Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 47
1.
Exp Hematol Oncol ; 12(1): 35, 2023 Apr 07.
Article En | MEDLINE | ID: mdl-37029450

INTRODUCTION: Circulating monocytic myeloid-derived suppressive cells (M-MDSCs) are implicated as a poor prognostic factor and cause CAR T-cell failure in diffuse large B-cell lymphoma (DLBCL). Triggering receptors expressed on myeloid cells 2 (TREM2) are a transmembrane glycoprotein that polarize macrophages to anti-inflammation phenotype but have never been explored on M-MDSCs. This study aims to elucidate the expression and clinical impact of surface TREM2 on circulating M-MDSCs derived from DLBCL adults. METHODS: This prospective, observational study enrolled 100 adults with newly diagnosed and treatment-naïve DLBCL from May 2019 to October 2021. Human circulating M-MDSCs were obtained from freshly isolated peripheral blood, and each patient's surface-TREM2 level on M-MDSCs was normalized via a healthy control at the same performance of flow-cytometry analysis. Murine MDSCs derived from bone marrow (BM-MDSCs) were adopted to assess the link between Trem2 and cytotoxic T lymphocytes. RESULTS: More circulating M-MDSCs at diagnosis of DLBCL predicted worse progression-free (PFS) and overall survival (OS). Patients with higher IPI scores, bone marrow involvement, or lower absolute counts of CD4+ or CD8+ T cells in PB had significantly higher normalized TREM2 levels on M-MDSCs. Additionally, normalized TREM2 levels on M-MDSCs could be grouped into low (< 2%), medium (2-44%), or high (> 44%) levels, and a high normalized TREM2 level on M-MDSCs was proven as an independent prognostic factor for both PFS and OS via multivariate Cox regression analysis and associated with worst PFS and OS. Interestingly, normalized levels of surface TREM2 on M-MDSCs were negatively associated with absolute counts of PB CD8+ T cells and positively correlated with levels of intracellular arginase 1 (ARG1) within M-MDSCs. Wild-type BM-MDSCs had significantly higher mRNA levels of Arg1 and showed more prominent ability to suppress the proliferation of co-cultured CD8+ T cells than BM-MDSCs from Trem2 knockout mice, and the suppressive ability could be impaired by adding Arg1 inhibitors (CB1158) or supplementing L-arginine. CONCLUSION: In treatment-naïve DLBCL adults, a high surface-TREM2 level on circulating M-MDSCs is a poor prognostic factor for both PFS and OS and warrants further investigation for its potential as a novel target in immunotherapy.

2.
Elife ; 112022 11 30.
Article En | MEDLINE | ID: mdl-36449334

Tissue-resident macrophages are essential to protect from pathogen invasion and maintain organ homeostasis. The ability of thymic macrophages to engulf apoptotic thymocytes is well appreciated, but little is known about their ontogeny, maintenance, and diversity. Here, we characterized the surface phenotype and transcriptional profile of these cells and defined their expression signature. Thymic macrophages were most closely related to spleen red pulp macrophages and Kupffer cells and shared the expression of the transcription factor (TF) SpiC with these cells. Single-cell RNA sequencing (scRNA-Seq) showed that the macrophages in the adult thymus are composed of two populations distinguished by the expression of Timd4 and Cx3cr1. Remarkably, Timd4+ cells were located in the cortex, while Cx3cr1+ macrophages were restricted to the medulla and the cortico-medullary junction. Using shield chimeras, transplantation of embryonic thymuses, and genetic fate mapping, we found that the two populations have distinct origins. Timd4+ thymic macrophages are of embryonic origin, while Cx3cr1+ macrophages are derived from adult hematopoietic stem cells. Aging has a profound effect on the macrophages in the thymus. Timd4+ cells underwent gradual attrition, while Cx3cr1+ cells slowly accumulated with age and, in older mice, were the dominant macrophage population in the thymus. Altogether, our work defines the phenotype, origin, and diversity of thymic macrophages.


Macrophages , Thymus Gland , Mice , Animals , Thymus Gland/metabolism , Thymocytes , Hematopoietic Stem Cells , Phenotype
3.
Front Immunol ; 13: 879648, 2022.
Article En | MEDLINE | ID: mdl-35720343

Background: Decoy receptor 3 (DcR3) belongs to the tumor necrosis factor (TNF) receptor superfamily and neutralizes TNF ligands, including FasL and TRAIL, to prevent T activation during T-cell priming. However, the cellular mechanisms underlying acute cell-mediated rejection (ACMR) remain unknown. Methods: We generated DcR3 transgenic (Tg) mice and mice with high DcR3 expression (HDE) to study both in vivo and in vitro. FasR RNA knockdown in immortalized CD4+CD8+ T-cells was used to survey the role of DcR3 on FasR/Fas-associated protein with death domain (FADD)/caspase 8 pathway and its cross-link to TNF receptor-associated factor 1 (TNFR1)-associated death domain protein (TRADD) in suppressing TNFR1. TNF/TRADD knockout mice were used to show the importance of TNF adaptor protein. Results: DcR3.Fc suppressed C57BL/6 female T-cell activation and transformation into CD4+CD69+, CD4+CD44+, and CD4+CD25+Foxp3+ when compared with isotype IgG1 and its co-treatment with FasL/TRAIL after exposing to bone marrow-derived dendritic cells (BMDCs) that carried alloantigen with male H-Y and minor antigenic determinant. Interleukin-17 and interferon-γ productions by BMDC-activated T-cells were lowered after co-treating with DcR3.Fc. DcR3.Fc induced effector T-cells (Teffs) and was susceptible to FasR-mediated apoptosis through the FADD/TRADD/caspase 8 pathway. After exposing to DcR3.Fc, TRADD was silenced, likely turning down the inflammatory response. The systemic effects of DcR3 Tg mice and HDE phenotype induced by the promoter of cytomegalovirus not only attenuated ACMR severity but also ameliorated the high serum creatinine and blood urea nitrogen levels even with high T-cell exposure frequencies. Besides this, DcR3 has minor biological effects on both MHC-matched and MHC-mismatched models. Conclusions: High DcR3 doses protect renal tubular epithelial cells from acute T-cell attack during the T-cell priming stage via interfering with TNF ligand-mediated reverse signaling and possibly promoting Teff apoptosis through FasR upregulation. Our findings supported that the decoy receptor is involved in T-cell modulation in kidney transplant rejection.


Receptors, Tumor Necrosis Factor, Member 6b , Receptors, Tumor Necrosis Factor, Type I , Animals , Apoptosis , CD8-Positive T-Lymphocytes/metabolism , Caspase 8/genetics , Caspase 8/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Receptors, Tumor Necrosis Factor/metabolism , Receptors, Tumor Necrosis Factor, Member 6b/genetics , Receptors, Tumor Necrosis Factor, Member 6b/metabolism , Receptors, Tumor Necrosis Factor, Type I/metabolism
4.
J Mol Med (Berl) ; 100(6): 917-931, 2022 06.
Article En | MEDLINE | ID: mdl-35532794

Chronic kidney disease (CKD) is a global public health issue. CKD is caused by the infiltration of various myeloid cell types into renal tissue, resulting in renal fibrosis and tubular atrophy. Unilateral ureteral obstruction (UUO) surgery in mice is a model of CKD and characterized by high expression of the anti-inflammatory receptor, Triggering receptor expressed on myeloid cells 2 (TREM-2), on myeloid cells in affected kidneys. Here, we show that iNOS expression and nitric oxide (NO) induction were decreased in Trem-2-/- bone marrow-derived DCs (BMDCs) and in Trem-2 knockdown DC2.4 cells stimulated in vitro with LPS. The nitration of RORγt was decreased in T cells co-cultured with LPS-stimulated Trem-2-/- BMDCs, enhancing IL-17 production. UUO-treated Trem-2-/- mice displayed aggravated renal pathogenesis accompanied by greater neutrophil infiltration and enhanced Th17 cells differentiation, phenotypes that could be rescued by the administration of L-arginine (a biological precursor of NO). Our data identify a key mechanism underlying TREM-2-mediated NO to modulate the cellular crosstalk between dendritic cells, Th17, and neutrophils. Furthermore, we also reveal TREM-2 as a potential novel target for the development of anti-inflammatory drugs in CKD treatment. KEY MESSAGES: The expression of TREM-2 is increased in nephritis TREM-2+ DCs maintain NO production to negatively regulate Th17 differentiation The severe pathologies of nephritis can be rescued by L-arginine supplementation.


Membrane Glycoproteins/metabolism , Nephritis , Receptors, Immunologic/metabolism , Renal Insufficiency, Chronic , Ureteral Obstruction , Animals , Arginine , Dendritic Cells/pathology , Lipopolysaccharides , Mice , Nephritis/complications , Nitric Oxide , Th17 Cells/pathology , Ureteral Obstruction/pathology
5.
Int J Mol Sci ; 23(3)2022 Jan 30.
Article En | MEDLINE | ID: mdl-35163542

The PD-1/PD-L1 pathway is critical in T cell biology; however, the role of the PD-1/PD-L1 pathway in clinical characteristics and treatment outcomes in pulmonary tuberculosis (PTB) patients is unclear. We prospectively enrolled PTB, latent TB infection (LTBI), and non-TB, non-LTBI subjects. The expression of PD-1/PD-L1 on peripheral blood mononuclear cells (PBMCs) was measured and correlated with clinical characteristics and treatment outcomes in PTB patients. Immunohistochemistry and immunofluorescence were used to visualize PD-1/PD-L1-expressing cells in lung tissues from PTB patients and from murine with heat-killed MTB (HK-MTB) treatment. A total of 76 PTB, 40 LTBI, and 28 non-TB, non-LTBI subjects were enrolled. The expression of PD-1 on CD4+ T cells and PD-L1 on CD14+ monocytes was significantly higher in PTB cases than non-TB subjects. PTB patients with sputum smear/culture unconversion displayed higher PD-L1 expression on monocytes. PD-L1-expressing macrophages were identified in lung tissue from PTB patients, and co-localized with macrophages in murine lung tissues. Mycobacterium tuberculosis (MTB) whole cell lysate/EsxA stimulation of human and mouse macrophages demonstrated increased PD-L1 expression. In conclusion, increased expression of PD-L1 on monocytes in PTB patients correlated with higher bacterial burden and worse treatment outcomes. The findings suggest the involvement of the PD-1/PD-L1 pathway in MTB-related immune responses.


Antitubercular Agents/pharmacology , B7-H1 Antigen/metabolism , Latent Tuberculosis/metabolism , Leukocytes, Mononuclear/metabolism , Mycobacterium tuberculosis/pathogenicity , Programmed Cell Death 1 Receptor/metabolism , Tuberculosis, Pulmonary/metabolism , Up-Regulation , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Antitubercular Agents/therapeutic use , Case-Control Studies , Cells, Cultured , Disease Models, Animal , Female , Humans , Latent Tuberculosis/microbiology , Male , Mice , Middle Aged , Mycobacterium tuberculosis/drug effects , THP-1 Cells , Treatment Outcome , Tuberculosis, Pulmonary/drug therapy , Tuberculosis, Pulmonary/microbiology , Young Adult
6.
EMBO J ; 41(4): e106825, 2022 02 15.
Article En | MEDLINE | ID: mdl-35023164

Despite extensive analysis of pRB phosphorylation in vitro, how this modification influences development and homeostasis in vivo is unclear. Here, we show that homozygous Rb∆K4 and Rb∆K7 knock-in mice, in which either four or all seven phosphorylation sites in the C-terminal region of pRb, respectively, have been abolished by Ser/Thr-to-Ala substitutions, undergo normal embryogenesis and early development, notwithstanding suppressed phosphorylation of additional upstream sites. Whereas Rb∆K4 mice exhibit telomere attrition but no other abnormalities, Rb∆K7 mice are smaller and display additional hallmarks of premature aging including infertility, kyphosis, and diabetes, indicating an accumulative effect of blocking pRb phosphorylation. Diabetes in Rb∆K7 mice is insulin-sensitive and associated with failure of quiescent pancreatic ß-cells to re-enter the cell cycle in response to mitogens, resulting in induction of DNA damage response (DDR), senescence-associated secretory phenotype (SASP), and reduced pancreatic islet mass and circulating insulin level. Pre-treatment with the epigenetic regulator vitamin C reduces DDR, increases cell cycle re-entry, improves islet morphology, and attenuates diabetes. These results have direct implications for cell cycle regulation, CDK-inhibitor therapeutics, diabetes, and longevity.


Aging/physiology , Ascorbic Acid/pharmacology , Diabetes Mellitus, Experimental/prevention & control , Retinoblastoma Protein/metabolism , Animals , Cellular Senescence/drug effects , Cyclin-Dependent Kinase 2/antagonists & inhibitors , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/pathology , E2F1 Transcription Factor/metabolism , Embryonic Development/genetics , Female , Fibroblasts/drug effects , Gene Knock-In Techniques , Insulin-Secreting Cells/pathology , Mice , Phosphorylation , Pregnancy , Retinoblastoma Protein/genetics , Telomere/genetics
7.
Proc Natl Acad Sci U S A ; 118(30)2021 07 27.
Article En | MEDLINE | ID: mdl-34301890

Cytosolic lipopolysaccharides (LPSs) bind directly to caspase-4/5/11 through their lipid A moiety, inducing inflammatory caspase oligomerization and activation, which is identified as the noncanonical inflammasome pathway. Galectins, ß-galactoside-binding proteins, bind to various gram-negative bacterial LPS, which display ß-galactoside-containing polysaccharide chains. Galectins are mainly present intracellularly, but their interactions with cytosolic microbial glycans have not been investigated. We report that in cell-free systems, galectin-3 augments the LPS-induced assembly of caspase-4/11 oligomers, leading to increased caspase-4/11 activation. Its carboxyl-terminal carbohydrate-recognition domain is essential for this effect, and its N-terminal domain, which contributes to the self-association property of the protein, is also critical, suggesting that this promoting effect is dependent on the functional multivalency of galectin-3. Moreover, galectin-3 enhances intracellular LPS-induced caspase-4/11 oligomerization and activation, as well as gasdermin D cleavage in human embryonic kidney (HEK) 293T cells, and it additionally promotes interleukin-1ß production and pyroptotic death in macrophages. Galectin-3 also promotes caspase-11 activation and gasdermin D cleavage in macrophages treated with outer membrane vesicles, which are known to be taken up by cells and release LPSs into the cytosol. Coimmunoprecipitation confirmed that galectin-3 associates with caspase-11 after intracellular delivery of LPSs. Immunofluorescence staining revealed colocalization of LPSs, galectin-3, and caspase-11 independent of host N-glycans. Thus, we conclude that galectin-3 amplifies caspase-4/11 oligomerization and activation through LPS glycan binding, resulting in more intense pyroptosis-a critical mechanism of host resistance against bacterial infection that may provide opportunities for new therapeutic interventions.


Caspases/metabolism , Galectin 3/metabolism , Inflammasomes/immunology , Inflammation/immunology , Lipopolysaccharides/metabolism , Macrophages/immunology , Animals , Cytosol/metabolism , Galectin 3/genetics , Inflammasomes/metabolism , Inflammation/metabolism , Inflammation/pathology , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Inbred C57BL , Pyroptosis
8.
Stem Cell Res Ther ; 12(1): 367, 2021 06 28.
Article En | MEDLINE | ID: mdl-34183058

BACKGROUND: Acute kidney injury (AKI) is an emerging global healthcare issue without effective therapy yet. Autophagy recycles damaged organelles and helps maintain tissue homeostasis in acute renal ischemia-reperfusion (I/R) injury. Hypoxic mesenchymal stem cells (HMSCs) represent an innovative cell-based therapy in AKI. Moreover, the conditioned medium of HMSCs (HMSC-CM) rich in beneficial trophic factors may serve as a cell-free alternative therapy. Nonetheless, whether HMSCs or HMSC-CM mitigate renal I/R injury via modulating tubular autophagy remains unclear. METHODS: Renal I/R injury was induced by clamping of the left renal artery with right nephrectomy in male Sprague-Dawley rats. The rats were injected with either PBS, HMSCs, or HMSC-CM immediately after the surgery and sacrificed 48 h later. Renal tubular NRK-52E cells subjected to hypoxia-reoxygenation (H/R) injury were co-cultured with HMSCs or treated with HMSC-CM to assess the regulatory effects of HSMCs on tubular autophagy and apoptosis. The association of tubular autophagy gene expression and renal recovery was also investigated in patients with ischemic AKI. RESULT: HMSCs had a superior anti-oxidative effect in I/R-injured rat kidneys as compared to normoxia-cultured mesenchymal stem cells. HMSCs further attenuated renal macrophage infiltration and inflammation, reduced tubular apoptosis, enhanced tubular proliferation, and improved kidney function decline in rats with renal I/R injury. Moreover, HMSCs suppressed superoxide formation, reduced DNA damage and lipid peroxidation, and increased anti-oxidants expression in renal tubular epithelial cells during I/R injury. Co-culture of HMSCs with H/R-injured NRK-52E cells also lessened tubular cell death. Mechanistically, HMSCs downregulated the expression of pro-inflammatory interleukin-1ß, proapoptotic Bax, and caspase 3. Notably, HMSCs also upregulated the expression of autophagy-related LC3B, Atg5 and Beclin 1 in renal tubular cells both in vivo and in vitro. Addition of 3-methyladenine suppressed the activity of autophagy and abrogated the renoprotective effects of HMSCs. The renoprotective effect of tubular autophagy was further validated in patients with ischemic AKI. AKI patients with higher renal LC3B expression were associated with better renal recovery. CONCLUSION: The present study describes that the enhancing effect of MSCs, and especially of HMCSs, on tissue autophagy can be applied to suppress renal tubular apoptosis and attenuate renal impairment during renal I/R injury in the rat. Our findings provide further mechanistic support to HMSCs therapy and its investigation in clinical trials of ischemic AKI.


Acute Kidney Injury , Mesenchymal Stem Cells , Reperfusion Injury , Acute Kidney Injury/therapy , Animals , Apoptosis , Autophagy , Humans , Hypoxia , Ischemia , Kidney , Male , Rats , Rats, Sprague-Dawley , Reperfusion , Reperfusion Injury/therapy
9.
Stem Cell Reports ; 16(5): 1363-1380, 2021 05 11.
Article En | MEDLINE | ID: mdl-33836143

Microglia, the immune cells of the central nervous system, play critical roles in brain physiology and pathology. We report a novel approach that produces, within 10 days, the differentiation of human induced pluripotent stem cells (hiPSCs) into microglia (iMG) by forced expression of both SPI1 and CEBPA. High-level expression of the main microglial markers and the purity of the iMG cells were confirmed by RT-qPCR, immunostaining, and flow cytometry analyses. Whole-transcriptome analysis demonstrated that these iMGs resemble human fetal/adult microglia but not human monocytes. Moreover, these iMGs exhibited appropriate physiological functions, including various inflammatory responses, ADP/ATP-evoked migration, and phagocytic ability. When co-cultured with hiPSC-derived neurons, the iMGs respond and migrate toward injured neurons. This study has established a protocol for the rapid conversion of hiPSCs into functional iMGs, which should facilitate functional studies of human microglia using different disease models and also help with drug discovery.


Cell Differentiation , Induced Pluripotent Stem Cells/cytology , Microglia/cytology , Transcription Factors/metabolism , Adenosine Diphosphate/pharmacology , Adenosine Triphosphate/pharmacology , Biomarkers/metabolism , Calcium Signaling/drug effects , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Movement/drug effects , Culture Media/pharmacology , Gene Expression Profiling , Gene Expression Regulation/drug effects , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Microglia/drug effects
10.
J Mol Cell Cardiol ; 155: 99-110, 2021 06.
Article En | MEDLINE | ID: mdl-33713645

Background Diabetes has a pronounced effect on the peripheral vasculature. The accumulation of advanced glycation end products (AGEs) is regarded as the crucial mechanism responsible for vascular damage in diabetes, but it is not easy to be avoided from food. In this study, we aimed to investigate the effects of an oral absorbent, AST-120, on the accumulation of AGEs and changes in blood flow recovery in diabetic mice. Methods The mice were divided into four groups, wild-type (WT) mice without treatment, WT mice treated with 5% AST-120 mixed into pulverized chow, streptozotocin-induced diabetes mellitus (DM) mice, and DM mice treated with 5% AST-120. Six weeks after hind-limb ischemia surgery, blood flow reperfusion, histology, plasma AGE, and cytokine were examined. Bone marrow cells were cultured and derived into macrophages to evaluate the effects of AGEs on macrophage polarization. Results Plasma AGEs were significantly increased in diabetic mice. AST-120 could bind to AGEs and reduced their plasma concentrations. Histological analysis revealed fewer collateral vessels with corresponding impairment of blood flow recovery in diabetic mice. In these mice, AGE-positive and AGE receptor-positive macrophages were numerous in ischemic limbs compared with non- diabetic mice. In diabetic mice, macrophages in ischemic tissues demonstrated greater M1 polarization than M2 polarization; this pattern was reversed in the AST-120 treatment group. The change in macrophage polarization was associated with the corresponding expression of pro-inflammatory cytokines in the ischemic tissues. In cell cultures, AGEs triggered the transformation of bone marrow-derived macrophages into the M1 phenotype. The alterations in the polarization of macrophages were reversed after treatment with AST-120. Conclusions Oral administration of AST-120 decreased the serum levels of AGEs in diabetic mice and improved neovascularization of ischemic limbs. This benefit may be due to, at least partially, the alterations in macrophage polarization and the associated changes in inflammatory cytokines.


Carbon/pharmacology , Cell Plasticity/drug effects , Ischemia/metabolism , Macrophages/drug effects , Macrophages/metabolism , Muscles/blood supply , Muscles/metabolism , Neovascularization, Physiologic/drug effects , Oxides/pharmacology , Animals , Cell Line , Cytokines/blood , Cytokines/metabolism , Diabetes Mellitus, Experimental , Glycation End Products, Advanced/blood , Glycation End Products, Advanced/metabolism , Inflammation Mediators/metabolism , Macrophage Activation/drug effects , Male , Mice , Models, Biological , Muscles/drug effects
11.
Microbes Infect ; 23(1): 104765, 2021.
Article En | MEDLINE | ID: mdl-33049389

Triggering receptor expressed on myeloid cells 1 (TREM-1) extensively interacts with toll-like receptors and amplifies pro-inflammatory responses. The effect of TREM-1 on Mycobacterium tuberculosis (MTB)-related immune responses remains to be elucidated. We isolated bone marrow-derived macrophages (BMDMs) from wild-type mice and Trem-1 KO mice and treated them with MTB whole cell lysate and EsxA (ESAT-6). Cytokine production and mRNA expression, including Trem-1, following stimulation were evaluated. Intratracheal instillation of heat-killed MTB (HKMTB) in mice was performed and the presence of TREM-1-positive macrophages was investigated by immunohistochemistry analysis. In our study, BMDMs isolated from wild-type mice produced more pro-inflammatory cytokines and demonstrated higher inflammatory gene expression levels compared with those isolated from Trem-1 KO mice when stimulated with MTB whole cell lysate. EsxA had a synergistic effect with MTB whole cell lysate on the induction of pro-inflammatory responses. The gene expression of Trem-1 was upregulated when treated with MTB-related proteins. TREM-1-positive macrophages were identified in the lung tissues from patients with active TB and from wild-type mice treated with intratracheal instillation of HKMTB. In conclusion, in mouse macrophages, TREM-1 could enhance pro-inflammatory immune responses when stimulated with MTB-related proteins. The gene expression of Trem-1 could also be induced by MTB-related stimulation.


Macrophages/immunology , Mycobacterium tuberculosis/immunology , Triggering Receptor Expressed on Myeloid Cells-1/immunology , Tuberculosis/immunology , Tuberculosis/microbiology , Animals , Female , Host-Pathogen Interactions , Humans , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/metabolism , Triggering Receptor Expressed on Myeloid Cells-1/genetics , Tuberculosis/genetics
12.
Int J Mol Sci ; 21(17)2020 Aug 19.
Article En | MEDLINE | ID: mdl-32825118

Mounting evidence indicates that an increase in histone deacetylation contributes to renal fibrosis. Although inhibition of histone deacetylase (HDAC) can reduce the extent of fibrosis, whether HDAC inhibitors exert the antifibrotic effect through modulating the phenotypes of macrophages, the key regulator of renal fibrosis, remains unknown. Moreover, the functional roles of the M2 macrophage subpopulation in fibrotic kidney diseases remain incompletely understood. Herein, we investigated the role of HDAC inhibitors on renal fibrogenesis and macrophage plasticity. We found that HDAC inhibition by trichostatin A (TSA) reduced the accumulation of interstitial macrophages, suppressed the activation of myofibroblasts and attenuated the extent of fibrosis in obstructive nephropathy. Moreover, TSA inhibited M1 macrophages and augmented M2 macrophage infiltration in fibrotic kidney tissue. Interestingly, TSA preferentially upregulated M2c macrophages and suppressed M2a macrophages in the obstructed kidneys, which was correlated with a reduction of interstitial fibrosis. TSA also repressed the expression of proinflammatory and profibrotic molecules in cultured M2a macrophages and inhibited the activation of renal myofibroblasts. In conclusion, our study was the first to show that HDAC inhibition by TSA alleviates renal fibrosis in obstructed kidneys through facilitating an M1 to M2c macrophage transition.


Histone Deacetylase Inhibitors/therapeutic use , Hydroxamic Acids/therapeutic use , Macrophages/drug effects , Renal Insufficiency, Chronic/drug therapy , Animals , Cell Line , Cells, Cultured , Fibrosis , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Myofibroblasts/drug effects , Myofibroblasts/metabolism , Rats , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/pathology
13.
Int J Mol Sci ; 21(13)2020 Jul 02.
Article En | MEDLINE | ID: mdl-32630825

Neutrophils are involved in the alveolitis of idiopathic pulmonary fibrosis (IPF). However, their pathogenic mechanisms are still poorly understood. Nintedanib has antifibrotic and anti-inflammatory activity in IPF. This study aimed to investigate the regulatory mechanism of nintedanib on neutrophil chemotaxis in bleomycin (BLM)-induced pulmonary fibrosis. Nintedanib was administered via oral gavage to male C57BL/6 mice 24 h after a bleomycin intratracheal injection (1.5 U/kg). Lung histopathological findings, the expression of cytokines, and the regulatory signaling pathways of neutrophil chemotaxis were analyzed. The effect of nintedanib was also investigated in a mouse model with adoptive neutrophil transfer in vivo. Nintedanib significantly decreased the histopathological changes and neutrophil recruitment in BLM-induced pulmonary fibrosis. Nintedanib mediated a downregulation of chemokine (C-X-C motif) receptor 2 (CXCR2) and very late antigen 4 (VLA-4) expression, as well as an upregulation of G protein-coupled receptor kinase 2 (GRK2) activity in peripheral blood neutrophils in BLM-induced pulmonary fibrosis. Nintedanib also decreased the activation of endothelial cells by the decreased expression of vascular cell adhesion molecule 1 (VCAM-1). The effect of nintedanib on regulating neutrophil chemotaxis was also confirmed by a mouse model with adoptive neutrophil transfer in vivo. In conclusion, nintedanib reduces neutrophil chemotaxis and endothelial cell activation to regulate the severity of BLM-induced pulmonary fibrosis. These effects are associated with an enhancement of GRK2 activity and a reduction in CXCR2 and VLA-4 expression on neutrophils and a decrease in VCAM-1 expression on endothelial cells.


G-Protein-Coupled Receptor Kinase 2/metabolism , Indoles/pharmacology , Neutrophils/metabolism , Animals , Bleomycin/pharmacology , Chemotaxis/drug effects , Chemotaxis, Leukocyte/drug effects , Endothelial Cells/metabolism , Idiopathic Pulmonary Fibrosis/metabolism , Indoles/metabolism , Lung/pathology , Male , Mice , Mice, Inbred C57BL , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/physiopathology , Signal Transduction/drug effects
14.
Immunol Lett ; 226: 22-30, 2020 10.
Article En | MEDLINE | ID: mdl-32622933

OBJECTIVE: MicroRNA-122 (miR-122) is the most abundant miRNA in the liver and it plays an important role in regulating liver metabolism and tumor formation. Previous studies also reveal an anti-inflammatory function of miR-122; however, relatively little is known about the mechanisms by which miR-122 suppresses inflammation. This study aims to search the effect of miR-122 on proinflammatory chemokines/cytokines production in mice. METHODS: Quantitative real-time PCR, Western blot analysis, and ELISA were performed to examine gene expression. TargetScan, miRanda, and microT v3.0 were used to search for possible miR-122 target sites in the 3'-untranslated regions (3'-UTR) of candidate genes. Luciferase reporter assay and site-directed mutagenesis were applied to verify miR-122 target sequences. LPS was applied to peritoneal macrophages and mice to evaluate inflammatory response. RESULTS: The expression of proinflammatory chemokines, including Ccl2, Ccl4, Ccl20, Cxcl2, and Cxcl10, and Relb in the livers of miR-122 knockout (KO) mice was increased. We identified Relb as a direct miR-122 target. Overexpressing RelB in the mouse liver increased the expression of Ccl2, Ccl4, Ccl20, Cxcl2, and Cxcl10. Peritoneal macrophages from miR-122 KO mice had a higher level of RelB, and they showed a stronger NF-κB activation and more TNF-α and IL-6 secretion after LPS stimulation. Overexpression of RelB in a macrophage cell line augmented LPS-induced TNF-α and IL-6 production. miR-122 KO mice showed a greatly increased mortality rate and generated a stronger and lasting inflammatory response to LPS. CONCLUSIONS: Deletion of miR-122 caused an upregulation of proinflammatory chemokines and RelB in the liver. Increased RelB may contribute to increases in these chemokine in the liver. Intriguingly, deletion of miR-122 also enhanced the sensitivity of macrophages and mice to LPS. Our results reveal that reducing RelB expression is a new mechanism by which miR-122 regulates inflammation.


Liver/physiology , Macrophages/physiology , MicroRNAs/genetics , Transcription Factor RelB/metabolism , Animals , Chemokines/metabolism , Cytokines/metabolism , HEK293 Cells , Humans , Inflammation Mediators/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction , Transcription Factor RelB/genetics , Up-Regulation
15.
Sci Rep ; 10(1): 8422, 2020 05 21.
Article En | MEDLINE | ID: mdl-32439945

Toll-like receptors (TLRs) play crucial roles in host immune defenses. Recently, TLR-mediated autophagy is reported to promote immune responses via increasing antigen processing and presentation in antigen presenting cells. The present study examined whether the synthetic TLR4 activator (CCL-34) could induce autophagy to promote innate and adaptive immunity. In addition, the potential of CCL-34 as an immune adjuvant in vivo was also investigated. Our data using RAW264.7 cells and bone marrow-derived macrophages showed that CCL-34 induced autophagy through a TLR4-NF-κB pathway. The autophagy-related molecules (Nrf2, p62 and Beclin 1) were activated in RAW264.7 cells and bone marrow-derived macrophages under CCL-34 treatment. CCL-34-stimulated macrophages exhibited significant antigen-processing activity and induced the proliferation of antigen-specific CD4+T cells as well as the production of activated T cell-related cytokines, IL-2 and IFN-γ. Furthermore, CCL-34 immunization in mice induced infiltration of monocytes in the peritoneal cavity and elevation of antigen-specific IgG in the serum. CCL-34 treatment in vivo did not cause toxicity based on serum biochemical profiles. Notably, the antigen-specific responses induced by CCL-34 were attenuated by the autophagy inhibitor, 3-methyladenine. In summary, we demonstrated CCL-34 can induce autophagy to promote antigen-specific immune responses and act as an efficient adjuvant.


Adjuvants, Immunologic/pharmacology , Autophagy/immunology , Glycolipids/pharmacology , Immunogenicity, Vaccine/immunology , Serine/analogs & derivatives , Toll-Like Receptor 4/metabolism , Adenine/analogs & derivatives , Adenine/pharmacology , Animals , Beclin-1/metabolism , CD4-Positive T-Lymphocytes/immunology , Cell Line , Cell Proliferation/drug effects , Enzyme Activation/drug effects , Immunoglobulin G/blood , Interferon-gamma/metabolism , Interleukin-2/metabolism , Macrophages/immunology , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Monocytes/immunology , NF-E2-Related Factor 2/metabolism , RAW 264.7 Cells , Serine/pharmacology , Vaccines/immunology
16.
Cancer Lett ; 474: 138-150, 2020 04 01.
Article En | MEDLINE | ID: mdl-31987921

Mitochondrial Lon is a chaperone protein whose upregulation increases the production of mitochondrial reactive oxygen species (ROS). However, there is a lack of information in detail on how mitochondrial Lon regulates cancer metastasis through ROS production in the tumor microenvironment (TME). Our results show that elevated Lon promotes epithelial-mesenchymal transition (EMT) via ROS-dependent p38 and NF-κB-signaling. We further identified pyrroline-5-carboxylate reductase 1 (PYCR1) as a client of chaperone Lon, which induces mitochondrial ROS and EMT by Lon. Mitochondrial Lon induces ROS-dependent production of inflammatory cytokines, such as TGF-ß, IL-6, IL-13, and VEGF-A, which consequently activates EMT, angiogenesis, and M2 macrophage polarization. In addition, Lon expression is induced upon the activation and M2 polarization of macrophages, which further promotes M2 macrophages to enhance the immunosuppressive microenvironment and metastatic behaviors in the TME. This raises the possibility that manipulation of the mitochondrial redox balance in the TME may serve as a therapeutic strategy to improve T cell function in cancer immunotherapy.


ATP-Dependent Proteases/metabolism , Lung Neoplasms/secondary , Mitochondria/pathology , Mitochondrial Proteins/metabolism , Mouth Neoplasms/pathology , Oxidative Stress , Pyrroline Carboxylate Reductases/metabolism , ATP-Dependent Proteases/genetics , Animals , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Proliferation , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/immunology , Lung Neoplasms/metabolism , Macrophage Activation/immunology , Male , Melanoma/immunology , Melanoma/metabolism , Melanoma/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Mouth Neoplasms/immunology , Mouth Neoplasms/metabolism , Prognosis , Pyrroline Carboxylate Reductases/genetics , Reactive Oxygen Species/metabolism , Tumor Cells, Cultured , Tumor Microenvironment , Xenograft Model Antitumor Assays , delta-1-Pyrroline-5-Carboxylate Reductase
17.
J Biomed Sci ; 26(1): 46, 2019 Jun 12.
Article En | MEDLINE | ID: mdl-31189465

BACKGROUND: Triggering receptor expressed on myeloid cells-1 (TREM-1) is highly expressed on macrophages in inflamed intestines and reportedly promotes inflammatory bowel disease (IBD) by augmenting pro-inflammatory responses. To study the mechanism mediated by TREM-1 on macrophages, we generated an independent TREM-1 deficient mouse. METHODS: Acute colitis was induced in C57BL/6 and TREM-1-deficient mice by the administration of dextran sodium sulfate (DSS). Colonic lamina propria immune cell composition and cytokines were analyzed. An innate lymphoid cell (ILC) co-culture experiment with macrophages was used to analyze IL-22 levels. Exogenous IL-22 and TREM-1-expressing macrophages were supplied to TREM-1-deficient mice for examining their effects on intestinal barrier integrity. RESULTS: In inflamed colons, TREM-1 loss compromised the activation of ILC3 and their production of IL-22, which is required for intestinal barrier integrity. ILC3-mediated IL-22 production depends on IL-1ß secreted by M1-polarized macrophages, and we found that TREM-1 deficiency results in a decreased number of IL-1ß producing-M1 macrophages in colons exposed to DSS. Accordingly, DSS-mediated damage was ameliorated by supplying exogenous IL-22 and TREM-1-expressing macrophages to TREM-1-deficient mice. CONCLUSIONS: TREM-1 plays a crucial role in regulating IL-22 production by ILC3 through modulating M1-macrophage polarization during DSS-induced acute colitis.


Colitis/pathology , Interleukins/metabolism , Intestinal Mucosa/physiology , Lymphocytes/immunology , Macrophages/physiology , Triggering Receptor Expressed on Myeloid Cells-1/metabolism , Animals , Colitis/chemically induced , Colitis/physiopathology , Dextran Sulfate/toxicity , Intestinal Mucosa/drug effects , Macrophages/drug effects , Male , Mice , Mice, Knockout , Interleukin-22
18.
Stem Cells ; 37(5): 631-639, 2019 05.
Article En | MEDLINE | ID: mdl-30681755

Induced pluripotent stem cells (iPSCs) can attenuate the pathological severity and neutrophil migration of lipopolysaccharide (LPS)-induced acute lung injury (ALI). However, interactions that may occur between iPSCs and the triggering receptor expressed on myeloid cells (TREM) family of proteins remain unclear. In this study, murine iPSCs (miPSCs) were delivered via tail vein injection to wild type, TREM-1 knockout (KO), and TREM-2 KO C57BL/6 mice 4 hours after an intratracheal delivery of LPS. Twenty-four hours later, the bronchoalveolar lavage fluid and lung tissue were collected to perform histology, immunohistochemistry, neutrophil counts, Western blot assays, and enzyme-linked immunosorbent assays. Neutrophils were also isolated from the bone marrow to perform in vitro migration assays. In the lung tissues collected, LPS increased the expression of TREM-1 and TREM-2, with the TREM-2 KO mice expressing more TREM-1 than the wild-type mice. The TREM-2 KO mice also exhibited greater severity of LPS-induced ALI, enhanced neutrophil infiltration in the lung tissues, and a higher ratio of phosphorylated p38 to total p38 (p-p38/p38) in neutrophils. The p-p38/p38 ratio and the expression of vascular cell adhesion molecule-1 and certain proinflammatory cytokines (macrophage inflammatory protein-2, tumor necrosis factor-α, interleukin-6, and interleukin-1ß) were increased in whole lung extracts following LPS-induced ALI, and these levels were even more in LPS-treated TREM-2 KO mice. These effects were reduced when miPSCs were administered. Thus, the results of this study suggest that miPSCs attenuate the role of neutrophils in lung inflammation and injury induced by LPS by reducing their expression of TREM-1 and p38 mitogen-activated protein kinase signaling. Stem Cells 2019;37:631-639.


Acute Lung Injury/therapy , Induced Pluripotent Stem Cells/transplantation , Neutrophil Infiltration/genetics , Triggering Receptor Expressed on Myeloid Cells-1/genetics , p38 Mitogen-Activated Protein Kinases/genetics , Acute Lung Injury/chemically induced , Acute Lung Injury/genetics , Acute Lung Injury/pathology , Animals , Chemokine CXCL2/genetics , Endotoxins/toxicity , Gene Expression Regulation, Developmental/drug effects , Humans , Interleukin-1beta/genetics , Lipopolysaccharides/toxicity , Lung/metabolism , Mice , Mice, Knockout , Myeloid Cells/metabolism , Phosphorylation , Receptors, Immunologic/genetics , Signal Transduction/genetics
19.
Sci Rep ; 8(1): 8223, 2018 05 29.
Article En | MEDLINE | ID: mdl-29844416

Triggering receptor expressed on myeloid cells 1 (TREM-1) amplifies inflammatory responses and is upregulated during sepsis and pulmonary infection. The association between serum soluble TREM-1 (sTREM-1) level and pulmonary tuberculosis (PTB) disease deserves investigation. In the present study, patients with PTB, latent TB infection (LTBI), and non-TB, non-LTBI subjects were prospectively enrolled and serum levels of sTREM-1, sTREM-2, and C-reactive protein (CRP) were measured. We correlated serum biomarkers and clinical presentations and treatment outcomes of PTB cases. We also utilized immunohistochemistry (IHC) to visualize TREM-1-expressing cells in lung tissues from PTB patients. A total of 86 PTB, 41 LTBI, and 20 non-TB, non-LTBI subjects were enrolled. Serum levels of sTREM-1 and CRP significantly increased in PTB patients; these higher serum levels were correlated with more advanced involvement in chest films and higher bacteria burden in sputum. In multivariate analysis, serum levels of sTREM-1 >260 pg/mL and CRP >2.6 mg/L were independent predictors for on-treatment mortality. Abundant TREM-1-expressing macrophages were identified in lung tissues from PTB samples. In conclusion, serum levels of sTREM-1 correlated with disease severity and treatment outcomes in PTB patients.


Triggering Receptor Expressed on Myeloid Cells-1/physiology , Tuberculosis, Pulmonary/blood , Aged , Aged, 80 and over , Antitubercular Agents/therapeutic use , Biomarkers/blood , Female , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Lung/metabolism , Male , Middle Aged , Treatment Outcome , Triggering Receptor Expressed on Myeloid Cells-1/blood , Triggering Receptor Expressed on Myeloid Cells-1/metabolism , Tuberculosis, Pulmonary/drug therapy
20.
PLoS One ; 12(10): e0186780, 2017.
Article En | MEDLINE | ID: mdl-29065139

Tuberculosis is a fatal human infectious disease caused by Mycobacterium tuberculosis (M. tuberculosis) that is prevalent worldwide. Mycobacteria differ from other bacteria in that they have a cell wall composed of specific surface glycans that are the major determinant of these organisms' pathogenicity. The interaction of M. tuberculosis with pattern recognition receptors (PRRs), in particular C-type lectin receptors (CLRs), on the surface of macrophages plays a central role in initiating innate and adaptive immunity, but the picture as a whole remains a puzzle. Defining novel mechanisms by which host receptors interact with pathogens in order to modulate a specific immune response is an area of intense research. In this study, based on an in vitro lectin binding assay, CLEC9A (DNGR-1) is identified as a novel CLR that binds with mycobacteria. Our results with CLEC9A-knocked down cells and a CLEC9A-Fc fusion protein as blocking agents show that CLEC9A is involved in the activation of SYK and MAPK signaling in response to heat-killed M. tuberculosis H37Ra treatment, and it then promotes the production of CXCL8 and IL-1ß in macrophages. The CXCL8 and IL-1ß secreted by the activated macrophages are critical to neutrophil recruitment and activation. In a in vivo mouse model, when the interaction between CLEC9A and H37Ra is interfered with by treatment with CLEC9A-Fc fusion protein, this reduces lung inflammation and cell infiltration. These findings demonstrate that CLEC9A is a specialized receptor that modulates the innate immune response when there is a mycobacterial infection.


Hot Temperature , Lectins, C-Type/physiology , Macrophages/physiology , Mycobacterium tuberculosis/physiology , Neutrophils/cytology , Receptors, Mitogen/physiology , Animals , Cell Line , Gene Knockdown Techniques , Humans , Lectins, C-Type/genetics , Macrophages/enzymology , Male , Mice , Mice, Inbred C57BL , Protein Kinases/metabolism , Receptors, Mitogen/genetics , Signal Transduction
...