18 research outputs found

    HMGB1-valkuaisaineen merkitys verenkierron soluissa

    Get PDF
    The matrix of blood is a liquid plasma that transports molecules and blood cells within vessels lined by endothelial cells. High-mobility group B1 (HMGB1) is a protein expressed in blood cells. Under normal circumstances, HMGB1 is virtually absent from plasma, but during inflammation or trauma its level in plasma is increased. In resting and quiescent cells, HMGB1 is usually localized in the intracellular compartment, with the exception of motile cells that express HMGB1 on their outer surface to mediate cell migration. During cell transformation or immune cell activation HMGB1 can be actively secreted outside of the cell. Further, when a cell is damaged, HMGB1 can passively leak into extracellular environment. Extracellular HMGB1 can then participate in regulation of the immune response and under some conditions it can mediate lethality in systemic inflammatory response. The aim of this study was to evaluate the expression and functions of HMGB1 in cells of the vascular system and to investigate the prognostic value of circulating HMGB1 in severe sepsis and septic shock. HMGB1 was detected in platelets, leukocytes, and endothelial cells. HMGB1 was released from platelets and leukocytes, and it was found to mediate their adhesive and migratory functions. During severe infections the plasma levels of HMGB1 were elevated; however, no direct correlation with lethality was found. Further, the analysis of proinflammatory mechanisms suggested that HMGB1 forms complexes with other molecules to activate the immune system. In conclusion, HMGB1 is expressed in the cells of the vascular system, and it participates in inflammatory mechanisms by activating platelets and leukocytes and by mediating monocyte migration.Veren plasma ja verisolut kiertävät verenkierrossa endoteelisolujen peittämää verisuonistoa myöten. HMGB1 on valkuaisaine, jota veri- ja endoteelisolut ilmentävät. Aktivoimattomissa soluissa HMGB1 esiintyy yleensä solun sisällä, poikkeuksena aktiivisesti liikkuvat solut, jotka käyttävät solun ulkoista HMGB1-proteiinia liikkumiseen. Solun vaurioituessa tai valkosolun aktivoituessa ne vapauttavat HMGB1-valkuaisainetta ulos soluista, joten tulehduksen tai kudosvaurion yhteydessä plasman HMGB1-taso kohoaa. Solun ulkoinen HMGB1 säätelee elimistön puolustusmekanismeja, mutta voi joissakin olosuhteissa esim. verenmyrkytyksessä olla myös haitallinen. Väitöskirjatutkimukseni tarkoituksena on ollut selvittää HMGB1:n ilmentymistä ja sen välittämiä toimintoja verenkiertoelimistön soluissa. Lisäksi tarkoituksena on ollut tutkia plasman HMGB1-tasojen määrityksen käyttökelpoisuutta vakaviin infektioihin sairastuneiden potilaiden ennusteen laatimisessa. Työ tehtiin yhteistyössä ruotsalaisten ja yhdysvaltalaisten tutkimusryhmien kanssa. Työssä kehitettiin uusi HMGB1:n analyysimenetelmä. Lisäksi HMGB1:n eritysmekanismia ja tulehdusta välittävää mekanismia pystyttiin selvittämään, ja HMGB1:n tärkeä merkitys tulehdukseen liittyvässä veren valkosolujen kulkeutumisessa endoteelin läpi kudokseen kyettiin osoittamaan. Väitöskirjatyön tulokset tuovat lisätietoa HMGB1-valkuaisaineen merkityksestä tulehdustaudeissa ja auttavat kliinisten HMGB1- analyysimenetelmien kehitystyötä

    Regulation of Neurogenesis in Mouse Brain by HMGB1

    Get PDF
    The High Mobility Group Box 1 (HMGB1) is the most abundant nuclear nonhistone protein that is involved in transcription regulation. In addition, HMGB1 has previously been found as an extracellularly acting protein enhancing neurite outgrowth in cultured neurons. Although HMGB1 is widely expressed in the developing central nervous system of vertebrates and invertebrates, its function in the developing mouse brain is poorly understood. Here, we have analyzed developmental defects of the HMGB1 null mouse forebrain, and further examined our findings in ex vivo brain cell cultures. We find that HMGB1 is required for the proliferation and differentiation of neuronal stem cells/progenitor cells. Enhanced apoptosis is also found in the neuronal cells lacking HMGB1. Moreover, HMGB1 depletion disrupts Wnt/β-catenin signaling and the expression of transcription factors in the developing cortex, including Foxg1, Tbr2, Emx2, and Lhx6. Finally, HMGB1 null mice display aberrant expression of CXCL12/CXCR4 and reduced RAGE signaling. In conclusion, HMGB1 plays a critical role in mammalian neurogenesis and brain development

    AMIGO, a transmembrane protein implicated in axon tract development, defines a novel protein family with leucine-rich repeats

    Get PDF
    Ordered differential display identified a novel sequence induced in neurons by the neurite-promoting protein amphoterin. We named this gene amphoterin-induced gene and ORF (AMIGO), and also cloned two other novel genes homologous to AMIGO (AMIGO2 and AMIGO3). Together, these three AMIGOs form a novel family of genes coding for type I transmembrane proteins which contain a signal sequence for secretion and a transmembrane domain. The deduced extracellular parts of the AMIGOs contain six leucine-rich repeats (LRRs) flanked by cysteine-rich LRR NH2- and COOH-terminal domains and by one immunoglobulin domain close to the transmembrane region. A substrate-bound form of the recombinant AMIGO ectodomain promoted prominent neurite extension in hippocampal neurons, and in solution, the same AMIGO ectodomain inhibited fasciculation of neurites. A homophilic and heterophilic binding mechanism is shown between the members of the AMIGO family. Our results suggest that the members of the AMIGO protein family are novel cell adhesion molecules among which AMIGO is specifically expressed on fiber tracts of neuronal tissues and participates in their formation

    Inhibition of Homophilic Interactions and Ligand Binding of the Receptor for Advanced Glycation End Products by Heparin and Heparin-Related Carbohydrate Structures

    Get PDF
    Background: Heparin and heparin-related sulphated carbohydrates inhibit ligand binding of the receptor for advanced glycation end products (RAGE). Here, we have studied the ability of heparin to inhibit homophilic interactions of RAGE in living cells and studied how heparin related structures interfere with RAGE–ligand interactions. Methods: Homophilic interactions of RAGE were studied with bead aggregation and living cell protein-fragment complementation assays. Ligand binding was analyzed with microwell binding and chromatographic assays. Cell surface advanced glycation end product binding to RAGE was studied using PC3 cell adhesion assay. Results: Homophilic binding of RAGE was mediated by V1- and modulated by C2-domain in bead aggregation assay. Dimerisation of RAGE on the living cell surface was inhibited by heparin. Sulphated K5 carbohydrate fragments inhibited RAGE binding to amyloid β-peptide and HMGB1. The inhibition was dependent on the level of sulfation and the length of the carbohydrate backbone. α-d-Glucopyranosiduronic acid (glycyrrhizin) inhibited RAGE binding to advanced glycation end products in PC3 cell adhesion and protein binding assays. Further, glycyrrhizin inhibited HMGB1 and HMGB1 A-box binding to heparin. Conclusions: Our results show that K5 polysaccharides and glycyrrhizin are promising candidates for RAGE targeting drug development.Peer reviewe

    High Mobility Group Box 1 Protein Induction by Mycobacterium Bovis BCG

    Get PDF
    High mobility group box 1 protein (HMGB1), a nuclear protein, is a critical cytokine that mediates the response to infection, injury, and inflammation. The aim of our study was to elaborate a reliable in vitro model to investigate whether Mycobacterium bovis BCG is able to induce HMGB1 secretion from the monocytic U-937 cells. Western blot technique was applied for the detection of HMGB1 from supernatants of cells, following induction with Mycobacterium bovis BCG. Densitometric analysis revealed higher concentrations of HMGB1 in cell supernatants stimulated with BCG than in the supernatants of the control, nonstimulated cells. Further quantitation of the secreted HMGB1 was performed by ELISA. The BCG strain resulted in a higher amount of secreted HMGB1 (450 ± 44 ng/mL) than that of LPS (84 ± 12 ng/mL) or Staphylococcus aureus (150 ± 14 ng/mL). BCG and Phorbol −12-myristate −13 acetate (PMA), added together, resulted in the highest HMGB1 secretion (645 ± 125 ng/mL). The translocation of the HMGB1 towards the cytoplasm following infection of cells with BCG was demonstrated by immunofluorescence examinations. Conclusion: Our pilot experiments draw attention to the HMGB1 inducing ability of Mycobacterium bovis. Assesment of the pathophysiological role of this late cytokine in mycobacterial infections demands further in vitro and in vivo examinations

    Low-Molecular Weight Protamine Overcomes Chondroitin Sulfate Inhibition of Neural Regeneration

    Get PDF
    Protamine is an arginine-rich peptide that replaces histones in the DNA-protein complex during spermatogenesis. Protamine is clinically used in cardiopulmonary bypass surgery to neutralize the effects of heparin that is required during the treatment. Here we demonstrate that protamine and its 14-22 amino acid long fragments overcome the neurite outgrowth inhibition by chondroitin sulfate proteoglycans (CSPGs) that are generally regarded as major inhibitors of regenerative neurite growth after injuries of the adult central nervous system (CNS). Since the full-length protamine was found to have toxic effects on neuronal cells we used the in vitro neurite outgrowth assay to select a protamine fragment that retains the activity to overcome the neurite outgrowth inhibition on CSPG substrate and ended up in the 14 amino acid fragment, low-molecular weight protamine (LMWP). In contrast to the full-length protamine, LMWP displays very low or no toxicity in our assays in vitro and in vivo. We therefore started studies on LMWP as a possible drug lead in treatment of CNS injuries, such as the spinal cord injury (SCI). LMWP mimicks HB-GAM (heparin-binding growth-associated molecule; pleiotrophin) in that it overcomes the CSPG inhibition on neurite outgrowth in primary CNS neurons in vitro and inhibits binding of protein tyrosine phosphatase (PTP) sigma, an inhibitory receptor in neurite outgrowth, to its CSPG ligand. Furthermore, the chondroitin sulfate (CS) chains of the cell matrix even enhance the LMWP-induced neurite outgrowth on CSPG substrate. In vivo studies using the hemisection and hemicontusion SCI models in mice at the cervical level C5 revealed that LMWP enhances recovery when administered through intracerebroventricular or systemic route. We suggest that LMWP is a promising drug lead to develop therapies for CNS injuries.Peer reviewe

    The bradykinin system in stress and anxiety in humans and mice

    Get PDF
    Pharmacological research in mice and human genetic analyses suggest that the kallikrein-kinin system (KKS) may regulate anxiety. We examined the role of the KKS in anxiety and stress in both species. In human genetic association analysis, variants in genes for the bradykinin precursor (KNG1) and the bradykinin receptors (BDKRB1 and BDKRB2) were associated with anxiety disorders (p <0.05). In mice, however, neither acute nor chronic stress affected B1 receptor gene or protein expression, and B1 receptor antagonists had no effect on anxiety tests measuring approach-avoidance conflict. We thus focused on the B2 receptor and found that mice injected with the B2 antagonist WIN 64338 had lowered levels of a physiological anxiety measure, the stress-induced hyperthermia (SIH), vs controls. In the brown adipose tissue, a major thermoregulator, WIN 64338 increased expression of the mitochondrial regulator Pgc1 alpha and the bradykinin precursor gene Kng2 was upregulated after cold stress. Our data suggests that the bradykinin system modulates a variety of stress responses through B2 receptor-mediated effects, but systemic antagonists of the B2 receptor were not anxiolytic in mice. Genetic variants in the bradykinin receptor genes may predispose to anxiety disorders in humans by affecting their function.Peer reviewe

    Circulating nucleosomes as predictive markers of severe acute pancreatitis

    Get PDF
    Abstract Background The components of nucleosomes, which contain DNA and histones, are released into the circulation from damaged cells and can promote inflammation. We studied whether the on-admission levels of circulating nucleosomes predict the development of severe acute pancreatitis (AP), in particular among the patients who present without clinical signs of organ dysfunction. Methods This is a prospective study of 74 AP patients admitted to Helsinki University Hospital from 2003 to 2007. Twenty-three patients had mild, 27 moderately severe, and 24 severe AP as defined by the revised Atlanta criteria. 14/24 severe AP patients had no sign of organ dysfunction on admission (modified marshall score <2). Blood samples were obtained on admission and the plasma levels of nucleosomes were measured using enzyme-linked immunosorbent assay. Results The on-admission levels of nucleosomes were significantly higher in severe AP than in mild or moderately severe AP (p < 0.001 for all), higher in non-survivors (n = 8) than in survivors (p = 0.019), and correlated with the on-admission levels of C-reactive protein (p < 0.001) and creatinine (p < 0.001). Among the AP patients who presented without organ dysfunction, the on-admission nucleosome level was an independent predictor of severe AP (p = 0.038, gender-adjusted forward-stepping logistic regression). Conclusions Circulating nucleosome levels may be helpful in identifying, on admission to hospital, the AP patients who present without clinical signs of organ dysfunction, and, yet, are bound to develop organ dysfunction during hospitalization

    HMGB4 is expressed by neuronal cells and affects the expression of genes involved in neural differentiation

    Get PDF
    HMGB4 is a new member in the family of HMGB proteins that has been characterized in sperm cells, but little is known about its functions in somatic cells. Here we show that HMGB4 and the highly similar rat Transition Protein 4 (HMGB4L1) are expressed in neuronal cells. Both proteins had slow mobility in nucleus of living NIH-3T3 cells. They interacted with histones and their differential expression in transformed cells of the nervous system altered the post-translational modification statuses of histones in vitro. Overexpression of HMGB4 in HEK 293T cells made cells more susceptible to cell death induced by topoisomerase inhibitors in an oncology drug screening array and altered variant composition of histone H3. HMGB4 regulated over 800 genes in HEK 293T cells with a p-valuePeer reviewe

    Identification of HMGB1-Binding Components Using Affinity Column Chromatography

    Get PDF
    High Mobility Group B1 (HMGB1) is a 30 kDa protein widely expressed in mammalian cells. HMGB1 has a high content of charged amino acids and has a bipolar structure consisting of two highly positive amino terminal HMG-box domains and an acidic carboxy terminal tail. HMGB1 has nuclear functions regulating chromatin structure and gene expression and extracellular functions regulating immune response and cell motility. Biochemical and cell biological studies have revealed that HMGB1 binds to various kinds of biomolecules and these interactions are crucial for determining the in vivo functions of HMGB1. Albeit several different biochemical methods have been used to detect HMGB1- binding components, HMGB1-affinity column chromatography has rarely been applied in such studies. Here, we describe an affinity chromatography method that we have applied to isolation and identification of HMGB1-binding molecules from different cell types. Biomolecules recovered with HMGB1-affinity chromatography include proinflammatory bacterial DNA and glioblastoma cell histones H1 and H3 which all have previously been reported as HMGB1-binding molecules by other methods. Furthermore, an entirely new HMGB1-binding protein, Multimerin-1 containing complex, was identified from platelet lysates by HMGB1-affinity chromatography. Endogenous Multimerin-1 and HMGB1 were shown to associate on the surface of endothelial cells and activated platelets, and endogenous Multimerin-1 also regulated the release of HMGB1 from activated platelets. In conclusion, HMGB1-affinity chromatography can be used to isolate and characterize novel HMGB1-binding partners from a variety of cellular sources. Such new interactions reveal further complexity in the multi-faceted biology of the HMGB1.Non peer reviewe
    corecore