Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Cancers (Basel) ; 13(24)2021 Dec 14.
Article in English | MEDLINE | ID: mdl-34944891

ABSTRACT

Cholangiocarcinoma (CCA) is a heterogeneous malignancy with a dismal prognosis. Therapeutic options are largely limited to surgery and conventional chemotherapy offers limited benefit. As immunotherapy has proven highly effective in various cancer types, we have undertaken a quantitative immunohistopathological assessment of immune cells expressing the immunoinhibitory T cell immune response cDNA 7 receptor (TIRC7), an emerging immunoinhibitory receptor, in a cohort of 135 CCA patients. TIRC7+ immune cells were present in both the tumor epithelia and stroma in the majority of CCA cases with the highest levels found in intrahepatic CCA. While intraepithelial density of TIRC7+ immune cells was decreased compared to matched non-neoplastic bile ducts, stromal quantity was higher in the tumor samples. Tumors exhibiting signet ring cell or adenosquamous morphology were exclusively associated with an intraepithelial TIRC7+ phenotype. Survival analysis showed intraepithelial TIRC7+ immune cell density to be a highly significant favorable prognosticator in intrahepatic but not proximal or distal CCA. Furthermore, intraepithelial TIRC7+ immune cell density correlated with the number of intraepithelial CD8+ immune cells and with the total number of CD4+ immune cells. Our results suggest the presence and prognostic relevance of TIRC7+ immune cells in CCA and warrant further functional studies on its pharmacological modulation.

2.
Front Immunol ; 12: 790775, 2021.
Article in English | MEDLINE | ID: mdl-35222353

ABSTRACT

A subset of T regulatory cells (Tregs), identified by TIRC7 (T cell immune response cDNA 7) expression is designated as Immune Regulatory 1 Cells (IR1 cells). TIRC7 is an immune checkpoint inhibitor, co-localized with the T- cell receptor, HLA-DR and CTLA-4 during T-cell activation, which delivers regulatory signals via binding to its ligand, HLA-DR α2 domain. IR1 cells express FOXP3, and multiple other markers associated with immune suppression. They constitute as much as 10% of Tregs. IR1 cells strongly inhibit proliferation in mixed lymphocyte reactions, where they express high levels of IL-10. Ex vivo expansion of Tregs over 2 weeks in the presence of an agonist TIRC7 antibody disproportionately expands the IR1 Treg subset, while maintaining high expression of suppressive markers including CD39, IL-10, LAP and GARP. Ex vivo expanded IR1 cells are a potent, homogeneous, stable set of suppressor Tregs with the potential to modulate immune dysregulation. The characteristics of IR1 cells suggest a therapeutic advantage over polyclonal Tregs for therapeutic interventions. Early restoration of immune homeostasis using IR1 cells has the potential to fundamentally alter the natural history of conditions characterized by abnormalities in the T regulatory cell compartment.


Subject(s)
Interleukin-10 , T-Lymphocytes, Regulatory , Forkhead Transcription Factors/metabolism , HLA-DR Antigens/metabolism , Humans , Lymphocyte Activation , Receptors, Antigen, T-Cell/metabolism
3.
Cancers (Basel) ; 12(11)2020 Oct 27.
Article in English | MEDLINE | ID: mdl-33121007

ABSTRACT

CAP7.1 is a novel topoisomerase II inhibitor, converted to active etoposide via carboxylesterase 2 (CES2), with signals of efficacy in treatment-refractory solid tumours. In a Phase II trial, 27 patients with advanced biliary tract cancers (BTC) were randomised 1:1 to CAP7.1 plus best supportive care (BSC), or BSC alone, with crossover to CAP7.1 upon disease progression. The primary objective was disease control rate (DCR) following 28-day cycles of CAP7.1 (200/150 mg/m2; iv), or BSC until progression. Secondary objectives included progression-free survival (PFS), time-to-treatment failure (TTF), overall survival (OS) and safety. Fourteen patients received CAP7.1 and 13 BSC. DCR favoured CAP7.1 vs. BSC (50% vs. 20%; treatment difference: 30%, 95%CI -18.44, 69.22, full analysis set [FAS]), with disease progression in 40% vs. 70%, respectively. Significantly longer median PFS was achieved for CAP7.1 vs. BSC: 66 vs. 39 days, respectively (hazard ratio [HR] 0.31; 95%CI 0.11, 0.86; p = 0.009; FAS). Similar trends were observed for TTF and OS. CES2-positive patients had longer median PFS (158 vs. 56 days) and OS (228 vs. 82 days) vs. CES2-negative patients. Adverse events were predictable, dose-dependent and consistent with those previously observed with etoposide. These efficacy and safety findings in second-line BTC warrant further clinical investigation of CAP7.1.

4.
J Gastrointest Oncol ; 11(4): 770-789, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32953160

ABSTRACT

Biliary tract carcinoma (BTC) has a poor prognosis and is increasing in incidence. Although surgery, chemotherapy and other treatment modalities have improved, surgery remains the only potential curative treatment and is appropriate for only those few patients who present with localized, resectable disease. However, for the majority of patients, unresectable disease is evident at diagnosis and about 95% of patients die within 10 years, despite the majority receiving chemotherapy. Long-term survival is significantly greater for patients with resected BTC compared to those with unresectable disease. In unresected disease, life expectancy is limited, with first-line gemcitabine/cisplatin (GEM/CIS) accepted as standard of care. Currently no standard second-line regimen which provides significant improvement of clinical outcomes exists for those who present with refractory disease or who relapse after first-line treatment. Of particular importance is establishing the impact of best supportive care (BSC) as a benchmark for survival outcomes to which the impact of treatment modalities can be compared. Survival outcome often differs significantly for patients with different prognostic factor profiles even when receiving the same therapy so that it can be difficult to predict which patient subgroup might benefit most from which therapy. Therefore, the influence of prognostic factors on survival under different therapies as well as under BSC needs to be further assessed in order to arrive at truly evidence-based, best therapeutic decisions for individual patients. Encouraging new research into the genomic landscape of BTC may help to further subdivide the BTC population into molecular-genetic clusters likely to be sensitive to different targeted therapy approaches leading to further improvements in survival. Consequently, an unmet need exists not only to develop new and more effective therapies for this devastating disease, but also to integrate original research findings into a more complex, dynamic, individualized therapeutic decision model to aid clinicians in making evidence-based, best therapeutic decisions for individual patients.

5.
Sci Rep ; 9(1): 4338, 2019 03 13.
Article in English | MEDLINE | ID: mdl-30867471

ABSTRACT

Carboxylesterase 2 (CES2) is instrumental for conversion of ester-containing prodrugs in cancer treatment. Novel treatment strategies are exceedingly needed for cholangiocarcinoma (CCA) patients. Here, we assessed CES2 expression by immunohistochemistry in a CCA cohort comprising 171 non-liver fluke associated, intrahepatic (n = 72) and extrahepatic (perihilar: n = 56; distal: n = 43) CCAs. Additionally, 80 samples of high-grade biliary intraepithelial neoplastic tissues and 158 corresponding samples of histological normal, non-neoplastic biliary tract tissues were included. CES2 expression was highest in non-neoplastic biliary tissue and significantly decreased in CCA. Patients showing any CES2 expression in tumor cells had a significantly better overall survival compared to negative cases (p = 0.008). This survival benefit was also maintained after stratification of CES2-positive cases, by comparing low, medium and high CES2 expression levels (p-trend = 0.0006). Evaluation of CCA subtypes showed the survival difference to be restricted to extrahepatic tumors. Correlation of CES2 expression with data of tumor-infiltrating immune cells showed that particularly CD8+ T cells were more frequently detected in CES2-positive CCAs. Furthermore, treatment of CCA cell lines with the prodrug Irinotecan reduced cell viability, increased cytotoxicity and modulated inflammatory gene expression. In conclusion, reduced CES2 expression is associated with poor outcome and low CD8+ T cell infiltration in CCA patients. Further clinical studies could show, whether CES2 expression may serve as a predictive marker in patients treated with prodrugs converted by CES2.


Subject(s)
Bile Duct Neoplasms/enzymology , Biomarkers, Tumor/metabolism , Carboxylesterase/metabolism , Cholangiocarcinoma/enzymology , Bile Duct Neoplasms/pathology , Cholangiocarcinoma/pathology , Cohort Studies , Female , Humans , Male , Middle Aged , Prognosis
6.
Cancer Lett ; 378(1): 51-8, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27149931

ABSTRACT

Carboxylesterase 2 (CES-2) is instrumental for conversion of ester-containing prodrugs in cancer treatment. CES-2 expression was analyzed by immunohistochemistry in colorectal cancer (CRC) compared to colonic inflammation as well as in liver and peripheral blood. In CRC, tumor grades showed no correlation with levels of CES-2 expression, which was heterogeneous within these tumors. Cellular infiltrates in the immediate tumor vicinity expressed high levels of CES-2. Thus, tissue adjacent to the tumor was a substantial source of CES-2 with high expression in plasma cells. CES-2(high) plasma cells were abundantly found in the colon of patients with inflammatory bowel disease. CES-2 expression is strong in hepatocytes of normal livers, while CES-2 expression in peripheral blood mononuclear cells of healthy donors was overall low at protein and mRNA levels. In summary, the conversion of ester-containing prodrugs by CES-2 is mainly to occur in the periphery, during liver passage and in the colon after enterohepatic recirculation. We here demonstrated plasma cells as strong producers of CES-2. Further studies should elucidate the role of CES-2(+) plasma cells in intestinal inflammation and cancer.


Subject(s)
Antineoplastic Agents/metabolism , Carboxylesterase/metabolism , Colorectal Neoplasms/enzymology , Gastrointestinal Agents/metabolism , Inflammatory Bowel Diseases/enzymology , Plasma Cells/enzymology , Prodrugs/metabolism , Activation, Metabolic , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/therapeutic use , Carboxylesterase/blood , Carboxylesterase/genetics , Colon/enzymology , Colon/pathology , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Enterohepatic Circulation , Female , Gastrointestinal Agents/pharmacology , Gene Expression Regulation, Enzymologic , HEK293 Cells , HT29 Cells , Hepatocytes/enzymology , Humans , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/pathology , Jurkat Cells , K562 Cells , Leukocytes, Mononuclear/enzymology , Male , Middle Aged , Neoplasm Grading , Prodrugs/therapeutic use , U937 Cells , Young Adult
7.
Future Oncol ; 8(1): 87-103, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22149037

ABSTRACT

Oncology therapeutics are less likely to reach the market than other therapeutics, at a higher cost, and only approximately one in ten cancer drugs in clinical development actually reach the market. To improve, there need to be new approaches to oncology research and development, based on understanding cancer biology and improving preclinical models and clinical trials, such as more use of biomarkers and evaluation of other targets including cancer stem cells and use of combination therapies. Biomarkers can be used to make early go/no-go decisions in drug development and can speed up drug development by selecting patients who will benefit and excluding patients likely to experience severe side effects, but they need validation before use. New approaches to preclinical and clinical trials can also speed up and improve the development of cancer therapeutics.


Subject(s)
Antineoplastic Agents/economics , Neoplasms/drug therapy , Neoplasms/economics , Animals , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/metabolism , Clinical Trials as Topic , Genomics , Humans , Molecular Targeted Therapy , Neoplasms/pathology , Neoplastic Stem Cells/pathology , Research/economics , Research Design , Treatment Outcome
8.
Eur J Immunol ; 41(8): 2323-32, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21538348

ABSTRACT

Adiponectin (APN), a cytokine constitutively produced in fat tissue, has been shown to exert anti-inflammatory effects in various disease models. While the influence of APN on monocytic cells has been extensively studied in vitro, little is known about its role in T cells. In this study, we show that while <10% of human peripheral blood T cells express adiponectin receptors (AdipoRs) on their surface, most T cells store AdipoRs in intracellular compartments. AdipoRs colocalized with immune regulatory molecules CTLA-4 and TIRC7 within clathrin-coated vesicles. After stimulation, the expression of adiponectin receptor 1 (AdipoR1) and AdipoR2 was upregulated on the surface of antigen-specific T cells, as determined by tetramer or CD137 staining, and AdipoR1 and AdipoR2 coexpressed with CTLA-4. Addition of APN resulted in a significant diminution of antigen-specific T-cell expansion. Mechanistically, APN enhanced apoptosis and inhibited proliferation of antigen-specific T-cell lines. Further, APN directly inhibited cytokine production in response to antigen stimulation. In line with the in vitro data, APN-deficient (knockout, KO) mice had higher frequencies of CD137(+) T cells upon Coxsackie B virus infection. Altogether, our data suggest that APN is a novel negative T-cell regulator. In contrast to the CTLA-4 ligand B7 only expressed on APCs, APN is abundant in human plasma.


Subject(s)
Adiponectin/immunology , Antigens/immunology , Lymphocyte Activation/immunology , T-Lymphocytes/immunology , Adiponectin/genetics , Adiponectin/pharmacology , Animals , Antigens, CD/immunology , Antigens, CD/metabolism , CTLA-4 Antigen , Cell Proliferation/drug effects , Cells, Cultured , Clathrin-Coated Vesicles/immunology , Clathrin-Coated Vesicles/metabolism , Coxsackievirus Infections/genetics , Coxsackievirus Infections/immunology , Coxsackievirus Infections/virology , Flow Cytometry , Gene Expression , Humans , Interferon-gamma/immunology , Interferon-gamma/metabolism , Jurkat Cells , K562 Cells , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Fluorescence , Receptors, Adiponectin/genetics , Receptors, Adiponectin/immunology , Receptors, Adiponectin/metabolism , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/metabolism , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism , Vacuolar Proton-Translocating ATPases/immunology , Vacuolar Proton-Translocating ATPases/metabolism
9.
Biotechnol Healthc ; 8(4): 25-7, 2011.
Article in English | MEDLINE | ID: mdl-22479231

ABSTRACT

Delivering drugs directly to tumors and overcoming drug resistance are two hurdles that face cancer researchers. Here's a look at where cancer treatment stands.

10.
PLoS One ; 3(2): e1576, 2008 Feb 13.
Article in English | MEDLINE | ID: mdl-18270567

ABSTRACT

Classically, HLA-DR expressed on antigen presenting cells (APC) initiates lymphocyte activation via presentation of peptides to TCR bearing CD4+ T-Cells. Here we demonstrate that HLA-DR alpha 2 domain (sHLA-DRalpha2) also induces negative signals by engaging TIRC7 on lymphocytes. This interaction inhibits proliferation and induces apoptosis in CD4+ and CD8+ T-cells via activation of the intrinsic pathway. Proliferation inhibition is associated with SHP-1 recruitment by TIRC7, decreased phosphorylation of STAT4, TCR-zeta chain & ZAP70, and inhibition of IFN-gamma and FasL expression. HLA-DRalpha2 and TIRC7 co-localize at the APC-T cell interaction site. Triggering HLA-DR - TIRC7 pathway demonstrates that sHLA-DRalpha2 treatment inhibits proinflammatory-inflammatory cytokine expression in APC & T cells after lipopolysaccaride (LPS) stimulation in vitro and induces apoptosis in vivo. These results suggest a novel antiproliferative role for HLA-DR mediated via TIRC7, revise the notion of an exclusive stimulatory interaction of HLA-DR with CD4+ T cells and highlights a novel physiologically relevant regulatory pathway.


Subject(s)
Apoptosis , HLA-DR Antigens/metabolism , Inflammation/immunology , Lymphocytes/immunology , Signal Transduction , Vacuolar Proton-Translocating ATPases/metabolism , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , Cell Proliferation , Cells, Cultured , Humans , Lymphocytes/cytology
11.
Curr Opin Investig Drugs ; 8(5): 401-10, 2007 May.
Article in English | MEDLINE | ID: mdl-17520869

ABSTRACT

The membrane protein T-cell immune response cDNA 7 (TIRC7) is transiently expressed in subsets of lymphocytes following antigen stimulation. The importance of TIRC7 in immune activation is demonstrated by the effect of antibodies directed against extracellular domains of TIRC7. In vitro targeting of TIRC7 inhibits proliferation and cytokine expression in human, mouse and rat lymphocytes, and these inhibitory effects have been associated with induction of cytotoxic T-lymphocyte antigen 4 mRNA and protein in the presence of TIRC7 antibodies. In vivo, anti-TIRC7 antibodies prevent kidney transplant rejection in rats and heart allograft rejection in mice. Treatment with an anti-TIRC7 antibody as monotherapy or in combination with TNFalpha blockade inhibits disease progression in collagen-induced arthritis. TIRC7 expression decreases in the peripheral blood of humans who have undergone cardiac transplant prior to clinical rejection, and is therefore a promising noninvasive tool for the prediction of rejection. Thus, targeting of TIRC7 may lead to the development of specific and effective therapeutic and diagnostic approaches by unifying relevant cellular and molecular responses in T- and B-cell subsets, and represents a promising new pathway for immune regulation in transplantation and autoimmune disease.


Subject(s)
Graft Rejection/metabolism , Inflammation/metabolism , Lymphocyte Activation , Lymphocytes/metabolism , Vacuolar Proton-Translocating ATPases/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/metabolism , Biomarkers/metabolism , Graft Rejection/immunology , Graft Rejection/prevention & control , Humans , Immunologic Factors/pharmacology , Immunologic Factors/therapeutic use , Inflammation/drug therapy , Inflammation/immunology , Lymphocyte Activation/drug effects , Lymphocytes/drug effects , Lymphocytes/immunology , Signal Transduction , Transplantation, Homologous , Vacuolar Proton-Translocating ATPases/antagonists & inhibitors , Vacuolar Proton-Translocating ATPases/immunology
12.
Transpl Immunol ; 16(3-4): 238-44, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17138060

ABSTRACT

TIRC7 delivers essential signals during immune activation as antibodies targeting TIRC7 inhibit lymphocyte proliferation and Th1 cytokine expression in vitro and prolonged kidney and heart allograft survival in vivo. Immunohistochemical analysis of biopsy specimens from human renal allografts undergoing rejection despite treatment with Calcineurin inhibitors (CI) showed elevated TIRC7 expression. Accordingly, with a view to clinical application, we evaluated the therapeutic effect of a chimerized anti-TIRC7 mAb in combination with Tacrolimus (FK506) using a rat kidney transplantation model (DA to Lewis). The combination of sub-therapeutic doses of both compounds significantly (p<0.05) prolonged the median graft survival to 19.5 days compared to monotherapy with FK506 (median survival, 7d) or mAb against TIRC7 (7d). These results suggest a potential synergism of anti-TIRC7 mAb and FK506 action, which could be developed into a novel combination therapy in the clinic by lowering side effects of present CI treatment. Moreover, the identification of TIRC7 in graft infiltrating lymphocytes might serve as a diagnostic marker to detect allograft rejection.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Graft Rejection/prevention & control , Immunosuppressive Agents/therapeutic use , Kidney Transplantation/immunology , Tacrolimus/therapeutic use , Vacuolar Proton-Translocating ATPases/immunology , Animals , Blotting, Western , Graft Rejection/immunology , Graft Survival/drug effects , Graft Survival/immunology , Humans , Immunohistochemistry , Kidney/immunology , Kidney/metabolism , Kidney/pathology , Kidney Transplantation/adverse effects , Male , Microscopy, Confocal , Rats , Rats, Inbred Lew , Transplantation, Homologous , Vacuolar Proton-Translocating ATPases/antagonists & inhibitors , Vacuolar Proton-Translocating ATPases/drug effects
13.
J Immunol ; 177(10): 6833-41, 2006 Nov 15.
Article in English | MEDLINE | ID: mdl-17082597

ABSTRACT

Ab targeting of TIRC7 has been shown previously to inhibit T cell proliferation and Th1 lymphocyte-associated cytokine production. In this study, we demonstrate that Ab targeting of TIRC7 induces early cell surface expression of CTLA-4. The majority of stimulated CD4+ and CD8+ human T cells coexpress CTLA-4 and TIRC7. Similar to CTLA-4, TIRC7 rapidly accumulates at the site of Ag adhesion upon T cell activation. TIRC7 seems to colocalize with CTLA-4 in human T cells, and both molecules are associated with clathrin-coated vesicles, indicating they share intracellular transport systems. Moreover, Ab targeting of TIRC7 results in an early activation of CTLA-4 transcription. The inhibition of cell proliferation mediated by TIRC7 is dependent on CTLA-4 expression because the TIRC7-mediated inhibitory effects on cell proliferation and cytokine expression are abolished by Ab blockade of CTLA-4. Splenocytes obtained from CTLA-4-deficient mice are not responsive to TIRC7 Ab targeting. Thus, TIRC7 acts as an upstream regulatory molecule of CTLA-4 expression.


Subject(s)
Antigens, CD/biosynthesis , Antigens, Differentiation/biosynthesis , Cell Proliferation , Growth Inhibitors/physiology , Immunosuppressive Agents , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Vacuolar Proton-Translocating ATPases/physiology , Antibodies, Blocking/pharmacology , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Antigens, CD/immunology , Antigens, CD/physiology , Antigens, Differentiation/immunology , Antigens, Differentiation/physiology , Binding Sites/immunology , CTLA-4 Antigen , Cell Membrane/immunology , Cell Membrane/metabolism , Cells, Cultured , Clathrin-Coated Vesicles/immunology , Clathrin-Coated Vesicles/metabolism , Cytokines/antagonists & inhibitors , Cytokines/biosynthesis , Humans , Immune Sera/pharmacology , Immunosuppressive Agents/pharmacology , Intracellular Fluid/immunology , Intracellular Fluid/metabolism , Lymphocyte Activation/immunology , Membrane Proteins/immunology , Membrane Proteins/metabolism , Protein Transport/immunology , T-Lymphocytes/immunology , Up-Regulation/immunology , Vacuolar Proton-Translocating ATPases/immunology , Vacuolar Proton-Translocating ATPases/metabolism
14.
Drug News Perspect ; 18(2): 103-8, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15883619

ABSTRACT

A number of leukocyte surface molecules play an essential role during immune activation. Targeting of these molecules utilizing antibodies serves as a specific therapeutic approach for the treatment of a variety of human diseases. Antibodies targeting a number of leukocyte surface molecules were shown to induce tolerance to transplants in several animal models. A novel membrane molecule, T-cell immune response cDNA 7 (TIRC7), has been shown to be an essential protein in the regulation of lymphocyte activation. TIRC7 does not share any homology with other known membrane proteins expressed during the course of lymphocyte activation and does not belong to any of the known costimulatory, cytokine, chemokine or receptor families. TIRC7, a highly conserved protein across species, is expressed in immune tissues such as spleen, lymph nodes, and T and B lymphocytes. Antibodies against extracellular domains of TIRC7 prolong allograft survival in rat and mouse transplantation models. The prevention of rejection is mediated at least partially via induction of cytotoxic T lymphocyte antigen 4 (CTLA4) in T cells. Functional cellular assays utilizing TIRC7-deficient mice splenocytes show that TIRC7 does have an impact not only on T-cell, but also on B-cell response. Subtherapeutic amounts of FK506 and anti-TIRC7 monoclonal antibody prolong graft survival, suggesting synergistic effects with calcineurin inhibitors. Targeting TIRC7 with monoclonal antibody might serve as a promising therapeutic strategy for preventing allograft rejection in humans and treatment of other immune-related diseases. Acutely rejected human kidney allografts show strong expression of TIRC7 despite treatment with calcineurin inhibitors. Therefore, monitoring TIRC7 expression may facilitate an early diagnostic tool of acute rejection. TIRC7 seems to belong to a group of targets with dual roles in disease pathogenesis, so-called theranostics, which can be utilized to treat and diagnose diseases.


Subject(s)
Antibodies/therapeutic use , Graft Rejection/prevention & control , Immunosuppression Therapy , Protein Subunits , Vacuolar Proton-Translocating ATPases , Animals , Graft Rejection/immunology , Graft Rejection/pathology , Humans , Protein Subunits/immunology , Protein Subunits/metabolism , Protein Subunits/physiology , Vacuolar Proton-Translocating ATPases/immunology , Vacuolar Proton-Translocating ATPases/metabolism , Vacuolar Proton-Translocating ATPases/physiology
15.
J Immunol ; 173(4): 2342-52, 2004 Aug 15.
Article in English | MEDLINE | ID: mdl-15294947

ABSTRACT

The membrane protein T cell immune response cDNA 7 (TIRC7) was recently identified and was shown to play an important role in T cell activation. To characterize the function of TIRC7 in more detail, we generated TIRC7-deficient mice by gene targeting. We observed disturbed T and B cell function both in vitro and in vivo in TIRC7(-/-) mice. Histologically, primary and secondary lymphoid organs showed a mixture of hypo-, hyper-, and dysplastic changes of multiple lymphohemopoietic compartments. T cells from TIRC7(-/-) mice exhibited significantly increased proliferation and expression of IL-2, IFN-gamma, and IL-4 in response to different stimuli. Resting T cells from TIRC7(-/-) mice exhibited decreased CD62L, but increased CD11a and CD44 expression, suggesting an in vivo expansion of memory/effector T cells. Remarkably, activated T cells from TIRC7(-/-) mice expressed lower levels of CTLA-4 in comparison with wild-type cells. B cells from TIRC7-deficient mice exhibited significantly higher in vitro proliferation following stimulation with anti-CD40 Ab or LPS plus IL-4. B cell hyperreactivity was reflected in vivo by elevated serum levels of various Ig classes and higher CD86 expression on B cells. Furthermore, TIRC7 deficiency resulted in an augmented delayed-type hypersensitivity response that was also reflected in increased mononuclear infiltration in the skin obtained from TIRC7-deficient mice food pads. In summary, the data strongly support an important role for TIRC7 in regulating both T and B cell responses.


Subject(s)
B-Lymphocytes/immunology , Cytokines/immunology , Lymphocyte Activation/immunology , Protein Subunits/immunology , T-Lymphocytes/immunology , Vacuolar Proton-Translocating ATPases/immunology , Animals , Antibody Formation/genetics , B-Lymphocytes/enzymology , Cells, Cultured , Flow Cytometry , Gene Targeting , Hypersensitivity, Delayed/immunology , Immunohistochemistry , Mice , Protein Subunits/deficiency , Spleen/immunology , Spleen/pathology , T-Lymphocytes/enzymology , Vacuolar Proton-Translocating ATPases/deficiency
16.
J Immunol ; 172(6): 3535-43, 2004 Mar 15.
Article in English | MEDLINE | ID: mdl-15004154

ABSTRACT

Carcinoembryonic Ag-related cellular adhesion molecule 1 (CEACAM1) represents a group of transmembrane protein isoforms that consist of variable numbers of extracellular Ig-like domains together with either a long cytoplasmic (cyt) tail containing two immunoreceptor tyrosine-based inhibitory motifs or a unique short cyt tail. Although CEACAM1 has been reported to be expressed on the surface of T lymphocytes upon activation, its roles in T cell regulation are controversial due to the lack of functional characterization of each individual CEACAM1 isoform. We thus cotransfected Jurkat T cells with CEACAM1 isoform-encoding constructs and an IL-2 promoter-bearing plasmid or a small interference RNA targeting src homology domain 2 containing phosphatase 1. In a luciferase reporter assay and through measurements of cytokine secretion (IL-2, IL-4, and IFN-gamma), CEACAM1 containing either a long or a short cyt tail inhibited or costimulated, respectively, TCR/CD3 complex plus CD28 mediated activation with the inhibitory functions of the long cyt tail dominating. The inhibitory function of CEACAM1, was dependent upon src homology domain 2 containing phosphatase 1 activity, required both tyrosine residues within the immunoreceptor tyrosine-based inhibitory motif domains of the cyt tail and was mediated through the mitogen-activated protein kinase pathway. CEACAM1-mediated inhibition could be functionally reconstituted by incubation of PBMC with either a CEACAM1-specific mAb or CEACAM1-Fc fusion protein in the presence of an allogeneic or mitogenic stimulus, respectively. These studies indicate that the long and short cyt tails of CEACAM1 serve as inhibitory and costimulatory receptors, respectively, in T cell regulation.


Subject(s)
Antigens, CD/physiology , Antigens, Differentiation/physiology , Antigens, Neoplasm/physiology , Cell Adhesion Molecules/physiology , Down-Regulation/immunology , Immunosuppressive Agents/pharmacology , Lymphocyte Activation/immunology , Nuclear Proteins , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Up-Regulation/immunology , Antibodies, Monoclonal/pharmacology , CD28 Antigens/immunology , CD3 Complex/immunology , Cells, Cultured , Cytoplasm/immunology , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Genes, Reporter , Humans , Intracellular Signaling Peptides and Proteins , JNK Mitogen-Activated Protein Kinases , Jurkat Cells , Ligands , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , NFATC Transcription Factors , Phosphorylation , Protein Isoforms/physiology , Protein Phosphatase 1 , Protein Structure, Tertiary , Protein Tyrosine Phosphatase, Non-Receptor Type 6 , Protein Tyrosine Phosphatases/physiology , Receptors, Immunologic/physiology , T-Lymphocytes/enzymology , Transcription Factor AP-1/antagonists & inhibitors , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Transcription Factors/metabolism , Transfection
17.
Am J Transplant ; 4(4): 505-14, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15023142

ABSTRACT

T cell immune response c-DNA (TIRC7) is up-regulated during the early stages of T-cell activation in response to alloantigens. In this study, we analyzed the effects of newly developed monoclonal antibodies (mAb) against TIRC7 in acute cardiac allograft rejection. Fully vascularized heterotopic allogeneic heart transplantation was performed in mice across a full-mismatch barrier (C57Bl/10 into CBA). Recipients received seven injections (day 0-7) of a novel anti-TIRC7 mAb or remained untreated. Graft survival, histology and ex vivo lymphocyte functions were tested. Targeting of TIRC7 with an anti-TIRC7 mAb diminishes lymphocyte infiltration into grafts resulting in delay of morphological graft damage and prolongation of allograft survival. The lymphocytes from anti-TIRC7 mAb-treated animals exhibit hypo-responsiveness without evidence of lymphocyte depletion against the donor allo-antigens. Proliferation and expression of interferon-gamma (IFN-gamma) and tumor necrosis factor-alpha (TNF-alpha) were down-regulated while interleukin-4 (IL-4) and IL-10 expression were spared. Moreover, anti-TIRC7 mAb enhanced up-regulation of CTLA-4 expression but suppressed up-regulation of CD25 on stimulated lymphocytes in vitro and in vivo. Ligation of TIRC7 has important effects on the regulation of co-stimulatory signaling pathways associated with suppressing of T-cell activation. Targeting of TIRC7 may therefore provide a novel therapeutic approach for modulating T cell immune responses during organ transplantation.


Subject(s)
Antibodies, Monoclonal/chemistry , Graft Rejection/prevention & control , Heart Transplantation/methods , Protein Subunits/immunology , Vacuolar Proton-Translocating ATPases/immunology , Animals , Antibodies, Monoclonal/metabolism , CD4-Positive T-Lymphocytes/metabolism , Cell Division , Cell Membrane/metabolism , DNA, Complementary/metabolism , Down-Regulation , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Graft Survival , Immunohistochemistry , Interferon-gamma/metabolism , Interleukin-10/metabolism , Interleukin-4/metabolism , Lymphocytes/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Myocardium/metabolism , Receptors, Interleukin-2/biosynthesis , Spleen/metabolism , Time Factors , Tumor Necrosis Factor-alpha/metabolism
18.
J Exp Med ; 199(4): 471-82, 2004 Feb 16.
Article in English | MEDLINE | ID: mdl-14970176

ABSTRACT

Carcinoembryonic antigen-related cellular adhesion molecule 1 (CEACAM1) is a cell surface molecule that has been proposed to negatively regulate T cell function. We have shown that CEACAM1 is associated with specific regulation of T helper cell (Th)1 pathways, T-bet-mediated Th1 cytokine signaling, and Th1-mediated immunopathology in vivo. Mice treated with anti-mouse CEACAM1-specific monoclonal antibody (mAb) CC1 during the effector phase exhibited a reduced severity of trinitrobenzene sulfonic acid colitis in association with decreased interferon (IFN)-gamma production. Although oxazolone colitis has been reported as Th2 mediated, mice treated with the CC1 mAb or a CEACAM1-Fc chimeric protein exhibited a reduced severity of colitis in association with a significant reduction of IFN-gamma and T-bet activation, whereas signal transducer and activator of antigen 4 activation was unaffected. Both interleukin-4 and IFN-gamma gene-deficient mice exhibited less severe colitis induction by oxazolone. Direct ligation of T cells in vitro with the murine hepatitis virus spike protein, a natural ligand for the N-domain of CEACAM1, inhibited the differentiation of naive cells into Th1 but not Th2 cells and activation of Th1 but not Th2 cytokine production. These results indicate that CEACAM1 isoforms are a novel class of activation-induced cell surface molecules on T cells that function in the specific regulation of Th1-mediated inflammation such as that associated with inflammatory bowel disease.


Subject(s)
Carcinoembryonic Antigen/immunology , Colitis/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Antibodies, Monoclonal/immunology , Colitis/chemically induced , Colitis/pathology , Disease Models, Animal , Female , Immunoglobulin Fc Fragments/immunology , Inflammation/immunology , Inflammation/pathology , Interferon-gamma/deficiency , Interferon-gamma/genetics , Interleukin-1/deficiency , Interleukin-1/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Oxazolone , Recombinant Fusion Proteins/immunology , Th1 Cells/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...