Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Front Immunol ; 9: 2118, 2018.
Article in English | MEDLINE | ID: mdl-30283452

ABSTRACT

Chemokine synergy-inducing molecules are emerging as regulating factors in cell migration. The alarmin HMGB1, in its reduced form, can complex with CXCL12 enhancing its activity on monocytes via the chemokine receptor CXCR4, while the form containing a disulfide bond, by binding to TLR2 or TLR4, initiates a cascade of events leading to production of cytokines and chemokines. So far, the possibility that the CXCL12/HMGB1 heterocomplex could be maintained in chronic inflammation was debated, due to the release of reactive oxygen species. Therefore, we have assessed if the heterocomplex could remain active in Rheumatoid Arthritis (RA) and its relevance in the disease assessment. Monocytes from RA patients with active disease require a low concentration of HMGB1 to enhance CXCL12-induced migration, in comparison to monocytes from patients in clinical remission or healthy donors. The activity of the heterocomplex depends on disease activity, on the COX2 and JAK/STAT pathways, and is determined by the redox potential of the microenvironment. In RA, the presence of an active thioredoxin system correlates with the enhanced cell migration, and with the presence of the heterocomplex in the synovial fluid. The present study highlights how, in an unbalanced microenvironment, the activity of the thioredoxin system plays a crucial role in sustaining inflammation. Prostaglandin E2 stimulation of monocytes from healthy donors is sufficient to recapitulate the response observed in patients with active RA. The activation of mechanisms counteracting the oxidative stress in the extracellular compartment preserves HMGB1 in its reduced form, and contributes to fuel the influx of inflammatory cells. Targeting the heterocomplex formation and its activity could thus be an additional tool for dampening the inflammation sustained by cell recruitment, for those patients with chronic inflammatory conditions who poorly respond to current therapies.


Subject(s)
Arthritis, Rheumatoid/metabolism , Cell Movement/drug effects , Chemokine CXCL12/pharmacology , HMGB1 Protein/pharmacology , Monocytes/drug effects , Adult , Aged , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/pathology , Cell Movement/immunology , Cells, Cultured , Dinoprostone/pharmacology , Drug Synergism , Female , Humans , Male , Middle Aged , Monocytes/immunology , Monocytes/metabolism , Oxidation-Reduction , Protein Binding/drug effects , Receptors, CXCR4/immunology , Receptors, CXCR4/metabolism , Signal Transduction/drug effects , Signal Transduction/immunology
2.
Mol Immunol ; 55(1): 76-82, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23207101

ABSTRACT

HMGB1 is a nuclear protein that is released or secreted following trauma or severe cellular stress. Extracellular HMGB1 triggers inflammation and recruits leukocytes to the site of tissue damage. We review recent evidence that the ability of HMGB1 to recruit leukocytes may be entirely due to the formation of a heterocomplex with the homeostatic chemokine CXCL12. The HMGB1-CXCL12 heterocomplex acts on the CXCR4 receptor more potently than CXCL12 alone. Notably, only one of the redox forms of HMGB1, the one where all cysteines are reduced (all-thiol), can bind CXCL12. Both HMGB1 containing a disulfide bond between C23 and C45, which induces chemokine and cytokine release by activating TLR4, and HMGB1 terminally oxidized to contain a cysteine sulfonate are inactive in recruiting leukocytes. Thus, the chemoattractant and cytokine-inducing activities of HMGB1 are separable, and we propose that they appear sequentially during the development of inflammation and its resolution. The HMGB1-CXCL12 heterocomplex constitutes a specific target that may hold promise for the treatment of several pathologies.


Subject(s)
Chemotaxis, Leukocyte/physiology , HMGB1 Protein/physiology , Inflammation/immunology , Wounds and Injuries/immunology , Animals , Cell Movement/genetics , Cell Movement/immunology , Chemokine CXCL12/metabolism , Chemokine CXCL12/physiology , Chemotaxis, Leukocyte/genetics , HMGB1 Protein/genetics , HMGB1 Protein/metabolism , Humans , Inflammation/pathology , Models, Biological , Oxidation-Reduction
3.
J Exp Med ; 209(9): 1519-28, 2012 Aug 27.
Article in English | MEDLINE | ID: mdl-22869893

ABSTRACT

Tissue damage causes inflammation, by recruiting leukocytes and activating them to release proinflammatory mediators. We show that high-mobility group box 1 protein (HMGB1) orchestrates both processes by switching among mutually exclusive redox states. Reduced cysteines make HMGB1 a chemoattractant, whereas a disulfide bond makes it a proinflammatory cytokine and further cysteine oxidation to sulfonates by reactive oxygen species abrogates both activities. We show that leukocyte recruitment and activation can be separated. A nonoxidizable HMGB1 mutant in which serines replace all cysteines (3S-HMGB1) does not promote cytokine production, but is more effective than wild-type HMGB1 in recruiting leukocytes in vivo. BoxA, a HMGB1 inhibitor, interferes with leukocyte recruitment but not with activation. We detected the different redox forms of HMGB1 ex vivo within injured muscle. HMGB1 is completely reduced at first and disulfide-bonded later. Thus, HMGB1 orchestrates both key events in sterile inflammation, leukocyte recruitment and their induction to secrete inflammatory cytokines, by adopting mutually exclusive redox states.


Subject(s)
Cytokines/metabolism , HMGB1 Protein/metabolism , Inflammation/metabolism , Leukocytes/cytology , Animals , Antibodies, Monoclonal/pharmacology , Cell Movement/drug effects , Chemotactic Factors/metabolism , Cysteine/metabolism , Disulfides/chemistry , Disulfides/metabolism , HMGB1 Protein/genetics , HMGB1 Protein/immunology , Leukocytes/metabolism , Male , Mice , Mice, Inbred C57BL , Muscle, Skeletal/injuries , Mutation , Rats , Reactive Oxygen Species/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
4.
Immunol Lett ; 145(1-2): 10-4, 2012 Jul 30.
Article in English | MEDLINE | ID: mdl-22698178

ABSTRACT

The arrest and directed migration of leukocytes during homeostasis, tumour development and inflammation is orchestrated by a multitude of chemokines, which govern leukocyte migratory activities. Immune cells are particularly adept at adjusting rapidly to changes within the environment by migration in response to chemokines. The confrontation of leukocytes with different combination of chemokines that are concomitantly produced under physiological or pathological conditions in vivo is complex. There are different ways to enhance or reduce leukocyte migration mediated by chemokines such as posttranslational modifications. Here, we described a positive regulatory mechanism in leukocyte trafficking, by the synergism between chemokines to rapidly augment the local leukocyte influx, thereby enhancing the outcome of an inflammatory response in vivo. The cellular mechanisms involved in chemokine synergy are still debated, but probably include chemokine and/or receptor heterodimerization and subsequent cooperation in signal transduction.


Subject(s)
Chemokines/immunology , Inflammation/pathology , Leukocytes/immunology , Animals , Cell Movement , Feedback, Physiological , Humans , Protein Processing, Post-Translational , Receptor Aggregation , Receptor Cross-Talk , Signal Transduction
5.
J Exp Med ; 209(3): 551-63, 2012 Mar 12.
Article in English | MEDLINE | ID: mdl-22370717

ABSTRACT

After tissue damage, inflammatory cells infiltrate the tissue and release proinflammatory cytokines. HMGB1 (high mobility group box 1), a nuclear protein released by necrotic and severely stressed cells, promotes cytokine release via its interaction with the TLR4 (Toll-like receptor 4) receptor and cell migration via an unknown mechanism. We show that HMGB1-induced recruitment of inflammatory cells depends on CXCL12. HMGB1 and CXCL12 form a heterocomplex, which we characterized by nuclear magnetic resonance and surface plasmon resonance, that acts exclusively through CXCR4 and not through other HMGB1 receptors. Fluorescence resonance energy transfer data show that the HMGB1-CXCL12 heterocomplex promotes different conformational rearrangements of CXCR4 from that of CXCL12 alone. Mononuclear cell recruitment in vivo into air pouches and injured muscles depends on the heterocomplex and is inhibited by AMD3100 and glycyrrhizin. Thus, inflammatory cell recruitment and activation both depend on HMGB1 via different mechanisms.


Subject(s)
Chemokine CXCL12/physiology , HMGB1 Protein/physiology , Inflammation/etiology , Receptors, CXCR4/physiology , Animals , Base Sequence , Calcium Signaling , Cell Movement/physiology , Chemokine CXCL12/chemistry , DNA, Complementary/genetics , Fibroblasts/physiology , Fluorescence Resonance Energy Transfer , HEK293 Cells , HMGB1 Protein/chemistry , Humans , Inflammation/pathology , Inflammation/physiopathology , MAP Kinase Signaling System , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Models, Molecular , Monocytes/physiology , Multiprotein Complexes/chemistry , NIH 3T3 Cells , Nuclear Magnetic Resonance, Biomolecular , Receptor for Advanced Glycation End Products , Receptors, CXCR4/chemistry , Receptors, CXCR4/genetics , Receptors, Immunologic/physiology , Signal Transduction , Surface Plasmon Resonance , Toll-Like Receptors/physiology , Transfection
6.
Int J Cancer ; 127(10): 2300-12, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20872671

ABSTRACT

Primary central nervous system lymphomas (PCNSL) are aggressive malignancies confined to the CNS, mostly of diffuse large B-cell histotype. Despite improved understanding of the malignant B cells, little is known on the tumor microenvironment and on the response of the adaptive immunity against PCNSL. We investigated the phenotype of tumor infiltrating lymphocytes (TILs), and the expression of chemokines that could affect malignant B cells and trafficking of TILs. TILs and chemokine expression were evaluated by immunohistochemistry and in situ hybridization. Furthermore, we performed in vitro migration assays to analyze the migratory capacity of lymphocytes and malignant B cells toward chemokines and chemokine heterocomplexes. We show in 22 cases of PCNSL from immunocompetent patients that CD8(+) T cells represent the majority of TILs in the tumor mass. They tend to accumulate in perivascular areas, show Granzyme B expression and proliferate in situ. Their localization and density correlates with the expression of the inflammatory chemokine CXCL9, which is transcribed and translated by perivascular macrophages and pericytes in the perivascular microenvironment. Moreover, CXCL9 and CXCL12 are coexpressed on the tumor vasculature and form heterocomplexes. In the presence of CXCL9, CXCL12-induced migration is enhanced not only on CXCR4(+)/CXCR3(+)/CD8(+) T cells but also on CXCR4(+)/CXCR3(-) malignant B cells. These findings indicate the presence of a strong chemoattractant stimulus in the perivascular microenvironment, which might serve as regulator for the recruitment of TILs and for the angiocentric positioning of malignant B cells in the perivascular cuff.


Subject(s)
B-Lymphocytes/immunology , Brain Neoplasms/immunology , Chemokine CXCL12/biosynthesis , Chemokine CXCL9/biosynthesis , Lymphocytes, Tumor-Infiltrating/immunology , Lymphoma, B-Cell/immunology , Adult , Aged , B-Lymphocytes/pathology , Brain Neoplasms/blood supply , Brain Neoplasms/pathology , CD8-Positive T-Lymphocytes/immunology , Cell Movement/immunology , Chemokine CXCL12/immunology , Chemokine CXCL9/immunology , Female , Humans , Immunohistochemistry , Lymphocyte Activation , Lymphocytes, Tumor-Infiltrating/pathology , Lymphoma, B-Cell/pathology , Male , Middle Aged , Receptors, CXCR3/biosynthesis , Receptors, CXCR3/immunology , Receptors, CXCR4/biosynthesis , Receptors, CXCR4/immunology
7.
Eur J Immunol ; 39(4): 1118-28, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19291700

ABSTRACT

The migration of monocytes to sites of inflammation is largely determined by their response to chemokines. Although the chemokine specificities and expression patterns of chemokine receptors are well defined, it is still a matter of debate how cells integrate the messages provided by different chemokines that are concomitantly produced in physiological or pathological situations in vivo. We present evidence for one regulatory mechanism of human monocyte trafficking. Monocytes can integrate stimuli provided by inflammatory chemokines in the presence of homeostatic chemokines. In particular, migration and cell responses could occur at much lower concentrations of the CCR2 agonists, in the presence of chemokines (CCL19 and CCL21) that per se do not act on monocytes. Binding studies on CCR2(+) cells showed that CCL19 and CCL21 do not compete with the CCR2 agonist CCL2. Furthermore, the presence of CCL19 or CCL21 could influence the degradation of CCL2 and CCL7 on cells expressing the decoy receptor D6. These findings disclose a new scenario to further comprehend the complexity of chemokine-based monocyte trafficking in a vast variety of human inflammatory disorders.


Subject(s)
Cell Movement/immunology , Chemotaxis, Leukocyte/immunology , Inflammation/immunology , Monocytes/immunology , Receptors, CCR2/immunology , Receptors, CCR7/immunology , Amino Acid Sequence , Cell Movement/drug effects , Chemokine CCL19/chemistry , Chemokine CCL19/immunology , Chemokine CCL19/pharmacology , Chemokine CCL2/immunology , Chemokine CCL2/pharmacology , Chemokine CCL21/chemistry , Chemokine CCL21/immunology , Chemokine CCL21/pharmacology , Chemokine CCL7/immunology , Chemokine CCL7/pharmacology , Chemotaxis, Leukocyte/drug effects , Extracellular Signal-Regulated MAP Kinases/immunology , Extracellular Signal-Regulated MAP Kinases/metabolism , Glycosaminoglycans/immunology , Glycosaminoglycans/metabolism , Humans , Inflammation/metabolism , Ligands , Molecular Sequence Data , Monocytes/drug effects , Monocytes/metabolism , Phosphorylation/immunology , Protein Structure, Tertiary , Receptors, CCR10/immunology , Receptors, CCR10/metabolism , Receptors, CCR2/agonists , Receptors, CCR2/chemistry , Receptors, CCR7/agonists , Receptors, CCR7/chemistry , Chemokine Receptor D6
SELECTION OF CITATIONS
SEARCH DETAIL
...