Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 24
1.
PLoS One ; 17(1): e0261789, 2022.
Article En | MEDLINE | ID: mdl-35030194

Nonalcoholic fatty liver disease (NAFLD) is one of the most common causes of liver diseases in the United States and can progress to cirrhosis, end-stage liver disease and need for liver transplantation. There are limited therapies for NAFLD, in part, due to incomplete understanding of the disease pathogenesis, which involves different cell populations in the liver. Endoplasmic reticulum stress and its adaptative unfolded protein response (UPR) signaling pathway have been implicated in the progression from simple hepatic steatosis to nonalcoholic steatohepatitis (NASH). We have previously shown that mice lacking the UPR protein X-box binding protein 1 (XBP1) in the liver demonstrated enhanced liver injury and fibrosis in a high fat sugar (HFS) dietary model of NAFLD. In this study, to better understand the role of liver XBP1 in the pathobiology of NAFLD, we fed hepatocyte XBP1 deficient mice a HFS diet or chow and investigated UPR and other cell signaling pathways in hepatocytes, hepatic stellate cells and immune cells. We demonstrate that loss of XBP1 in hepatocytes increased inflammatory pathway expression and altered expression of the UPR signaling in hepatocytes and was associated with enhanced hepatic stellate cell activation after HFS feeding. We believe that a better understanding of liver cell-specific signaling in the pathogenesis of NASH may allow us to identify new therapeutic targets.


Diet, High-Fat/adverse effects , Dietary Carbohydrates/adverse effects , Endoplasmic Reticulum Stress/immunology , Liver , Signal Transduction/immunology , Unfolded Protein Response/immunology , X-Box Binding Protein 1/deficiency , Animals , Endoplasmic Reticulum Stress/genetics , Liver/immunology , Liver/injuries , Mice , Mice, Knockout , Non-alcoholic Fatty Liver Disease/chemically induced , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/immunology , Signal Transduction/genetics , Unfolded Protein Response/genetics , X-Box Binding Protein 1/immunology
2.
Front Immunol ; 12: 705484, 2021.
Article En | MEDLINE | ID: mdl-34659198

Allogeneic hematopoietic cell transplantation (allo-HCT) is an effective therapeutic procedure to treat hematological malignancies. However, the benefit of allo-HCT is limited by a major complication, chronic graft-versus-host disease (cGVHD). Since transmembrane and secretory proteins are generated and modified in the endoplasmic reticulum (ER), the ER stress response is of great importance to secretory cells including B cells. By using conditional knock-out (KO) of XBP-1, IRE-1α or both specifically on B cells, we demonstrated that the IRE-1α/XBP-1 pathway, one of the major ER stress response mediators, plays a critical role in B cell pathogenicity on the induction of cGVHD in murine models of allo-HCT. Endoribonuclease activity of IRE-1α activates XBP-1 signaling by converting unspliced XBP-1 (XBP-1u) mRNA into spliced XBP-1 (XBP-1s) mRNA but also cleaves other ER-associated mRNAs through regulated IRE-1α-dependent decay (RIDD). Further, ablation of XBP-1s production leads to unleashed activation of RIDD. Therefore, we hypothesized that RIDD plays an important role in B cells during cGVHD development. In this study, we found that the reduced pathogenicity of XBP-1 deficient B cells in cGVHD was reversed by RIDD restriction in IRE-1α kinase domain KO mice. Restraining RIDD activity per se in B cells resulted in an increased severity of cGVHD. Besides, inhibition of RIDD activity compromised B cell differentiation and led to dysregulated expression of MHC II and costimulatory molecules such as CD86, CD40, and ICOSL in B cells. Furthermore, restraining the RIDD activity without affecting XBP-1 splicing increased B cell ability to induce cGVHD after allo-HCT. These results suggest that RIDD is an important mediator for reducing cGVHD pathogenesis through targeting XBP-1s.


B-Lymphocytes/immunology , Endoribonucleases/immunology , Graft vs Host Disease/immunology , Hematopoietic Stem Cell Transplantation , Protein Serine-Threonine Kinases/immunology , Proteolysis , X-Box Binding Protein 1/immunology , Allografts , Animals , Chronic Disease , Endoplasmic Reticulum Stress/genetics , Endoplasmic Reticulum Stress/immunology , Endoribonucleases/genetics , Graft vs Host Disease/genetics , Mice , Mice, Inbred BALB C , Mice, Transgenic , Protein Serine-Threonine Kinases/genetics , Signal Transduction , X-Box Binding Protein 1/genetics
3.
Signal Transduct Target Ther ; 6(1): 357, 2021 10 20.
Article En | MEDLINE | ID: mdl-34667145

Macrophages are among the most abundant immune cells in colorectal cancer (CRC). Re-educating tumor-associated macrophages (TAMs) to switch from protumoral to anti-tumoral activity is an attractive treatment strategy that warrants further investigation. However, little is known about the key pathway that is activated in TAMs. In this study, infitrating CD206+ TAMs in CRC were sorted and subjected to RNA-seq analysis. Differentially expressed genes were found to be enriched in unfolded protein response/endoplasmic reticulum stress response processes, and XBP1 splicing/activation was specifically observed in TAMs. XBP1 activation in TAMs promoted the growth and metastasis of CRC. Ablation of XBP1 inhibited the expression of the pro-tumor cytokine signature of TAMs, including IL-6, VEGFA, and IL-4. Simultaneously, XBP1 depletion could directly inhibit the expression of SIRPα and THBS1, thereby blocking "don't eat me" recognition signals and enhancing phagocytosis. Therapeutic XBP1 gene editing using AAV2-sgXBP1 enhanced the anti-tumor activity. Together, XBP1 activation in TAMs drives CRC progression by elevating pro-tumor cytokine expression and secretion, as well as inhibiting macrophage phagocytosis. Targeting XBP1 signaling in TAMs may be a potential strategy for CRC therapy.


Antigens, Differentiation/genetics , Colorectal Neoplasms/genetics , Receptors, Immunologic/genetics , Thrombospondins/genetics , Tumor-Associated Macrophages/transplantation , X-Box Binding Protein 1/genetics , Aged , Aged, 80 and over , Animals , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Colorectal Neoplasms/therapy , Endoplasmic Reticulum Stress/genetics , Female , Gene Expression Regulation, Neoplastic/immunology , HCT116 Cells , Heterografts , Humans , Interleukin-4 , Interleukin-6/genetics , Male , Mannose Receptor/immunology , Mice , Middle Aged , Phagocytosis , RNA-Seq , Tumor-Associated Macrophages/immunology , Unfolded Protein Response/genetics , Vascular Endothelial Growth Factor A/genetics , X-Box Binding Protein 1/antagonists & inhibitors , X-Box Binding Protein 1/immunology
4.
Nat Commun ; 12(1): 5321, 2021 09 07.
Article En | MEDLINE | ID: mdl-34493732

CARM1 is often overexpressed in human cancers including in ovarian cancer. However, therapeutic approaches based on CARM1 expression remain to be an unmet need. Cancer cells exploit adaptive responses such as the endoplasmic reticulum (ER) stress response for their survival through activating pathways such as the IRE1α/XBP1s pathway. Here, we report that CARM1-expressing ovarian cancer cells are selectively sensitive to inhibition of the IRE1α/XBP1s pathway. CARM1 regulates XBP1s target gene expression and directly interacts with XBP1s during ER stress response. Inhibition of the IRE1α/XBP1s pathway was effective against ovarian cancer in a CARM1-dependent manner both in vitro and in vivo in orthotopic and patient-derived xenograft models. In addition, IRE1α inhibitor B-I09 synergizes with immune checkpoint blockade anti-PD1 antibody in an immunocompetent CARM1-expressing ovarian cancer model. Our data show that pharmacological inhibition of the IRE1α/XBP1s pathway alone or in combination with immune checkpoint blockade represents a therapeutic strategy for CARM1-expressing cancers.


Carcinoma, Ovarian Epithelial/therapy , Endoribonucleases/genetics , Ovarian Neoplasms/therapy , Programmed Cell Death 1 Receptor/genetics , Protein Serine-Threonine Kinases/genetics , Protein-Arginine N-Methyltransferases/genetics , X-Box Binding Protein 1/genetics , Animals , Antibodies, Monoclonal/pharmacology , Base Sequence , Benzopyrans/pharmacology , Carcinoma, Ovarian Epithelial/genetics , Carcinoma, Ovarian Epithelial/immunology , Carcinoma, Ovarian Epithelial/pathology , Cell Line, Tumor , Endoplasmic Reticulum Stress/drug effects , Endoribonucleases/antagonists & inhibitors , Endoribonucleases/immunology , Female , Gene Expression Regulation, Neoplastic , Humans , Hymecromone/analogs & derivatives , Hymecromone/pharmacology , Immune Checkpoint Inhibitors , Mice , Molecular Targeted Therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/immunology , Ovarian Neoplasms/pathology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology , Protein Binding , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/immunology , Protein-Arginine N-Methyltransferases/antagonists & inhibitors , Protein-Arginine N-Methyltransferases/immunology , Signal Transduction , X-Box Binding Protein 1/antagonists & inhibitors , X-Box Binding Protein 1/immunology , Xenograft Model Antitumor Assays
5.
Int J Biochem Cell Biol ; 131: 105906, 2021 02.
Article En | MEDLINE | ID: mdl-33370716

BACKGROUND: Apolipoprotein H (APOH), also known as beta2-glycoprotein I (beta2-GPI), is an acute phase protein in hepatitis B virus (HBV) infection and binds to hepatitis B surface antigen (HBsAg) with high-affinity. APOH expression is upregulated by HBV and the large surface protein (LHBs), but also elevated in HBV-related hepatoma cells. Previous studies show that intracellular retention of HBsAg induces endoplasmic reticulum (ER) stress, a key driver of hepatocyte damage during chronic liver injury, but the mechanisms are unclear. We hypothesize that APOH mediates HBV-induced ER stress through increased retention of HBsAg. METHODS: VR-APOH-myc and VR-LHBs-flag plasmids were constructed by PCR using pcDNA3.1(-)-APOH or an HBV expression vector, respectively. APOH and ER stress markers were examined at protein and mRNA levels by Western Blot or RT-qPCR. HBsAg titer was assayed by ELISA. RNA-seq was performed to elucidate the transcriptional impact of APOH manipulation in HBV-producing cells (HepG2.2.15 cells). RESULTS: We found that HBV upregulates APOH expression in 293 T cells, and APOH overexpression subsequently inhibits secretion of HBsAg. Next, we show that LHBs overexpression in conjunction with APOH leads to ER stress in 293 T cells, as evidenced by production of the binding immunoglobulin protein (BiP) and C/EBP homologous protein (CHOP), as well as increased splicing of X-box binding protein 1 (XBP1). We further observed that loss of beta2-GPI reduced CHOP expression in HepG2.2.15 cells, while beta2-GPI overexpression enhanced CHOP production. CONCLUSION: The interaction of beta2-GPI and HBV initiates ER stress through driving intracellular retention of HBsAg and activates the UPR.


Endoplasmic Reticulum Stress/genetics , Endoplasmic Reticulum/genetics , Hepatitis B Surface Antigens/genetics , Hepatitis B virus/genetics , Host-Pathogen Interactions/genetics , beta 2-Glycoprotein I/genetics , Endoplasmic Reticulum/immunology , Endoplasmic Reticulum/virology , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress/immunology , Gene Expression Regulation , HEK293 Cells , Heat-Shock Proteins/genetics , Heat-Shock Proteins/immunology , Hep G2 Cells , Hepatitis B Surface Antigens/immunology , Hepatitis B virus/immunology , Host-Pathogen Interactions/immunology , Humans , Plasmids/chemistry , Plasmids/metabolism , Protein Binding , Signal Transduction , Transcription Factor CHOP/genetics , Transcription Factor CHOP/immunology , Transfection , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology , X-Box Binding Protein 1/genetics , X-Box Binding Protein 1/immunology , beta 2-Glycoprotein I/immunology
6.
Fish Shellfish Immunol ; 107(Pt A): 202-210, 2020 Dec.
Article En | MEDLINE | ID: mdl-33011436

X-box protein 1 (Xbp1), an essential transcription factor including an unstable form (Xbp1-u) and a stable form (Xbp1-s), plays an vital role in B cell activation and differentiation to plasma cells. In this study, we cloned and identified Xbp1-u gene from Nile tilapia (Oreochromis niloticus), containing 783 bp of nucleotide sequence encoding 260 amino acids. The deduced protein possesses a basic region leucine zipper domain (bZIP) and 26 ribonucleotides of OnXbp1-u transcript. Transcription analysis revealed OnXbp1-u and OnXbp1-s were widely distributed in all examined tissues, with a high expression in immune-related tissues. When stimulated with Streptococcus agalactiae in vivo, the expressions of OnXbp1-u and OnXbp1-s were significantly up-regulated in liver, spleen, head kidney, blood, skin and intestine. After in vitro challenge upon S.agalactiae, the similar up-regulations of OnXbp1-u and OnXbp1-s were also demonstrated in head kidney leukocytes. Moreover, the OnXbp1-u and OnXbp1-s could get involved in LPS-inducible B cell activation and (r)OnIL6-inducible B cell differentiation. Taken together, the results indicated that OnXbp1-u and OnXbp1-s might not only involved in the immune response against S. agalactiae challenge, but also in the B cell activation and differentiation in Nile tilapia.


Fish Diseases/immunology , Gene Expression Regulation/immunology , Immunity, Innate/genetics , Perciformes/genetics , Perciformes/immunology , X-Box Binding Protein 1/genetics , X-Box Binding Protein 1/immunology , Amino Acid Sequence , Animals , Base Sequence , Cichlids , Fish Proteins/chemistry , Fish Proteins/genetics , Fish Proteins/immunology , Gene Expression Profiling/veterinary , Phylogeny , Sequence Alignment/veterinary , X-Box Binding Protein 1/chemistry
7.
Microbiol Immunol ; 64(4): 270-279, 2020 Apr.
Article En | MEDLINE | ID: mdl-31909489

Anaplasma phagocytophilum, an obligate intracellular bacterium that propagates within host granulocytes, is considered to modify the host intracellular environment for pathogenesis. However, the mechanism(s) underlying such host modifications remain unclear. Here, we aimed to investigate the relation between A. phagocytophilum and endoplasmic reticulum (ER) stress in THP-1 cells. A. phagocytophilum activated the three ER stress sensors: inositol-requiring enzyme-1 (IRE1), protein kinase RNA-like endoplasmic reticulum kinase (PERK), and activating transcription factor-6 (ATF6). IRE1 activation occurred immediately after host cell invasion by A. phagocytophilum; however, the activated IRE1-induced splicing of X-box-binding protein 1 was not promoted during A. phagocytophilum infection. This suppression was sustained even after the doxycycline-mediated elimination of intracellular A. phagocytophilum. IRE1 knockdown accelerated A. phagocytophilum-induced apoptosis and decreased intracellular A. phagocytophilum. These data suggest that A. phagocytophilum utilizes IRE1 activation to promote its own intracellular proliferation. Moreover, PERK and ATF6 partially mediated A. phagocytophilum-induced apoptosis by promoting the expression of CCAAT/enhancer-binding protein homologous protein, which induces the transcription of several proapoptotic genes. Thus, A. phagocytophilum possibly manipulates the host ER stress signals to facilitate intracellular proliferation and infection of surrounding cells before/after host cell apoptosis.


Anaplasma phagocytophilum/pathogenicity , Apoptosis/immunology , Ehrlichiosis/immunology , Endoplasmic Reticulum Stress/immunology , Host Microbial Interactions/immunology , Activating Transcription Factor 6/immunology , Cell Line , Ehrlichiosis/microbiology , Endoribonucleases/immunology , Humans , Protein Serine-Threonine Kinases/immunology , X-Box Binding Protein 1/immunology , eIF-2 Kinase/immunology
8.
Sci Rep ; 9(1): 11716, 2019 08 12.
Article En | MEDLINE | ID: mdl-31406212

The pathogenesis of intestinal Behçet's disease (BD) remains poorly understood. Therefore, we aimed to discover and validate biomarkers using proteomics analysis and subsequent functional studies. After two-dimensional electrophoresis, candidate proteins were identified using matrix-assisted laser desorption/ionization tandem time-of-flight mass spectrometry (MALDI-TOF/TOF MS). We validated these results by evaluating the protein levels and their functions in vitro using HT-29 colorectal cancer cells, colon tissues from patients and mice, and murine bone marrow derived macrophages (BMDMs). Of the 30 proteins differentially expressed in intestinal BD tissues, we identified seven using MALDI-TOF/TOF MS. Focusing on galectin-3, we found that TGF-B and IL-10 expression was significantly lower in shLGALS3-transfected cells. Expression of GRP78 and XBP1s and apoptosis rates were all higher in shLGALS3-transfected cells upon the induction of endoplasmic reticulum stress. In response to lipopolysaccharide stimulation, microtubule-associated protein 1 light chain 3B accumulated and lysosomes decreased in these cells. Finally, Salmonella typhimurium infection induced caspase-1 activation and increased IL-1ß production, which facilitated activation of the NLRC4 inflammasome, in Lgals3-/- murine BMDMs compared to wild type BMDMs. Our data suggest that galectin-3 may play a protective role in the pathogenesis of intestinal BD via modulation of ER stress, autophagy, and inflammasome activation.


Behcet Syndrome/immunology , Epithelial Cells/immunology , Galectin 3/immunology , Intestines/immunology , Proteome/immunology , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/immunology , Behcet Syndrome/genetics , Behcet Syndrome/pathology , Blood Proteins , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/immunology , Endoplasmic Reticulum Chaperone BiP , Epithelial Cells/drug effects , Epithelial Cells/pathology , Female , Galectin 3/antagonists & inhibitors , Galectin 3/genetics , Galectins , Gene Expression Regulation , HT29 Cells , Heat-Shock Proteins/genetics , Heat-Shock Proteins/immunology , Humans , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Intestines/drug effects , Intestines/pathology , Lipopolysaccharides/pharmacology , Lysosomes/drug effects , Lysosomes/metabolism , Macrophages/drug effects , Macrophages/immunology , Mice , Mice, Inbred C57BL , Primary Cell Culture , Proteome/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/immunology , X-Box Binding Protein 1/genetics , X-Box Binding Protein 1/immunology
9.
Fish Shellfish Immunol ; 92: 706-711, 2019 Sep.
Article En | MEDLINE | ID: mdl-31276789

Recently, studies have shown that IκB kinase ß (IKKß), a critical kinase in the nucleus factor kappa-B (NF-κB) pathway, participates in inflammatory responses associated with unfolded protein response (UPR) and plays an important role in ER stress-induced cell death. The unfolded protein response (UPR), which is a regulatory system to restore cellular homeostasis in the endoplasmic reticulum (ER), such as oxidative stress, bacterial infection, and virus invasion. The UPR pathways have been reported to be involved in immune responses in mammals, including the classical NF-κB pathway. However, the molecular mechanism of their crosstalk remains to be elucidated. Previously, we demonstrated that IKKß also has some conserved functions between fish and human, as grass carp (Ctenopharyngodon idella) IKKß (CiIKKß) can activate NF-κB pathway. In this study, we found that CiIKKß level in nucleus was elevated under ER stress and CiIKKß can interact with grass carp X-box-binding protein 1 (CiXBP1S), a key transcription factor in UPR. Consistently, fluorescent histochemical analysis of grass carp kidney (CIK) cells indicated that CiIKKß and CiXBP1S colocalized under ER stress. Furthermore, overexpression of CiIKKß in CIK cells enhanced ER stress tolerance by regulating UPR signaling and resulted in the significant increase of cell viability.


Carps/genetics , Endoplasmic Reticulum Stress , Fish Proteins/genetics , Gene Expression Regulation/immunology , I-kappa B Kinase/genetics , X-Box Binding Protein 1/genetics , Animals , Carps/immunology , Cell Nucleus/genetics , Cell Survival , Fish Proteins/immunology , I-kappa B Kinase/immunology , Unfolded Protein Response , X-Box Binding Protein 1/immunology
10.
Front Immunol ; 10: 483, 2019.
Article En | MEDLINE | ID: mdl-30941130

Age-related deficits in the immune system have been associated with an increased incidence of infections, autoimmune diseases, and cancer. Human B cell populations change quantitatively and qualitatively in the elderly. However, the function of human B-1 cells, which play critical anti-microbial and housekeeping roles, have not been studied in the older age population. In the present work, we analyzed how the frequency, function and repertoire of human peripheral blood B-1 cells (CD19+CD20+CD27+CD38low/intCD43+) change with age. Our results show that not only the percentage of B-1 cells but also their ability to spontaneously secrete IgM decreased with age. Further, expression levels of the transcription factors XBP-1 and Blimp-1 were significantly lower, while PAX-5, characteristic of non-secreting B cells, was significantly higher, in healthy donors over 65 years (old) as compared to healthy donors between 20 and 45 years (young). To further characterize the B-1 cell population in older individuals, we performed single cell sequencing analysis of IgM heavy chains from healthy young and old donors. We found reduced repertoire diversity of IgM antibodies in B-1 cells from older donors as well as differences in usage of certain VH and DH specific genes, as compared to younger. Overall, our results show impairment of the human B-1 cell population with advancing age, which might impact the quality of life and onset of disease within the elderly population.


Aging/immunology , Antibodies/immunology , B-Lymphocyte Subsets/immunology , B-Lymphocytes/immunology , Adult , Aged , Aged, 80 and over , Antigens, CD/immunology , Cells, Cultured , Female , Humans , Immunoglobulin M/immunology , Immunologic Memory/immunology , Leukocytes, Mononuclear/immunology , Male , Middle Aged , Positive Regulatory Domain I-Binding Factor 1/immunology , Quality of Life , X-Box Binding Protein 1/immunology , Young Adult
11.
Cell ; 177(5): 1201-1216.e19, 2019 05 16.
Article En | MEDLINE | ID: mdl-31031005

Innate immune responses are intricately linked with intracellular metabolism of myeloid cells. Toll-like receptor (TLR) stimulation shifts intracellular metabolism toward glycolysis, while anti-inflammatory signals depend on enhanced mitochondrial respiration. How exogenous metabolic signals affect the immune response is unknown. We demonstrate that TLR-dependent responses of dendritic cells (DCs) are exacerbated by a high-fatty-acid (FA) metabolic environment. FAs suppress the TLR-induced hexokinase activity and perturb tricarboxylic acid cycle metabolism. These metabolic changes enhance mitochondrial reactive oxygen species (mtROS) production and, in turn, the unfolded protein response (UPR), leading to a distinct transcriptomic signature with IL-23 as hallmark. Interestingly, chemical or genetic suppression of glycolysis was sufficient to induce this specific immune response. Conversely, reducing mtROS production or DC-specific deficiency in XBP1 attenuated IL-23 expression and skin inflammation in an IL-23-dependent model of psoriasis. Thus, fine-tuning of innate immunity depends on optimization of metabolic demands and minimization of mtROS-induced UPR.


Cellular Microenvironment/immunology , Dendritic Cells/immunology , Immunity, Innate , Mitochondria/immunology , Reactive Oxygen Species/immunology , Unfolded Protein Response/immunology , Animals , Cellular Microenvironment/genetics , Citric Acid Cycle/genetics , Citric Acid Cycle/immunology , Dendritic Cells/pathology , Hexokinase/genetics , Hexokinase/immunology , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Mice , Mice, Knockout , Mitochondria/genetics , Toll-Like Receptors/genetics , Toll-Like Receptors/immunology , Unfolded Protein Response/genetics , X-Box Binding Protein 1/genetics , X-Box Binding Protein 1/immunology
12.
Cell Prolif ; 52(2): e12542, 2019 Mar.
Article En | MEDLINE | ID: mdl-30430692

OBJECTIVES: Tumour necrosis factor alpha (TNF-α) expressed by nucleus pulposus cells (NPCs) plays a critical role in intervertebral disc (IVD) degeneration. A key unfolded protein response (UPR) component, X-box binding protein 1 (XBP1) and nuclear factor-kappa B (NF-κB) are essential for cell survival and proliferation. The aim of our study was to elucidate the roles of XBP1 and NF-κB in IVD degeneration (IDD). MATERIALS AND METHODS: Rat NPCs were cultured with TNF-α in the presence or absence of XBP1 and NF-κB-p65 small interfering RNA. The associated genes and proteins were evaluated through quantitative real-time PCR, Western blot analyses and immunofluorescence staining to monitor UPR and NF-κB signalling and identify the regulatory mechanism of p65 by XBP1. Cell counting kit-8 assay, cell cycle analysis and related gene and protein expression were performed to examine the proliferation of NPCs. RESULTS: The acute exposure of TNF-α accelerated the proliferation of rat NPCs by activating the UPR/XBP1 pathway. XBP1 signalling favoured the phosphorylation and nuclear translocation of p65 subunit of NF-κB. The activation of NF-κB in the later phase also enhanced NPC proliferation. CONCLUSIONS: Unfolded protein response reinforces the survival and proliferation of NPCs under TNF-α stimulation by activating the XBP1 pathway, and NF-κB serves as a vital mediator in these events. The XBP1 signalling of UPR can be a novel therapeutic target in IDD.


Cell Proliferation , NF-kappa B/immunology , Nucleus Pulposus/cytology , Signal Transduction , Tumor Necrosis Factor-alpha/immunology , X-Box Binding Protein 1/immunology , Animals , Cells, Cultured , Intervertebral Disc Degeneration/immunology , Nucleus Pulposus/immunology , Rats, Sprague-Dawley
13.
Biochem Biophys Res Commun ; 509(2): 414-420, 2019 02 05.
Article En | MEDLINE | ID: mdl-30594400

BST2 is an antiviral factor that inhibits the release of enveloped virus at the plasma membrane via an unusual topology in which its N-terminal is in the cytosol while its C-terminal is anchored by glycophosphatidylinositol (GPI). BST2-deficient cells showed substantially higher release of virions than wild type cells. Influenza-infected BST2-deficient cells showed greatly reduced cytopathic effect (CPE) than wild type cells despite their generally robust virus production. This finding prompted us to determine whether BST2 was involved in the apoptotic process of virus-infected host cells. Our results revealed that BST2 might be involved in IRE1α-mediated ER stress pathway by increasing spliced form XBP-1. Consequently, levels of cytochrome C, caspase-3, caspase-9, and PARP as representative molecules of apoptosis were significantly increased in wild type cells than those in BST2-deficient cells. These results suggest that BST2 might participate in innate host defense by augmenting ER-stress-induced apoptotic signaling to inhibit the replication and spread of virus.


Antigens, CD/genetics , Endoribonucleases/genetics , Host-Pathogen Interactions/genetics , Influenza A Virus, H1N1 Subtype/genetics , Protein Serine-Threonine Kinases/genetics , X-Box Binding Protein 1/genetics , Animals , Antigens, CD/immunology , Apoptosis/genetics , Apoptosis/immunology , Caspase 3/genetics , Caspase 3/immunology , Caspase 9/genetics , Caspase 9/immunology , Chlorocebus aethiops , Cytochromes c/genetics , Cytochromes c/immunology , Dogs , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/immunology , Endoplasmic Reticulum Stress/genetics , Endoplasmic Reticulum Stress/immunology , Endoribonucleases/immunology , GPI-Linked Proteins/genetics , GPI-Linked Proteins/immunology , Gene Expression Regulation , HEK293 Cells , Host-Pathogen Interactions/immunology , Humans , Immunity, Innate , Influenza A Virus, H1N1 Subtype/immunology , Madin Darby Canine Kidney Cells , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/immunology , Protein Serine-Threonine Kinases/immunology , Signal Transduction , Vero Cells , Virus Replication , X-Box Binding Protein 1/immunology
14.
Front Immunol ; 9: 2887, 2018.
Article En | MEDLINE | ID: mdl-30574153

Acute graft- vs. -host disease (GVHD) is an important cause of morbidity and death after allogeneic hematopoietic cell transplantation (HCT). We identify a new approach to prevent GVHD that impairs monocyte-derived dendritic cell (moDC) alloactivation of T cells, yet preserves graft- vs.-leukemia (GVL). Exceeding endoplasmic reticulum (ER) capacity results in a spliced form of X-box binding protein-1 (XBP-1s). XBP-1s mediates ER stress and inflammatory responses. We demonstrate that siRNA targeting XBP-1 in moDCs abrogates their stimulation of allogeneic T cells. B-I09, an inositol-requiring enzyme-1α (IRE1α) inhibitor that prevents XBP-1 splicing, reduces human moDC migration, allo-stimulatory potency, and curtails moDC IL-1ß, TGFß, and p40 cytokines, suppressing Th1 and Th17 cell priming. B-I09-treated moDCs reduce responder T cell activation via calcium flux without interfering with regulatory T cell (Treg) function or GVL effects by cytotoxic T lymphocytes (CTL) and NK cells. In a human T cell mediated xenogeneic GVHD model, B-I09 inhibition of XBP-1s reduced target-organ damage and pathogenic Th1 and Th17 cells without impacting donor Tregs or anti-tumor CTL. DC XBP-1s inhibition provides an innovative strategy to prevent GVHD and retain GVL.


Dendritic Cells/immunology , Graft vs Host Disease/prevention & control , Hematopoietic Stem Cell Transplantation/adverse effects , Immunosuppression Therapy/methods , Leukemia/therapy , X-Box Binding Protein 1/antagonists & inhibitors , Animals , Cell Line, Tumor , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/immunology , Endoribonucleases/antagonists & inhibitors , Endoribonucleases/metabolism , Enzyme Inhibitors/pharmacology , Female , Gene Knockdown Techniques , Graft vs Host Disease/immunology , Graft vs Leukemia Effect/immunology , Humans , Inflammasomes/drug effects , Inflammasomes/immunology , Inflammasomes/metabolism , Isoantibodies/immunology , Isoantibodies/metabolism , Isoantigens/immunology , Leukemia/immunology , Lymphocyte Activation/drug effects , Male , Mice , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , RNA, Small Interfering/metabolism , Skin Transplantation , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Transplantation Chimera , Transplantation, Homologous/adverse effects , X-Box Binding Protein 1/genetics , X-Box Binding Protein 1/immunology , X-Box Binding Protein 1/metabolism , Xenograft Model Antitumor Assays
15.
Mol Cells ; 41(8): 705-716, 2018 Aug 31.
Article En | MEDLINE | ID: mdl-30078231

The endoplasmic reticulum (ER) is a critical organelle for protein synthesis, folding and modification, and lipid synthesis and calcium storage. Dysregulation of ER functions leads to the accumulation of misfolded- or unfolded-protein in the ER lumen, and this triggers the unfolded protein response (UPR), which restores ER homeostasis. The UPR is characterized by three distinct downstream signaling pathways that promote cell survival or apoptosis depending on the stressor, the intensity and duration of ER stress, and the cell type. Mammalian cells express the UPR transducers IRE1, PERK, and ATF6, which control transcriptional and translational responses to ER stress. Direct links between ER stress and immune responses are also evident, but the mechanisms by which UPR signaling cascades are coordinated with immunity remain unclear. This review discusses recent investigations of the roles of ER stress in immune responses that lead to differentiation, maturation, and cytokine expression in immune cells. Further understanding of how ER stress contributes to the pathogenesis of immune disorders will facilitate the development of novel therapies that target UPR pathways.


Endoplasmic Reticulum Stress/immunology , Unfolded Protein Response/immunology , X-Box Binding Protein 1/immunology , Cell Differentiation , Humans
16.
Leukemia ; 32(9): 1932-1947, 2018 09.
Article En | MEDLINE | ID: mdl-29487385

Histone deacetylases (HDAC) are therapeutic targets in multiple cancers. ACY241, an HDAC6 selective inhibitor, has shown anti-multiple myeloma (MM) activity in combination with immunomodulatory drugs and proteasome inhibitors. Here we show ACY241 significantly reduces the frequency of CD138+ MM cells, CD4+CD25+FoxP3+ regulatory T cells, and HLA-DRLow/-CD11b+CD33+ myeloid-derived suppressor cells; and decreases expression of PD1/PD-L1 on CD8+ T cells and of immune checkpoints in bone marrow cells from myeloma patients. ACY241 increased B7 (CD80, CD86) and MHC (Class I, Class II) expression on tumor and dendritic cells. We further evaluated the effect of ACY241 on antigen-specific cytotoxic T lymphocytes (CTL) generated with heteroclitic XBP1unspliced184-192 (YISPWILAV) and XBP1spliced367-375 (YLFPQLISV) peptides. ACY241 induces co-stimulatory (CD28, 41BB, CD40L, OX40) and activation (CD38) molecule expression in a dose- and time-dependent manner, and anti-tumor activities, evidenced by increased perforin/CD107a expression, IFN-γ/IL-2/TNF-α production, and antigen-specific central memory CTL. These effects of ACY241 on antigen-specific memory T cells were associated with activation of downstream AKT/mTOR/p65 pathways and upregulation of transcription regulators including Bcl-6, Eomes, HIF-1 and T-bet. These studies therefore demonstrate mechanisms whereby ACY241 augments immune response, providing the rationale for its use, alone and in combination, to restore host anti-tumor immunity and improve patient outcome.


Epitopes, T-Lymphocyte/immunology , Histone Deacetylase Inhibitors/pharmacology , Multiple Myeloma/immunology , Neoplasms/immunology , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Biomarkers , Cell Line, Tumor , Cytotoxicity, Immunologic/drug effects , Epitopes, T-Lymphocyte/genetics , Gene Expression Regulation, Neoplastic/drug effects , Histone Deacetylases/metabolism , Humans , Immunologic Memory , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , Multiple Myeloma/metabolism , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism , Peptides/immunology , Signal Transduction/drug effects , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Cytotoxic/metabolism , X-Box Binding Protein 1/chemistry , X-Box Binding Protein 1/genetics , X-Box Binding Protein 1/immunology
17.
Cell Immunol ; 328: 9-17, 2018 06.
Article En | MEDLINE | ID: mdl-29499909

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.


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
18.
Front Immunol ; 9: 3050, 2018.
Article En | MEDLINE | ID: mdl-30687308

The IRE1α/XBP1s signaling pathway is an arm of the unfolded protein response (UPR) that safeguards the fidelity of the cellular proteome during endoplasmic reticulum (ER) stress, and that has also emerged as a key regulator of dendritic cell (DC) homeostasis. However, in the context of DC activation, the regulation of the IRE1α/XBP1s axis is not fully understood. In this work, we report that cell lysates generated from melanoma cell lines markedly induce XBP1s and certain members of the UPR such as the chaperone BiP in bone marrow derived DCs (BMDCs). Activation of IRE1α endonuclease upon innate recognition of melanoma cell lysates was required for amplification of proinflammatory cytokine production and was necessary for efficient cross-presentation of melanoma-associated antigens without modulating the MHC-II antigen presentation machinery. Altogether, this work provides evidence indicating that ex-vivo activation of the IRE1α/XBP1 pathway in BMDCs enhances CD8+ T cell specific responses against tumor antigens.


CD8-Positive T-Lymphocytes/immunology , Endoribonucleases/metabolism , Melanoma/immunology , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/immunology , Animals , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Cross-Priming/drug effects , Cytokines/immunology , Cytokines/metabolism , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/metabolism , Endoplasmic Reticulum Stress/immunology , Endoribonucleases/antagonists & inhibitors , Endoribonucleases/genetics , Endoribonucleases/immunology , Humans , Hymecromone/analogs & derivatives , Hymecromone/pharmacology , Lymphocyte Activation/drug effects , Melanoma/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Primary Cell Culture , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/immunology , Signal Transduction/drug effects , Unfolded Protein Response/immunology , X-Box Binding Protein 1/immunology , X-Box Binding Protein 1/metabolism
19.
Leukemia ; 32(3): 752-764, 2018 03.
Article En | MEDLINE | ID: mdl-29089645

X-box binding protein 1 (XBP1), CD138 (Syndecan-1) and CS1 (SLAMF7) are highly expressed antigens in cancers including multiple myeloma (MM). Here, we identify and characterize immunogenic HLA-A24 peptides derived from these antigens for potential vaccination therapy of HLA-A24+ patients with MM. The identified immunogenic HLA-A24-specific XBP1 unspliced (UN)185-193 (I S P W I L A V L), XBP1 spliced (SP)223-231 (V Y P E G P S S L), CD138265-273 (I F A V C L V G F) and CS1240-248 (L F V L G L F L W) peptides induced antigen-specific CTL with anti-MM activity in an HLA-A24 restricted manner. Furthermore, a cocktail containing the four HLA-A24 peptides evoked MM-specific CTL with distinct phenotypic profiles (CD28, CD40L, 41BB, CD38, CD69) and anti-tumor activities, evidenced by perforin upregulation, CD107a degranulation (cytotoxicity) and Th1-type cytokines (IFN-γ/IL-2/TNF-α) production in response to HLA-A24+ MM cells. The multipeptide-specific CTL included antigen-specific memory CD8+ T cells expressing both T-cell activation (CD38, CD69) and immune checkpoints antigens (CTLA, PD-1, LAG-3, TIM-3). These results provide the framework for a multipeptide vaccination therapy to induce tumor-specific CTL in HLA-A24-positive patients with myeloma and other cancers expressing these antigens.


ADP-ribosyl Cyclase 1/immunology , HLA-A24 Antigen/immunology , Multiple Myeloma/immunology , Peptides/immunology , T-Cell Antigen Receptor Specificity/immunology , T-Lymphocytes, Cytotoxic/immunology , X-Box Binding Protein 1/immunology , ADP-ribosyl Cyclase 1/chemistry , ADP-ribosyl Cyclase 1/metabolism , Amino Acid Sequence , Biomarkers , Cell Line, Tumor , Cytokines/metabolism , Cytotoxicity, Immunologic , Epitopes, T-Lymphocyte/chemistry , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/metabolism , HLA-A24 Antigen/genetics , HLA-A24 Antigen/metabolism , Humans , Immunologic Memory , Intercellular Signaling Peptides and Proteins , Lymphocyte Activation/immunology , Multiple Myeloma/genetics , Multiple Myeloma/metabolism , Peptides/chemistry , Peptides/metabolism , Phenotype , Protein Binding , T-Lymphocytes, Cytotoxic/metabolism , X-Box Binding Protein 1/chemistry , X-Box Binding Protein 1/metabolism
20.
Circ Res ; 121(3): 270-281, 2017 Jul 21.
Article En | MEDLINE | ID: mdl-28620068

RATIONALE: Diverse B cell responses and functions may be involved in atherosclerosis. Protective antibody responses, such as those against oxidized lipid epitopes, are thought to mainly derive from T cell-independent innate B cell subsets. In contrast, both pathogenic and protective roles have been associated with T cell-dependent antibodies, and their importance in both humans and mouse models is still unclear. OBJECTIVE: To specifically target antibody production by plasma cells and determine the impact on atherosclerotic plaque development in mice with and without CD4+ T cells. METHODS AND RESULTS: We combined a model of specific antibody deficiency, B cell-specific CD79a-Cre x XBP1 (X-box binding protein-1) floxed mice (XBP1-conditional knockout), with antibody-mediated depletion of CD4+ T cells. Ldlr knockout mice transplanted with XBP1-conditional knockout (or wild-type control littermate) bone marrow were fed western diet for 8 weeks with or without anti-CD4 depletion. All groups had similar levels of serum cholesterol. In Ldlr/XBP1-conditional knockout mice, serum levels of IgG, IgE, and IgM were significantly attenuated, and local antibody deposition in atherosclerotic plaque was absent. Antibody deficiency significantly accelerated atherosclerosis at both the aortic root and aortic arch. T cell and monocyte responses were not modulated, but necrotic core size was greater, even when adjusting for plaque size, and collagen deposition significantly lower. Anti-CD4 depletion in Ldlr/wild-type mice led to a decrease of serum IgG1 and IgG2c but not IgG3, as well as decreased IgM, associated with increased atherosclerosis and necrotic cores, and a decrease in plaque collagen. The combination of antibody deficiency and anti-CD4 depletion has no additive effects on aortic root atherosclerosis. CONCLUSIONS: The endogenous T cell-dependent humoral response can be protective. This has important implications for novel vaccine strategies for atherosclerosis and in understanding the impacts of immunotherapies used in patients at high risk for cardiovascular disease.


Atherosclerosis/metabolism , B-Lymphocytes/metabolism , T-Lymphocytes/metabolism , X-Box Binding Protein 1/deficiency , Animals , Atherosclerosis/immunology , Atherosclerosis/pathology , B-Lymphocytes/immunology , Immunity, Humoral/physiology , Male , Mice , Mice, Knockout , Plasma Cells/immunology , Plasma Cells/metabolism , T-Lymphocytes/immunology , X-Box Binding Protein 1/immunology
...