Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Cell Immunol ; 300: 54-62, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26711331

ABSTRACT

Macrophages play an important role in immune responses including allograft rejection and they are one of the potential targets of anti-rejection therapies in organ transplantation. Macrophage alloreactivity relies on their phenotype/polarity, motility, phagocytosis and matrix degradation, which in turn depend on proper functioning of actin cytoskeleton and its regulators, the small GTPase RhoA and its downstream effector the Rho-associated protein kinase (ROCK). Several laboratories showed that administration of ROCK inhibitor Y-27632 to the graft recipient inhibits chronic rejection or rodent cardiac allografts. Here we studied the effect of Y-27632 on mouse peritoneal macrophage structure, polarity and functions in in vitro assays. We show that Y-27632 inhibitor affects macrophage phenotype/polarity, phagocytosis, migration, and matrix degradation. These novel findings suggest that the impediment of macrophage structure and function via interference with the RhoA/ROCK pathway has a potential to be therapeutically effective in organ transplantation.


Subject(s)
Cell Polarity/physiology , Graft Rejection/immunology , Macrophage Activation/immunology , Macrophages, Peritoneal/cytology , Macrophages, Peritoneal/immunology , rho-Associated Kinases/antagonists & inhibitors , Allografts , Amides/pharmacology , Animals , Cell Polarity/drug effects , Enzyme Inhibitors/pharmacology , Heart Transplantation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Microscopy, Electron , Models, Animal , Pyridines/pharmacology
2.
Endocrinology ; 156(9): 3137-46, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26151356

ABSTRACT

The osteoblast-specific hormone osteocalcin (OC) was found to regulate glucose metabolism, fat mass, and ß-cell proliferation in mice. Here, we investigate the effect of decarboxylated OC (D-OC) on human ß-cell function and mass in culture and in vivo using a Nonobese diabetic-severe combined immunodeficiency mouse model. We found that D-OC at dose ranges from 1.0 to 15 ng/mL significantly augmented insulin content and enhanced human ß-cell proliferation of cultured human islets. This was paralleled by increased expression of sulfonylurea receptor protein; a marker of ß-cell differentiation and a component of the insulin-secretory apparatus. Moreover, in a Nonobese diabetic-severe combined immunodeficiency mouse model, systemic administration of D-OC at 4.5-ng/h significantly augmented production of human insulin and C-peptide from the grafted human islets. Finally, histological staining of the human islet grafts showed that the improvement in the ß-cell function was attributable to an increase in ß-cell mass as a result of ß-cell proliferation indicated by MKI67 staining together with the increased ß-cell number and decreased α-cell number data obtained using laser scanning cytometry. Our data for the first time show D-OC-enhanced ß-cell function in human islets and support future exploitation of D-OC-mediated ß-cell regulation for developing useful clinical treatments for patients with diabetes.


Subject(s)
Insulin-Secreting Cells/drug effects , Insulin/biosynthesis , Osteocalcin/pharmacology , Adult , Animals , C-Peptide/metabolism , Cattle , Cell Proliferation , Cells, Cultured , Female , Humans , Insulin/metabolism , Insulin Secretion , Islets of Langerhans Transplantation , Male , Mice, Inbred NOD , Mice, SCID , Middle Aged , Osteocalcin/therapeutic use
3.
Transplantation ; 96(9): 782-90, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-23985719

ABSTRACT

BACKGROUND: Current immunosuppressive regimens fail to avert chronic rejection (CR) of transplanted organs; however, selective targeting of actin-cytoskeletal regulators decreases T-cell motility and abrogates CR in rat model system. Administration of mutated class I major histocompatibility complex molecules or selective targeting of the RhoA pathway, which controls T-cell cytoskeletal activity, using Y27632 (a selective Rock1 inhibitor) resulted in reduced T-cell infiltration and abrogation of CR as judged from the neointimal index (13.9±19.7 vs. 45±37.5; P<0.001) and the number of affected vessels (30% vs. 60%; P<0.01). Here, we examined the role of mammalian target of rapamycin (mTOR) pathway in inhibition of CR. METHODS: A mutated class I major histocompatibility complex molecule that eliminates CR was delivered into ACI recipients of Wistar-Furth hearts at the time of transplantation with subtherapeutic cyclosporine (10 mg/kg on days 0-2). Controls included untreated and cyclosporine A-treated (10 mg/kg on days 0-2) heart allograft recipients. RESULTS: Western blotting and immunostaining showed that rat heart allografts with abolished CR exhibited down-regulation of the RAPA-sensitive mTORC1 components such as mTOR and Raptor and down-regulation of the RAPA-insensitive mTORC2 elements Rictor and Sin1. The mTOR regulator Deptor and its downstream target Rac1 were also inhibited. CONCLUSIONS: Abrogation of CR in rat model system involves modulation of two mTOR pathways: a RAPA-sensitive mTORC1 pathway regulating cellular proliferation and a RAPA-insensitive mTORC2 pathway regulating T-cell motility. Selective targeting of T-cell actin cytoskeletal pathways shows potential for pathway-targeted immunosuppression therapies.


Subject(s)
Graft Rejection/prevention & control , Heart Transplantation/adverse effects , Histocompatibility Antigens/pharmacology , Immunosuppressive Agents/pharmacology , Multiprotein Complexes/metabolism , TOR Serine-Threonine Kinases/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Carrier Proteins/metabolism , Chronic Disease , Cyclosporine/pharmacology , Graft Rejection/immunology , Graft Rejection/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Male , Mechanistic Target of Rapamycin Complex 1 , Mechanistic Target of Rapamycin Complex 2 , Phosphoproteins/metabolism , Rapamycin-Insensitive Companion of mTOR Protein , Rats , Rats, Inbred ACI , Rats, Wistar , Regulatory-Associated Protein of mTOR , Signal Transduction/drug effects , Sirolimus/pharmacology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Time Factors , rac1 GTP-Binding Protein/metabolism
4.
Folia Histochem Cytobiol ; 50(3): 358-67, 2012 Oct 08.
Article in English | MEDLINE | ID: mdl-23042265

ABSTRACT

The translationally controlled tumor protein (TCTP) plays a role in cell growth, cell cycle and cancer progression. TCTP controls negatively the stability of the p53 tumor suppressor protein and interacts with the cellular cytoskeleton. The deregulation of the actin and cytokeratin cytoskeleton is responsible for the increased migratory activity of tumor cells and is linked with poor patient outcome. Recent studies indicate that cyclin A,a key regulator of cell cycle, controls actin organization and negatively regulates cell motility via regulation of RhoA expression. We studied the organization of actin and cytokeratin cytoskeleton and the expression of TCTP, p53,cyclin A, RhoA and actin in HIO180 non-transformed ovarian epithelial cells, and OVCAR3 and SKOV3 (expressing low level of inducible p53) ovarian epithelial cancer cells with different metastatic potential. Immunostaining and ultrastructural analyses illustrated a dramatic difference in the organization of the cytokeratin and actin filaments in non-transformed versus cancer cell lines. We also determined that there is an inverse relationship between the level of TCTP/RhoA and actin/p53/cyclin A expression in ovarian cancer cell lines. This previously unidentified negative relationship between TCTP/RhoA and actin/p53/cyclin A may suggest that this interaction is linked with the high aggressiveness of ovarian cancers.


Subject(s)
Actins/metabolism , Biomarkers, Tumor/metabolism , Cyclin A1/metabolism , Gene Expression Regulation, Neoplastic , Ovarian Neoplasms/metabolism , Tumor Suppressor Protein p53/metabolism , rhoA GTP-Binding Protein/metabolism , Blotting, Western , Cell Line, Tumor , Female , Humans , Keratins/metabolism , Ovarian Neoplasms/pathology , Ovarian Neoplasms/ultrastructure , Protein Transport , Tumor Protein, Translationally-Controlled 1
5.
J Heart Lung Transplant ; 31(1): 73-84, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21497519

ABSTRACT

BACKGROUND: Dendritic cells are professional antigen presenting cells that perform antigen processing and antigen presentation functions and rely on the proper functioning and distribution of the endoplasmic reticulum (ER) and Golgi apparatus and of vesicular trafficking pathways. We previously developed a model system to study the mechanisms governing inhibition of chronic rejection of heart allografts. METHODS: Heterotopic cardiac transplants were placed intra-abdominally and the major histocompatibility class (MHC) class I allochimeric molecule, [α1h1/u]-RT1.Aa, which contains donor-type (Wistar Furth, WF; RT1u) immunogenic epitopes displayed on recipient-type (ACI, RT1a) sequences, was delivered by portal vein to the recipients of heterotopic hearts. Dendritic cells were isolated from the recipient bone marrow at 1 and 3 days after transplantation and were immunostained or processed for Western blotting with anti-RhoB, translationally controlled tumor protein (TCTP), Sprouty-related (Spred1) protein, ER, and Golgi antibodies. RESULTS: Western blotting analyses showed the downregulation of RhoB GTPase, TCTP, and Spred1 in dendritic cells isolated from allochimeric molecule-treated rats. Immunostaining showed that in these cells, Spred 1 was shifted to the base of cellular processes, Rho B formed nonvesicular band in the cell equator, and TCTP was highly enriched in the cell nucleus. The Golgi apparatus was drastically reduced in size and formed a tiny nonvesicular aggregate, and the ER partially lost vesicular appearance. CONCLUSIONS: The function of allochimeric molecule in the abrogation of heart allograft rejection may rely on the downregulation of RhoB pathway components that regulate the structure and function of the ER/Golgi/vesicular trafficking pathways involved in antigen processing and presentation by dendritic cells.


Subject(s)
Biomarkers, Tumor/immunology , Dendritic Cells/immunology , Graft Rejection/metabolism , Heart Transplantation/immunology , Histocompatibility Antigens/immunology , Repressor Proteins/immunology , Transplantation Tolerance/immunology , Animals , Biomarkers, Tumor/metabolism , Blotting, Western , Dendritic Cells/metabolism , Disease Models, Animal , Graft Rejection/immunology , Graft Survival/immunology , Histocompatibility Antigens/metabolism , Immune Tolerance , Immunoglobulin E , MAP Kinase Signaling System , Male , Rats , Rats, Inbred WF , Repressor Proteins/metabolism , Transplantation, Homologous , Tumor Protein, Translationally-Controlled 1
6.
J Clin Lab Anal ; 25(3): 207-11, 2011.
Article in English | MEDLINE | ID: mdl-21567470

ABSTRACT

United States Food and Drug Administration (FDA) in 2010 approved the use of immunosuppressant drug everolimus, which requires therapeutic drug monitoring in whole blood. Taking advantage of structural similarity between sirolimus and everolimus we attempted to rapidly estimate everolimus concentration from apparent sirolimus concentration obtained by using Architect sirolimus immunoassay and mathematical equations (both polynomial and linear). Mathematical equations were derived by curve-fitting methods based on observed apparent sirolimus concentration and true everolimus concentration determined by a liquid chromatography combined with mass spectrometry (LC/MS) method using eight everolimus standards (concentration range 1-30 ng/mL) prepared in whole blood. In order to determine the validity of our approach, we analyzed 12 specimens from patients receiving everolimus using both Architect sirolimus assay and LC/MS method. We observed good correlation between calculated everolimus values and true everolimus values as determined by LC/MS. However, if a patient is switched from sirolimus to everolimus, then sirolimus immunoassay can roughly estimate everolimus concentration plus any residual sirolimus present in whole blood and it is not possible to calculate everolimus concentration.


Subject(s)
Immunoassay/methods , Mass Spectrometry/methods , Models, Biological , Sirolimus/analogs & derivatives , Sirolimus/blood , Calibration , Chromatography, Liquid , Everolimus , Humans , Linear Models , Sirolimus/chemistry
7.
Transpl Immunol ; 23(4): 185-93, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20619345

ABSTRACT

Proper actin cytoskeleton architecture and dynamics are indispensable for events in the immunological response such as T cell migration, redistribution of T cell receptors, and interaction with antigen presenting cells. Thus, T cell activation, downstream signaling events and effector functions are all actin-dependent. Actin cytoskeleton architecture and dynamics are regulated by proteins belonging to the superfamily of small GTP-binding proteins, such as RhoA GTPase. We previously showed that the administration of an MHC class I allochimeric molecule [alpha1h1/u]-RT1.Aa, which contains donor-type (Wistar Furth, WF; RT1u) immunogenic epitopes displayed on recipient-type (ACI, RT1a) sequences, to the ACI recipient of heterotopic WF heart resulted in the restriction of the TCR repertoire, inhibition of T cell infiltration into the heterotopic cardiac allografts, abrogation of acute and chronic rejection, and induction of indefinite survival of the allograft. Here we show that the allochimeric molecule treatment caused downregulation of RhoA GTPase in T cells. This resulted in dramatic changes in the distribution of actin and the actin-binding protein, Hip55, in these cells, which in turn, inhibited T cell infiltration into the graft. This indicates that the immunosuppressive activity of the allochimeric molecule is achieved via downregulation of the RhoA pathway and disruption of the proper organization of T cell actin cytoskeleton to inhibit T cell functions such as motility and/or TCR signaling events.


Subject(s)
Actins/metabolism , Cytoskeleton/metabolism , Graft Rejection/drug therapy , Heart Transplantation , Microfilament Proteins/metabolism , T-Lymphocytes/drug effects , rhoA GTP-Binding Protein/metabolism , Animals , Cells, Cultured , Cytoskeleton/drug effects , Down-Regulation , Graft Rejection/immunology , Histocompatibility Antigens Class I/administration & dosage , Microfilament Proteins/genetics , Protein Transport/drug effects , Rats , Rats, Wistar , Recombinant Fusion Proteins/administration & dosage , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , rhoA GTP-Binding Protein/genetics , src Homology Domains/genetics
8.
Genesis ; 48(1): 8-19, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19882666

ABSTRACT

The MHC class I allochimeric protein containing donor-type epitopes on recipient-type heavy chains induces indefinite survival of heterotopic cardiac allografts in rats. We analyzed gene expression profile of heart allograft tissue. Mutated peptide [alpha1h1/u]-RT1.Aa that contains donor-type (Wistar Furth, WF; RT1u) immunogenic epitopes displayed on recipient-type (ACI, RT1a) was delivered into ACI recipients of WF hearts at the time of transplantation in addition to a 3 days course of oral cyclosporine. Microarray analysis was performed using Affymetrix Rat 230 2.0 Microarray. Allochimeric molecule treatment caused upregulation of genes involved in structural integrity of heart muscle, downregulation of IL-1beta a key modulator of the immune response, and downregulation of partitioning defective six homolog gamma PAR6, which is involved in T cell polarity, motility, and ability to scan dendritic cells (DC). These indicate that the immunosuppressive function of allochimeric molecule and/or the establishment of allograft tolerance depend on the induction of genes responsible for the heart tissue integrity, the suppression of cytokine pathway(s), and possibly the impairment of T cells mobility and their DC scanning ability. These novel findings may have important clinical implications for inhibition of chronic rejection in transplant recipients.


Subject(s)
Gene Expression Profiling , Heart Transplantation , Histocompatibility Antigens/immunology , Transplantation Tolerance/genetics , Adaptor Proteins, Signal Transducing , Animals , Carrier Proteins/genetics , Cyclosporine/pharmacology , Gene Expression/drug effects , Immunosuppressive Agents/pharmacology , Interleukin-1beta/genetics , Male , Oligonucleotide Array Sequence Analysis , Rats , Rats, Inbred Strains , Reverse Transcriptase Polymerase Chain Reaction , Transplantation Tolerance/immunology
9.
PLoS One ; 4(12): e8020, 2009 Dec 02.
Article in English | MEDLINE | ID: mdl-19956540

ABSTRACT

BACKGROUND: The allochimeric MHC class I molecule [alpha1h1/u]-RT1.Aa that contains donor-type (Wistar Furth, WF; RT1u) epitopes displayed on recipient-type (ACI, RT1a) administered in conjunction with sub-therapeutic dose of cyclosporine (CsA) induces indefinite survival of heterotopic cardiac allografts in rat model. In vascularized transplantation models, the spleen contributes to graft rejection by generating alloantigen reactive T cells. The immune response in allograft rejection involves a cascade of molecular events leading to the formation of immunological synapses between T cells and the antigen-presenting cells. METHODOLOGY/PRINCIPAL FINDINGS: To elucidate the molecular pathways involved in the immunosuppressive function of allochimeric molecule we performed microarray and quantitative RTPCR analyses of gene expression profile of splenic T cells from untreated, CsA treated, and allochimeric molecule + subtherapeutic dose of CsA treated animals at day 1, 3 and 7 of post transplantation. Allochimeric molecule treatment caused down regulation of genes involved in actin filament polymerization (RhoA and Rac1), cell adhesion (Catna1, Vcam and CD9), vacuolar transport (RhoB, Cln8 and ATP6v1b2), and MAPK pathway (Spred1 and Dusp6) involved in tubulin cytoskeleton reorganization and interaction between actin and microtubule cytoskeleton. All these genes are involved in T cell polarity and motility, i.e., their ability to move, scan and to form functional immunological synapse with antigen presenting cells (APCs). CONCLUSIONS: These results indicate that the immunosuppressive function of allochimeric molecule may depend on the impairment of T cells' movement and scanning ability, and possibly also the formation of immunological synapse. We believe that these novel findings may have important clinical implications for organ transplantation.


Subject(s)
Cell Movement/genetics , Cell Polarity/genetics , Down-Regulation/genetics , Heart Transplantation/immunology , Histocompatibility Antigens Class I/genetics , T-Lymphocytes/cytology , Transplantation Tolerance/genetics , Animals , Cell Movement/drug effects , Cell Polarity/drug effects , Cyclosporine/pharmacology , Down-Regulation/drug effects , Gene Expression Profiling , Histocompatibility Antigens Class I/immunology , Peptides/pharmacology , Principal Component Analysis , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Spleen/cytology , Spleen/drug effects , Spleen/metabolism , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Time Factors , Transplantation Tolerance/drug effects
10.
J Immunol ; 176(1): 128-37, 2006 Jan 01.
Article in English | MEDLINE | ID: mdl-16365403

ABSTRACT

STATs play key roles in immune function. We examined the role of STAT5a/b in allograft rejection. STAT5a/b-deficient mice showed a 4-fold increased survival time of heart allografts (p < 0.01). Unlike wild type, purified STAT5a/b-/- T cells transferred to Rag1-/- recipients failed to mediate heart allograft rejection until supplemented with STAT5a/b-/- B cells. In vitro, STAT5a/b-/- T cells did not proliferate in response to Con A or alloantigens but entered apoptosis within 48 h (95%). Activated STAT5a/b-/- T cells showed increased expression of proapoptotic (caspases, DNA repair genes, TNF/TNFR-associated factor family genes) and decreased antiapoptotic mRNAs in microarrays, while Western blots confirmed reduced antiapoptotic Bcl-2 and elevated proapoptotic Bax protein expression. Interestingly, at 24 h postactivation, STAT5a/b+/+ and STAT5a/b-/- T cells produced similar levels of IL-2, IL-4, IL-10, and IFN-gamma mRNA; ELISPOT assay showed an equivalent number of IL-4- and IFN-gamma-producing T cells in both STAT5a/b+/+ and STAT5a/b-/- splenic populations. Sera from STAT5a/b+/+ and STAT5a/b-/- rejectors had donor-specific IgM, IgG1, IgG2a, and IgG2b Ab, while STAT5a/b deficiency had no impact on B cell survival or proliferation in response to LPS. Compared with allografts from STAT5a/b+/+ recipients, heart allografts from STAT5a/b-/- recipients had markedly reduced infiltration by CD4 and CD8 T cells but increased infiltration by B cells and dense endothelial deposition of C4d, a marker of humoral rejection. Thus, activated STAT5a/b-/- T cells produce cytokines prior to entering apoptosis, thereby promoting differentiation of B cells yielding donor-specific IgM and IgG Ab that mediate allograft rejection.


Subject(s)
Apoptosis/immunology , B-Lymphocytes/immunology , Graft Rejection/immunology , Heart Transplantation/immunology , STAT5 Transcription Factor/immunology , T-Lymphocytes/immunology , Adoptive Transfer , Animals , Blotting, Western , Immunoglobulin G/blood , Immunoglobulin G/immunology , Immunoglobulin M/blood , Immunoglobulin M/immunology , In Situ Nick-End Labeling , Mice , Mice, Knockout , Oligonucleotide Array Sequence Analysis , STAT5 Transcription Factor/deficiency , Transplantation, Homologous/immunology
11.
J Immunol ; 175(7): 4236-46, 2005 Oct 01.
Article in English | MEDLINE | ID: mdl-16177063

ABSTRACT

JAK3 is a cytoplasmic tyrosine kinase with limited tissue expression but is readily found in activated T cells. Patients lacking JAK3 are immune compromised, suggesting that JAK3 represents a therapeutic target for immunosuppression. Herein, we show that a Mannich base, NC1153, blocked IL-2-induced activation of JAK3 and its downstream substrates STAT5a/b more effectively than activation of the closely related prolactin-induced JAK2 or TNF-alpha-driven NF-kappaB. In addition, NC1153 failed to inhibit several other enzymes, including growth factor receptor tyrosine kinases, Src family members, and serine/threonine protein kinases. Although NC1153 inhibited proliferation of normal human T cells challenged with IL-2, IL-4, or IL-7, it did not block T cells void of JAK3. In vivo, a 14-day oral therapy with NC1153 significantly extended survival of MHC/non-MHC mismatched rat kidney allografts, whereas a 90-day therapy induced transplantation tolerance (>200 days). Although NC1153 acted synergistically with cyclosporin A (CsA) to prolong allograft survival, it was not nephrotoxic, myelotoxic, or lipotoxic and did not increase CsA-induced nephrotoxicity. In contrast to CsA, NC1153 was not metabolized by cytochrome P450 3A4. Thus, NC1153 prolongs allograft survival without several toxic effects associated with current immunosuppressive drugs.


Subject(s)
Graft Survival/drug effects , Immunosuppressive Agents/pharmacology , Mannich Bases/pharmacology , Animals , Cell Line , Cyclosporine/pharmacology , Humans , Interleukin-2/physiology , Janus Kinase 3 , Kidney Transplantation , Phosphorylation/drug effects , Protein-Tyrosine Kinases/metabolism , Rats , Rats, Inbred ACI , Rats, Inbred Lew , Spleen/cytology , Spleen/transplantation , Transplantation, Homologous
12.
Transplantation ; 73(8): 1227-35, 2002 Apr 27.
Article in English | MEDLINE | ID: mdl-11981413

ABSTRACT

BACKGROUND: The various toxicities associated with the general immune suppression resulting from current clinical immunosuppressive therapies continue to plague transplant recipients as well as jeopardize allograft survival. METHODS: The present study utilized allochimeric class I MHC antigens (alpha1hu70-77-RT1.Aa) bearing only four donor RT1.Au polymorphic amino acids (a.a.; His70, Val73, Asn74, and Asn77) superimposed on the recipient RT1.Aa background to induce transplantation tolerance in the rat cardiac transplant model. RESULTS: Oral delivery of alpha1hu70-77-RT1.Aa protein alone (days 0-6) induced tolerance, as evidenced by inhibition of both acute and chronic rejection processes. Delivery of alpha1hu70-77-RT1.Aa with therapeutic doses of cyclosporine (CsA) also prevented chronic rejection, otherwise readily developed after treatment with CsA alone. A polymerase chain reaction (PCR)-based analysis showed that tolerant recipients had reduced numbers of interleukin (IL)-2/interferon (IFN)-gamma-producing T helper (Th)1 cells and elevated numbers of IL-4/IL-10-producing Th2 cells. Adoptive transfer experiments revealed that potent regulatory T cells mediated tolerance. The same T cells displayed diminished T cell receptor (TCR)-driven signaling via extracellular regulated kinase, AP-1, and NF-kappaB, as well as the common gamma-chain (gammac) cytokine-receptor-induced signaling by Janus kinase 3 (Jak3)/stimulators and activators of transcription Stat/5 pathways. Tolerance induction was prevented in vivo by inhibition of signal 2 by CTL4Ig or of signal 3 by either rapamycin, which disrupts the mammalian target of rapamycin, or AG490, which inhibits Jak3. Finally, partial or complete tyrosine phosphorylation of Zap70 was observed in alloantigen-specific T cell clones in response to tolerogenic versus immunogenic peptides, respectively. CONCLUSIONS: Tolerance induction by allochimeric proteins is achieved by partial TCR activation in the presence of signals 2 and 3, resulting in a skewed Th2 phenotype.


Subject(s)
Antigens, Differentiation/immunology , Cytokines/immunology , Heart Transplantation/immunology , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens/immunology , Immunoconjugates , Immunoglobulin lambda-Chains/immunology , Receptors, Antigen, T-Cell/immunology , Transplantation Chimera , Abatacept , Adoptive Transfer , Animals , Antigens, CD , CTLA-4 Antigen , Drug Therapy, Combination , Immunosuppression Therapy/methods , Immunosuppressive Agents/therapeutic use , Lymphocyte Activation , Male , Polymerase Chain Reaction , Rats , Rats, Inbred BN , Rats, Inbred WF , Signal Transduction/immunology , T-Lymphocytes/immunology , Transplantation, Homologous
13.
Blood ; 99(2): 680-9, 2002 Jan 15.
Article in English | MEDLINE | ID: mdl-11781254

ABSTRACT

Janus kinase 3 (Jak3) is a cytoplasmic tyrosine (Tyr) kinase associated with the interleukin-2 (IL-2) receptor common gamma chain (gamma(c)) that is activated by multiple T-cell growth factors (TCGFs) such as IL-2, -4, and -7. Using human T cells, it was found that a recently discovered variant of the undecylprodigiosin family of antibiotics, PNU156804, previously shown to inhibit IL-2-induced cell proliferation, also blocks IL-2-mediated Jak3 auto-tyrosine phosphorylation, activation of Jak3 substrates signal transducers and activators of transcription (Stat) 5a and Stat5b, and extracellular regulated kinase 1 (Erk1) and Erk2 (p44/p42). Although PNU156804 displayed similar efficacy in blocking Jak3-dependent T-cell proliferation by IL-2, -4, -7, or -15, it was more than 2-fold less effective in blocking Jak2-mediated cell growth, its most homologous Jak family member. A 14-day alternate-day oral gavage with 40 to 120 mg/kg PNU156804 extended the survival of heart allografts in a dose-dependent fashion. In vivo, PNU156804 acted synergistically with the signal 1 inhibitor cyclosporine A (CsA) and additively with the signal 3 inhibitor rapamycin to block allograft rejection. It is concluded that inhibition of signal 3 alone by targeting Jak3 in combination with a signal 1 inhibitor provides a unique strategy to achieve potent immunosuppression.


Subject(s)
Cyclosporine/therapeutic use , Enzyme Inhibitors/therapeutic use , Graft Survival/drug effects , Immunosuppressive Agents/therapeutic use , Milk Proteins , Prodigiosin/analogs & derivatives , Prodigiosin/therapeutic use , Protein Processing, Post-Translational/drug effects , Protein-Tyrosine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins , Pyrroles/therapeutic use , Sirolimus/therapeutic use , T-Lymphocytes/drug effects , Administration, Oral , Animals , Cell Line/drug effects , Cells, Cultured/drug effects , Cells, Cultured/enzymology , Cyclosporine/pharmacology , DNA-Binding Proteins/metabolism , Dose-Response Relationship, Drug , Drug Interactions , Drug Synergism , Enzyme Inhibitors/pharmacology , Heart Transplantation , Humans , Immunosuppressive Agents/pharmacology , Interleukins/pharmacology , Janus Kinase 2 , Janus Kinase 3 , Jurkat Cells/drug effects , Jurkat Cells/enzymology , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , MAP Kinase Signaling System/drug effects , Phosphorylation/drug effects , Prodigiosin/pharmacology , Pyrroles/pharmacology , Rats , Rats, Inbred BUF , Rats, Inbred WF , STAT5 Transcription Factor , Sirolimus/pharmacology , T-Lymphocytes/enzymology , Trans-Activators/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...