Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Ann Hematol ; 103(1): 297-305, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37991510

ABSTRACT

This study was aimed to explore the prognosis of allogenic hematopoietic stem cell transplantation (allo-HSCT) in pediatric patients with acute lymphoblastic leukemia (ALL). This retrospective case series study included children with ALL who underwent allo-HSCT at Beijing Children's Hospital of Capital Medical University, Beijing, China, between January 2009 and December 2019. The outcomes included 5-year overall survival (OS) and event-free survival (EFS). A total of 75 children (52 males) were included. The median age at presentation was 5.30 years, and the median time from diagnosis to transplantation was 1.64 years. There were 15 human leukocyte antigen (HLA)-matched and 60 HLA-semi-matched transplants, 73 complete remissions (CR), and 2 MRD-positive transplants. The median follow-up time was 41 months. Out of 75 patients, 51 children survived, and 24 died/given up at the terminal stage. The 5-year OS and EFS rates were 67.77% and 57.30%, respectively, whereas the 5-year recurrence rate was 35.69%. Acute and chronic graft versus host diseases occurred in 40 and 28 cases, respectively. Children with MLL gene fusion had higher survival rates compared to other subgroups. Haplo-HSCT is not inferior to HLA-matched transplant. The children with MLL rearrangement had an acceptable 5-year OS, while complications and relapse should be monitored.


Subject(s)
Hematopoietic Stem Cell Transplantation , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Male , Humans , Child , Retrospective Studies , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Prognosis , Progression-Free Survival
2.
Article in English | MEDLINE | ID: mdl-32103929

ABSTRACT

Background and Purpose: Chronic obstructive pulmonary disease (COPD) is a common chronic inflammatory disease, which is associated with various comorbidities including osteoporosis. Interleukin(IL)-17 has been reported to play important roles in the pathogenesis of COPD and also associated with bone destruction in inflammatory diseases. However, the role of IL-17A in COPD-related osteoporosis is yet unknown. The purpose of our study was to investigate the potential contribution of IL-17A in COPD-related bone loss. Materials and Methods: We examined the bone mass and bone microarchitecture in wild-type and IL-17A-/- mice exposed to long-term cigarette smoke (CS). Osteoclast activities and the expression of receptor activator of nuclear factor-κB ligand (RANKL) in bone tissues were assessed, and the blood levels of inflammatory cytokines were measured. Results: Less bone loss as well as attenuated emphysema were shown in IL-17A-/- mice compared with wild-type mice. CS-exposed IL-17A-/- mice had decreased TRAP+ osteoclast numbers and lower RANKL expression compared with CS-exposed wild-type mice. Inflammatory cytokines including IL-6 and IL-1ß in circulation were decreased in IL-17A-/- mice exposed to CS compared with wild-type mice. Conclusion: This study indicates that IL-17A is involved in CS-induced bone loss and may be a common link between COPD and osteoporosis.


Subject(s)
Femur/metabolism , Interleukin-17/deficiency , Lung/metabolism , Osteoporosis/prevention & control , Pulmonary Disease, Chronic Obstructive/prevention & control , Pulmonary Emphysema/prevention & control , Smoke/adverse effects , Tobacco Products/adverse effects , Animals , Bone Density , Bone Remodeling , Cytokines/metabolism , Disease Models, Animal , Female , Femur/diagnostic imaging , Femur/physiopathology , Inflammation Mediators/metabolism , Interleukin-17/genetics , Lung/pathology , Lung/physiopathology , Mice, Inbred C57BL , Mice, Knockout , Osteoporosis/genetics , Osteoporosis/metabolism , Osteoporosis/physiopathology , Pulmonary Disease, Chronic Obstructive/genetics , Pulmonary Disease, Chronic Obstructive/metabolism , Pulmonary Disease, Chronic Obstructive/physiopathology , Pulmonary Emphysema/genetics , Pulmonary Emphysema/metabolism , Pulmonary Emphysema/physiopathology , RANK Ligand/metabolism
3.
Front Immunol ; 11: 588522, 2020.
Article in English | MEDLINE | ID: mdl-33613513

ABSTRACT

IL-17 is critical in lung lymphoid neogenesis in COPD, but the cellular and molecular mechanisms remain to be elucidated. Receptor activator of nuclear factor-κB ligand (RANKL) functions in lymphoid follicle formation in other organs, whether it is involved in IL-17A-dependent lymphoid neogenesis in COPD is unknown. To elucidate the expression and functional role of IL-17A/RANKL pathway in COPD. We first quantified and localized RANKL, its receptor RANK and IL-17A in lungs of patients with COPD, smokers and non-smokers. Next, IL-17A-/- and wild-type (WT) mice were exposed to air or cigarette smoke (CS) for 24 weeks, and lung lymphoid follicles and RANKL-RANK expression were measured. Lastly, we studied the in vitro biological function of RANKL pertaining to lymphoid neogenesis. We found that the expressions of RANKL-RANK and IL-17A, together with lymphoid follicles, were increased in lung tissues from patients with COPD. In WT mice exposed to CS, RANKL-RANK expressions were prominent in lung lymphoid follicles, which were absent in IL-17A-/- mice exposed to CS. In the lymphoid follicles, RANKL+ cells were identified mostly as B cells and RANK was localized in dendritic cells (DCs). In vitro IL-17A increased the expressions of RANKL in B cells and RANK in DCs, which in turn responded to RANKL stimulation by upregulation of CXCL13. Altogether, these results suggest that B lymphocyte RANKL pathway is involved in IL-17A-dependent lymphoid neogenesis in COPD.


Subject(s)
Interleukin-17/immunology , Lymphoid Tissue/immunology , Nicotiana , Pulmonary Disease, Chronic Obstructive/immunology , RANK Ligand/immunology , Smoke/adverse effects , Animals , B-Lymphocytes/immunology , Chemokine CXCL13/immunology , Dendritic Cells/immunology , Female , Humans , Interleukin-17/genetics , Lung/immunology , Male , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Signal Transduction , Smoking/immunology
4.
J Cell Mol Med ; 24(2): 1319-1331, 2020 01.
Article in English | MEDLINE | ID: mdl-31769590

ABSTRACT

High-mobility group box 1 (HMGB1) shows pro-inflammatory activity in various inflammatory diseases and has been found up-regulated in chronic obstructive pulmonary disease (COPD). Lung macrophages play an important role in airway inflammation and lung destruction in COPD, yet whether HMGB1 is involved in cigarette smoke (CS)-induced lung macrophage dysfunction is unknown. We sought to evaluate the intracellular localization and release of HMGB1 in lung macrophages from COPD patients and CS-exposed mice, and to investigate the role of HMGB1 in regulating autophagy in CS extract (CSE)-treated lung macrophages (MH-S cells). Our results showed that HMGB1 was highly expressed in lung tissues and sera of COPD patients and CS-exposed mice, along with predominantly cytoplasmic exporting from nuclei in lung macrophages. In vitro experiments revealed that CSE promoted the expression, nucleocytoplasmic translocation and release of HMGB1 partly via the nicotinic acetylcholine receptor (nAChR). Blockade of HMGB1 with chicken anti-HMGB1 polyclonal antibody (anti-HMGB1) or glycyrrhizin (Gly) attenuated the increase of LC3B-II and Beclin1, migration and p65 phosphorylation, suggesting the involvement of HMGB1 in autophagy, migration and NF-κB activation of lung macrophages. Hydroxychloroquine (CQ), an autophagy inhibitor, enhanced the increase of LC3B-II but not Beclin1 in CSE or rHMGB1-treated MH-S cells, and inhibition of autophagy by CQ and 3-methyladenine (3-MA) abrogated the migration and p65 phosphorylation of CSE-treated cells. These results indicate that CS-induced HMGB1 translocation and release contribute to migration and NF-κB activation through inducing autophagy in lung macrophages, providing novel evidence for HMGB1 as a potential target of intervention in COPD.


Subject(s)
Autophagy , Cell Movement , HMGB1 Protein/metabolism , Macrophages, Alveolar/pathology , NF-kappa B/metabolism , Pulmonary Disease, Chronic Obstructive/pathology , Smoke/adverse effects , Animals , Case-Control Studies , Female , Gene Expression Regulation , HMGB1 Protein/genetics , Humans , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/metabolism , Male , Mice , Mice, Inbred C57BL , Middle Aged , NF-kappa B/genetics , Protein Transport , Pulmonary Disease, Chronic Obstructive/etiology , Pulmonary Disease, Chronic Obstructive/metabolism , Nicotiana/adverse effects
5.
Article in English | MEDLINE | ID: mdl-30587964

ABSTRACT

BACKGROUND AND PURPOSE: Cigarette smoke (CS) induces alveolar destruction through overproduction of proteinases including matrix metalloproteinase (MMP)-9 by alveolar macrophages (AMs). Receptor activator of nuclear factor-κB ligand (RANKL) functions in immune regulation and cytokine secretion; whether it is involved in CS-induced MMP-9 expression is unknown. The purpose of our study was to investigate the expression and functional role of RANKL pathway in MMP-9 production pertaining to the pathogenesis of COPD. MATERIALS AND METHODS: We first localized RANKL and its receptor RANK in the lungs of mice exposed to long-term CS exposure. Next, we studied RANKL and RANK expression under CS extract (CSE) stimulation in vitro. Lastly, we studied the in vitro biological function of RANKL in CS-induced production of MMP-9. RESULTS: Both RANKL and RANK were highly expressed in AMs in CS-exposed mice, but not in the control mice. In vitro, CSE increased the expressions of RANKL and RANK in macrophages. AMs responded to CSE and RANKL stimulation by overexpressing MMP-9, and CSE-induced MMP-9 expression was partly blocked by using monoclonal anti-RANKL antibody. CONCLUSION: RANKL/RANK pathway mediates CS-induced MMP-9 expression in AMs, suggesting a novel mechanism for CS-associated emphysema.


Subject(s)
Cigarette Smoking/adverse effects , Lung/drug effects , Macrophages, Alveolar/drug effects , Matrix Metalloproteinase 9/biosynthesis , RANK Ligand/metabolism , Smoke/adverse effects , Animals , Cigarette Smoking/genetics , Cigarette Smoking/metabolism , Cigarette Smoking/physiopathology , Enzyme Induction , Female , Lung/enzymology , Lung/physiopathology , Macrophages, Alveolar/enzymology , Matrix Metalloproteinase 9/genetics , Mice , Mice, Inbred C57BL , Models, Animal , RAW 264.7 Cells , Receptor Activator of Nuclear Factor-kappa B/metabolism , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...