Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Biologicals ; 37(3): 139-40, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19328011

ABSTRACT

Appropriate adjuvants and formulations improve the immunogenicity of antigens, by increasing both the intensity and duration of immune responses. To that aim, the design and use of adjuvants must obey to the rules regulating physiological responses of the immune system, in particular the long-term development of memory lymphocytes. Here I will briefly discuss the main mechanisms of adjuvanticity at the light of recent knowledge on antigen presentation by B memory lymphocytes, the role of nonspecific stimulation of such cells for memory persistence, and the use of particulate antigens to target B cells, thus facilitating immunological T/B lymphocyte cooperation for getting optimal immune responses.


Subject(s)
Adjuvants, Immunologic , Antigens , B-Lymphocytes/immunology , Humans , Immunologic Memory
2.
Influenza Other Respir Viruses ; 1(2): 61-4, 2007 Mar.
Article in English | MEDLINE | ID: mdl-19432636

ABSTRACT

The fear of a potential pandemic with a highly pathogenic influenza A virus, such as the avian virus H5N1, has rightly prompted multidisciplinary reflections and calls for better preparedness all over the world. In terms of therapeutic aspects, most of the focus has been on vaccines and antivirals. The present 'opinion paper' intends to discuss a different therapeutic approach, although not mutually exclusive to the two others quoted above. We here propose an approach, based on well-documented experimental evidence in animal models, in which a short series of human monoclonal antibodies adapted to the probable pandemic strain, specific for external antigens of that influenza virus and shown in vitro and in experimental models to have neutralizing properties, are prepared and stockpiled for administration to people recently exposed to the pandemic virus.


Subject(s)
Immunization, Passive , Influenza, Human/immunology , Animals , Antibodies, Viral/immunology , Birds , Hemagglutinins/immunology , Humans , Influenza A Virus, H5N1 Subtype/immunology , Influenza in Birds/epidemiology , Influenza in Birds/immunology , Influenza in Birds/transmission , Influenza, Human/epidemiology , Influenza, Human/transmission , Mice , Virus Replication/immunology
3.
J Virol ; 80(6): 2949-57, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16501104

ABSTRACT

The C-type lectin DC-SIGN expressed on immature dendritic cells (DCs) captures human immunodeficiency virus (HIV) particles and enhances the infection of CD4+ T cells. This process, known as trans-enhancement of T-cell infection, has been related to HIV endocytosis. It has been proposed that DC-SIGN targets HIV to a nondegradative compartment within DCs and DC-SIGN-expressing cells, allowing incoming virus to persist for several days before infecting target cells. In this study, we provide several lines of evidence suggesting that intracellular storage of intact virions does not contribute to HIV transmission. We show that endocytosis-defective DC-SIGN molecules enhance T-cell infection as efficiently as their wild-type counterparts, indicating that DC-SIGN-mediated HIV internalization is dispensable for trans-enhancement. Furthermore, using immature DCs that are genetically resistant to infection, we demonstrate that several days after viral uptake, HIV transfer from DCs to T cells requires viral fusion and occurs exclusively through DC infection and transmission of newly synthesized viral particles. Importantly, our results suggest that DC-SIGN participates in this process by cooperating with the HIV entry receptors to facilitate cis-infection of immature DCs and subsequent viral transfer to T cells. We suggest that such a mechanism, rather than intracellular storage of incoming virus, accounts for the long-term transfer of HIV to CD4+ T cells and may contribute to the spread of infection by DCs.


Subject(s)
CD4-Positive T-Lymphocytes/virology , Cell Adhesion Molecules/metabolism , Dendritic Cells/virology , HIV Infections/transmission , HIV-1/pathogenicity , Lectins, C-Type/metabolism , Receptors, Cell Surface/metabolism , Amino Acid Sequence , Animals , Cell Adhesion Molecules/chemistry , Cell Adhesion Molecules/genetics , Cell Line , HIV Infections/virology , Humans , Lectins, C-Type/chemistry , Lectins, C-Type/genetics , Molecular Sequence Data , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/genetics
4.
J Biol Chem ; 280(25): 23698-708, 2005 Jun 24.
Article in English | MEDLINE | ID: mdl-15855154

ABSTRACT

Dengue virus (DV) is a mosquito-borne flavivirus that causes hemorrhagic fever in humans. In the natural infection, DV is introduced into human skin by an infected mosquito vector where it is believed to target immature dendritic cells (DCs) and Langerhans cells (LCs). We found that DV productively infects DCs but not LCs. We show here that the interactions between DV E protein, the sole mannosylated glycoprotein present on DV particles, and the C-type lectin dendritic cell-specific intercellular adhesion molecule 3-grabbing non-integrin (DC-SIGN) are essential for DV infection of DCs. Binding of mannosylated N-glycans on DV E protein to DC-SIGN triggers a rapid and efficient internalization of the viral glycoprotein. However, we observed that endocytosis-defective DC-SIGN molecules allow efficient DV replication, indicating that DC-SIGN endocytosis is dispensable for the internalization step in DV entry. Together, these results argue in favor of a mechanism by which DC-SIGN enhances DV entry and infection in cis. We propose that DC-SIGN concentrates mosquito-derived DV particles at the cell surface to allow efficient interaction with an as yet unidentified entry factor that is ultimately responsible for DV internalization and pH-dependent fusion into DCs.


Subject(s)
Cell Adhesion Molecules/physiology , Dengue Virus/physiology , Dengue/physiopathology , Lectins, C-Type/physiology , Receptors, Cell Surface/physiology , Base Sequence , Cell Line , DNA Primers , Flow Cytometry , Humans , Virion/physiology
5.
Blood ; 105(6): 2449-57, 2005 Mar 15.
Article in English | MEDLINE | ID: mdl-15536153

ABSTRACT

The WHIM syndrome is a rare immunodeficiency disorder characterized by warts, hypogammaglobulinemia, infections, and myelokathexis. Dominant heterozygous mutations of the gene encoding CXCR4, a G-protein-coupled receptor with a unique ligand, CXCL12, have been associated with this pathology. We studied patients belonging to 3 different pedigrees. Two siblings inherited a CXCR4 mutation encoding a novel C-terminally truncated receptor. Two unrelated patients were found to bear a wild-type CXCR4 open reading frame. Circulating lymphocytes and neutrophils from all patients displayed similar functional alterations of CXCR4-mediated responses featured by a marked enhancement of G-protein-dependent responses. This phenomenon relies on the refractoriness of CXCR4 to be both desensitized and internalized in response to CXCL12. Therefore, the aberrant dysfunction of the CXCR4-mediated signaling constitutes a common biologic trait of WHIM syndromes with different causative genetic anomalies. Responses to other chemokines, namely CCL4, CCL5, and CCL21, were preserved, suggesting that, in clinical forms associated with a wild-type CXCR4 open reading frame, the genetic anomaly might target an effector with some degree of selectivity for the CXCL12/CXCR4 axis. We propose that the sustained CXCR4 activity in patient cells accounts for the immune-hematologic clinical manifestations and the profusion of warts characteristic of the WHIM syndrome.


Subject(s)
Chemokines, CXC/immunology , Codon, Nonsense , Genetic Diseases, Inborn/genetics , Immunologic Deficiency Syndromes/genetics , Quantitative Trait, Heritable , Receptors, CXCR4/genetics , Adolescent , Adult , Chemokine CXCL12 , Chemokines, CC/immunology , Chemokines, CXC/genetics , Female , GTP-Binding Proteins/immunology , Genetic Diseases, Inborn/immunology , Genetic Diseases, Inborn/pathology , Heterozygote , Humans , Immunologic Deficiency Syndromes/immunology , Immunologic Deficiency Syndromes/pathology , Lymphocytes/immunology , Lymphocytes/pathology , Male , Neutrophils/immunology , Neutrophils/pathology , Open Reading Frames/genetics , Open Reading Frames/immunology , Pedigree , Receptors, CXCR4/immunology , Signal Transduction/genetics , Signal Transduction/immunology , Warts/genetics , Warts/immunology , Warts/pathology
6.
J Immunol ; 173(12): 7150-60, 2004 Dec 15.
Article in English | MEDLINE | ID: mdl-15585836

ABSTRACT

Interaction of HIV-1 envelope glycoprotein gp120 with the chemokine receptor CXCR4 triggers not only viral entry but also an array of signal transduction cascades. Whether gp120 induces an incomplete or aberrant set of signals, or whether it can function as a full CXCR4 agonist, remains unclear. We report that, in unstimulated human primary CD4(+) T cells, the spectrum of signaling responses induced by gp120 through CXCR4 paralleled that induced by the natural ligand stromal cell-derived factor 1/CXCL12. gp120 activated heterotrimeric G proteins and the major G protein-dependent pathways, including calcium mobilization, phosphoinositide-3 kinase, and Erk-1/2 MAPK activation. Interestingly, gp120 caused rapid actin cytoskeleton rearrangements and profuse membrane ruffling, as evidenced by dynamic confocal imaging. This coordinated set of events resulted in a bona fide chemotactic response. Inactivated HIV-1 virions that harbored conformationally intact envelope glycoproteins also caused actin polymerization and chemotaxis, while similar virions devoid of envelope glycoproteins did not. Thus gp120, in monomeric as well as oligomeric, virion-associated form, elicited a complex cellular response that mimicked the effects of a chemokine. HIV-1 has therefore the capacity to dysregulate the vast CD4(+) T cell population that expresses CXCR4. In addition, HIV-1 may exploit its chemotactic properties to retain potential target cells and locally perturb their cytoskeleton, thereby facilitating viral transmission.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Chemotaxis, Leukocyte/immunology , HIV Envelope Protein gp120/physiology , HIV-1/immunology , Receptors, CXCR4/physiology , Actins/metabolism , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/enzymology , Calcium/metabolism , Calcium Signaling/immunology , Cytoskeleton/metabolism , Heterotrimeric GTP-Binding Proteins/metabolism , Humans , Lymphocyte Activation/immunology , MAP Kinase Signaling System/immunology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphatidylinositol 3-Kinases/biosynthesis , Phosphatidylinositol 3-Kinases/physiology , Receptors, CXCR4/agonists , Virion/physiology
7.
J Biol Chem ; 279(31): 32035-45, 2004 Jul 30.
Article in English | MEDLINE | ID: mdl-15166245

ABSTRACT

The molecular mechanisms involved in the hepatic tropism of hepatitis C virus (HCV) have not been identified. We have shown previously that liver-expressed C-type lectins L-SIGN and DC-SIGN bind the HCV E2 glycoprotein with high affinity (Lozach, P. Y., Lortat-Jacob, H., de Lacroix de Lavalette, A., Staropoli, I., Foung, S., Amara, A., Houles, C., Fieschi, F., Schwartz, O., Virelizier, J. L., Arenzana-Seisdedos, F., and Altmeyer, R. (2003) J. Biol. Chem. 278, 20358-20366). To analyze the functional relevance of this interaction, we generated pseudotyped lentivirus particles presenting HCV glycoproteins E1 and E2 at the virion surface (HCV-pp). High mannose N-glycans are present on E1 and, to a lesser extent, on E2 proteins of mature infectious HCV-pp. Such particles bind to both L-SIGN and DC-SIGN, but they cannot use these receptors for entry into cells. However, infectious virus is transmitted efficiently when permissive Huh-7 cells are cocultured with HCV-pp bound to L-SIGN or to DC-SIGN-positive cell lines. HCV-pp transmission via L-SIGN or DC-SIGN is inhibited by characteristic inhibitors such as the calcium chelator EGTA and monoclonal antibodies directed against lectin carbohydrate recognition domains of both lectins. In support of the biological relevance of this phenomenon, dendritic cells expressing endogenous DC-SIGN transmitted HCV-pp with high efficiency in a DC-SIGN-dependent manner. Our results support the hypothesis that C-type lectins such as the liver sinusoidal endothelial cell-expressed L-SIGN could act as a capture receptor for HCV in the liver and transmit infectious virions to neighboring hepatocytes.


Subject(s)
Cell Adhesion Molecules/chemistry , Hepacivirus/metabolism , Lectins, C-Type/chemistry , Lectins/chemistry , Receptors, Cell Surface/chemistry , Viral Envelope Proteins/chemistry , Antibodies, Monoclonal/chemistry , Calcium/chemistry , Cell Line , Cell Separation , Chelating Agents/pharmacology , Dendritic Cells/metabolism , Egtazic Acid/chemistry , Flow Cytometry , Genetic Vectors , Glycoproteins/chemistry , Hepatocytes/metabolism , Humans , Lentivirus/genetics , Leukocytes, Mononuclear/metabolism , Liver/metabolism , Protein Binding
8.
EMBO Rep ; 4(7): 723-8, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12783086

ABSTRACT

Dengue virus (DV) is a mosquito-borne flavivirus that causes haemorrhagic fever in humans. DV primarily targets immature dendritic cells (DCs) after a bite by an infected mosquito vector. Here, we analysed the interactions between DV and human-monocyte-derived DCs at the level of virus entry. We show that the DC-specific ICAM3-grabbing non-integrin (DC-SIGN) molecule, a cell-surface, mannose-specific, C-type lectin, binds mosquito-cell-derived DVs and allows viral replication. Conclusive evidence for the involvement of DC-SIGN in DV infection was obtained by the inhibition of viral infection by anti-DC-SIGN antibodies and by the soluble tetrameric ectodomain of DC-SIGN. Our data show that DC-SIGN functions as a DV-binding lectin by interacting with the DV envelope glycoprotein. Mosquito-cell-derived DVs may have differential infectivity for DC-SIGN-expressing cells. We suggest that the differential use of DC-SIGN by viral envelope glycoproteins may account for the immunopathogenesis of DVs.


Subject(s)
Antigens, CD/metabolism , Cell Adhesion Molecules/metabolism , Culicidae/virology , Dendritic Cells/metabolism , Dendritic Cells/virology , Dengue Virus/physiology , Lectins, C-Type/metabolism , Receptors, Cell Surface/metabolism , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Cell Adhesion Molecules/antagonists & inhibitors , Cell Adhesion Molecules/immunology , Cells, Cultured , Dendritic Cells/cytology , Fluorescent Antibody Technique, Direct , Humans , Hydrogen-Ion Concentration , Lectins, C-Type/antagonists & inhibitors , Lectins, C-Type/immunology , Monocytes/cytology , Monocytes/metabolism , Monocytes/virology , Receptors, Cell Surface/antagonists & inhibitors , Receptors, Cell Surface/immunology
9.
J Biol Chem ; 278(22): 20358-66, 2003 May 30.
Article in English | MEDLINE | ID: mdl-12609975

ABSTRACT

The hepatitis C virus (HCV) genome codes for highly mannosylated envelope proteins, which are naturally retained in the endoplasmic reticulum. We found that the HCV envelope glycoprotein E2 binds the dendritic cell-specific intercellular adhesion molecule 3-grabbing nonintegrin (DC-SIGN) and the related liver endothelial cell lectin L-SIGN through high-mannose N-glycans. Competing ligands such as mannan and an antibody directed against the carbohydrate recognition domains (CRD) abrogated binding. While no E2 interaction with distant monomeric CRDs on biosensor chips could be detected, binding is observed if CRDs are closely seeded (Kd = 48 nm) and if the CRD is part of the oligomeric-soluble extracellular domain of DC-SIGN (Kd = 30 nm). The highest affinity is seen for plasma membrane-expressed DC-SIGN and L-SIGN (Kd = 3 and 6 nm, respectively). These results indicate that several high-mannose N-glycans in a structurally defined cluster on E2 bind to several subunits of the oligomeric lectin CRD. High affinity interaction of viral glycoproteins with oligomeric lectins might represent a strategy by which HCV targets to and concentrates in the liver and infects dendritic cells.


Subject(s)
Cell Adhesion Molecules/metabolism , Lectins, C-Type/metabolism , Receptors, Cell Surface/metabolism , Viral Envelope Proteins/metabolism , Cell Membrane/metabolism , DNA Primers , Mannose/metabolism , Protein Binding
10.
J Exp Med ; 197(4): 527-35, 2003 Feb 17.
Article in English | MEDLINE | ID: mdl-12591909

ABSTRACT

The clinical phenotype of interleukin 12 receptor beta1 chain (IL-12Rbeta1) deficiency and the function of human IL-12 in host defense remain largely unknown, due to the small number of patients reported. We now report 41 patients with complete IL-12Rbeta1 deficiency from 17 countries. The only opportunistic infections observed, in 34 patients, were of childhood onset and caused by weakly virulent Salmonella or Mycobacteria (Bacille Calmette-Guérin -BCG- and environmental Mycobacteria). Three patients had clinical tuberculosis, one of whom also had salmonellosis. Unlike salmonellosis, mycobacterial infections did not recur. BCG inoculation and BCG disease were both effective against subsequent environmental mycobacteriosis, but not against salmonellosis. Excluding the probands, seven of the 12 affected siblings have remained free of case-definition opportunistic infection. Finally, only five deaths occurred in childhood, and the remaining 36 patients are alive and well. Thus, a diagnosis of IL-12Rbeta1 deficiency should be considered in children with opportunistic mycobacteriosis or salmonellosis; healthy siblings of probands and selected cases of tuberculosis should also be investigated. The overall prognosis is good due to broad resistance to infection and the low penetrance and favorable outcome of infections. Unexpectedly, human IL-12 is redundant in protective immunity against most microorganisms other than Mycobacteria and Salmonella. Moreover, IL-12 is redundant for primary immunity to Mycobacteria and Salmonella in many individuals and for secondary immunity to Mycobacteria but not to Salmonella in most.


Subject(s)
Immunity, Innate , Receptors, Interleukin/deficiency , Adolescent , Adult , Cells, Cultured , Child , Child, Preschool , Humans , Mutation , Mycobacterium Infections/immunology , Opportunistic Infections/immunology , Polymorphism, Single-Stranded Conformational , Receptors, Interleukin/genetics , Receptors, Interleukin/physiology , Receptors, Interleukin-12 , Salmonella Infections/immunology
11.
J Virol ; 77(4): 2550-8, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12551993

ABSTRACT

The requirement of human immunodeficiency virus (HIV)-induced CCR5 activation for infection by R5 HIV type 1 (HIV-1) strains remains controversial. Ectopic CCR5 expression in CD4(+)-transformed cells or pharmacological inhibition of G(alpha)i proteins coupled to CCR5 left unsolved whether CCR5-dependent cell activation is necessary for the HIV life cycle. In this study, we investigated the role played by HIV-induced CCR5-dependent cell signaling during infection of primary CD4-expressing leukocytes. Using lentiviral vectors, we restored CCR5 expression in T lymphocytes and macrophages from individuals carrying the homozygous 32-bp deletion of the CCR5 gene (ccr5 Delta32/Delta32). Expression of wild-type (wt) CCR5 in ccr5 Delta32/Delta32 cells permitted infection by R5 HIV isolates. We assessed the capacity of a CCR5 derivative carrying a mutated DRY motif (CCR5-R126N) in the second intracellular loop to work as an HIV-1 coreceptor. The R126N mutation is known to disable G protein coupling and agonist-induced signal transduction through CCR5 and other G protein-coupled receptors. Despite its inability to promote either intracellular calcium mobilization or cell chemotaxis, the inactive CCR5-R126N mutant provided full coreceptor function to several R5 HIV-1 isolates in primary cells as efficiently as wt CCR5. We conclude that in a primary, CCR5-reconstituted CD4(+) cell environment, G protein signaling is dispensable for R5 HIV-1 isolates to actively infect primary CD4(+) T lymphocytes or macrophages.


Subject(s)
CD4-Positive T-Lymphocytes/virology , GTP-Binding Proteins/metabolism , HIV-1/pathogenicity , Macrophages/virology , Receptors, CCR5/metabolism , Signal Transduction , Cell Line , Cells, Cultured , HIV Infections/virology , HIV-1/physiology , Humans , Lentivirus/genetics , Receptors, CCR5/genetics , Virus Replication
12.
J Biol Chem ; 278(5): 3153-61, 2003 Jan 31.
Article in English | MEDLINE | ID: mdl-12431990

ABSTRACT

The contribution of raft domains to human immunodeficiency virus (HIV) 1 entry was assessed. In particular, we asked whether the CD4 and CCR5 HIV-1 receptors need to associate with sphingolipid-enriched, detergent-resistant membrane domains (rafts) to allow viral entry into primary and T-cell lines. Based on Triton X-100 solubilization and confocal microscopy, CD4 was shown to distribute partially to rafts. In contrast, CCR5 did not associate with rafts and localized in nonraft plasma membrane domains. HIV-1-receptor partitioning remained unchanged upon viral adsorption, suggesting that viral entry probably takes place outside rafts. To directly investigate this possibility, we targeted CD4 to nonraft domains of the membrane by preventing CD4 palmitoylation and interaction with p56(lck). Directed mutagenesis of both targeting signals significantly prevented association of CD4 with rafts, but did not suppress the HIV-1 receptor function of CD4. Collectively, these results strongly suggest that the presence of HIV-1 receptors in rafts is not required for viral infection. We show, however, that depleting plasma membrane cholesterol inhibits HIV-1 entry. We therefore propose that cholesterol modulates the HIV-1 entry process independently of its ability to promote raft formation.


Subject(s)
CD4 Antigens/immunology , HIV-1/physiology , Membrane Microdomains/virology , Receptors, CCR5/immunology , Receptors, HIV/physiology , Sphingolipids/pharmacology , T-Lymphocytes/immunology , Base Sequence , Cell Line , Cell Membrane/drug effects , Cell Membrane/virology , Cholera Toxin/pharmacology , Cholesterol/pharmacology , DNA Primers , HIV-1/drug effects , Humans , Membrane Microdomains/drug effects , Receptors, HIV/immunology
13.
AIDS ; 16(17): 2337-40, 2002 Nov 22.
Article in English | MEDLINE | ID: mdl-12441808

ABSTRACT

The susceptibility of phenotypically CCR5-negative resting CD4 T cells for membrane fusion with a CCR5-specific HIV-1 envelope was analysed using a novel sensitive fusion assay. A very low overall density of CCR5 on T cells expressing high levels of CD4 was shown to be sufficient for HIV envelope-mediated membrane fusion. These findings are relevant to the understanding of how HIV-1 R5 strains enter and replicate in resting CD4 T cells in vivo.


Subject(s)
CD4-Positive T-Lymphocytes/virology , HIV Envelope Protein gp120/metabolism , HIV-1/metabolism , Receptors, CCR5/metabolism , Antibodies, Monoclonal/immunology , CD4-Positive T-Lymphocytes/metabolism , Humans , Lymphocyte Activation , Receptors, CXCR4/metabolism
14.
J Biol Chem ; 277(18): 15677-89, 2002 May 03.
Article in English | MEDLINE | ID: mdl-11867624

ABSTRACT

Activation of CXCR4 by the CXC chemokine stromal cell-derived factor-1 (SDF-1) requires interaction of the amino-terminal domains of both molecules. We report that proteinases released from either mononucleated blood cells or polymorphonuclear neutrophils degranulated by inflammatory stimuli generate an SDF-1 fragment that is deleted from amino-terminal residues Lys(1)-Pro(2)-Val(3), as characterized by mass spectrometry analysis. The proteolyzed chemokine fails to induce agonistic functions and is unable to prevent the fusogenic capacity of CXCR4-tropic human immunodeficiency viruses. Furthermore, we observed that exposure of CXCR4-expressing cells to leukocyte proteinases results in the proteolysis of the extracellular amino-terminal domain of the receptor, as assessed by flow cytometry analysis and electrophoretic separation of immunoprecipitated CXCR4. Blockade of SDF-1 and CXCR4 proteolysis by the specific leukocyte elastase inhibitor, N-methoxysuccinyl-alanine-alanine-proline-valine-chloromethyl ketone, identified elastase as the major enzyme among leukocyte-secreted proteinases that accounts for inactivation of both SDF-1 and CXCR4. Indeed, purified leukocyte elastase generated in either SDF-1 or CXCR4 a pattern of cleavage indistinguishable from that observed with leukocyte-secreted proteinases. Our findings suggest that elastase-mediated proteolysis of SDF-1/CXCR4 is part of a mechanism regulating their biological functions in both homeostatic and pathologic processes.


Subject(s)
Chemokines, CXC/metabolism , Leukocyte Elastase/metabolism , Receptors, CXCR4/metabolism , Animals , Antibodies, Monoclonal/pharmacology , Binding Sites , Calcium/metabolism , Cell Fusion , Chemokine CXCL12 , Chemokines, CXC/pharmacology , Cytosol/metabolism , HIV-1/physiology , Humans , Jurkat Cells , Leukocytes, Mononuclear/enzymology , Leukocytes, Mononuclear/physiology , Mice , Oligopeptides/metabolism , Oligopeptides/pharmacology , Sequence Deletion , Spectrometry, Mass, Electrospray Ionization , Stromal Cells/metabolism , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...