Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 78
Filter
Add more filters










Publication year range
1.
J Med Chem ; 44(12): 1986-92, 2001 Jun 07.
Article in English | MEDLINE | ID: mdl-11384243

ABSTRACT

The prevention of xenograft rejection is substantially dependent on inhibiting antibodies (Ab) produced by B-cells independently of T-cell signals (TI-1). Due to their ubiquitous biochemical mechanisms of action, the immunosuppressants currently employed not only fail to discriminate between B- and T-cells but also have a narrow therapeutic window and, thus, their prolonged use in complex immunosuppressive regimens is problematic. By capitalizing on the target enzyme-bound (DHODH) structure 1b of one of these compounds, leflunomide, and modulating part of its multiple mechanisms of action to gain selectivity, the quinoline-8-carboxamide 3 was designed as a potentially weak enzyme inhibitor but effective immunosuppressant. Compound 3 fulfilled the mechanistic criteria set and had 10-fold B-cell over T-cell selectivity. Its pyridyl analogue 4 was found to be a highly potent and selective B-cell immunosuppressant with a 75-fold selectivity for B- over T-cells (as judged by the MLR data) and no general cytotoxicity at concentrations up to 160-fold higher than those required to inhibit B-cells. In the mouse, 4 effectively blocked TI-1 Ab production and suppressed Ab-mediated xenograft rejection in a xenotransplantation model under a once-daily dosing regimen, with efficacy down to 0.3 mg/kg/day po. These are the first data demonstrating the feasibility of the development of drugs specific for impeding Ab production.


Subject(s)
B-Lymphocytes/drug effects , Graft Rejection/prevention & control , Immunosuppressive Agents/chemical synthesis , Quinolines/chemical synthesis , Transplantation, Heterologous/immunology , Animals , Antibody Formation/drug effects , B-Lymphocytes/immunology , Cricetinae , Drug Design , Graft Survival/drug effects , Heart Transplantation/immunology , Immunosuppressive Agents/blood , Immunosuppressive Agents/chemistry , Immunosuppressive Agents/pharmacology , Lipopolysaccharides/pharmacology , Mesocricetus , Mice , Mice, Nude , Models, Molecular , Molecular Conformation , Quinolines/chemistry , Quinolines/pharmacokinetics , Quinolines/pharmacology , Structure-Activity Relationship , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
3.
Transplantation ; 69(4): 488-96, 2000 Feb 27.
Article in English | MEDLINE | ID: mdl-10708100

ABSTRACT

BACKGROUND: In previous studies of cynomolgus monkey lung allograft recipients, we demonstrated significant immunosuppressive efficacy but reduced tolerability after combined treatment with high doses of microemulsion cyclosporine (CsA) and SDZ RAD (40-O-(2-hydroxyethyl)-rapamycin). The current study was designed to compare efficacy and tolerability of a combination of low-dose CsA and high-dose SDZ RAD (CTL group) to triple therapy using the chimeric anti-interleukin-2 (IL-2) receptor (CD25) monoclonal antibody (mAb) basiliximab (anti-IL-2 receptor mAb) for induction therapy (basiliximab: 5 mg intravenously on days 0 and 4) plus low-dose CsA and low-dose SDZ RAD for maintenance immunosuppression (CD25 group). CsA and anti-IL-2 receptor mAb are drugs that reduce cytokine synthesis and block IL-2-mediated lymphocyte stimulation, respectively. SDZ RAD blocks lymphocyte stimulation by other cytokines (e.g., IL-15) that are not inhibited by anti-IL-2 receptor mAb. METHODS: Twelve unilateral lung transplants were performed. Recipients were observed for 49 days by daily weight assessment, hemograms, blood chemistries, radiographs, and lung biopsies. Monkeys were euthanized before day 49 in the event of excessive weight loss (>25%) or organ failure. Target CsA trough levels were 100-200 ng/ml. Target SDZ RAD trough levels in the CTL group (no mAb) were 20-40 ng/ml, and 10-20 ng/ml in the CD25 group. RESULTS: None of the monkeys in the CD25 group needed to be euthanized early due to signs of drug toxicity. In contrast, four monkeys in the CTL group were sacrificed on days 28-35 as a result of excessive weight loss (n=3) and renal functional impairment (n=1). Three recipients in the CD25 group were euthanized on days 36, 38, and 46 as a result of persistent high fever associated with severe rejection. The median animal survival in the CTL group was 32 vs. 46 days in the CD25 group (P<0.04). The only two long-term survivors in the CTL group showed moderate rejection at day 49. The median rejection scores at day 14 (A0) and day 28 (A2) were identical in the two groups, despite the fact that the mean SDZ RAD trough level was significantly lower in the CD25 group (CTL: 38+/-3 ng/ml, CD25: 18+/-2 ng/ml, P<0.0001). After basiliximab levels fell below the minimum therapeutic level (1 mg/ml) on day 28, the median rejection score at day 49 increased to A4 in the CD25 group. CONCLUSION: This is the first study to combine an anti-IL-2 receptor mAb with a drug from the rapamycin class plus CsA. Our study shows that induction therapy with basiliximab enabled SDZ RAD blood levels to be significantly reduced, which led to improved tolerability without the penalty of increased rejection.


Subject(s)
Lung Transplantation/immunology , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/therapeutic use , Autopsy , Basiliximab , Biopsy , Body Weight , Bronchoscopy , Creatinine/blood , Cyclosporine/administration & dosage , Cyclosporine/pharmacokinetics , Cyclosporine/therapeutic use , Dose-Response Relationship, Drug , Drug Synergism , Drug Therapy, Combination , Emulsions , Everolimus , Graft Rejection/prevention & control , Immune Tolerance/drug effects , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/therapeutic use , Lung/pathology , Macaca fascicularis , Male , Microchemistry , Postoperative Period , Receptors, Interleukin-2/immunology , Recombinant Fusion Proteins/immunology , Sirolimus/administration & dosage , Sirolimus/analogs & derivatives , Sirolimus/therapeutic use , Tissue Donors
5.
J Med Chem ; 41(18): 3530-8, 1998 Aug 27.
Article in English | MEDLINE | ID: mdl-9719606

ABSTRACT

T-cell immunosuppressant-based therapies efficiently control early graft rejection in allotransplantation settings. They fail, however, to prevent those rejection events which are mediated by transplant-induced antibody (Ab) responses such as those involved in xenograft and chronic allograft rejection. This is mainly due to their inability to block T-cell-independent Ab production against the transplanted organs. The bioactive metabolite 2(Z) of leflunomide (1) inhibits the formation of such Ab, but the drug has pharmacokinetic properties and a therapeutic window incompatible with transplantation indications. Pyrazole 3, a constrained analogue of 2(Z), was designed and shown to be conformationally and biologically similar to 2(Z). Further investigations with derivatives of 3 demonstrated that the pyrazoles had very tight structure-activity relationships, the only equipotent compound being 3o. However, in contrast to 2(Z), both 3 and 3o were inactive in vivo due to short half-life and drug concentrations lower than the in vitro obtained IC50 values. Compound 3o inhibits T-cell-independent Ab production by a different biochemical mechanism from that of 2(Z) and 3 and may therefore represent a valuable tool for the identification of new targets for B-cell inhibition.


Subject(s)
B-Lymphocytes/drug effects , Graft Rejection/prevention & control , Immunosuppressive Agents , Isoxazoles/chemistry , Oxidoreductases Acting on CH-CH Group Donors , Pyrazoles , Transplantation, Heterologous/immunology , Administration, Oral , Animals , Antigens, T-Independent/immunology , B-Lymphocytes/immunology , Cell Division/drug effects , Cell Division/immunology , Dihydroorotate Dehydrogenase , Graft Rejection/immunology , Humans , Immunoglobulin G/immunology , Immunoglobulin M/immunology , Immunosuppressive Agents/chemical synthesis , Immunosuppressive Agents/chemistry , Immunosuppressive Agents/pharmacokinetics , Immunosuppressive Agents/pharmacology , In Vitro Techniques , Injections, Intravenous , Isoxazoles/pharmacology , Jurkat Cells/cytology , Jurkat Cells/immunology , Leflunomide , Lipopolysaccharides/immunology , Lymphocyte Culture Test, Mixed , Mice , Oxidoreductases/antagonists & inhibitors , Pyrazoles/administration & dosage , Pyrazoles/chemical synthesis , Pyrazoles/pharmacokinetics , Pyrazoles/pharmacology , Structure-Activity Relationship
6.
Bioorg Med Chem Lett ; 8(16): 2203-8, 1998 Aug 18.
Article in English | MEDLINE | ID: mdl-9873513

ABSTRACT

A library of thioamide derivatives of leflunomide 1a and of its bioactive metabolite 1b has been synthesised on solid phase. Thus, para-substituted phenylacetic acids were coupled to TentaGel and were subsequently reacted with aromatic isothiocyanates. Treatment of the resulting enaminothioamides with hydroxylamine led to their simultaneous cyclisation and cleavage from the resin affording 2-25. Their in vitro profiling demonstrated that the amide-thioamide isologous substitution was detrimental of the biological activity.


Subject(s)
Immunosuppressive Agents/chemical synthesis , Isothiocyanates/chemical synthesis , Isoxazoles/chemical synthesis , Oxidoreductases Acting on CH-CH Group Donors , Animals , Dihydroorotate Dehydrogenase , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , Hydroxylamine , Immunosuppressive Agents/chemistry , Immunosuppressive Agents/pharmacology , Indicators and Reagents , Isothiocyanates/chemistry , Isothiocyanates/pharmacology , Isoxazoles/chemistry , Isoxazoles/pharmacology , Leflunomide , Lymphocyte Culture Test, Mixed , Lymphocytes/drug effects , Lymphocytes/immunology , Mice , Molecular Structure , Oxidoreductases/antagonists & inhibitors , Phenylacetates/chemistry , Recombinant Proteins/antagonists & inhibitors , Spectrometry, Mass, Fast Atom Bombardment , Structure-Activity Relationship
7.
Transplantation ; 64(1): 36-42, 1997 Jul 15.
Article in English | MEDLINE | ID: mdl-9233698

ABSTRACT

BACKGROUND: This report describes the preclinical pharmacological profile of the new rapamycin analog, SDZ RAD, i.e., 40-O-(2-hydroxyethyl)-rapamycin. METHODS: The pharmacological effects of SDZ RAD were assessed in a variety of in vitro and in vivo models, which included an autoimmune disease model as well as kidney and heart allotransplantation models using different rat strain combinations. RESULTS: SDZ RAD has a mode of action that is different from that of cyclosporine or FK506. In contrast to the latter, SDZ RAD inhibits growth factor-driven cell proliferation in general, as demonstrated for the in vitro cell proliferation of a lymphoid cell line and of vascular smooth muscle cells. SDZ RAD is immunosuppressive in vitro as demonstrated by the inhibition of mouse and human mixed lymphocyte reactions and the inhibition of antigen-driven proliferation of human T-cell clones. The concentrations needed to achieve 50% inhibition in all of these assays fall into the subnanomolar range. SDZ RAD is effective in the in vivo models when given by the oral route in doses ranging between 1 mg/kg/day and 5 mg/kg/day. When compared with rapamycin, the in vitro activity of SDZ RAD is generally about two to three times lower; however, when administered orally, SDZ RAD is at least as active in vivo as rapamycin. CONCLUSIONS: In conclusion, SDZ RAD is a new, orally active rapamycin-derivative that is immunosuppressive and that efficiently prevents graft rejection in rat models of allotransplantation. SDZ RAD has therefore been selected for development for use in combination with cyclosporine A to prevent acute and chronic rejection after solid organ allotransplantation.


Subject(s)
Immunosuppressive Agents/pharmacology , Polyenes/pharmacology , Abdomen , Administration, Oral , Animals , Carrier Proteins/metabolism , Cell Division/drug effects , DNA-Binding Proteins/metabolism , Dose-Response Relationship, Drug , Everolimus , Glomerulonephritis/chemically induced , Graft Rejection/prevention & control , Graft vs Host Reaction/drug effects , Growth Inhibitors/physiology , Growth Substances/pharmacology , Heat-Shock Proteins/metabolism , Humans , Kidney Transplantation , Mercuric Chloride , Protein Binding , Rats , Rats, Inbred BN , Rats, Inbred F344 , Rats, Wistar , Sirolimus/analogs & derivatives , Tacrolimus Binding Proteins , Transplantation, Heterotopic , Transplantation, Homologous/immunology
8.
J Immunol ; 158(5): 2126-33, 1997 Mar 01.
Article in English | MEDLINE | ID: mdl-9036957

ABSTRACT

TNF is implicated in in vitro Ig production, but its role in vivo is not clearly defined. Our previous studies had shown that TNF-LT alpha double-deficient mice have defective IgM and IgG primary Ab responses to the T cell-dependent (TD) Ag SRBC. We now extend these studies to secondary responses and to T cell-independent (TI) B cell responses. Injections of the TD Ag SRBC did not induce germinal center formation in the spleen of TNF-LT alpha-deficient mice. Associated with the morphologic defect, there was a defective IgG Ab response and a secondary hyper-IgM response to the TD Ag in TNF-LT alpha-deficient mice. The response to the TI Ag type 2 DNP-alanyl-glycyl-glycyl-Ficoll was essentially absent in TNF-LT alpha-deficient mice, while that to the TI Ag type 1 TNP-LPS was significantly reduced only for IgG2b isotype. Transplantation of bone marrow cells from wild-type mice into irradiated TNF-LT alpha-deficient mice restored the formation of splenic germinal centers and corrected the IgM and IgG responses to both TD and TI Ags. These data suggest that TNF and/or LT alpha signaling are critically required for germinal center formation and for the IgM and IgG responses to both TD and TI type 2 Ags.


Subject(s)
Antigens, T-Independent/immunology , B-Lymphocytes/immunology , Ficoll/analogs & derivatives , Immunoglobulin Class Switching/immunology , Lymphotoxin-alpha/genetics , Tumor Necrosis Factor-alpha/deficiency , Animals , Antibody Formation/genetics , B-Lymphocytes/metabolism , Bone Marrow Transplantation , Erythrocytes/immunology , Ficoll/immunology , Germinal Center/metabolism , Immunization , Mice , Mice, Inbred C57BL , Mice, Knockout , Sheep , Tumor Necrosis Factor-alpha/genetics
9.
Transplantation ; 60(7): 748-56, 1995 Oct 15.
Article in English | MEDLINE | ID: mdl-7570988

ABSTRACT

A high affinity chimeric CD25 mAb (chRFT5: SDZ CHI 621) blocking interleukin-2 binding to the interleukin-2 receptor alpha-chain was evaluated in a phase I/II study in human renal cadaveric transplantation. The chRFT5 was well tolerated with no immediate adverse effects during 6 spaced infusions (from before transplantation to day 24) in 24 patients escalating from 2.5- to 25-mg dosages. The chRFT5 had a long terminal half-life with a mean of 13.1 days. There was good correlation between the detection of chRFT5 in the serum by radioimmunoassay, the coating and suppression of CD25 on T cells, and antibody activity in patient serum samples. The chRFT5 activity persisted in vivo for up to 120 days. No antibody response to the chRFT5 was detected in any of the patients, even though two patients who required treatment with antithymocyte globulin or OKT3 developed xenogeneic antiglobulin responses while chRFT5 was still present in vivo. There was a 33% incidence of rejection and the first rejection episode always occurred during chRFT5 therapy. Patients who did not reject during therapy did not reject during the first year following transplantation. Equal numbers of patients received dual and triple immunosuppressive therapy together with chRFT5. Posttransplant lymphoproliferative disorder developed in 2 patients, both on triple therapy, at 9 months after transplantation. The disorder did not develop in any patient receiving dual therapy, and no further cases have been observed to a minimum of 2 years' follow-up. No other viral, fungal, or bacterial infectious complications were prevalent in patients treated with chRFT5.


Subject(s)
Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/therapeutic use , Kidney Transplantation , Receptors, Interleukin-2/immunology , Recombinant Fusion Proteins/therapeutic use , Adult , Antibodies, Monoclonal/pharmacokinetics , Antibody Formation , Female , Graft Rejection/prevention & control , Humans , Male , Middle Aged , Receptors, Interleukin-2/metabolism , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacokinetics
10.
Eur J Immunol ; 25(2): 474-80, 1995 Feb.
Article in English | MEDLINE | ID: mdl-7533086

ABSTRACT

To study the role of perforin in cell-mediated graft rejection, vascularized hearts were grafted to perforin-deficient C57BL/6 and control C57BL/6 recipient mice. Fully allogeneic heart grafts (BALB/c) were acutely rejected by both recipients within 6 days. Peritoneal exudate lymphocytes from control mice but not from perforin-deficient mice exhibit a strong alloreactive cytotoxic activity in vitro. Histological analysis of the rejected tissues demonstrated extensive mononuclear cell infiltrates in both recipients. Flow cytometry analysis and immunohistology of graft-infiltrating cells showed similar proportions of lymphocyte subsets (CD8 >> CD4). Collectively, these data indicate that perforin is not essential in the cell-mediated acute rejection of a fully mismatched heart allograft. However, perforin-dependent effector mechanisms appeared to be limiting in the T cell-mediated rejection of heart allografts differing only at a single major histocompatibility complex class I antigen (bm1), because these grafts survived longer (mean 87.8 days) in perforin-deficient than in control mice (mean 31.5 days).


Subject(s)
Graft Rejection , Heart Transplantation/immunology , Membrane Glycoproteins/deficiency , Animals , Antigens, Surface/physiology , Cytotoxicity, Immunologic , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Myocardium/pathology , Perforin , Pore Forming Cytotoxic Proteins , T-Lymphocyte Subsets/immunology , Transplantation, Homologous , fas Receptor
12.
Blood ; 84(5): 1543-52, 1994 Sep 01.
Article in English | MEDLINE | ID: mdl-7520778

ABSTRACT

The immunosuppressive drug rapamycin suppresses T-cell activation by impairing the T-cell response to lymphokines such as interleukin-2 (IL-2) and interleukin-4 (IL-4). In addition, rapamycin blocks the proliferative response of cell lines to a variety of hematopoietic growth factors, including interleukin-3 (IL-3), interleukin-6 (IL-6), granulocyte-colony stimulating factor (G-CSF), granulocyte macrophage-colony stimulating factor (GM-CSF), and kit ligand (KL), suggesting that it should be a strong inhibitor of hematopoiesis. In this report, we studied the effects of rapamycin on different hematopoietic cell populations in vitro and in vivo. In vitro, rapamycin inhibited the proliferation of primary bone marrow cells induced by IL-3, GM-CSF, KL, or a complex mixture of factors present in cell-conditioned media. Rapamycin also inhibited the multiplication of colony-forming cells in suspension cultures containing IL-3 plus interleukin-1 (IL-1) or interleukin-11 (IL-11) plus KL. In vivo, treatment for 10 to 28 days with high doses of rapamycin (50 mg/kg/d, orally) had no effect on myelopoiesis in normal mice, as measured by bone marrow cellularity, proliferative capacity, and number of colony-forming progenitors. In contrast, the same treatment strongly suppressed the hematopoietic recovery normally seen 10 days after an injection of 5-fluorouracil (5-FU; 150 mg/kg, intravenously [i.v.]). Thus, rapamycin may be detrimental in myelocompromised individuals. In addition, the results suggest that the rapamycin-sensitive cytokine-driven pathways are essential for hematopoietic recovery after myelodepression, but not for steady-state hematopoiesis.


Subject(s)
Bone Marrow Cells , Cell Division/drug effects , Cytokines/pharmacology , Heart Transplantation/immunology , Hematopoiesis/drug effects , Hematopoietic Stem Cells/cytology , Immunosuppressive Agents/pharmacology , Polyenes/pharmacology , Animals , Bone Marrow/drug effects , Cell Line , Culture Media, Conditioned , Cytokines/antagonists & inhibitors , Female , Fluorouracil/pharmacology , Granulocyte Colony-Stimulating Factor/pharmacology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Hematopoietic Cell Growth Factors/pharmacology , Hematopoietic Stem Cells/drug effects , Humans , Interleukin-1/pharmacology , Interleukin-3/pharmacology , Lymphocyte Activation/drug effects , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Recombinant Proteins/pharmacology , Reference Values , Sirolimus , Stem Cell Factor , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Transplantation, Homologous
15.
Infect Immun ; 61(9): 3863-72, 1993 Sep.
Article in English | MEDLINE | ID: mdl-8359907

ABSTRACT

During the last decade, episodes of sepsis have increased and Escherichia coli has remained the most frequent clinical isolate. Lipopolysaccharides (LPS; endotoxin) are the major toxic and antigenic components of gram-negative bacteria and qualify as targets for therapeutic interventions. Molecules that neutralize the toxic effects of LPS are actively investigated. In this paper, we describe a murine monoclonal antibody (MAb; WN1 222-5), broadly cross-reactive and cross-protective for smooth (S)-form and rough (R)-form LPS. As shown in enzyme-linked immunosorbent assay and the passive hemolysis assay, WN1 222-5 binds to the five known E. coli core chemotypes, to Salmonella core, and to S-form LPS having these core structures. In immunoblots, it is shown to react with both the nonsubstituted core LPS and with LPS carrying O-side chains, indicating the exposure of the epitope in both S-form and R-form LPS. This MAb of the immunoglobulin G2a class is not lipid A reactive but binds to E. coli J5, an RcP+ mutant which carries an inner core structure common to many members of the family Enterobacteriaceae. Phosphate groups present in the inner core contribute to the epitope but are not essential for the binding of WN1 222-5 to complete core LPS. Cross-reactivity for clinical bacterial isolates is broad. WN1 222-5 binds to all E. coli clinical isolates tested so far (79 blood isolates, 80 urinary isolates, and 21 fecal isolates) and to some Citrobacter, Enterobacter, and Klebsiella isolates. This pattern of reactivity indicates that its binding epitope is widespread among members of the Enterobacteriaceae. WN1 222-5 exhibits biologically relevant activities. In vitro, it inhibits the Limulus amoebocyte lysate assay activity of S-form and R-form LPS in a dose-dependent manner and it neutralizes the LPS-induced release of clinically relevant monokines (interleukin 6 and tumor necrosis factor). In vivo, WN1 222-5 blocks endotoxin-induced pyrogenicity in rabbits and lethality in galactosamine-sensitized mice. The discovery of WN1 222-5 settles the long-lasting controversy over the existence of anti-core LPS MAbs with both cross-reactive and cross-protective activity, opening new possibilities for the immunotherapy of sepsis caused by gram-negative bacteria.


Subject(s)
Antibodies, Monoclonal/immunology , Escherichia coli/immunology , Immunoglobulin G/immunology , Lipopolysaccharides/immunology , Salmonella/immunology , Animals , Cross Reactions , Enzyme-Linked Immunosorbent Assay , Female , Hemolysis , Immunoblotting , Lipopolysaccharides/toxicity , Mice , Mice, Inbred C57BL , Mice, Inbred NZB , Monokines/metabolism , Rabbits
16.
Ann N Y Acad Sci ; 685: 330-5, 1993 Jun 23.
Article in English | MEDLINE | ID: mdl-7689806

ABSTRACT

Despite the successful clinical application of the immunosuppressive drug cyclosporin A (CsA, Sandimmun), its precise mechanism of action in the process of T cell activation remains elusive. CsA binds to the high-affinity cytosolic receptor cyclophilin whose peptidyl-prolyl cis-trans isomerase activity is inhibited upon binding. The linkage of this effect with the inhibition of the T cell receptor-mediated signal transduction pathway, which leads to a suppression of lymphokine gene transcription, is still unclear. We analyzed the relationship between cyclophilin-binding and immunosuppressive activity (e.g., effect on IL-2 transcription) of cyclosporin derivatives in vitro. The results show that binding to cyclophilin is required, but not sufficient for immunosuppression. Cyclosporin analogues which completely lack immunosuppressive activity but fully retained their cyclophilin-binding capacity antagonize the immunosuppressive activity of CsA. These derivatives inhibit the isomerase activity of cyclophilin, which clearly demonstrates that inhibition of the cyclophilin isomerase activity does not lead to immunosuppression. In analogy to the other immunosuppressants of microbial origin, FK-506 and rapamycin, a specific structure of the "effector" domain of CsA, which is unrelated to the cyclophilin-binding domain, determines the biological activity. In the nucleus, CsA interferes with the DNA-binding of inducible transcription factors to their respective DNA motifs within lymphokine promoters by affecting intracellular translocation of transcription factor subunits.


Subject(s)
Cyclosporins/pharmacology , Immune Tolerance , T-Lymphocytes/drug effects , Amino Acid Isomerases/metabolism , Animals , Carrier Proteins/metabolism , Cyclosporins/metabolism , Humans , Lymphocyte Activation/drug effects , Peptidylprolyl Isomerase , T-Lymphocytes/immunology , Tacrolimus/pharmacology
19.
Br J Haematol ; 82(1): 6-12, 1992 Sep.
Article in English | MEDLINE | ID: mdl-1384647

ABSTRACT

Human serum induces human peripheral blood leucocytes (PBL) to release an activity stimulating neutrophil colony formation (G-CSA) from human bone marrow cells. By titrating individual growth factors and using specific neutralizing antibodies we showed that: human serum contains very low levels of G-CSF which are by themselves insufficient to stimulate myeloid colony formation in primary human bone marrow cultures and cannot account for the serum releaser activity; that although no detectable levels of IL-1, IL-2, IL-3, IL-4, IL-6 or IL-8 are found in the serum, anti IL-1 antibodies partially block the release of G-CSA when added early during PBL incubation; that PBL incubated in the absence of serum for 2 d produce small amounts of IL-1, IL-6, IL-8 and G-CSF and this is increased 6-16 fold in the presence of human serum; and that the neutrophil colony-stimulating activity released by PBL incubated with human serum is G-CSF.


Subject(s)
Granulocyte Colony-Stimulating Factor/blood , Interleukins/biosynthesis , Leukocytes/immunology , Biological Assay , Blood , Cells, Cultured , Colony-Forming Units Assay , Humans , Interleukin-1/biosynthesis , Interleukin-6/biosynthesis , Interleukin-8/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...