Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
1.
Sci Adv ; 6(47)2020 Nov.
Article in English | MEDLINE | ID: mdl-33219030

ABSTRACT

Electron interferometry via phase-contrast microscopy, holography, or picodiffraction can provide a direct visualization of the static electric and magnetic fields inside or around a material at subatomic precision, but understanding the electromagnetic origin of light-matter interaction requires time resolution as well. Here, we demonstrate that pump-probe electron diffraction with all-optically compressed electron pulses can capture dynamic electromagnetic potentials in a nanophotonic material with sub-light-cycle time resolution via centrosymmetry-violating Bragg spot dynamics. The origin of this effect is a sizable quantum mechanical phase shift that the electron de Broglie wave obtains from the oscillating electromagnetic potentials within less than 1 fs. Coherent electron imaging and scattering can therefore reveal the electromagnetic foundations of light-matter interaction on the level of the cycles of light.

2.
Leukemia ; 31(10): 2191-2199, 2017 10.
Article in English | MEDLINE | ID: mdl-28202953

ABSTRACT

Impressive results have been achieved by adoptively transferring T-cells expressing CD19-specific CARs with binding domains from murine mAbs to treat B-cell malignancies. T-cell mediated immune responses specific for peptides from the murine scFv antigen-binding domain of the CAR can develop in patients and result in premature elimination of CAR T-cells increasing the risk of tumor relapse. As fully human scFv might reduce immunogenicity, we generated CD19-specific human scFvs with similar binding characteristics as the murine FMC63-derived scFv using human Ab/DNA libraries. CARs were constructed in various formats from several scFvs and used to transduce primary human T-cells. The resulting CD19-CAR T-cells were specifically activated by CD19-positive tumor cell lines and primary chronic lymphocytic leukemia cells, and eliminated human lymphoma xenografts in immunodeficient mice. Certain fully human CAR constructs were superior to the FMC63-CAR, which is widely used in clinical trials. Imaging of cell surface distribution of the human CARs revealed no evidence of clustering without target cell engagement, and tonic signaling was not observed. To further reduce potential immunogenicity of the CARs, we also modified the fusion sites between different CAR components. The described fully human CARs for a validated clinical target may reduce immune rejection compared with murine-based CARs.


Subject(s)
Antigens, CD19/immunology , Burkitt Lymphoma/therapy , Immunotherapy, Adoptive/methods , Recombinant Fusion Proteins/immunology , Single-Chain Antibodies/immunology , T-Lymphocytes/transplantation , Animals , Burkitt Lymphoma/pathology , Cell Line, Tumor , Female , Gene Library , HEK293 Cells , Humans , K562 Cells , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Recombinant Fusion Proteins/genetics , Single-Chain Antibodies/genetics , Species Specificity , Transduction, Genetic , Xenograft Model Antitumor Assays
3.
Bioorg Med Chem Lett ; 13(16): 2799-803, 2003 Aug 18.
Article in English | MEDLINE | ID: mdl-12873518

ABSTRACT

The SAR of a series of potent sulfonamide hydroxamate TACE inhibitors, all bearing a butynyloxy P1' group, was explored. In particular, compound 5j has excellent in vitro potency against isolated TACE enzyme and in cells, good selectivity over MMP-1 and MMP-9, and oral activity in an in vivo model of TNF-alpha production and a collagen-induced arthritis model.


Subject(s)
Acetylene/chemistry , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Metalloendopeptidases/antagonists & inhibitors , ortho-Aminobenzoates/chemistry , ADAM Proteins , ADAM17 Protein , Crystallography, X-Ray , Hydroxamic Acids/chemistry , Molecular Structure , Structure-Activity Relationship , Sulfonamides/chemistry , ortho-Aminobenzoates/pharmacology
5.
Clin Immunol ; 103(1): 13-21, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11987981

ABSTRACT

Etanercept, a recombinant human tumor necrosis factor (TNF) inhibitor that binds both soluble and cell-bound TNF, has been shown to reduce disease activity and inhibit joint destruction when administered to patients with rheumatoid arthritis (RA). Because TNF receptors are found on many types of cells that modulate the immune response, we evaluated the general immune function of a subset of RA patients in a blinded clinical study. No significant differences were seen between patients treated with etanercept or placebo in the surface antigen phenotypes of peripheral blood leukocytes, T cell proliferative responses, neutrophil function, delayed-type hypersensitivity (DTH) reactions, serum immunoglobulin levels, or incidence of infections. Although this observational study was relatively small and could detect only major changes in immunological status, the stability of immune function over time in patients receiving etanercept corroborates the findings in clinical studies, which suggest that etanercept does not alter overall global immune function.


Subject(s)
Antirheumatic Agents/therapeutic use , Arthritis, Rheumatoid/drug therapy , Immunoglobulin G/therapeutic use , Receptors, Tumor Necrosis Factor/therapeutic use , Adult , Aged , Arthritis, Rheumatoid/immunology , Double-Blind Method , Etanercept , Humans , Immunoglobulins/blood , Immunophenotyping , Infections/epidemiology , Lymphocyte Activation , Middle Aged , Neutrophils/physiology , T-Lymphocytes/immunology
6.
Bioorg Med Chem Lett ; 12(8): 1199-202, 2002 Apr 22.
Article in English | MEDLINE | ID: mdl-11934588

ABSTRACT

The SAR of a series of potent sulfonamide hydroxamate TACE inhibitors bearing novel acetylenic P1' groups was explored. In particular, compound 4t bearing a butynyloxy P1' moiety has excellent in vitro potency against isolated TACE enzyme and in cells, good selectivity over MMP-1 and oral activity in an in vivo model of TNF-alpha production.


Subject(s)
Acetylene/chemistry , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Hydroxamic Acids/chemistry , Hydroxamic Acids/pharmacology , Metalloendopeptidases/antagonists & inhibitors , Sulfonamides/chemical synthesis , Sulfonamides/pharmacology , ortho-Aminobenzoates/chemistry , ortho-Aminobenzoates/pharmacology , ADAM Proteins , ADAM17 Protein , Structure-Activity Relationship
10.
Am J Physiol ; 276(5): L715-27, 1999 05.
Article in English | MEDLINE | ID: mdl-10330027

ABSTRACT

To determine the roles of the type 1 tumor necrosis factor (TNF) receptor (TNFR1) in lung inflammation and antibacterial defense, we exposed transgenic mice lacking TNFR1 [TNFR1(-/-)] and wild-type control mice to aerosolized lipopolysaccharide or Pseudomonas aeruginosa. After LPS, bronchoalveolar lavage fluid (BALF) from TNFR1(-/-) mice contained fewer neutrophils and less macrophage inflammatory protein-2 than BALF from control mice. TNF-alpha, interleukin-1beta, and total protein levels in BALF as well as tissue intercellular adhesion molecule-1 expression did not differ between the two groups. In contrast, lung inflammation and bacterial clearance after infection were augmented in TNFR1(-/-) mice. BALF from infected TNFR1(-/-) mice contained more neutrophils and TNF-alpha and less interleukin-1beta and macrophage inflammatory protein-2 than that from control mice, but protein levels were similarly elevated in both groups. Lung inflammation and bacterial clearance were also augmented in mice lacking both TNF receptors. Thus TNFR1 facilitates neutrophil recruitment after inhalation of lipopolysaccharide, in part by augmenting chemokine induction. In contrast, TNFR1 attenuates lung inflammation in response to live bacteria but does not contribute to increased lung permeability and is not required for the elimination of P. aeruginosa.


Subject(s)
Endotoxins/administration & dosage , Pneumonia, Bacterial/microbiology , Pseudomonas Infections , Receptors, Tumor Necrosis Factor/physiology , Administration, Inhalation , Animals , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , Chemokine CXCL2 , Chemotaxis, Leukocyte , Cytokines/analysis , Intercellular Adhesion Molecule-1/analysis , Interleukin-1/analysis , Leukocyte Count , Mice , Mice, Inbred C57BL , Mice, Transgenic , Monokines/analysis , Neutrophils , Pneumonia, Bacterial/metabolism , Pneumonia, Bacterial/pathology , Proteins/analysis , Receptors, Tumor Necrosis Factor/deficiency , Receptors, Tumor Necrosis Factor/genetics , Tumor Necrosis Factor-alpha/analysis
11.
Clin Excell Nurse Pract ; 3(2): 73-9, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10646394

ABSTRACT

Nurse practitioners (NPs) are in demand nationwide as primary care providers both in the inpatient and outpatient settings. Lack of prescriptive privileges and/or a narrow scope of practice can be practice limiting. This article provides a written template for prescribing authority, generic scope of practice, and furnishing policy, including evaluative process, for nurse practitioners who wish to obtain prescriptive privileges. Developed at the Palo Alto Veterans Affairs Health Care System, these documents can be used by NPs nationwide. They can be adapted to each provider's unique practice, whether in the hospital, clinic, or private practice setting.


Subject(s)
Drug Prescriptions/standards , Nurse Practitioners/standards , Practice Guidelines as Topic , Professional Autonomy , Acute Disease/nursing , Ambulatory Care/standards , Drug Monitoring/standards , Humans , Nursing Assessment , Nursing Records , Patient Care Planning , Primary Health Care/standards
12.
Am J Respir Cell Mol Biol ; 19(6): 881-91, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9843922

ABSTRACT

The pulmonary host response to infection and inflammation appears, at least in part, to be compartmentalized from the systemic host response. Tumor necrosis factor-alpha (TNF-alpha) has been implicated in lung inflammation and injury, but its site(s) of action has not been clearly defined. To investigate this, transgenic mice (surfactant apoprotein C promotor/soluble TNF receptor type II-Fc fusion protein ([SPCTNFRIIFc] mice) were generated in which TNF-alpha was selectively antagonized in the distal lung through tissue-specific expression of sTNFRIIFc, a soluble TNF inhibitor. The lung inflammatory response in these mice to pulmonary challenge with Micropolyspora faeni antigen or lipopolysaccharide (LPS) was compared with the response of wild-type mice, wild-type mice treated with recombinant sTNFRIIFc intravenously, and type I TNF-receptor knockout mice. Recruitment of polymorphonuclear leukocytes (PMN) to the lung after challenge with M. faeni antigen was essentially abolished in the TNFRI knockout mice and markedly reduced in the SPCTNFRIIFc mice. Wild-type mice given sTNFRIIFc intravenously in amounts resulting in lung concentrations similar to those in SPCTNFRIIFc mice also showed significantly reduced lung PMN recruitment, whereas those given doses that achieved such concentrations in the blood but low levels in the lung did not. In contrast, PMN recruitment to the lung following aerosol challenge with LPS was reduced significantly in the TNFRI knockout mice and in mice given high-dose sTNFRIIFc intravenously, but was not reduced significantly in SPCTNFRIIFc mice. Thus, inhibition of PMN recruitment in response to M. faeni antigen correlated largely with the extent of intrapulmonary inhibition of TNF-alpha, whereas the response to LPS correlated best with the extent of extrapulmonary inhibition of TNF-alpha. These studies indicate that TNF-alpha may act at different loci to mediate lung inflammation, with the site of action depending in part on the nature of the inflammatory stimulus, and that SPCTNFRIIFc mice provide a tool by which the locus of TNF action can be addressed.


Subject(s)
Pneumonia/immunology , Tumor Necrosis Factor-alpha/physiology , Administration, Intranasal , Animals , Antigens, Bacterial/administration & dosage , Antigens, Bacterial/immunology , Apolipoproteins C/genetics , Immunoglobulin Fc Fragments/genetics , Lipopolysaccharides/administration & dosage , Lung/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Micromonosporaceae/immunology , Neutrophils/pathology , Pneumonia/pathology , Promoter Regions, Genetic , Receptors, Tumor Necrosis Factor/genetics , Recombinant Fusion Proteins/pharmacology
13.
Int Immunol ; 10(6): 785-90, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9678759

ABSTRACT

Mouse NK cells may use both cytokine, e.g. IFN-gamma, tumor necrosis factor (TNF)-alpha and IL-12, and cytotoxic, e.g. perforin and Fas-FasL, pathways to reject incompatible bone marrow cell (BMC) grafts. To begin a dissection of these two major pathways, mice bearing deletional mutations of IFN-gamma, TNF-RI/II or perforin, or mice treated with mAb to IL-12, IFN-gamma or NK1.1 were irradiated and challenged with class I-deficient BMC grafts, a system in which only NK cells are the effector cells. Proliferation of the donor-derived cells was judged in terms of splenic incorporation of [125I]iododeoxyuridine 5 or 7 days after cell transfer. All of these mice maintained in a specific pathogen-free (s.p.f.) environment were able to reject the BMC, except those treated with anti-NK1.1 mAb. However, perforin deficient mice maintained in a conventional breeding facility failed to reject class I (Tap-1)-deficient marrow cells. Transfer of mice from the pathogen-free to the conventional facility resulted in a slow and incomplete loss of the ability to reject marrow cells. Thus, the breeding colony environment can elicit otherwise undetectable defects in the rejection ability of perforin-deficient NK cells. This report will hopefully alert those investigators who have only studied immune gene knockout mice in s.p.f. facilities and found no significant abnormalities.


Subject(s)
Bone Marrow Transplantation/immunology , Cytokines/metabolism , Cytotoxins/metabolism , Environmental Exposure , Graft Rejection/immunology , Killer Cells, Natural/immunology , Animals , Antibodies, Monoclonal , Cytokines/immunology , Cytotoxins/immunology , Housing, Animal , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Specific Pathogen-Free Organisms
14.
J Immunol ; 160(2): 943-52, 1998 Jan 15.
Article in English | MEDLINE | ID: mdl-9551933

ABSTRACT

The pleiotropic activities of the potent proinflammatory cytokine TNF are mediated by two structurally related, but functionally distinct, receptors, p55 and p75, that are coexpressed on most cell types. The majority of biologic responses classically attributed to TNF are mediated by p55. In contrast, p75 has been proposed to function as both a TNF antagonist by neutralizing TNF and as a TNF agonist by facilitating the interaction between TNF and p55 at the cell surface. We have examined the roles of p55 and p75 in mediating and modulating the activity of TNF in vivo by generating and examining mice genetically deficient in these receptors. Selective deficits in several host defense and inflammatory responses are observed in mice lacking p55 or both p55 and p75, but not in mice lacking p75. In these models, the activity of p55 is not impaired by the absence of p75, arguing against a physiologic role for p75 as an essential element of p55-mediated signaling. In contrast, exacerbated pulmonary inflammation and dramatically increased endotoxin induced serum TNF levels in mice lacking p75 suggest a dominant role for p75 in suppressing TNF-mediated inflammatory responses. In summary, these data help clarify the biologic roles of p55 and p75 in mediating and modulating the biologic activity of TNF and provide genetic evidence for an antagonistic role of p75 in vivo.


Subject(s)
Antigens, CD/metabolism , Antigens, CD/physiology , Inflammation/immunology , Receptors, Tumor Necrosis Factor/metabolism , Receptors, Tumor Necrosis Factor/physiology , Acute-Phase Reaction/genetics , Acute-Phase Reaction/immunology , Animals , Antigens, CD/blood , Antigens, CD/genetics , Cell Differentiation/genetics , Cell Differentiation/immunology , Crosses, Genetic , Disease Models, Animal , Endotoxemia/genetics , Endotoxemia/immunology , Endotoxemia/mortality , Farmer's Lung/genetics , Farmer's Lung/immunology , Farmer's Lung/pathology , Female , Immunity, Innate , Inflammation/genetics , Listeriosis/immunology , Lymphocyte Subsets/cytology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Inbred Strains , Mice, Knockout , Receptors, Tumor Necrosis Factor/blood , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor, Type I , Receptors, Tumor Necrosis Factor, Type II , Thymus Gland/cytology , Thymus Gland/growth & development , Tumor Necrosis Factor-alpha/metabolism
15.
Clin Excell Nurse Pract ; 2(5): 263-9, 1998 Sep.
Article in English | MEDLINE | ID: mdl-10455573

ABSTRACT

Healthcare priorities and use have changed dramatically over the last several years. Expansion of the nurse practitioner (NP) role has been a hallmark of the restructuring efforts, which have emphasized primary care and the use of nonphysician specialists. NPs are practicing in a wider range of settings than ever before, including acute hospital settings, outpatient clinics, and specialty services. While the Veterans Health Administration (VHA) has employed NPs since the early 1970s, their practice has been limited at many VHA medical centers. Such was the case at the Veterans Affairs Palo Alto Health Care System (VAPAHCS), when physicians produced a scope of practice (SOP) for NPs that was practice limiting. This article highlights the historical progress of NPs in defining their practice, discusses barriers to NP practice at the VAPAHCS, outlines strategies to overcome barriers, and discusses future possibilities for advanced practice nursing within the VA.


Subject(s)
Job Description , Nurse Practitioners/organization & administration , Professional Autonomy , United States Department of Veterans Affairs/organization & administration , Adult , California , Certification , Female , Hospital Restructuring/organization & administration , Humans , Male , Middle Aged , Models, Nursing , Nursing Evaluation Research , Program Evaluation , United States
16.
Virology ; 236(2): 316-27, 1997 Sep 29.
Article in English | MEDLINE | ID: mdl-9325239

ABSTRACT

Poxvirus genomes encode several proteins which inhibit specific elements of the host immune response. We show the "35K" virulence gene in variola and cowpox viruses, whose vaccinia and Shope fibroma virus equivalents are strongly conserved in sequence, actually encodes a secreted soluble protein with high-affinity binding to virtually all known beta chemokines, but only weak or no affinity to the alpha and gamma classes. The viral protein completely inhibits the biological activity of monocyte chemotactic protein-1 (MCP-1) by competitive inhibition of chemokine binding to cellular receptors. As all beta chemokines are also shown to cross-compete with MCP1 binding to the viral protein, we conclude that this viral chemokine inhibitor (vCCI) not only interacts through a common binding site, but is likely a potent general inhibitor of beta chemokine activity. Unlike many poxvirus virulence genes to date, which are clearly altered forms of acquired cellular genes of the vertebrate immune system, this viral chemokine inhibitor (vCCI) shares no sequence homology with known proteins, including known cellular chemokine receptors, all of which are multiple membrane-spanning proteins. Thus, vCCI presumably has no cellular analogue and instead may be the product of unrelenting sequence variations which gave rise to a completely new protein with similar binding properties to native chemokine receptors. The proposed function of vCCI is inhibition of the proinflammatory (antiviral) activities of beta chemokines.


Subject(s)
Chemokines/antagonists & inhibitors , Genome, Viral , Poxviridae/genetics , Poxviridae/immunology , Viral Proteins/genetics , Viral Proteins/immunology , Amino Acid Sequence , Animals , Base Sequence , Binding, Competitive , Calcium/metabolism , Cell Line , Chemokines/classification , Chemotaxis, Leukocyte , Chimera/genetics , Chimera/immunology , Cowpox virus/genetics , Cowpox virus/immunology , DNA Primers/genetics , Mice , Molecular Sequence Data , Polymerase Chain Reaction , Receptors, Chemokine/genetics , Sequence Homology, Amino Acid , Solubility , Variola virus/genetics , Variola virus/immunology , Viral Proteins/metabolism , Virulence/genetics
17.
N Engl J Med ; 337(3): 141-7, 1997 Jul 17.
Article in English | MEDLINE | ID: mdl-9219699

ABSTRACT

BACKGROUND: Tumor necrosis factor (TNF) is a proinflammatory cytokine involved in the pathogenesis of rheumatoid arthritis, and antagonism of TNF may reduce the activity of the disease. This study evaluated the safety and efficacy of a novel TNF antagonist - a recombinant fusion protein that consists of the soluble TNF receptor (p75) linked to the Fc portion of human IgG1 (TNFR:Fc). METHODS: In this multicenter, double-blind trial, we randomly assigned 180 patients with refractory rheumatoid arthritis to receive subcutaneous injections of placebo or one of three doses of TNFR:Fc (0.25, 2, or 16 mg per square meter of body-surface area) twice weekly for three months. The clinical response was measured by changes in composite symptoms of arthritis defined according to American College of Rheumatology criteria. RESULTS: Treatment with TNFR:Fc led to significant reductions in disease activity, and the therapeutic effects of TNFR:Fc were dose-related. At three months, 75 percent of the patients in the group assigned to 16 mg of TNFR:Fc per square meter had improvement of 20 percent or more in symptoms, as compared with 14 percent in the placebo group (P<0.001). In the group assigned to 16 mg per square meter, the mean percent reduction in the number of tender or swollen joints at three months was 61 percent, as compared with 25 percent in the placebo group (P<0.001). The most common adverse events were mild injection-site reactions and mild upper respiratory tract symptoms. There were no dose-limiting toxic effects, and no antibodies to TNFR:Fc were detected in serum samples. CONCLUSIONS: In this three-month trial TNFR:Fc was safe, well tolerated, and associated with improvement in the inflammatory symptoms of rheumatoid arthritis.


Subject(s)
Antigens, CD , Arthritis, Rheumatoid/drug therapy , Immunoglobulin G/therapeutic use , Receptors, Tumor Necrosis Factor , Recombinant Fusion Proteins/therapeutic use , Antibodies/blood , Antigens, CD/immunology , Dose-Response Relationship, Drug , Double-Blind Method , Female , Humans , Immunoglobulin Fc Fragments/therapeutic use , Male , Middle Aged , Receptors, Tumor Necrosis Factor/immunology , Receptors, Tumor Necrosis Factor, Type II , Treatment Outcome
18.
Transplantation ; 63(4): 570-7, 1997 Feb 27.
Article in English | MEDLINE | ID: mdl-9047153

ABSTRACT

A preliminary clinical study of renal allograft recipients revealed that a dimeric form of the human 80 kDa soluble receptor (sTNFR:Fc) for tumor necrosis factor (TNF) is well tolerated and attenuates the OKT3-induced acute clinical syndrome. The current study determined the in vivo biological effects and fate of sTNFR:Fc in these patients. Serial assessment of both antigenic and biological activities of circulating TNF and sTNFR:Fc have led to the following observations. (1) Although control patients typically responded to the first OKT3 injection with a rapid increase of biologically active TNFalpha, patients on sTNFR:Fc therapy had markedly higher serum TNFalpha antigenic levels, but no detectable bioactivity. Thus, sTNFR:Fc functioned as a potent antagonist, despite its cytokine-carrier effect. (2) Peak sTNFR:Fc levels averaging 800 and 2500 ng/ml were routinely achieved in vivo, using the low-dose (0.05 mg/kg) and high-dose (0.15 mg/kg) protocols. (3) The half-life of circulating sTNFR:Fc was estimated to be approximately 4.4 days, and levels of p80 receptors in treated patients remained significantly above those in control patients for at least 20 days. (4) In vitro blocking studies demonstrated that circulating sTNFR:Fc remained biologically active for 2 weeks. These results demonstrate that under current protocols, significant serum levels of sTNFR:Fc, capable of effectively neutralizing TNF activity over prolonged periods, can be achieved. The persistent OKT3 side effects observed, despite sTNFR:Fc therapy, are therefore likely to be caused by factors other than TNF.


Subject(s)
Immunosuppressive Agents/adverse effects , Kidney Transplantation , Muromonab-CD3/adverse effects , Receptors, Tumor Necrosis Factor/physiology , Dimerization , Graft Rejection/prevention & control , Humans , Prospective Studies , Receptors, Tumor Necrosis Factor/analysis , Recombinant Proteins/therapeutic use , Transplantation, Homologous , Tumor Necrosis Factor-alpha/analysis , Tumor Necrosis Factor-alpha/antagonists & inhibitors
19.
FEBS Lett ; 401(2-3): 235-8, 1997 Jan 20.
Article in English | MEDLINE | ID: mdl-9013894

ABSTRACT

Pro-TNF alpha, Steel factor, type II IL-1R and IL-2R alpha were expressed in COS-7 cells and the generation of their soluble forms was examined. The release of all four proteins was strongly stimulated by the phorbol ester PMA and completely blocked by a hydroxamate-based inhibitor of metalloproteases. COS-7 cell membranes were found to cleave various synthetic pro-TNF alpha peptides with the same specificity as a partially purified TNF alpha converting enzyme purified from human monocytic cells, suggesting that the same enzyme may be responsible for at least some of the COS-7 cell shedding activity.


Subject(s)
Membrane Proteins/metabolism , Animals , COS Cells , Cell Line , Dipeptides/pharmacology , Humans , Hydroxamic Acids/pharmacology , Receptors, Interleukin/metabolism , Stem Cell Factor/metabolism , Tumor Necrosis Factor-alpha/metabolism
20.
Biochem Biophys Res Commun ; 225(2): 400-5, 1996 Aug 14.
Article in English | MEDLINE | ID: mdl-8753775

ABSTRACT

Tumor necrosis factor-alpha is released from cells by a proteolytic cleavage. Previous work suggested that a specific, non-matrix metalloproteinase carries out this cleavage, but matrix metalloproteinases have also been implicated. In this paper, we report that none of the matrix metalloproteinases tested cleaved peptide substrates as specifically as the non-matrix metalloproteinase. A matrix metalloproteinase did process tumor necrosis factor-alpha extracted from COS cells, but neither tissue inhibitor of metalloproteinases-1 nor -2 blocked tumor necrosis factor-alpha processing by human monocytes. Moreover, tissue inhibitor of metalloproteinases-1 had at most a partial effect on the in vivo release of the cytokine in mice. We conclude that a non-matrix metalloproteinase is the major physiological tumor necrosis factor-alpha convertase.


Subject(s)
Glycoproteins/pharmacology , Metalloendopeptidases/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , ADAM Proteins , ADAM17 Protein , Animals , CHO Cells , Cell Line , Cells, Cultured , Cricetinae , Humans , Mice , Mice, Inbred BALB C , Protein Processing, Post-Translational , Recombinant Proteins/pharmacology , Substrate Specificity , Tissue Inhibitor of Metalloproteinases , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...