals , Cell Proliferation/drug effects , Drug Synergism , Drug Therapy, Combination , Female , Flow Cytometry , Mice , Mice, Inbred BALB C , Mice, Nude , Survival Rate
17.
Blood ; 110(1): 180-5, 2007 Jul 01.
Article in English | MEDLINE | ID: mdl-17289811

ABSTRACT

Although self-reactive T-cell precursors can be eliminated upon recognition of self-antigen presented in the thymus, this central tolerance process is often incomplete, and additional mechanisms are required to prevent autoimmunity. Recent studies indicates that the interaction between B7-H1 and its receptor PD-1 on activated T cells plays an important role in the inhibition of T-cell responses in peripheral organs. Here, we show that, before their exit to the periphery, T cells in lymphoid organs rapidly up-regulate PD-1 upon tolerogen recognition. Ablation of the B7-H1 and PD-1 interaction when T cells are still in lymphoid organs prevents anergy. Furthermore, blockade of B7-H1 and PD-1 interaction could render anergic T cells responsive to antigen. Our results thus reveal previously unappreciated roles of B7-H1 and PD-1 interaction in the control of initiation and reversion of T-cell anergy.


Subject(s)
Antigens, Differentiation/immunology , B7-1 Antigen/immunology , Clonal Anergy/immunology , Membrane Glycoproteins/immunology , Peptides/immunology , Self Tolerance/immunology , T-Lymphocytes/immunology , Animals , Antigens, Differentiation/genetics , Antigens, Differentiation/metabolism , B7-1 Antigen/metabolism , B7-H1 Antigen , Lymph Nodes/cytology , Lymphocyte Activation/immunology , Membrane Glycoproteins/metabolism , Mice , Peptides/metabolism , Programmed Cell Death 1 Receptor , Up-Regulation/genetics
18.
J Exp Med ; 201(10): 1531-41, 2005 May 16.
Article in English | MEDLINE | ID: mdl-15897272

ABSTRACT

B7-DC, one of the recently described B7 family members, has the capacity to inhibit T cell responses via engagement of the immunoreceptor tyrosine-based inhibitory motif-containing inhibitory PD-1 receptor as well as enhance responses via an as yet unidentified costimulatory receptor. B7-DC is highly homologous to a coinhibitory B7 family member, B7-H1, which also binds PD-1. It is currently unclear which B7-DC function-costimulation or inhibition-predominates in vivo. To study in vivo functions of B7-DC, we evaluated immune responses in B7-DC knockout (KO) mice. Although not eliminated, interferon-gamma (IFN-gamma) production by CD4 T cells and IFN-gamma-dependent humoral responses were reduced in B7-DC KO mice relative to wild type mice. Antigen-specific CD8 T cell responses and cytotoxic T lymphocyte (CTL) activity were also diminished in B7-DC KO mice. Hepatic tumors grew more quickly in B7-DC KO mice, associated with a decrease in intrahepatic tumor-specific CD8 T cells. These results highlight the contrasting in vivo roles of B7-DC and B7-H1 and indicate that B7-DC functions as a tuning molecule, selectively augmenting T helper 1 and CTL responses.


Subject(s)
B7-1 Antigen/immunology , Cell Communication/immunology , Lymphocyte Activation/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Cytotoxic/immunology , Th1 Cells/immunology , Animals , Antigens, Differentiation/immunology , B7-1 Antigen/genetics , Cell Communication/genetics , Interferon-gamma/biosynthesis , Liver Neoplasms, Experimental/genetics , Liver Neoplasms, Experimental/immunology , Liver Neoplasms, Experimental/pathology , Lymphocyte Activation/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Programmed Cell Death 1 Ligand 2 Protein , Programmed Cell Death 1 Receptor
19.
Blood ; 104(7): 2187-93, 2004 Oct 01.
Article in English | MEDLINE | ID: mdl-15172973

ABSTRACT

Graft-versus-host disease (GVHD) is characterized by an impairment of mechanisms that underlie the development of immunologic tolerance. Although the cytokine storm associated with GVHD leads to expression of cell surface markers on both effector and regulatory T cells, regulatory CD4+ T cells that play an instrumental role in the maintenance of tolerance appear to uniquely express the Foxp3 transcriptional repressor. Foxp3 mRNA expression was significantly decreased in peripheral blood mononuclear cells from patients with either allogeneic GVHD or autologous GVHD compared with patients without GVHD. Expression of Foxp3 negatively correlated with the severity of GVHD but positively correlated with recent thymic emigrants. The results suggest that defective thymic function contributes to the impaired reconstitution of immune regulatory mechanisms following transplantation. The decrease in regulatory mechanisms after transplantation appears to provide an environment permissive to the development of GVHD.


Subject(s)
DNA-Binding Proteins/genetics , Gene Expression Regulation , Graft vs Host Disease/genetics , Adolescent , Adult , Aged , Bone Marrow Cells/metabolism , Bone Marrow Transplantation , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Membrane/metabolism , Female , Forkhead Transcription Factors , Humans , Leukocytes, Mononuclear/metabolism , Male , Middle Aged , RNA, Messenger/metabolism , Receptors, Interleukin-2/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Stem Cell Transplantation , T-Lymphocytes/metabolism , Thymidine/chemistry , Thymus Gland/metabolism , Time Factors , Transcription, Genetic
20.
Mol Immunol ; 40(1): 35-47, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12909129

ABSTRACT

Professional antigen presenting cells such as dendritic cells (DC) and macrophages (Mphi) share similar characteristics; however, they differ in their ability to initiate an immune response. DCs are much more potent in priming and stimulating nai;ve T-cells. Thus, DCs are good targets for the expression of foreign genes to elicit and specifically modify immune responses. To identify DC markers cDNA subtraction was performed using murine MHC class II(high), B7(high) bone marrow derived DCs as tester and interferon-gamma/E. coli lipopolysaccaride (LPS) treated bone marrow derived macrophages as driver. Analysis of 114 resulting clones revealed a diverse pattern of DC selective (DC(DeltaMphi)) gene expression including known genes whose expression in DCs had not been previously demonstrated as well as multiple novel genes. For several identified DC(DeltaMphi) genes, proximal promoter elements were isolated and incorporated into self-inactivating lentiviral GFP reporter vectors. Promoter activity was measured in bone marrow derived macrophages or dendritic cells. Of the promoters analyzed those for B7-DC and CCL17 drove strong GFP expression in DCs but not in resting or activated macrophages. The CCL17 promoter offered the highest level of expression in DCs and was further activated by culture with LPS or interleukin-4 (IL-4). In contrast, the B7-DC promoter was induced by IL-4 but not by LPS. Endogenous CCL17 and B7-DC mRNAs were increased similarly in IL-4 cultured DCs but only CCL17 was induced by LPS. Additionally, IL-4 increased cell surface expression of B7-DC in both immature and mature DCs.


Subject(s)
B7-1 Antigen/genetics , Chemokines, CC/genetics , Dendritic Cells/metabolism , Gene Transfer Techniques , Lentivirus/genetics , Animals , Base Sequence , Chemokine CCL17 , Female , Gene Expression Profiling , Interleukin-4/pharmacology , Lipopolysaccharides/pharmacology , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Programmed Cell Death 1 Ligand 2 Protein , Promoter Regions, Genetic , Tumor Necrosis Factor-alpha/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...