Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 272
Filter
Add more filters

Country/Region as subject
Affiliation country
Publication year range
1.
Biochem Biophys Res Commun ; 691: 149326, 2024 Jan 08.
Article in English | MEDLINE | ID: mdl-38035406

ABSTRACT

Sleep deprivation (SD) weakens the immune system and leads to increased susceptibility to infectious or inflammatory diseases. However, it is still unclear how SD affects humoral immunity. In the present study, sleep disturbance was conducted using an sleep deprivation instrument, and the bacterial endotoxin lipopolysaccharide (LPS) was used to activate the immune response. It was found that SD-pretreatment reduced LPS-induced IgG2b+ B cells and IgG2b isotype antibody production in lymphocytes of spleen. And, SD-pretreatment decreased the proportion of CD4+T cells, production of CD4+T cells derived TGF-ß1 and its contribution in helping IgG2b production. Additionally, BMAL1 and CLOCK were selectively up-regulated in lymphocytes after SD. Importantly, BMAL1 and CLOCK deficiency contributed to TGF-ß1 expression and production of IgG2b+ B cells. Thus, our results provide a novel insight to explain the involvement of BMAL1 and CLOCK under SD stress condition, and their roles in inhibiting TGF-ß1 expression and contributing to reduction of LPS induced IgG2b production.


Subject(s)
ARNTL Transcription Factors , Antibody Formation , CLOCK Proteins , Immunoglobulin G , Sleep Deprivation , Sleep Deprivation/genetics , Sleep Deprivation/immunology , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Rats, Sprague-Dawley , Mice, Inbred C57BL , ARNTL Transcription Factors/genetics , ARNTL Transcription Factors/immunology , CLOCK Proteins/genetics , CLOCK Proteins/immunology , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Antibody Formation/drug effects , Antibody Formation/genetics , Stress, Physiological/immunology , Animals , Mice , Rats , Cells, Cultured
2.
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
3.
Proc Natl Acad Sci U S A ; 117(5): 2473-2483, 2020 02 04.
Article in English | MEDLINE | ID: mdl-31941714

ABSTRACT

Neddylation is a ubiquitination-like pathway that controls cell survival and proliferation by covalently conjugating NEDD8 to lysines in specific substrate proteins. However, the physiological role of neddylation in mammalian metabolism remains elusive, and no mitochondrial targets have been identified. Here, we report that mouse models with liver-specific deficiency of NEDD8 or ubiquitin-like modifier activating enzyme 3 (UBA3), the catalytic subunit of the NEDD8-activating enzyme, exhibit neonatal death with spontaneous fatty liver as well as hepatic cellular senescence. In particular, liver-specific UBA3 deficiency leads to systemic abnormalities similar to glutaric aciduria type II (GA-II), a rare autosomal recessive inherited fatty acid oxidation disorder resulting from defects in mitochondrial electron transfer flavoproteins (ETFs: ETFA and ETFB) or the corresponding ubiquinone oxidoreductase. Neddylation inhibition by various strategies results in decreased protein levels of ETFs in neonatal livers and embryonic hepatocytes. Hepatic neddylation also enhances ETF expression in adult mice and prevents fasting-induced steatosis and mortality. Interestingly, neddylation is active in hepatic mitochondria. ETFs are neddylation substrates, and neddylation stabilizes ETFs by inhibiting their ubiquitination and degradation. Moreover, certain mutations of ETFs found in GA-II patients hinder the neddylation of these substrates. Taken together, our results reveal substrates for neddylation and add insight into GA-II.


Subject(s)
Electron-Transferring Flavoproteins/metabolism , Fatty Acids/metabolism , Liver/metabolism , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/metabolism , Animals , Electron-Transferring Flavoproteins/genetics , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/genetics , NEDD8 Protein/genetics , NEDD8 Protein/metabolism , Oxidation-Reduction , Ubiquitination , Ubiquitins/genetics , Ubiquitins/metabolism
4.
Mol Pharmacol ; 102(3): 161-171, 2022 09.
Article in English | MEDLINE | ID: mdl-35764384

ABSTRACT

Sialic acid-binding Ig-like lectin-15 is an important immunosuppressive molecule considered to be a key target in next-generation tumor immunotherapy. In this study, we screened 22 high-affinity antibodies that specifically recognize human Siglec-15 by using a large human phage antibody library, and five representative sequences were selected for further study. The results showed the binding activity of five antibodies to Siglec-15 (EC50 ranged from 0.02368 µg/mL to 0.07949 µg/mL), and in two Siglec-15-overexpressed cell lines, three antibodies had the strongest binding activity, so the two clones were discarded for further study. Subsequently, the affinity of three antibodies were measured by bio-layer interferometry technology (5-9 × 10E-09M). As the reported ligands of Siglec-15, the binding activity of Siglec-15 and sialyl-Tn, cluster of differentiation 44, myelin-associated glycoprotein, and leucine-rich repeat-containing protein 4C can be blocked by three of the antibodies. Among these, 3F1 had a competitive advantage. Then, the antibody 3F1 showed an obvious antibody-dependent cell-mediated cytotoxicity effect (EC50 was 0.85 µg/mL). Further, antibody 3F1 can reverse the inhibitory effect of Siglec-15 on lymphocyte proliferation (especially CD4+T and CD8+T) and cytokine release Interferon-γ. Given the above results, 3F1 was selected as a candidate for the in vivo pharmacodynamics study. In the tumor model of Balb/c Nude mice, 3F1 (10 mg/kg) showed certain antitumor effects [tumor growth inhibition (TGI) was 31.5%], while the combination of 3F1 (5 mg/kg) and Erbitux (5 mg/kg) showed significant antitumor effects (TGI was 48.7%) compared with the PBS group. In conclusion, novel human antibody 3F1 has antitumor activity and is expected to be an innovative candidate drug targeting Siglec-15 for tumor immunotherapy. SIGNIFICANCE STATEMENT: Siglec-15 is considered as an important target in the next generation of tumor immunotherapy. 3F1 is expected to be the most promising potential candidate for targeting Siglec-15 for cancer treatment and could provide a reference for the development of antitumor drugs.


Subject(s)
Antigens, CD , Neoplasms , Animals , Antigens, CD/metabolism , Humans , Immunoglobulins , Lectins/chemistry , Lectins/metabolism , Ligands , Membrane Proteins , Mice , Mice, Nude , Neoplasms/drug therapy
5.
Biotechnol Lett ; 44(9): 1063-1072, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35918621

ABSTRACT

AIM: To investigate the impact of deficiency of LIG4 gene on site-specific integration in CHO cells. RESULTS: CHO cells are considered the most valuable mammalian cells in the manufacture of biological medicines, and genetic engineering of CHO cells can improve product yield and stability. The traditional method of inserting foreign genes by random integration (RI) requires multiple rounds of screening and selection, which may lead to location effects and gene silencing, making it difficult to obtain stable, high-yielding cell lines. Although site-specific integration (SSI) techniques may overcome the challenges with RI, its feasibility is limited by the very low efficiency of the technique. Recently, SSI efficiency has been enhanced in other mammalian cell types by inhibiting DNA ligase IV (Lig4) activity, which is indispensable in DNA double-strand break repair by NHEJ. However, this approach has not been evaluated in CHO cells. In this study, the LIG4 gene was knocked out of CHO cells using CRISPR/Cas9-mediated genome editing. Efficiency of gene targeting in LIG4-/--CHO cell lines was estimated by a green fluorescence protein promoterless reporter system. Notably, the RI efficiency, most likely mediated by NHEJ in CHO, was inhibited by LIG4 knockout, whereas SSI efficiency strongly increased 9.2-fold under the precise control of the promoter in the ROSA26 site in LIG4-/--CHO cells. Moreover, deletion of LIG4 had no obvious side effects on CHO cell proliferation. CONCLUSIONS: Deficiency of LIG4 represents a feasible strategy to improve SSI efficiency and suggests it can be applied to develop and engineer CHO cell lines in the future.


Subject(s)
CRISPR-Cas Systems , Gene Editing , Animals , CHO Cells , CRISPR-Cas Systems/genetics , Cricetinae , Cricetulus , DNA End-Joining Repair/genetics , DNA Ligase ATP/genetics
6.
Mol Pharmacol ; 100(3): 193-202, 2021 09.
Article in English | MEDLINE | ID: mdl-34315811

ABSTRACT

Phagocytic resistance plays a key role in tumor-mediated immune escape, so phagocytosis immune checkpoints are a potential target for cancer immunotherapy. CD47 is one of the important phagocytosis immune checkpoints; thus, blocking the interaction between CD47 and signal regulatory protein α (SIRPα) may provide new options for cancer treatment. Using computer-aided targeted epitope mammalian cell-displayed antibody library, we screened and obtained an engineered SIRPα variant fragment crystallizable fusion protein, FD164, with higher CD47-binding activity than wild-type SIRPα Compared with wild-type SIRPα, FD164 has approximately 3-fold higher affinity for binding to CD47, which further enhanced its phagocytic effect in vitro and tumor suppressor activity in vivo. FD164 maintains the similar antitumor activity of the clinical research drug Hu5F9 in the mouse xenograft model. Furthermore, FD164 combined with rituximab can significantly improve the effect of single-agent therapy. On the other hand, compared with Hu5F9, FD164 does not cause hemagglutination, and its ability to bind to red blood cells or white blood cells is weaker at the same concentration. Finally, it was confirmed by computer structure prediction and alanine scanning experiments that the N45, E47, 52TEVYVK58, K60, 115EVTELTRE122, and E124 residues of CD47 are important for SIRPα or FD164 recognition. Briefly, we obtained a high-affinity SIRPα variant FD164 with balanced safety and effectiveness. SIGNIFICANCE STATEMENT: Up to now, few clinically marketed drugs targeting CD47 have been determined to be effective and safe. FD164, a potential signal regulatory protein α variant fragment crystallizable protein with balanced safety and effectiveness, could provide a reference for the development of antitumor drugs.


Subject(s)
Antigens, Differentiation/genetics , Antigens, Differentiation/immunology , CD47 Antigen/immunology , Receptors, Immunologic/genetics , Receptors, Immunologic/immunology , Animals , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/therapeutic use , Antigens, Differentiation/adverse effects , Antigens, Differentiation/chemistry , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Burkitt Lymphoma/drug therapy , Burkitt Lymphoma/immunology , Burkitt Lymphoma/pathology , CD47 Antigen/chemistry , CHO Cells , Cell Line , Cricetulus , Drug Design , Epitopes/chemistry , Epitopes/genetics , Epitopes/immunology , Hemagglutination/drug effects , Immunotherapy , Mice, SCID , Models, Molecular , Phagocytosis/drug effects , Phagocytosis/immunology , Receptors, Immunologic/chemistry , Recombinant Fusion Proteins/adverse effects , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Rituximab/therapeutic use , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
7.
Biochem Biophys Res Commun ; 559: 217-221, 2021 06 25.
Article in English | MEDLINE | ID: mdl-33957483

ABSTRACT

Ornithine transcarbamylases (OTC), a key enzyme in urea cycle, is an important marker for some liver injury or diseases. However, whether OTC could be a sensitive indicator for liver dysfunction under sleep disturbance condition remains unknown. The present study aimed to explore the circadian oscillation expression of OTC and its significance in disturbed sleep condition. Sleep disturbance was conducted by a sleep deprivation (SD) instrument. Our results found that SD for 72h induced abnormal increasing of OTC levels in serum and liver of rats. And, serum OTC concentration and liver OTC expression could return to normal levels after recovery sleep following SD. Moreover, hepatic OTC expression showed circadian oscillation in day and night, characterized with occurrence of a peak between ZT 22 and ZT 2, and a nadir between ZT 14 and ZT 18. Further analysis suggested the existence of ROR response element (RORE) for potential RORɑ binding sites in OTC promoter region, and elevated RORɑ expression in rat livers under sleep disturbance condition. Additionally, oscillation expression of OTC induced by serum shock in HepG2 cells was characterized with a peak occurred between ZT 12 and ZT 16, and RORɑ knockdown at ZT 16 significantly lowered OTC expression. The results together indicate that OTC is closely correlated with circadian clock, and could be a sensitive indicator for sleep disturbance stress.


Subject(s)
Circadian Rhythm , Ornithine Carbamoyltransferase/metabolism , Sleep Wake Disorders/enzymology , Sleep Wake Disorders/physiopathology , Animals , Base Sequence , Gene Expression Regulation, Enzymologic , Hep G2 Cells , Homeostasis , Humans , Liver/enzymology , Male , Nuclear Receptor Subfamily 1, Group F, Member 1/metabolism , Ornithine Carbamoyltransferase/genetics , Rats, Sprague-Dawley , Sleep/genetics , Sleep Wake Disorders/genetics
8.
Biochem Biophys Res Commun ; 549: 120-127, 2021 04 16.
Article in English | MEDLINE | ID: mdl-33667709

ABSTRACT

Staphylococcal enterotoxin B (SEB), one of the exotoxins produced by Staphylococcus aureus, is the key toxin that causes poisoning reactions and toxic shock syndrome. In the current research work, a novel human antibody named LXY8 was screened from a human phage display antibody library, and LXY8 blocked the interaction between SEB and the T cell receptor (TCR). The binding activity between LXY8 and SEB was 0.525 nM. Furthermore, LXY8 could effectively inhibit the SEB-induced activation of peripheral blood mononuclear cells and release of cytokines. In the BALB/c mouse model, LXY8 effectively neutralized SEB toxicity in vivo. Finally, based on computer-guided molecular modeling, we designed a series of SEB mutation sites; these sites facilitated the determination of the key residues (i.e.176EFNN179) of SEB recognized by LXY8. The research revealed that the 176EFNN179 residues of SEB are important for specific antibody-antigen recognition. The results may be helpful for the development of antibody-based therapy for SEB-induced toxic shock syndrome.


Subject(s)
Antibodies, Bacterial/analysis , Antibodies, Monoclonal/analysis , Antibodies, Neutralizing/analysis , Enterotoxins/immunology , Epitopes/immunology , Animals , CHO Cells , Cell Proliferation , Cell Surface Display Techniques , Cricetulus , Cytokines/metabolism , Enterotoxins/antagonists & inhibitors , Epitope Mapping , Female , Histocompatibility Antigens Class II/metabolism , Humans , Mice, Inbred BALB C , Protein Binding , Receptors, Antigen, T-Cell/metabolism
9.
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
10.
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
11.
Scand J Immunol ; 90(2): e12777, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31075180

ABSTRACT

TAM family members (TYRO3, AXL and MERTK) play essential roles in the resolution of inflammation and in infectious diseases and cancer. AXL, a tyrosine kinase receptor, is commonly overexpressed in several solid tumours and numerous hematopoietic malignancies including acute myeloid leukaemia, acute lymphocytic leukaemia, chronic myeloid leukaemia, chronic lymphocytic leukaemia and multiple myeloma. AXL significantly promotes tumour cell migration, invasion and metastasis, as well as angiogenesis. AXL also plays an important role in inflammation and macrophage ontogeny. Recent studies have revealed that AXL contributes to leukaemic phenotypes through activation of oncogenic signalling pathways that lead to increased cell migration and proliferation. To evaluate the mechanisms underlying the role of AXL signalling in tumour metastasis, we screened a phage display library to generate a novel human monoclonal antibody, named DAXL-88, that recognizes both human and murine AXL. The concentrations of DAXL-88 required for 50% maximal binding to human and murine AXL were 0.118 and 0.164 µg/mL, respectively. Furthermore, DAXL-88 bound to human AXL with high affinity (KD  ~ 370 pM). DAXL-88 blocked the interaction between AXL and its ligand, growth arrest-specific gene 6 (GAS6), with a half maximal inhibitory concentration of 2.16 µg/mL. Moreover, DAXL-88 inhibited AXL/GAS6-dependent cell signalling, which is implicated in cell migration and invasion. In conclusion, the novel anti-AXL DAXL-88 high-affinity antibody blocks the interaction between AXL and GAS6 and inhibits tumour cell migration and invasion induced by GAS6. Thus, DAXL-88 offers promise for the development of targeted therapeutic strategies in solid tumours, leukaemias and other lymphoid neoplasms.


Subject(s)
Antibodies, Monoclonal/pharmacology , Intercellular Signaling Peptides and Proteins/metabolism , Ovarian Neoplasms/pathology , Proto-Oncogene Proteins/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , A549 Cells , Antibodies, Monoclonal/immunology , Cell Line, Tumor , Cell Movement , Female , Humans , Molecular Docking Simulation , Neoplasm Invasiveness/pathology , Neoplasm Invasiveness/prevention & control , Neoplasm Metastasis/prevention & control , Protein Binding , Proto-Oncogene Proteins/immunology , Receptor Protein-Tyrosine Kinases/immunology , Signal Transduction , Axl Receptor Tyrosine Kinase
12.
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
13.
Mol Cell Biochem ; 451(1-2): 155-163, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30019299

ABSTRACT

As a classic differentiation agent, all-trans retinoic acid (ATRA) has been widely used in the treatment of acute promyelocytic leukemia (APL). However, the clinical application of ATRA has strict limitations, for its severe side effects due to the accumulation of peripheral blood leukocytes. The scaffold protein RACK1 (Receptor for activated C kinase 1), which regulates multiple signaling pathways, has been proposed to contribute to the survival of leukemic progenitors. But it remains unclear whether it is also involved in the oncogenic growth of APL. In the present study, we demonstrate that silencing of endogenous RACK1 expression synergized with ATRA to promote the death of NB4 and HL-60 APL cells without effect on cell differentiation induced by ATRA. Interestingly, RACK1 knockdown combined with ATRA treatment mainly induces apoptosis. It is distinct to the necrotic cell death induced by idarubicin in combination with ATRA, a regimen extensively used in the clinic to prevent neutrophil accumulation. Further exploration revealed that the lysosome-autophagy pathway is likely to be responsible for the anti-apoptotic role of RACK1. Taken together, our findings indicate that RACK1 is essential in maintaining the malignant features of APL, and targeting RACK1 may have promising therapeutic implications in the treatment of APL.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Autophagy , Leukemia, Promyelocytic, Acute/pathology , Neoplasm Proteins/deficiency , Receptors for Activated C Kinase/deficiency , Tretinoin/pharmacology , Cell Differentiation , Humans , Leukemia, Promyelocytic, Acute/drug therapy , Leukemia, Promyelocytic, Acute/metabolism , Signal Transduction , Tumor Cells, Cultured
14.
Immunology ; 153(1): 71-83, 2018 01.
Article in English | MEDLINE | ID: mdl-28799242

ABSTRACT

The Nod-like receptor protein 3 (NLRP3) inflammasome plays roles in host defence against invading pathogens and in the development of autoimmune damage. Strict regulation of these responses is important to avoid detrimental effects. Here, we demonstrate that T cell Ig mucin-3 (Tim-3), an immune checkpoint inhibitor, inhibits NLRP3 inflammasome activation by damping basal and lipopolysaccharide-induced nuclear factor-κB-mediated up-regulation of NLRP3 and interleukin-1ß during the priming step and basal and ATP/lipopolysaccharide-induced ATP production, K+ efflux, and reactive oxygen species production during the activation step. Residues Y256/Y263 in the C-terminal region of Tim-3 are required for these inhibitory effects on the NLRP3 inflammasome. In mice with alum-induced peritonitis, blockade of Tim-3 exacerbates peritonitis by overcoming the inhibitory effect of Tim-3 on NLRP3 inflammasome activation, while transgenic expression of Tim-3 attenuates inflammation by inhibiting NLRP3 inflammasome activation. Our results show that Tim-3 is a critical negative regulator of NLRP3 inflammasome and provides a potential target for intervention of diseases with uncontrolled inflammasome activation.


Subject(s)
Hepatitis A Virus Cellular Receptor 2/metabolism , Inflammasomes/immunology , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Peritonitis/immunology , Peritonitis/metabolism , Adenosine Triphosphate/biosynthesis , Adult , Animals , Case-Control Studies , Caspase 1 , Cell Line , Disease Models, Animal , Female , Hepatitis A Virus Cellular Receptor 2/chemistry , Humans , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Middle Aged , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Peritonitis/pathology , Potassium/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Reactive Oxygen Species/metabolism , Signal Transduction , Young Adult
15.
Cell Immunol ; 328: 9-17, 2018 06.
Article in English | MEDLINE | ID: mdl-29499909

ABSTRACT

IL-1α in vitro promotes immunoglobulin secretion by inducing proliferation of mature B cells, whereas IL-1α deficiency has no effect on in vivo antibody production. However, the reason IL-1α deficiency does not reduce in vivo antibody production is still unclear. In this study, we found that similar as in vivo data, IL-1α deficiency did not affect antibody production in in vitro LPS-stimulated B cells. Surprisingly, LPS-stimulated IL-1α-/- B cells reduced a key antibody production-related transcription factor X-box binding protein 1 (Xbp-1) expression. Furthermore, we found that IL-1α deficiency up-regulated mTOR expression, which bypassed Xbp-1 for immunoglobulin secretion. Finally, we showed that Xbp-1 suppressed mTOR expression, whereas mTOR suppressed the activation of Xbp-1 promoter via JunB. Together, these data suggest that IL-1a deficiency reduced Xbp-1 and up-regulated mTOR. This may explain why IL-1α deficiency has no effect on antibody production.


Subject(s)
B-Lymphocytes/immunology , TOR Serine-Threonine Kinases/physiology , X-Box Binding Protein 1/metabolism , Animals , Antibody Formation , B-Lymphocytes/metabolism , B-Lymphocytes/physiology , Cell Differentiation/immunology , DNA-Binding Proteins/genetics , Female , Gene Expression Regulation/immunology , Interleukin-1alpha/immunology , Interleukin-1alpha/metabolism , Interleukin-1alpha/physiology , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred BALB C , Plasma Cells/immunology , Protein Transport , TOR Serine-Threonine Kinases/immunology , Transcription Factors/genetics , Transcriptional Activation , X-Box Binding Protein 1/genetics , X-Box Binding Protein 1/immunology
16.
J Cell Mol Med ; 21(12): 3658-3669, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28707394

ABSTRACT

As the first line of defence, marginal zone (MZ) B cells play principal roles in clearing blood-borne pathogens during infection and are over-primed in autoimmune diseases. However, the basic mechanisms underlying MZ B-cell development are still unclear. We found here that CD19 deficiency blocked the differentiation of marginal zone precursors (MZP) to MZ B cells, whereas CD19 expression in CD19-deficient MZP rescues MZ B-cell generation. Furthermore, CD19 regulates Notch2 cleavage by up-regulating ADAM28 expression in MZP. Finally, we found that CD19 suppressed Foxo1 expression to promote ADAM28 expression in MZP. These results suggest that CD19 controls the differentiation of MZP to MZ B cells by regulating ADAM28-mediated Notch2 cleavage. Thus, we demonstrated the basic mechanisms underlying the differentiation of MZP to MZ B cells.


Subject(s)
ADAM Proteins/genetics , Antigens, CD19/genetics , B-Lymphocytes/immunology , Lymphoid Tissue/immunology , Receptor, Notch2/genetics , ADAM Proteins/immunology , Animals , Antigens, CD19/immunology , B-Lymphocytes/cytology , Cell Differentiation/immunology , Forkhead Box Protein O1/genetics , Forkhead Box Protein O1/immunology , Gene Expression Regulation , Lymphoid Tissue/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout , Proteolysis , Receptor, Notch2/immunology , Signal Transduction
17.
Immunology ; 150(4): 478-488, 2017 04.
Article in English | MEDLINE | ID: mdl-27995618

ABSTRACT

Interleukin-10-positive (IL-10+ ) regulatory B (Breg) cells play an important role in restraining excessive inflammatory responses by secreting IL-10. However, it is still unclear what key transcription factors determine Breg cell differentiation. Hence, we explore what transcription factor plays a key role in the expression of IL-10, a pivotal cytokine in Breg cells. We used two types of web-based prediction software to predict transcription factors binding the IL-10 promoter and found that IL-10 promoter had many binding sites for Foxd3. Chromatin immunoprecipitation PCR assay demonstrated that Foxd3 directly binds the predicted binding sites around the start codon upstream by -1400 bp. Further, we found that Foxd3 suppressed the activation of IL-10 promoter by using an IL-10 promoter report system. Finally, knocking out Foxd3 effectively promotes Breg cell production by up-regulating IL-10 expression. Conversely, up-regulated Foxd3 expression was negatively associated with IL-10+ Breg cells in lupus-prone MRL/lpr mice. Hence, our data suggest that Foxd3 suppresses the production of IL-10+ Breg cells by directly binding the IL-10 promoter. This study demonstrates the mechanism for Breg cell production and its application to the treatment of autoimmune diseases by regulating Foxd3 expression.


Subject(s)
B-Lymphocytes, Regulatory/immunology , Forkhead Transcription Factors/metabolism , Interleukin-10/metabolism , Lupus Erythematosus, Systemic/immunology , Repressor Proteins/metabolism , Animals , Cell Differentiation/genetics , Cells, Cultured , Computational Biology , Disease Models, Animal , Female , Forkhead Transcription Factors/genetics , Gene Knockdown Techniques , Humans , Interleukin-10/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred MRL lpr , Promoter Regions, Genetic/genetics , Protein Binding , Repressor Proteins/genetics
18.
Eur J Immunol ; 46(6): 1343-50, 2016 06.
Article in English | MEDLINE | ID: mdl-27019190

ABSTRACT

Interleukin-12 family cytokines have emerged as critical regulators of immunity with some members (IL-12, IL-23) associated with disease pathogenesis while others (IL-27, IL-35) mitigate autoimmune diseases. Each IL-12 family member is comprised of an α and a ß chain, and chain-sharing is a key feature. Although four bona fide members have thus far been described, promiscuous chain-pairing between alpha (IL-23p19, IL-27p28, IL-12/IL-35p35) and beta (IL-12/IL-23p40, IL-27/IL-35Ebi3) subunits, predicts six possible heterodimeric IL-12 family cytokines. Here, we describe a new IL-12 member composed of IL-23p19 and Ebi3 heterodimer (IL-39) that is secreted by LPS-stimulated B cells and GL7(+) activated B cells of lupus-like mice. We further show that IL-39 mediates inflammatory responses through activation of STAT1/STAT3 in lupus-like mice. Taken together, our results show that IL-39 might contribute to immunopathogenic mechanisms of systemic lupus erythematosus, and could be used as a possible target for its treatment.


Subject(s)
Interleukin-23 Subunit p19/metabolism , Lupus Erythematosus, Systemic/etiology , Lupus Erythematosus, Systemic/metabolism , Minor Histocompatibility Antigens/metabolism , Receptors, Cytokine/metabolism , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Disease Models, Animal , Female , Gene Expression , Immunophenotyping , Inflammation/etiology , Inflammation/metabolism , Inflammation/pathology , Interleukin-23 Subunit p19/chemistry , Interleukin-23 Subunit p19/genetics , Lupus Erythematosus, Systemic/pathology , Lymphocyte Activation/immunology , Mice , Mice, Inbred MRL lpr , Minor Histocompatibility Antigens/chemistry , Minor Histocompatibility Antigens/genetics , Protein Multimerization , Receptors, Cytokine/chemistry , Receptors, Cytokine/genetics , Receptors, Interleukin/metabolism , STAT1 Transcription Factor/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction
19.
Biotechnol Lett ; 39(9): 1309-1323, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28560579

ABSTRACT

OBJECTIVES: To find a "me-better" antibody by epitope-specific antibody optimization and multi-parametric analysis. RESULTS: Using epitope-specific library based on the commercial drug, Pertuzumab/2C4, we screened a novel human anti-HER2 antibody, MIL5, which has slightly higher affinity than the drug. MIL5 and 2C4 share the same epitope to bind HER2; however, MIL5 bound to HER2 His235-His245 more tightly than 2C4, which could be the main reason of its enhanced affinity. In vivo experiments also showed MIL5 had stronger anti-cancer activity than 2C4; however, the classical flow cytometry assays to detect cell apoptosis or cycling did not show convincing evidence of the advantages of MIL5. Thus we introduced the multi-parameter in-cell analysis method to evaluate the superiority of MIL5 to 2C4 in arresting cancer cells in G2-phase to inhibit cell growth and/or proliferation. CONCLUSION: Multi-parametric method confirmed stronger arrest of G2 by MIL5 to show better anti-cancer function both in vitro and in vivo than 2C4.


Subject(s)
Antibodies/administration & dosage , Breast Neoplasms/drug therapy , Cell Cycle Checkpoints , G2 Phase/drug effects , Receptor, ErbB-2/antagonists & inhibitors , Animals , Antibodies/metabolism , Cell Line, Tumor , Disease Models, Animal , Epitopes/metabolism , Humans , Mice, Inbred BALB C , Mice, Nude , Protein Binding , Treatment Outcome
20.
J Immunol ; 192(9): 4192-201, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24670802

ABSTRACT

Clinical trials have shown that BAFF inhibitors do not reduce memory B cell levels but can reduce the number of mature B cells. It remains uncertain whether BAFF affects memory-maintaining cytokines such as IL-15. We found that BAFF suppressed IL-15 expression in B cells from lupus-like or experimental allergic encephalomyelitis mice. When BAFF was blocked with atacicept-IgG, IL-15 expression was upregulated in lupus-like or experimental allergic encephalomyelitis mice. Finally, we showed that BAFF suppressed IL-15 expression in transitional 2 B cells by reducing Foxo1 expression and inducing Foxo1 phosphorylation. This study suggests that BAFF suppresses IL-15 expression in autoimmune diseases, and this opens up the possible opportunity for the clinical application of BAFF- and IL-15-specific therapeutic agents.


Subject(s)
Autoimmune Diseases/immunology , B-Cell Activating Factor/immunology , B-Lymphocytes/immunology , Interleukin-15/biosynthesis , Animals , Autoimmune Diseases/metabolism , B-Cell Activating Factor/metabolism , B-Lymphocytes/metabolism , Blotting, Western , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Interleukin-15/immunology , Lupus Erythematosus, Systemic/blood , Lupus Erythematosus, Systemic/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Multiple Sclerosis/blood , Multiple Sclerosis/immunology , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL