Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 245
Filter
1.
Nat Commun ; 15(1): 7664, 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39227567

ABSTRACT

The immunosuppressive tumor microenvironment (TME) remains a major obstacle to tumor control and causes suboptimal responses to immune checkpoint blockade (ICB) therapy. Thus, developing feasible therapeutic strategies that trigger inflammatory responses in the TME could improve the ICB efficacy. Mitochondria play an essential role in inflammation regulation and tumor immunogenicity induction. Herein, we report the discovery and characterization of a class of small molecules that can recapitulate aqueous self-assembly behavior, specifically target cellular organelles (e.g., mitochondria), and invigorate tumor cell immunogenicity. Mechanistically, this nanoassembly platform dynamically rewires mitochondria, induces endoplasmic reticulum stress, and causes apoptosis/paraptosis-associated immunogenic cell death. After treatment, stressed and dying tumor cells can act as prophylactic or therapeutic cancer vaccines. In preclinical mouse models of cancers with intrinsic or acquired resistance to PD-1 blockade, the local administration of nanoassemblies inflames the immunologically silent TME and synergizes with ICB therapy, generating potent antitumor immunity. This chemically programmed small-molecule immune enhancer acts distinctly from regular cytotoxic therapeutics and offers a promising strategy for synchronous and dynamic tailoring of innate immunity to achieve traceless cancer therapy and overcome immunosuppression in cancers.


Subject(s)
Mitochondria , Neoplasms , Tumor Microenvironment , Animals , Mitochondria/drug effects , Mitochondria/metabolism , Mice , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Humans , Cell Line, Tumor , Neoplasms/immunology , Neoplasms/drug therapy , Immune Checkpoint Inhibitors/pharmacology , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/immunology , Apoptosis/drug effects , Female , Immunogenic Cell Death/drug effects , Mice, Inbred C57BL , Nanoparticles/chemistry , Cancer Vaccines/immunology , Cancer Vaccines/administration & dosage , Immunotherapy/methods
2.
Front Immunol ; 15: 1393248, 2024.
Article in English | MEDLINE | ID: mdl-39114661

ABSTRACT

Objective: Beta cell destruction in type 1 diabetes (T1D) results from the combined effect of inflammation and recurrent autoimmunity. In recent years, the role played by beta cells in the development of T1D has evolved from passive victims of the immune system to active contributors in their own destruction. We and others have demonstrated that perturbations in the islet microenvironment promote endoplasmic reticulum (ER) stress in beta cells, leading to enhanced immunogenicity. Among the underlying mechanisms, secretion of extracellular vesicles (EVs) by beta cells has been suggested to mediate the crosstalk with the immune cell compartment. Methods: To study the role of cellular stress in the early events of T1D development, we generated a novel cellular model for constitutive ER stress by modulating the expression of HSPA5, which encodes BiP/GRP78, in EndoC-ßH1 cells. To investigate the role of EVs in the interaction between beta cells and the immune system, we characterized the EV miRNA cargo and evaluated their effect on innate immune cells. Results: Analysis of the transcriptome showed that HSPA5 knockdown resulted in the upregulation of signaling pathways involved in the unfolded protein response (UPR) and changes the miRNA content of EVs, including reduced levels of miRNAs involved in IL-1ß signaling. Treatment of primary human monocytes with EVs from stressed beta cells resulted in increased surface expression of CD11b, HLA-DR, CD40 and CD86 and upregulation of IL-1ß and IL-6. Conclusion: These findings indicate that the content of EVs derived from stressed beta cells can be a mediator of islet inflammation.


Subject(s)
Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress , Extracellular Vesicles , Insulin-Secreting Cells , MicroRNAs , Monocytes , Extracellular Vesicles/metabolism , Extracellular Vesicles/immunology , Monocytes/immunology , Monocytes/metabolism , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/immunology , Humans , Endoplasmic Reticulum Stress/immunology , MicroRNAs/genetics , Inflammation/immunology , Inflammation/metabolism , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/metabolism , Animals , Cell Line , Islets of Langerhans/immunology , Islets of Langerhans/metabolism , Signal Transduction , Unfolded Protein Response/immunology
3.
Chem Biol Interact ; 400: 111177, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39097071

ABSTRACT

Tartrolon D (TRL) is produced by Teredinibacter turnerae, a symbiotic cellulose-degrading bacteria in shipworm gills. Immunogenic cell death (ICD) induction contributes to a better and longer-lasting response to anticancer treatment. Tumor cells undergoing ICD trigger activation of the immune system, as a vaccine. AIMS: This study aimed to evaluate ICD induction by TRL. MAIN METHODS: Cell viability was evaluated by SRB assay. Cell stress, cell death, ICD features and antigen-presenting molecules were evaluated by flow cytometry and immunoblot. KEY FINDINGS: TRL showed antiproliferative activity on 7 tumor cell lines (L929, HCT 116, B16-F10, WM293A, SK-MEL-28, PC-3M, and MCF-7) and a non-tumor cell (HEK293A), with an inhibition concentration mean (IC50) ranging from 0.03 µM to 13 µM. Metastatic melanomas, SK-MEL-28, B16-F10, and WM293A, were more sensitive cell lines, with IC50 ranging from 0.07 to 1.2 µM. TRL induced apoptosis along with autophagy and endoplasmic reticulum stress and release of typical damage-associated molecular patterns (DAMPs) of ICD such calreticulin, ERp57, and HSP70 exposure, and HMGB1 release. Additionally, melanoma B16-F10 exposed to TRL increased expression of antigen-presenting molecules MHC II and CD1d and induced activation of splenocytes of C57BL/6 mice. SIGNIFICANCE: In spite of recent advances provided by target therapy and immunotherapy, advanced metastatic melanoma is incurable for more than half of patients. ICD inducers yield better and long-lasting responses to anticancer treatment. Our findings shed light on an anticancer candidate of marine origin that induces ICD in melanoma.


Subject(s)
Immunogenic Cell Death , Melanoma , Humans , Immunogenic Cell Death/drug effects , Cell Line, Tumor , Melanoma/immunology , Melanoma/pathology , Melanoma/drug therapy , Animals , Apoptosis/drug effects , Mice , Autophagy/drug effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/immunology , Cell Proliferation/drug effects , Cell Survival/drug effects , HEK293 Cells , Calreticulin/metabolism
4.
Eur J Pharmacol ; 981: 176913, 2024 Oct 15.
Article in English | MEDLINE | ID: mdl-39154830

ABSTRACT

Tumor immunotherapy, especially immune checkpoint inhibitors (ICIs), has been applied in clinical practice, but low response to immune therapies remains a thorny issue. Oncolytic viruses (OVs) are considered promising for cancer treatment because they can selectively target and destroy tumor cells followed by spreading to nearby tumor tissues for a new round of infection. Immunogenic cell death (ICD), which is the major mechanism of OVs' anticancer effects, is induced by endoplasmic reticulum stress and reactive oxygen species overload after virus infection. Subsequent release of specific damage-associated molecular patterns (DAMPs) from different types of tumor cells can transform the tumor microenvironment from "cold" to "hot". In this paper, we broadly define ICD as those types of cell death that is immunogenic, and describe their signaling pathways respectively. Focusing on ICD, we also elucidate the advantages and disadvantages of recent combination therapies and their future prospects.


Subject(s)
Immunogenic Cell Death , Immunotherapy , Neoplasms , Oncolytic Virotherapy , Oncolytic Viruses , Oncolytic Virotherapy/methods , Humans , Immunogenic Cell Death/drug effects , Neoplasms/therapy , Neoplasms/immunology , Immunotherapy/methods , Animals , Oncolytic Viruses/physiology , Oncolytic Viruses/immunology , Tumor Microenvironment/immunology , Endoplasmic Reticulum Stress/immunology , Signal Transduction
5.
Proc Natl Acad Sci U S A ; 121(34): e2321686121, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39141352

ABSTRACT

To broadly measure the spectrum of cellular self-antigens for natural killer T cells (NKT), we developed a sensitive lipidomics system to analyze lipids trapped between CD1d and NKT T cell receptors (TCRs). We captured diverse antigen complexes formed in cells from natural endogenous lipids, with or without inducing endoplasmic reticulum (ER) stress. After separating protein complexes with no, low, or high CD1d-TCR interaction, we eluted lipids to establish the spectrum of self-lipids that facilitate this interaction. Although this unbiased approach identified fifteen molecules, they clustered into only two related groups: previously known phospholipid antigens and unexpected neutral lipid antigens. Mass spectrometry studies identified the neutral lipids as ceramides, deoxyceramides, and diacylglycerols, which can be considered headless lipids because they lack polar headgroups that usually form the TCR epitope. The crystal structure of the TCR-ceramide-CD1d complex showed how the missing headgroup allowed the TCR to predominantly contact CD1d, supporting a model of CD1d autoreactivity. Ceramide and related headless antigens mediated physiological TCR binding affinity, weak NKT cell responses, and tetramer binding to polyclonal human and mouse NKT cells. Ceramide and sphingomyelin are oppositely regulated components of the "sphingomyelin cycle" that are altered during apoptosis, transformation, and ER stress. Thus, the unique molecular link of ceramide to NKT cell response, along with the recent identification of sphingomyelin blockers of NKT cell activation, provide two mutually reinforcing links for NKT cell response to sterile cellular stress conditions.


Subject(s)
Antigens, CD1d , Lipidomics , Natural Killer T-Cells , Receptors, Antigen, T-Cell , Natural Killer T-Cells/immunology , Natural Killer T-Cells/metabolism , Antigens, CD1d/immunology , Antigens, CD1d/metabolism , Animals , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Mice , Lipidomics/methods , Humans , Autoantigens/immunology , Autoantigens/metabolism , Ceramides/metabolism , Ceramides/immunology , Lipids/chemistry , Lipids/immunology , Endoplasmic Reticulum Stress/immunology
6.
J Clin Invest ; 134(13)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38949019

ABSTRACT

Type 3 innate lymphoid cells (ILC3s) are key regulators of intestinal homeostasis and epithelial barrier integrity. In this issue of the JCI, Cao and colleagues found that a sensor of endoplasmic reticulum (ER) stress, the inositol-requiring kinase 1α/X-box-binding protein 1 (IRE1α/XBP1) pathway, fine-tuned the functions of ILC3s. Activation of IRE1α and XBP1 in ILC3s limited intestinal inflammation in mice and correlated with the efficacy of ustekinumab, an IL-12/IL-23 blocker, in patients with Crohn's disease. These results advance our understanding in the use of ILCs as biomarkers not only to predict disease outcomes but also to indicate the response to biologicals in patients with inflammatory bowel disease.


Subject(s)
Endoplasmic Reticulum Stress , Endoribonucleases , Protein Serine-Threonine Kinases , X-Box Binding Protein 1 , X-Box Binding Protein 1/genetics , X-Box Binding Protein 1/metabolism , X-Box Binding Protein 1/immunology , Animals , Endoribonucleases/metabolism , Endoribonucleases/genetics , Endoribonucleases/immunology , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/immunology , Humans , Mice , Endoplasmic Reticulum Stress/immunology , Lymphocytes/immunology , Lymphocytes/metabolism , Signal Transduction/immunology , Crohn Disease/immunology , Crohn Disease/pathology , Crohn Disease/metabolism , Immunity, Innate , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology
7.
Infect Immun ; 92(8): e0030024, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39057915

ABSTRACT

The cGAS/STING sensor system drives innate immune responses to intracellular microbial double-stranded DNA (dsDNA) and bacterial cyclic nucleotide second messengers (e.g., c-di-AMP). STING-dependent cell-intrinsic responses can increase resistance to microbial infection and speed pathogen clearance. Correspondingly, STING activation and signaling are known to be targeted for suppression by effectors from several bacterial pathogens. Whether STING responses are also positively regulated through sensing of specific bacterial effectors is less clear. We find that STING activation through dsDNA, by its canonical ligand 2'-3' cGAMP, or the small molecule DMXAA is potentiated following intracellular delivery of the AB5 toxin family member pertussis toxin from Bordetella pertussis or the B subunit of cholera toxin from Vibrio cholerae. Entry of pertussis toxin or cholera toxin B into mouse macrophages triggers markers of endoplasmic reticulum (ER) stress and enhances ligand-dependent STING responses at the level of STING receptor activation in a manner that is independent of toxin enzymatic activity. Our results provide an example in which STING responses integrate information about the presence of relevant ER-transiting bacterial toxins into the innate inflammatory response and may help to explain the in vivo adjuvant effects of catalytically inactive toxins.


Subject(s)
Bacterial Toxins , Endoplasmic Reticulum Stress , Immunity, Innate , Membrane Proteins , Animals , Mice , Endoplasmic Reticulum Stress/immunology , Membrane Proteins/metabolism , Membrane Proteins/immunology , Bacterial Toxins/immunology , Bacterial Toxins/metabolism , Signal Transduction , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/immunology , Macrophages/immunology , Macrophages/metabolism , Mice, Inbred C57BL , Humans
8.
Front Immunol ; 15: 1427859, 2024.
Article in English | MEDLINE | ID: mdl-39026685

ABSTRACT

Endoplasmic reticulum stress occurs due to large amounts of misfolded proteins, hypoxia, nutrient deprivation, and more. The unfolded protein is a complex intracellular signaling network designed to operate under this stress. Composed of three individual arms, inositol-requiring enzyme 1, protein kinase RNA-like ER kinase, and activating transcription factor-6, the unfolded protein response looks to resolve stress and return to proteostasis. The CD8+ T cell is a critical cell type for the adaptive immune system. The unfolded protein response has been shown to have a wide-ranging spectrum of effects on CD8+ T cells. CD8+ T cells undergo cellular stress during activation and due to environmental insults. However, the magnitude of the effects this response has on CD8+ T cells is still understudied. Thus, studying these pathways is important to unraveling the inner machinations of these powerful cells. In this review, we will highlight the recent literature in this field, summarize the three pathways of the unfolded protein response, and discuss their roles in CD8+ T cell biology and functionality.


Subject(s)
CD8-Positive T-Lymphocytes , Endoplasmic Reticulum Stress , Signal Transduction , Unfolded Protein Response , Unfolded Protein Response/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Humans , Animals , Endoplasmic Reticulum Stress/immunology , Protein Serine-Threonine Kinases/metabolism , Activating Transcription Factor 6/metabolism , Endoribonucleases/metabolism , Endoribonucleases/immunology , Lymphocyte Activation/immunology
9.
Int Immunopharmacol ; 140: 112763, 2024 Oct 25.
Article in English | MEDLINE | ID: mdl-39083925

ABSTRACT

Nogo-B, a ubiquitously expressed member of the reticulon family, plays an important role in maintaining endoplasmic reticulum (ER) structure, regulating protein folding, and calcium homeostasis. In this study, we demonstrate that Nogo-B expression and secretion are upregulated in lung cancer and correlate to overall survival. Nogo-B is secreted by various cells, particularly lung cancer cells. ER stress and phosphorylation at serine 107 can induce Nogo-B secretion. Secretory Nogo-B suppresses the differentiation of Th2 cells and the release of type 2 cytokines, thus influencing the anti-tumor effects of Th2-related immune cells, including IgE+B cell class switching and eosinophil activation.


Subject(s)
Cell Differentiation , Lung Neoplasms , Nogo Proteins , Th2 Cells , Tumor Microenvironment , Humans , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Tumor Microenvironment/immunology , Th2 Cells/immunology , Nogo Proteins/metabolism , Nogo Proteins/genetics , Endoplasmic Reticulum Stress/immunology , Cell Line, Tumor , Cytokines/metabolism , Male , Female , Phosphorylation
10.
Int Immunopharmacol ; 137: 112436, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-38857552

ABSTRACT

Selenium and selenoproteins are closely related to melanoma progression. However, it is unclear how SELENOK affects lipid metabolism, endoplasmic reticulum stress (ERS), immune cell infiltration, survival, and prognosis in melanoma patients. Transcriptome data from melanoma patients was used to investigate SELENOK levels and their effect on prognosis, followed by an investigation of SELENOK's effects on immune cell infiltration. Furthermore, a risk model based on ERS, lipid metabolism, and immune-related genes was constructed, and its utility in melanoma prognosis was evaluated. Finally, the drug sensitivity of the risk model was analyzed to provide a reference for melanoma therapy. The results showed that melanoma with a high SELENOK level had a greater degree of immune cell infiltration and a better prognosis. Additionally, SELENOK was found to regulate ERS, lipid metabolism, and immune cell infiltration in melanoma. The risk model based on SELENOK signature genes successfully predicted the prognosis of melanoma, and the low-risk group exhibited a favorable immunological microenvironment. Furthermore, high-risk patients with melanoma were candidates for chemotherapy with RAS pathway inhibitors, whereas low-risk patients were more susceptible to routinely used chemotherapy medicines. In summary, SELENOK was shown to regulate ERS, lipid metabolism, and immune cell infiltration in melanoma, and SELENOK was positively associated with the prognosis of melanoma. The risk model based on SELENOK signature genes was valuable for melanoma prognosis and therapy.


Subject(s)
Immunotherapy , Melanoma , Humans , Melanoma/immunology , Melanoma/therapy , Melanoma/genetics , Melanoma/drug therapy , Melanoma/mortality , Prognosis , Immunotherapy/methods , Selenoproteins/genetics , Selenoproteins/metabolism , Endoplasmic Reticulum Stress/immunology , Endoplasmic Reticulum Stress/drug effects , Gene Expression Regulation, Neoplastic , Transcriptome , Tumor Microenvironment/immunology , Lipid Metabolism/genetics , Male , Skin Neoplasms/immunology , Skin Neoplasms/therapy , Skin Neoplasms/genetics , Skin Neoplasms/drug therapy , Skin Neoplasms/mortality , Female
11.
Cancer Lett ; 597: 217045, 2024 Aug 10.
Article in English | MEDLINE | ID: mdl-38871246

ABSTRACT

To maintain protein homeostasis, X-box binding protein 1 (XBP1) undergoes splicing following the activation of the unfolded protein response (UPR) in response to endoplasmic reticulum (ER) stress. Although targeting ER stress represents a promising therapeutic strategy, a comprehensive understanding of XBP1 at the cellular level and the link between XBP1 and the innate nervous system is lacking. Here, TCGA pancancer datasets from 33 cancer types, scRNA pancancer datasets from 454 patients and bulk RNA-seq datasets from 155 paired esophageal squamous cell carcinoma (ESCC) patients were analyzed. To cope with ER stress, plasma cells tend to activate XBP1 after undergoing bacterial infection and inflammatory signaling from the innate immune system. Patients with high XBP1 expression in their plasma cells have a higher tumor grade and worse survival. However, activation of the innate immune system with increased XBP1 expression in plasma cells correlates with an increased lymphocyte ratio, indicative of a more robust immune response. Moreover, XBP1 activation appears to initiate leukocyte migration at the transcriptional level. Our study revealed that the XBP1-induced UPR could mediate the crosstalk between optimal acquired humoral immune responses and innate immunity in ESCC.


Subject(s)
Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Immunity, Innate , Plasma Cells , Unfolded Protein Response , X-Box Binding Protein 1 , Humans , X-Box Binding Protein 1/genetics , X-Box Binding Protein 1/metabolism , Esophageal Neoplasms/immunology , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Esophageal Neoplasms/mortality , Esophageal Squamous Cell Carcinoma/immunology , Esophageal Squamous Cell Carcinoma/genetics , Esophageal Squamous Cell Carcinoma/pathology , Plasma Cells/immunology , Plasma Cells/metabolism , Male , Female , Endoplasmic Reticulum Stress/immunology , Gene Expression Regulation, Neoplastic , Middle Aged , Aged , Prognosis
12.
Cancer Immunol Res ; 12(9): 1184-1201, 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-38900485

ABSTRACT

Endoplasmic reticulum (ER) stress leads to hepatocellular carcinoma (HCC) progression. Small extracellular vesicles (sEV) play a crucial role in modulating the tumor microenvironment (TME) by influencing cellular communication and immune responses. However, it is unclear whether ER stress modulates the TME through sEVs. In the current study, we investigated the effects and underlying mechanisms of ER stress on the HCC TME. In vivo and in vitro experiments showed that overactivated ER stress was a salient attribute of the immunosuppressive HCC TME. This was caused by the ATF4-promoted release of small nucleolar RNA host gene 6 (SNHG6)-carrying sEVs, which attenuated T cell-mediated immune responses. Overall, SNHG6 modulated the immunosuppressive TME and aggravated ER stress. Meanwhile, targeting SNHG6 facilitated M1-like macrophage and CD8+ T-cell infiltration and decreased the proportion of M2-like macrophages. In addition, SNHG6 knockdown enhanced anti-PD1 immunotherapeutic efficacy. Moreover, in HCC patients, overexpression of SNHG6 was associated with a lack of response to anti-PD1 therapy and poor prognosis, whereas low SNHG6 expression was associated with improved therapeutic efficacy and prognoses. These data indicate that a correlation exists among ER stress, sEVs, immunosuppressive HCC TME, and immunotherapeutic efficacy. Hence, SNHG6-targeted therapy may represent an effective strategy for patients with HCC.


Subject(s)
Carcinoma, Hepatocellular , Endoplasmic Reticulum Stress , Extracellular Vesicles , Liver Neoplasms , RNA, Long Noncoding , Tumor Microenvironment , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/genetics , Liver Neoplasms/immunology , Liver Neoplasms/pathology , Liver Neoplasms/metabolism , Liver Neoplasms/genetics , Endoplasmic Reticulum Stress/immunology , Tumor Microenvironment/immunology , Extracellular Vesicles/metabolism , Extracellular Vesicles/immunology , Humans , RNA, Long Noncoding/genetics , Animals , Mice , Cell Line, Tumor , Male , Female , Gene Expression Regulation, Neoplastic
13.
J Immunol ; 213(3): 384-393, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38864663

ABSTRACT

Allogeneic hematopoietic cell transplantation is an effective treatment for hematologic malignancies, but the complications such as graft-versus-host disease (GVHD) can limit its benefit. The conditioning regimens before transplant, including chemotherapy or irradiation, can trigger endoplasmic reticulum stress. IRE-1α is a major endoplasmic reticulum stress mediator that can further activate both spliced XBP-1 (XBP-1s) and regulated IRE-1-dependent decay (RIDD). IRE-1α-XBP-1s signaling controls dendritic cell (DC) differentiation and Ag presentation, crucial in GVHD progression. In this study, we used DC-specific XBP-1-deficient mice as donors or recipients and observed that XBP-1s was crucial for host DCs in the induction of GVHD but dispensable for the graft-versus-leukemia response. To specifically target IRE-1α in the host, we treated recipient mice with the IRE-1α inhibitor B-I09 for 3 d prior to bone marrow transplantation, which significantly suppressed GVHD development while maintaining the graft-versus-leukemia effect. XBP-1-deficient or BI09-treated recipients showed reduced DC survival after irradiation and bone marrow transplantation. Inhibition of IRE-1α also led to a reduction in DC alloreactivity, subsequently decreasing the proliferation and activation of allogeneic T cells. With further study using RIDD-deficient DCs, we observed that RIDD was also required for optimal DC activation. Taken together, XBP-1s and RIDD both promote host DC survival and alloreactivity that contribute to GVHD development.


Subject(s)
Dendritic Cells , Endoplasmic Reticulum Stress , Endoribonucleases , Graft vs Host Disease , Protein Serine-Threonine Kinases , X-Box Binding Protein 1 , Animals , Dendritic Cells/immunology , Graft vs Host Disease/immunology , Mice , Endoplasmic Reticulum Stress/immunology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Endoribonucleases/genetics , X-Box Binding Protein 1/genetics , X-Box Binding Protein 1/metabolism , Mice, Knockout , Mice, Inbred C57BL , Hematopoietic Stem Cell Transplantation , Bone Marrow Transplantation , Signal Transduction , Cell Differentiation/immunology , Graft vs Leukemia Effect/immunology
14.
Int Immunopharmacol ; 136: 112342, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-38820956

ABSTRACT

NLRP3 inflammasome is a key component of the innate immune system, mediating the activation of caspase-1, and the maturity and secretion of the pro-inflammatory cytokine interleukin (IL)-1beta (IL-1ß) and IL-18 to cope with microbial infections and cell injury. The NLRP3 inflammasome is activated by various endogenous danger signals, microorganisms and environmental stimuli, including urate, extracellular adenosine triphosphate (ATP) and cholesterol crystals. Increasing evidence indicates that the abnormal activation of NLRP3 is involved in multiple diseases including renal diseases. Hence, clarifying the mechanism of action of NLRP3 inflammasome in different diseases can help prevent and treat various diseases. Endoplasmic reticulum (ER) is an important organelle which participates in cell homeostasis maintenance and protein quality control. The unfolded protein response (UPR) and ER stress are caused by the excessive accumulation of unfolded or misfolded proteins in ER to recover ER homeostasis. Many factors can cause ER stress, including inflammation, hypoxia, environmental toxins, viral infections, glucose deficiency, changes in Ca2+ level and oxidative stress. The dysfunction of ER stress participates in multiple diseases, such as renal diseases. Many previous studies have shown that NLRP3 inflammasome and ER stress play an important role in renal diseases. However, the relevant mechanisms are not yet fully clear. Herein, we focus on the current understanding of the role and mechanism of ER stress and NLRP3 inflammasome in renal diseases, hoping to provide theoretical references for future related researches.


Subject(s)
Endoplasmic Reticulum Stress , Inflammasomes , Kidney Diseases , NLR Family, Pyrin Domain-Containing 3 Protein , Humans , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Endoplasmic Reticulum Stress/immunology , Kidney Diseases/immunology , Kidney Diseases/metabolism , Inflammasomes/metabolism , Inflammasomes/immunology , Animals , Unfolded Protein Response/immunology
15.
Int Immunopharmacol ; 135: 112315, 2024 Jun 30.
Article in English | MEDLINE | ID: mdl-38805908

ABSTRACT

Exosomes generated from mesenchymal stem cells (MSCs) are thought to be a unique therapeutic strategy for several autoimmune deficiency illnesses. The purpose of this study was to elucidate the protective effects of human umbilical cord mesenchymal stem cell-derived exosomes (hUCMSC-Exo) on CD4+ T cells dysfunction during graft-versus-host disease (GVHD) and to identify the underlying processes involved. Here, we showed that hUCMSC-Exo treatment can effectively attenuate GVHD injury by alleviating redox metabolism disorders and inflammatory cytokine bursts in CD4+ T cells. Furthermore, hUCMSC-Exo ameliorate ER stress and ATF6/CHOP signaling-mediated apoptosis in CD4+ T cells and promote the development of CD4+IL-10+ T cells during GVHD. Moreover, downregulating miR-16-5p in hUCMSC-Exo impaired their ability to prevent CD4+ T cells apoptosis and weakened their ability to promote the differentiation of CD4+IL-10+ T cells. Collectively, the obtained data suggested that hUCMSC-Exo suppress ATF6/CHOP signaling-mediated ER stress and apoptosis in CD4+ T cells, enhance the differentiation of CD4+IL-10+ T cells, and reverse the imbalance of immune homeostasis in the GVHD process by transferring miR-16-5p. Our study provided further evidence that GVHD patients can benefit from hUCMSC-Exo-mediated therapy.


Subject(s)
Activating Transcription Factor 6 , CD4-Positive T-Lymphocytes , Endoplasmic Reticulum Stress , Exosomes , Graft vs Host Disease , Mesenchymal Stem Cells , MicroRNAs , Signal Transduction , Transcription Factor CHOP , MicroRNAs/metabolism , MicroRNAs/genetics , Exosomes/metabolism , Endoplasmic Reticulum Stress/immunology , Graft vs Host Disease/immunology , Graft vs Host Disease/prevention & control , Humans , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Animals , Activating Transcription Factor 6/metabolism , Activating Transcription Factor 6/genetics , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/immunology , Transcription Factor CHOP/metabolism , Transcription Factor CHOP/genetics , Apoptosis , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Umbilical Cord/cytology , Cells, Cultured
16.
J Clin Invest ; 134(13)2024 May 09.
Article in English | MEDLINE | ID: mdl-38722686

ABSTRACT

Group 3 innate lymphoid cells (ILC3s) are key players in intestinal homeostasis. ER stress is linked to inflammatory bowel disease (IBD). Here, we used cell culture, mouse models, and human specimens to determine whether ER stress in ILC3s affects IBD pathophysiology. We show that mouse intestinal ILC3s exhibited a 24-hour rhythmic expression pattern of the master ER stress response regulator inositol-requiring kinase 1α/X-box-binding protein 1 (IRE1α/XBP1). Proinflammatory cytokine IL-23 selectively stimulated IRE1α/XBP1 in mouse ILC3s through mitochondrial ROS (mtROS). IRE1α/XBP1 was activated in ILC3s from mice exposed to experimental colitis and in inflamed human IBD specimens. Mice with Ire1α deletion in ILC3s (Ire1αΔRorc) showed reduced expression of the ER stress response and cytokine genes including Il22 in ILC3s and were highly vulnerable to infections and colitis. Administration of IL-22 counteracted their colitis susceptibility. In human ILC3s, IRE1 inhibitors suppressed cytokine production, which was upregulated by an IRE1 activator. Moreover, the frequencies of intestinal XBP1s+ ILC3s in patients with Crohn's disease before administration of ustekinumab, an anti-IL-12/IL-23 antibody, positively correlated with the response to treatment. We demonstrate that a noncanonical mtROS-IRE1α/XBP1 pathway augmented cytokine production by ILC3s and identify XBP1s+ ILC3s as a potential biomarker for predicting the response to anti-IL-23 therapies in IBD.


Subject(s)
Endoribonucleases , Immunity, Innate , Inflammatory Bowel Diseases , Protein Serine-Threonine Kinases , X-Box Binding Protein 1 , Animals , X-Box Binding Protein 1/genetics , X-Box Binding Protein 1/immunology , X-Box Binding Protein 1/metabolism , Mice , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/immunology , Endoribonucleases/genetics , Endoribonucleases/metabolism , Endoribonucleases/immunology , Humans , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/pathology , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/metabolism , Lymphocytes/immunology , Lymphocytes/metabolism , Endoplasmic Reticulum Stress/immunology , Cytokines/metabolism , Cytokines/immunology , Cytokines/genetics , Signal Transduction/immunology , Mice, Knockout , Male , Female
17.
J Affect Disord ; 356: 190-203, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38604455

ABSTRACT

BACKGROUND: Several lines of evidence point to an interaction between genetic predisposition and environmental factors in the onset of major depressive disorder (MDD). This study is aimed to investigate the pathogenesis of MDD by identifying key biomarkers, associated immune infiltration using bioinformatic analysis and human postmortem sample. METHODS: The Gene Expression Omnibus (GEO) database of GSE98793 was adopted to identify hub genes linked to endoplasmic reticulum (ER) stress-related genes (ERGs) in MDD. Another GEO database of GSE76826 was employed to validate the novel target associated with ERGs and immune infiltration in MDD. Moreover, human postmortem sample from MDD patients was utilized to confirm the differential expression analysis of hub genes. RESULTS: We discovered 12 ER stress-related differentially expressed genes (ERDEGs). A LASSO Cox regression analysis helped construct a diagnostic model for these ERDEGs, incorporating immune infiltration analysis revealed that three hub genes (ERLIN1, SEC61B, and USP13) show the significant and consistent expression differences between the two groups. Western blot analysis of postmortem brain samples indicated notably higher expression levels of ERLIN1 and SEC61B in the MDD group, with USP13 also tending to increase compared to control group. LIMITATIONS: The utilization of the MDD gene chip in this analysis was sourced from the GEO database, which possesses a restricted number of pertinent gene chip samples. CONCLUSIONS: These findings indicate that ERDEGs especially including ERLIN1, SEC61B, and USP13 associated the infiltration of immune cells may be potential diagnostic indicators for MDD.


Subject(s)
Depressive Disorder, Major , Endoplasmic Reticulum Stress , Humans , Depressive Disorder, Major/genetics , Depressive Disorder, Major/immunology , Endoplasmic Reticulum Stress/genetics , Endoplasmic Reticulum Stress/immunology , Endoplasmic Reticulum/metabolism , Membrane Proteins/genetics , Computational Biology , Male , Female , Biomarkers/metabolism , Gene Expression Profiling , Brain/immunology , Brain/metabolism , Brain/pathology
18.
Am J Respir Cell Mol Biol ; 71(2): 169-181, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38593442

ABSTRACT

Heightened unfolded protein responses (UPRs) are associated with the risk for asthma, including severe asthma. Treatment-refractory severe asthma manifests a neutrophilic phenotype with T helper (Th)17 responses. However, how UPRs participate in the deregulation of Th17 cells leading to neutrophilic asthma remains elusive. This study found that the UPR sensor IRE1 is induced in the murine lung with fungal asthma and is highly expressed in Th17 cells relative to naive CD4+ T cells. Cytokine (e.g., IL-23) signals induce the IRE1-XBP1s axis in a JAK2-dependent manner. This noncanonical activation of the IRE1-XBP1s pathway promotes UPRs and cytokine secretion by both human and mouse Th17 cells. Ern1 (encoding IRE1) deficiency decreases the expression of endoplasmic reticulum stress factors and impairs the differentiation and cytokine secretion of Th17 cells. Genetic ablation of Ern1 leads to alleviated Th17 responses and airway neutrophilia in a fungal airway inflammation model. Consistently, IL-23 activates the JAK2-IRE1-XBP1s pathway in vivo and enhances Th17 responses and neutrophilic infiltration into the airway. Taken together, our data indicate that IRE1, noncanonically activated by cytokine signals, promotes neutrophilic airway inflammation through the UPR-mediated secretory function of Th17 cells. The findings provide a novel insight into the fundamental understanding of IRE1 in Th17-biased TH2-low asthma.


Subject(s)
Asthma , Endoribonucleases , Neutrophils , Protein Serine-Threonine Kinases , Th17 Cells , Animals , Th17 Cells/immunology , Th17 Cells/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Neutrophils/immunology , Neutrophils/metabolism , Humans , Endoribonucleases/metabolism , Endoribonucleases/genetics , Asthma/immunology , Asthma/pathology , Asthma/metabolism , Unfolded Protein Response , Mice , Mice, Inbred C57BL , Interleukin-23/metabolism , Interleukin-23/immunology , Endoplasmic Reticulum Stress/immunology , X-Box Binding Protein 1/metabolism , X-Box Binding Protein 1/genetics , Signal Transduction , Mice, Knockout , Lung/immunology , Lung/pathology , Lung/metabolism
19.
Int Immunopharmacol ; 133: 112056, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38626546

ABSTRACT

OBJECTIVES: The aim of this study was to investigate the effect of 4µ8c, an inhibitor targeting the endoplasmic reticulum stress-associated factor IRE1α, on macrophage polarization in an experimental model of diabetic periodontitis through ex vivo experiments. MATERIALS AND METHODS: Local alveolar bone parameters were evaluated using Micro-CT following intraperitoneal administration of 4µ8c in mice with experimental diabetic periodontitis. Surface markers indicating macrophage polarization were identified using immunofluorescence. In vitro experiments were performed employing bone marrow-derived macrophages and gingival fibroblasts. Macrophage polarization was determined using flow cytometry. Principal impacted signaling pathways were identified through Western blot analysis. RESULTS: Results from both in vitro and in vivo experiments demonstrated that 4µ8c mitigated alveolar bone resorption and inflammation in mice with diabetic periodontitis. Furthermore, it modulated macrophage polarization towards the M2 phenotype and augmented M2 macrophage polarization through the MAPK signaling pathway. CONCLUSIONS: These findings suggest that inhibiting IRE1α can modulate macrophage polarization and alleviate ligature-induced diabetic periodontitis via the MAPK signaling pathway. This unveils a novel mechanism, offering a scientific foundation for the treatment of experimental diabetic periodontitis.


Subject(s)
Diabetes Mellitus, Type 2 , Endoplasmic Reticulum Stress , Endoribonucleases , Macrophages , Periodontitis , Protein Serine-Threonine Kinases , Animals , Humans , Male , Mice , Alveolar Bone Loss/immunology , Cells, Cultured , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/immunology , Diabetes Mellitus, Type 2/metabolism , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/immunology , Endoribonucleases/metabolism , Macrophages/immunology , Macrophages/metabolism , Macrophages/drug effects , MAP Kinase Signaling System/drug effects , Mice, Inbred C57BL , Periodontitis/immunology , Periodontitis/metabolism , Protein Serine-Threonine Kinases/metabolism
20.
Cell Mol Immunol ; 21(6): 604-619, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38689020

ABSTRACT

Autoreactive CD8+ T cells play a key role in type 1 diabetes (T1D), but the antigen spectrum that activates autoreactive CD8+ T cells remains unclear. Endoplasmic reticulum stress (ERS) has been implicated in ß-cell autoantigen generation. Here, we analyzed the major histocompatibility complex class I (MHC-I)-associated immunopeptidome (MIP) of islet ß-cells under steady and ERS conditions and found that ERS reshaped the MIP of ß-cells and promoted the MHC-I presentation of a panel of conventional self-peptides. Among them, OTUB258-66 showed immunodominance, and the corresponding autoreactive CD8+ T cells were diabetogenic in nonobese diabetic (NOD) mice. High glucose intake upregulated pancreatic OTUB2 expression and amplified the OTUB258-66-specific CD8+ T-cell response in NOD mice. Repeated OTUB258-66 administration significantly reduced the incidence of T1D in NOD mice. Interestingly, peripheral blood mononuclear cells (PBMCs) from patients with T1D, but not from healthy controls, showed a positive IFN-γ response to human OTUB2 peptides. This study provides not only a new explanation for the role of ERS in promoting ß-cell-targeted autoimmunity but also a potential target for the prevention and treatment of T1D. The data are available via ProteomeXchange with the identifier PXD041227.


Subject(s)
CD8-Positive T-Lymphocytes , Diabetes Mellitus, Type 1 , Endoplasmic Reticulum Stress , Insulin-Secreting Cells , Mice, Inbred NOD , Animals , Diabetes Mellitus, Type 1/immunology , Humans , CD8-Positive T-Lymphocytes/immunology , Endoplasmic Reticulum Stress/immunology , Mice , Insulin-Secreting Cells/immunology , Insulin-Secreting Cells/metabolism , Female , Autoantigens/immunology , Peptides/immunology , Peptides/pharmacology , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL