Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 422
1.
J Coll Physicians Surg Pak ; 34(5): 527-532, 2024 May.
Article En | MEDLINE | ID: mdl-38720211

OBJECTIVE: To develop an intervention based on Notch-1 signalling pathway blockade by investigating the potential application of the neurogenic locus notch homologue protein 1(Notch-1) signalling pathway as a key regulator of chronic inflammation and adipogenesis in the treatment of hepatic insulin resistance (HIR). STUDY DESIGN: Experimental study. Place and Duration of the Study: Animal Laboratory of the Fourth Hospital of Hebei Medical University, Shijiazhuang, China, from April 2021 to June 2022. METHODOLOGY: HIR models were established in Notch-1WT and Notch-1MAC-KO mice by high fat diet (HFD) for 16 weeks. Haematoxylin and eosin (HE) staining and oil red O (ORO) staining were used to detect inflammatory infiltration and lipid accumulation in each group. Enzyme-linked immunosorbent assay (ELISA) was used to detect the levels of TNF-α and IL-6. Free fatty acid (FFA) and total cholesterol (TC) were measured with relevant kits. Moreover, real-time quantitative polymerase chain reaction (PCR) was performed to detect the relative expressions of F4/80, Mcp1, and CD11b in hepatic tissues. Mass spectrometry was used to analyse the levels of triglyceride (TG), diacylglycerol (DAG) and conformite europeenne (CE) in liver tissue. Western blotting was used to detect the expression of related proteins. RESULTS: Specific knockdown of Notch-1 in macrophages decreases the relative fluorescence intensity of CD68 and attenuates inflammatory infiltration and lipid degeneration. There was no difference in plasma levels of FFA and TG. Specific knockdown of Notch-1 in macrophages decreases the expression of F4/80, Mcp1, and CD11b, as well as the levels of TG, DAG, CE, IL-6, and TNF-α. CONCLUSION: Specific knockout of Notch-1 in macrophages may reduce HIR by inhibiting the IRE1α-XBP1 signalling pathway. KEY WORDS: Hepatic insulin resistance, Macrophages, Notch-1, IRE1α, XBP1.


Diet, High-Fat , Insulin Resistance , Macrophages , Mice, Knockout , Protein Serine-Threonine Kinases , Receptor, Notch1 , Signal Transduction , Animals , Mice , Disease Models, Animal , Endoribonucleases/metabolism , Endoribonucleases/genetics , Insulin Resistance/physiology , Liver/metabolism , Macrophages/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Receptor, Notch1/metabolism , Receptor, Notch1/genetics , X-Box Binding Protein 1/metabolism , X-Box Binding Protein 1/genetics
2.
J Exp Med ; 221(7)2024 Jul 01.
Article En | MEDLINE | ID: mdl-38695876

Platinum-based chemotherapy drugs can lead to the development of anorexia, a detrimental effect on the overall health of cancer patients. However, managing chemotherapy-induced anorexia and subsequent weight loss remains challenging due to limited effective therapeutic strategies. Growth differentiation factor 15 (GDF15) has recently gained significant attention in the context of chemotherapy-induced anorexia. Here, we report that hepatic GDF15 plays a crucial role in regulating body weight in response to chemo drugs cisplatin and doxorubicin. Cisplatin and doxorubicin treatments induce hepatic Gdf15 expression and elevate circulating GDF15 levels, leading to hunger suppression and subsequent weight loss. Mechanistically, selective activation by chemotherapy of hepatic IRE1α-XBP1 pathway of the unfolded protein response (UPR) upregulates Gdf15 expression. Genetic and pharmacological inactivation of IRE1α is sufficient to ameliorate chemotherapy-induced anorexia and body weight loss. These results identify hepatic IRE1α as a molecular driver of GDF15-mediated anorexia and suggest that blocking IRE1α RNase activity offers a therapeutic strategy to alleviate the adverse anorexia effects in chemotherapy.


Anorexia , Doxorubicin , Endoribonucleases , Growth Differentiation Factor 15 , Liver , Protein Serine-Threonine Kinases , Signal Transduction , Unfolded Protein Response , Weight Loss , X-Box Binding Protein 1 , Growth Differentiation Factor 15/metabolism , Growth Differentiation Factor 15/genetics , Endoribonucleases/metabolism , Endoribonucleases/genetics , Animals , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , X-Box Binding Protein 1/metabolism , X-Box Binding Protein 1/genetics , Anorexia/metabolism , Anorexia/chemically induced , Weight Loss/drug effects , Signal Transduction/drug effects , Liver/metabolism , Liver/drug effects , Liver/pathology , Mice , Unfolded Protein Response/drug effects , Doxorubicin/adverse effects , Cisplatin/adverse effects , Mice, Inbred C57BL , Antineoplastic Agents/adverse effects , Male , Humans
3.
In Vivo ; 38(3): 1316-1324, 2024.
Article En | MEDLINE | ID: mdl-38688649

BACKGROUND/AIM: Our objectives in this study were to (i) evaluate the clinical significance of X-box-binding protein 1 (XBP1) expression in cases of hepatocellular carcinoma (HCC) and (ii) assess the potential of XBP1 to be used as a prognostic biomarker. PATIENTS AND METHODS: The expression of XBP1 protein in 267 HCC tissue specimens was measured using immunohistochemistry in order to characterize the associations among XBP1 expression, clinicopathological factors and survival outcomes. Survival analysis using follow-up data was used to assess the prognostic value of XBP1 in cases of HCC. Immunohistochemistry revealed a significant decrease in cytoplasmic XBP1 protein expression in HCC tumor tissue. RESULTS: Immunoreactivity results showed that low cytoplasmic XBP1 expression was significantly associated with vascular invasion, as well as poor 5-year overall survival and long-term disease-specific (DSS) and disease-free (DFS) survival rates. Kaplan-Meier survival curves further confirmed a significant association between low cytoplasmic XBP1 protein expression and poor DSS and DFS. Univariate and multivariate analyses revealed that XBP1 expression, tumor differentiation, vascular invasion, tumor stage, and the rate of recurrence were linked to DSS, while low cytoplasmic XBP1 expression remained an independent predictor of poor DSS. Our analysis also revealed that XBP1 expression, tumor differentiation, vascular invasion, and T classification were linked to DFS, while low cytoplasmic XBP1 expression remained an independent predictor of poor DFS. CONCLUSION: Low cytoplasmic XBP1 protein expression may play an important role in the pathogenesis of HCC, which suggests that XBP1 could potentially be targeted to benefit therapeutic strategies for HCC.


Biomarkers, Tumor , Carcinoma, Hepatocellular , Cytoplasm , Liver Neoplasms , X-Box Binding Protein 1 , Humans , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/mortality , Liver Neoplasms/pathology , Liver Neoplasms/metabolism , Liver Neoplasms/mortality , Liver Neoplasms/genetics , X-Box Binding Protein 1/metabolism , X-Box Binding Protein 1/genetics , Male , Female , Middle Aged , Cytoplasm/metabolism , Prognosis , Biomarkers, Tumor/metabolism , Aged , Adult , Immunohistochemistry , Kaplan-Meier Estimate , Neoplasm Staging
4.
Mol Biol Rep ; 51(1): 599, 2024 Apr 30.
Article En | MEDLINE | ID: mdl-38689181

BACKGROUND: CPUK02 (15-Oxosteviol benzyl ester) is a semi-synthetic derivative of stevioside known for its anticancer effects. It has been reported that the natural compound of stevioside and its associated derivatives enhances the sensitivity of cancer cells to conventional anti-cancer agents by inducing endoplasmic reticulum (ER) stress. In response to ER stress, autophagy and unfolded protein responses (UPR) are activated to restore cellular homeostasis. Consequently, the primary aim of this study is to investigate the impact of CPUK02 treatment on UPR and autophagy markers in two colorectal cancer cell lines. METHODS: HCT116 and SW480 cell lines were treated with various concentrations of CPUK02 for 72 h. The expression levels of several proteins and enzymes were evaluated to investigate the influence of CPUK02 on autophagy and UPR pathways. These include glucose-regulated protein 78 (GRP78), Inositol-requiring enzyme 1-α (IRE1-α), spliced X-box binding protein 1 (XBP-1 s), protein kinase R-like ER kinase (PERK), C/EBP homologous protein (CHOP), Beclin-1, P62 and Microtubule-associated protein 1 light chain 3 alpha (LC3ßII). The evaluation was conducted using western blotting and quantitative real-time PCR techniques. RESULTS: The results obtained indicate that the treatment with CPUK02 reduced the expression of UPR markers, including GRP78 and IRE1-α at protein levels and XBP-1 s, PERK, and CHOP at mRNA levels in both HCT116 and SW480 cell lines. Furthermore, CPUK02 also influenced autophagy by decreasing Beclin-1 and increasing P62 and LC3ßII at mRNA levels in both HCT116 and SW480 treated cells. CONCLUSIONS: The study findings suggest CPUK02 may exert its cytotoxic effects by inhibiting UPR and autophagy flux in colorectal cancer cells.


Autophagy , Colorectal Neoplasms , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress , Unfolded Protein Response , Humans , Autophagy/drug effects , Unfolded Protein Response/drug effects , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Endoplasmic Reticulum Stress/drug effects , HCT116 Cells , Cell Line, Tumor , Diterpenes, Kaurane/pharmacology , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Endoribonucleases/metabolism , Endoribonucleases/genetics , Heat-Shock Proteins/metabolism , Heat-Shock Proteins/genetics , X-Box Binding Protein 1/metabolism , X-Box Binding Protein 1/genetics
5.
J Cell Mol Med ; 28(8): e18247, 2024 Apr.
Article En | MEDLINE | ID: mdl-38520212

Malignant melanoma (MM) is a highly aggressive and deadly form of skin cancer, primarily caused by recurrence and metastasis. Therefore, it is crucial to investigate the regulatory mechanisms underlying melanoma recurrence and metastasis. Our study has identified a potential targeted regulatory relationship between LINC02202, miR-526b-3p and XBP1 in malignant melanoma. Through the regulation of the miR-526b-3p/XBP1 signalling pathway, LINC02202 may play a role in tumour progression and immune infiltration and inhibiting the expression of LINC02202 can increase the efficacy of immunotherapy for melanoma. Our findings shed light on the impact of LINC02202/XBP1 on the phenotype and function of malignant melanoma cells. Furthermore, this study provides a theoretical foundation for the development of novel immunotherapy strategies for malignant melanoma.


Melanoma , MicroRNAs , Skin Neoplasms , Humans , Melanoma/drug therapy , Melanoma/genetics , Melanoma/pathology , MicroRNAs/metabolism , Programmed Cell Death 1 Receptor/metabolism , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Cell Line, Tumor , Skin Neoplasms/genetics , Drug Delivery Systems , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , X-Box Binding Protein 1/genetics , X-Box Binding Protein 1/metabolism
6.
Chin J Integr Med ; 30(5): 398-407, 2024 May.
Article En | MEDLINE | ID: mdl-38386253

OBJECTIVE: To investigate the pharmacological mechanism of Qili Qiangxin Capsule (QLQX) improvement of heart failure (HF) based on miR133a-endoplasmic reticulum stress (ERS) pathway. METHODS: A left coronary artery ligation-induced HF after myocardial infarction model was used in this study. Rats were randomly assigned to the sham group, the model group, the QLQX group [0.32 g/(kg·d)], and the captopril group [2.25 mg/(kg·d)], 15 rats per group, followed by 4 weeks of medication. Cardiac function such as left ventricular ejection fraction (EF), fractional shortening (FS), left ventricular systolic pressure (LVSP), left ventricular end diastolic pressure (LVEDP), the maximal rate of increase of left ventricular pressure (+dp/dt max), and the maximal rate of decrease of left ventricular pressure (-dp/dt max) were monitored by echocardiography and hemodynamics. Hematoxylin and eosin (HE) and Masson stainings were used to visualize pathological changes in myocardial tissue. The mRNA expression of miR133a, glucose-regulated protein78 (GRP78), inositol-requiring enzyme 1 (IRE1), activating transcription factor 6 (ATF6), X-box binding protein1 (XBP1), C/EBP homologous protein (CHOP) and Caspase 12 were detected by RT-PCR. The protein expression of GRP78, p-IRE1/IRE1 ratio, cleaved-ATF6, XBP1-s (the spliced form of XBP1), CHOP and Caspase 12 were detected by Western blot. TdT-mediated dUTP nick-end labeling (TUNEL) staining was used to detect the rate of apoptosis. RESULTS: QLQX significantly improved cardiac function as evidenced by increased EF, FS, LVSP, +dp/dt max, -dp/dt max, and decreased LVEDP (P<0.05, P<0.01). HE staining showed that QLQX ameliorated cardiac pathologic damage to some extent. Masson staining indicated that QLQX significantly reduced collagen volume fraction in myocardial tissue (P<0.01). Results from RT-PCR and Western blot showed that QLQX significantly increased the expression of miR133a and inhibited the mRNA expressions of GRP78, IRE1, ATF6 and XBP1, as well as decreased the protein expressions of GRP78, cleaved-ATF6 and XBP1-s and decreased p-IRE1/IRE1 ratio (P<0.05, P<0.01). Further studies showed that QLQX significantly reduced the expression of CHOP and Caspase12, resulting in a significant reduction in apoptosis rate (P<0.05, P<0.01). CONCLUSION: The pharmacological mechanism of QLQX in improving HF is partly attributed to its regulatory effect on the miR133a-IRE1/XBP1 pathway.


Drugs, Chinese Herbal , Endoplasmic Reticulum Stress , Heart Failure , MicroRNAs , Animals , MicroRNAs/genetics , MicroRNAs/metabolism , Endoplasmic Reticulum Stress/drug effects , Drugs, Chinese Herbal/pharmacology , Heart Failure/drug therapy , Heart Failure/genetics , Male , Rats, Sprague-Dawley , Capsules , Activating Transcription Factor 6/metabolism , Activating Transcription Factor 6/genetics , Endoplasmic Reticulum Chaperone BiP , Apoptosis/drug effects , Caspase 12/metabolism , Caspase 12/genetics , Myocardium/pathology , Myocardium/metabolism , Heat-Shock Proteins/metabolism , Heat-Shock Proteins/genetics , Rats , X-Box Binding Protein 1/metabolism , X-Box Binding Protein 1/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Myocardial Infarction/drug therapy , Myocardial Infarction/pathology , Myocardial Infarction/genetics , Myocardial Infarction/physiopathology
7.
Immunology ; 172(2): 210-225, 2024 Jun.
Article En | MEDLINE | ID: mdl-38366844

Numerous diseases of the immune system can be traced back to the malfunctioning of the regulatory T cells. The aetiology is unclear. Psychological stress can cause disruption to the immune regulation. The synergistic effects of psychological stress and immune response on immune regulation have yet to be fully understood. The intention of this study is to analyse the interaction between psychological stress and immune responses and how it affects the functional status of type 1 regulatory T (Tr1) cells. In this study, ovalbumin peptide T-cell receptor transgenic mice were utilised. Mice were subjected to restraint stress to induce psychological stress. An airway allergy murine model was established, in which a mouse strain with RING finger protein 20 (Rnf20)-deficient CD4+ T cells were used. The results showed that concomitant exposure to restraint stress and immune response could exacerbate endoplasmic reticulum stress in Tr1 cells. Corticosterone was responsible for the elevated expression of X-box protein-1 (XBP1) in mouse Tr1 cells after exposure to both restraint stress and immune response. XBP1 mediated the effects of corticosterone on inducing Rnf20 in Tr1 cells. The reduction of the interleukin-10 expression in Tr1 cells was facilitated by Rnf20. Inhibition of Rnf20 alleviated experimental airway allergy by restoring the immune regulatory ability of Tr1 cells. In conclusion, the functions of Tr1 cells are negatively impacted by simultaneous exposure to psychological stress and immune response. Tr1 cells' immune suppressive functions can be restored by inhibiting Rnf20, which has the translational potential for the treatment of diseases of the immune system.


Interleukin-10 , Mice, Transgenic , Ovalbumin , Stress, Psychological , T-Lymphocytes, Regulatory , Animals , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Ovalbumin/immunology , Stress, Psychological/immunology , Mice , Interleukin-10/metabolism , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell/immunology , X-Box Binding Protein 1/metabolism , X-Box Binding Protein 1/genetics , Corticosterone/blood , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Endoplasmic Reticulum Stress/immunology , Disease Models, Animal , Restraint, Physical , Mice, Knockout , Mice, Inbred C57BL , Respiratory Hypersensitivity/immunology
8.
Cell Death Differ ; 31(4): 447-459, 2024 Apr.
Article En | MEDLINE | ID: mdl-38413797

Hypoxia is a hallmark of cancer development. However, the molecular mechanisms by which hypoxia promotes tumor metastasis are not fully understood. In this study, we demonstrate that hypoxia promotes breast cancer metastasis through suppression of ΔNp63α in a HIF1α-independent manner. We show that hypoxia-activated XBP1s forms a stable repressor protein complex with HDAC2 and EZH2 to suppress ΔNp63α transcription. Notably, H3K27ac is predominantly occupied on the ΔNp63 promoter under normoxia, while H3K27me3 on the promoter under hypoxia. We show that XBP1s binds to the ΔNp63 promoter to recruit HDAC2 and EZH2 in facilitating the switch of H3K27ac to H3K27me3. Pharmacological inhibition or the knockdown of either HDAC2 or EZH2 leads to increased H3K27ac, accompanied by the reduced H3K27me3 and restoration of ΔNp63α expression suppressed by hypoxia, resulting in inhibition of cell migration. Furthermore, the pharmacological inhibition of IRE1α, but not HIF1α, upregulates ΔNp63α expression in vitro and inhibits tumor metastasis in vivo. Clinical analyses reveal that reduced p63 expression is correlated with the elevated expression of XBP1, HDAC2, or EZH2, and is associated with poor overall survival in human breast cancer patients. Together, these results indicate that hypoxia-activated XBP1s modulates the epigenetic program in suppression of ΔNp63α to promote breast cancer metastasis independent of HIF1α and provides a molecular basis for targeting the XBP1s/HDAC2/EZH2-ΔNp63α axis as a putative strategy in the treatment of breast cancer metastasis.


Breast Neoplasms , Enhancer of Zeste Homolog 2 Protein , Epigenesis, Genetic , Histone Deacetylase 2 , Hypoxia-Inducible Factor 1, alpha Subunit , Tumor Suppressor Proteins , X-Box Binding Protein 1 , Humans , Breast Neoplasms/pathology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Enhancer of Zeste Homolog 2 Protein/metabolism , Enhancer of Zeste Homolog 2 Protein/genetics , X-Box Binding Protein 1/metabolism , X-Box Binding Protein 1/genetics , Histone Deacetylase 2/metabolism , Histone Deacetylase 2/genetics , Female , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Tumor Suppressor Proteins/metabolism , Tumor Suppressor Proteins/genetics , Animals , Cell Line, Tumor , Neoplasm Metastasis , Mice , Gene Expression Regulation, Neoplastic , Transcription Factors/metabolism , Transcription Factors/genetics , Cell Hypoxia/genetics
9.
Iran J Med Sci ; 49(1): 10-21, 2024 Jan.
Article En | MEDLINE | ID: mdl-38322164

Background: Three main cell signaling pathways including the endoplasmic reticulum stress (ERS) response, autophagy, and apoptosis play critical roles in both cell survival and death. They were found to crosstalk with one another during tumorigenesis and cancer progression. This study aimed to investigate the expression of the spliced form of X-box binding protein 1 (XBP1s), p62, and caspase-3, as the essential biomarkers of ERS, autophagy, and apoptosis in patients with colorectal cancer (CRC), as well as the correlation between their expression and clinicopathological data. Methods: This retrospective study was conducted on formalin-fixed paraffin-embedded (FFPE) blocks, which were collected from patients and their tumor margins, from the tumor bank of Imam Khomeini Hospital (Tehran, Iran) from 2017 to 2019. Tissue microarray (TMA) was used to measure the XBP1s, p62, and caspase-3 biomarkers. Data were analyzed using SPSS software version 20, and P≤0.05 was considered statistically significant. Results: Evaluating the total of 91 patients, a significant relationship was found between XBP1s expression and TNM stage (P=0.003), primary tumor (pT) (P=0.054), and the degree of differentiation (P=0.006); and between caspase-3 with pT (P=0.004), and lymphovascular invasion (P=0.02). However, no significant correlation was found between p62 and clinicopathological data. Furthermore, a positive relationship between XBP1s and p62 was confirmed (correlation coefficient: 22.2% and P=0.05). Conclusion: Our findings indicated that XBP1s could be considered as a target for therapy in personalized medicine.


Caspase 3 , Colorectal Neoplasms , X-Box Binding Protein 1 , Humans , Biomarkers , Caspase 3/genetics , Clinical Relevance , Colorectal Neoplasms/genetics , Iran , Protein Serine-Threonine Kinases/metabolism , Retrospective Studies , X-Box Binding Protein 1/genetics
10.
J Thromb Haemost ; 22(5): 1475-1488, 2024 May.
Article En | MEDLINE | ID: mdl-38278417

BACKGROUND: Endoplasmic reticulum (ER) stress is a key feature of lipid-laden macrophages and contributes to the development of atherosclerotic plaques. Blood platelets are known to interact with macrophages and fine-tune effector functions such as inflammasome activation and phagocytosis. However, the effect of platelets on ER stress induction is unknown. OBJECTIVES: The objective of this study is to elucidate the potential of platelets in regulating ER stress in macrophages in vitro. METHODS: Bone marrow-derived macrophages and RAW 264.7 cells were incubated with isolated murine platelets, and ER stress and inflammation markers were determined by reverse transcription-quantitative polymerase chain reaction, Western blotting, and enzyme-linked immunosorbent assay. ER morphology was investigated by electron microscopy. Cell viability, lipid accumulation, and activation were measured by flow cytometry. To gain mechanistic insights, coincubation experiments were performed with platelet decoys/releasates as well as lipopolysaccharide, blocking antibodies, and TLR4 inhibitors. RESULTS: Coincubation of platelets and macrophages led to elevated levels of ER stress markers (BIP, IRE1α, CHOP, and XBP1 splicing) in murine and human macrophages, which led to a pronounced enlargement of the ER. Macrophage ER stress was accompanied by increased release of proinflammatory cytokines and intracellular lipid accumulation, but not cell death. Platelet decoys, but not platelet releasates or lysate from other cells, phenocopied the effect of platelets. Blocking TLR4 inhibited inflammatory activation of macrophages but did not affect ER stress induction by platelet coincubation. CONCLUSION: To our knowledge, this study is the first to demonstrate that platelets induce ER stress and unfolded protein response in macrophages by heat-sensitive membrane proteins, independent of inflammatory activation of macrophages.


Blood Platelets , Endoplasmic Reticulum Stress , Macrophages , Mice, Inbred C57BL , Protein Serine-Threonine Kinases , X-Box Binding Protein 1 , Animals , Blood Platelets/metabolism , Macrophages/metabolism , Humans , Mice , RAW 264.7 Cells , X-Box Binding Protein 1/metabolism , X-Box Binding Protein 1/genetics , Toll-Like Receptor 4/metabolism , Endoplasmic Reticulum/metabolism , Endoribonucleases/metabolism , Cytokines/metabolism , Endoplasmic Reticulum Chaperone BiP , Transcription Factor CHOP/metabolism , Signal Transduction , Lipopolysaccharides/pharmacology , Heat-Shock Proteins/metabolism , Lipid Metabolism , Inflammation Mediators/metabolism , Inflammation/metabolism , Cell Survival
11.
Environ Int ; 184: 108445, 2024 Feb.
Article En | MEDLINE | ID: mdl-38262168

Methylparaben (MP), a preservative widely used in daily supplies, exists in both the environment and the human body. However, the potential health risks posed by MP remain unclear. This study aimed to unravel the mechanisms by which MP disrupts glucose and lipid homeostasis. For this, we administered MP to mice and observed changes in glucose and lipid metabolism. MP exposure led to hyperglycemia, hyperlipidemia, visceral organ injury, and hepatic lipid accumulation. RNA sequencing results from mice livers indicated a close association between MP exposure and endoplasmic reticulum (ER) stress, inflammatory response, and glucose and lipid homeostasis. Western blotting and quantitative reverse transcription-polymerase chain reaction revealed that MP activated ER stress, particularly the inositol-requiring enzyme 1 (IRE1)/X-box binding protein 1 (XBP1) pathway, which further promoted the activation of the nuclear factor-kappa B (NF-κB) and mitogen-activated protein kinase (MAPK) pathways. The activation of these pathways phosphorylated insulin receptor substrate-1 (IRS1) (ser 307), resulting in decreased phosphorylation of protein kinase B (Akt) (ser 473), leading to insulin resistance. Additionally, MP exposure promoted lipogenesis through ER stress. To explore potential remedies, we administered the ER stress inhibitor 4-phenylbutyric acid (4-PBA) and the IRE1α-XBP1 pathway inhibitor toyocamycin to mice, both of which protected against metabolic disorders and organ injury caused by MP. These findings suggest that MP induces disruptions in glucose and lipid metabolism through ER stress, primarily through the IRE1α-XBP1 pathway.


Endoribonucleases , Parabens , Protein Serine-Threonine Kinases , Animals , Male , Mice , Endoribonucleases/genetics , Endoribonucleases/metabolism , Glucose , Glycolipids , Lipid Metabolism , Lipids , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , X-Box Binding Protein 1/genetics , X-Box Binding Protein 1/metabolism
12.
J Autoimmun ; 142: 103152, 2024 Jan.
Article En | MEDLINE | ID: mdl-38071801

Anti-nuclear antibodies are the hallmark of autoimmune diseases such as systemic lupus erythematosus (SLE) and scleroderma. However, the molecular mechanisms of B cell tolerance breakdown in these pathological contexts are poorly known. The study of rare familial forms of autoimmune diseases could therefore help to better describe common biological mechanisms leading to B cell tolerance breakdown. By Whole-Exome Sequencing, we identified a new heterozygous mutation (p.R594C) in ERN1 gene, encoding IRE1α (Inositol-Requiring Enzyme 1α), in a multiplex family with several members presenting autoantibody-mediated autoimmunity. Using human cell lines and a knock-in (KI) transgenic mouse model, we showed that this mutation led to a profound defect of IRE1α ribonuclease activity on X-Box Binding Protein 1 (XBP1) splicing. The KI mice developed a broad panel of autoantibodies, however in a subclinical manner. These results suggest that a decrease of spliced form of XBP1 (XBP1s) production could contribute to B cell tolerance breakdown and give new insights into the function of IRE1α which are important to consider for the development of IRE1α targeting strategies.


Autoimmune Diseases , Protein Serine-Threonine Kinases , Humans , Mice , Animals , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Endoribonucleases/genetics , Endoribonucleases/metabolism , Signal Transduction , X-Box Binding Protein 1/genetics , X-Box Binding Protein 1/metabolism , Mice, Transgenic
13.
Int Immunopharmacol ; 126: 111149, 2024 Jan 05.
Article En | MEDLINE | ID: mdl-38006750

BACKGROUND: Tumor-associated macrophages (TAMs) have unique functions in the development of hepatocellular carcinoma (HCC). The tumor microenvironment is in a complex state in chronic disease. As a major participant in tumor-associated inflammation, TAMs have a unique effect on promoting tumor cell proliferation, angiogenesis and immunosuppression. The in-depth study of TAMs has important scientific and clinical value and provides new ideas for the treatment of cancer. METHODS: Bioinformatics analysis, dual-luciferase reporter assays, RT-qPCR and clinical samples were used to analyze the potential mechanism of the miR-21-5p/SP1/XBP1 molecular axis in HCC. In this study, miR-21-5p was highly expressed in HCC exosomes compared with normal hepatocyte exosomes, and HCC exosomes containing miR-21-5p promoted the proliferation and migration of HCC cells and inhibited cell apoptosis. In addition, this treatment promoted the M2 polarization of macrophages, induced the expression of transcription factor-specific protein 1 (SP1), and inhibited the expression of X-box binding protein 1 (XBP1). However, these expression trends were reversed after inhibition of miR-21-5p expression in exosomes of hepatoma cells, and the effects of exosomal miR-21-5p were partially restored after overexpression of SP1. Animal experiments also verified that exosomal miR-21-5p in HCC cells affected the expression level of the SP1/XBP1 protein and promoted M2 polarization of TAMs. CONCLUSION: Exosomal miR-21-5p in HCC cells can affect the development of HCC cells by regulating SP1/XBP1 and promoting the M2 polarization of TAMs, thereby affecting the adverse prognostic response of HCC patients.


Carcinoma, Hepatocellular , Liver Neoplasms , MicroRNAs , Animals , Humans , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , X-Box Binding Protein 1/genetics , X-Box Binding Protein 1/metabolism , Cell Line, Tumor , Macrophages/metabolism , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Tumor Microenvironment/genetics
14.
Cell Signal ; 113: 110935, 2024 01.
Article En | MEDLINE | ID: mdl-37866666

The renin-angiotensin system (RAS) has been recognized as a crucial contributor to the development of liver fibrosis, and AT2R, an essential component of RAS, is involved in the progression of liver fibrosis. However, the underlying mechanisms by which AT2R modulates liver fibrosis remain elusive. Here, we report that AT2R was induced to be highly expressed during the progression of liver fibrosis, and the elevated AT2R attenuates liver fibrosis by suppressing IRE1α-XBP1 pathway. In this study, we found that AT2R is not expressed in the no cirrhotic adult liver, but is induced expression during liver fibrosis in both cirrhotic patients and fibrotic mice models. Upregulated AT2R inhibits the activation and proliferation of hepatic stellate cells (HSCs). In addition, our study showed that during liver fibrosis, AT2R deletion increased the dimerization activation of IRE1α and promoted XBP1 splicing, and the spliced XBP1s could promote their transcription by binding to the AT2R promoter and repress the IRE1α-XBP1 axis, forming an AT2R-IRE1α-XBP1 negative feedback loop. Importantly, the combination treatment of an AT2R agonist and an endoplasmic reticulum stress (ER stress) alleviator significantly attenuated liver fibrosis in a mouse model of liver fibrosis. Therefore, we conclude that the AT2R-IRE1α signaling pathway can regulate the progression of liver fibrosis, and AT2R is a new potential therapeutic target for treating liver fibrosis.


Endoribonucleases , Protein Serine-Threonine Kinases , Humans , Adult , Mice , Animals , Protein Serine-Threonine Kinases/metabolism , Endoribonucleases/metabolism , Angiotensin II , Signal Transduction , Endoplasmic Reticulum Stress , Liver Cirrhosis , X-Box Binding Protein 1/genetics , X-Box Binding Protein 1/metabolism
15.
Cell Signal ; 113: 110929, 2024 01.
Article En | MEDLINE | ID: mdl-37875231

Abnormal differentiation and proliferation of chondrocytes leads to various diseases related to growth and development. The process of chondrocyte differentiation involves a series of complex cellular and molecular interactions. X-box binding protein 1 (XBP1), an essential molecule of the unfolded protein response (UPR) in Endoplasmic Reticulum (ER) stress, participated in cartilage development and causes other related diseases. We previously reported that XBP1 deficiency in cartilage impacts the function and associated diseases of many different tissues including cartilage. However, how differential expression of genes modulates the roles of cartilage and other tissues when XBP1 is lack of in chondrocytes remains unclear. We aimed to screen for differentially expressed (DE) genes in cartilage, brain, heart, and muscle by high-throughput sequencing in XBP1 cartilage-specific knockout (CKO) mice. Further, gene co-expression networks were constructed by weighted gene co-expression network analysis (WGCNA) algorithm and pivot genes were identified in the above four tissues. Protein detection, Hematoxylin-eosin (HE) staining and immunohistochemistry (IHC) experiments have proved that these differentially co-expressed genes participate in the downstream regulatory pathway of different tissues and affect tissue function.Significantly differentially expressed mRNAs [differentially expressed genes (DEGs)] were identified between XBP1 CKO mice and controls in cartilage, brain, heart, and muscle tissues, including 610, 126, 199 and 219 DEGs, respectively. 39 differentially co-expressed genes were identified in the above four tissues, and they were important pivot genes. Comprehensive analysis discovered that XBP1 deficiency in cartilage influences the difference of co-expressed genes between cartilage and other different tissues. These differentially co-expressed genes participate in downstream regulatory pathways of different tissues and affect tissue functions. Collectively, our conclusions may contribute potential biomarkers and molecular mechanisms for the mutual modulation between cartilage and different tissues and the diagnosis and treatment of diseases caused by abnormalities in different tissues. The analysis also provides meaningful insights for future genetic discoveries.


Cartilage , Unfolded Protein Response , Animals , Mice , Cartilage/metabolism , Chondrocytes/metabolism , Endoplasmic Reticulum Stress/genetics , X-Box Binding Protein 1/genetics , X-Box Binding Protein 1/metabolism
16.
Genes Immun ; 25(1): 43-54, 2024 Feb.
Article En | MEDLINE | ID: mdl-38146001

The utilization of host-cell machinery during SARS-CoV-2 infection can overwhelm the protein-folding capacity of the endoplasmic reticulum and activate the unfolded protein response (UPR). The IRE1α-XBP1 arm of the UPR could also be activated by viral RNA via Toll-like receptors. Based on these premises, a study to gain insight into the pathogenesis of COVID-19 disease was conducted using nasopharyngeal exudates and bronchioloalveolar aspirates. The presence of the mRNA of spliced XBP1 and a high expression of cytokine mRNAs were observed during active infection. TLR8 mRNA showed an overwhelming expression in comparison with TLR7 mRNA in bronchioloalveolar aspirates of COVID-19 patients, thus suggesting the presence of monocytes and monocyte-derived dendritic cells (MDDCs). In vitro experiments in MDDCs activated with ssRNA40, a synthetic mimic of SARS-CoV-2 RNA, showed induction of XBP1 splicing and the expression of proinflammatory cytokines. These responses were blunted by the IRE1α inhibitor MKC8866, the TLR8 antagonist CU-CPT9a, and knockdown of TLR8 receptor. In contrast, the IRE1α-XBP1 activator IXA4 enhanced these responses. Based on these findings, the TLR8/IRE1α system seems to play a significant role in the induction of the proinflammatory cytokines associated with severe COVID-19 disease and might be a druggable target to control cytokine storm.


COVID-19 , Endoribonucleases , Humans , Cytokines , Endoribonucleases/genetics , Endoribonucleases/metabolism , Lung/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/genetics , RNA, Viral , SARS-CoV-2/genetics , Toll-Like Receptor 8/genetics , X-Box Binding Protein 1/genetics , X-Box Binding Protein 1/metabolism
17.
Expert Opin Ther Targets ; 27(12): 1207-1215, 2023.
Article En | MEDLINE | ID: mdl-38078890

INTRODUCTION: Despite improvements in clinical management of hepatocellular carcinoma (HCC), prognosis remains poor with a 5-year survival rate less than 40%. Drug resistance in HCC makes it challenging to treat; therefore, it is imperative to develop new therapeutic strategies. Higher expression of X-box binding protein 1 (XBP1) in tumor cells is highly correlated with poor prognosis. In tumor cells, XBP1 modulates the unfolded protein response (UPR) to restore homeostasis in endoplasmic reticulum. Targeting XBP1 could be a promising therapeutic strategy to overcome HCC resistance and improve the survival rate of patients. AREAS COVERED: This review provides the recent evidence that indicates XBP1 is involved in HCC drug resistance via DNA damage response, drug inactivation, and inhibition of apoptosis. In addition, the potential roles of XBP1 in inducing resistance in HCC cells were highlighted, and we showed how its inhibition could sensitize tumor cells to controlled cell death. EXPERT OPINION: Due to the diversity in molecular mechanism of multidrug-resistance, targeting one specific pathway is inadequate. XBP1 inhibition could be a potential therapeutic target to overcome verity of resistance mechanisms. The main function of this transcription factor in HCC treatment response is an attractive area for further studies and should be discussed more.


Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , DNA-Binding Proteins/genetics , X-Box Binding Protein 1/genetics , X-Box Binding Protein 1/metabolism , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Unfolded Protein Response , Drug Resistance , Endoplasmic Reticulum Stress
18.
Exp Mol Med ; 55(11): 2376-2389, 2023 11.
Article En | MEDLINE | ID: mdl-37907740

Osteoarthritis (OA) is a full-joint, multifactorial, degenerative and inflammatory disease that seriously affects the quality of life of patients due to its disabling and pain-causing properties. ER stress has been reported to be closely related to the progression of OA. The inositol-requiring enzyme 1α/X-box-binding protein-1 spliced (IRE1α/XBP1s) pathway, which is highly expressed in the chondrocytes of OA patients, promotes the degradation and refolding of abnormal proteins during ER stress and maintains the stability of the ER environment of chondrocytes, but its function and the underlying mechanisms of how it contributes to the progression of OA remain unclear. This study investigates the role of IRE1α/ERN1 in OA. Specific deficiency of ERN1 in chondrocytes spontaneously resulted in OA-like cartilage destruction and accelerated OA progression in a surgically induced arthritis model. Local delivery of AdERN1 relieved degradation of the cartilage matrix and prevented OA development in an ACLT-mediated model. Mechanistically, progranulin (PGRN), an intracellular chaperone, binds to IRE1α, promoting its phosphorylation and splicing of XBP1u to generate XBP1s. XBP1s protects articular cartilage through TNF-α/ERK1/2 signaling and further maintains collagen homeostasis by regulating type II collagen expression. The chondroprotective effect of IRE1α/ERN1 is dependent on PGRN and XBP1s splicing. ERN1 deficiency accelerated cartilage degeneration in OA by reducing PGRN expression and XBP1s splicing, subsequently decreasing collagen II expression and triggering collagen structural abnormalities and an imbalance in collagen homeostasis. This study provides new insights into OA pathogenesis and the UPR and suggests that IRE1α/ERN1 may serve as a potential target for the treatment of joint degenerative diseases, including OA.


Cartilage, Articular , Osteoarthritis , Humans , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Progranulins/metabolism , Endoribonucleases/genetics , Endoribonucleases/metabolism , Quality of Life , Osteoarthritis/metabolism , Chondrocytes/metabolism , Cartilage, Articular/metabolism , Collagen/metabolism , Homeostasis , X-Box Binding Protein 1/genetics , X-Box Binding Protein 1/metabolism
19.
Invest Ophthalmol Vis Sci ; 64(14): 40, 2023 Nov 01.
Article En | MEDLINE | ID: mdl-38015176

Purpose: Diabetic retinopathy (DR) is a leading cause of blindness in working-age adults characterized by retinal dysfunction and neurovascular degeneration. We previously reported that deletion of X-box binding protein 1 (XBP1) leads to accelerated retinal neurodegeneration in diabetes; however, the mechanisms remain elusive. The goal of this study is to determine the role of XBP1 in the regulation of photoreceptor synaptic integrity in early DR. Methods: Diabetes was induced by streptozotocin in retina-specific XBP1 conditional knockout (cKO) or wild-type (WT) mice to generate diabetic cKO (cKO/DM) or WT/DM mice for comparison with nondiabetic cKO (cKO/NDM) and WT/NDM mice. Retinal morphology, structure, and function were assessed by immunohistochemistry, optical coherence tomography, and electroretinogram (ERG) after 3 months of diabetes. The synapses between photoreceptors and bipolar cells were examined by confocal microscopy, and synaptic integrity was quantified using the QUANTOS algorithm. Results: We found a thinning of the outer nuclear layer and a decline in the b-wave amplitude in dark- and light-adapted ERG in cKO/DM mice compared to all other groups. In line with these changes, cKO mice showed increased loss of synaptic integrity compared to WT mice, regardless of diabetes status. In searching for candidate molecules responsible for the loss of photoreceptor synaptic integrity in diabetic and XBP1-deficient retinas, we found decreased mRNA and protein levels of DLG4/PSD-95 in cKO/DM retina compared to WT/DM. Conclusions: These findings suggest that XBP1 is a crucial regulator in maintaining synaptic integrity and retinal function, possibly through regulation of synaptic scaffold proteins.


Diabetes Mellitus , Diabetic Retinopathy , X-Box Binding Protein 1 , Animals , Mice , Algorithms , Diabetic Retinopathy/genetics , Electroretinography , Retina , X-Box Binding Protein 1/genetics
20.
J Clin Invest ; 133(24)2023 Dec 15.
Article En | MEDLINE | ID: mdl-37874641

Heart failure with preserved ejection fraction (HFpEF) is a widespread syndrome with limited therapeutic options and poorly understood immune pathophysiology. Using a 2-hit preclinical model of cardiometabolic HFpEF that induces obesity and hypertension, we found that cardiac T cell infiltration and lymphoid expansion occurred concomitantly with cardiac pathology and that diastolic dysfunction, cardiomyocyte hypertrophy, and cardiac phospholamban phosphorylation were T cell dependent. Heart-infiltrating T cells were not restricted to cardiac antigens and were uniquely characterized by impaired activation of the inositol-requiring enzyme 1α/X-box-binding protein 1 (IRE1α/XBP1) arm of the unfolded protein response. Notably, selective ablation of XBP1 in T cells enhanced their persistence in the heart and lymphoid organs of mice with preclinical HFpEF. Furthermore, T cell IRE1α/XBP1 activation was restored after withdrawal of the 2 comorbidities inducing HFpEF, resulting in partial improvement of cardiac pathology. Our results demonstrated that diastolic dysfunction and cardiomyocyte hypertrophy in preclinical HFpEF were T cell dependent and that reversible dysregulation of the T cell IRE1α/XBP1 axis was a T cell signature of HFpEF.


Cardiomyopathies , Heart Failure , Animals , Mice , Cardiomyopathies/genetics , Cardiomyopathies/metabolism , Endoribonucleases/genetics , Endoribonucleases/metabolism , Heart Failure/metabolism , Hypertrophy , Inflammation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Stroke Volume/physiology , T-Lymphocytes/pathology , X-Box Binding Protein 1/genetics , X-Box Binding Protein 1/metabolism
...