Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Eur Rev Med Pharmacol Sci ; 24(9): 4729-4737, 2020 05.
Article in English | MEDLINE | ID: mdl-32432736

ABSTRACT

OBJECTIVE: To investigate the function of HMGB2 in renal tumor ACHN cells in vitro and in vivo and to study the underlying molecular mechanisms. PATIENTS AND METHODS: Kaplan-Meier analysis was used to study the relationship between expression of HMGB2 and prognosis of renal tumor. MTT assay was employed to examine cell proliferation and flow cytometry analysis was used to study the role of HMGB2 in cell apoptosis in ACHN cells. Transwell assays were used to explore the migration and invasion of ACHN cells. The effect of HMGB2 on tumor growth was investigated in vivo. Western blot was performed to evaluate the expression levels of p-JNK, p-ERK and p-p38MAPK. RESULTS: HMGB2 was upregulated in renal tumor and correlated with worse overall survival in renal tumor patients. Down-regulation of HMGB2 suppressed ACHN cells proliferation, invasion and migration in vitro. Moreover, down-regulation of HMGB2 inhibited tumor growth in vivo and HMGB2 exerts the oncogene function partly via the inhibition of p-p38MAPK activation. CONCLUSIONS: Our results provide novel insights into neuropathic pain and help to explore therapeutic targets in the treatment.


Subject(s)
Carcinoma, Renal Cell/metabolism , Cell Proliferation/physiology , Gene Knockdown Techniques/methods , HMGB2 Protein/deficiency , Kidney Neoplasms/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Female , HMGB2 Protein/antagonists & inhibitors , HMGB2 Protein/genetics , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Xenograft Model Antitumor Assays/methods , p38 Mitogen-Activated Protein Kinases/genetics
2.
J Immunol Methods ; 456: 72-76, 2018 05.
Article in English | MEDLINE | ID: mdl-29453955

ABSTRACT

HMGB1 and HMGB2 are DNA-interacting proteins but can also have extracellular actions during inflammation. Despite their relatively high homology, they may have distinct roles, making it essential to be able to differentiate between the two. Here we examine the specificity of five commercially-available anti-HMGB1 antibodies. By Western blotting of recombinant proteins and HMGB1-/- mouse embryonic fibroblasts, we identified only one HMGB1 antibody that, under our experimental conditions, did not also detect HMGB2. Selecting specific antibodies for HMGB1 and HMGB2 allowed identification of distinct HMGB1 and HMGB2 subcellular pools in primary neutrophils.


Subject(s)
Antibodies/immunology , Cross Reactions , HMGB1 Protein/immunology , HMGB2 Protein/immunology , Human Umbilical Vein Endothelial Cells/immunology , Antigen-Antibody Reactions , Cells, Cultured , HMGB1 Protein/deficiency , HMGB2 Protein/deficiency , Healthy Volunteers , Human Umbilical Vein Endothelial Cells/cytology , Humans
3.
Arterioscler Thromb Vasc Biol ; 37(4): 717-729, 2017 04.
Article in English | MEDLINE | ID: mdl-28183701

ABSTRACT

OBJECTIVE: In a previous study, we established diabetic and nondiabetic minipig models with coronary artery in-stent restenosis (ISR). Mass spectrometry showed that high-mobility group box (HMGB) 2 level was higher in ISR than in non-ISR tissue from diabetic minipigs. We here investigated whether serum HMGB2 levels were related to ISR in coronary artery disease patients. The effect of HMGB2 was evaluated in mice with femoral artery wire injury and in human aortic smooth muscle cells. APPROACH AND RESULTS: From 2513 patients undergoing coronary artery intervention and follow-up angiography at ≈1 year, 262 patients were diagnosed with ISR, and 298 patients with no ISR were randomly included as controls. Serum HMGB2 levels were significantly higher in patients with ISR than in those without ISR and were associated with ISR severity. Multivariable logistic regression analysis showed that HMGB2 level was independently associated with ISR. In experiments, HMGB2 expression was increased in vascular tissue after injury. Perivascular HMGB2 administration promoted injury-induced neointimal hyperplasia in C57Bl/6 mice compared with in the control, whereas such pathophysiological features were attenuated in Hmgb2-/- mice. Mechanistically, HMGB2 enhanced neointimal hyperplasia in mice and proliferation and migration in human aortic smooth muscle cells by inducing reactive oxygen species through increased p47phox phosphorylation. Knocking down p47phox, however, inhibited HMGB2-induced effects in human aortic smooth muscle cells. Finally, HMGB2-induced effects were significantly declined in receptor of advanced glycation end products knockdown or deficient cells, but not in Toll-like receptor 4 knockdown or deficient cells. CONCLUSIONS: Serum HMGB2 levels were associated with ISR in patients. HMGB2 promoted neointimal hyperplasia in mice with arterial wire injury through reactive oxygen species activation.


Subject(s)
Cell Movement , Cell Proliferation , Coronary Artery Disease/therapy , Coronary Restenosis/etiology , HMGB2 Protein/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Neointima , Percutaneous Coronary Intervention/adverse effects , Vascular System Injuries/blood , Aged , Animals , Biomarkers/blood , Case-Control Studies , Cells, Cultured , Coronary Angiography , Coronary Artery Disease/blood , Coronary Artery Disease/diagnostic imaging , Coronary Restenosis/blood , Coronary Restenosis/diagnostic imaging , Disease Models, Animal , Female , Femoral Artery/injuries , Femoral Artery/metabolism , Femoral Artery/pathology , Genetic Predisposition to Disease , HMGB2 Protein/deficiency , HMGB2 Protein/genetics , Humans , Hyperplasia , Logistic Models , Male , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Multivariate Analysis , Muscle, Smooth, Vascular/injuries , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , NADPH Oxidases/metabolism , Percutaneous Coronary Intervention/instrumentation , Phenotype , Phosphorylation , RNA Interference , Reactive Oxygen Species/metabolism , Receptor for Advanced Glycation End Products/genetics , Receptor for Advanced Glycation End Products/metabolism , Risk Factors , Signal Transduction , Stents , Swine , Swine, Miniature , Transfection , Vascular System Injuries/genetics , Vascular System Injuries/pathology
4.
PLoS One ; 8(12): e84838, 2013.
Article in English | MEDLINE | ID: mdl-24391977

ABSTRACT

Neural stem and progenitor cells (NSCs/NPCs) are distinct groups of cells found in the mammalian central nervous system (CNS). Previously we determined that members of the High Mobility Group (HMG) B family of chromatin structural proteins modulate NSC proliferation and self-renewal. Among them HMGB2 was found to be dynamically expressed in proliferating and differentiating NSCs, suggesting that it may regulate NSC maintenance. We report now that Hmgb2(-/-) mice exhibit SVZ hyperproliferation, increased numbers of SVZ NSCs, and a trend towards aberrant increases in newly born neurons in the olfactory bulb (OB) granule cell layer. Increases in the levels of the transcription factor p21 and the Neural cell adhesion molecule (NCAM), along with down-regulation of the transcription/pluripotency factor Oct4 in the Hmgb2-/- SVZ point to a possible pathway for this increased proliferation/differentiation. Our findings suggest that HMGB2 functions as a modulator of neurogenesis in young adult mice through regulation of NSC proliferation, and identify a potential target via which CNS repair could be amplified following trauma or disease-based neuronal degeneration.


Subject(s)
Cell Proliferation , HMGB2 Protein/deficiency , Neural Stem Cells/physiology , Neurogenesis/genetics , Animals , Blotting, Western , Cells, Cultured , DNA Primers/genetics , HMGB2 Protein/metabolism , Lateral Ventricles/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Fluorescence , Neural Stem Cells/metabolism , Neurogenesis/physiology , Reverse Transcriptase Polymerase Chain Reaction , Statistics, Nonparametric
5.
Nature ; 462(7269): 99-103, 2009 Nov 05.
Article in English | MEDLINE | ID: mdl-19890330

ABSTRACT

The activation of innate immune responses by nucleic acids is crucial to protective and pathological immunities and is mediated by the transmembrane Toll-like receptors (TLRs) and cytosolic receptors. However, it remains unknown whether a mechanism exists that integrates these nucleic-acid-sensing systems. Here we show that high-mobility group box (HMGB) proteins 1, 2 and 3 function as universal sentinels for nucleic acids. HMGBs bind to all immunogenic nucleic acids examined with a correlation between affinity and immunogenic potential. Hmgb1(-/-) and Hmgb2(-/-) mouse cells are defective in type-I interferon and inflammatory cytokine induction by DNA or RNA targeted to activate the cytosolic nucleic-acid-sensing receptors; cells in which the expression of all three HMGBs is suppressed show a more profound defect, accompanied by impaired activation of the transcription factors interferon regulatory factor 3 (IRF3) and nuclear factor (NF)-kappaB. The absence of HMGBs also severely impairs the activation of TLR3, TLR7 and TLR9 by their cognate nucleic acids. Our results therefore indicate a hierarchy in the nucleic-acid-mediated activation of immune responses, wherein the selective activation of nucleic-acid-sensing receptors is contingent on the more promiscuous sensing of nucleic acids by HMGBs. These findings may have implications for understanding the evolution of the innate immune system and for the treatment of immunological disorders.


Subject(s)
HMGB Proteins/immunology , HMGB Proteins/metabolism , Immunity, Innate/immunology , Nucleic Acids/immunology , Animals , Cell Line , Cytosol/immunology , DNA/immunology , HMGB Proteins/deficiency , HMGB Proteins/genetics , HMGB1 Protein/deficiency , HMGB1 Protein/genetics , HMGB1 Protein/immunology , HMGB1 Protein/metabolism , HMGB2 Protein/deficiency , HMGB2 Protein/genetics , HMGB2 Protein/immunology , HMGB2 Protein/metabolism , Interferon Regulatory Factor-3/metabolism , Mice , Mice, Inbred C57BL , Models, Immunological , NF-kappa B/metabolism , Nucleotides/chemistry , Nucleotides/immunology , Nucleotides/metabolism , RNA/immunology , Signal Transduction , Toll-Like Receptors/immunology , Virus Diseases/immunology , Virus Diseases/virology
6.
Proc Natl Acad Sci U S A ; 106(4): 1181-6, 2009 Jan 27.
Article in English | MEDLINE | ID: mdl-19139395

ABSTRACT

Osteoarthritis (OA) is the most common joint disease and typically begins with an aging-related disruption of the articular cartilage surface. Mechanisms leading to the aging-related cartilage surface degeneration remain to be determined. Here, we demonstrate that nonhistone chromatin protein high-mobility group box (HMGB) protein 2 is uniquely expressed in the superficial zone (SZ) of human articular cartilage. In human and murine cartilage, there is an aging-related loss of HMGB2 expression, ultimately leading to its complete absence. Mice genetically deficient in HMGB2 (Hmgb2(-/-)) show earlier onset of and more severe OA. This is associated with a profound reduction in cartilage cellularity attributable to increased cell death. These cellular changes precede glycosaminoglycan depletion and progressive cartilage erosions. Chondrocytes from Hmgb2(-/-) mice are more susceptible to apoptosis induction in vitro. In conclusion, HMGB2 is a transcriptional regulator specifically expressed in the SZ of human articular cartilage and supports chondrocyte survival. Aging is associated with a loss of HMGB2 expression and reduced cellularity, and this contributes to the development of OA.


Subject(s)
Aging/pathology , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Chromatin/metabolism , HMGB2 Protein/deficiency , Osteoarthritis/metabolism , Osteoarthritis/pathology , Aging/metabolism , Animals , Apoptosis , Cartilage, Articular/enzymology , Cell Survival , Gene Expression Regulation , HMGB2 Protein/genetics , HMGB2 Protein/metabolism , Humans , Joints/enzymology , Joints/pathology , Matrix Metalloproteinases/metabolism , Mice , Osteoarthritis/enzymology , Protein Transport , Proteoglycans/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...