Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Br J Pharmacol ; 173(11): 1778-92, 2016 06.
Article in English | MEDLINE | ID: mdl-26990079

ABSTRACT

BACKGROUND AND PURPOSE: Sphingosine1-phosphate (S1P) receptors mediate multiple events including lymphocyte trafficking, cardiac function, and endothelial barrier integrity. Stimulation of S1P1 receptors sequesters lymphocyte subsets in peripheral lymphoid organs, preventing their trafficking to inflamed tissue sites, modulating immunity. Targeting S1P receptors for treating autoimmune disease has been established in clinical studies with the non-selective S1P modulator, FTY720 (fingolimod, Gilenya™). The purpose of this study was to assess RPC1063 for its therapeutic utility in autoimmune diseases. EXPERIMENTAL APPROACH: The specificity and potency of RPC1063 (ozanimod) was evaluated for all five S1P receptors, and its effect on cell surface S1P1 receptor expression, was characterized in vitro. The oral pharmacokinetic (PK) parameters and pharmacodynamic effects were established in rodents, and its activity in three models of autoimmune disease (experimental autoimmune encephalitis, 2,4,6-trinitrobenzenesulfonic acid colitis and CD4(+) CD45RB(hi) T cell adoptive transfer colitis) was assessed. KEY RESULTS: RPC1063 was specific for S1P1 and S1P5 receptors, induced S1P1 receptor internalization and induced a reversible reduction in circulating B and CCR7(+) T lymphocytes in vivo. RPC1063 showed high oral bioavailability and volume of distribution, and a circulatory half-life that supports once daily dosing. Oral RPC1063 reduced inflammation and disease parameters in all three autoimmune disease models. CONCLUSIONS AND IMPLICATIONS: S1P receptor selectivity, favourable PK properties and efficacy in three distinct disease models supports the clinical development of RPC1063 for the treatment of relapsing multiple sclerosis and inflammatory bowel disease, differentiates RPC1063 from other S1P receptor agonists, and could result in improved safety outcomes in the clinic.


Subject(s)
Autoimmune Diseases/drug therapy , Indans/pharmacology , Inflammatory Bowel Diseases/drug therapy , Oxadiazoles/pharmacology , Receptors, Lysosphingolipid/agonists , Animals , Autoimmune Diseases/chemically induced , Disease Models, Animal , Female , Inflammatory Bowel Diseases/chemically induced , Male , Mice , Mice, Inbred C57BL , Mice, SCID , Myelin-Oligodendrocyte Glycoprotein/immunology , Rats , Rats, Sprague-Dawley , Trinitrobenzenesulfonic Acid
2.
Cell Death Differ ; 15(4): 708-17, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18202705

ABSTRACT

Human GraB (hGraB) preferentially induces apoptosis via Bcl-2-regulated mitochondrial damage but can also directly cleave caspases and caspase substrates in cell-free systems. How hGraB kills cells when it is delivered by cytotoxic lymphocytes (CL) and the contribution of hGraB to CL-induced death is still not clear. We show that primary human natural killer (hNK) cells, which specifically used hGraB to induce target cell death, were able to induce apoptosis of cells whose mitochondria were protected by Bcl-2. Purified hGraB also induced apoptosis of Bcl-2-overexpressing targets but only when delivered at 5- to 10-fold the concentration required to kill cells expressing endogenous Bcl-2. Caspases were critical in this process as inhibition of caspase activity permitted clonogenic survival of Bcl-2-overexpressing cells treated with hGraB or hNK cells but did not protect cells that only expressed endogenous Bcl-2. Our data therefore show that hGraB triggers caspase activation via mitochondria-dependent and mitochondria-independent mechanisms that are activated in a hierarchical manner, and that the combined effects of Bcl-2 and direct caspase inhibition can block cell death induced by hGraB and primary hNK cells.


Subject(s)
Apoptosis , Caspases/metabolism , Granzymes/metabolism , Killer Cells, Natural/enzymology , Mitochondria/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Secretory Vesicles/enzymology , Amino Acid Chloromethyl Ketones/pharmacology , Apoptosis/drug effects , Caspase Inhibitors , Cell Culture Techniques , Cells, Cultured , Cysteine Proteinase Inhibitors/pharmacology , Dipeptides/pharmacology , Enzyme Activation , Granzymes/antagonists & inhibitors , Granzymes/genetics , HeLa Cells , Humans , Killer Cells, Natural/drug effects , Mitochondria/enzymology , Mitochondrial Membranes/metabolism , Permeability , Protease Inhibitors/pharmacology , Proto-Oncogene Proteins c-bcl-2/genetics , Secretory Vesicles/drug effects , Time Factors , Transfection , X-Linked Inhibitor of Apoptosis Protein/metabolism
3.
Proc Natl Acad Sci U S A ; 98(25): 14250-5, 2001 Dec 04.
Article in English | MEDLINE | ID: mdl-11734640

ABSTRACT

A critical step in the induction of apoptosis is the activation of the apoptotic initiator caspase 9. We show that at its normal physiological concentration, caspase 9 is primarily an inactive monomer (zymogen), and that activity is associated with a dimeric species. At the high concentrations used for crystal formation, caspase 9 is dimeric, and the structure reveals two very different active-site conformations within each dimer. One site closely resembles the catalytically competent sites of other caspases, whereas in the second, expulsion of the "activation loop" disrupts the catalytic machinery. We propose that the inactive domain resembles monomeric caspase 9. Activation is induced by dimerization, with interactions at the dimer interface promoting reorientation of the activation loop. These observations support a model in which recruitment by Apaf-1 creates high local concentrations of caspase 9 to provide a pathway for dimer-induced activation.


Subject(s)
Caspases/chemistry , Caspases/metabolism , Apoptosis , Caspase 9 , Catalytic Domain , Dimerization , Enzyme Activation , Humans , In Vitro Techniques , Kinetics , Models, Molecular , Protein Conformation , Protein Structure, Quaternary , Protein Structure, Tertiary
4.
DNA Cell Biol ; 20(3): 123-31, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11313015

ABSTRACT

Hurpin (protease inhibitor 13; PI13) is the most recently identified member of the ovalbumin family of serine protease inhibitors (serpins). It is expressed in human epidermal keratinocytes and is downregulated by exposure to ultraviolet irradiation. A role for hurpin in the proliferation or differentiation of keratinocytes has been proposed because of its strong expression in proliferating cells and its deregulated expression in the lesional epidermis of psoriatic patients. Here, we report the cloning, chromosomal localization, and complete sequence of the human hurpin gene. By PCR-based screening of the GeneBridge 4 radiation hybrid panel, we mapped the gene to chromosome 18q21.3, close to a known cluster of ov-serpin genes. Using the full-length cDNA for hurpin, we identified two clones from an arrayed genomic P1 placental library that contain the entire hurpin gene. Sequencing revealed that the gene covers 12.253 kb and is comprised of eight exons and seven introns. The exon--intron boundaries are identical in position and phasing to those in other members of the 18q serpin gene cluster, and analysis of hurpin variants indicated that modified functional inhibitors, differing only in the CD interhelical loop, can be generated by differential splicing of exon 3. These data show that hurpin is a typical member of the 18q ovalbumin-serpins most closely related to the serpins squamous-cell carcinoma antigens 1 and 2.


Subject(s)
Alternative Splicing , Chromosomes, Human, Pair 18 , Psoriasis/metabolism , Serpins/genetics , Amino Acid Sequence , Base Sequence , Cloning, Molecular , Exons , Humans , Introns , Molecular Sequence Data , Plasminogen Activator Inhibitor 2/genetics , RNA, Messenger , Serpins/metabolism , Transcription, Genetic , Up-Regulation
5.
Blood ; 93(6): 2089-97, 1999 Mar 15.
Article in English | MEDLINE | ID: mdl-10068683

ABSTRACT

The monocyte and granulocyte azurophilic granule proteinases elastase, proteinase 3, and cathepsin G are implicated in acute and chronic diseases thought to result from an imbalance between the secreted proteinase(s) and circulating serpins such as alpha1-proteinase inhibitor and alpha1-antichymotrypsin. We show here that the intracellular serpin, proteinase inhibitor 6 (PI-6), is present in monocytes, granulocytes, and myelomonocytic cell lines. In extracts from these cells, PI-6 bound an endogenous membrane-associated serine proteinase to form an sodium dodecyl sulfate (SDS)-stable complex. Using antibodies to urokinase, elastase, proteinase 3, or cathepsin G, we demonstrated that the complex contains cathepsin G. Native cathepsin G and recombinant PI-6 formed an SDS-stable complex in vitro similar in size to that observed in the extracts. Further kinetic analysis demonstrated that cathepsin G and PI-6 rapidly form a tight 1:1 complex (ka = 6.8 +/- 0.2 x 10(6) mol/L-1s-1 at 17 degrees C; Ki = 9.2 +/- 0.04 x 10(-10) mol/L). We propose that PI-6 complements alpha1-proteinase inhibitor and alpha1-antichymotrypsin (which control extracellular proteolysis) by neutralizing cathepsin G that leaks into the cytoplasm of monocytes or granulocytes during biosynthesis or phagocytosis. Control of intracellular cathepsin G may be particularly important, because it has recently been shown to activate the proapoptotic proteinase, caspase-7.


Subject(s)
Cathepsins/antagonists & inhibitors , Granulocytes/chemistry , Monocytes/chemistry , Serpins/analysis , Cathepsin G , Cathepsins/metabolism , Cell Membrane/enzymology , Cytoplasmic Granules/chemistry , Dimerization , HL-60 Cells/chemistry , Humans , K562 Cells/chemistry , Kinetics , Recombinant Proteins/metabolism , Serine Endopeptidases/metabolism , Serpins/metabolism , Sodium Dodecyl Sulfate/pharmacology , U937 Cells/chemistry
6.
Genomics ; 62(3): 490-9, 1999 Dec 15.
Article in English | MEDLINE | ID: mdl-10644448

ABSTRACT

The human ovalbumin (ov) serpins are associated with tumorigenesis, inflammation, and protection from autolysis by granule proteinases. Their genes are located at 18q21 or 6p25, falling into two structurally very similar but distinct categories depending on the presence or absence of a particular exon. Analysis of ov-serpin gene structure provides an opportunity to elucidate the mechanisms contributing to the formation of the larger serpin gene superfamily. Here we have identified a new gene (PI8L1) at 6p25 that is 72% identical to the 18q21 gene PI8. FISH analysis using the 3' untranslated region of PI8 yielded an additional signal at 18q23, separable from the known 18q21.3 signal by the t(1;18)(p32;q23) chromosomal translocation. The presence of more than one PI8-related gene was confirmed by analysis of human genomic DNA using the same probe. Cloning and analysis of PI8 showed that its intron number and phasing are identical to those of the 6p25 genes PI6, PI9, and ELANH2, and it lacks the interhelical variable loop exon found in other 18q21 genes. PCR analysis demonstrated that PI5 at 18q21 also lacks this exon, indicating that it is organized identically to the 6p25 genes. By contrast, PI10 and megsin have this exon and resemble the other 18q21 genes, PLANH2, SCCA-1, and SCCA-2, in structure. Using these data with an ov-serpin phylogenic tree we have constructed, we propose that the ov-serpin gene clusters arose via interchromosomal duplication of PI5 (or a precursor) to 6p25, followed by duplication at 6p25, and a more recent interchromosomal duplication from 6p25 to 18q to yield PI8.


Subject(s)
Chromosomes, Human, Pair 18/genetics , Chromosomes, Human, Pair 6/genetics , Evolution, Molecular , Serpins/genetics , 3' Untranslated Regions/genetics , Base Sequence , Exons/genetics , Gene Duplication , Genes, Tumor Suppressor , Humans , In Situ Hybridization, Fluorescence , Introns/genetics , Molecular Sequence Data , Multigene Family , Ovalbumin/genetics , Phylogeny , Proteins/genetics , Sequence Homology, Amino Acid , Serine Proteinase Inhibitors/genetics
7.
Exp Cell Res ; 245(2): 263-71, 1998 Dec 15.
Article in English | MEDLINE | ID: mdl-9851866

ABSTRACT

Proteinase inhibitor 6 (PI-6) is a 42-kDa intracellular protein present in epithelial cells and endothelial cells. It is capable of inhibiting a number of serine proteinases, including trypsin and chymotrypsin. In this study we examined PI-6 expression in human skin and its primary cell type, the keratinocyte. By immunohistochemical analysis, PI-6 staining is absent from the basal cells, weak in the spinous layer, and strongest in the granulosa layer of human epidermis. Immunoblotting of cultured primary keratinocytes revealed that PI-6 production increases 24-fold on differentiation. Analysis of an immortalized keratinocyte cell line, HaCat, showed a 5-fold increase in PI-6 mRNA and a 7-fold increase in PI-6 protein upon differentiation, and indirect immunofluorescence revealed that this is due to an increase in the number of differentiated cells expressing high levels of PI-6. Of particular interest is the appearance of a preformed complex between PI-6 and an endogenous serine proteinase in differentiating HaCat cells, which was detected by a monoclonal antibody demonstrated to preferentially recognize PI-6 in complex with a proteinase. This identification of a PI-6/proteinase complex is the first example of a serpin bound to a proteinase in keratinocytes. We postulate that a physiological role of PI-6 is to regulate a serine proteinase associated with keratinocyte differentiation.


Subject(s)
Epidermal Cells , Keratinocytes/cytology , Serpins/metabolism , Animals , Antibodies, Monoclonal , Blotting, Northern , COS Cells , Cell Differentiation , Cell Line , Cell Size , Cells, Cultured , Cytosol/metabolism , Epidermis/enzymology , Epidermis/metabolism , Gene Expression Regulation , Humans , Keratinocytes/enzymology , Keratinocytes/metabolism , Keratins/metabolism , Molecular Weight , Multienzyme Complexes/metabolism , Protein Conformation , Serine Endopeptidases/metabolism , Serpins/chemistry , Serpins/genetics , Serpins/immunology , Thrombin/metabolism
9.
J Biol Chem ; 271(3): 1605-12, 1996 Jan 19.
Article in English | MEDLINE | ID: mdl-8576159

ABSTRACT

We have recently described a new serine proteinase inhibitor, proteinase inhibitor 6 (PI-6). This serpin has features that suggest it may function intracellularly, but its close resemblance to ovalbumin serpins like plasminogen activator inhibitor 2 (PAI-2) raises the possibility that it is secreted to regulate an extracellular proteinase. To determine whether PI-6 is secreted, we have examined its cellular distribution by immunohistochemistry and have attempted to induce its release from platelets and from cultured cells. We find that PI-6 is present in endothelial and epithelial cells, but it is apparently cytoplasmic and it is not released from cells in response to phorbol ester, dibutyryl cAMP or tumor necrosis factor alpha treatment. It is also not released from activated platelets. The addition of a conventional signal peptide to the amino terminus of PI-6 directed its translocation into the endoplasmic reticulum (ER), resulting in glycosylation but not secretion of the molecule. By contrast, the addition of the same signal peptide to PAI-2 markedly enhanced its translocation and secretion. Glycosylated PI-6 was sequestered in the ER and was incapable of interacting with thrombin. The failure of PI-6 to move along the secretory pathway, and the loss of inhibitory function of ER-localized PI-6, demonstrates that unlike PAI-2, PI-6 is not naturally secreted. Taken together, these results suggest that PI-6 has evolved to fulfil an intracellular role and that it represents a new type of cellular serpin.


Subject(s)
Blood Platelets/physiology , Serpins/biosynthesis , Skin/cytology , Amino Acid Sequence , Animals , Base Sequence , Blood Platelets/drug effects , Bucladesine/pharmacology , Cell Line , Chlorocebus aethiops , Cloning, Molecular , DNA Primers , DNA, Complementary , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/ultrastructure , Fluorescent Antibody Technique, Indirect , Glycosylation , Humans , Immunohistochemistry , In Vitro Techniques , Kinetics , Molecular Sequence Data , Platelet Activation/drug effects , Recombinant Proteins/analysis , Recombinant Proteins/biosynthesis , Recombinant Proteins/pharmacology , Sequence Homology, Amino Acid , Serpins/analysis , Serpins/pharmacology , Skin/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Thrombin/metabolism , Transfection , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...