Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
J Virol ; 92(22)2018 11 15.
Article in English | MEDLINE | ID: mdl-30158289

ABSTRACT

Rabies virus is a neurovirulent RNA virus, which causes about 59,000 human deaths each year. Treatment for rabies does not exist due to incomplete understanding of the pathogenesis. MALT1 mediates activation of several immune cell types and is involved in the proliferation and survival of cancer cells. MALT1 acts as a scaffold protein for NF-κB signaling and a cysteine protease that cleaves substrates, leading to the expression of immunoregulatory genes. Here, we examined the impact of genetic or pharmacological MALT1 inhibition in mice on disease development after infection with the virulent rabies virus strain CVS-11. Morbidity and mortality were significantly delayed in Malt1-/- compared to Malt1+/+ mice, and this effect was associated with lower viral load, proinflammatory gene expression, and infiltration and activation of immune cells in the brain. Specific deletion of Malt1 in T cells also delayed disease development, while deletion in myeloid cells, neuronal cells, or NK cells had no effect. Disease development was also delayed in mice treated with the MALT1 protease inhibitor mepazine and in knock-in mice expressing a catalytically inactive MALT1 mutant protein, showing an important role of MALT1 proteolytic activity. The described protective effect of MALT1 inhibition against infection with a virulent rabies virus is the precise opposite of the sensitizing effect of MALT1 inhibition that we previously observed in the case of infection with an attenuated rabies virus strain. Together, these data demonstrate that the role of immunoregulatory responses in rabies pathogenicity is dependent on virus virulence and reveal the potential of MALT1 inhibition for therapeutic intervention.IMPORTANCE Rabies virus is a neurotropic RNA virus that causes encephalitis and still poses an enormous challenge to animal and public health. Efforts to establish reliable therapeutic strategies have been unsuccessful and are hampered by gaps in the understanding of virus pathogenicity. MALT1 is an intracellular protease that mediates the activation of several innate and adaptive immune cells in response to multiple receptors, and therapeutic MALT1 targeting is believed to be a valid approach for autoimmunity and MALT1-addicted cancers. Here, we study the impact of MALT1 deficiency on brain inflammation and disease development in response to infection of mice with the highly virulent CVS-11 rabies virus. We demonstrate that pharmacological or genetic MALT1 inhibition decreases neuroinflammation and extends the survival of CVS-11-infected mice, providing new insights in the biology of MALT1 and rabies virus infection.


Subject(s)
Brain/immunology , Inflammation/prevention & control , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/physiology , Rabies virus/immunology , Rabies/prevention & control , T-Lymphocytes/immunology , Animals , Brain/metabolism , Brain/virology , Cells, Cultured , Inflammation/immunology , Inflammation/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/antagonists & inhibitors , Rabies/immunology , Rabies/metabolism , T-Lymphocytes/pathology , T-Lymphocytes/virology
2.
J Virol ; 92(8)2018 04 15.
Article in English | MEDLINE | ID: mdl-29367251

ABSTRACT

MALT1 is involved in the activation of immune responses, as well as in the proliferation and survival of certain cancer cells. MALT1 acts as a scaffold protein for NF-κB signaling and a cysteine protease that cleaves substrates, further promoting the expression of immunoregulatory genes. Deregulated MALT1 activity has been associated with autoimmunity and cancer, implicating MALT1 as a new therapeutic target. Although MALT1 deficiency has been shown to protect against experimental autoimmune encephalomyelitis, nothing is known about the impact of MALT1 on virus infection in the central nervous system. Here, we studied infection with an attenuated rabies virus, Evelyn-Rotnycki-Abelseth (ERA) virus, and observed increased susceptibility with ERA virus in MALT1-/- mice. Indeed, after intranasal infection with ERA virus, wild-type mice developed mild transient clinical signs with recovery at 35 days postinoculation (dpi). Interestingly, MALT1-/- mice developed severe disease requiring euthanasia at around 17 dpi. A decreased induction of inflammatory gene expression and cell infiltration and activation was observed in MALT1-/- mice at 10 dpi compared to MALT1+/+ infected mice. At 17 dpi, however, the level of inflammatory cell activation was comparable to that observed in MALT1+/+ mice. Moreover, MALT1-/- mice failed to produce virus-neutralizing antibodies. Similar results were obtained with specific inactivation of MALT1 in T cells. Finally, treatment of wild-type mice with mepazine, a MALT1 protease inhibitor, also led to mortality upon ERA virus infection. These data emphasize the importance of early inflammation and activation of T cells through MALT1 for controlling the virulence of an attenuated rabies virus in the brain.IMPORTANCE Rabies virus is a neurotropic virus which can infect any mammal. Annually, 59,000 people die from rabies. Effective therapy is lacking and hampered by gaps in the understanding of virus pathogenicity. MALT1 is an intracellular protein involved in innate and adaptive immunity and is an interesting therapeutic target because MALT1-deregulated activity has been associated with autoimmunity and cancers. The role of MALT1 in viral infection is, however, largely unknown. Here, we study the impact of MALT1 on virus infection in the brain, using the attenuated ERA rabies virus in different models of MALT1-deficient mice. We reveal the importance of MALT1-mediated inflammation and T cell activation to control ERA virus, providing new insights in the biology of MALT1 and rabies virus infection.


Subject(s)
Brain/immunology , Lymphocyte Activation , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/immunology , Rabies virus/immunology , Rabies/immunology , T-Lymphocytes/immunology , Animals , Brain/pathology , Brain/virology , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Inflammation/virology , Mice , Mice, Knockout , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/genetics , Rabies/genetics , Rabies/pathology , Rabies virus/genetics , T-Lymphocytes/pathology
3.
Pol J Pathol ; 64(2): 84-9, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23900864

ABSTRACT

The pro-inflammatory context of sickle cell disease promotes the liberation of cytokines such as CCL5, encoded by a gene located on chromosome 17. Herein, the occurrence of three variations of CCL5 in sickle cell anemia (SCA) and their relations to two major complications - painful crisis and presence of infections - were investigated. 100 SCA Tunisian patients and 100 healthy subjects were included in the case control study. Then the sample of patients was divided into two groups according to the presence or absence of each complication. The polymorphisms, namely g.-403G>A, g.-28C>G and g.In1.+1T>C, were analyzed by PCR/sequencing. Our findings show the presence of eight genotypes, namely GG, GA and AA of g.-403G>A, CC, CG and GG of g.-28C>G, and TT and TC of g.In1.+1T>C. The frequencies of studied single nucleotide polymorphisms (SNPs) and haplotypes in SCA patients do not differ significantly from healthy control group results. There is also no significant association between the analyzed polymorphisms and complications as for painful crisis and presence of infections (p > 0.05). Altogether, our data support the conclusion that the three polymorphisms of CCL5, namely g.-403G>A, g.-28C>G and g.In1.+1T>C, do not seem to be involved in the clinical variability of SCA in Tunisia.


Subject(s)
Anemia, Sickle Cell/complications , Anemia, Sickle Cell/genetics , Chemokine CCL5/genetics , Polymorphism, Single Nucleotide , Adult , Case-Control Studies , Female , Humans , Male , Phenotype , Reverse Transcriptase Polymerase Chain Reaction , Tunisia
4.
Sci Total Environ ; 830: 154448, 2022 Jul 15.
Article in English | MEDLINE | ID: mdl-35307416

ABSTRACT

Arsenic (As) exposure, even at low environmentally relevant levels, may cause detrimental health outcomes through developmental toxicity and by acting as an endocrine disrupting compound (EDC). Although several studies indicate that wildlife bioaccumulate As, few evaluate the health impact on fish species in their natural environment. In the U.S., As has a drinking water regulatory limit of 10 µg/L. In many parts of Arizona, surface water and groundwater have naturally elevated levels of As from geologic deposits and contamination is exacerbated by anthropogenic activity. In aquatic environments, the Western mosquitofish, Gambusia affinis, is a good bioindicator for EDC exposure because of the distinct androgen-related development of an intromittent organ, the gonapodium, in males. We evaluated morphological and reproductive outcomes in mosquitofish exposed to As. In a laboratory experiment, juvenile male mosquitofish were exposed to sodium arsenite (0 µg/L, 0.75 µg/L, 7.50 µg/L, and 75 µg/L) for 30 days, and in a field study, populations of adult male mosquitofish were collected in Arizona waterways with As levels above and below the World Health Organization's regulatory limit. In both studies, higher As exposure was significantly associated with altered hepatosomatic indices, altered fish morphology, shortened gonopodia, and lower gonopodia-somatic indices. In the field experiment, populations from surface water with higher As concentrations exhibited lower condition factors, lower gonadal-somatic indices, distinct gonopodia shapes, and altered estrogen receptor alpha and vitellogenin gene expression; androgen receptor expression was unchanged. Together, laboratory and field results suggest that As exposure at environmentally-relevant levels affects general growth and reproductive development in mosquitofish. Observed effects may further influence individual health, mobility, or reproductive function, and because G. affinis is a species known to tolerate and adapt to a wide range of environments, it serves as a local bioindicator species as well as a model organism for parallel field and laboratory studies.


Subject(s)
Arsenic , Cyprinodontiformes , Water Pollutants, Chemical , Androgens/metabolism , Animals , Arsenic/metabolism , Arsenic/toxicity , Cyprinodontiformes/physiology , Gonads , Male , Reproduction , Water/metabolism , Water Pollutants, Chemical/metabolism , Water Pollutants, Chemical/toxicity
5.
Cell Death Differ ; 14(5): 1050-9, 2007 May.
Article in English | MEDLINE | ID: mdl-17318221

ABSTRACT

We have studied the involvement of receptor interacting protein kinase-1 (RIP1) and dsRNA-activated protein kinase (PKR) in external dsRNA-induced apoptotic and necrotic cell death in Jurkat T cell lymphoma. Our results suggest that RIP1 plays an imported role in dsRNA-induced apoptosis and necrosis. We demonstrated that contrary to necrosis, protein synthesis is inhibited in apoptosis. Here, we show that phosphorylation of translation initiation factor 2-alpha (eukaryotic initiation factor 2-alpha (eIF2-alpha)) and its kinase, PKR, occur in dsRNA-induced apoptosis but not in necrosis. These events are caspase-dependent and coincide with the appearance of the caspase-mediated PKR fragments, N-terminal domain (ND) and kinase domain (KD). Our immunoprecipitation experiments demonstrated that both fragments could independently co-precipitate with full-length PKR. Expression of PKR-KD leads to PKR and eIF2-alpha phosphorylation and inhibits protein translation, whereas that of PKR-ND does not. Co-expression of PKR-ND and PKR-KD promotes their interaction with PKR, PKR and eIF2-alpha phosphorylation and suppresses protein translation better than PKR-KD alone. Our findings suggest a caspase-dependent mode of activation of PKR in apoptosis in which the PKR-KD fragment interacts with and activates intact PKR. PKR-ND facilitates the interaction of PKR-KD with full-length PKR and thus the activation of the kinase and amplifies the translation inhibitory signal.


Subject(s)
Caspases/metabolism , Peptide Fragments/metabolism , Protein Biosynthesis , eIF-2 Kinase/metabolism , Apoptosis/drug effects , Caspase Inhibitors , Cell Line , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Humans , Immunoprecipitation , Jurkat Cells , Necrosis , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Biosynthesis/drug effects , Protein Processing, Post-Translational/drug effects , Protein Structure, Tertiary/drug effects , RNA, Double-Stranded/pharmacology , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Signal Transduction/drug effects , eIF-2 Kinase/biosynthesis
6.
Cell Death Discov ; 3: 17012, 2017.
Article in English | MEDLINE | ID: mdl-28280602

ABSTRACT

Rabies virus is a highly neurovirulent RNA virus, which causes about 59000 deaths in humans each year. Previously, we described macrophage cytotoxicity upon infection with rabies virus. Here we examined the type of cell death and the role of specific caspases in cell death and disease development upon infection with two laboratory strains of rabies virus: Challenge Virus Standard strain-11 (CVS-11) is highly neurotropic and lethal for mice, while the attenuated Evelyn-Rotnycki-Abelseth (ERA) strain has a broader cell tropism, is non-lethal and has been used as an oral vaccine for animals. Infection of Mf4/4 macrophages with both strains led to caspase-1 activation and IL-1ß and IL-18 production, as well as activation of caspases-3, -7, -8, and -9. Moreover, absence of caspase-3, but not of caspase-1 and -11 or -7, partially inhibited virus-induced cell death of bone marrow-derived macrophages. Intranasal inoculation with CVS-11 of mice deficient for either caspase-1 and -11 or -7 or both IL-1ß and IL-18 led to general brain infection and lethal disease similar to wild-type mice. Deficiency of caspase-3, on the other hand, significantly delayed the onset of disease, but did not prevent final lethal outcome. Interestingly, deficiency of caspase-1/11, the key executioner of pyroptosis, aggravated disease severity caused by ERA virus, whereas wild-type mice or mice deficient for either caspase-3, -7, or both IL-1ß and IL-18 presented the typical mild symptoms associated with ERA virus. In conclusion, rabies virus infection of macrophages induces caspase-1- and caspase-3-dependent cell death. In vivo caspase-1/11 and caspase-3 differently affect disease development in response to infection with the attenuated ERA strain or the virulent CVS-11 strain, respectively. Inflammatory caspases seem to control attenuated rabies virus infection, while caspase-3 aggravates virulent rabies virus infection.

7.
Int J Lab Hematol ; 39(2): 223-231, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28133914

ABSTRACT

INTRODUCTION: Pyruvate kinase (PK) deficiency is one of the most common hereditary nonspherocytic hemolytic anemias worldwide with clinical manifestations ranging from mild to severe hemolysis. However, investigation of this enzymopathy is lacking in Tunisia. We report here a pioneer investigation of PK deficiency among Tunisian cases referred to our laboratory for biological analysis of unknown cause of hemolytic anemia. METHODS: Two hundred and fifty-three patients with unknown cause of hemolytic anemia have been addressed to our laboratory in order to investigate for red blood cells genetic disorders. Red cell enzyme activities were measured by standard methods, and molecular analysis was performed by DNA sequencing. The interpretation of mutation effect and the molecular modeling were performed by using specific software. RESULTS: Six different PKLR mutations were found (c.966-1G>T; c.965+1G>A; c.721G>T; c.1163C>A; c.1456C>T; c.1537T>A), among which four are described for the first time. Genotype-phenotype correlations for the novel missense mutations were investigated by three-dimensional structure analysis. CONCLUSION: This study provides important data of PK deficiency among Tunisians. It might be followed by a large neonatal screening to determine the spectrum of PK mutations and identify potential deficient patients for an early medical follow-up.


Subject(s)
Anemia, Hemolytic, Congenital Nonspherocytic/genetics , Mutation, Missense/genetics , Pyruvate Kinase/deficiency , Pyruvate Metabolism, Inborn Errors/genetics , Anemia, Hemolytic/etiology , Anemia, Hemolytic/genetics , Anemia, Hemolytic, Congenital Nonspherocytic/etiology , DNA Mutational Analysis , Erythrocytes/enzymology , Genetic Association Studies , Humans , Models, Molecular , Pyruvate Kinase/genetics , Pyruvate Metabolism, Inborn Errors/etiology , Tunisia/epidemiology
8.
Cell Death Differ ; 9(9): 981-94, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12181749

ABSTRACT

Interferons enhance the cellular antiviral response by inducing expression of protective proteins. Many of these proteins are activated by dsRNA, a typical by-product of viral infection. Here we show that type-I and type-II interferons can sensitize cells to dsRNA-induced cytotoxicity. In caspase-8- or FADD-deficient Jurkat cells dsRNA induces necrosis, instead of apoptosis. In L929sA cells dsRNA-induced necrosis involves high reactive oxygen species production. The antioxidant butylated hydroxyanisole protects cells from necrosis, but shifts the response to apoptosis. Treatment with the caspase inhibitor benzyloxycarbonyl-Val-Ala-DL-Asp(OMe)-fluoromethylketone or overexpression of Bcl-2 prevent this shift and promote necrosis. Our results suggest that a single stimulus can initiate different death-signaling pathways, leading to either necrotic or apoptotic cell death. Inhibition of key events in these signaling pathways, such as caspase activation, cytochrome c release or mitochondrial reactive oxygen species production, tips the balance between necrosis and apoptosis, leading to dominance of one of these death programs.


Subject(s)
Adaptor Proteins, Signal Transducing , Apoptosis/drug effects , Interferons/pharmacology , Jurkat Cells/drug effects , Necrosis , RNA Virus Infections/drug therapy , RNA Viruses/drug effects , RNA, Double-Stranded/drug effects , Signal Transduction/drug effects , Animals , Antioxidants/pharmacology , Apoptosis/genetics , Butylated Hydroxyanisole/pharmacology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Caspase 8 , Caspase 9 , Caspase Inhibitors , Caspases/deficiency , Caspases/genetics , Caspases/metabolism , Enzyme Inhibitors/pharmacology , Fas-Associated Death Domain Protein , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Humans , Interferon-beta/pharmacology , Interferon-gamma/pharmacology , Jurkat Cells/metabolism , Jurkat Cells/virology , Mice , Proto-Oncogene Proteins c-bcl-2/genetics , RNA Virus Infections/genetics , RNA Viruses/genetics , RNA, Double-Stranded/genetics , RNA, Double-Stranded/pharmacology , Reactive Oxygen Species/metabolism , Signal Transduction/genetics
9.
Laryngoscope ; 108(1 Pt 1): 107-10, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9432077

ABSTRACT

A prospective, double-blind, randomized, placebo-controlled study was performed to evaluate the effect of antibiotic prophylaxis in ear surgery. The present study reports on the results of 750 patients, half of whom received cefuroxime for 1 day, the other half, placebo. All postoperative infections occurring within 2 weeks after the intervention were recorded, together with several preoperative and perioperative parameters. It is concluded that exploratory tympanoplasties (including stapedotomy) and "dry perforation" tympanoplasties should be considered "clean" operations according to the American National Research Council and do not benefit from antibiotic prophylaxis. On the other hand, tympanoplasties performed on draining ears and on ears with cholesteatoma should be considered "dirty" operations for which antibiotic prophylaxis may decrease the postoperative infection rate by factor 3. All postoperative infections healed without sequels under proper treatment, except for three that resulted in graft necrosis--one in the placebo group and two in the cefuroxime group. In consequence, prophylaxis may not be mandatory in the dirty group, although the authors advocate its use for the sake of patient and surgeon comfort.


Subject(s)
Antibiotic Prophylaxis , Cefuroxime/therapeutic use , Ear Diseases/surgery , Surgical Wound Infection/prevention & control , Double-Blind Method , Humans , Prospective Studies , Tympanoplasty
14.
J Biol Chem ; 276(45): 41620-8, 2001 Nov 09.
Article in English | MEDLINE | ID: mdl-11555640

ABSTRACT

The protein kinase PKR is a major player in the cellular antiviral response, acting mainly by phosphorylation of the alpha-subunit of the eukaryotic translation initiation factor 2 (eIF2-alpha) to block de novo protein synthesis. PKR activation requires binding of double-stranded RNA or PACT/RAX proteins to its regulatory domain. Since several reports have demonstrated that translation is inhibited in apoptosis, we investigated whether PKR and eIF2-alpha phosphorylation contribute to this process. We show that PKR is proteolysed and that eIF2-alpha is phosphorylated at the early stages of apoptosis induced by various stimuli. Both events coincide with the onset of caspase activity and are prevented by caspase inhibitors. Using site-directed mutagenesis we show that PKR is specifically proteolysed at Asp(251) during cellular apoptosis. This site is cleaved in vitro by recombinant caspase-3, caspase-7, and caspase-8 and not by the proinflammatory caspase-1 and caspase-11. The released kinase domain efficiently phosphorylates eIF2-alpha at the cognate Ser(51) residue, and its overexpression in mammalian cells impairs the translation of its own mRNA and of reporter mRNAs. Our results demonstrate a new and caspase-dependent activation mode for PKR, leading to eIF2-alpha phosphorylation and translation inhibition in apoptosis.


Subject(s)
Apoptosis , Caspases/physiology , Eukaryotic Initiation Factor-2/metabolism , Protein Biosynthesis , eIF-2 Kinase/metabolism , Enzyme Activation , Humans , Jurkat Cells , Phosphorylation
15.
Nat Immun Cell Growth Regul ; 7(3): 185-92, 1988.
Article in English | MEDLINE | ID: mdl-3262823

ABSTRACT

Hemopoiesis is a sequence of events initiated by the self-renewal of pluripotent stem cells followed by a series of differentiation steps and completed in the formation of distinct tissue patterns. Differentiation and self-renewal are antagonistic processes. A mechanism that attenuates the differentiation flow is obligatory to prevent the exhaustion of the stem cell pool. We suggest that stromal cells from the bone marrow control stem cell renewal through a mechanism that does not require colony-stimulating factors. The organization of cells within the tissue and their specific localization is suggested to be directed by stromal cell activities other than differentiation inducers. These stromal cell activities restrict differentiation or accumulation of mature cells. They are therefore designated as 'Restrictins'.


Subject(s)
Biological Factors/physiology , Hematopoiesis , Animals , Colony-Forming Units Assay , Colony-Stimulating Factors/physiology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/physiology , In Vitro Techniques , Mice , Models, Biological
16.
Proteins ; 27(1): 96-109, 1997 Jan.
Article in English | MEDLINE | ID: mdl-9037715

ABSTRACT

The cytokines IL-6, LIF, CNTF, OSM, IL-11, and CT-1 have been grouped into the family of IL-6-type cytokines, since they all require gp130 for signal transduction. Interestingly, gp130 binds directly to OSM, whereas complex formation with the other cytokines depends on additional receptor subunits. Only limited structural information on these cytokines and their receptors is available. X-ray structures have been solved for the cytokines LIF and CNTF, whose up-up-down-down four-helix bundle is common to all of these cytokines, and for the receptors of hGH and prolactin, which contain two domains with a fibronectin III-like fold. Since cocrystallization and x-ray analysis of the up to four different proteins forming the receptor complexes of the IL-6-type cytokines is unlikely to be achieved in the near future, model building based on the existing structural information is the only approach for the time being. Here we present model structures of the complexes of human and murine IL-6 with their receptors. Their validity can be deduced from the fact that published mutagenesis data and the different receptor specificity of human and murine IL-6 can be understood. It is now possible to predict the relative positions and contacts for all molecules in their respective complexes. Such information can be used for the rational design of cytokine and receptor antagonists, which may have a valuable therapeutic perspective.


Subject(s)
Antigens, CD/metabolism , Cytokines/metabolism , Interleukin-6/metabolism , Receptors, Interleukin/metabolism , Amino Acid Sequence , Animals , Antigens, CD/chemistry , Cytokine Receptor gp130 , Cytokines/chemistry , Humans , Interleukin-6/chemistry , Membrane Glycoproteins/chemistry , Mice , Models, Molecular , Molecular Sequence Data , Receptors, Interleukin/chemistry , Receptors, Interleukin-6 , Sequence Homology, Amino Acid
17.
Infect Immun ; 62(3): 849-54, 1994 Mar.
Article in English | MEDLINE | ID: mdl-8112854

ABSTRACT

We report the cloning and sequencing of the gene coding for antigen 88 from Mycobacterium tuberculosis by using monoclonal antibodies to screen an expression library in lambda gt11. The gene encodes a 403-amino-acid-residue protein with a calculated molecular mass of 43,790 Da which contains seven putative transmembrane alpha-helical domains and presents a significant homology to the PstA protein of Escherichia coli. In its N-terminal region, it contains a 61-amino-acid region highly homologous to the fifth transmembrane helix of E. coli PstC. PstA and PstC are the two hydrophobic subunits of an E. coli periplasmic phosphate permease. Since the phosphate-binding subunit of this putative permease in M. tuberculosis has previously been characterized, i.e., the 38-kDa mycobacterial protein (also called protein antigen b, Ag 5, and Ag 78) homologous to PstS of E. coli, it seems likely that functional permeases analogous to the periplasmic permeases of gram-negative bacteria also exist in mycobacteria.


Subject(s)
ATP-Binding Cassette Transporters , Antigens, Bacterial/genetics , Bacterial Proteins , Carrier Proteins/genetics , Escherichia coli Proteins , Membrane Transport Proteins/genetics , Mycobacterium tuberculosis/immunology , Phosphate Transport Proteins , Amino Acid Sequence , Antibodies, Monoclonal/immunology , Antigens, Bacterial/chemistry , Base Sequence , Blotting, Southern , Cloning, Molecular , DNA/chemistry , Molecular Sequence Data
18.
J Biol Chem ; 272(29): 18304-10, 1997 Jul 18.
Article in English | MEDLINE | ID: mdl-9218470

ABSTRACT

The leptin receptor is a class I transmembrane protein with either a short or a long cytoplasmic domain. Using chemical cross-linking we have analyzed the binding of leptin to its receptor. Cross-linking of radiolabeled leptin to different isoforms of the leptin receptor expressed on COS-1 cells reveals leptin receptor monomer, homodimer, and oligomer complexes. Cotransfection of the long and short form of the leptin receptor did not provide any evidence for the formation of heterodimer complexes. Soluble forms consisting of either the entire extracellular domain or the two cytokine receptor homologous domains of the leptin receptor were purified to homogeneity from recombinant baculovirus-infected insect cells by leptin affinity chromatography. Gel filtration chromatography showed that these proteins exist in a dimeric form. Analysis of the complex formed between soluble leptin receptor and leptin by native polyacrylamide gel electrophoresis, and data obtained from the amino acid composition of the complex provide direct evidence that the extracellular domain of the leptin receptor binds leptin in a 1:1 ratio.


Subject(s)
Carrier Proteins/chemistry , Carrier Proteins/metabolism , Proteins/metabolism , Receptors, Cell Surface , Amino Acid Sequence , Animals , Antibodies , COS Cells , Cell Line , Cross-Linking Reagents , Cytoplasm/metabolism , Dimerization , Humans , Kinetics , Leptin , Molecular Sequence Data , Obesity , Peptide Fragments/chemistry , Peptide Fragments/immunology , Receptors, Leptin , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Spodoptera , Transfection
19.
J Biol Chem ; 274(49): 34838-45, 1999 Dec 03.
Article in English | MEDLINE | ID: mdl-10574956

ABSTRACT

We constructed chimeric receptors wherein the extracellular domain of the erythropoietin receptor (EpoR) was fused to the transmembrane and intracellular domains of the interferon (IFN) type I receptor subunits, IFNaR1 or IFNaR2-2. Transfection into 2fTGH and Tyk2-deficient 11,1 cells showed that EpoR/IFNaR2-2 alone was able to transduce a signal upon stimulation with erythropoietin (Epo), as judged by induction of the interferon type I-inducible 6-16 promoter. In contrast, protection against infection with encephalomyocarditis virus or vesicular stomatitis virus was reduced or absent, respectively. To further investigate the role of IFNaR1 in the induction of an antiviral state, we analyzed the Epo- versus IFNalpha-induced transcription of a set of genes, involved in antiviral protection. Up to 24 h after stimulation with Epo or IFNalpha, comparable transcription of the p56, dsRNA-dependent protein kinase, 2'-5'A synthetase, and MxA genes was seen. However, at later time points, only in the case of Epo induction, a sharp decrease of mRNA levels was observed. Western blotting analysis of dsRNA-dependent protein kinase showed a similar pattern at the protein level. Taken together, our results imply a role for IFNaR1 in the induction of sustained mRNA and protein levels that are likely required for optimal antiviral activity.


Subject(s)
Gene Expression Regulation , Interferon-alpha/metabolism , Interferon-beta/metabolism , Receptors, Interferon/metabolism , Alkaline Phosphatase/metabolism , Dimerization , Dose-Response Relationship, Drug , Fibrosarcoma/metabolism , Humans , Membrane Proteins , Promoter Regions, Genetic , RNA, Messenger/metabolism , Receptor, Interferon alpha-beta , Receptors, Erythropoietin/metabolism , Receptors, Interferon/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction , Temperature , Time Factors , Transcription, Genetic , Transfection
20.
Eur J Biochem ; 249(3): 690-700, 1997 Nov 01.
Article in English | MEDLINE | ID: mdl-9395315

ABSTRACT

Anti-human interleukin-6 (human IL-6) and anti-human IL-6 receptor (IL-6R)-neutralising monoclonal antibodies (mAbs) are among the most promising human IL-6-specific inhibitors and have been shown to exert short-term beneficial effects in clinical trials. Simultaneous treatment with different anti-human IL-6 or anti-human IL-6R mAbs was recently suggested to be a potent way to inhibit the action of the cytokine in vivo. Although some of these mAbs are already used, their mechanisms of action and the location of their epitopes on the surface of human IL-6 and human IL-6R are still unknown. Here, we analysed the capacity of several anti-human IL-6 and anti-human IL-6R mAbs to inhibit the interaction between human IL-6, human IL-6R, and human glycoprotein 130 (gp130). We mapped the epitopes of several of these mAbs by studying their binding to human IL-6 and human IL-6R mutant proteins. Our results show that several anti-human IL-6 and anti-human IL-6R-neutralising mAbs block the binding between human IL-6 and human IL-6R, whereas others block the binding to gp130. We provide evidence that some of the latter mAbs inhibit interaction with gp130beta1, whereas others interfere with the binding to gp130beta2. Our results suggest that residues included in the C'D' loop of human IL-6R interact with gp130beta2.


Subject(s)
Antibodies, Monoclonal/pharmacology , Interleukin-6/immunology , Receptors, Interleukin-6/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/metabolism , Antigens, CD/metabolism , Antigens, CD/pharmacology , Cell Line , Cytokine Receptor gp130 , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay , Epitope Mapping , Humans , Interleukin-6/antagonists & inhibitors , Interleukin-6/chemistry , Interleukin-6/genetics , Interleukin-6/metabolism , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/pharmacology , Mice , Models, Molecular , Neutralization Tests , Precipitin Tests , Protein Conformation , Protein Structure, Tertiary , Receptors, Interleukin-6/antagonists & inhibitors , Receptors, Interleukin-6/chemistry , Receptors, Interleukin-6/genetics , Receptors, Interleukin-6/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL