首页 > 最新文献

Immunological Investigations最新文献

英文 中文
PTBP1-TRAF6 Axis Aggravates Chronic Obstructive Pulmonary Disease by Promoting M1 Macrophage Polarization and Impairing Clearance of Neutrophil Extracellular Traps. pptbp1 - traf6轴通过促进M1巨噬细胞极化和损害中性粒细胞胞外陷阱的清除而加重慢性阻塞性肺疾病。
IF 2.4 4区 医学 Q3 IMMUNOLOGY Pub Date : 2026-01-12 DOI: 10.1080/08820139.2025.2611872
Mingming Zhao, Liangfeng Yang, Shanshan Xu, Gang Chen, Yanping Hang, Minmin Yang, Haixia Zheng, Miaomiao Kong, Tao Li, Nan Wang

Objective: To elucidate the role of the polypyrimidine tract-binding protein 1 (PTBP1)-tumor necrosis factor receptor-associated factor 6 (TRAF6) pathway in chronic obstructive pulmonary disease (COPD), with a particular focus on its regulatory effects on macrophage polarization and clearance of neutrophil extracellular traps (NETs).

Methods: A COPD-like mouse model was established via cigarette smoke (CS) exposure. Lung injury, macrophage polarization, and levels of NETs were assessed. The role of TRAF6 in macrophage polarization and NETs clearance was assessed through lentiviral modulation of TRAF6 expression. RNA immunoprecipitation-quantitative polymerase chain reaction (RIP-qPCR), RNA stability assays, and Western blotting were performed to investigate the regulatory mechanism of PTBP1 on TRAF6 expression.

Results: The COPD-like mouse model exhibited increased M1 and decreased M2 macrophage populations, along with elevated NETs formation. Both PTBP1 and TRAF6 were upregulated in lung tissues of the COPD-like mouse model and cigarette smoke extract (CSE)-treated macrophages. Mechanistically, PTBP1 bound to and stabilized TRAF6 mRNA, enhancing TRAF6 expression. Inhibition of the PTBP1-TRAF6 pathway alleviated lung injury, restored macrophage polarization balance, and promoted NETs clearance.

Conclusion: PTBP1 enhances TRAF6 expression by stabilizing its mRNA, promoting M1 macrophage polarization and impairing NETs clearance, ultimately aggravating COPD.

目的:阐明多嘧啶束结合蛋白1 (PTBP1)-肿瘤坏死因子受体相关因子6 (TRAF6)通路在慢性阻塞性肺疾病(COPD)中的作用,特别关注其对巨噬细胞极化和中性粒细胞胞外陷阱(NETs)清除的调节作用。方法:通过香烟烟雾暴露建立copd样小鼠模型。评估肺损伤、巨噬细胞极化和NETs水平。通过慢病毒调节TRAF6表达来评估TRAF6在巨噬细胞极化和NETs清除中的作用。通过RNA免疫沉淀-定量聚合酶链反应(RIP-qPCR)、RNA稳定性和Western blotting检测PTBP1对TRAF6表达的调控机制。结果:copd样小鼠模型M1巨噬细胞数量增加,M2巨噬细胞数量减少,NETs形成升高。在copd样小鼠模型和香烟烟雾提取物(CSE)处理的巨噬细胞肺组织中,PTBP1和TRAF6均上调。机制上,PTBP1结合并稳定TRAF6 mRNA,增强TRAF6的表达。抑制PTBP1-TRAF6通路可减轻肺损伤,恢复巨噬细胞极化平衡,促进NETs清除。结论:PTBP1通过稳定TRAF6 mRNA表达,促进M1巨噬细胞极化,损害NETs清除,最终加重COPD。
{"title":"PTBP1-TRAF6 Axis Aggravates Chronic Obstructive Pulmonary Disease by Promoting M1 Macrophage Polarization and Impairing Clearance of Neutrophil Extracellular Traps.","authors":"Mingming Zhao, Liangfeng Yang, Shanshan Xu, Gang Chen, Yanping Hang, Minmin Yang, Haixia Zheng, Miaomiao Kong, Tao Li, Nan Wang","doi":"10.1080/08820139.2025.2611872","DOIUrl":"https://doi.org/10.1080/08820139.2025.2611872","url":null,"abstract":"<p><strong>Objective: </strong>To elucidate the role of the polypyrimidine tract-binding protein 1 (PTBP1)-tumor necrosis factor receptor-associated factor 6 (TRAF6) pathway in chronic obstructive pulmonary disease (COPD), with a particular focus on its regulatory effects on macrophage polarization and clearance of neutrophil extracellular traps (NETs).</p><p><strong>Methods: </strong>A COPD-like mouse model was established via cigarette smoke (CS) exposure. Lung injury, macrophage polarization, and levels of NETs were assessed. The role of TRAF6 in macrophage polarization and NETs clearance was assessed through lentiviral modulation of TRAF6 expression. RNA immunoprecipitation-quantitative polymerase chain reaction (RIP-qPCR), RNA stability assays, and Western blotting were performed to investigate the regulatory mechanism of PTBP1 on TRAF6 expression.</p><p><strong>Results: </strong>The COPD-like mouse model exhibited increased M1 and decreased M2 macrophage populations, along with elevated NETs formation. Both PTBP1 and TRAF6 were upregulated in lung tissues of the COPD-like mouse model and cigarette smoke extract (CSE)-treated macrophages. Mechanistically, PTBP1 bound to and stabilized TRAF6 mRNA, enhancing TRAF6 expression. Inhibition of the PTBP1-TRAF6 pathway alleviated lung injury, restored macrophage polarization balance, and promoted NETs clearance.</p><p><strong>Conclusion: </strong>PTBP1 enhances TRAF6 expression by stabilizing its mRNA, promoting M1 macrophage polarization and impairing NETs clearance, ultimately aggravating COPD.</p>","PeriodicalId":13387,"journal":{"name":"Immunological Investigations","volume":" ","pages":"1-26"},"PeriodicalIF":2.4,"publicationDate":"2026-01-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145951816","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Emerging Immunotherapeutic Approaches in Colorectal Cancer: From Checkpoint Inhibitors to CAR-T Cell and Viral Vector Vaccines. 结直肠癌新出现的免疫治疗方法:从检查点抑制剂到CAR-T细胞和病毒载体疫苗。
IF 2.4 4区 医学 Q3 IMMUNOLOGY Pub Date : 2026-01-11 DOI: 10.1080/08820139.2026.2613047
Rajeev Ranjan, Amit Kumar, Vikas Kumar, Bhawesh Kumar Mahato, Atreyee Sarkar, Ratnesh Kumar Yadav, Vikas Gond, Sreeharsha Nagaraja, Ravi Raj Pal

Introduction: Colorectal Cancer has been a significant health issue at a global level in terms of increasing incidence, high metastatic capacity, and inadequate treatment reactions in most patient groups. Though chemotherapy and targeted agents have given better survival chances, resistance to treatment, avoidance of the immune system as well as systemic toxicity still limit long-term efficacy. Immunotherapeutic strategies have developed as effective methods in the past few years to address these shortomings.

Methods: A comprehensive literature search was conducted across different electronics databases including Web of Science, PubMed, Scopus, and ScienceDirect utilizing keywords colorectal cancer immunotherapy, PD-1 / PD-L1, CAR-T cells, viral vector vaccines, carcinoembryonic antigen alone and in combinations.

Results: PD-1/PD-L1, CTLA-4, immune checkpoint inhibitors have shown long-term responses, especially in mismatch repair-deficient and microsatellite instability-high tumours, and cell-based therapies such as CAR -T cells and dendritic cell vaccines provide other opportunities of personalised immune modulation.

Discussion: This critical review summarizes the current developments in terms of immunotherapy and interventions of CRC with a focus on the mechanistic understanding of the issue, clinical outcomes, and combinatorial strategies aimed to improve antitumor immunity and surmount therapy resistance. Specific attention is paid to the optimization of the checkpoint blockade, innovations with vaccines and CAR-T engineering to accumulate and release tumor-specific cells and control the microenvironment. Finally, key translational challenges current clinical trials, and future perspectives are identified to promote the development of the next-generation multimodal therapies in the efficient and customized management of CRC.

导论:在全球范围内,结直肠癌的发病率不断上升,转移能力高,在大多数患者群体中治疗反应不足,是一个重大的健康问题。尽管化疗和靶向药物提供了更好的生存机会,但对治疗的耐药性、免疫系统的回避以及全身毒性仍然限制了长期疗效。在过去几年中,免疫治疗策略已发展成为解决这些缺陷的有效方法。方法:在Web of Science、PubMed、Scopus和ScienceDirect等不同的电子数据库中进行综合文献检索,检索关键词为结直肠癌免疫治疗、PD-1 / PD-L1、CAR-T细胞、病毒载体疫苗、癌胚抗原单独和联合。结果:PD-1/PD-L1、CTLA-4、免疫检查点抑制剂显示出长期应答,特别是在错配修复缺陷和微卫星不稳定性高的肿瘤中,基于细胞的疗法,如CAR -T细胞和树突状细胞疫苗,提供了个性化免疫调节的其他机会。讨论:这篇重要的综述总结了CRC免疫治疗和干预措施的最新进展,重点是对该问题的机制理解、临床结果和旨在提高抗肿瘤免疫和克服治疗耐药性的联合策略。特别关注检查点阻断的优化,疫苗和CAR-T工程的创新,以积累和释放肿瘤特异性细胞并控制微环境。最后,确定了关键的转化挑战,当前的临床试验和未来的观点,以促进下一代多模式治疗在有效和定制化的CRC管理中的发展。
{"title":"Emerging Immunotherapeutic Approaches in Colorectal Cancer: From Checkpoint Inhibitors to CAR-T Cell and Viral Vector Vaccines.","authors":"Rajeev Ranjan, Amit Kumar, Vikas Kumar, Bhawesh Kumar Mahato, Atreyee Sarkar, Ratnesh Kumar Yadav, Vikas Gond, Sreeharsha Nagaraja, Ravi Raj Pal","doi":"10.1080/08820139.2026.2613047","DOIUrl":"https://doi.org/10.1080/08820139.2026.2613047","url":null,"abstract":"<p><strong>Introduction: </strong>Colorectal Cancer has been a significant health issue at a global level in terms of increasing incidence, high metastatic capacity, and inadequate treatment reactions in most patient groups. Though chemotherapy and targeted agents have given better survival chances, resistance to treatment, avoidance of the immune system as well as systemic toxicity still limit long-term efficacy. Immunotherapeutic strategies have developed as effective methods in the past few years to address these shortomings.</p><p><strong>Methods: </strong>A comprehensive literature search was conducted across different electronics databases including Web of Science, PubMed, Scopus, and ScienceDirect utilizing keywords colorectal cancer immunotherapy, PD-1 / PD-L1, CAR-T cells, viral vector vaccines, carcinoembryonic antigen alone and in combinations.</p><p><strong>Results: </strong>PD-1/PD-L1, CTLA-4, immune checkpoint inhibitors have shown long-term responses, especially in mismatch repair-deficient and microsatellite instability-high tumours, and cell-based therapies such as CAR -T cells and dendritic cell vaccines provide other opportunities of personalised immune modulation.</p><p><strong>Discussion: </strong>This critical review summarizes the current developments in terms of immunotherapy and interventions of CRC with a focus on the mechanistic understanding of the issue, clinical outcomes, and combinatorial strategies aimed to improve antitumor immunity and surmount therapy resistance. Specific attention is paid to the optimization of the checkpoint blockade, innovations with vaccines and CAR-T engineering to accumulate and release tumor-specific cells and control the microenvironment. Finally, key translational challenges current clinical trials, and future perspectives are identified to promote the development of the next-generation multimodal therapies in the efficient and customized management of CRC.</p>","PeriodicalId":13387,"journal":{"name":"Immunological Investigations","volume":" ","pages":"1-36"},"PeriodicalIF":2.4,"publicationDate":"2026-01-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145951844","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Naloxone and Naltrexone as Potential Adjuvants for Vaccination Against Intracellular Pathogens: A Narrative Review. 纳洛酮和纳曲酮作为细胞内病原体疫苗接种的潜在佐剂:综述。
IF 2.4 4区 医学 Q3 IMMUNOLOGY Pub Date : 2026-01-01 DOI: 10.1080/08820139.2025.2607684
Hajar Yaghoobi, Maryam Hataminejad, Hakim Azizi

Background: Adjuvants potentiate adaptive immunity by enhancing antigen-specific immune responses, yet many exhibit high toxicity for clinical use. Naloxone (NLx) and naltrexone (NLt) opioid receptor antagonists have emerged as promising adjuvants in preclinical vaccination models against intracellular pathogens.

Objective: This narrative review synthesizes evidence from 2000 to 2025 evaluating NLx/NLt as adjuvants against bacterial, viral, and parasitic infections, while examining proposed immunoregulatory mechanisms.

Methods: A comprehensive search of Web of Science, PubMed, Scopus, ScienceDirect, and ProQuest employed Boolean operators combining: (vaccine OR vaccination) AND (adjuvant) AND (naloxone OR naltrexone OR NLx OR NLt OR "opioid receptor antagonist") AND ("immune response" OR "cellular immunity" OR "Th1").

Results: Key findings demonstrate that NLx/NLt monotherapy or combinatorial use with Alum consistently, enhanced Th1 polarization and cellular immunity, Elevated pathogen clearance and protection rates, synergistically overcame Alum's Th2 bias. Mechanistically, μ-opioid receptor (MOR) antagonism disrupts tonic immunosuppression, reversing opioid-mediated inhibition of dendritic cell maturation, IL-12 production, and cytotoxic T-cell function.

Conclusions: This immunorestorative action positions NLx/NLt as strategic adjuvants for vaccines targeting intracellular pathogens requiring robust cell-mediated immunity. Future horizons include combinatorial platforms (e.g. DNA vaccines + low-dose naltrexone) and clinical translation.

背景:佐剂通过增强抗原特异性免疫反应来增强适应性免疫,但许多佐剂在临床使用时表现出高毒性。纳洛酮(NLx)和纳曲酮(NLt)阿片受体拮抗剂已成为临床前疫苗模型中抗细胞内病原体的有希望的佐剂。目的:这篇叙述性综述综合了2000年至2025年的证据,评估了NLx/NLt作为抗细菌、病毒和寄生虫感染的佐剂,同时研究了拟议的免疫调节机制。方法:综合检索Web of Science, PubMed, Scopus, ScienceDirect和ProQuest,采用布尔运算符组合:(疫苗或疫苗接种)和(佐剂)和(纳洛酮或纳曲酮或NLx或NLt或“阿片受体拮抗剂”)和(“免疫反应”或“细胞免疫”或“Th1”)。结果:主要研究结果表明,NLx/NLt单药或与Alum联合使用,可增强Th1极化和细胞免疫,提高病原体清除率和保护率,协同克服Alum的Th2偏倚。从机制上讲,μ-阿片受体(MOR)拮抗破坏补性免疫抑制,逆转阿片介导的树突状细胞成熟、IL-12产生和细胞毒性t细胞功能的抑制。结论:这种免疫恢复作用使NLx/NLt成为针对需要强大细胞介导免疫的细胞内病原体的疫苗的战略佐剂。未来的发展方向包括组合平台(例如DNA疫苗+低剂量纳曲酮)和临床翻译。
{"title":"Naloxone and Naltrexone as Potential Adjuvants for Vaccination Against Intracellular Pathogens: A Narrative Review.","authors":"Hajar Yaghoobi, Maryam Hataminejad, Hakim Azizi","doi":"10.1080/08820139.2025.2607684","DOIUrl":"https://doi.org/10.1080/08820139.2025.2607684","url":null,"abstract":"<p><strong>Background: </strong>Adjuvants potentiate adaptive immunity by enhancing antigen-specific immune responses, yet many exhibit high toxicity for clinical use. Naloxone (NLx) and naltrexone (NLt) opioid receptor antagonists have emerged as promising adjuvants in preclinical vaccination models against intracellular pathogens.</p><p><strong>Objective: </strong>This narrative review synthesizes evidence from 2000 to 2025 evaluating NLx/NLt as adjuvants against bacterial, viral, and parasitic infections, while examining proposed immunoregulatory mechanisms.</p><p><strong>Methods: </strong>A comprehensive search of Web of Science, PubMed, Scopus, ScienceDirect, and ProQuest employed Boolean operators combining: (vaccine OR vaccination) AND (adjuvant) AND (naloxone OR naltrexone OR NLx OR NLt OR \"opioid receptor antagonist\") AND (\"immune response\" OR \"cellular immunity\" OR \"Th1\").</p><p><strong>Results: </strong>Key findings demonstrate that NLx/NLt monotherapy or combinatorial use with Alum consistently, enhanced Th1 polarization and cellular immunity, Elevated pathogen clearance and protection rates, synergistically overcame Alum's Th2 bias. Mechanistically, μ-opioid receptor (MOR) antagonism disrupts tonic immunosuppression, reversing opioid-mediated inhibition of dendritic cell maturation, IL-12 production, and cytotoxic T-cell function.</p><p><strong>Conclusions: </strong>This immunorestorative action positions NLx/NLt as strategic adjuvants for vaccines targeting intracellular pathogens requiring robust cell-mediated immunity. Future horizons include combinatorial platforms (e.g. DNA vaccines + low-dose naltrexone) and clinical translation.</p>","PeriodicalId":13387,"journal":{"name":"Immunological Investigations","volume":" ","pages":"1-28"},"PeriodicalIF":2.4,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145888594","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
High-Fat Diet Promotes Tumor Immune Evasion via CD155 Upregulation in Colorectal Cancer. 高脂肪饮食通过上调CD155促进结直肠癌的肿瘤免疫逃避。
IF 2.4 4区 医学 Q3 IMMUNOLOGY Pub Date : 2025-12-20 DOI: 10.1080/08820139.2025.2600597
Liwei Liu, Ling Mo, Jingcun Fang, Mingqiang Ling, Yingying Lei, Zhenzhen Wen, Guohui Liang, Jingfeng Guo

Background: Obesity-induced metabolic stress impairs the efficacy of immune checkpoint blockade (ICB) therapy, but the mechanisms linking a high-fat diet (HFD) to immune suppression remain unclear.

Objective: To investigate how HFD-induced metabolic changes modulate the tumor immune microenvironment through the STAT3/CD155 axis.

Methods: Murine colorectal cancer models using control, CD155-overexpressing (CD155 OE), and STAT3 knockdown CT26 cells were established under normal or HFD conditions with anti - PD-L1 treatment. Tumor growth, immune infiltration, and gene expression were analyzed by flow cytometry, Western blotting, and chromatin immunoprecipitation.

Results: HFD impaired anti - PD-L1 efficacy and accelerated tumor growth. Mechanistically, HFD promoted STAT3 nuclear translocation and CD155 upregulation, reducing CD8+ T cell infiltration and enhancing regulatory T cell accumulation. Chromatin immunoprecipitation confirmed direct STAT3 binding to the CD155 promoter, while STAT3 knockdown reversed these effects and restored antitumor immunity in HFD-fed mice.

Conclusion: HFD-induced metabolic stress drives immune evasion via the STAT3/CD155 axis. Targeting this pathway may improve ICB efficacy in obesity-related cancers.

背景:肥胖诱导的代谢应激会损害免疫检查点阻断(ICB)治疗的效果,但高脂肪饮食(HFD)与免疫抑制之间的机制尚不清楚。目的:探讨hfd诱导的代谢变化如何通过STAT3/CD155轴调控肿瘤免疫微环境。方法:通过抗PD-L1治疗,在正常或HFD条件下建立小鼠结直肠癌模型,使用对照、CD155过表达(CD155 OE)和STAT3敲低的CT26细胞。通过流式细胞术、Western blotting和染色质免疫沉淀分析肿瘤生长、免疫浸润和基因表达。结果:HFD降低了抗PD-L1的作用,加速了肿瘤的生长。机制上,HFD促进STAT3核易位和CD155上调,减少CD8+ T细胞浸润,增强调节性T细胞积累。染色质免疫沉淀证实了STAT3与CD155启动子的直接结合,而STAT3敲低逆转了这些作用,并恢复了hfd喂养小鼠的抗肿瘤免疫。结论:hfd诱导的代谢应激通过STAT3/CD155轴驱动免疫逃避。靶向这一途径可能提高ICB在肥胖相关癌症中的疗效。
{"title":"High-Fat Diet Promotes Tumor Immune Evasion via CD155 Upregulation in Colorectal Cancer.","authors":"Liwei Liu, Ling Mo, Jingcun Fang, Mingqiang Ling, Yingying Lei, Zhenzhen Wen, Guohui Liang, Jingfeng Guo","doi":"10.1080/08820139.2025.2600597","DOIUrl":"https://doi.org/10.1080/08820139.2025.2600597","url":null,"abstract":"<p><strong>Background: </strong>Obesity-induced metabolic stress impairs the efficacy of immune checkpoint blockade (ICB) therapy, but the mechanisms linking a high-fat diet (HFD) to immune suppression remain unclear.</p><p><strong>Objective: </strong>To investigate how HFD-induced metabolic changes modulate the tumor immune microenvironment through the STAT3/CD155 axis.</p><p><strong>Methods: </strong>Murine colorectal cancer models using control, CD155-overexpressing (CD155 OE), and STAT3 knockdown CT26 cells were established under normal or HFD conditions with anti - PD-L1 treatment. Tumor growth, immune infiltration, and gene expression were analyzed by flow cytometry, Western blotting, and chromatin immunoprecipitation.</p><p><strong>Results: </strong>HFD impaired anti - PD-L1 efficacy and accelerated tumor growth. Mechanistically, HFD promoted STAT3 nuclear translocation and CD155 upregulation, reducing CD8<sup>+</sup> T cell infiltration and enhancing regulatory T cell accumulation. Chromatin immunoprecipitation confirmed direct STAT3 binding to the CD155 promoter, while STAT3 knockdown reversed these effects and restored antitumor immunity in HFD-fed mice.</p><p><strong>Conclusion: </strong>HFD-induced metabolic stress drives immune evasion via the STAT3/CD155 axis. Targeting this pathway may improve ICB efficacy in obesity-related cancers.</p>","PeriodicalId":13387,"journal":{"name":"Immunological Investigations","volume":" ","pages":"1-19"},"PeriodicalIF":2.4,"publicationDate":"2025-12-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145800458","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
OPN Promotes Hepatocellular Carcinoma Progression Through the p65/c-Myc/CD155 Axis by Suppressing CD8+ T Cell Infiltration and Activation. OPN通过p65/c-Myc/CD155轴抑制CD8+ T细胞浸润和活化促进肝细胞癌进展
IF 2.4 4区 医学 Q3 IMMUNOLOGY Pub Date : 2025-12-19 DOI: 10.1080/08820139.2025.2599834
Jincun Fang, Liwei Liu, Zhiying He, Mingqiang Ling, Jiajun Liang, Yingying Lei, Zhenzhen Wen, Jingfeng Guo

Introduction: The immunosuppressive microenvironment of hepatocellular carcinoma (HCC) is shaped by multiple oncogenic pathways, but howosteopontin (OPN), a molecule frequently overexpressed in HCC, regulates anti-tumor immunity remains unclear.

Methods: To define the mechanism by which OPN modulates the HCC immune microenvironment, with a focus on its regulation of CD155 and its impact on CD8+ T-cell - mediated anti-tumor responses.

Results: We identified that OPN activates the p65/NF-κB - c-Myc/CD155 signaling axis, leading to robust upregulation of CD155 and impaired intratumoral CD8+ T-cell infiltration and effector activity. These immunosuppressive effects occurred independently of changes in tumor stemness markers or Treg accumulation. Genetic suppression of p65 abrogated OPN-induced CD155 expression and mitigated tumor progression in vivo, demonstrating the pathway's functional requirement.

Discussion: OPN drives HCC progression by suppressing CD8+ T-cell immunity through the NF-κB (p65)/c-Myc/CD155 axis. Targeting this pathway may enhance anti-tumor immunity and represents a promising therapeutic strategy for HCC.

肝细胞癌(HCC)的免疫抑制微环境是由多种致癌途径形成的,但在HCC中经常过表达的分子肌桥蛋白(OPN)如何调节抗肿瘤免疫尚不清楚。方法:明确OPN调节HCC免疫微环境的机制,重点研究OPN对CD155的调节及其对CD8+ t细胞介导的抗肿瘤反应的影响。结果:我们发现OPN激活p65/NF-κ b - c-Myc/CD155信号轴,导致CD155的强烈上调,损害肿瘤内CD8+ t细胞浸润和效应物活性。这些免疫抑制作用独立于肿瘤干性标志物或Treg积累的变化而发生。p65的遗传抑制消除了opn诱导的CD155表达,并在体内减缓了肿瘤的进展,证明了该途径的功能需求。讨论:OPN通过NF-κB (p65)/c-Myc/CD155轴抑制CD8+ t细胞免疫,从而驱动HCC进展。靶向这一途径可能增强抗肿瘤免疫,是一种有希望的HCC治疗策略。
{"title":"OPN Promotes Hepatocellular Carcinoma Progression Through the p65/c-Myc/CD155 Axis by Suppressing CD8+ T Cell Infiltration and Activation.","authors":"Jincun Fang, Liwei Liu, Zhiying He, Mingqiang Ling, Jiajun Liang, Yingying Lei, Zhenzhen Wen, Jingfeng Guo","doi":"10.1080/08820139.2025.2599834","DOIUrl":"https://doi.org/10.1080/08820139.2025.2599834","url":null,"abstract":"<p><strong>Introduction: </strong>The immunosuppressive microenvironment of hepatocellular carcinoma (HCC) is shaped by multiple oncogenic pathways, but howosteopontin (OPN), a molecule frequently overexpressed in HCC, regulates anti-tumor immunity remains unclear.</p><p><strong>Methods: </strong>To define the mechanism by which OPN modulates the HCC immune microenvironment, with a focus on its regulation of CD155 and its impact on CD8<sup>+</sup> T-cell - mediated anti-tumor responses.</p><p><strong>Results: </strong>We identified that OPN activates the p65/NF-κB - c-Myc/CD155 signaling axis, leading to robust upregulation of CD155 and impaired intratumoral CD8<sup>+</sup> T-cell infiltration and effector activity. These immunosuppressive effects occurred independently of changes in tumor stemness markers or Treg accumulation. Genetic suppression of p65 abrogated OPN-induced CD155 expression and mitigated tumor progression in vivo, demonstrating the pathway's functional requirement.</p><p><strong>Discussion: </strong>OPN drives HCC progression by suppressing CD8<sup>+</sup> T-cell immunity through the NF-κB (p65)/c-Myc/CD155 axis. Targeting this pathway may enhance anti-tumor immunity and represents a promising therapeutic strategy for HCC.</p>","PeriodicalId":13387,"journal":{"name":"Immunological Investigations","volume":" ","pages":"1-19"},"PeriodicalIF":2.4,"publicationDate":"2025-12-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145793861","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The Therapeutic Targeting Effect of Toll-Like Receptors and Their Related Signaling Pathways in Cardiomyopathy. toll样受体及其相关信号通路在心肌病中的靶向治疗作用。
IF 2.4 4区 医学 Q3 IMMUNOLOGY Pub Date : 2025-12-10 DOI: 10.1080/08820139.2025.2583276
Haoran Cheng, Jieqi Wang, Wang Ting

Background: Cardiomyopathies contribute substantially to heart failure, arrhythmias, and sudden cardiac death. Innate immune activation-particularly via Toll-like receptors (TLRs)-is increasingly recognized as a driver of pathological cardiac remodeling.

Objective: To evaluate how TLR expression and downstream signaling pathways (MyD88 and TRIF) shape distinct cardiomyopathy phenotypes and to assess therapeutic strategies targeting these mechanisms.

Methods: Evidence from studies of ischemic, viral, septic, diabetic, drug-induced, hypertrophic, and obesity-associated cardiomyopathies was surveyed to determine the roles of TLR subtypes and the impact of modulating their signaling pathways.

Results: Activation of TLR2, TLR3, TLR4, TLR7, and TLR9 consistently amplifies myocardial inflammation, fibrosis, and functional decline. However, context-dependent protective effects in certain settings highlight the complexity of uniformly suppressing TLR activity. Preclinical investigations demonstrate favorable outcomes from selective pathway modulation, including TLR4 antagonists (such as TAK-242 and eritoran), MyD88 pathway inhibitors, and nucleic-acid-directed TLR9 strategies.

Conclusions: Targeting TLR signaling has strong potential to limit inflammation, curb fibrotic remodeling, and preserve cardiac function across diverse cardiomyopathies. The most promising therapeutic direction involves developing subtype-specific and cell- or pathway-targeted interventions, supported by translational research to bridge preclinical findings with clinical application.

背景:心肌病是心力衰竭、心律失常和心源性猝死的主要原因。先天免疫激活-特别是通过toll样受体(TLRs)-越来越被认为是病理性心脏重构的驱动因素。目的:评估TLR表达和下游信号通路(MyD88和TRIF)如何塑造不同的心肌病表型,并评估针对这些机制的治疗策略。方法:对缺血性、病毒性、败血症、糖尿病、药物性、肥厚性和肥胖相关心肌病的研究证据进行调查,以确定TLR亚型的作用以及调节其信号通路的影响。结果:TLR2、TLR3、TLR4、TLR7和TLR9的激活持续放大心肌炎症、纤维化和功能下降。然而,在某些情况下,环境依赖的保护作用突出了均匀抑制TLR活性的复杂性。临床前研究表明,选择性途径调节包括TLR4拮抗剂(如TAK-242和itoran)、MyD88途径抑制剂和核酸导向TLR9策略的有利结果。结论:针对TLR信号具有限制炎症、抑制纤维化重塑和保护各种心肌病心功能的强大潜力。最有希望的治疗方向包括发展亚型特异性和细胞或途径靶向干预,并得到转化研究的支持,将临床前发现与临床应用联系起来。
{"title":"The Therapeutic Targeting Effect of Toll-Like Receptors and Their Related Signaling Pathways in Cardiomyopathy.","authors":"Haoran Cheng, Jieqi Wang, Wang Ting","doi":"10.1080/08820139.2025.2583276","DOIUrl":"https://doi.org/10.1080/08820139.2025.2583276","url":null,"abstract":"<p><strong>Background: </strong>Cardiomyopathies contribute substantially to heart failure, arrhythmias, and sudden cardiac death. Innate immune activation-particularly via Toll-like receptors (TLRs)-is increasingly recognized as a driver of pathological cardiac remodeling.</p><p><strong>Objective: </strong>To evaluate how TLR expression and downstream signaling pathways (MyD88 and TRIF) shape distinct cardiomyopathy phenotypes and to assess therapeutic strategies targeting these mechanisms.</p><p><strong>Methods: </strong>Evidence from studies of ischemic, viral, septic, diabetic, drug-induced, hypertrophic, and obesity-associated cardiomyopathies was surveyed to determine the roles of TLR subtypes and the impact of modulating their signaling pathways.</p><p><strong>Results: </strong>Activation of TLR2, TLR3, TLR4, TLR7, and TLR9 consistently amplifies myocardial inflammation, fibrosis, and functional decline. However, context-dependent protective effects in certain settings highlight the complexity of uniformly suppressing TLR activity. Preclinical investigations demonstrate favorable outcomes from selective pathway modulation, including TLR4 antagonists (such as TAK-242 and eritoran), MyD88 pathway inhibitors, and nucleic-acid-directed TLR9 strategies.</p><p><strong>Conclusions: </strong>Targeting TLR signaling has strong potential to limit inflammation, curb fibrotic remodeling, and preserve cardiac function across diverse cardiomyopathies. The most promising therapeutic direction involves developing subtype-specific and cell- or pathway-targeted interventions, supported by translational research to bridge preclinical findings with clinical application.</p>","PeriodicalId":13387,"journal":{"name":"Immunological Investigations","volume":" ","pages":"1-35"},"PeriodicalIF":2.4,"publicationDate":"2025-12-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145714247","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
DOCK2 in Disease: Emerging Insights and Research Advances. 疾病中的DOCK2:新见解和研究进展。
IF 2.4 4区 医学 Q3 IMMUNOLOGY Pub Date : 2025-12-10 DOI: 10.1080/08820139.2025.2596074
Zhaolong Yu, Songzhi Jin, Shujing Wan, Zixuan Zhang, Yingjie Jiang, Jialin Li, Suzhen Wu

Background: This review aims to synthesize the structural and functional characteristics of DOCK2, as well as its mechanistic roles in various diseases, in order to provide a theoretical foundation for targeted therapies.

Methods: We systematically reviewed existing literature to summarize the molecular features and expression patterns of DOCK2, its regulation of cellular processes through both Rac-dependent and Rac-independent pathways, and its implications in inflammation, cancer, fibrosis, and related disorders.

Results: DOCK2, a member of the Dock-A subfamily of GEFs, is widely expressed across tissues and prominently in immune cells (e.g., dendritic cells, macrophages, T cells, B cells), where it regulates proliferation, differentiation, migration, and cytokine secretion primarily via Rac activation. Emerging evidence also links DOCK2 to non-immune cell types such as glial cells and vascular smooth muscle cells, highlighting its relevance in immune-related and broader pathological conditions including inflammatory diseases, cancer, atherosclerosis, idiopathic pulmonary fibrosis, and obesity.

Conclusion: As a critical GEF, DOCK2 plays a central role in immune and non-immune cellular processes through Rac and non-Rac signaling pathways. Dysregulation of DOCK2 is closely associated with multiple diseases. Further elucidation of its regulatory networks may reveal novel therapeutic targets for treating related disorders.

背景:本文旨在综合DOCK2的结构、功能特征及其在各种疾病中的作用机制,为靶向治疗提供理论依据。方法:我们系统地回顾了现有文献,总结了DOCK2的分子特征和表达模式,它通过rac依赖性和rac非依赖性途径对细胞过程的调节,以及它在炎症、癌症、纤维化和相关疾病中的意义。结果:DOCK2是GEFs Dock-A亚家族的一员,广泛表达于组织中,在免疫细胞(如树突状细胞、巨噬细胞、T细胞、B细胞)中表现突出,主要通过Rac激活来调节增殖、分化、迁移和细胞因子分泌。新出现的证据还将DOCK2与非免疫细胞类型(如胶质细胞和血管平滑肌细胞)联系起来,突出了其在免疫相关和更广泛的病理状况(包括炎症性疾病、癌症、动脉粥样硬化、特发性肺纤维化和肥胖)中的相关性。结论:DOCK2作为一个关键的GEF,通过Rac和非Rac信号通路在免疫和非免疫细胞过程中发挥核心作用。DOCK2的失调与多种疾病密切相关。进一步阐明其调控网络可能为治疗相关疾病揭示新的治疗靶点。
{"title":"DOCK2 in Disease: Emerging Insights and Research Advances.","authors":"Zhaolong Yu, Songzhi Jin, Shujing Wan, Zixuan Zhang, Yingjie Jiang, Jialin Li, Suzhen Wu","doi":"10.1080/08820139.2025.2596074","DOIUrl":"https://doi.org/10.1080/08820139.2025.2596074","url":null,"abstract":"<p><strong>Background: </strong>This review aims to synthesize the structural and functional characteristics of DOCK2, as well as its mechanistic roles in various diseases, in order to provide a theoretical foundation for targeted therapies.</p><p><strong>Methods: </strong>We systematically reviewed existing literature to summarize the molecular features and expression patterns of DOCK2, its regulation of cellular processes through both Rac-dependent and Rac-independent pathways, and its implications in inflammation, cancer, fibrosis, and related disorders.</p><p><strong>Results: </strong>DOCK2, a member of the Dock-A subfamily of GEFs, is widely expressed across tissues and prominently in immune cells (e.g., dendritic cells, macrophages, T cells, B cells), where it regulates proliferation, differentiation, migration, and cytokine secretion primarily via Rac activation. Emerging evidence also links DOCK2 to non-immune cell types such as glial cells and vascular smooth muscle cells, highlighting its relevance in immune-related and broader pathological conditions including inflammatory diseases, cancer, atherosclerosis, idiopathic pulmonary fibrosis, and obesity.</p><p><strong>Conclusion: </strong>As a critical GEF, DOCK2 plays a central role in immune and non-immune cellular processes through Rac and non-Rac signaling pathways. Dysregulation of DOCK2 is closely associated with multiple diseases. Further elucidation of its regulatory networks may reveal novel therapeutic targets for treating related disorders.</p>","PeriodicalId":13387,"journal":{"name":"Immunological Investigations","volume":" ","pages":"1-23"},"PeriodicalIF":2.4,"publicationDate":"2025-12-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145714029","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Phillyrin Ameliorates Traumatic Spinal Cord Injury by Inhibiting Inflammation, Oxidative Stress, and Pyroptosis Through Modulation of the Nrf2/HO-1 Signaling Pathway. 通过调节Nrf2/HO-1信号通路,连翘苷通过抑制炎症、氧化应激和焦亡改善创伤性脊髓损伤。
IF 2.4 4区 医学 Q3 IMMUNOLOGY Pub Date : 2025-12-09 DOI: 10.1080/08820139.2025.2599833
Yang Gao, Yanyang Wang, Liumin Bai, Kunpeng Fu

Background: Treating traumatic spinal cord injury (SCI) requires mitigating microglia-mediated inflammation and nerve damage. Phillyrin (phi) shows neuroprotective potential, but its effects on SCI are unknown. We investigated phi's impact on microglial activation, inflammation, neuronal apoptosis, and pyroptosis in mice after SCI.

Methods: A mouse contusion SCI model was used to assess phi efficacy. Functional recovery was assessed with BBB and LSS scales. Histological and TUNEL staining were used to evaluate tissue damage and neuronal survival. Microglial phenotypes, inflammation, pyroptosis, oxidative stress and Nrf2/HO-1 pathway were analyzed.

Results: Phi treatment accelerated motor function recovery, improved tissue pathology, increased live cells, and reduced apoptosis. Phi treatment showed a decrease in the proportion of M1 pro-inflammatory microglia characterized by iNOS, an increase in the proportion of M2 anti-inflammatory microglia characterized by Arg1 and a reduction in inflammatory factors. Phi enhanced Nrf2 expression in neurons and microglia, activated HO-1 and NQO1 expression, weaken oxidative stress, and improved pyroptosis.

Conclusion: Phi alleviates SCI by activating the Nrf2/HO-1 pathway, suppressing microglial activation-mediated neuroinflammation, neuronal apoptosis, oxidative stress and pyroptosis. This study provides the first evidence of the therapeutic effect of phi on traumatic SCI, providing theoretical support for its future clinical application in SCI.

背景:治疗创伤性脊髓损伤(SCI)需要减轻小胶质细胞介导的炎症和神经损伤。philyrin (phi)具有神经保护潜能,但其对脊髓损伤的作用尚不清楚。我们研究了phi对脊髓损伤后小鼠小胶质细胞激活、炎症、神经元凋亡和焦亡的影响。方法:采用小鼠挫裂性脊髓损伤模型评价其疗效。用BBB和LSS量表评估功能恢复。采用组织学和TUNEL染色评估组织损伤和神经元存活。分析小胶质细胞表型、炎症、焦亡、氧化应激和Nrf2/HO-1通路。结果:Phi处理加速运动功能恢复,改善组织病理,增加活细胞,减少凋亡。Phi处理后,以iNOS为特征的M1促炎小胶质细胞比例降低,以Arg1为特征的M2抗炎小胶质细胞比例增加,炎症因子减少。Phi增强神经元和小胶质细胞中Nrf2的表达,激活HO-1和NQO1的表达,减弱氧化应激,改善焦亡。结论:Phi通过激活Nrf2/HO-1通路,抑制小胶质细胞激活介导的神经炎症、神经元凋亡、氧化应激和焦亡,减轻脊髓损伤。本研究首次证实了phi对创伤性脊髓损伤的治疗作用,为其今后在脊髓损伤中的临床应用提供了理论支持。
{"title":"Phillyrin Ameliorates Traumatic Spinal Cord Injury by Inhibiting Inflammation, Oxidative Stress, and Pyroptosis Through Modulation of the Nrf2/HO-1 Signaling Pathway.","authors":"Yang Gao, Yanyang Wang, Liumin Bai, Kunpeng Fu","doi":"10.1080/08820139.2025.2599833","DOIUrl":"https://doi.org/10.1080/08820139.2025.2599833","url":null,"abstract":"<p><strong>Background: </strong>Treating traumatic spinal cord injury (SCI) requires mitigating microglia-mediated inflammation and nerve damage. Phillyrin (phi) shows neuroprotective potential, but its effects on SCI are unknown. We investigated phi's impact on microglial activation, inflammation, neuronal apoptosis, and pyroptosis in mice after SCI.</p><p><strong>Methods: </strong>A mouse contusion SCI model was used to assess phi efficacy. Functional recovery was assessed with BBB and LSS scales. Histological and TUNEL staining were used to evaluate tissue damage and neuronal survival. Microglial phenotypes, inflammation, pyroptosis, oxidative stress and Nrf2/HO-1 pathway were analyzed.</p><p><strong>Results: </strong>Phi treatment accelerated motor function recovery, improved tissue pathology, increased live cells, and reduced apoptosis. Phi treatment showed a decrease in the proportion of M1 pro-inflammatory microglia characterized by iNOS, an increase in the proportion of M2 anti-inflammatory microglia characterized by Arg1 and a reduction in inflammatory factors. Phi enhanced Nrf2 expression in neurons and microglia, activated HO-1 and NQO1 expression, weaken oxidative stress, and improved pyroptosis.</p><p><strong>Conclusion: </strong>Phi alleviates SCI by activating the Nrf2/HO-1 pathway, suppressing microglial activation-mediated neuroinflammation, neuronal apoptosis, oxidative stress and pyroptosis. This study provides the first evidence of the therapeutic effect of phi on traumatic SCI, providing theoretical support for its future clinical application in SCI.</p>","PeriodicalId":13387,"journal":{"name":"Immunological Investigations","volume":" ","pages":"1-16"},"PeriodicalIF":2.4,"publicationDate":"2025-12-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145714066","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Microbiome Mediated Immune Crosstalk on the Gut-Thyroid Axis in Autoimmune Thyroid Disease. 自身免疫性甲状腺疾病中肠道-甲状腺轴上微生物介导的免疫串扰。
IF 2.4 4区 医学 Q3 IMMUNOLOGY Pub Date : 2025-11-26 DOI: 10.1080/08820139.2025.2593335
Mubeen Hussein Arawker, Fitrat Habibullah, Shantanu Baral, Lijun Fu, Ning Sun, Hongting Li, Feihong Ji, Xinguang Qiu

Background: The gut microbiota plays an important role in systemic immune homeostasis and is increasingly implicated in autoimmune thyroid disease (AITD). Evidence suggests that gut dysbiosis, impaired intestinal barrier function, and altered microbial metabolites particularly short-chain fatty acids contribute to immune imbalance along the gut-thyroid axis. Although molecular mimicry between microbial and thyroid antigens has been proposed, current human evidence remains associative rather than causal.

Methods: This review synthesized current observational, translational, and preclinical studies evaluating microbial composition, barrier integrity, microbial metabolites, and immune pathways relevant to AITD. Mechanistic insights into T-lymphocyte regulation and microbial-host interactions were integrated with emerging interventional data.

Results: Gut dysbiosis in AITD is linked to reduced regulatory T-lymphocytes, expansion of Th17 cells, increased intestinal permeability, and the loss of short-chain-fatty-acid-producing taxa. Observational studies consistently report disease-associated taxonomic alterations, while preclinical models support causal pathways through barrier disruption and microbiota-driven immune activation. Early interventional approaches such as high-fiber dietary patterns, probiotics, prebiotics, and experimental fecal microbiota transplantation show modest reductions in thyroid autoantibodies in small trials, though effects are strain-specific, short-term, and not disease-modifying.

Conclusion: Despite largely associative human evidence, converging mechanistic findings highlight the gut microbiota as a modifiable contributor to thyroid autoimmunity. Future priorities include clarifying causality, identifying keystone microbial taxa and metabolites, and establishing standardized interventional frameworks to facilitate translation into endocrine practice.

背景:肠道微生物群在全身免疫稳态中起着重要作用,并且越来越多地与自身免疫性甲状腺疾病(AITD)有关。有证据表明,肠道生态失调、肠道屏障功能受损和微生物代谢物(尤其是短链脂肪酸)的改变导致了肠道-甲状腺轴上的免疫失衡。虽然已经提出了微生物和甲状腺抗原之间的分子模仿,但目前人类的证据仍然是相关的,而不是因果关系。方法:本综述综合了目前的观察、转化和临床前研究,评估了与AITD相关的微生物组成、屏障完整性、微生物代谢物和免疫途径。对t淋巴细胞调节和微生物-宿主相互作用的机制见解与新兴的介入数据相结合。结果:AITD的肠道生态失调与调节性t淋巴细胞减少、Th17细胞扩增、肠道通透性增加以及短链脂肪酸产生分类群的丧失有关。观察性研究一致报告了疾病相关的分类改变,而临床前模型支持通过屏障破坏和微生物群驱动的免疫激活的因果途径。早期干预方法,如高纤维饮食模式、益生菌、益生元和实验性粪便微生物群移植,在小型试验中显示甲状腺自身抗体的适度减少,尽管效果是菌株特异性的、短期的,并且不改变疾病。结论:尽管有大量相关的人类证据,但趋同的机制发现强调肠道微生物群是甲状腺自身免疫的可改变贡献者。未来的优先事项包括澄清因果关系,确定关键的微生物分类群和代谢物,并建立标准化的干预框架,以促进转化为内分泌实践。
{"title":"Microbiome Mediated Immune Crosstalk on the Gut-Thyroid Axis in Autoimmune Thyroid Disease.","authors":"Mubeen Hussein Arawker, Fitrat Habibullah, Shantanu Baral, Lijun Fu, Ning Sun, Hongting Li, Feihong Ji, Xinguang Qiu","doi":"10.1080/08820139.2025.2593335","DOIUrl":"https://doi.org/10.1080/08820139.2025.2593335","url":null,"abstract":"<p><strong>Background: </strong>The gut microbiota plays an important role in systemic immune homeostasis and is increasingly implicated in autoimmune thyroid disease (AITD). Evidence suggests that gut dysbiosis, impaired intestinal barrier function, and altered microbial metabolites particularly short-chain fatty acids contribute to immune imbalance along the gut-thyroid axis. Although molecular mimicry between microbial and thyroid antigens has been proposed, current human evidence remains associative rather than causal.</p><p><strong>Methods: </strong>This review synthesized current observational, translational, and preclinical studies evaluating microbial composition, barrier integrity, microbial metabolites, and immune pathways relevant to AITD. Mechanistic insights into T-lymphocyte regulation and microbial-host interactions were integrated with emerging interventional data.</p><p><strong>Results: </strong>Gut dysbiosis in AITD is linked to reduced regulatory T-lymphocytes, expansion of Th17 cells, increased intestinal permeability, and the loss of short-chain-fatty-acid-producing taxa. Observational studies consistently report disease-associated taxonomic alterations, while preclinical models support causal pathways through barrier disruption and microbiota-driven immune activation. Early interventional approaches such as high-fiber dietary patterns, probiotics, prebiotics, and experimental fecal microbiota transplantation show modest reductions in thyroid autoantibodies in small trials, though effects are strain-specific, short-term, and not disease-modifying.</p><p><strong>Conclusion: </strong>Despite largely associative human evidence, converging mechanistic findings highlight the gut microbiota as a modifiable contributor to thyroid autoimmunity. Future priorities include clarifying causality, identifying keystone microbial taxa and metabolites, and establishing standardized interventional frameworks to facilitate translation into endocrine practice.</p>","PeriodicalId":13387,"journal":{"name":"Immunological Investigations","volume":" ","pages":"1-21"},"PeriodicalIF":2.4,"publicationDate":"2025-11-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145603942","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The Role of IL-12 Family Cytokines in the Pathogenesis of Periodontal Disease: A Therapeutic Approach. IL-12家族细胞因子在牙周病发病中的作用:一种治疗方法。
IF 2.4 4区 医学 Q3 IMMUNOLOGY Pub Date : 2025-11-21 DOI: 10.1080/08820139.2025.2590612
Haichao Wang, Hongyi Zhang

Background: Periodontal disease is a prevalent chronic inflammatory condition of the tooth-supporting tissues characterized by progressive loss of connective attachment and alveolar bone. The IL-12 cytokine family-comprising IL-12, IL-23, IL-27, and IL-35-plays key yet divergent roles in shaping periodontal immune responses through heterodimeric subunits and distinct JAK-STAT signaling pathways. These cytokines differentially regulate inflammation, microbial interactions, and bone resorption.

Methods: This review integrates experimental and clinical evidence assessing IL-12 family members in saliva, gingival crevicular fluid, and periodontal tissues. Studies examining cytokine expression profiles, their correlation with disease activity, microbial dysbiosis, and treatment response were analyzed. Mechanistic data elucidating how IL-12 family signaling modulates inflammatory cascades and osteoclastogenic pathways were also evaluated.

Results: IL-12 and IL-23 predominantly amplify pro-inflammatory responses by promoting Th1/Th17 polarization, enhancing neutrophil recruitment, and driving osteoclastogenesis, thereby linking dysbiotic biofilms to tissue destruction. In contrast, IL-27 and IL-35 exhibit context-dependent immunoregulatory properties, inducing IL-10-mediated anti-inflammatory signaling and expanding regulatory T and B cell compartments to support the resolution of inflammation. Across clinical samples, cytokine levels consistently reflect disease severity and demonstrate modulation following periodontal therapy, underscoring their potential as adjunctive biomarkers.

Conclusion: Members of the IL-12 cytokine family exert both pathogenic and protective influences in periodontal disease. Therapeutic strategies that suppress IL-12/IL-23-driven inflammation while augmenting IL-27/IL-35-mediated regulation show promise as host-modulatory approaches in periodontal treatment. A deeper understanding of these immunologic dynamics may advance precision-based periodontal therapies.

背景:牙周病是一种常见的牙齿支撑组织慢性炎症,其特征是结缔组织和牙槽骨的逐渐丧失。IL-12细胞因子家族包括IL-12、IL-23、IL-27和il -35,通过异二聚体亚基和不同的JAK-STAT信号通路,在形成牙周免疫应答中发挥关键但不同的作用。这些细胞因子以不同的方式调节炎症、微生物相互作用和骨吸收。方法:本综述综合了唾液、龈沟液和牙周组织中IL-12家族成员的实验和临床证据。研究人员分析了细胞因子表达谱及其与疾病活动、微生物生态失调和治疗反应的相关性。机制数据阐明如何IL-12家族信号调节炎症级联反应和破骨生成途径也进行了评估。结果:IL-12和IL-23主要通过促进Th1/Th17极化、增强中性粒细胞募集和驱动破骨细胞生成来增强促炎反应,从而将生态失调的生物膜与组织破坏联系起来。相反,IL-27和IL-35表现出上下文依赖的免疫调节特性,诱导il -10介导的抗炎信号,扩大调节性T细胞和B细胞区室,以支持炎症的解决。在临床样本中,细胞因子水平一致地反映了疾病的严重程度,并在牙周治疗后显示出调节,强调了它们作为辅助生物标志物的潜力。结论:IL-12细胞因子家族成员在牙周病中具有致病和保护作用。抑制IL-12/ il -23驱动的炎症同时增强IL-27/ il -35介导的调节的治疗策略有望成为牙周治疗中宿主调节的方法。对这些免疫动力学的深入了解可能会促进基于精确的牙周治疗。
{"title":"The Role of IL-12 Family Cytokines in the Pathogenesis of Periodontal Disease: A Therapeutic Approach.","authors":"Haichao Wang, Hongyi Zhang","doi":"10.1080/08820139.2025.2590612","DOIUrl":"https://doi.org/10.1080/08820139.2025.2590612","url":null,"abstract":"<p><strong>Background: </strong>Periodontal disease is a prevalent chronic inflammatory condition of the tooth-supporting tissues characterized by progressive loss of connective attachment and alveolar bone. The IL-12 cytokine family-comprising IL-12, IL-23, IL-27, and IL-35-plays key yet divergent roles in shaping periodontal immune responses through heterodimeric subunits and distinct JAK-STAT signaling pathways. These cytokines differentially regulate inflammation, microbial interactions, and bone resorption.</p><p><strong>Methods: </strong>This review integrates experimental and clinical evidence assessing IL-12 family members in saliva, gingival crevicular fluid, and periodontal tissues. Studies examining cytokine expression profiles, their correlation with disease activity, microbial dysbiosis, and treatment response were analyzed. Mechanistic data elucidating how IL-12 family signaling modulates inflammatory cascades and osteoclastogenic pathways were also evaluated.</p><p><strong>Results: </strong>IL-12 and IL-23 predominantly amplify pro-inflammatory responses by promoting Th1/Th17 polarization, enhancing neutrophil recruitment, and driving osteoclastogenesis, thereby linking dysbiotic biofilms to tissue destruction. In contrast, IL-27 and IL-35 exhibit context-dependent immunoregulatory properties, inducing IL-10-mediated anti-inflammatory signaling and expanding regulatory T and B cell compartments to support the resolution of inflammation. Across clinical samples, cytokine levels consistently reflect disease severity and demonstrate modulation following periodontal therapy, underscoring their potential as adjunctive biomarkers.</p><p><strong>Conclusion: </strong>Members of the IL-12 cytokine family exert both pathogenic and protective influences in periodontal disease. Therapeutic strategies that suppress IL-12/IL-23-driven inflammation while augmenting IL-27/IL-35-mediated regulation show promise as host-modulatory approaches in periodontal treatment. A deeper understanding of these immunologic dynamics may advance precision-based periodontal therapies.</p>","PeriodicalId":13387,"journal":{"name":"Immunological Investigations","volume":" ","pages":"1-39"},"PeriodicalIF":2.4,"publicationDate":"2025-11-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145573453","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Immunological Investigations
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:604180095
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1