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










Publication year range
1.
Transpl Immunol ; 75: 101704, 2022 12.
Article in English | MEDLINE | ID: mdl-36057381

ABSTRACT

Non-human primate (NHP) renal transplantation models are widely used vivo models for researching new immunosuppressive therapies including allograft tolerance strategies. To enroll animals into a tolerance study, an immunosuppressive regimen that efficiently establishes stable renal function in NHPs is needed. Here, we assessed the effect of triple therapy comprising 2.0 mg/kg tacrolimus, mycophenolate mofetil and a steroid and its success rate for achieving stable renal function. In addition, to predict the pathophysiological consequences of withdrawing immunosuppressants, an indispensable process after induction of tolerance, we also assessed changes in the stable renal state maintained by triple therapy after drug withdrawal. Six cynomolgus monkeys were used. The median survival time was >176 days over the dosing period and 45 days after drug withdrawal. The triple therapy successfully induced stable graft function without calcineurin inhibitor nephrotoxicity in three of six recipients, although adopting trough-dependent tacrolimus dose adjustment rather than a preset dose regimen could improve on the present strategy. Further, drug withdrawal led to deterioration of renal function, de novo donor specific antibody production and increased the memory/naïve T cell ratio within two weeks post drug withdrawal. We expect that these findings contribute to establish one of the choices for animal model for evaluating future tolerance therapy for renal transplantation.


Subject(s)
Kidney Transplantation , Animals , Tacrolimus/therapeutic use , Graft Rejection/drug therapy , Graft Rejection/prevention & control , Mycophenolic Acid/therapeutic use , Immunosuppressive Agents/therapeutic use , Immunosuppressive Agents/pharmacology , Primates , Calcineurin Inhibitors/therapeutic use , Graft Survival , Drug Therapy, Combination
2.
J Pharmacol Exp Ther ; 380(1): 26-33, 2022 01.
Article in English | MEDLINE | ID: mdl-34728559

ABSTRACT

Thymic stromal lymphopoietin (TSLP), positioned at the top of the inflammatory cascade, is a key regulator that enhances allergic inflammatory responses by activating T helper type 2 cells, Group 2 innate lymphoid cells (ILC2), and myeloid dendritic cells (mDCs) via the TSLP receptor (TSLPR). We evaluated the inhibitory effects of ASP7266, a novel recombinant fully human IgG1 monoclonal antibody against TSLPR, on TSLP signaling and inflammation. The inhibitory effects of ASP7266 and the control antibody tezepelumab on TSLP and TSLPR interactions were investigated using a proliferation assay with TSLP stimulation and a chemokine production assay. The pharmacological effects of ASP7266 were investigated by examining differentiation of naive CD4+ T cells, ILC2 cytokine production, and ascaris extract-induced skin allergic reaction in cynomolgus monkeys. ASP7266 potently inhibited TSLP-induced cell proliferation and C-C motif chemokine ligand 17 production. Furthermore, ASP7266 inhibited TSLP-stimulated mDC-mediated naive CD4+ T-cell differentiation and interleukin 5 production by lineage-negative peripheral blood mononuclear cells, which can be considered ILC2 in vitro. In sensitized monkeys, ASP7266 completely suppressed ascaris extract-induced allergic skin reactions. Based on these results, ASP7266, a novel human therapeutic antibody against TSLPR, is a potential therapy for patients with allergic diseases. SIGNIFICANCE STATEMENT: TSLP, positioned at the top of the inflammatory cascade, plays a key role in various allergic diseases, including asthma, chronic rhinosinusitis with nasal polyposis, and atopic dermatitis. Here we show that the anti-TSLPR antibody ASP7266 exhibited excellent pharmacological activity in preclinical studies. Therefore, ASP7266 has the potential to be a promising treatment option for patients with allergic disorders.


Subject(s)
Antibodies, Monoclonal/immunology , Dermatitis, Allergic Contact/drug therapy , Receptors, Cytokine/immunology , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Cell Line , Cell Proliferation , Cells, Cultured , Cytokines/metabolism , Dendritic Cells/drug effects , Dendritic Cells/immunology , Humans , Macaca fascicularis , Male , Mice , Receptors, Cytokine/antagonists & inhibitors , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Helper-Inducer/immunology
3.
J Pharmacol Sci ; 148(1): 134-141, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34924117

ABSTRACT

Peficitinib, a pan-JAK inhibitor, is known to suppress the activation of fibroblast-like synoviocytes (FLSs) and thereby reduces joint inflammation associated with rheumatoid arthritis (RA). However, the effect on osteoporosis in RA remains to be elucidated. In this study, the effect of peficitinib or etanercept on joint inflammation, and consequently decreased bone mineral density (BMD) was evaluated in mice with collagen-induced arthritis (CIA). Additionally, the effect on RANKL production from osteoblasts differentiated from the mesenchymal stem cells of RA patients was evaluated. Administration of peficitinib for established CIA ameliorated arthritis and improved BMD in the femoral metaphysis, but not in the femoral diaphysis. Conversely, etanercept suppressed an increase in synovial inflammatory markers but did not improve arthritic conditions or the reduction of BMD in either region. All elevated bone formation and bone resorption markers were decreased with peficitinib but only partially decreased with etanercept. Furthermore, production of RANKL by human osteoblasts was suppressed by peficitinib but enhanced by etanercept. Unlike etanercept, peficitinib is thought to increase BMD by ameliorating the high bone turnover associated with RA states, resulting in improvement of bone fragility. Our data provide evidence that peficitinib would be expected to show efficacy for osteoporosis associated with RA.


Subject(s)
Adamantane/analogs & derivatives , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/metabolism , Bone Density/drug effects , Bone Remodeling/drug effects , Bone and Bones/metabolism , Janus Kinase Inhibitors/pharmacology , Janus Kinase Inhibitors/therapeutic use , Niacinamide/analogs & derivatives , Osteoporosis/drug therapy , Adamantane/pharmacology , Adamantane/therapeutic use , Animals , Arthritis, Rheumatoid/complications , Bone Resorption/prevention & control , Disease Models, Animal , Male , Mice, Inbred DBA , Niacinamide/pharmacology , Niacinamide/therapeutic use , Osteoblasts/metabolism , Osteoporosis/etiology , Osteoporosis/prevention & control , RANK Ligand/metabolism
4.
Eur J Pharmacol ; 838: 145-152, 2018 Nov 05.
Article in English | MEDLINE | ID: mdl-30196110

ABSTRACT

Donor-specific antibodies (DSA) are a major risk factor for antibody-mediated rejection (ABMR) in solid organ transplantation, and ABMR remains a medical challenge. Therefore, effective anti-ABMR therapies are needed to improve overall graft survival. Cathepsin S (Cat S) is an essential protease for antigen peptide loading onto lysosomal/endosomal major histocompatibility complex (MHC) class II molecules to promote antigen presentation. Cat S deficiency produces immuno-deficient phenotypes including a suppressed humoral immune response, and Cat S inhibition reportedly prevents autoimmunity. However, little is known about the effects of Cat S inhibitors on organ transplantation, especially ABMR. Here, we report the pharmacological profile of novel Cat S inhibitors, AS2761325 and AS2863995, and explore their preventive potential on DSA production and acute rejection in a mouse cardiac transplantation model. Cat S inhibitors potently inhibited upregulation of antigen peptide loading MHC class II expression on the surface of splenic B cells and suppressed ovalbumin-induced T cell-dependent antibody production in mice. In a mouse cardiac transplantation model, oral administration of AS2761325 monotherapy inhibited DSA production without affecting graft survival. When combined with a suboptimal dose of tacrolimus, AS2761325 significantly prolonged graft survival. The more potent Cat S inhibitor AS2863995 also prolonged graft survival and almost completely suppressed DSA production. These results suggest that Cat S inhibitors may be promising ABMR prophylaxis drug candidates. Combination therapy comprising a Cat S inhibitor and calcineurin inhibitors may be a more effective immunosuppressive maintenance therapy for controlling both cell-mediated and antibody-mediated rejection.


Subject(s)
Allografts/immunology , Cathepsins/antagonists & inhibitors , Graft Rejection/drug therapy , Heart Transplantation/adverse effects , Immunosuppressive Agents/pharmacology , Administration, Oral , Animals , Antibodies/immunology , Antibodies/metabolism , Antigen Presentation/drug effects , Antigen Presentation/immunology , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Calcineurin Inhibitors/pharmacology , Calcineurin Inhibitors/therapeutic use , Disease Models, Animal , Drug Therapy, Combination/methods , Graft Rejection/immunology , Graft Survival/drug effects , Histocompatibility Antigens Class II/immunology , Histocompatibility Antigens Class II/metabolism , Humans , Immunity, Humoral/drug effects , Immunity, Humoral/immunology , Immunosuppressive Agents/therapeutic use , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Tacrolimus/pharmacology , Treatment Outcome
5.
Eur J Pharmacol ; 833: 320-327, 2018 Aug 15.
Article in English | MEDLINE | ID: mdl-29932925

ABSTRACT

Integrin, alpha9 subunit (hereinafter, alpha9) has been identified as a novel putative therapeutic target for rheumatoid arthritis (RA). Support for this target comes from the observations that alpha9 is overexpressed both in the joints of RA patients and in animal models of arthritis. In the experimental models, the increase in alpha9 expression precedes the onset of arthritic symptoms. The current study presents data on the pharmacological profile of an anti-alpha9 antibody in a collagen-induced arthritis (CIA) mouse model. Administration of an alpha9-blocking antibody in CIA mice suppressed the development of arthritis and significantly decreased plasma level of activated fibroblast-like synoviocyte (FLS)-derived biomarkers without reducing the formation of anti-type II collagen antibodies. While anti-alpha9 antibody administration significantly suppress the accumulation of immune cells in arthritic joints it had no effect on immune cell number in the spleen. Furthermore, in non-arthritic mice, alpha9 had no inhibitory effect in either a mixed lymphocyte reaction (MLR) or in a delayed type hypersensitivity (DTH) reaction. These results suggest that blocking alpha9 exerts its anti-arthritic effect through suppression of FLS-activation via a non-immune mediated mechanism. Finally, therapeutic administration of anti-alpha9 antibody alleviated established arthritis in CIA mice. Our data provide evidence that alpha9 blockade is a promising therapy for joint inflammation with minimal systemic immunomodulation.


Subject(s)
Antirheumatic Agents/pharmacology , Arthritis, Experimental/drug therapy , Arthritis, Rheumatoid/drug therapy , Immunomodulation/drug effects , Integrins/antagonists & inhibitors , Animals , Antibodies/pharmacology , Antibodies/therapeutic use , Antirheumatic Agents/therapeutic use , Arthritis, Experimental/immunology , Arthritis, Rheumatoid/immunology , Biomarkers/metabolism , Cells, Cultured , Collagen Type II/immunology , Humans , Integrins/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred DBA , Tacrolimus/pharmacology , Treatment Outcome
6.
Transpl Immunol ; 46: 14-20, 2018 02.
Article in English | MEDLINE | ID: mdl-28988984

ABSTRACT

BACKGROUND: Janus kinase (JAK) inhibitors are thought to be promising candidates to aid renal transplantation. However, the effectiveness of JAK inhibitors against features of chronic rejection, including interstitial fibrosis/tubular atrophy (IF/TA) and glomerulosclerosis, has not been elucidated. Here, we investigated the effect of AS2553627, a novel JAK inhibitor, on the development of chronic rejection in rat renal transplantation. METHODS: Lewis (LEW) to Brown Norway (BN) rat renal transplantation was performed. Tacrolimus (TAC) at 0.1mg/kg was administered intramuscularly once a day for 10 consecutive days starting on the day of transplantation (days 0 to 9) to prevent initial acute rejection. After discontinuation of TAC treatment from days 10 to 28, AS2553627 (1 and 10mg/kg) was orally administered with TAC. At 13weeks after renal transplantation, grafts were harvested for histopathological and mRNA analysis. Creatinine and donor-specific antibodies were measured from plasma samples. Urinary protein and kidney injury markers were also evaluated. RESULTS: AS2553627 in combination with TAC exhibited low plasma creatinine and a marked decrease in urinary protein and kidney injury markers, such as tissue inhibitor of metalloproteinase-1 and kidney injury molecule-1. At 13weeks, histopathological analysis revealed that AS2553627 treatment inhibited glomerulosclerosis and IF/TA. In addition, upregulation of cell surface markers, fibrosis/epithelial-mesenchymal transition and inflammation-related genes were reduced by the combination of AS2553672 and TAC, particularly CD8 and IL-6 mRNAs, indicating that AS2553627 prevented cell infiltration and inflammation in renal allografts. CONCLUSIONS: These results indicate the therapeutic potential of JAK inhibitors in chronic rejection progression, and suggest that AS2553627 is a promising agent to improve long-term graft survival after renal transplantation.


Subject(s)
Allografts/immunology , Glomerulosclerosis, Focal Segmental/prevention & control , Graft Rejection/prevention & control , Kidney Transplantation , Piperidines/therapeutic use , Pyrroles/therapeutic use , Animals , Chronic Disease , Disease Models, Animal , Drug Therapy, Combination , Glomerulosclerosis, Focal Segmental/immunology , Graft Rejection/immunology , Humans , Interleukin-6/genetics , Interleukin-6/metabolism , Janus Kinases/antagonists & inhibitors , Rats , Rats, Inbred Lew , Tacrolimus/therapeutic use
7.
Transpl Immunol ; 40: 57-65, 2017 02.
Article in English | MEDLINE | ID: mdl-28077266

ABSTRACT

FTY720, a S1P-receptor modulator, has shown to be effective in several transplant and autoimmune disease models, via modulating lymphocyte homing into secondary lymphoid organs (SLOs), and thereby reducing these cells in peripheral blood. ASP0028, a newly developed S1P1/S1P5-selective agonist, presented comparable efficacy to FTY720 and wider safety margins than FTY720. In this study, we assessed the efficacy and safety of ASP0028 co-administered with suboptimal-dose of tacrolimus in the Cynomolgus monkey renal transplantation model. Seven animals in group-1 or group-2 received mono-tacrolimus 1.0mg/kg once a day (QD), or ASP0028 0.6mg/kg plus tacrolimus 1.0mg/kg QD, respectively. Eight animals in group-3 received ASP0028 1.2mg/kg plus tacrolimus 1.0mg/kg QD. The allograft median survival time (MST) in group-2 and group-3 were significantly extended to 41 and 61.5days, versus that of 28days in group-1 (p=0.036 and 0.001, respectively). ASP0028 administration remarkably reduced absolute numbers of peripheral lymphocytes, particularly subsets of CD4+/ or CD8+/naive and central memory cells, CD4+/Treg cells, and to a lesser extent on B cells, but not CD4+/ or CD8+/effector memory cells and NK cells. These data show ASP0028 combined with suboptimal-dose of tacrolimus effectively prolongs renal allograft survival in nonhuman primates (NHPs) with well tolerated safety, supporting its further investigation to optimize CNI-sparing regimens.


Subject(s)
B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Graft Rejection/drug therapy , Immunosuppressive Agents/therapeutic use , Kidney Transplantation , T-Lymphocytes, Regulatory/immunology , Tacrolimus/therapeutic use , Animals , Disease Models, Animal , Drug Therapy, Combination , Fingolimod Hydrochloride/therapeutic use , Humans , Immunologic Memory , Macaca fascicularis , Receptors, Lysosphingolipid/agonists , Receptors, Lysosphingolipid/antagonists & inhibitors , Treatment Outcome
8.
Transpl Immunol ; 38: 19-26, 2016 09.
Article in English | MEDLINE | ID: mdl-27545900

ABSTRACT

BACKGROUND: The Fischer-to-Lewis (LEW) rat model of kidney transplantation is a widely accepted and well-characterized model of chronic rejection. In contrast to transplantation in a clinical setting, however, the absence of treatment with immunosuppressants and only minor mismatch of major histocompatibility complexes (MHCs) are critical discrepancies. Here, we established a rat model of chronic rejection using fully MHC-mismatched strains in which kidney disease progresses even under immunosuppressive therapy. METHODS: LEW (RT1(l)) rats were used as donors and Brown Norway (BN, RT1(n)) rats as recipients. Intramuscular administration of 0.1mg/kg of tacrolimus was initiated on the day of transplantation. Post-transplantation, this dose was maintained until Day 9, suspended until Day 28 and then resumed from Day 29. Renal function, histopathology, and levels of donor-specific antibody (DSA) and several biomarkers of renal injury were assessed. RESULTS: On Day 91 post-transplantation, recipients received tacrolimus treatment with short-term suspension exhibited reduced renal function and changes in histology. Those were characteristics of chronic rejection including glomerulosclerosis, interstitial fibrosis, and tubular atrophy in human transplantation recipients. Urinary protein excretion increased in a linear fashion, and elevated levels of several biomarkers of renal injury and DSA were observed even under administration of an immunosuppressant. CONCLUSIONS: We established an allograft rejection model with impaired renal function and typical histopathological changes of chronic rejection in fully MHC-mismatched rats by controlling administration of an immunosuppressant. These findings suggest that this model more accurately reflects transplantation in a clinical setting than existing models and enables the evaluation of therapeutic agents.


Subject(s)
Graft Rejection/drug therapy , Immunosuppressive Agents/therapeutic use , Kidney Transplantation , Kidney/pathology , Tacrolimus/therapeutic use , Animals , Atrophy , Biomarkers/metabolism , Chronic Disease , Disease Models, Animal , Feasibility Studies , Fibrosis , Graft Rejection/immunology , Histocompatibility Antigens/immunology , Humans , Isoantibodies/blood , Kidney/immunology , Rats , Rats, Inbred BN , Rats, Inbred Lew , Sclerosis , Transplantation, Homologous
9.
Eur J Immunol ; 44(6): 1770-80, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24595757

ABSTRACT

The activation of T cells is known to be accompanied by the temporary downmodulation of the TCR/CD3 complex on the cell surface. Here, we established a novel monoclonal antibody, Dow2, that temporarily induces downmodulation of the TCR/CD3 complex in mouse CD4(+) T cells without activating T cells. Dow2 recognized the determinant on CD3ε; however, differences were observed in the binding mode between Dow2 and the agonistic anti-CD3ε Ab, 145-2C11. An injection of Dow2 in vivo resulted in T-cell anergy, and prolonged the survival of cardiac allografts without a marked increase in cytokine release. The phosphorylated forms of the signaling proteins PLC-γ1 and LAT in Dow2-induced anergic T cells were markedly decreased upon stimulation. However, the levels of phosphorylated LAT and PLCγ1 in Dow2-induced anergic T cells could be rescued in the presence of the proteasome inhibitor MG-132. These results suggest that proteasome-mediated degradation is involved in hypophosphorylated LAT and PLCγ1 in Dow2-induced anergic T cells. The novel CD3-specific Ab, Dow2, may provide us with a unique tool for inducing immunosuppression.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Antibodies, Monoclonal, Murine-Derived/pharmacology , CD3 Complex/immunology , Clonal Anergy/drug effects , Membrane Proteins/immunology , Phospholipase C gamma/immunology , Phosphoproteins/immunology , T-Lymphocytes/immunology , Animals , Antibodies, Monoclonal, Murine-Derived/immunology , Male , Mice , Mice, Inbred BALB C , Phosphorylation/drug effects , Phosphorylation/immunology , Proteasome Endopeptidase Complex/immunology , Proteolysis/drug effects
10.
J Toxicol Sci ; 33(5): 575-84, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19043279

ABSTRACT

The aim of this study is to investigate the effect of the pharmacokinetic profile of tacrolimus on its pancreatic toxicity and efficacy in rats. For toxicity evaluation, doses of 0.03, 0.1, or 0.3 mg/kg/day were given once daily for 8 days in the bolus intravenous injection groups. In the continuous intravenous infusion groups, tacrolimus was infused using an Alzet osmotic mini-pump for 9 days at the same doses. Pancreatic insulin content decreased dose-dependently in both the bolus intravenous injection and continuous intravenous infusion groups, and there was no significant difference between the decreases caused by the two dosing regimens. At 0.03 mg/kg, continuous intravenous infusion did not cause glucose intolerance, but bolus intravenous injection induced significant and dose-dependent glucose intolerance. The pharmacokinetic data indicated that continuous intravenous infusion resulted in a sustained blood drug concentration with an area under the curve (AUC) similar to that obtained with the bolus administration at the same dose. For efficacy evaluation, donor ear grafts were transplanted to the lateral thoraxes of recipients. Tacrolimus doses of 0.01, 0.1, or 1 mg/kg/day were administered from day 0 to day 13. Both bolus intramuscular administration and continuous intravenous infusion prolonged skin allograft survival dose-dependently, and there was no significant difference between the median survival times of groups given the same doses. To summarize, the sustained-release of tacrolimus resulted in a steady blood drug concentration with an AUC similar to that of the bolus administration. In rats, it was better tolerated and just as efficacious as the bolus administration without producing a higher maximal blood concentration (Cmax). These results indicate that the sustained-release formulation has the potential to improve the safety of tacrolimus.


Subject(s)
Immunosuppressive Agents , Pancreas/drug effects , Tacrolimus , Animals , Blood Glucose/metabolism , Body Weight/drug effects , Dose-Response Relationship, Drug , Glucose Tolerance Test , Graft Rejection/prevention & control , Immunosuppressive Agents/pharmacokinetics , Immunosuppressive Agents/therapeutic use , Immunosuppressive Agents/toxicity , Infusions, Intravenous , Injections, Intramuscular , Insulin/metabolism , Male , Pancreas/metabolism , Rats , Rats, Inbred F344 , Rats, Sprague-Dawley , Skin Transplantation , Tacrolimus/pharmacokinetics , Tacrolimus/therapeutic use , Tacrolimus/toxicity
11.
J Invest Dermatol ; 128(8): 1969-76, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18337836

ABSTRACT

In psoriasis, CD28/B7 costimulatory molecules are well characterized. Here, using the severe combined immunodeficient (SCID) mouse-psoriasis xenograft model, we report therapeutic efficacy of a humanized anti-CD28 monoclonal antibody (FR255734; Astellas Pharmaceuticals Inc., Tokyo, Japan). Transplanted psoriasis plaques on the SCID mouse were treated weekly for 4 weeks with intraperitoneal injections of FR255734 at 10, 3, and 1-mg kg(-1) doses. Groups treated with doses of 10 and 3 mg kg(-1) had significant thinning of the epidermis and reduced HLA-DR-positive lymphocytic infiltrates. The length of the rete pegs changed from 415.2+/-59.6 to 231.4+/-40.4 microm (P<0.005) in the 10-mg kg(-1) group, and from 323.4+/-69.6 to 237.5+/-73.6 microm in the 3-mg kg(-1) group (P=0.002). Positive controls treated with CTLA4-Ig and cyclosporine had significant histological improvement, whereas plaques treated with saline and isotype controls (human and mouse IgG2) remained unchanged. In vitro studies have shown that FR255734 effectively blocked T-cell proliferation and proinflammatory cytokine production. These observations warrant studies to evaluate the efficacy of FR255734 in human autoimmune diseases.


Subject(s)
Antibodies, Monoclonal/therapeutic use , CD28 Antigens/immunology , Psoriasis/drug therapy , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Cell Proliferation/drug effects , Cyclosporine/therapeutic use , Disease Models, Animal , Dose-Response Relationship, Drug , Humans , Immunoglobulin G/therapeutic use , Immunosuppressive Agents/therapeutic use , Mice , Mice, SCID , Psoriasis/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/pathology , T-Lymphocytes/physiology , Transplantation, Heterologous/immunology
12.
Eur J Pharmacol ; 509(1): 11-9, 2005 Feb 10.
Article in English | MEDLINE | ID: mdl-15713424

ABSTRACT

Immunophilin ligands are neuroregenerative agents, characterized by binding to FK506 binding proteins (FKBPs), which stimulate recovery of neurons in a variety of injury paradigms. Here we report the discovery of a novel, non-immunosuppressive immunophilin ligand, FK1706. FK1706, a derivative of FK506, showed similarly high affinity for two FKBP subtypes, FKBP-12 and FKBP-52, but inhibited T-cell proliferation and interleukin-2 cytokine production with much lower potency and efficacy than FK506. FK1706 (0.1 to 10 nM) significantly potentiated nerve growth factor (NGF)-induced neurite outgrowth in SH-SY5Y cells, as did FK506. This neurite potentiation could be blocked by an anti-FKBP-52 antibody, as well as by specific pharmacological inhibitors of phospholipase C (PLC), phosphatidylinositol 3-kinase (PI3K), and the Ras/Raf/Mitogen-Activated Protein Kinase (MAPK) signaling pathway. FK1706 also potentiated NGF-induced MAPK activation, with a similar dose-dependency to that necessary for potentiating neurite outgrowth. Taken together, these data suggest that FK1706 is a non-immunosuppressive immunophilin ligand with significant neurotrophic effects, putatively mediated via FKBP-52 and the Ras/Raf/MAPK signaling pathway, and therefore that FK1706 may have therapeutic potential in a variety of neurological disorders.


Subject(s)
Immunophilins/pharmacology , Nerve Growth Factors/pharmacology , Nerve Growth Factors/physiology , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Synergism , Humans , Immunophilins/chemistry , Immunophilins/metabolism , Mitogen-Activated Protein Kinases/drug effects , Mitogen-Activated Protein Kinases/metabolism , Nerve Growth Factors/antagonists & inhibitors , Receptor, trkB/metabolism , Signal Transduction/physiology , Tacrolimus/analogs & derivatives , Tacrolimus/chemistry , Tacrolimus/immunology , Tacrolimus/metabolism , Tacrolimus/pharmacology , Tacrolimus Binding Protein 1A/chemistry , Tacrolimus Binding Protein 1A/metabolism , Tacrolimus Binding Protein 1A/pharmacology , Tritium
13.
Eur J Pharmacol ; 458(1-2): 197-200, 2003 Jan 01.
Article in English | MEDLINE | ID: mdl-12498926

ABSTRACT

Inhibition of colonic propulsive motility is the main contributor to postoperative ileus in humans. Experimental models for investigating colonic propulsion in surgically induced postoperative ileus have not previously been available. This study was designed to assess whether adenosine A(1) receptor antagonists (R)-1-[(E)-3-(2-phenylpyrazolo[1,5-a]pyridin-3-yl) acryloyl]-piperidin-2-yl acetic acid (FK352) and 8-cyclopentyl-1, 3-dipropylxanthine (DPCPX) reverse the colonic motility disorder in newly developed rat experimental ileus models. When rats underwent anesthesia (pentobarbital) or surgical traumas (partial gastrectomy, cecectomy or gentle touching of the colon with fingers), colonic propulsive motility was evaluated by migration of intracolonically injected dye in awake unrestrained rats. Propulsive motility resulted in significant decrease after the treatment of the anesthesia or partial gastrectomy. Intravenous administration of either adenosine A(1) receptor antagonist reversed the slowed colonic propulsion in these experimental ileus models. The present study suggests that the blockade of adenosine A(1) receptors has therapeutic potential for postoperative ileus.


Subject(s)
Colon/drug effects , Intestinal Obstruction/prevention & control , Purinergic P1 Receptor Antagonists , Pyrazoles/pharmacology , Pyridines/pharmacology , Anesthesia , Animals , Cecostomy , Colon/physiopathology , Dose-Response Relationship, Drug , Gastrectomy , Gastrointestinal Motility/drug effects , Gastrointestinal Transit/drug effects , Intestinal Obstruction/etiology , Male , Postoperative Complications , Rats , Rats, Sprague-Dawley , Xanthines/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...