Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Ann Rheum Dis ; 2024 May 22.
Article in English | MEDLINE | ID: mdl-38777379

ABSTRACT

OBJECTIVE: Tissue-resident memory cells (Trm) are a subset of T cells residing persistently and long-term within specific tissues that contribute to persistent inflammation and tissue damage. We characterised the phenotype and function of Trm and the role of CD103 in primary Sjogren's syndrome (pSS). METHODS: In both pSS and non-pSS sicca syndrome patients, we examined Trm frequency, cytokine production in salivary glands (SG) and peripheral blood (PB). We also analysed Trm-related gene expression in SG biopsies through bulk and single-cell RNA sequencing (scRNAseq). Additionally, we investigated Trm properties in an immunisation-induced animal model of pSS (experimental SS, ESS) mouse model and assessed the effects of Trm inhibition via intraglandular anti-CD103 monoclonal antibody administration. RESULTS: Transcriptomic pSS SG showed an upregulation of genes associated with tissue recruitment and long-term survival of Trm cells, confirmed by a higher frequency of CD8+CD103+CD69+ cells in pSS SG, compared with non-specific sialadenitis (nSS). In SG, CD8+ CD103+ Trm contributed to the secretion of granzyme-B and interferon-γ, CD8+ Trm cells were localised within inflammatory infiltrates, where PD1+CD8+ T cells were also increased compared with nSS and MALT lymphoma. scRNAseq of PB and pSS SG T cells confirmed expression of CD69, ITGAE, GZMB, GZMK and HLA-DRB1 among CD3+CD8+ SG T cells. In the SG of ESS, CD8+CD69+CD103+ Trm producing Granzyme B progressively expanded. However, intraglandular blockade of CD103 in ESS reduced Trm, reduced glandular damage and improved salivary flow. CONCLUSIONS: CD103+CD8+Trm cells are expanded in the SG of pSS and ESS, participate in tissue inflammation and can be therapeutically targeted.

2.
Cell Transplant ; 31: 9636897221116085, 2022.
Article in English | MEDLINE | ID: mdl-36062473

ABSTRACT

Nasopharyngeal carcinoma (NPC) is a unique malignant tumor of the head and neck. Despite higher survival rates by the combination of radiotherapy and chemotherapy, the recurrence or metastasis of NPC still occurs at about 10%. Therefore, there is urgent demand to develop more effective in vivo models for preclinical trials to investigate the mechanisms of NPC development and progression and to explore better treatment approaches. In this study, we transplanted human NPC CNE1 cells into zebrafish embryos to establish a xenograft model of NPC, where the proliferation and invasion behaviors of NPC cells were investigated in vivo. Combining in vitro and in vivo analyses, we found that activating transcription factor 7 (ATF7) was involved in the occurrence and development of NPC regulated by peptidyl-prolyl cis-trans isomerase NIMA-interacting 1 (Pin1). The zebrafish NPC xenograft model established here thereby provides an in vivo tool for exploring the occurrence and development of NPC, which may help to identify new tumor markers and develop new therapeutic strategies for the treatment of NPC.


Subject(s)
Carcinoma , Nasopharyngeal Neoplasms , Animals , Carcinoma/drug therapy , Carcinoma/genetics , Carcinoma/pathology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Disease Models, Animal , Gene Expression Regulation, Neoplastic , Heterografts , Humans , NIMA-Interacting Peptidylprolyl Isomerase/metabolism , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/drug therapy , Zebrafish/metabolism
4.
EMBO Rep ; 23(3): e53602, 2022 02 03.
Article in English | MEDLINE | ID: mdl-34935271

ABSTRACT

Cortical expansion and folding are key processes in human brain development and evolution and are considered to be principal elements of intellectual ability. How cortical folding has evolved and is induced during embryo development is not well understood. Here, we show that the expression of human FOXM1 promotes basal progenitor cell proliferation and induces cortical thickening and folding in mice. Human-specific protein sequences further promote the generation of basal progenitor cells. Human FOXM1 increases the proliferation of neural progenitors by binding to the Lin28a promoter and increasing Lin28a expression. Furthermore, overexpression of LIN28A rescues the proliferation of human FOXM1 knockout neural progenitor cells. Together, our findings demonstrate that a human gene can increase the number of basal progenitor cells in mice, leading to brain size increase and gyrification, and may thus contribute to evolutionary brain development and cortical expansion.


Subject(s)
Brain/cytology , Cell Proliferation , Forkhead Box Protein M1 , Neural Stem Cells , Animals , Forkhead Box Protein M1/genetics , Forkhead Box Protein M1/metabolism , Humans , Mice , Neural Stem Cells/cytology , RNA-Binding Proteins
5.
Front Immunol ; 12: 697725, 2021.
Article in English | MEDLINE | ID: mdl-34804004

ABSTRACT

The intestinal mucosal immune environment requires multiple immune cells to maintain homeostasis. Although intestinal B cells are among the most important immune cells, little is known about the mechanism that they employ to regulate immune homeostasis. In this study, we found that CD11b+ B cells significantly accumulated in the gut lamina propria and Peyer's patches in dextran sulfate sodium-induced colitis mouse models and patients with ulcerative colitis. Adoptive transfer of CD11b+ B cells, but not CD11b-/- B cells, effectively ameliorated colitis and exhibited therapeutic effects. Furthermore, CD11b+ B cells were found to produce higher levels of IgA than CD11b- B cells. CD11b deficiency in B cells dampened IgA production, resulting in the loss of their ability to ameliorate colitis. Mechanistically, CD11b+ B cells expressed abundant TGF-ß and TGF-ß receptor II, as well as highly activate phosphorylated Smad2/3 signaling pathway, consequently promoting the class switch to IgA. Collectively, our findings demonstrate that CD11b+ B cells are essential intestinal suppressive immune cells and the primary source of intestinal IgA, which plays an indispensable role in maintaining intestinal homeostasis.


Subject(s)
B-Lymphocytes/immunology , CD11b Antigen/immunology , Colitis, Ulcerative/immunology , Colitis/immunology , Immunoglobulin A, Secretory/immunology , Peyer's Patches/immunology , Adoptive Transfer , Animals , B-Lymphocytes/pathology , CD11b Antigen/genetics , Colitis/chemically induced , Colitis/pathology , Colitis, Ulcerative/pathology , Dextran Sulfate/toxicity , Disease Models, Animal , Humans , Immunoglobulin Class Switching , Intestinal Mucosa/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Peyer's Patches/pathology , Signal Transduction , Smad2 Protein/metabolism
6.
Front Immunol ; 12: 684999, 2021.
Article in English | MEDLINE | ID: mdl-34168653

ABSTRACT

Primary Sjögren's syndrome (pSS) is a chronic autoimmune disease characterized by lymphocytic infiltration and tissue destruction of exocrine glands such as salivary glands. Although the formation of ectopic lymphoid tissue in exocrine glands and overproduction of autoantibodies by autoreactive B cells highlight the critical involvement of B cells in disease development, the precise roles of various B cell subsets in pSS pathogenesis remain partially understood. Current studies have identified several novel B cell subsets with multiple functions in pSS, among which autoreactive age-associated B cells, and plasma cells with augmented autoantibody production contribute to the disease progression. In addition, tissue-resident Fc Receptor-Like 4 (FcRL4)+ B cell subset with enhanced pro-inflammatory cytokine production serves as a key driver in pSS patients with mucosa-associated lymphoid tissue (MALT)-lymphomas. Recently, regulatory B (Breg) cells with impaired immunosuppressive functions are found negatively correlated with T follicular helper (Tfh) cells in pSS patients. Further studies have revealed a pivotal role of Breg cells in constraining Tfh response in autoimmune pathogenesis. This review provides an overview of recent advances in the identification of pathogenic B cell subsets and Breg cells, as well as new development of B-cell targeted therapies in pSS patients.


Subject(s)
B-Lymphocytes, Regulatory/immunology , B-Lymphocytes, Regulatory/metabolism , Plasma Cells/immunology , Receptors, Fc/metabolism , Sjogren's Syndrome/etiology , Biomarkers , Cytokines/metabolism , Disease Susceptibility , Humans , Lymphoma, B-Cell, Marginal Zone/pathology , Plasma Cells/metabolism , Receptors, Fc/genetics , Sjogren's Syndrome/metabolism , Sjogren's Syndrome/pathology , T Follicular Helper Cells/immunology
7.
Clin Transl Immunology ; 10(4): e1277, 2021.
Article in English | MEDLINE | ID: mdl-33968407

ABSTRACT

OBJECTIVES: This study aims to determine a role of interleukin-17A (IL-17) in salivary gland (SG) dysfunction and therapeutic effects of targeting IL-17 in SG for treating autoimmune sialadenitis in primary Sjögren's syndrome (pSS). METHODS: Salivary IL-17 levels and IL-17-secreting cells in labial glands of pSS patients were examined. Kinetic changes of IL-17-producing cells in SG from mice with experimental Sjögren's syndrome (ESS) were analysed. To determine a role of IL-17 in salivary secretion, IL-17-deficient mice and constructed chimeric mice with IL-17 receptor C (IL-17RC) deficiency in non-hematopoietic and hematopoietic cells were examined for saliva flow rates during ESS development. Both human and murine primary SG epithelial cells were treated with IL-17 for measuring cholinergic activation-induced calcium movement. Moreover, SG functions were assessed in ESS mice with salivary retrograde cannulation of IL-17 neutralisation antibodies. RESULTS: Increased salivary IL-17 levels were negatively correlated with saliva flow rates in pSS patients. Both IL-17-deficient mice and chimeric mice with non-hematopoietic cell-restricted IL-17RC deficiency exhibited no obvious salivary reduction while chimeric mice with hematopoietic cell-restricted IL-17RC deficiency showed significantly decreased saliva secretion during ESS development. In SG epithelial cells, IL-17 inhibited acetylcholine-induced calcium movement and downregulated the expression of transient receptor potential canonical 1 via promoting Nfkbiz mRNA stabilisation. Moreover, local IL-17 neutralisation in SG markedly attenuated hyposalivation and ameliorated tissue inflammation in ESS mice. CONCLUSION: These findings identify a novel function of IL-17 in driving salivary dysfunction during pSS development and may provide a new therapeutic strategy for targeting SG dysfunction in pSS patients.

8.
Nat Commun ; 12(1): 1914, 2021 03 26.
Article in English | MEDLINE | ID: mdl-33772013

ABSTRACT

Innate immunity is important for host defense by eliciting rapid anti-viral responses and bridging adaptive immunity. Here, we show that endogenous lipids released from virus-infected host cells activate lung γδ T cells to produce interleukin 17 A (IL-17A) for early protection against H1N1 influenza infection. During infection, the lung γδ T cell pool is constantly supplemented by thymic output, with recent emigrants infiltrating into the lung parenchyma and airway to acquire tissue-resident feature. Single-cell studies identify IL-17A-producing γδ T (Tγδ17) cells with a phenotype of TCRγδhiCD3hiAQP3hiCXCR6hi in both infected mice and patients with pneumonia. Mechanistically, host cell-released lipids during viral infection are presented by lung infiltrating CD1d+ B-1a cells to activate IL-17A production in γδ T cells via γδTCR-mediated IRF4-dependent transcription. Reduced IL-17A production in γδ T cells is detected in mice either lacking B-1a cells or with ablated CD1d in B cells. Our findings identify a local host-immune crosstalk and define important cellular and molecular mediators for early innate defense against lung viral infection.


Subject(s)
Influenza A Virus, H1N1 Subtype/immunology , Influenza, Human/immunology , Lipids/immunology , Orthomyxoviridae Infections/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , Animals , Antigens, CD1d/immunology , Antigens, CD1d/metabolism , Female , Host-Pathogen Interactions/immunology , Humans , Immunity, Innate/immunology , Influenza A Virus, H1N1 Subtype/physiology , Influenza, Human/metabolism , Influenza, Human/virology , Interferon Regulatory Factors/immunology , Interferon Regulatory Factors/metabolism , Interleukin-17/immunology , Interleukin-17/metabolism , Lung/immunology , Lung/metabolism , Lung/virology , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Orthomyxoviridae Infections/metabolism , Orthomyxoviridae Infections/virology , Receptors, Antigen, T-Cell, gamma-delta/metabolism
9.
Mod Rheumatol ; 31(5): 927-932, 2021 Sep.
Article in English | MEDLINE | ID: mdl-33427554

ABSTRACT

The pandemic of COVID-19 has caused global social impact and high health risk. Clinical observations have suggested that elevated levels of inflammatory mediators are associated with disease severities in COVID-19 patients, in which the immunological profiles indicate the hyperactivation of innate immune cells and dysregulated adaptive immune responses. The increasing prevalence and disease progression of COVID-19 has emerged as a pressing challenge for the management of rheumatic patients with immune dysregulations. Here we review the immune dysregulations in COVID-19 and discuss the management of COVID-19 patients with rheumatic diseases.


Subject(s)
COVID-19 , Immune System/physiopathology , Rheumatic Diseases , COVID-19/immunology , COVID-19/physiopathology , Humans , Pandemics , Rheumatic Diseases/epidemiology , Rheumatic Diseases/therapy
10.
Cell Mol Immunol ; 18(7): 1739-1750, 2021 07.
Article in English | MEDLINE | ID: mdl-32917979

ABSTRACT

Recent studies have demonstrated a central role for plasma cells in the development of autoimmune diseases, such as systemic lupus erythematosus (SLE). Currently, both the phenotypic features and functional regulation of autoreactive plasma cells during SLE pathogenesis remain largely unclear. In this study, we first found that a major subset of IL-17 receptor-expressing plasma cells potently produced anti-dsDNA IgG upon IL-17A (IL-17) stimulation in SLE patients and lupus mice. Using a humanized lupus mouse model, we showed that the transfer of Th17 cell-depleted PBMCs from lupus patients resulted in a significantly reduced plasma cell response and attenuated renal damage in recipient mice compared to the transfer of total SLE PBMCs. Moreover, long-term BrdU incorporation in lupus mice detected highly enriched long-lived BrdU+ subsets among IL-17 receptor-expressing plasma cells. Lupus mice deficient in IL-17 or IL-17 receptor C (IL-17RC) exhibited a diminished plasma cell response and reduced autoantibody production with attenuated renal damage, while the adoptive transfer of Th17 cells triggered the plasma cell response and renal damage in IL-17-deficient lupus mice. In reconstituted chimeric mice, IL-17RC deficiency resulted in severely impaired plasma cell generation but showed no obvious effect on germinal center B cells. Further mechanistic studies revealed that IL-17 significantly promoted plasma cell survival via p38-mediated Bcl-xL transcript stabilization. Together, our findings identified a novel function of IL-17 in enhancing plasma cell survival for autoantibody production in lupus pathogenesis, which may provide new therapeutic strategies for the treatment of SLE.


Subject(s)
Lupus Erythematosus, Systemic , Plasma Cells , Animals , Germinal Center , Humans , Interleukin-17/metabolism , Mice , RNA Stability
11.
Front Immunol ; 12: 816839, 2021.
Article in English | MEDLINE | ID: mdl-35095918

ABSTRACT

Inflammasome is a cytoplasmic multiprotein complex that facilitates the clearance of exogenous microorganisms or the recognition of endogenous danger signals, which is critically involved in innate inflammatory response. Excessive or abnormal activation of inflammasomes has been shown to contribute to the development of various diseases including autoimmune diseases, neurodegenerative changes, and cancers. Rheumatoid arthritis (RA) is a chronic and complex autoimmune disease, in which inflammasome activation plays a pivotal role in immune dysregulation and joint inflammation. This review summarizes recent findings on inflammasome activation and its effector mechanisms in the pathogenesis of RA and potential development of therapeutic targeting of inflammasome for the immunotherapy of RA.


Subject(s)
Arthritis, Rheumatoid/etiology , Arthritis, Rheumatoid/metabolism , Biomarkers , Disease Susceptibility , Inflammasomes/metabolism , Animals , Arthritis, Rheumatoid/diagnosis , Arthritis, Rheumatoid/drug therapy , Autoimmunity , Disease Management , Genetic Predisposition to Disease , Humans , Inflammasomes/immunology , Molecular Targeted Therapy , Polymorphism, Single Nucleotide
12.
Int J Mol Sci ; 21(15)2020 Jul 22.
Article in English | MEDLINE | ID: mdl-32708044

ABSTRACT

Tissue injury and inflammatory response trigger the development of fibrosis in various diseases. It has been recognized that both innate and adaptive immune cells are important players with multifaceted functions in fibrogenesis. The activated immune cells produce various cytokines, modulate the differentiation and functions of myofibroblasts via diverse molecular mechanisms, and regulate fibrotic development. The immune cells exhibit differential functions during different stages of fibrotic diseases. In this review, we summarized recent advances in understanding the roles of immune cells in regulating fibrotic development and immune-based therapies in different disorders and discuss the underlying molecular mechanisms with a focus on mTOR and JAK-STAT signaling pathways.


Subject(s)
Adaptive Immunity , Fibrosis/immunology , Immunity, Innate , Signal Transduction/immunology , Animals , B-Lymphocytes/immunology , Cytokines/metabolism , Dendritic Cells/immunology , Fibrosis/pathology , Fibrosis/therapy , Humans , Lymphopoiesis/immunology , Macrophages/immunology , Myofibroblasts/metabolism , Neutrophils/immunology , T-Lymphocytes/immunology
13.
Clin Exp Rheumatol ; 37 Suppl 118(3): 209-216, 2019.
Article in English | MEDLINE | ID: mdl-31376261

ABSTRACT

Sjögren's syndrome (SS) is a chronic autoimmune disease characterised by lymphocytic infiltration in exocrine glands with secretory dysfunction. Although both environmental triggers and genetic predisposition have been recognised as important factors in the initiation and development of SS, the pathogenesis of SS is complex and still largely unclear. Animal models have served as useful tools for studying SS pathogenesis with several advantages. A number of animal models recapitulating key characteristics of primary SS patients including secretory dysfunction, glandular inflammation and presence of autoantibodies were developed in the past years. The studies based on the animal models of SS have provided significant insight in SS pathogenesis and therapeutic intervention. This review summarises current animal models with primary SS-like symptoms including spontaneous models, genetically modified models, induced models and humanised models, and discusses their contribution to the understanding of SS aetiology and therapies.


Subject(s)
Autoimmune Diseases , Disease Models, Animal , Sjogren's Syndrome , Animals , Autoantibodies , Humans
14.
Mol Immunol ; 112: 266-273, 2019 08.
Article in English | MEDLINE | ID: mdl-31212097

ABSTRACT

Myeloid derived suppressor cells (MDSCs) play a key role in tumor immunosuppressive microenvironment, which helps tumors avoid immune destruction. Blocking the suppressive activities of MDSCs could be a promising strategy to enhance the effect of anti-tumor immunotherapies. In this study, we found that TLR1/TLR2 expression predicted favorable prognosis of lung cancer patients. In the related mice tumor model, TLR1/TLR2 activation by synthetic bacterial lipoprotein (BLP), a TLR1/2 agonist, greatly inhibited tumor growth and selectively decreased monocytic MDSCs (M-MDSCs). Furthermore, BLP treatment redirected M-MDSC differentiation towards M1 macrophage through JNK pathway, and thus blocked the suppressive activity of M-MDSCs in a TLR2-dependent manner. Therefore, our data demonstrated that TLR2 could be a promising biomarker and a potential immunotherapeutic target for lung cancer.


Subject(s)
Cell Differentiation/genetics , Macrophages/physiology , Myeloid-Derived Suppressor Cells/physiology , Signal Transduction/genetics , Toll-Like Receptor 1/genetics , Toll-Like Receptor 2/genetics , Animals , Biomarkers, Tumor/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Immunotherapy/methods , Mice , Mice, Inbred C57BL , Monocytes/physiology , Myeloid Cells , Tumor Microenvironment/genetics
15.
Immunology ; 156(1): 56-68, 2019 01.
Article in English | MEDLINE | ID: mdl-30171602

ABSTRACT

B lymphocytes, known as antibody producers, mediate tumor cell destruction in the manner of antibody-dependent cell-mediated cytotoxicity; however, their anti-tumor function seems to be weakened during tumorigenesis, while the underlying mechanisms remain unclear. In this study, we found that IgG mediated anti-tumor effects, but IgG-producing B cells decreased in various tumors. Considering the underlying mechanism, glycometabolism was noteworthy. We found that tumor-infiltrating B cells were glucose-starved and accompanied by a deceleration of glycometabolism. Both inhibition of glycometabolism and deprivation of glucose through tumor cells, or glucose-free treatment, reduced the differentiation of B cells into IgG-producing cells. In this process, special AT-rich sequence-binding protein-1 (SATB1) was significantly silenced in B cells. Down-regulating SATB1 by inhibiting glycometabolism or RNA interference reduced the binding of signal transducer and activator of transcription 6 (STAT6) to the promoter of germline Cγ gene, subsequently resulting in fewer B cells producing IgG. Our findings provide the first evidence that glycometabolic inhibition by tumorigenesis suppresses differentiation of B cells into IgG-producing cells, and altering glycometabolism may be promising in improving the anti-tumor effect of B cells.


Subject(s)
Adenocarcinoma/immunology , B-Lymphocytes/metabolism , Colorectal Neoplasms/immunology , Glucose/metabolism , Lung Neoplasms/immunology , Matrix Attachment Region Binding Proteins/metabolism , Neoplasms/immunology , Aged , Animals , Azoxymethane , B-Lymphocytes/immunology , Cells, Cultured , Colorectal Neoplasms/chemically induced , Dextran Sulfate , Disease Models, Animal , Female , Humans , Immunoglobulin G/genetics , Immunoglobulin G/metabolism , Male , Matrix Attachment Region Binding Proteins/genetics , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic/genetics , RNA, Small Interfering/genetics , STAT6 Transcription Factor/metabolism
16.
Immunology ; 155(3): 356-366, 2018 11.
Article in English | MEDLINE | ID: mdl-29969845

ABSTRACT

Regulatory T (Treg) cells play an essential role in the maintenance of intestinal homeostasis. In Peyer's patches (PPs), which comprise the most important IgA induction site in the gut-associated lymphoid tissue, Treg cells promote IgA isotype switching. However, the mechanisms underlying their entry into PPs and isotype switching facilitation in activated B cells remain unknown. This study, based on the dextran sulphate sodium (DSS)-induced colitis model, revealed that Treg cells are significantly increased in PPs, along with CD11b+ B-cell induction. Immunofluorescence staining showed that infiltrated Treg cells were located around CD11b+ B cells and produced transforming growth factor-ß, thereby inducing IgA+ B cells. Furthermore, in vivo and in vitro studies revealed that CD11b+ B cells in PPs had the capacity to recruit Treg cells into PPs rather than promoting their proliferation. Finally, we found that Treg cell recruitment was mediated by the chemokine CXCL9 derived from CD11b+ B cells in PPs. These findings demonstrate that CD11b+ B cells induced in PPs during colitis actively recruit Treg cells to accomplish IgA isotype switch in a CXCL9-dependent manner.


Subject(s)
B-Lymphocytes/immunology , CD11b Antigen/immunology , Chemokine CXCL9/immunology , Colitis/immunology , Peyer's Patches/immunology , T-Lymphocytes, Regulatory/immunology , Animals , B-Lymphocytes/pathology , CD11b Antigen/genetics , Chemokine CXCL9/genetics , Colitis/chemically induced , Colitis/genetics , Colitis/pathology , Dextran Sulfate/toxicity , Disease Models, Animal , Mice , Mice, Knockout , Peyer's Patches/pathology , T-Lymphocytes, Regulatory/pathology
17.
Hepatology ; 67(3): 1027-1040, 2018 03.
Article in English | MEDLINE | ID: mdl-29023933

ABSTRACT

Interleukin-22 (IL-22), as a link between leukocytic and nonleukocytic cells, has gained increasing attention for its pronounced tissue-protective properties. MicroRNAs, emerging as crucial immune modulators, have been reported to be involved in the production and action of various cytokines. However, the precise control of IL-22 by microRNAs and its subsequent actions remained to be elucidated. In this study, we found a negative correlation between the expression of microRNA 15a/16-1 (miR-15a/16-1) and IL-22 in the model of concanavalin A-induced, immune-mediated liver injury. Knockout of miR-15a/16-1 ameliorated liver injury in an IL-22-dependent manner. Further results revealed that cluster of differentiation 4-positive (CD4+ ) T cells were the major source of IL-22 during liver injury and that the aryl hydrocarbon receptor was the direct target of miR-15a/16-1 in CD4+ T cells. In vivo and in vitro data showed that miR-15a/16-1 knockout CD4+ T cells produced more IL-22, while overexpression of miR-15a/16-1 down-regulated the IL-22 production by inhibiting the aryl hydrocarbon receptor. Moreover, transfer of miR-15a/16-1 knockout CD4+ T cells promoted tissue repair compared to wild-type CD4+ T cells by up-regulating IL-22. In addition, as a synergistic effect, IL-22 could down-regulate miR-15a/16-1 expression by activating phosphorylated signal transducer and activator of transcription 3-c-myc signaling, and the decrease of miR-15a/16-1 in damaged hepatocytes contributed to IL-22-mediated tissue repair by reducing cell apoptosis and promoting cell proliferation. As further proof, we demonstrated the role of miR-15a/16-1 in controlling IL-22 production and IL-22-mediated reconstruction of the intestinal epithelial barrier in a dextran sodium sulfate-induced colitis model. CONCLUSION: Our results suggest that miR-15a/16-1 acts as a essential regulator of IL-22 and that the miR-15a/16-1-aryl hydrocarbon receptor-IL-22 regulatory axis plays a central role in tissue repair; modulation of miR-15a/16-1 might hold promise in developing new strategies to enhance IL-22-mediated tissue repair. (Hepatology 2018;67:1027-1040).


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Interleukins/metabolism , MicroRNAs/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Animals , Cell Line, Tumor , Colitis/chemically induced , Colitis/metabolism , Flow Cytometry , Gene Expression Regulation , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Regeneration/genetics , Signal Transduction/genetics , Interleukin-22
18.
Gastroenterology ; 154(3): 637-651.e7, 2018 02.
Article in English | MEDLINE | ID: mdl-29031499

ABSTRACT

BACKGROUND & AIMS: B cells infiltrate tumors, but little is known about how they affect tumor growth and progression. microRNA15A (MIR15A or miRNA15A) and microRNA16-1 (MIR16-1 or miRNA16-1) regulate cell proliferation, apoptosis, and drug resistance. We investigated their involvement in B-cell-mediated immune suppression by colorectal tumors. METHODS: Mice with disruptions of the gene cluster that encodes MIR15A and MIR16-1 (knockout mice), and control (C57BL/B6) mice were given azoxymethane with dextran sodium sulfate (AD) to induce formation of colorectal tumors. Mice were given anti-CD20 to delete B cells, or injections of agomir to increase MIR15A and MIR16-1. Proliferation of CD8+T cells was measured by carboxyfluorescein-succinimidyl-ester analysis. Colon tissues were collected from mice and analyzed by flow cytometry, microRNA (miRNA) sequencing, and for cytokine production. Intestinal epithelial cells (IECs) were isolated and transfected with miRNA mimics, to identify their targets. We analyzed miRNA expression patterns and quantified B cells in colorectal cancer tissue microarrays derived from 90 patients who underwent surgical resection, from July 2006 through April 2008, in Shanghai, China; expression data were compared with clinical outcomes. RESULTS: Tumors that developed in knockout mice following administration of AD were larger and contained greater numbers of B cells than tumors that grew in control mice. Most of the B cells in the tumors were positive for immunoglobulin A (IgA+). IgA+ B cells expressed high levels of immune regulatory molecules (programmed death ligand 1, interleukin 10, and transforming growth factor beta), and repressed the proliferation and activation of CD8+ T cells. Levels of MIR15A and MIR16-1 were reduced in colon tumors from mice, compared with nontumor colon tissue. Incubation of IECs with IL17A reduced expression of MIR15A and MIR16-1. Transgenic expression of MIR15A and MIR16-1 in IECs decreased activation of NF-κB and STAT1 by reducing expression of I-kappaB kinases; this resulted in reduced production of chemokine (C-X-C motif) ligands 9 and 10 and decreased chemotaxis of IgA+ B cells. Tumors in mice injected with AD and agomir grew more slowly than tumors in mice not given in agomir and contained fewer IgA+ B cells. We found a negative correlation between levels of MIR15A and MIR16-1 and numbers of IgA+B cells in human colorectal tumor tissues; high levels of MIR15A and MIR16-1 and low numbers of IgA+B cells were associated with longer survival times of patients. CONCLUSIONS: We found increased levels of MIR15A and MIR16-1 to reduce numbers of IgA+ B cells in colorectal tumor tissues and correlate with increased survival time of patients. In mice that lack MIR15A and MIR16-1, colon tumors grow more rapidly and contain increased numbers of IgA+ B cells. MIR15A and MIR16-1 appear to activate signaling pathways required for B-cell-mediated immune suppression.


Subject(s)
B-Lymphocytes, Regulatory/metabolism , Chemotaxis, Leukocyte , Colorectal Neoplasms/metabolism , Lymphocytes, Tumor-Infiltrating/metabolism , MicroRNAs/metabolism , Signal Transduction , Tumor Escape , Animals , Azoxymethane , B-Lymphocytes, Regulatory/immunology , Cell Proliferation , Chemokine CXCL10/immunology , Chemokine CXCL10/metabolism , Chemokine CXCL9/immunology , Chemokine CXCL9/metabolism , Colorectal Neoplasms/chemically induced , Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Dextran Sulfate , Gene Expression Regulation, Neoplastic , Genotype , Humans , I-kappa B Kinase/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/genetics , NF-kappa B/metabolism , Phenotype , STAT1 Transcription Factor/metabolism , Time Factors , Tumor Burden
19.
Cell Death Dis ; 7(12): e2506, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27906181

ABSTRACT

Signal transducer and activator of transcription 6 (STAT6) is involved in epithelial cell growth. However, little is known regarding the STAT6 phosphorylation status in Graves' disease (GD) and its role in thyroid epithelial cells (TECs). In this study, we found that STAT6 phosphorylation (p-STAT6) was significantly increased in TECs from both GD patients and experimental autoimmune Graves' disease mice and that STAT6 deficiency ameliorated GD symptoms. Autocrine IL-4 signalling in TECs activated the phosphorylation of STAT6 via IL-4 R engagement, and the downstream targets of STAT6 were Bcl-xL and cyclin D1. Thus, the IL-4-STAT6-Bcl-xL/cyclin D1 pathway is crucial for TEC hyperplasia, which aggravates GD. More importantly, in vitro and in vivo experiments demonstrated that STAT6 phosphorylation inhibited by AS1517499 decreased TEC hyperplasia, thereby reducing serum T3 and T4 and ameliorating GD. Thus, our study reveals that in addition to the traditional pathogenesis of GD, in which autoantibody TRAb stimulates thyroid-stimulating hormone receptors and consequently produces T3, T4, TRAb could also trigger TECs producing IL-4, and IL-4 then acts in an autocrine manner to activate p-STAT6 signalling and stimulate unrestricted cell growth, thus aggravating GD. These findings suggest that STAT6 inhibitors could be potent therapeutics for treating GD.


Subject(s)
Graves Disease/metabolism , Graves Disease/pathology , STAT6 Transcription Factor/deficiency , Severity of Illness Index , Thyroid Epithelial Cells/metabolism , Thyroid Epithelial Cells/pathology , Animals , Cyclin D1/metabolism , Humans , Hyperplasia , Interleukin-4/metabolism , Mice, Inbred BALB C , Models, Biological , Phosphorylation/drug effects , Pyrimidines/pharmacology , Receptors, Interleukin/metabolism , STAT6 Transcription Factor/metabolism , Thyroid Epithelial Cells/drug effects , Up-Regulation/drug effects , bcl-X Protein/metabolism
20.
Oncotarget ; 7(28): 43401-43411, 2016 Jul 12.
Article in English | MEDLINE | ID: mdl-27270324

ABSTRACT

INTRODUCTION: Esophageal squamous cell carcinoma (ESCC) is one of the most aggressive malignant tumors. Eukaryotic translation initiation factors 3B (EIF3B) is considered to influence tumor proliferation, invasion, apoptosis and cell cycle, which act together to promote the progression of tumors. However, the role of EIF3B in ESCC is unknown. This study aims to explore the clinical and biological role of EIF3B in ESCC. RESULTS: EIF3B expressions were up-regulated in both ESCC tissues and cell lines. Overexpression of EIF3B was associated with tumor depth, lymph node metastasis and advanced TNM stage. Importantly, patients with high EIF3B expression suffered shorter overall and disease-free survival. Knockdown of EIF3B could inhibit cell proliferation and invasion, promote cell apoptosis, and interfere the cell cycle in vitro. EIF3B-knockdown cells could form smaller subcutaneous tumors in vivo. Finally, we demonstrated EIF3B could activate ß-catenin signaling pathway. METHODS: Immunohistochemical staining and Western blot were performed to detect the EIF3B expression in ESCC patient tissues and cell lines. The association between EIF3B expression and patients' prognosis was analyzed by Kaplan-Meier and Cox regression. Then, CCK-8, colony-formation, Transwell and wound-healing assay were performed to compare the bio-functional change after knockdown of EIF3B. Flow cytometry was applied to analyze the change of cell apoptosis and cycle induced by EIF3B knockdown. Tumor xenograft assay was done to verify the in-vitro results. CONCLUSIONS: EIF3B might serve as a novel marker for predicting prognosis of ESCC patients and as a potential therapeutic target, individually or together with other subunits of EIF3 complex.


Subject(s)
Carcinoma, Squamous Cell/mortality , Carcinoma, Squamous Cell/pathology , Esophageal Neoplasms/mortality , Esophageal Neoplasms/pathology , Eukaryotic Initiation Factor-3/metabolism , beta Catenin/metabolism , Apoptosis , Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/surgery , Cell Cycle , Cell Line, Tumor , Cell Movement , Cell Proliferation , Disease Progression , Disease-Free Survival , Esophageal Neoplasms/surgery , Esophageal Squamous Cell Carcinoma , Esophagectomy , Female , Follow-Up Studies , Gene Knockdown Techniques , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Lymphatic Metastasis , Male , Middle Aged , Neoplasm Staging , Prognosis , RNA Interference , RNA, Small Interfering/metabolism , Signal Transduction , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...