We aimed to investigate the potential pro-oncogenic properties of GREM1 by Pan-cancer multi-omics analysis. Accumulating evidence has highlighted that GREM1 (Gremlin 1), serves as an inhibitor of BMP (Bone Morphogenetic Protein) family, involve in bone related diseases, carcinogenesis, cell stemness, and cell differentiation. However, the effect and underlying mechanism of GREM1 on the cancer biology remain largely elusive. The mRNA expression of GREM1 were extracted from GTEx (Genotype-Tissue Expression) and TCGA (The Cancer Genome Atlas) database. Analysis of OS (Overall Survival), PFI (Progression Free Interval), DSS (Disease-Specific Survival), and ROC (Receiver Operating Characteristic) were performed to predicted prognostic value of GREM1 in various cancers. The TIMER (Tumor Immune Estimation Resource) online tool was used to investigate the relationship between GREM1 transcriptional level and infiltration of immune cells. KEGG (Kyoto Encyclopedia of Genes and Genomes) analysis and GO (Gene Ontology) analysis were used to investigate the GREM1 related molecular events, and then constructed a PPI (Protein-Protein Interaction) network via the STRING (Search Tool for the Retrieval of Interaction Genes/Proteins) online tool. Western blot was performed to investigate the indicated protein expression. In the present study, our results showed that GREM1 tended to be upregulated in various cancers, which would correlate with the poor prognosis. Mechanistically, our results showed that GREM1 involve in regulating the ECM-receptor interaction pathway, upregulation of MMP activity, angiogenesis, and immune cell infiltration. In vitro studies, our results further showed that BMP agonist significantly decreased the protein level of GREM1 in GES-1 cells and BGC cells, which accompanied by inhibiting migration and proliferation in GES-1 cells and BGC cells. BMP inhibitor significantly promoted GREM1 expression and migration in BGC cells, but not GES-1 cells. GREM1 might serve as a potential and promising prognostic biomarker for drug development and cancer treatment.
我们的目的是通过泛癌症多组学分析来研究GREM1潜在的促癌特性。越来越多的证据表明,GREM1 (Gremlin 1)作为骨形态发生蛋白(BMP)家族的抑制剂,参与骨相关疾病、癌变、细胞干性和细胞分化。然而,GREM1在癌症生物学中的作用和潜在机制在很大程度上仍然是未知的。从GTEx (Genotype-Tissue expression)和TCGA (The Cancer Genome Atlas)数据库中提取GREM1的mRNA表达。通过分析总生存期(OS)、无进展间期(PFI)、疾病特异性生存期(DSS)和受试者工作特征(ROC)来预测GREM1在各种癌症中的预后价值。利用TIMER (Tumor Immune Estimation Resource)在线工具研究GREM1转录水平与免疫细胞浸润的关系。通过KEGG (Kyoto Encyclopedia of Genes and Genomes)分析和GO (Gene Ontology)分析对GREM1相关的分子事件进行分析,并通过STRING (Search Tool for Retrieval of interacting Genes/Proteins)在线工具构建PPI (Protein-Protein Interaction)网络。Western blot检测指示蛋白的表达。在本研究中,我们的研究结果表明,GREM1在各种癌症中都有上调的趋势,这可能与预后不良有关。在机制上,我们的研究结果表明,GREM1参与调节ecm受体相互作用途径,上调MMP活性,血管生成和免疫细胞浸润。在体外研究中,我们的研究结果进一步表明,BMP激动剂显著降低了GES-1细胞和BGC细胞中GREM1的蛋白水平,同时抑制了GES-1细胞和BGC细胞的迁移和增殖。BMP抑制剂能显著促进BGC细胞中GREM1的表达和迁移,而对GES-1细胞无显著影响。GREM1可能作为药物开发和癌症治疗的潜在和有前途的预后生物标志物。
{"title":"Pan-cancer multi-omics analysis to identify the potential pro-oncogenic properties of GREM1 as a promising targets for cancer prognosis and therapeutics.","authors":"Menglu Zhu, Hengli Zhou, Yue Zhuo, Changhua Liu, Jiaxin Li, Peiyao He, Naihua Liu, Ziming Zhao, Pan Huafeng","doi":"10.1177/03946320251331850","DOIUrl":"https://doi.org/10.1177/03946320251331850","url":null,"abstract":"<p><p>We aimed to investigate the potential pro-oncogenic properties of GREM1 by Pan-cancer multi-omics analysis. Accumulating evidence has highlighted that GREM1 (Gremlin 1), serves as an inhibitor of BMP (Bone Morphogenetic Protein) family, involve in bone related diseases, carcinogenesis, cell stemness, and cell differentiation. However, the effect and underlying mechanism of GREM1 on the cancer biology remain largely elusive. The mRNA expression of GREM1 were extracted from GTEx (Genotype-Tissue Expression) and TCGA (The Cancer Genome Atlas) database. Analysis of OS (Overall Survival), PFI (Progression Free Interval), DSS (Disease-Specific Survival), and ROC (Receiver Operating Characteristic) were performed to predicted prognostic value of GREM1 in various cancers. The TIMER (Tumor Immune Estimation Resource) online tool was used to investigate the relationship between GREM1 transcriptional level and infiltration of immune cells. KEGG (Kyoto Encyclopedia of Genes and Genomes) analysis and GO (Gene Ontology) analysis were used to investigate the GREM1 related molecular events, and then constructed a PPI (Protein-Protein Interaction) network via the STRING (Search Tool for the Retrieval of Interaction Genes/Proteins) online tool. Western blot was performed to investigate the indicated protein expression. In the present study, our results showed that GREM1 tended to be upregulated in various cancers, which would correlate with the poor prognosis. Mechanistically, our results showed that GREM1 involve in regulating the ECM-receptor interaction pathway, upregulation of MMP activity, angiogenesis, and immune cell infiltration. In vitro studies, our results further showed that BMP agonist significantly decreased the protein level of GREM1 in GES-1 cells and BGC cells, which accompanied by inhibiting migration and proliferation in GES-1 cells and BGC cells. BMP inhibitor significantly promoted GREM1 expression and migration in BGC cells, but not GES-1 cells. GREM1 might serve as a potential and promising prognostic biomarker for drug development and cancer treatment.</p>","PeriodicalId":48647,"journal":{"name":"International Journal of Immunopathology and Pharmacology","volume":"39 ","pages":"3946320251331850"},"PeriodicalIF":3.5,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12033649/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144035565","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Objective: This study aims to develop a prognostic model for HCC based on TME-related factors.
Introduction: Hepatocellular carcinoma (HCC) is characterized by a poor prognosis, largely due to the complex and heterogeneous interactions between stromal and immune cells within the tumor microenvironment (TME).
Methods: Genome and transcriptome data, as well as clinical information of HCC patients, were obtained from the Cancer Genome Atlas (TCGA) and the Gene Expression Omnibus (GEO). The TME score was evaluated using the "ESTIMATE" R package. Differentially expressed genes (DEGs) associated with TME phenotype were analyzed using the LIMMA R-package. Survival outcomes were compared using Kaplan-Meier curves with log-rank test and Cox proportional hazards model. Protein-Protein Interaction (PPI) networks integrated with multivariate survival and LASSO analyses were utilized to identify TME-related hub genes for a risk score model. A nomogram predicting prognosis of HCC patients was developed through four independent cohorts.
Results: The TME scores showed a negative correlation with tumor progression and survival in HCC patients. We identified 50 core genes with high connectivity in the PPI network, as along with 33 key DEGs associated with survival in HCC. Intersection analysis revealed six hub genes -CXCL8, CXCL1, CCR7, IL7R, MMP9, and CD69. The risk score based on these six TME-related hub genes was significantly associated with overall survival and clinicopathological characteristics of HCC patients. Furthermore, the nomogram demonstrated its ability to discriminate HCC patients from healthy individuals using peripheral blood mononuclear cells.
Conclusion: We have developed a TME-related risk scoring model for HCC patients and identified six hub gene panel that serve as a potential biomarker for personalized prognosis of immunotherapy and non-invasive diagnostics of HCC.
{"title":"Construction of a tumor immune microenvironment-related risk scoring model for prognosis of hepatocellular carcinoma.","authors":"Xinyi Li, Zifan Qin, Haozhi Chen, Daichuan Chen, Nafisa Alimu, Duoduo Li, Xiyu Cheng, Qiong Yan, Lishu Zhang, Xingwei Liu, Zitong Zhou, Jiayi Zhu, Hangqi Ma, Xinyue Pei, Hanli Xu, Jiaqiang Huang","doi":"10.1177/03946320251333975","DOIUrl":"https://doi.org/10.1177/03946320251333975","url":null,"abstract":"<p><strong>Objective: </strong>This study aims to develop a prognostic model for HCC based on TME-related factors.</p><p><strong>Introduction: </strong>Hepatocellular carcinoma (HCC) is characterized by a poor prognosis, largely due to the complex and heterogeneous interactions between stromal and immune cells within the tumor microenvironment (TME).</p><p><strong>Methods: </strong>Genome and transcriptome data, as well as clinical information of HCC patients, were obtained from the Cancer Genome Atlas (TCGA) and the Gene Expression Omnibus (GEO). The TME score was evaluated using the \"ESTIMATE\" R package. Differentially expressed genes (DEGs) associated with TME phenotype were analyzed using the LIMMA R-package. Survival outcomes were compared using Kaplan-Meier curves with log-rank test and Cox proportional hazards model. Protein-Protein Interaction (PPI) networks integrated with multivariate survival and LASSO analyses were utilized to identify TME-related hub genes for a risk score model. A nomogram predicting prognosis of HCC patients was developed through four independent cohorts.</p><p><strong>Results: </strong>The TME scores showed a negative correlation with tumor progression and survival in HCC patients. We identified 50 core genes with high connectivity in the PPI network, as along with 33 key DEGs associated with survival in HCC. Intersection analysis revealed six hub genes -<i>CXCL8</i>, <i>CXCL1</i>, <i>CCR7</i>, <i>IL7R</i>, <i>MMP9</i>, and <i>CD69</i>. The risk score based on these six TME-related hub genes was significantly associated with overall survival and clinicopathological characteristics of HCC patients. Furthermore, the nomogram demonstrated its ability to discriminate HCC patients from healthy individuals using peripheral blood mononuclear cells.</p><p><strong>Conclusion: </strong>We have developed a TME-related risk scoring model for HCC patients and identified six hub gene panel that serve as a potential biomarker for personalized prognosis of immunotherapy and non-invasive diagnostics of HCC.</p>","PeriodicalId":48647,"journal":{"name":"International Journal of Immunopathology and Pharmacology","volume":"39 ","pages":"3946320251333975"},"PeriodicalIF":3.5,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12035210/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144042658","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-01Epub Date: 2025-03-20DOI: 10.1177/03946320251321083
{"title":"Thanks to Reviewers.","authors":"","doi":"10.1177/03946320251321083","DOIUrl":"10.1177/03946320251321083","url":null,"abstract":"","PeriodicalId":48647,"journal":{"name":"International Journal of Immunopathology and Pharmacology","volume":"39 ","pages":"3946320251321083"},"PeriodicalIF":3.5,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11926831/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143781652","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-01Epub Date: 2025-04-11DOI: 10.1177/03946320251327621
Yuanda Liu, Lanlan Chen, Wei Hao, Kun Zhao, Changfeng Li
Explore the causal relationship of risk between type 1 diabetes and primary biliary cholangitis (PBC) and primary sclerosing cholangitis (PSC). A causal association between type 1 diabetes and autoimmune liver disease remains ambiguous. This study explored potential causality between different autoimmune conditions, indicating that caution should be taken of the occurrence of autoimmune liver diseases in daily management of T1D patients. Genetic variants were extracted as instrumental variables from the genome-wide association study (GWAS) of PBC, PSC, type 1 diabetes (T1D), and type 2 diabetes (T2D). Associations between four primary liver enzymes, alkaline phosphatase (ALP), aspartate aminotransferase (AST), alanine transferase (ALT), and glutamyl transaminase (GGT), and blood glucose-related indicators such as 2h-glucose post-challenge (2hGlu), fasting glucose (FG), fasting insulin (FI), and glycated hemoglobin (HbA1c) were also evaluated (GWAS p-value < 5 × 10-8). A bi-directional two-sample Mendelian randomization (MR) design was used to assess causality between type 1 diabetes and autoimmune cholestasis. Genetic susceptibility to T1D increased the risk of PSC and PBC. Genetic susceptibility to T2D reduced the risk of PSC and showed no correlation with PBC. Genetically susceptibility to PBC increased the risk of T1D and showed no correlation with T2D. Genetically susceptibility to PSC did not impact the risk of T1D and T2D. T1D patients have an increased risk of PBC/PSC, but such causation is not mediated or explained by the altered blood glucose levels. A bi-directional causal association was identified between type 1 diabetes and autoimmune cholestasis. The findings provide new insight into the management of patients with these conditions.
{"title":"Causal association between type 1 diabetes and autoimmune cholestasis: A bi-directional Mendelian randomized study.","authors":"Yuanda Liu, Lanlan Chen, Wei Hao, Kun Zhao, Changfeng Li","doi":"10.1177/03946320251327621","DOIUrl":"https://doi.org/10.1177/03946320251327621","url":null,"abstract":"<p><p>Explore the causal relationship of risk between type 1 diabetes and primary biliary cholangitis (PBC) and primary sclerosing cholangitis (PSC). A causal association between type 1 diabetes and autoimmune liver disease remains ambiguous. This study explored potential causality between different autoimmune conditions, indicating that caution should be taken of the occurrence of autoimmune liver diseases in daily management of T1D patients. Genetic variants were extracted as instrumental variables from the genome-wide association study (GWAS) of PBC, PSC, type 1 diabetes (T1D), and type 2 diabetes (T2D). Associations between four primary liver enzymes, alkaline phosphatase (ALP), aspartate aminotransferase (AST), alanine transferase (ALT), and glutamyl transaminase (GGT), and blood glucose-related indicators such as 2h-glucose post-challenge (2hGlu), fasting glucose (FG), fasting insulin (FI), and glycated hemoglobin (HbA1c) were also evaluated (GWAS p-value < 5 × 10<sup>-8</sup>). A bi-directional two-sample Mendelian randomization (MR) design was used to assess causality between type 1 diabetes and autoimmune cholestasis. Genetic susceptibility to T1D increased the risk of PSC and PBC. Genetic susceptibility to T2D reduced the risk of PSC and showed no correlation with PBC. Genetically susceptibility to PBC increased the risk of T1D and showed no correlation with T2D. Genetically susceptibility to PSC did not impact the risk of T1D and T2D. T1D patients have an increased risk of PBC/PSC, but such causation is not mediated or explained by the altered blood glucose levels. A bi-directional causal association was identified between type 1 diabetes and autoimmune cholestasis. The findings provide new insight into the management of patients with these conditions.</p>","PeriodicalId":48647,"journal":{"name":"International Journal of Immunopathology and Pharmacology","volume":"39 ","pages":"3946320251327621"},"PeriodicalIF":3.5,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12033451/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144026506","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-01Epub Date: 2025-04-15DOI: 10.1177/03946320251326699
Marina Markovic, Danijela Niciforovic, Violeta Mladenovic, Dragica Pavlovic, Dragana Papic, Katarina Milojevic, Dalibor Jovanovic, Marija Spasojevic, Rade Milic
Immune Checkpoint Inhibitors (ICIs) are monoclonal antibodies that block inhibitory immune targets, such as cytotoxic T lymphocyte antigen 4 (CTLA-4), programmed cell death protein 1 (PD-1), and programmed death ligand 1 (PD-L). Pembrolizumab targets the PD-1 receptor of lymphocytes in lung cancer treatment. ICI checkpoint blockade enhances immunity against cancer cells. However, loss of immunoregulatory control can cause autoimmune reactions in various organs, leading to immune-related adverse events (irAEs). The most common irAE is ICIs-induced colitis, which usually develops 6-8 weeks after ICI initiation and can involve any part of the gastrointestinal system. Herein, we report a presentation of pembrolizumab-induced colitis in a female patient with metastatic lung cancer and review the most recent findings in the model of checkpoint-induced colitis. It was interesting to learn that the colon mucosa may show normal macroscopic findings, but microscopically, immunotherapy-induced autoimmune colitis could be present. Additionally, patients with grade 2 or higher symptoms should have a colonoscopy, receive systemic corticosteroids as treatment, and, based on their response, receive biologic therapy. Here, we present a case report of in a 45-year-old female who has been a smoker for 25 years, without comorbidities, and with metastatic lung cancer who developed colitis after the seventh cycle of pembrolizumab. This case presentation highlights the importance of early recognition and appropriate intervention in order to prevent permanent interruption of treatment with checkpoint inhibitors, as well as prevention of colitis complications.
{"title":"Immune-related adverse events-pembrolizumab-induced colitis-the importance of early diagnosis and treatment: A case report and review of the literature.","authors":"Marina Markovic, Danijela Niciforovic, Violeta Mladenovic, Dragica Pavlovic, Dragana Papic, Katarina Milojevic, Dalibor Jovanovic, Marija Spasojevic, Rade Milic","doi":"10.1177/03946320251326699","DOIUrl":"https://doi.org/10.1177/03946320251326699","url":null,"abstract":"<p><p>Immune Checkpoint Inhibitors (ICIs) are monoclonal antibodies that block inhibitory immune targets, such as cytotoxic T lymphocyte antigen 4 (CTLA-4), programmed cell death protein 1 (PD-1), and programmed death ligand 1 (PD-L). Pembrolizumab targets the PD-1 receptor of lymphocytes in lung cancer treatment. ICI checkpoint blockade enhances immunity against cancer cells. However, loss of immunoregulatory control can cause autoimmune reactions in various organs, leading to immune-related adverse events (irAEs). The most common irAE is ICIs-induced colitis, which usually develops 6-8 weeks after ICI initiation and can involve any part of the gastrointestinal system. Herein, we report a presentation of pembrolizumab-induced colitis in a female patient with metastatic lung cancer and review the most recent findings in the model of checkpoint-induced colitis. It was interesting to learn that the colon mucosa may show normal macroscopic findings, but microscopically, immunotherapy-induced autoimmune colitis could be present. Additionally, patients with grade 2 or higher symptoms should have a colonoscopy, receive systemic corticosteroids as treatment, and, based on their response, receive biologic therapy. Here, we present a case report of in a 45-year-old female who has been a smoker for 25 years, without comorbidities, and with metastatic lung cancer who developed colitis after the seventh cycle of pembrolizumab. This case presentation highlights the importance of early recognition and appropriate intervention in order to prevent permanent interruption of treatment with checkpoint inhibitors, as well as prevention of colitis complications.</p>","PeriodicalId":48647,"journal":{"name":"International Journal of Immunopathology and Pharmacology","volume":"39 ","pages":"3946320251326699"},"PeriodicalIF":3.5,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12033556/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144005021","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-01DOI: 10.1177/03946320251315036
Sara I Taha, Sara F Samaan, Sally Saber Hawash, Eman M El-Sehsah, Sara Shamloul, Dalia Hussein Helmy Elsheikh, Basim Othman, Mohammad A Albanghali, Saeedah H Aljadani, Abdalla Elmanna, Rasha Ahmed Ghorab
Cell-free DNA (cfDNA) has emerged as a potential biomarker for assessing disease activity and prognosis in rheumatoid arthritis (RA). However, the association between cfDNA levels and the established RA markers of inflammation and disease severity remains unclear. The current study aimed to detect plasma levels of cfDNA in patients with RA and to investigate their association with RA activity indicators (erythrocyte sedimentation rate (ESR), C-reactive protein (CRP), disease activity score-28 (DAS28)), prognostic markers (rheumatoid factor (RF), anticitrullinated protein antibodies (ACPA)), and the musculoskeletal ultrasonographic (US7) scores. This controlled cross-sectional study included 108 RA patients and 108 healthy controls. Plasma levels of cfDNA were quantified by real-time PCR using ALU repeats. Levels of ESR, CRP, RF, and ACPA were measured using routine laboratory assays. Synovial inflammation and joint damage evaluation was performed using the US7 scoring system. Plasma levels of cfDNA were higher in RA patients than controls (P < 0.001) and significantly increased with higher DAS28 scores among all RA activity groups. Also, cfDNA levels were significantly positively correlated with ESR, CRP, RF, and ACPA levels (P-values <0.001). Regarding US7, cfDNA was significantly positively correlated with synovitis and erosion scores (P-values <0.05) but did not correlate significantly with tenosynovitis scores (P-values >0.05). In addition, plasma cfDNA was significantly higher in seropositive RA patients than in seronegative patients (P = 0.007). The odds ratio for cfDNA as a risk factor for erosions was 2.254. This study revealed that cfDNA levels are elevated in RA patients and positively associated with disease activity indicators and prognosis markers. Further research is warranted to validate these findings in larger cohorts and explore the clinical implications of cfDNA measurement in RA management.
{"title":"Circulating cell-free DNA as a potential biomarker for prediction of disease activity and prognosis among Egyptian rheumatoid arthritis patients.","authors":"Sara I Taha, Sara F Samaan, Sally Saber Hawash, Eman M El-Sehsah, Sara Shamloul, Dalia Hussein Helmy Elsheikh, Basim Othman, Mohammad A Albanghali, Saeedah H Aljadani, Abdalla Elmanna, Rasha Ahmed Ghorab","doi":"10.1177/03946320251315036","DOIUrl":"10.1177/03946320251315036","url":null,"abstract":"<p><p>Cell-free DNA (cfDNA) has emerged as a potential biomarker for assessing disease activity and prognosis in rheumatoid arthritis (RA). However, the association between cfDNA levels and the established RA markers of inflammation and disease severity remains unclear. The current study aimed to detect plasma levels of cfDNA in patients with RA and to investigate their association with RA activity indicators (erythrocyte sedimentation rate (ESR), C-reactive protein (CRP), disease activity score-28 (DAS28)), prognostic markers (rheumatoid factor (RF), anticitrullinated protein antibodies (ACPA)), and the musculoskeletal ultrasonographic (US7) scores. This controlled cross-sectional study included 108 RA patients and 108 healthy controls. Plasma levels of cfDNA were quantified by real-time PCR using ALU repeats. Levels of ESR, CRP, RF, and ACPA were measured using routine laboratory assays. Synovial inflammation and joint damage evaluation was performed using the US7 scoring system. Plasma levels of cfDNA were higher in RA patients than controls (<i>P</i> < 0.001) and significantly increased with higher DAS28 scores among all RA activity groups. Also, cfDNA levels were significantly positively correlated with ESR, CRP, RF, and ACPA levels (<i>P-</i>values <0.001). Regarding US7, cfDNA was significantly positively correlated with synovitis and erosion scores (<i>P-</i>values <0.05) but did not correlate significantly with tenosynovitis scores (<i>P-</i>values >0.05). In addition, plasma cfDNA was significantly higher in seropositive RA patients than in seronegative patients (<i>P</i> = 0.007). The odds ratio for cfDNA as a risk factor for erosions was 2.254. This study revealed that cfDNA levels are elevated in RA patients and positively associated with disease activity indicators and prognosis markers. Further research is warranted to validate these findings in larger cohorts and explore the clinical implications of cfDNA measurement in RA management.</p>","PeriodicalId":48647,"journal":{"name":"International Journal of Immunopathology and Pharmacology","volume":"39 ","pages":"3946320251315036"},"PeriodicalIF":3.5,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11786270/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143076006","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Objective: To study the role and underlying mechanisms of dihydromyricetin on the myocardial function in mice induced by CCl4.
Methods: Eighteen C57BL/6 mice (6-8 W, female) were randomly divided into the following three groups: control group, CCl4-induced positive group (CCl4 group), dihydromyricetin group, six mice/group. NLRP3-deficient (NLRP3-/-) C57BL/6 mice used the same age, gender, and modeling method. The HL-1 cells were used for in vitro experiments. The HL-1 cells were treated with PBS, CCl4, and CCl4 + DMY respectively.
Results: The RT-qPCR results showed that compared to the mice induced by CCl4, the dihydromyricetin increased the Arg-1 mRNA level in the mouse myocardial tissues. The mRNA expressions of the iNOS, IL-33, and ST2 were reduced by the dihydromyricetin. The results of immunohistochemistry showed that dihydromyricetin decreased IL-33 protein expressions in the myocardial tissues. Western blot results also showed that compared with the control group, the activation of NLRP3 inflammasomes in the myocardial tissues of mice injured by CCl4 was increased, and dihydromyricetin can reduce NLRP3 inflammasomes activation in the myocardial tissues induced by CCl4. The results of ELISA showed that dihydromyricetin could reduce the IL-1β level in the serum of the mice induced by CCl4. Consistent with the in vivo results, compared with the control group, the NLRP3 inflammasome activation and IL-33/ST2 expression were increased in the CCl4-treated HL-1 cells, while DMY significantly weakened this effect. Interestingly, NLRP3 deficiency enhanced the protective effect of DMY on myocardial function in mice.
Conclusions: IL-33/ST2 signaling pathways and NLRP3 inflammasome activation may be involved in dihydromyricetin improving the myocardial function of the mice induced by CCl4.
{"title":"Dihydromyricetin improving myocardial function in the mice induced by CCl<sub>4</sub>.","authors":"Wen-Juan Zhang, Ke-Yun Li, Le-Ying Lin, Tao Song, Heng Hu, Yi-Man Song, Zi-Qing Xiao, Jiang-Rui Zhu, Li-Tao Long, Gao-Lu Cao, Bin-Hong Huang","doi":"10.1177/03946320251317397","DOIUrl":"10.1177/03946320251317397","url":null,"abstract":"<p><strong>Objective: </strong>To study the role and underlying mechanisms of dihydromyricetin on the myocardial function in mice induced by CCl<sub>4</sub>.</p><p><strong>Methods: </strong>Eighteen C57BL/6 mice (6-8 W, female) were randomly divided into the following three groups: control group, CCl<sub>4</sub>-induced positive group (CCl<sub>4</sub> group), dihydromyricetin group, six mice/group. NLRP3-deficient (NLRP3<sup>-/-</sup>) C57BL/6 mice used the same age, gender, and modeling method. The HL-1 cells were used for in vitro experiments. The HL-1 cells were treated with PBS, CCl<sub>4</sub>, and CCl<sub>4</sub> + DMY respectively.</p><p><strong>Results: </strong>The RT-qPCR results showed that compared to the mice induced by CCl<sub>4</sub>, the dihydromyricetin increased the Arg-1 mRNA level in the mouse myocardial tissues. The mRNA expressions of the iNOS, IL-33, and ST2 were reduced by the dihydromyricetin. The results of immunohistochemistry showed that dihydromyricetin decreased IL-33 protein expressions in the myocardial tissues. Western blot results also showed that compared with the control group, the activation of NLRP3 inflammasomes in the myocardial tissues of mice injured by CCl<sub>4</sub> was increased, and dihydromyricetin can reduce NLRP3 inflammasomes activation in the myocardial tissues induced by CCl<sub>4</sub>. The results of ELISA showed that dihydromyricetin could reduce the IL-1β level in the serum of the mice induced by CCl<sub>4.</sub> Consistent with the in vivo results, compared with the control group, the NLRP3 inflammasome activation and IL-33/ST2 expression were increased in the CCl<sub>4</sub>-treated HL-1 cells, while DMY significantly weakened this effect. Interestingly, NLRP3 deficiency enhanced the protective effect of DMY on myocardial function in mice.</p><p><strong>Conclusions: </strong>IL-33/ST2 signaling pathways and NLRP3 inflammasome activation may be involved in dihydromyricetin improving the myocardial function of the mice induced by CCl<sub>4</sub>.</p>","PeriodicalId":48647,"journal":{"name":"International Journal of Immunopathology and Pharmacology","volume":"39 ","pages":"3946320251317397"},"PeriodicalIF":3.5,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12033494/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143634863","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Objective: The aim of this study was to investigate the effects of halofuginone (HF) on gastric cancer cells and whether the combination of HF and trametinib has synergistic effects.
Introduction: Halofuginone, a natural small molecule isolated from the plant Dichroa febrifuga, has been found to have anticancer activity in a variety of cancers, but few studies on HF in gastric cancer.
Methods: cell viability was performed using the CellTiterGlo assay and apoptosis and cell cycle analysis was performed by Annexin V-FITC staining and PI staining. We also analyzed RNA sequencing and differentially expressed genes was shown using the heatmap. Western blot and qPCR were carried out to determine the expression of pro-apoptotic and anti-apoptotic proteins.
Results: HF inhibited proliferation and induced apoptosis in gastric cancer cells in a dose-dependent manner. HF induced the expression of p-ERK in gastric cancer cells. HF and trametinib synergistically inhibited the gastric cancer cell proliferation. Trametinib inhibited HF-induced p-ERK expression. HF reduced anti-apoptotic protein Mcl-1 expression and reduced trametinib-induced upregulation of Mcl-1 expression.
Conclusion: HF exerts its anti-cancer effects in gastric cancer and has a synergistic inhibition with trametinib, which may provide a novel therapeutic strategy for gastric cancer.
{"title":"Halofuginone induces ERK phosphorylation and synergizes with trametinib in gastric cancer cells.","authors":"Yuemin Sun, Peng Wang, Xiaofeng Bai, Liang Tian, Yuxin Zhong","doi":"10.1177/03946320251359860","DOIUrl":"10.1177/03946320251359860","url":null,"abstract":"<p><strong>Objective: </strong>The aim of this study was to investigate the effects of halofuginone (HF) on gastric cancer cells and whether the combination of HF and trametinib has synergistic effects.</p><p><strong>Introduction: </strong>Halofuginone, a natural small molecule isolated from the plant <i>Dichroa febrifuga</i>, has been found to have anticancer activity in a variety of cancers, but few studies on HF in gastric cancer.</p><p><strong>Methods: </strong>cell viability was performed using the CellTiterGlo assay and apoptosis and cell cycle analysis was performed by Annexin V-FITC staining and PI staining. We also analyzed RNA sequencing and differentially expressed genes was shown using the heatmap. Western blot and qPCR were carried out to determine the expression of pro-apoptotic and anti-apoptotic proteins.</p><p><strong>Results: </strong>HF inhibited proliferation and induced apoptosis in gastric cancer cells in a dose-dependent manner. HF induced the expression of p-ERK in gastric cancer cells. HF and trametinib synergistically inhibited the gastric cancer cell proliferation. Trametinib inhibited HF-induced p-ERK expression. HF reduced anti-apoptotic protein Mcl-1 expression and reduced trametinib-induced upregulation of Mcl-1 expression.</p><p><strong>Conclusion: </strong>HF exerts its anti-cancer effects in gastric cancer and has a synergistic inhibition with trametinib, which may provide a novel therapeutic strategy for gastric cancer.</p>","PeriodicalId":48647,"journal":{"name":"International Journal of Immunopathology and Pharmacology","volume":"39 ","pages":"3946320251359860"},"PeriodicalIF":2.6,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12340186/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144817993","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-01Epub Date: 2025-04-09DOI: 10.1177/03946320251331873
Haidy A Abdullah, Fatma Sm Moawed, Esraa Sa Ahmed, Fatma F Abdel Hamid, Riham Abdel-Hamid Haroun
Excess iron has been associated with cardiovascular diseases. Flavonoids are antioxidants and cardioprotectants. Therefore, the goal of the current study is to evaluate the anti-apoptotic, antioxidant, and iron-chelating qualities of two flavonoids, rutin (R) and hesperidin (H), as well as their potential to prevent induced ferroptosis in rats. It is an in vivo cross-sectional study, in which rats were divided into 12 groups; control, H, R, H + R, Fe, Fe + IR, Fe + IR + Ref, Fe + H, Fe + IR + H, Fe + R, Fe + IR + R and Fe + IR + H + R. Cardiac and serum iron levels, serum troponin I, creatine kinase-MB (CK-MB), total iron binding capacity (TIBC), transferrin, ferritin, and hepicidin were determined. Moreover, the levels of malondialdehyde (MDA), nitric oxide (NO) and glutathione (GSH) and the activities of superoxide dismutase (SOD) and glutathione peroxidase (GPx), were also determined. The expression levels of DMT1, ACSL4, GPX4, Nrf2, and caspase-3 genes were evaluated by RT-qPCR. Lastly, a histological analysis of the heart tissues from several groups of rats was conducted. After hesperidin and/or rutin treatment, our results revealed that cardiac markers (serum troponin I and CK-MB), iron metabolism markers (serum and cardiac iron, TIBC, ferritin, transferrin, hepicidin and DMT1 expression levels) and oxidative stress markers (MDA, NO and ACSL4 expression levels) were significantly (P ⩽ 0.05) reduced, while the antioxidant markers (GSH level, GPx and SOD activities and GPX4 and Nrf2 expression levels) were significantly (P ⩽ 0.05) increased. Also, hesperidin and rutin exerted its protective anti-apoptotic role by significantly (P ⩽ 0.05) decreasing caspase-3 expression levels. Hesperidin and/or rutin treatment can be proposed as a therapeutic candidate to attenuate ferroptosis.
过量的铁与心血管疾病有关。类黄酮是抗氧化剂和心脏保护剂。因此,本研究的目的是评估两种黄酮类化合物,芦丁(R)和橙皮苷(H)的抗凋亡、抗氧化和铁螯合特性,以及它们预防大鼠诱导的铁凋亡的潜力。这是一种体内横断面研究,将大鼠分为12组;控制、H R, H + R,铁、铁+红外光谱、铁+红外+ Ref,铁+ H Fe +红外+ H,铁+ R、铁+红外+ R和铁+红外+ H + R。测定心脏和血清铁水平、血清肌钙蛋白I、肌酸激酶- mb (CK-MB)、总铁结合能力(TIBC)、转铁蛋白、铁蛋白和hepicidin。测定各组丙二醛(MDA)、一氧化氮(NO)、谷胱甘肽(GSH)水平及超氧化物歧化酶(SOD)、谷胱甘肽过氧化物酶(GPx)活性。RT-qPCR检测DMT1、ACSL4、GPX4、Nrf2、caspase-3基因的表达水平。最后,对几组大鼠的心脏组织进行组织学分析。经橙皮苷和/或芦丁处理后,心脏标志物(血清肌钙蛋白I和CK-MB)、铁代谢标志物(血清和心脏铁、TIBC、铁蛋白、转铁蛋白、hepicidin和DMT1表达水平)和氧化应激标志物(MDA、NO和ACSL4表达水平)显著(P < 0.05)降低,而抗氧化标志物(GSH水平、GPx和SOD活性以及GPX4和Nrf2表达水平)显著(P < 0.05)升高。橙皮苷和芦丁可显著(P < 0.05)降低caspase-3的表达水平,发挥抗凋亡的保护作用。橙皮苷和/或芦丁治疗可以作为一种候选的治疗方案,以减轻铁下垂。
{"title":"Iron chelating, antioxidant and anti-apoptotic activities of hesperidin and/or rutin against induced-ferroptosis in heart tissue of rats.","authors":"Haidy A Abdullah, Fatma Sm Moawed, Esraa Sa Ahmed, Fatma F Abdel Hamid, Riham Abdel-Hamid Haroun","doi":"10.1177/03946320251331873","DOIUrl":"10.1177/03946320251331873","url":null,"abstract":"<p><p>Excess iron has been associated with cardiovascular diseases. Flavonoids are antioxidants and cardioprotectants. Therefore, the goal of the current study is to evaluate the anti-apoptotic, antioxidant, and iron-chelating qualities of two flavonoids, rutin (R) and hesperidin (H), as well as their potential to prevent induced ferroptosis in rats. It is an in vivo cross-sectional study, in which rats were divided into 12 groups; control, H, R, H + R, Fe, Fe + IR, Fe + IR + Ref, Fe + H, Fe + IR + H, Fe + R, Fe + IR + R and Fe + IR + H + R. Cardiac and serum iron levels, serum troponin I, creatine kinase-MB (CK-MB), total iron binding capacity (TIBC), transferrin, ferritin, and hepicidin were determined. Moreover, the levels of malondialdehyde (MDA), nitric oxide (NO) and glutathione (GSH) and the activities of superoxide dismutase (SOD) and glutathione peroxidase (GPx), were also determined. The expression levels of DMT1, ACSL4, GPX4, Nrf2, and caspase-3 genes were evaluated by RT-qPCR. Lastly, a histological analysis of the heart tissues from several groups of rats was conducted. After hesperidin and/or rutin treatment, our results revealed that cardiac markers (serum troponin I and CK-MB), iron metabolism markers (serum and cardiac iron, TIBC, ferritin, transferrin, hepicidin and DMT1 expression levels) and oxidative stress markers (MDA, NO and ACSL4 expression levels) were significantly (<i>P</i> ⩽ 0.05) reduced, while the antioxidant markers (GSH level, GPx and SOD activities and GPX4 and Nrf2 expression levels) were significantly (<i>P</i> ⩽ 0.05) increased. Also, hesperidin and rutin exerted its protective anti-apoptotic role by significantly (<i>P</i> ⩽ 0.05) decreasing caspase-3 expression levels. Hesperidin and/or rutin treatment can be proposed as a therapeutic candidate to attenuate ferroptosis.</p>","PeriodicalId":48647,"journal":{"name":"International Journal of Immunopathology and Pharmacology","volume":"39 ","pages":"3946320251331873"},"PeriodicalIF":3.5,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12032457/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143812718","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-01Epub Date: 2025-12-10DOI: 10.1177/03946320251398285
Seona Park, Hyun Jung Lee, Seong-Joon Koh, Jong Pil Im, Joo Sung Kim
Objective: This study aimed to elucidate the anti-inflammatory mechanisms of asiatic acid (AA) by focusing on its modulation of the nuclear factor-κB (NF-κB) signaling pathway and to evaluate its therapeutic effects in murine models of both acute and chronic colitis.
Introduction: AA, a naturally occurring triterpenoid compound derived from Centella asiatica, is known for its anti-inflammatory activity. However, its comprehensive effects on both acute and chronic intestinal inflammation, particularly through detailed modulation of the NF-κB pathway, have not been fully elucidated.
Methods: Human intestinal epithelial cells COLO 205 and murine macrophage cells RAW 264.7 were pretreated with AA, followed by stimulation with tumor necrosis factor-α (TNF-α) or lipopolysaccharide (LPS), respectively. The mRNA expression of pro-inflammatory cytokines, including interleukin (IL)-8, TNF-α, and IL-6, was quantified using real-time RT-PCR. Western blotting was performed to assess the phosphorylation and degradation of the NF-κB inhibitor IκBα, and NF-κB DNA-binding activity was assessed via electrophoretic mobility shift assay (EMSA). In vivo, acute colitis was induced using dextran sulfate sodium (DSS) in wild-type mice, and chronic colitis was established by piroxicam administration in IL-10⁻/⁻ mice. Following AA treatment, colon length, body weight, histology (H&E) with histologic scoring, and colonic NF-κB p65 immunohistochemistry (IHC) were evaluated.
Results: AA significantly downregulated cytokine expression in both cell lines. It inhibited IκBα phosphorylation and degradation, and EMSA demonstrated a marked reduction in NF-κB DNA-binding activity. In mice, AA attenuated body weight loss, colonic shortening, and histologic inflammation in both DSS and IL-10⁻/⁻ models. Concomitantly, colonic IHC showed reduced nuclear NF-κB p65.
Conclusions: AA alleviates intestinal inflammation by suppressing NF-κB signaling in vitro and exhibits therapeutic efficacy in both acute and chronic colitis models, suggesting its potential as a therapeutic candidate for inflammatory bowel disease.
{"title":"Asiatic acid inhibits NF-κB signaling and ameliorates experimental acute and chronic colitis in mice.","authors":"Seona Park, Hyun Jung Lee, Seong-Joon Koh, Jong Pil Im, Joo Sung Kim","doi":"10.1177/03946320251398285","DOIUrl":"10.1177/03946320251398285","url":null,"abstract":"<p><strong>Objective: </strong>This study aimed to elucidate the anti-inflammatory mechanisms of asiatic acid (AA) by focusing on its modulation of the nuclear factor-κB (NF-κB) signaling pathway and to evaluate its therapeutic effects in murine models of both acute and chronic colitis.</p><p><strong>Introduction: </strong>AA, a naturally occurring triterpenoid compound derived from <i>Centella asiatica</i>, is known for its anti-inflammatory activity. However, its comprehensive effects on both acute and chronic intestinal inflammation, particularly through detailed modulation of the NF-κB pathway, have not been fully elucidated.</p><p><strong>Methods: </strong>Human intestinal epithelial cells COLO 205 and murine macrophage cells RAW 264.7 were pretreated with AA, followed by stimulation with tumor necrosis factor-α (TNF-α) or lipopolysaccharide (LPS), respectively. The mRNA expression of pro-inflammatory cytokines, including interleukin (IL)-8, TNF-α, and IL-6, was quantified using real-time RT-PCR. Western blotting was performed to assess the phosphorylation and degradation of the NF-κB inhibitor IκBα, and NF-κB DNA-binding activity was assessed via electrophoretic mobility shift assay (EMSA). In vivo, acute colitis was induced using dextran sulfate sodium (DSS) in wild-type mice, and chronic colitis was established by piroxicam administration in IL-10⁻/⁻ mice. Following AA treatment, colon length, body weight, histology (H&E) with histologic scoring, and colonic NF-κB p65 immunohistochemistry (IHC) were evaluated.</p><p><strong>Results: </strong>AA significantly downregulated cytokine expression in both cell lines. It inhibited IκBα phosphorylation and degradation, and EMSA demonstrated a marked reduction in NF-κB DNA-binding activity. In mice, AA attenuated body weight loss, colonic shortening, and histologic inflammation in both DSS and IL-10⁻/⁻ models. Concomitantly, colonic IHC showed reduced nuclear NF-κB p65.</p><p><strong>Conclusions: </strong>AA alleviates intestinal inflammation by suppressing NF-κB signaling in vitro and exhibits therapeutic efficacy in both acute and chronic colitis models, suggesting its potential as a therapeutic candidate for inflammatory bowel disease.</p>","PeriodicalId":48647,"journal":{"name":"International Journal of Immunopathology and Pharmacology","volume":"39 ","pages":"3946320251398285"},"PeriodicalIF":2.6,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12696327/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145716509","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}