Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
J Adv Res ; 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38432392

ABSTRACT

INTRODUCTION: Excessive immune activation induces tissue damage during infection. Compared to external strategies to reconstruct immune homeostasis, host balancing ways remain largely unclear. OBJECTIVES: Here we found a neuroimmune way that prevents infection-induced tissue damage. METHODS: By FACS and histopathology analysis of brain Streptococcus pneumonia meningitis infection model and behavioral testing. Western blot, co-immunoprecipitation, and ubiquitination analyze the Fluoxetine initiate 5-HT7R-STUB1-CCR5 K48-linked ubiquitination degradation. RESULTS: Fluoxetine, a selective serotonin reuptake inhibitor, or the agonist of serotonin receptor 5-HT7R, protects mice from meningitis by inhibiting CCR5-mediated excessive immune response and tissue damage. Mechanistically, the Fluoxetine-5-HT7R axis induces proteasome-dependent degradation of CCR5 via mTOR signaling, and then recruits STUB1, an E3 ubiquitin ligase, to initiate K48-linked polyubiquitination of CCR5 at K138 and K322, promotes its proteasomal degradation. STUB1 deficiency blocks 5-HT7R-mediated CCR5 degradation. CONCLUSION: Our results reveal a neuroimmune pathway that balances anti-infection immunity via happiness neurotransmitter receptor and suggest the 5-HT7R-CCR5 axis as a potential target to promote neuroimmune resilience.

2.
Biosci Rep ; 44(3)2024 Mar 29.
Article in English | MEDLINE | ID: mdl-38323526

ABSTRACT

T cell is vital in the adaptive immune system, which relays on T-cell receptor (TCR) to recognize and defend against infection and tumors. T cells are mainly divided into well-known CD4+ and CD8+ T cells, which can recognize short peptide antigens presented by major histocompatibility complex (MHC) class II and MHC class I respectively in humoral and cell-mediated immunity. Due to the Human Leukocyte Antigen (HLA) diversity and restriction with peptides complexation, TCRs are quite diverse and complicated. To better elucidate the TCR in humans, the present study shows the difference between the TCR repertoire in CD4+ and CD8+ T cells from 30 healthy donors. The result showed count, clonality, diversity, frequency, and VDJ usage in CD4+ and CD8+ TCR-ß repertoire is different, but CDR3 length is not. The Common Clone Cluster result showed that CD4+ and CD8+ TCR repertoires are connected separately between the bodies, which is odd considering the HLA diversity. More knowledge about TCR makes more opportunities for immunotherapy. The TCR repertoire is still a myth for discovery.


Subject(s)
CD8-Positive T-Lymphocytes , Receptors, Antigen, T-Cell, alpha-beta , Humans , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell/genetics , HLA Antigens , CD4-Positive T-Lymphocytes
3.
Int Immunopharmacol ; 128: 111524, 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38232537

ABSTRACT

BACKGROUND: A growing number of studies have found that antidepressants have anti-inflammatory effects while protecting nerves. Hypidone hydrochloride (YL-0919) is a novel highly selective 5-HT reuptake blocker. Our previous studies have demonstrated that YL-0919 exerts notable antidepressant- and anxiolytic-like as well as procognitive effects. However, whether YL-0919 can be used to treat inflammatory and infectious diseases remain unknown. In this study, we aimed to verify the anti-inflammatory effect of YL-0919 on bacterial meningitis and further explore the potential molecular mechanisms. METHODS: We performed the experiments on pneumococcal meningitis mice in vivo and S. pneumoniae infected macrophages/microglia in vitro, with or without YL-0919 treatment. Cognitive function was evaluated by open-field task, Morris water maze test, and novel object recognition test. Histopathological staining and immunofluorescence staining were used to detect the pathological damage of meningitis and NLRP3 inflammasome activation in microglia/macrophages. The expression of the STAT1/NLRP3/GSDMD signal pathway was measured by western blots. Proinflammatory cytokines associated with pyroptosis were detected by ELISA. RESULTS: YL-0919 protected mice from fatal pneumococcal meningitis, characterized by attenuated cytokine storms, decreased bacterial loads, improved neuroethology, and reduced mortality. NLRP3 plays a key role in the regulation of inflammation. When the underlying mechanisms were investigated, we found that YL-0919 inhibited the activation of NLRP3 via STAT1, and thus inhibited macrophages/microglia pyroptosis, resulting in downregulation of proinflammatory cytokines. In addition, Sigma1R was identified as a pivotal receptor that can be engaged by YL-0919 to inhibit NLRP3 activation and pyroptosis pathway in microglia/macrophages. CONCLUSIONS: These results provide new insights into the mechanisms of inflammation regulation mediated by the antidepressant YL-0919. Moreover, targeting the STAT1/NLRP3 pyroptosis pathway is a promising strategy for the treatment of infectious diseases like bacterial meningitis.


Subject(s)
Communicable Diseases , Meningitis, Pneumococcal , Piperidines , Pyridones , Animals , Mice , NLR Family, Pyrin Domain-Containing 3 Protein , Inflammation , Cytokines , Antidepressive Agents , Anti-Inflammatory Agents , Pyroptosis , Inflammasomes
4.
Neurotox Res ; 42(1): 8, 2024 Jan 09.
Article in English | MEDLINE | ID: mdl-38194189

ABSTRACT

Chemotherapy-induced neuropathic pain (CIPN) is a common side effect of antitumor chemotherapeutic agents. It describes a pathological state of pain related to the cumulative dosage of the drug, significantly limiting the efficacy of antitumor treatment. Sofas strategies alleviating CIPN still lack. Calcitonin gene-related peptide (CGRP) is a neuropeptide involved in many pathologic pains. In this study, we explored the effects of CGRP blocking on CIPN and potential mechanisms. Total dose of 20.7 mg/kg cisplatin was used to establish a CIPN mouse model. Mechanical and thermal hypersensitivity was measured using von Frey hairs and tail flick test. Western blot and immunofluorescence were utilized to evaluate the levels of CGRP and activated astrocytes in mouse spinal cord, respectively. In addition, real-time quantitative PCR (RT-qPCR) was used to detect the level of inflammatory cytokines such as IL-6, IL-1ß, and NLRP3 in vitro and in vivo. There are markedly increased CGRP expression and astrocyte activation in the spinal cord of mice following cisplatin treatment. Pretreatment with a monoclonal antibody targeting CGRP (ZR8 mAb) effectively reduced cisplatin-induced mechanical hypersensitivity and thermal nociceptive sensitization and attenuated neuroinflammation as marked by downregulated expression of IL-6, IL-1ß, and NLRP3 in the mice spinal cord and spleen. Lastly, ZR8 mAb does not interfere with the antitumor effects of cisplatin in tumor-bearing mice. Our findings indicate that neutralizing CGRP with monoclonal antibody could effectively alleviate CIPN by attenuating neuroinflammation. CGRP is a promising therapeutic target for CIPN.


Subject(s)
Calcitonin Gene-Related Peptide , Neuralgia , Animals , Mice , Cisplatin/toxicity , NLR Family, Pyrin Domain-Containing 3 Protein , Interleukin-6 , Neuroinflammatory Diseases , Neuralgia/chemically induced , Neuralgia/drug therapy , Antibodies, Monoclonal , Interleukin-1beta
5.
J Infect Dis ; 229(3): 855-865, 2024 Mar 14.
Article in English | MEDLINE | ID: mdl-37603461

ABSTRACT

BACKGROUND: Calcitonin gene-related peptide (CGRP), an immunomodulatory neuropeptide, is important for regulating pain transmission, vasodilation, and the inflammatory response. However, the molecular mechanisms of the CGRP-mediated immune response remain unknown. METHODS: The effects of CGRP on bacterial meningitis (BM) and its underlying mechanisms were investigated in BM mice in vivo and macrophages in vitro. RESULTS: Peripheral injection of CGRP attenuated cytokine storms and protected mice from fatal pneumococcal meningitis, marked by increased bacterial clearance, improved neuroethology, and reduced mortality. When the underlying mechanisms were investigated, we found that CGRP induces proteasome-dependent degradation of major histocompatibility complex class II (MHC-II) in macrophages and then inhibits CD4+ T-cell activation. MARCH1 was identified as an E3 ligase that can be induced by CGRP engagement and promote K48-linked ubiquitination and degradation of MHC-II in macrophages. These results provide new insights into neuropeptide CGRP-mediated immune regulation mechanisms. CONCLUSIONS: We conclude that targeting the nervous system and manipulating neuroimmune communication is a promising strategy for treating intracranial infections like BM.


Subject(s)
Calcitonin Gene-Related Peptide , Meningitis, Bacterial , Mice , Animals , Calcitonin Gene-Related Peptide/metabolism , Histocompatibility Antigens Class II , Ubiquitination , Major Histocompatibility Complex , Homeostasis , Ubiquitin-Protein Ligases/metabolism
6.
J Clin Med ; 12(3)2023 Jan 25.
Article in English | MEDLINE | ID: mdl-36769584

ABSTRACT

Little is known about the distribution of etiology in obstructive sleep apnea (OSA) combined with chronic breathlessness. A significant portion of patients in this group have so-called "overlap syndrome (OVS)", characterized by chronic obstructive pulmonary disease (COPD). OVS has more complications and a poorer prognosis compared to patients with either OSA or COPD alone, which makes it important to identify OVS early in OSA. The current study was a retrospective cross-sectional analysis of consecutive adult patients who were diagnosed with OSA (n = 1062), of whom 275 were hospitalized due to chronic breathlessness. Respiratory and cardiac diseases accounted for the vast majority of causes, followed by gastrointestinal and renal disorders. The final study population comprised 115 patients with OSA alone (n = 64) and OVS (n = 51), who had chronic breathlessness as the primary complaint, not secondary as one of many other complaints. Lymphocytes, CD4 counts, neutrophil-to-lymphocyte ratio (NLR), and PLR were differently expressed between the OSA-alone group and OVS group. The NLR, lymphocytes, and CD4 counts had a moderate diagnostic value for OVS in OSA patients, with AUCs of 0.708 (95% CI, 0.614-0.802), 0.719 (95% CI, 0.624-0.813), and 0.744 (95% CI, 0.653-0.834), respectively. The NLR had the highest AUC for predicting a 6-month re-admission of OVS, with a cut-off of 3.567 and a moderate prognostic value. The sensitivity and specificity were 0.8 and 0.732, respectively. In the animal model, the spleen hematoxylin- and eosin-stained, electron microscopy images showed germinal-center damage, chromatin activation, and mitochondrial swelling under the overlapping effect of intermittent hypoxia and cigarette smoke exposure. OSA with chronic breathlessness cannot be overstated. A significant proportion of patients with COPD in this group had poor lung function at initial diagnosis. The NLR is a useful biomarker to differentiate OVS among OSA patients combined with chronic breathlessness.

7.
Immunol Invest ; 52(1): 1-19, 2023 Jan.
Article in English | MEDLINE | ID: mdl-35997714

ABSTRACT

The mechanisms by which retinoic acid-inducible gene I (RIG-I), a critical RNA virus sensor, is regulated in many biological and pathological processes remain to be determined. Here, we demonstrate that T cell immunoglobulin and mucin protein-3 (Tim-3), an immune checkpoint inhibitor, mediates infection tolerance by suppressing RIG-I-type I interferon pathway. Overexpression or blockade of Tim-3 affects type I interferon expression, virus replication, and tissue damage in mice following H1N1 infection. Tim-3 signaling decreases RIG-I transcription via STAT1 in macrophages and promotes the proteasomal dependent degradation of RIG-I by enhancing K-48-linked ubiquitination via the E3 ligase RNF-122. Silencing RIG-I reversed Tim-3 blockage-mediated upregulation of type I interferon in macrophages. We thus identified a new mechanism through which Tim-3 mediates the immune evasion of H1N1, which may have clinical implications for the treatment of viral diseases.


Subject(s)
Influenza A Virus, H1N1 Subtype , Interferon Type I , Mice , Animals , Hepatitis A Virus Cellular Receptor 2/genetics , Hepatitis A Virus Cellular Receptor 2/metabolism , Macrophages , Interferon Type I/genetics , Ubiquitin-Protein Ligases/genetics
8.
Front Oncol ; 11: 751834, 2021.
Article in English | MEDLINE | ID: mdl-34722306

ABSTRACT

Acute myeloid leukemia (AML), a malignant disorder of hemopoietic stem cells. AML can escape immunosurveillance of natural killer (NK) by gene mutation, fusions, and epigenetic modification, while the mechanism is not clearly understood. Here we show that the expression of Intercellular adhesion molecule-1 (ICAM-1, CD54) is silenced in AML cells. Decitabine could upregulate ICAM-1 expression, which contributes to the NK-AML cell conjugates and helps NK cells kill AML cells. We also show that ICAM-1 high expression can reverse the AML immune evasion and activate NK cells function in vivo. This study suggests that a combination of the hypomethylating agent and NK cell infusion could be a new strategy to cure AML.

9.
Front Immunol ; 12: 667478, 2021.
Article in English | MEDLINE | ID: mdl-34025669

ABSTRACT

Viral encephalitis is the most common cause of encephalitis. It is responsible for high morbidity rates, permanent neurological sequelae, and even high mortality rates. The host immune response plays a critical role in preventing or clearing invading pathogens, especially when effective antiviral treatment is lacking. However, due to blockade of the blood-brain barrier, it remains unclear how peripheral immune cells contribute to the fight against intracerebral viruses. Here, we report that peripheral injection of an antibody against human Tim-3, an immune checkpoint inhibitor widely expressed on immune cells, markedly attenuated vesicular stomatitis virus (VSV) encephalitis, marked by decreased mortality and improved neuroethology in mice. Peripheral injection of Tim-3 antibody enhanced the recruitment of immune cells to the brain, increased the expression of major histocompatibility complex-I (MHC-I) on macrophages, and as a result, promoted the activation of VSV-specific CD8+ T cells. Depletion of macrophages abolished the peripheral injection-mediated protection against VSV encephalitis. Notably, for the first time, we found a novel post-translational modification of MHC-I by Tim-3, wherein, by enhancing the expression of MARCH9, Tim-3 promoted the proteasome-dependent degradation of MHC-I via K48-linked ubiquitination in macrophages. These results provide insights into the immune response against intracranial infections; thus, manipulating the peripheral immune cells with Tim-3 antibody to fight viruses in the brain may have potential applications for combating viral encephalitis.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antibodies, Neutralizing/administration & dosage , Antigen-Presenting Cells/drug effects , Brain/drug effects , Encephalitis, Viral/prevention & control , Hepatitis A Virus Cellular Receptor 2/antagonists & inhibitors , Macrophages/drug effects , Rhabdoviridae Infections/prevention & control , Vesiculovirus/immunology , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Antigen-Presenting Cells/virology , Brain/immunology , Brain/metabolism , Brain/virology , Chlorocebus aethiops , Disease Models, Animal , Encephalitis, Viral/immunology , Encephalitis, Viral/metabolism , Encephalitis, Viral/virology , HEK293 Cells , Hepatitis A Virus Cellular Receptor 2/immunology , Histocompatibility Antigens Class I/metabolism , Host-Pathogen Interactions , Humans , Injections, Intraperitoneal , Macrophages/immunology , Macrophages/metabolism , Macrophages/virology , Male , Mice , Mice, Inbred C57BL , Proteasome Endopeptidase Complex/metabolism , Proteolysis , RAW 264.7 Cells , Rhabdoviridae Infections/immunology , Rhabdoviridae Infections/metabolism , Rhabdoviridae Infections/virology , Ubiquitination , Vero Cells , Vesiculovirus/pathogenicity , Viral Load
10.
Front Immunol ; 12: 618710, 2021.
Article in English | MEDLINE | ID: mdl-33868234

ABSTRACT

T cell immunoglobulin and mucin protein 3 (Tim-3) is an immune checkpoint and plays a vital role in immune responses during acute myeloid leukemia (AML). Targeting Tim-3 kills two birds with one stone by balancing the immune system and eliminating leukemia stem cells (LSCs) in AML. These functions make Tim-3 a potential target for curing AML. This review mainly discusses the roles of Tim-3 in the immune system in AML and as an AML LSC marker, which sheds new light on the role of Tim-3 in AML immunotherapy.


Subject(s)
Disease Susceptibility , Hepatitis A Virus Cellular Receptor 2/genetics , Hepatitis A Virus Cellular Receptor 2/metabolism , Leukemia, Myeloid, Acute/etiology , Leukemia, Myeloid, Acute/metabolism , Animals , Biomarkers , Disease Management , Gene Expression Regulation, Leukemic , Humans , Immune System/immunology , Immune System/metabolism , Immunomodulation/genetics , Immunomodulation/immunology , Leukemia, Myeloid, Acute/pathology , Leukemia, Myeloid, Acute/therapy , Ligands , Molecular Targeted Therapy , Neoplastic Stem Cells/immunology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Protein Binding , Signal Transduction
11.
Front Immunol ; 12: 770402, 2021.
Article in English | MEDLINE | ID: mdl-35095844

ABSTRACT

Tim-3, an immune checkpoint inhibitor, is widely expressed on the immune cells and contributes to immune tolerance. However, the mechanisms by which Tim-3 induces immune tolerance remain to be determined. Major histocompatibility complex II (MHC-II) plays a key role in antigen presentation and CD4+T cell activation. Dysregulated expressions of Tim-3 and MHC-II are associated with the pathogenesis of many autoimmune diseases including multiple sclerosis. Here we demonstrated that, by suppressing MHC-II expression in macrophages via the STAT1/CIITA pathway, Tim-3 inhibits MHC-II-mediated autoantigen presentation and CD4+T cell activation. As a result, overexpression or blockade of Tim-3 signaling in mice with experimental autoimmune encephalomyelitis (EAE) inhibited or increased MHC-II expression respectively and finally altered clinical outcomes. We thus identified a new mechanism by which Tim-3 induces immune tolerance in vivo and regulating the Tim-3-MHC-II signaling pathway is expected to provide a new solution for multiple sclerosis treatment.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Hepatitis A Virus Cellular Receptor 2/immunology , Nuclear Proteins/immunology , Trans-Activators/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , Cell Line , HEK293 Cells , Humans , Immune Tolerance/immunology , Lymphocyte Activation/immunology , Macrophages/immunology , Mice , Mice, Inbred C57BL , RAW 264.7 Cells , Signal Transduction/immunology
12.
Scand J Immunol ; 93(2): e12981, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33031600

ABSTRACT

T cell immunoglobulin and mucin domain-3 (Tim-3), an immune checkpoint molecule, plays critical roles in maintaining innate immune homeostasis; however, the mechanisms underlying these roles remain to be determined. Here, we determined that Tim-3 controls glycolysis in macrophages and thus contributes to phenotype shifting. Tim-3 signal blockade significantly increases lactate production by macrophages, but does not influence cell proliferation or apoptosis. Tim-3 attenuates glucose uptake by inhibiting hexokinase 2 (HK2) expression in macrophages. Tim-3-mediated inhibition of macrophage glycolysis and the expression of proinflammatory cytokines, tumour necrosis factor (TNF)-α and interleukin (IL)-1ß are reversed by HK2 silencing. Finally, we demonstrated that Tim-3 inhibits HK2 expression via the STAT1 pathway. We have thus discovered a new way by which Tim-3 modulates macrophage function.


Subject(s)
Glycolysis/immunology , Hepatitis A Virus Cellular Receptor 2/immunology , Hexokinase/immunology , Macrophages/immunology , Signal Transduction/immunology , T-Lymphocytes/immunology , Animals , Apoptosis/immunology , Cell Line , Cell Proliferation/physiology , Cytokines/immunology , HEK293 Cells , Humans , Immunity, Innate/immunology , Inflammation/immunology , Interleukin-1beta/immunology , Male , Mice , Mice, Inbred C57BL , RAW 264.7 Cells , Tumor Necrosis Factor-alpha/immunology
13.
Leuk Lymphoma ; 62(4): 937-943, 2021 04.
Article in English | MEDLINE | ID: mdl-33225787

ABSTRACT

The t(8;21)(q22;q22) translocation is the most common chromosomal translocation in acute myeloid leukemia (AML), and it gives rise to acute myeloid gene 1 (AML1)-myeloid transforming gene 8 (ETO)-positive AML, which has a relatively favorable prognosis. CD48 is a favorable prognosis factor that is downregulated in AML patients. AML can escape immunosurveillance of natural killer (NK) cells by decreasing CD48 expression. The correlation between AML1-ETO and CD48-mediated immune evasion is not well understood. Here, we show that AML1-ETO can increase CD48 expression, which is regulated by AML1-ETO/P300-mediated acetylation. AML1-ETO can inhibit AML immune escape from NK cell recognition and killing by increasing CD48 expression. This study describes a novel mechanism by which AML1-ETO can inhibit AML immune escape by increasing CD48 acetylation, thereby providing new evidence about AML patients with AML1-ETO oncogene infusion having better clinical outcomes.


Subject(s)
Core Binding Factor Alpha 2 Subunit , Leukemia, Myeloid, Acute , Chromosomes, Human, Pair 21 , Chromosomes, Human, Pair 8 , Core Binding Factor Alpha 2 Subunit/genetics , Humans , Leukemia, Myeloid, Acute/genetics , Oncogene Proteins, Fusion/genetics , RUNX1 Translocation Partner 1 Protein/genetics , Translocation, Genetic
14.
Biosci Rep ; 40(11)2020 11 27.
Article in English | MEDLINE | ID: mdl-33185693

ABSTRACT

Cancer immunotherapy has recently undergone rapid development into a validated therapy for clinical use. The adoptive transfer of engineered autologous T cells, such as chimeric antigen receptor (CAR) T cells, has been remarkably successful in patients with leukemia and lymphoma with cluster of differentiation (CD)19 expression. Because of the higher number of antigen choices and reduced incidence of cytokine release syndrome (CRS) than CAR-T cells, T cell receptor (TCR)-T cells are also considered a promising immunotherapy. More therapeutic targets for other cancers need to be explored due to the human leukocyte antigen (HLA)-restricted recognition of TCR-T. Major histocompatibility complex (MHC), class I-related (MR1)-restricted T cells can recognize metabolites presented by MR1 in the context of host cells infected with pathogens. MR1 is expressed by all types of human cells. Recent studies have shown that one clone of a MR1-restricted T (MR1-T) cell can recognize many types of cancer cells without HLA-restriction. These studies provide additional information on MR1-T cells for cancer immunotherapy. This review describes the complexity of MR1-T cell TCR in diseases and the future of cancer immunotherapy.


Subject(s)
Histocompatibility Antigens Class I/immunology , Immunotherapy, Adoptive , Lymphocytes, Tumor-Infiltrating/immunology , Minor Histocompatibility Antigens/immunology , Mucosal-Associated Invariant T Cells/immunology , Neoplasms/therapy , T-Lymphocytes/transplantation , Animals , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/metabolism , Humans , Lymphocytes, Tumor-Infiltrating/metabolism , Minor Histocompatibility Antigens/genetics , Minor Histocompatibility Antigens/metabolism , Mucosal-Associated Invariant T Cells/metabolism , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/metabolism , Phenotype , Prognosis , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
15.
Sci Rep ; 10(1): 17691, 2020 10 19.
Article in English | MEDLINE | ID: mdl-33077816

ABSTRACT

Lymphocyte apoptosis appears to play an important role in immunodysfunction in sepsis. We investigated the role of miR-223 in cell proliferation and apoptosis to identify potential target downstream proteins in sepsis. We recruited 143 patients with sepsis and 44 healthy controls from the Chinese PLA General Hospital. Flow cytometry was used to sort monocytes, lymphocytes, and neutrophils from fresh peripheral blood. A miR-223 mimic and inhibitor were used for transient transfection of Jurkat T cells. Quantitative reverse transcriptase-polymerase chain reaction (qRT-PCR) was used to assess expression of the miRNAs in cells. Western blot analysis was performed to measure protein expression. We evaluated the cell cycle and apoptosis by using flow cytometry (FCM) and terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL). Expression of miR-223 was significantly higher in the survivor group than in the nonsurvivor group. Multiple linear regression analysis revealed that SOFA scores correlated negatively with miR-223 and monocyte counts, with ß coefficients (95% CI) of - 0.048 (- 0.077, - 0.019) and - 47.707 (- 83.871, - 11.543), respectively. miR-223 expression also correlated negatively with the percentage of apoptosis in lymphocytes. The rate of apoptosis in the miR-223 mimic group was significantly lower than that of the negative control, with an adverse outcome observed in the miR-223 inhibitor group. We also found that miR-223 enhanced the proliferation of Jurkat T cells and that inhibiting miR-223 had an inhibitory effect on the G1/S transition. We conclude that miR-223 can serve as a protective factor in sepsis by reducing apoptosis and enhancing cell proliferation in lymphocytes by interacting with FOXO1. Potential downstream molecules are HSP60, HSP70, and HTRA.


Subject(s)
MicroRNAs/physiology , Sepsis/genetics , Sepsis/mortality , Adult , Aged , Aged, 80 and over , Apoptosis/physiology , Case-Control Studies , Cell Cycle , Female , Humans , Lymphocytes/cytology , Lymphocytes/immunology , Male , Middle Aged , Sepsis/immunology
16.
Clin Sci (Lond) ; 134(2): 261-271, 2020 Jan 31.
Article in English | MEDLINE | ID: mdl-31922199

ABSTRACT

Acute myeloid leukemia (AML) is a malignant disorder of hemopoietic stem cells. AML can escape immunosurveillance of natural killer (NK) by gene mutation, fusions and epigenetic modification. The mechanism of AML immune evasion is not clearly understood. Here we show that CD48 high expression is a favorable prognosis factor that is down-regulated in AML patients, which can help AML evade from NK cell recognition and killing. Furthermore, we demonstrate that CD48 expression is regulated by methylation and that a hypomethylating agent can increase the CD48 expression, which increases the NK cells killing in vitro. Finally, we show that CD48 high expression can reverse the AML immune evasion and activate NK cells function in vivo. The present study suggests that a combination the hypomethylating agent and NK cell infusion could be a new strategy to cure AML.


Subject(s)
CD48 Antigen/immunology , Epigenesis, Genetic/immunology , Gene Silencing/immunology , Leukemia, Myeloid/immunology , Tumor Escape/immunology , Acute Disease , Animals , Antimetabolites, Antineoplastic/pharmacology , CD48 Antigen/genetics , Cell Line, Tumor , Cells, Cultured , DNA Methylation/drug effects , DNA Methylation/genetics , DNA Methylation/immunology , Decitabine/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Humans , Kaplan-Meier Estimate , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Leukemia, Myeloid/drug therapy , Leukemia, Myeloid/genetics , Male , Mice, Inbred BALB C , Tumor Escape/genetics , Xenograft Model Antitumor Assays
17.
J Infect Dis ; 221(5): 830-840, 2020 02 18.
Article in English | MEDLINE | ID: mdl-31586389

ABSTRACT

BACKGROUND: T-cell immunoglobulin and mucin protein 3 (Tim-3) is an immune checkpoint inhibitor that has therapeutic implications for many tumors and infectious diseases. However, the mechanisms by which Tim-3 promotes immune evasion remain unclear. METHODS: In this study, we demonstrated that Tim-3 inhibits the expression of major histocompatibility complex class I (MHC-I) in macrophages at both the messenger ribonucleic acid and protein levels by inhibiting the STAT1-NLRC5 signaling pathway. RESULTS: As a result, MHC-I-restricted antigen presentation by macrophages was inhibited by Tim-3 both in vitro and in a Listeria monocytogenes infection model in vivo. Systemic overexpression of Tim-3 or specific knockout of Tim-3 in macrophages significantly attenuated or enhanced CD8+ T-cell activation and infection damage in L monocytogenes-infected mice, respectively. CONCLUSIONS: Thus, we identified a new mechanism by which Tim-3 promotes L monocytogenes immune evasion. Further studies on this pathway might shed new light on the physio-pathological roles of Tim-3 and suggest new approaches for intervention.


Subject(s)
HLA-A Antigens/metabolism , Hepatitis A Virus Cellular Receptor 2/metabolism , Immune Evasion/immunology , Listeria monocytogenes/immunology , Listeriosis/immunology , Macrophages/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , HEK293 Cells , Hepatitis A Virus Cellular Receptor 2/genetics , Humans , Listeriosis/microbiology , Lymphocyte Activation/genetics , Mice , Mice, Transgenic , RAW 264.7 Cells , Transfection
18.
Sci Rep ; 9(1): 421, 2019 01 23.
Article in English | MEDLINE | ID: mdl-30674954

ABSTRACT

The E3 ubiquitin ligase Itch interacts with Foxo1 and targets it for ubiquitination and degradation during follicular helper T-cell differentiation, whereas the transcription factor Foxo1 plays a critical role in B-cell development. Thus, we proposed that Itch mediates B-cell differentiation. Unexpectedly, we found that Itch deficiency downregulated Foxo1 expression in B cells. Itch cKO (conditional knock out in B cells) mice had fewer pro-B cells in the bone marrow, more small resting IgM-IgD-B cells in the periphery, and lower B-cell numbers in the lymph nodes through decreased Foxo1-mediated IL-7Rα, RAG, and CD62L expression, respectively. Importantly, Itch deficiency reduced Foxo1 mRNA expression by up-regulating JunB-mediated miR-182. Finally, Foxo1 negatively regulated JunB expression by up-regulating Itch. Thus, we have identified a novel regulatory axis between Itch and Foxo1 in B cells, suggesting that Itch is essential for B-cell development.


Subject(s)
B-Lymphocytes/immunology , Bone Marrow Cells/immunology , Cell Differentiation/immunology , Ubiquitin-Protein Ligases/immunology , Animals , B-Lymphocytes/cytology , Bone Marrow Cells/cytology , Cell Differentiation/genetics , Forkhead Box Protein O1/genetics , Forkhead Box Protein O1/immunology , L-Selectin/genetics , L-Selectin/immunology , Mice , Mice, Knockout , MicroRNAs/genetics , MicroRNAs/immunology , Receptors, Interleukin-7/genetics , Receptors, Interleukin-7/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Transcription Factors/genetics , Transcription Factors/immunology , Ubiquitin-Protein Ligases/genetics
19.
Scand J Immunol ; 89(2): e12738, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30506563

ABSTRACT

T cell immunoglobulin and mucin domain protein 3 (Tim-3) is an immune checkpoint inhibitor in T cells and innate immune cells. The deregulated upregulation of Tim-3 is related to immune exhaustion in tumour and viral infection. To overcome Tim-3-mediated immune tolerance, we developed a novel monoclonal antibody against human Tim-3 (L3G) and investigated its roles in inhibiting Tim-3 signalling and overcoming immune tolerance in T cells and monocytes/macrophages. The administration of L3G to cultured peripheral blood mononuclear cells (PBMCs) significantly increased the production of IFN-γ and IL-2 and the expression of type I interferon. The administration of L3G also increased the production of IFN-γ, IL-8 and type I interferon in U937 cells and primary monocytes. We investigated the mechanisms by which L3G enhances pro-inflammatory cytokine expression, and our data show that L3G enhances STAT1 phosphorylation in both monocytes/macrophages and T cells. Finally, in an H1N1 infection model of PBMCs and U937 cells, L3G decreased the viral load and enhanced the expression of interferon. Thus, we developed a functional antibody with therapeutic potential against Tim-3-mediated infection tolerance.


Subject(s)
Antibodies, Monoclonal/metabolism , Hepatitis A Virus Cellular Receptor 2/immunology , Influenza A Virus, H1N1 Subtype/physiology , Influenza, Human/immunology , Macrophages/immunology , Orthomyxoviridae Infections/immunology , T-Lymphocytes/immunology , Animals , Cytokines/metabolism , Disease Models, Animal , Female , Humans , Inflammation Mediators/metabolism , Lymphocyte Activation , Macrophages/virology , Mice , Mice, Inbred BALB C , STAT1 Transcription Factor/metabolism , Signal Transduction , U937 Cells , Viral Load
20.
Mol Med Rep ; 17(1): 1660-1666, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29138852

ABSTRACT

The interleukin (IL)­12 family cytokines have been examined as therapeutic targets in the treatment of several autoimmune diseases. Our previous study showed that a novel IL­12 family cytokine, IL­39 (IL­23p19/Ebi3) mediates inflammation in lupus­like mice. In the present study, the effect of anti­mouse IL­39 polyclonal antibodies on autoimmune symptoms in lupus­like mice was investigated. Rabbit anti­mouse IL­39 polyclonal antibodies were produced by immunization with recombinant mouse IL­39, and purified using protein A chromatography. These antibodies were subsequently used to treat lupus­like mice. Flow cytometry, captured images, ELISA and H&E staining were used to determine the effect of anti­IL­39 polyclonal antibodies on inflammatory cells, autoantibody titers, proteinuria, infiltrating inflammatory cells and the structure of the glomerular region. The anti­IL­39 polyclonal antibodies effectively reduced the numbers of inflammatory cells, splenomegaly, autoantibody titers, proteinuria, infiltrating inflammatory cells, and restored the structure of the glomerular region in MRL/lpr mice. Taken together, these results suggested that anti­IL­39 polyclonal antibodies ameliorated autoimmune symptoms in lupus­like mice. Therefore, IL­39 may be used as a possible target for the treatment of systemic lupus erythematosus.


Subject(s)
Antibodies/pharmacology , Immunologic Factors/pharmacology , Lupus Erythematosus, Systemic/drug therapy , Animals , Antibodies/therapeutic use , Autoantibodies/blood , Drug Evaluation, Preclinical , Female , Glomerulonephritis/etiology , Glomerulonephritis/immunology , Glomerulonephritis/prevention & control , Immunologic Factors/therapeutic use , Interleukin-23 Subunit p19/antagonists & inhibitors , Interleukin-23 Subunit p19/immunology , Kidney/drug effects , Kidney/pathology , Lupus Erythematosus, Systemic/blood , Lupus Erythematosus, Systemic/complications , Lupus Erythematosus, Systemic/immunology , Mice, Transgenic , Minor Histocompatibility Antigens/immunology , Proteinuria/etiology , Proteinuria/immunology , Proteinuria/prevention & control , Rabbits , Receptors, Cytokine/antagonists & inhibitors , Receptors, Cytokine/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...