Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Eur J Immunol ; 54(1): e2350658, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37816219

ABSTRACT

Expression levels of the chemokine receptor CX3CR1 serve as high-resolution marker delineating functionally distinct antigen-experienced T-cell states. The factors that influence CX3CR1 expression in T cells are, however, incompletely understood. Here, we show that in vitro priming of naïve CD8+ T cells failed to robustly induce CX3CR1, which highlights the shortcomings of in vitro priming settings in recapitulating in vivo T-cell differentiation. Nevertheless, in vivo generated memory CD8+ T cells maintained CX3CR1 expression during culture. This allowed us to investigate whether T-cell receptor ligation, cell death, and CX3CL1 binding influence CX3CR1 expression. T-cell receptor stimulation led to downregulation of CX3CR1. Without stimulation, CX3CR1+ CD8+ T cells had a selective survival disadvantage, which was enhanced by factors released from necrotic but not apoptotic cells. Exposure to CX3CL1 did not rescue their survival and resulted in a dose-dependent loss of CX3CR1 surface expression. At physiological concentrations of CX3CL1, CX3CR1 surface expression was only minimally reduced, which did not hamper the interpretability of T-cell differentiation states delineated by CX3CR1. Our data further support the broad utility of CX3CR1 surface levels as T-cell differentiation marker and identify factors that influence CX3CR1 expression and the maintenance of CX3CR1 expressing CD8+ T cells.


Subject(s)
CD8-Positive T-Lymphocytes , Receptors, Chemokine , CD8-Positive T-Lymphocytes/metabolism , Receptors, Chemokine/genetics , Cellular Microenvironment , Receptors, Antigen, T-Cell/metabolism , CX3C Chemokine Receptor 1/metabolism
2.
Br J Anaesth ; 126(2): 467-476, 2021 02.
Article in English | MEDLINE | ID: mdl-33183737

ABSTRACT

BACKGROUND: Postoperative neurocognitive decline is a frequent complication in adult patients undergoing major surgery with increased risk for morbidity and mortality. The mechanisms behind cognitive decline after anaesthesia and surgery are not known. We studied the association between CSF and blood biomarkers of neuronal injury or brain amyloidosis and long-term changes in neurocognitive function. METHODS: In patients undergoing major orthopaedic surgery (knee or hip replacement), blood and CSF samples were obtained before surgery and then at 4, 8, 24, 32, and 48 h after skin incision through an indwelling spinal catheter. CSF and blood concentrations of total tau (T-tau), neurofilament light, neurone-specific enolase and amyloid ß (Aß1-42) were measured. Neurocognitive function was assessed using the International Study of Postoperative Cognitive Dysfunction (ISPOCD) test battery 1-2 weeks before surgery, at discharge from the hospital (2-5 days after surgery), and at 3 months after surgery. RESULTS: CSF and blood concentrations of T-tau, neurone-specific enolase, and Aß1-42 increased after surgery. A similar increase in serum neurofilament light was seen with no overall changes in CSF concentrations. There were no differences between patients having a poor or good late postoperative neurocognitive outcome with respect to these biomarkers of neuronal injury and Aß1-42. CONCLUSIONS: The findings of the present explorative study showed that major orthopaedic surgery causes a release of CSF markers of neural injury and brain amyloidosis, suggesting neuronal damage or stress. We were unable to detect an association between the magnitude of biomarker changes and long-term postoperative neurocognitive dysfunction.


Subject(s)
Amyloidosis/cerebrospinal fluid , Arthroplasty, Replacement, Hip/adverse effects , Arthroplasty, Replacement, Knee/adverse effects , Biomarkers/cerebrospinal fluid , Brain Injuries/cerebrospinal fluid , Postoperative Cognitive Complications/etiology , Aged , Amyloid beta-Peptides/cerebrospinal fluid , Amyloidosis/complications , Amyloidosis/diagnosis , Brain Injuries/complications , Brain Injuries/diagnosis , Cognition , Female , Humans , Male , Neurofilament Proteins/cerebrospinal fluid , Peptide Fragments/cerebrospinal fluid , Phosphopyruvate Hydratase/cerebrospinal fluid , Postoperative Cognitive Complications/cerebrospinal fluid , Postoperative Cognitive Complications/diagnosis , Postoperative Cognitive Complications/psychology , Prospective Studies , Risk Assessment , Risk Factors , Time Factors , Treatment Outcome , tau Proteins/cerebrospinal fluid
3.
Mol Med ; 25(1): 13, 2019 04 11.
Article in English | MEDLINE | ID: mdl-30975096

ABSTRACT

BACKGROUND: Extracellular high mobility group box 1 protein  (HMGB1) serves a central role in inflammation as a transporter protein, which binds other immune-activating molecules that are endocytosed via the receptor for advanced glycation end-products (RAGE). These pro-inflammatory complexes are targeted to the endolysosomal compartment, where HMGB1 permeabilizes the lysosomes. This enables HMGB1-partner molecules to avoid degradation, to leak into the cytosol, and to reach cognate immune-activating sensors. Lipopolysaccharide (LPS) requires this pathway to generate pyroptosis by accessing its key cytosolic receptors, murine caspase 11, or the human caspases 4 and 5. This lytic, pro-inflammatory cell death plays a fundamental pathogenic role in gram-negative sepsis. The aim of the study was to identify molecules inhibiting HMGB1 or HMGB1/LPS cellular internalization. METHODS: Endocytosis was studied in cultured macrophages using Alexa Fluor-labeled HMGB1 or complexes of HMGB1 and Alexa Fluor-labeled LPS in the presence of an anti-HMGB1 monoclonal antibody (mAb), recombinant HMGB1 box A protein, acetylcholine, the nicotinic acetylcholine receptor subtype alpha 7 (α7 nAChR) agonist GTS-21, or a dynamin-specific inhibitor of endocytosis. Images were obtained by fluorescence microscopy and quantified by the ImageJ processing program (NIH). Data were analyzed using student's t test or one-way ANOVA followed by the least significant difference or Tukey's tests. RESULTS: Anti-HMGB1 mAb, recombinant HMGB1 antagonist box A protein, acetylcholine, GTS-21, and the dynamin-specific inhibitor of endocytosis inhibited internalization of HMGB1 or HMGB1-LPS complexes in cultured macrophages. These agents prevented macrophage activation in response to HMGB1 and/or HMGB1-LPS complexes. CONCLUSION: These results demonstrate that therapies based on HMGB1 antagonists and the cholinergic anti-inflammatory pathway share a previously unrecognized molecular mechanism of substantial clinical relevance.


Subject(s)
HMGB1 Protein/metabolism , Lipopolysaccharides/pharmacology , Receptor for Advanced Glycation End Products/metabolism , Acetylcholine/pharmacology , Animals , Cells, Cultured , Cholinergic Agonists/pharmacology , Endocytosis/drug effects , Enzyme-Linked Immunosorbent Assay , Inflammation/metabolism , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Microscopy, Fluorescence , RAW 264.7 Cells
4.
J Neuroinflammation ; 16(1): 45, 2019 Feb 19.
Article in English | MEDLINE | ID: mdl-30782181

ABSTRACT

BACKGROUND: Amyotrophic lateral sclerosis (ALS) is a fatal and rapidly progressing neurodegenerative disease without effective treatment. The receptor for advanced glycation end products (RAGE) and the toll-like receptor (TLR) system are major components of the innate immune system, which have been implicated in ALS pathology. Extracellularly released high-mobility group box 1 (HMGB1) is a pleiotropic danger-associated molecular pattern (DAMP), and is an endogenous ligand for both RAGE and TLR4. METHODS: The present study examined the effect of HMGB1 inhibition on disease progression in the preclinical SOD1G93A transgenic mouse model of ALS using a potent anti-HMGB1 antibody (2G7), which targets the extracellular DAMP form of HMGB1. RESULTS: We found that chronic intraperitoneal dosing of the anti-HMGB1 antibody to SOD1G93A mice transiently improved hind-limb grip strength early in the disease, but did not extend survival. Anti-HMGB1 treatment also reduced tumour necrosis factor α and complement C5a receptor 1 gene expression in the spinal cord, but did not affect overall glial activation. CONCLUSIONS: In summary, our results indicate that therapeutic targeting of an extracellular DAMP, HMGB1, improves early motor dysfunction, but overall has limited efficacy in the SOD1G93A mouse model of ALS.


Subject(s)
Amyotrophic Lateral Sclerosis , HMGB1 Protein/antagonists & inhibitors , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Amyotrophic Lateral Sclerosis/physiopathology , Animals , Disease Models, Animal , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Activity/physiology , Muscle Strength/physiology , Mutation , Spinal Cord/pathology , Superoxide Dismutase/genetics , Superoxide Dismutase-1/genetics
5.
Gerontology ; 63(4): 337-349, 2017.
Article in English | MEDLINE | ID: mdl-28427050

ABSTRACT

BACKGROUND: It is known from clinical practice and observational studies that elderly patients with a diagnosis of inflammatory rheumatic diseases (IRD) bear a significantly increased risk for cardiovascular diseases such as coronary artery disease (CAD) and heart failure. The molecular mechanism, however, is still not known. Recently, high mobility group protein B1 (HMGB1), a ubiquitous, highly conserved single polypeptide expressed in all mammal eukaryotic cells, has been identified to mediate myocardial dysfunction in vitro once released from the nuclei of cardiomyocytes. OBJECTIVE: To investigate whether HMGB1 and its receptors are expressed in cardiac muscles of elderly patients with CAD with or without IRD. METHODS: HMGB1 and its 3 well-known receptors, receptor for advanced glycation end products, Toll-like receptor 2 (TLR2), and TLR4, were examined by immunohistochemistry on myocardial biopsy specimens from 18 elderly patients with CAD (10 with IRD, 8 without IRD). Furthermore, total HMGB1 protein levels were measured by Western blot from the cardiac biopsies in 5 patients with and 5 without IRD. RESULTS: Pathologic cytosolic HMGB1 in cardiomyocytes was massively recorded in all patients with IRD, but only slightly expressed in 1 patient without IRD. Total HMGB1 levels were also consistently lower in myocardial muscle biopsies of patients with IRD compared to those without IRD. Furthermore, all 3 HMGB1 receptors were expressed in cardiomyocytes of all patients. CONCLUSION: The increased cytosolic expression of HMGB1 in cardiomyocytes and the lower total amount of HMGB1 in the cardiac specimens of IRD patients is consistent with a greater release of HMGB1 from the myocardial nuclei in IRD than non-IRD individuals. Thus, the HMGB1 signaling pathways may be more easily activated in elderly CAD patients with concomitant IRD and trigger a detrimental inflammatory process causing severe cardiovascular problems. Therefore, targeting HMGB1 in IRD patients might reduce the risk for cardiovascular events.


Subject(s)
Coronary Artery Disease/complications , Coronary Artery Disease/metabolism , HMGB1 Protein/metabolism , Myocardium/metabolism , Rheumatic Diseases/complications , Rheumatic Diseases/metabolism , Aged , Blotting, Western , Coronary Vessels/metabolism , Endocardium/metabolism , Female , Humans , Immunohistochemistry , Male , Middle Aged , Myocytes, Cardiac/metabolism , Pericardium/metabolism , Receptor for Advanced Glycation End Products/metabolism , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism
6.
Ann Rheum Dis ; 72(8): 1390-9, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23148306

ABSTRACT

OBJECTIVES: Polymyositis and dermatomyositis are characterised by muscle weakness and fatigue even in patients with normal muscle histology via unresolved pathogenic mechanisms. In this study, we investigated the mechanisms by which high mobility group box protein 1 (HMGB1) acts to accelerate muscle fatigue development. METHODS: Intact single fibres were dissociated from flexor digitorum brevis (FDB) of wild type, receptor for advanced glycation endproduct (RAGE) knockout and toll like receptor 4 (TLR4) knockout mice and cultured in the absence or presence of recombinant HMGB1. A decrease in sarcoplasmic reticulum Ca(2+) release during a series of 300 tetanic contractions, which reflects the development of muscle fatigue, was determined by measuring myoplasmic free tetanic Ca(2+). TLR4 and major histocompatibility complex (MHC)-class I expression in mouse FDB fibres were investigated by immunofluorescence and confocal microscopy. Immunohistochemistry was used to investigate TLR4, MHC-class I and myosin heavy chain expression in muscle fibres of patients. RESULTS: Our results demonstrate that TLR4 is expressed in human and mouse skeletal muscle fibres, and coexpressed with MHC-class I in muscle fibres of patients with myositis. Furthermore, we show that HMGB1 acts via TLR4 but not RAGE to accelerate muscle fatigue and to induce MHC-class I expression in vitro. In order to bind and signal via TLR4, HMGB1 must have a reduced cysteine 106 and a disulphide linkage between cysteine 23 and 45. CONCLUSIONS: The HMGB1-TLR4 pathway may play an important role in causing muscle fatigue in patients with polymyositis or dermatomyositis and thus is a potential novel target for future therapy.


Subject(s)
HMGB1 Protein/pharmacology , Muscle Fatigue/drug effects , Myositis/metabolism , Toll-Like Receptor 4/metabolism , Adult , Aged , Animals , Calcium/metabolism , Cells, Cultured , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Muscle Contraction/drug effects , Muscle Contraction/physiology , Muscle Fibers, Skeletal , Myosin Heavy Chains/metabolism , Myositis/pathology , Receptor for Advanced Glycation End Products , Receptors, Immunologic/deficiency , Receptors, Immunologic/metabolism , Recombinant Proteins , Sarcoplasmic Reticulum/drug effects , Sarcoplasmic Reticulum/metabolism , Toll-Like Receptor 4/deficiency
7.
J Exp Med ; 212(1): 5-14, 2015 Jan 12.
Article in English | MEDLINE | ID: mdl-25559892

ABSTRACT

Innate immune receptors for pathogen- and damage-associated molecular patterns (PAMPs and DAMPs) orchestrate inflammatory responses to infection and injury. Secreted by activated immune cells or passively released by damaged cells, HMGB1 is subjected to redox modification that distinctly influences its extracellular functions. Previously, it was unknown how the TLR4 signalosome distinguished between HMGB1 isoforms. Here we demonstrate that the extracellular TLR4 adaptor, myeloid differentiation factor 2 (MD-2), binds specifically to the cytokine-inducing disulfide isoform of HMGB1, to the exclusion of other isoforms. Using MD-2-deficient mice, as well as MD-2 silencing in macrophages, we show a requirement for HMGB1-dependent TLR4 signaling. By screening HMGB1 peptide libraries, we identified a tetramer (FSSE, designated P5779) as a specific MD-2 antagonist preventing MD-2-HMGB1 interaction and TLR4 signaling. P5779 does not interfere with lipopolysaccharide-induced cytokine/chemokine production, thus preserving PAMP-mediated TLR4-MD-2 responses. Furthermore, P5779 can protect mice against hepatic ischemia/reperfusion injury, chemical toxicity, and sepsis. These findings reveal a novel mechanism by which innate systems selectively recognize specific HMGB1 isoforms. The results may direct toward strategies aimed at attenuating DAMP-mediated inflammation while preserving antimicrobial immune responsiveness.


Subject(s)
HMGB1 Protein/metabolism , Lymphocyte Antigen 96/metabolism , Signal Transduction , Toll-Like Receptor 4/metabolism , Acetaminophen , Animals , Blotting, Western , Cell Line , Cell Line, Tumor , Cells, Cultured , Chemical and Drug Induced Liver Injury/blood , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/metabolism , Cytokines/blood , Cytokines/pharmacology , Disulfides/metabolism , HMGB1 Protein/pharmacology , Lipopolysaccharides/pharmacology , Lymphocyte Antigen 96/chemistry , Lymphocyte Antigen 96/genetics , Macrophages/drug effects , Macrophages/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Models, Molecular , Peptides/chemistry , Peptides/metabolism , Peptides/pharmacology , Protein Binding/drug effects , Protein Structure, Tertiary , RNA Interference , Reperfusion Injury/blood , Reperfusion Injury/metabolism , Survival Analysis
8.
Proc Natl Acad Sci U S A ; 101(1): 296-301, 2004 Jan 06.
Article in English | MEDLINE | ID: mdl-14695889

ABSTRACT

Despite significant advances in intensive care therapy and antibiotics, severe sepsis accounts for 9% of all deaths in the United States annually. The pathological sequelae of sepsis are characterized by a systemic inflammatory response, but experimental therapeutics that target specific early inflammatory mediators [tumor necrosis factor (TNF) and IL-1beta] have not proven efficacious in the clinic. We recently identified high mobility group box 1 (HMGB1) as a late mediator of endotoxin-induced lethality that exhibits significantly delayed kinetics relative to TNF and IL-1beta. Here, we report that serum HMGB1 levels are increased significantly in a standardized model of murine sepsis, beginning 18 h after surgical induction of peritonitis. Specific inhibition of HMGB1 activity [with either anti-HMGB1 antibody (600 microg per mouse) or the DNA-binding A box (600 microg per mouse)] beginning as late as 24 h after surgical induction of peritonitis significantly increased survival (nonimmune IgG-treated controls = 28% vs. anti-HMGB1 antibody group = 72%, P < 0.03; GST control protein = 28% vs. A box = 68%, P < 0.03). Animals treated with either HMGB1 antagonist were protected against the development of organ injury, as evidenced by improved levels of serum creatinine and blood urea nitrogen. These observations demonstrate that specific inhibition of endogenous HMGB1 therapeutically reverses lethality of established sepsis indicating that HMGB1 inhibitors can be administered in a clinically relevant time frame.


Subject(s)
HMGB1 Protein/antagonists & inhibitors , Sepsis/drug therapy , Animals , Antibodies/administration & dosage , Cell Line , DNA, Complementary/genetics , Disease Models, Animal , HMGB1 Protein/blood , HMGB1 Protein/genetics , Humans , In Vitro Techniques , Kinetics , Mice , Neutralization Tests , Peptide Fragments/chemistry , Peptide Fragments/genetics , Sepsis/blood , Sepsis/etiology
9.
Mol Med ; 9(1-2): 37-45, 2003.
Article in English | MEDLINE | ID: mdl-12765338

ABSTRACT

High mobility group box 1 (HMGB), a ubiquitous DNA-binding protein, has been implicated as a proinflammatory cytokine and late mediator of lethal endotoxemia. HMGB1 is released by activated macrophages. It amplifies and extends the inflammatory response by inducing cytokine release and mediating acute lung injury, anorexia, and the inflammatory response to tissue necrosis. The kinetics of HMGB1 release provide a wide therapeutic window for endotoxemia because extracellular levels of HMGB1 begin to increase 12 to 24 h after exposure to inflammatory stimuli. Here, we demonstrate that a DNA-binding domain of HMGB1, the B box, recapitulates the cytokine activity of full length HMGB1 and efficiently activates macrophages to release tumor necrosis factor (TNF) and other proinflammatory cytokines. Truncation of the B box revealed that the TNF-stimulating activity localizes to 20 amino acids (HMGB1 amino acids 89 to 108). Passive immunization of mice with antibodies raised against B box conferred significant protection against lethal endotoxemia or sepsis, induced by cecal perforation. These results indicate that a proinflammatory domain of HMGB1 maps to the highly conserved DNA-binding B box, making this primary sequence a suitable target in the design of therapeutics.


Subject(s)
Cytokines/metabolism , HMGB1 Protein/chemistry , HMGB1 Protein/physiology , Animals , Cell Nucleus/metabolism , DNA Primers/chemistry , Electrophoretic Mobility Shift Assay , Endotoxemia/prevention & control , Glutathione Transferase/metabolism , Immunization, Passive , Lipopolysaccharides/pharmacology , Macrophage Activation , Macrophages/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Peptide Fragments/immunology , Peptide Fragments/metabolism , Polymerase Chain Reaction , Rabbits , Sepsis/immunology , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL