Mixed reality is an immersive visualization technology based on simultaneous localization and mapping systems and standalone head-mounted displays. It enables seamless integration and dynamic interaction among users, virtual elements, and the physical environment. Although numerous clinical studies and in vitro experiments have confirmed the value of mixed reality in surgical practice, the hierarchy of evidence remains limited. This review draws on published English-language literature to summarize its technical foundations, clinical applications, current innovations, and existing challenges. Specifically, the primary procedures of mixed reality-assisted surgery consist of three-dimensional reconstruction, holographic visualization, and spatial registration. Its clinical applications span preoperative planning, intraoperative navigation, surgical training, and postoperative rehabilitation. However, current limitations include insufficient computational and display capabilities of head-mounted displays, inadequate accuracy in spatial registration, high costs, workflow complexity, and unresolved ethical concerns. Therefore, we recommend increased resource allocation for technological innovations, multicenter randomized controlled clinical trials, and detailed risk-benefit assessments, aiming to establish and validate standardized clinical workflows. As the first comprehensive narrative review to compare the clinical applicability of mixed reality across all surgical specialties, this article outlines future research directions by analyzing representative clinical studies and offers a reliable report on current progress.
{"title":"Advancing Surgical Practice With Mixed Reality: Current Innovations and Future Prospects","authors":"Yifan Ke, Kunpeng Hu","doi":"10.1002/mef2.70043","DOIUrl":"https://doi.org/10.1002/mef2.70043","url":null,"abstract":"<p>Mixed reality is an immersive visualization technology based on simultaneous localization and mapping systems and standalone head-mounted displays. It enables seamless integration and dynamic interaction among users, virtual elements, and the physical environment. Although numerous clinical studies and in vitro experiments have confirmed the value of mixed reality in surgical practice, the hierarchy of evidence remains limited. This review draws on published English-language literature to summarize its technical foundations, clinical applications, current innovations, and existing challenges. Specifically, the primary procedures of mixed reality-assisted surgery consist of three-dimensional reconstruction, holographic visualization, and spatial registration. Its clinical applications span preoperative planning, intraoperative navigation, surgical training, and postoperative rehabilitation. However, current limitations include insufficient computational and display capabilities of head-mounted displays, inadequate accuracy in spatial registration, high costs, workflow complexity, and unresolved ethical concerns. Therefore, we recommend increased resource allocation for technological innovations, multicenter randomized controlled clinical trials, and detailed risk-benefit assessments, aiming to establish and validate standardized clinical workflows. As the first comprehensive narrative review to compare the clinical applicability of mixed reality across all surgical specialties, this article outlines future research directions by analyzing representative clinical studies and offers a reliable report on current progress.</p>","PeriodicalId":74135,"journal":{"name":"MedComm - Future medicine","volume":"4 4","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-12-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mef2.70043","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145887289","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Air pollution poses a significant threat to respiratory health globally, contributing to the development and worsening of diseases, such as asthma, chronic obstructive pulmonary disease, pulmonary fibrosis, and lung cancer. A key mechanism behind this involves epigenetic reprogramming, where environmental exposures alter gene activity without changing the underlying DNA sequence itself. This review deciphers the multilayered epigenetic mechanisms linking pollutants (e.g., PM2.5, cigarette smoke, and ozone) to respiratory pathology, emphasizing reversible modifications that bridge environmental exposure and disease phenotypes. Air pollution-induced epigenetic reprogramming regulates critical biological processes, such as immune imbalance, chronic inflammation, oxidative stress, cellular dysfunction (senescence, apoptosis, and ferroptosis), tissue remodeling (epithelial–mesenchymal transition and fibrosis), and genomic instability. Specifically, aberrant DNA methylation, dysregulated RNA methylation, perturbed noncoding RNA networks, and histone modification abnormalities collectively drive disease pathogenesis. Furthermore, emerging epigenetic therapies targeting these modifications, such as DNA methyltransferase inhibitors (5-AZADC), histone deacetylase inhibitors, and RNA methylation regulators (methyltransferase-like 3 inhibitor STM2457), show promising therapeutic potential. This review highlights the reversibility of epigenetic changes as a strategic basis for intervention, emphasizing the need for future research on mixed exposures, tissue-specific mechanisms, and clinical translation to mitigate the global burden of pollution-related respiratory diseases.
{"title":"Air Pollution-Induced Epigenetic Regulation in Respiratory Diseases: Mechanisms, Dysregulation, and Therapeutic Opportunities","authors":"Ruitong Zeng, Jiliu Liu, Guoping Li, Junyi Wang, Anying Xiong, Ying Xiong, Xiang He","doi":"10.1002/mef2.70044","DOIUrl":"https://doi.org/10.1002/mef2.70044","url":null,"abstract":"<p>Air pollution poses a significant threat to respiratory health globally, contributing to the development and worsening of diseases, such as asthma, chronic obstructive pulmonary disease, pulmonary fibrosis, and lung cancer. A key mechanism behind this involves epigenetic reprogramming, where environmental exposures alter gene activity without changing the underlying DNA sequence itself. This review deciphers the multilayered epigenetic mechanisms linking pollutants (e.g., PM<sub>2.5</sub>, cigarette smoke, and ozone) to respiratory pathology, emphasizing reversible modifications that bridge environmental exposure and disease phenotypes. Air pollution-induced epigenetic reprogramming regulates critical biological processes, such as immune imbalance, chronic inflammation, oxidative stress, cellular dysfunction (senescence, apoptosis, and ferroptosis), tissue remodeling (epithelial–mesenchymal transition and fibrosis), and genomic instability. Specifically, aberrant DNA methylation, dysregulated RNA methylation, perturbed noncoding RNA networks, and histone modification abnormalities collectively drive disease pathogenesis. Furthermore, emerging epigenetic therapies targeting these modifications, such as DNA methyltransferase inhibitors (5-AZADC), histone deacetylase inhibitors, and RNA methylation regulators (methyltransferase-like 3 inhibitor STM2457), show promising therapeutic potential. This review highlights the reversibility of epigenetic changes as a strategic basis for intervention, emphasizing the need for future research on mixed exposures, tissue-specific mechanisms, and clinical translation to mitigate the global burden of pollution-related respiratory diseases.</p>","PeriodicalId":74135,"journal":{"name":"MedComm - Future medicine","volume":"4 4","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-12-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mef2.70044","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145824856","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Traditional Chinese Medicine (TCM), with its fragmented knowledge system, subjective diagnostics, and limited standardization, faces ongoing challenges in modernization. The rapid development of Large Language Models (LLMs) offers an unprecedented opportunity for the digitalization and standardization of TCM knowledge. By leveraging deep semantic understanding, contextual reasoning, and knowledge-graph–based inference, LLMs can systematize knowledge, integrate multimodal data, and support both standardized and personalized decision-making. Several TCM–LLMs, such as Qibo and MedChatZH, have been developed. Although previous reviews compared individual TCM–LLMs, a comprehensive analysis of common characteristics is lacking. This review addresses this gap by examining the “construction–application–challenges–prospects” paradigm of TCM–LLMs. Construction generally involves collecting authoritative data from diverse sources, standardizing textual content, training and fine-tuning on foundational LLMs, and subsequent evaluation. Key applications include auxiliary diagnosis and treatment, health management, medical education, and drug discovery. Five major challenges are data quality, reasoning performance, multimodal integration, ethical and regulatory compliance, and cultural adaptability. On the basis of this analysis, we propose a classification scheme that categorizes applications into four types and adopt the identified challenges as evaluation dimensions for future TCM–LLMs. This framework aims to clarify conceptual structures, guide methodology, and provide a foundation for innovation and cross-cultural integration in TCM research.
{"title":"Artificial Intelligence in Traditional Chinese Medicine: Bridging Ancient Practice and Future Innovation","authors":"Zhehan Jiang, Quanming Peng, Li Li, Shate Xiang","doi":"10.1002/mef2.70042","DOIUrl":"https://doi.org/10.1002/mef2.70042","url":null,"abstract":"<p>Traditional Chinese Medicine (TCM), with its fragmented knowledge system, subjective diagnostics, and limited standardization, faces ongoing challenges in modernization. The rapid development of Large Language Models (LLMs) offers an unprecedented opportunity for the digitalization and standardization of TCM knowledge. By leveraging deep semantic understanding, contextual reasoning, and knowledge-graph–based inference, LLMs can systematize knowledge, integrate multimodal data, and support both standardized and personalized decision-making. Several TCM–LLMs, such as Qibo and MedChatZH, have been developed. Although previous reviews compared individual TCM–LLMs, a comprehensive analysis of common characteristics is lacking. This review addresses this gap by examining the “construction–application–challenges–prospects” paradigm of TCM–LLMs. Construction generally involves collecting authoritative data from diverse sources, standardizing textual content, training and fine-tuning on foundational LLMs, and subsequent evaluation. Key applications include auxiliary diagnosis and treatment, health management, medical education, and drug discovery. Five major challenges are data quality, reasoning performance, multimodal integration, ethical and regulatory compliance, and cultural adaptability. On the basis of this analysis, we propose a classification scheme that categorizes applications into four types and adopt the identified challenges as evaluation dimensions for future TCM–LLMs. This framework aims to clarify conceptual structures, guide methodology, and provide a foundation for innovation and cross-cultural integration in TCM research.</p>","PeriodicalId":74135,"journal":{"name":"MedComm - Future medicine","volume":"4 4","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-12-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mef2.70042","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145750836","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
<p>In a recent publication in <i>Cell</i>, Wong et al. presented an optogenetic system for screening compounds that specifically modulate the integrated stress response (ISR) [<span>1</span>]. The authors identified eight non-cytotoxic ISR enhancers as broad-spectrum antiviral agents and revealed their key mechanism: the selective targeting of general control nonderepressible 2 (GCN2) to upregulate activating transcription factor 4 (ATF4) expression, thereby sensitizing cells to stress and apoptosis [<span>1</span>].</p><p>Optogenetics enables precise spatiotemporal control of cellular activity by conferring light sensitivity via heterologous expression of photosensitive proteins. This approach is increasingly being integrated with synthetic biology to facilitate novel paradigms in phenotypic drug discovery [<span>2</span>]. Meanwhile, the ISR pathway represents a conserved signaling pathway activated by four stress sensor kinases—heme-regulated inhibitor (HRI), protein kinase R (PKR), protein kinase R-like ER kinase (PERK), and GCN2—which respond to diverse stressors such as viral double-stranded RNA [<span>3</span>]. Upon activation, these kinases phosphorylate the eukaryotic translation initiation factor 2 subunit alpha (eIF2α), leading to the selective translation of ISR-related proteins, such as ATF4, C/EBP homologous protein (CHOP), and growth arrest and DNA damage-inducible protein 34 (GADD34), thereby modulating cell survival and function. Given this regulatory capacity, ISR-enhancing compounds represent a novel strategy for the development of broad-spectrum antiviral therapeutics [<span>3</span>]. The combination of optogenetics-driven precise manipulation and deeper exploration of the ISR network—particularly its crosstalk with other signaling pathways—may open breakthrough directions for future broad-spectrum antiviral research.</p><p>To achieve precise control of the ISR pathway, Taivan et al. developed an optogenetic platform that dynamically stimulates the ISR signaling using a light-activated optogenetic PKR (opto-PKR) [<span>4</span>]. The dsRBM1 and dsRBM2 regions of PKR were replaced with an optimized mutant of the <i>Arabidopsis</i> blue light receptor Cry, namely Cry2Olig (E490G). Upon transduction of opto-PKR into cells, exposure to blue light prompted Cry2 aggregation, inducing in PKR oligomerization, kinase activation, and subsequent initiation of ISR. This light-controlled system simulates PKR-mediated ISR activation as observed during viral infection, while avoiding off-target cytotoxicity [<span>4</span>]. The platform's efficacy was validated through both pharmacological activators and inhibitors of the ISR pathway (Figure 1a) [<span>1</span>]. Crucially, unlike traditional small-molecule stressors that often cause cross-pathway interference, this approach minimizes such off-target effects and offers immediate deactivation in darkness, enabling transient response unattainable with conventional small-molecule activators.
{"title":"Optogenetic Regulation of Integrated Stress Responses: Developing Novel Broad-Spectrum Antiviral Strategies","authors":"Shizhan Cui, Zehan Pang, Bixia Hong","doi":"10.1002/mef2.70040","DOIUrl":"https://doi.org/10.1002/mef2.70040","url":null,"abstract":"<p>In a recent publication in <i>Cell</i>, Wong et al. presented an optogenetic system for screening compounds that specifically modulate the integrated stress response (ISR) [<span>1</span>]. The authors identified eight non-cytotoxic ISR enhancers as broad-spectrum antiviral agents and revealed their key mechanism: the selective targeting of general control nonderepressible 2 (GCN2) to upregulate activating transcription factor 4 (ATF4) expression, thereby sensitizing cells to stress and apoptosis [<span>1</span>].</p><p>Optogenetics enables precise spatiotemporal control of cellular activity by conferring light sensitivity via heterologous expression of photosensitive proteins. This approach is increasingly being integrated with synthetic biology to facilitate novel paradigms in phenotypic drug discovery [<span>2</span>]. Meanwhile, the ISR pathway represents a conserved signaling pathway activated by four stress sensor kinases—heme-regulated inhibitor (HRI), protein kinase R (PKR), protein kinase R-like ER kinase (PERK), and GCN2—which respond to diverse stressors such as viral double-stranded RNA [<span>3</span>]. Upon activation, these kinases phosphorylate the eukaryotic translation initiation factor 2 subunit alpha (eIF2α), leading to the selective translation of ISR-related proteins, such as ATF4, C/EBP homologous protein (CHOP), and growth arrest and DNA damage-inducible protein 34 (GADD34), thereby modulating cell survival and function. Given this regulatory capacity, ISR-enhancing compounds represent a novel strategy for the development of broad-spectrum antiviral therapeutics [<span>3</span>]. The combination of optogenetics-driven precise manipulation and deeper exploration of the ISR network—particularly its crosstalk with other signaling pathways—may open breakthrough directions for future broad-spectrum antiviral research.</p><p>To achieve precise control of the ISR pathway, Taivan et al. developed an optogenetic platform that dynamically stimulates the ISR signaling using a light-activated optogenetic PKR (opto-PKR) [<span>4</span>]. The dsRBM1 and dsRBM2 regions of PKR were replaced with an optimized mutant of the <i>Arabidopsis</i> blue light receptor Cry, namely Cry2Olig (E490G). Upon transduction of opto-PKR into cells, exposure to blue light prompted Cry2 aggregation, inducing in PKR oligomerization, kinase activation, and subsequent initiation of ISR. This light-controlled system simulates PKR-mediated ISR activation as observed during viral infection, while avoiding off-target cytotoxicity [<span>4</span>]. The platform's efficacy was validated through both pharmacological activators and inhibitors of the ISR pathway (Figure 1a) [<span>1</span>]. Crucially, unlike traditional small-molecule stressors that often cause cross-pathway interference, this approach minimizes such off-target effects and offers immediate deactivation in darkness, enabling transient response unattainable with conventional small-molecule activators.","PeriodicalId":74135,"journal":{"name":"MedComm - Future medicine","volume":"4 4","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-12-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mef2.70040","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145695083","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Mengyi Zhu, Haoxuan Wang, Haishan Lin, Guibin Hong, Yun Wang, Fan Jiang, Ye Xie, Runnan Shen, Hongkun Yang, Shaoxu Wu
Urological cancers represent a significant and growing global health challenge. This study aims to provide a comprehensive assessment of the global burden, trends, and inequalities of prostate, bladder, and kidney cancers from 1990 to 2021, and to project their future burden to 2050. Utilizing data from the Global Burden of Disease Study 2021, we analyzed incidence, prevalence, mortality, and disability-adjusted life years (DALYs). We employed joinpoint regression to analyze temporal trends, frontier analysis to evaluate healthcare efficiency, and Bayesian age-period-cohort models to project future burden. Risk factor attributions were also quantified. In 2021, prostate cancer demonstrated the highest age-standardized rates across all metrics. From 1990 to 2021, age-standardized mortality and DALY rates declined for all three cancers. The burden was disproportionately higher among males and older populations, with smoking, high fasting plasma glucose, and high body-mass index identified as leading risk factors. Projections to 2050 indicate a continued decline in all age-standardized rates globally. Despite favorable trends in standardized rates, persistent sex disparities, growing absolute case numbers due to population aging, and the influence of modifiable risk factors necessitate targeted public health interventions and strategic healthcare planning.
{"title":"Global Burden and Future Projections of Urological Cancers: A Comprehensive Analysis From the Global Burden of Disease Study 2021","authors":"Mengyi Zhu, Haoxuan Wang, Haishan Lin, Guibin Hong, Yun Wang, Fan Jiang, Ye Xie, Runnan Shen, Hongkun Yang, Shaoxu Wu","doi":"10.1002/mef2.70041","DOIUrl":"https://doi.org/10.1002/mef2.70041","url":null,"abstract":"<p>Urological cancers represent a significant and growing global health challenge. This study aims to provide a comprehensive assessment of the global burden, trends, and inequalities of prostate, bladder, and kidney cancers from 1990 to 2021, and to project their future burden to 2050. Utilizing data from the Global Burden of Disease Study 2021, we analyzed incidence, prevalence, mortality, and disability-adjusted life years (DALYs). We employed joinpoint regression to analyze temporal trends, frontier analysis to evaluate healthcare efficiency, and Bayesian age-period-cohort models to project future burden. Risk factor attributions were also quantified. In 2021, prostate cancer demonstrated the highest age-standardized rates across all metrics. From 1990 to 2021, age-standardized mortality and DALY rates declined for all three cancers. The burden was disproportionately higher among males and older populations, with smoking, high fasting plasma glucose, and high body-mass index identified as leading risk factors. Projections to 2050 indicate a continued decline in all age-standardized rates globally. Despite favorable trends in standardized rates, persistent sex disparities, growing absolute case numbers due to population aging, and the influence of modifiable risk factors necessitate targeted public health interventions and strategic healthcare planning.</p>","PeriodicalId":74135,"journal":{"name":"MedComm - Future medicine","volume":"4 4","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-11-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mef2.70041","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145619225","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Yilong Lin, Yuning Wang, Ruidan Zhao, Liyi Zhang, Qingmo Yang
<p>Individuals suffering from major depressive disorder (MDD) have been reported to be more susceptible to cancer. A recent meta-analysis revealed that individuals with MDD or anxiety had a moderately elevated likelihood of developing cancer (RR = 1.13, 95% CI = 1.06–1.19) and a higher probability of cancer-related death (RR = 1.21, 95% CI = 1.16–1.26) [<span>1</span>]. The increased susceptibility to cancer among individuals with MDD may arise from multiple mechanisms. On one hand, patients suffering from this disorder are often less likely to participate in early cancer detection and screening programs. On the other hand, dysregulation of neuroendocrine pathways in MDD may further contribute to tumor progression by altering immune function, promoting chronic inflammation, and affecting tumor biology [<span>2</span>].</p><p>Specially, individuals with MDD also had a higher risk of breast cancer compared to those without MDD [<span>3</span>], and those patients with both diseases tended to have poorer BC outcomes [<span>2</span>]. In addition, up to 60% of women experience depression before and throughout their BC diagnosis, affecting women of all races and ages [<span>2</span>]. However, these observational studies may have been affected by confounding factors and biases.</p><p>In this study, we employed a genome-wide cross-trait analysis to chart the shared genetic architecture between BC and MDD, further exploring the genetic and phenotypic relationships. The overview of this study was shown in Figure S1. The genome-wide association studies (GWAS) data of BC and MDD were obtained from The Breast Cancer Association Consortium and Discovery, Biology and Risk of Inherited Variants in Breast Cancer Consortium, and Psychiatric Genomics Consortium. First, we conducted a genetic correlation analysis using linkage disequilibrium score regression (LDSC). Then, we employed two cross-trait meta-analysis methods (the multi-trait analysis of GWAS [MTAG] and the Cross-Phenotype Association analysis [CPASSOC]) to identify common risk SNPs associated with BC and MDD. In addition, pleiotropic analysis under composite null hypothesis (PLACO) was used to identify pleiotropic loci associated with BC and MDD. Based on PLACO analysis, we identified potential pleiotropic loci and genes through Functional Mapping and Annotation of Genetic Associations (FUMA) tool and multimarker analysis of GenoMic annotation (MAGMA) analysis. Finally, we conducted Generalized summary-data-based Mendelian randomization (GSMR) and two-sample Mendelian Randomization (TSMR) to identify the causal associations. MR analysis relies on three core assumptions: (1) the genetic variants used as instrumental variables (IVs) are strongly associated with the exposure; (2) these IVs are independent of confounding factors; and (3) they affect the outcome only through the exposure. The SNPs were selected based on commonly accepted criteria in MR studies: (i) genome-wide significance with a <i>p</i>
据报道,患有重度抑郁症(MDD)的人更容易患癌症。最近的一项荟萃分析显示,患有重度抑郁症或焦虑症的个体患癌症的可能性中等升高(RR = 1.13, 95% CI = 1.06-1.19),癌症相关死亡的可能性较高(RR = 1.21, 95% CI = 1.16-1.26)。重度抑郁症患者对癌症的易感性增加可能有多种机制。一方面,患有这种疾病的患者通常不太可能参与早期癌症检测和筛查项目。另一方面,MDD中神经内分泌通路的失调可能通过改变免疫功能、促进慢性炎症和影响肿瘤生物学来进一步促进肿瘤进展。特别的是,患有重度抑郁症的人患乳腺癌的风险也比没有重度抑郁症的人高,而且患有这两种疾病的患者往往有较差的BC预后。此外,高达60%的女性在BC诊断之前和整个过程中都经历过抑郁症,影响到所有种族和年龄的女性。然而,这些观察性研究可能受到混杂因素和偏差的影响。在这项研究中,我们采用全基因组交叉性状分析来绘制BC和MDD之间的共享遗传结构,进一步探索遗传和表型关系。本研究概述如图S1所示。BC和MDD的全基因组关联研究(GWAS)数据来自乳腺癌协会联盟、乳腺癌协会的发现、生物学和遗传变异风险以及精神病学基因组学联盟。首先,我们使用连锁不平衡评分回归(LDSC)进行了遗传相关分析。然后,我们采用两种跨性状荟萃分析方法(GWAS的多性状分析[MTAG]和跨表型关联分析[CPASSOC])来确定与BC和MDD相关的常见风险snp。此外,采用复合零假设下的多效性分析(PLACO)来鉴定与BC和MDD相关的多效性位点。基于PLACO分析,我们通过功能定位和遗传关联注释(fua)工具和基因组注释的多标记分析(MAGMA)分析确定了潜在的多效位点和基因。最后,我们采用基于广义汇总数据的孟德尔随机化(GSMR)和双样本孟德尔随机化(TSMR)来确定因果关系。核磁共振分析依赖于三个核心假设:(1)作为工具变量(IVs)的遗传变异与暴露密切相关;(2)这些IVs独立于混杂因素;(3)它们仅通过暴露影响结果。根据MR研究中普遍接受的标准选择snp:(i)全基因组显著性,p值为5 × 10−8;(ii)与其他变异的独立性,通过在10mb窗口内去除LD中R2≥0.001的snp来确保;(iii)仪器强度,需要f统计量。无约束截距LDSC分析显示,乳腺癌与MDD之间存在全基因组正相关(rg = 0.0545, SE = 0.0151, P = 3 × 10−4)。随后的敏感性分析与约束截距证实了正遗传相关性(rg = 0.0545, SE = 0.0151, P = 7 × 10−4)。使用MTAG方法,我们分析了BC和MDD的GWAS数据,鉴定出17个具有全基因组意义的snp (P < 5 × 10−8)(表S1)。为了验证MTAG的发现,我们使用CPASSOC进行了多重性分析,鉴定出16470个具有全基因组意义的snp (P < 5 × 10−8)(表S2)。通过PLACO分析,我们鉴定出775个snp具有符合基因组意义的潜在多态性变异(图S2A,表S3)。整合MTAG、CPASSOC和PLACO分析结果后,我们发现MTAG鉴定为全基因组显著的17个snp在CPASSOC和PLACO分析中也保持其显著性(表S4)。基于PLACO结果,MAGMA组织表达分析显示成纤维细胞可能参与了BC和MDD之间的关联(图S2B)。此外,snp对基因的功能影响主要集中在内含子区和基因间区(图1A)。fua分析显示22个独立的基因组风险位点分布在22个不同的染色体区域(图S2C,表S5)。MAGMA分析鉴定出130个显著的多效性基因(表S6)。Gene Ontology结果表明,这些基因在嗅觉感知、钠离子跨膜转运、脂肪细胞增殖调控、成纤维细胞生长因子受体结合等方面发挥着重要作用(图S2D,表S7)。 京都基因与基因组百科全书(KEGG)结果显示,嗅觉转导、wnt信号通路和p53信号通路可能参与了BC和MDD之间的关联(图1B)。此外,这些基因可能导致其他癌症,包括黑色素瘤和胃癌(图1B)。虽然嗅觉转导最初可能与MDD-BC合并症无关,但先前的研究已经确定了嗅觉受体(ORs)与乳腺癌进展之间的潜在联系。例如,据报道,两个OR基因OR2W3和OR2B6可能与乳腺癌的进展有关。此外,抑郁症与嗅觉功能受损密切相关,而嗅觉功能受损往往伴随着显著的社交功能障碍。总之,我们的富集分析表明,MDD和BC发展的共同神经内分泌或感觉信号失调。最后,我们通过MR分析探讨了BC与MDD之间的因果关系。GSMR分析显示,MDD患者发生BC的风险显著增加(OR = 1.27, 95% CI: [1.11-1.46], P = 5.90 × 10−4,图1C,D)。TSMR的逆方差加权(IVW)方法验证了因果关系(OR = 1.12, 95% CI: [1.02-1.22], P = 5.90 × 10−4,图1C,E)。虽然MR-Egger回归和加权中位数方法的结果没有统计学意义,但它们的效应估计与IVW分析的结果方向一致,支持IVW研究结果的稳健性(表S8)。敏感性分析提示TSMR结果不受水平多效性的影响(p = 0.75)。然而,我们在任何方法中都没有发现BC对MDD的显著因果关系(图1C, p > 0.05)。因此,这些MR结果提供了一致的证据,表明MDD可能会导致BC的风险增加,强调了从MDD到肿瘤发展的潜在单向因果途径。鉴于几个方法学上的局限性,我们应该谨慎地解释MR的发现。尽管我们采用了严格的聚集阈值,并使用了多种互补的MR方法,包括IVW、MR- egger回归和加权中位数,以减轻潜在的水平多效性,但残留偏倚不能完全排除。值得注意的是,水平多效性试验没有显示显著的水平多效性,MR-Egger和加权中位数方法的效应估计与IVW结果方向一致,支持了我们研究结果的稳健性。此外,所有数据集都来自欧洲血统的人群,这最大限度地减少了人口分层的影响,但可能会限制我们的结果推广到其他种族群体。最后,观察到的混杂因素,如生活方式或社会经济因素,不能直接被遗传工具捕获,仍然可能有助于观察到的关联。我们的研究绘制了BC和MDD之间的共同遗传结构,并探讨了它们之间潜在的因果关系。这些发现加强了对BC和MDD共同涉及的遗传途径的理解,突出了它们之间复杂的相互联系。这项研究为旨在开发新的BC和MDD预防措施和治疗干预措施的实验研究提供了有价值的见解。林一龙撰写了初稿,并分析了主要数据。王玉宁和赵瑞丹收集数据。张立毅和杨庆谟对论文进行了修改,并监督了整个研究过程。所有作者都阅读并批准了最终稿件。本研究利用了来自参与者研究的公开数据,这些数据已经得到了人体实验伦理标准委员会的批准,从而消除了单独的伦理批准的需要。不适用。作者声明无利益冲突。支持本研究结果的数据可在本文的支持材料中获得。
{"title":"Charting the Shared Genetic Architecture Between Breast Cancer and Major Depressive Disorder","authors":"Yilong Lin, Yuning Wang, Ruidan Zhao, Liyi Zhang, Qingmo Yang","doi":"10.1002/mef2.70039","DOIUrl":"https://doi.org/10.1002/mef2.70039","url":null,"abstract":"<p>Individuals suffering from major depressive disorder (MDD) have been reported to be more susceptible to cancer. A recent meta-analysis revealed that individuals with MDD or anxiety had a moderately elevated likelihood of developing cancer (RR = 1.13, 95% CI = 1.06–1.19) and a higher probability of cancer-related death (RR = 1.21, 95% CI = 1.16–1.26) [<span>1</span>]. The increased susceptibility to cancer among individuals with MDD may arise from multiple mechanisms. On one hand, patients suffering from this disorder are often less likely to participate in early cancer detection and screening programs. On the other hand, dysregulation of neuroendocrine pathways in MDD may further contribute to tumor progression by altering immune function, promoting chronic inflammation, and affecting tumor biology [<span>2</span>].</p><p>Specially, individuals with MDD also had a higher risk of breast cancer compared to those without MDD [<span>3</span>], and those patients with both diseases tended to have poorer BC outcomes [<span>2</span>]. In addition, up to 60% of women experience depression before and throughout their BC diagnosis, affecting women of all races and ages [<span>2</span>]. However, these observational studies may have been affected by confounding factors and biases.</p><p>In this study, we employed a genome-wide cross-trait analysis to chart the shared genetic architecture between BC and MDD, further exploring the genetic and phenotypic relationships. The overview of this study was shown in Figure S1. The genome-wide association studies (GWAS) data of BC and MDD were obtained from The Breast Cancer Association Consortium and Discovery, Biology and Risk of Inherited Variants in Breast Cancer Consortium, and Psychiatric Genomics Consortium. First, we conducted a genetic correlation analysis using linkage disequilibrium score regression (LDSC). Then, we employed two cross-trait meta-analysis methods (the multi-trait analysis of GWAS [MTAG] and the Cross-Phenotype Association analysis [CPASSOC]) to identify common risk SNPs associated with BC and MDD. In addition, pleiotropic analysis under composite null hypothesis (PLACO) was used to identify pleiotropic loci associated with BC and MDD. Based on PLACO analysis, we identified potential pleiotropic loci and genes through Functional Mapping and Annotation of Genetic Associations (FUMA) tool and multimarker analysis of GenoMic annotation (MAGMA) analysis. Finally, we conducted Generalized summary-data-based Mendelian randomization (GSMR) and two-sample Mendelian Randomization (TSMR) to identify the causal associations. MR analysis relies on three core assumptions: (1) the genetic variants used as instrumental variables (IVs) are strongly associated with the exposure; (2) these IVs are independent of confounding factors; and (3) they affect the outcome only through the exposure. The SNPs were selected based on commonly accepted criteria in MR studies: (i) genome-wide significance with a <i>p</i> ","PeriodicalId":74135,"journal":{"name":"MedComm - Future medicine","volume":"4 4","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-11-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mef2.70039","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145625976","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
The long-term survival benefit of neoadjuvant therapy (NAT) in breast cancer patients eligible for breast-conserving surgery (BCS) remains uncertain. This retrospective cohort study analyzed 94,677 BCS-eligible patients from the SEER database (2010–2020), including 8565 who received NAT. After propensity score matching (n = 5734 each), NAT significantly improved overall survival (OS) only in patients with triple-negative (HR = 0.79), ER-negative (HR = 0.80), and stage IIA (HR = 0.81) disease. No OS benefit was observed in HER2-positive patients despite high response rates. To guide treatment decisions, two machine learning models using Random Survival Forest were developed to predict 5-year OS, showing good discrimination (C-index: 0.743 for BCS, 0.690 for NAT-BCS). SHAP analysis identified age, stage, and breast subtype as key prognostic factors. Cross-stratification based on predicted OS revealed that 8.9% of BCS patients could benefit from NAT, while 90.8% of NAT-BCS patients might safely omit it. Patients whose treatment matched model recommendations had significantly better survival. These findings suggest that NAT provides limited survival benefit in BCS-eligible patients, with the advantage concentrated in specific subgroups. Predictive modeling offers a clinically useful approach to personalize NAT use, potentially reducing unnecessary treatment while identifying those most likely to benefit.
{"title":"Evaluating the Survival Impact of Neoadjuvant Therapy and Development of Personalized Machine Learning Survival Predictive Model for Breast Cancer Patients Eligible for Breast-Conserving Surgery","authors":"Zhicheng Du, Yongqu Zhang, Weibin Li, Xue Zhao, Xueqi Fan, Jingwen Bai, Guojun Zhang","doi":"10.1002/mef2.70036","DOIUrl":"https://doi.org/10.1002/mef2.70036","url":null,"abstract":"<p>The long-term survival benefit of neoadjuvant therapy (NAT) in breast cancer patients eligible for breast-conserving surgery (BCS) remains uncertain. This retrospective cohort study analyzed 94,677 BCS-eligible patients from the SEER database (2010–2020), including 8565 who received NAT. After propensity score matching (<i>n</i> = 5734 each), NAT significantly improved overall survival (OS) only in patients with triple-negative (HR = 0.79), ER-negative (HR = 0.80), and stage IIA (HR = 0.81) disease. No OS benefit was observed in HER2-positive patients despite high response rates. To guide treatment decisions, two machine learning models using Random Survival Forest were developed to predict 5-year OS, showing good discrimination (C-index: 0.743 for BCS, 0.690 for NAT-BCS). SHAP analysis identified age, stage, and breast subtype as key prognostic factors. Cross-stratification based on predicted OS revealed that 8.9% of BCS patients could benefit from NAT, while 90.8% of NAT-BCS patients might safely omit it. Patients whose treatment matched model recommendations had significantly better survival. These findings suggest that NAT provides limited survival benefit in BCS-eligible patients, with the advantage concentrated in specific subgroups. Predictive modeling offers a clinically useful approach to personalize NAT use, potentially reducing unnecessary treatment while identifying those most likely to benefit.</p>","PeriodicalId":74135,"journal":{"name":"MedComm - Future medicine","volume":"4 4","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-11-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mef2.70036","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145572234","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
In the recent article published in Cell [1], Kaltenecker et al. systematically uncovered the liver as an active pathological driver of cancer cachexia (CCx). Through integrated multi-omics analysis, the study identifies the downregulation of the circadian regulator REV-ERBα (nuclear receptor subfamily 1 group D member 1, NR1D1) as a central mechanism that disrupts hepatic homeostasis and induces abnormal secretion of hepatokines, thereby promoting peripheral tissue wasting. These findings provide a conceptual framework for developing therapeutic strategies targeting the “liver–peripheral tissue” axis in CCx.
Cancer cachexia is a highly catabolic metabolic syndrome associated with advanced malignancies, characterized by progressive weight loss, skeletal muscle atrophy, and adipose tissue degradation [2, 3]. It is particularly prevalent in pancreatic, gastric, and lung cancers, affecting 50%–80% of patients and accounting for approximately 20% of cancer-related deaths. While previous studies have predominantly focused on local mechanisms of muscle atrophy, such as the ubiquitin-proteasome system (UPS), autophagy, and the NF-κB or FOXO pathways, the systemic metabolic functions of the liver in CCx pathogenesis remain underexplored. Notably, a concurrent study by Garrett et al. published in Cell demonstrated that tumor-driven inflammation can alter the vagal nerve–liver axis, resulting in suppression of hepatic HNF4α and exacerbation of systemic catabolism, further highlighting the critical involvement of the liver in this condition [4].
Given the pivotal role of the liver in systemic metabolism, the authors hypothesized that tissue wasting in cachexia may originate from hepatic responses to tumor-derived signals, which drive peripheral tissue catabolism through the secretion of specific factors. To investigate this, Kaltenecker et al. performed integrated transcriptomic and epigenomic profiling of hepatocytes from both weight-stable and cachectic cancer mouse models. These analyses uncovered a distinct transcriptional program specific to cachexia, characterized by dysregulated expression of the circadian regulator REV-ERBα. Hepatic restoration of REV-ERBα expression markedly attenuated both muscle and fat wasting. Mechanistically, REV-ERBα modulates tissue degradation by controlling hepatokines that activate catabolic pathways in myotubes and adipocytes. Notably, clinical data further revealed elevated circulating levels of key hepatokines in cachectic patients, independent of classical inflammatory cytokines such as IL-6 or TNF-α, underscoring a novel “hepatokine-peripheral tissue” axis in CCx pathophysiology.
Delving further into molecular effectors, the authors identified three key liver-secreted proteins-lipopolysaccharide-binding protein (LBP), inter-α-trypsin inhibitor heavy-chain H3 (ITIH3), and insulin-like-growth-factor-binding protein 1 (IGFBP1)–as direct targets of REV-ERB
{"title":"REV-ERBα–Mediated Hepatic Regulation of Cachexia: A Circadian-Metabolic Axis of Tissue Wasting","authors":"Wenhui Wang, Junke Song, Gaofei Wei","doi":"10.1002/mef2.70038","DOIUrl":"https://doi.org/10.1002/mef2.70038","url":null,"abstract":"<p>In the recent article published in <i>Cell</i> [<span>1</span>], Kaltenecker et al. systematically uncovered the liver as an active pathological driver of cancer cachexia (CCx). Through integrated multi-omics analysis, the study identifies the downregulation of the circadian regulator REV-ERBα (nuclear receptor subfamily 1 group D member 1, NR1D1) as a central mechanism that disrupts hepatic homeostasis and induces abnormal secretion of hepatokines, thereby promoting peripheral tissue wasting. These findings provide a conceptual framework for developing therapeutic strategies targeting the “liver–peripheral tissue” axis in CCx.</p><p>Cancer cachexia is a highly catabolic metabolic syndrome associated with advanced malignancies, characterized by progressive weight loss, skeletal muscle atrophy, and adipose tissue degradation [<span>2, 3</span>]. It is particularly prevalent in pancreatic, gastric, and lung cancers, affecting 50%–80% of patients and accounting for approximately 20% of cancer-related deaths. While previous studies have predominantly focused on local mechanisms of muscle atrophy, such as the ubiquitin-proteasome system (UPS), autophagy, and the NF-κB or FOXO pathways, the systemic metabolic functions of the liver in CCx pathogenesis remain underexplored. Notably, a concurrent study by Garrett et al. published in Cell demonstrated that tumor-driven inflammation can alter the vagal nerve–liver axis, resulting in suppression of hepatic HNF4α and exacerbation of systemic catabolism, further highlighting the critical involvement of the liver in this condition [<span>4</span>].</p><p>Given the pivotal role of the liver in systemic metabolism, the authors hypothesized that tissue wasting in cachexia may originate from hepatic responses to tumor-derived signals, which drive peripheral tissue catabolism through the secretion of specific factors. To investigate this, Kaltenecker et al. performed integrated transcriptomic and epigenomic profiling of hepatocytes from both weight-stable and cachectic cancer mouse models. These analyses uncovered a distinct transcriptional program specific to cachexia, characterized by dysregulated expression of the circadian regulator REV-ERBα. Hepatic restoration of REV-ERBα expression markedly attenuated both muscle and fat wasting. Mechanistically, REV-ERBα modulates tissue degradation by controlling hepatokines that activate catabolic pathways in myotubes and adipocytes. Notably, clinical data further revealed elevated circulating levels of key hepatokines in cachectic patients, independent of classical inflammatory cytokines such as IL-6 or TNF-α, underscoring a novel “hepatokine-peripheral tissue” axis in CCx pathophysiology.</p><p>Delving further into molecular effectors, the authors identified three key liver-secreted proteins-lipopolysaccharide-binding protein (LBP), inter-α-trypsin inhibitor heavy-chain H3 (ITIH3), and insulin-like-growth-factor-binding protein 1 (IGFBP1)–as direct targets of REV-ERB","PeriodicalId":74135,"journal":{"name":"MedComm - Future medicine","volume":"4 4","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-11-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mef2.70038","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145469712","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
<p>In a recent publication in Nature, Kelepouras et al. [<span>1</span>] reveal a new function of STING in regulating programmed cell death. The study identifies that the absence of <i>Caspase-8</i> leads to abnormal activation of the cGAS–STING signaling pathway, which induces the upregulation of ZBP1 and MLKL and licenses necroptosis independently of TNFR1 and FADD. Crucially, the authors also demonstrate that aberrant activation of STING, whether caused by Caspase-8 loss or by gain-of-function mutation in <i>Sting1</i>, invariably drives inflammatory necroptosis [<span>1</span>].</p><p>Necroptosis is a programmed cell death pathway executed by RIPK3 and mixed lineage kinase domain-like (MLKL), that has been shown to be involved in various inflammatory diseases and tissue injuries [<span>2</span>]. Caspase-8 is a key negative regulator of necroptosis Its deficiency permits aberrant activation of the pathway and causes lethal inflammation in development or in a tissue-selective manner. Deletion of TNFR1 can modestly delay dermatitis by inhibiting canonical necrosome assembly. These findings indicate that necroptosis can be activated independently of TNF-induced, FADD-mediated recruitment of RIPK1 and RIPK3 [<span>3</span>]. However, the upstream signal source and the main regulatory factors remain unclear at this point. STING as a core factor in cellular DNA recognition and the interferon pathway, has traditionally been considered to mainly participate in antiviral and innate immunity, but whether it can be associated with necroptosis has no direct evidence in the past. Deletion of Sting ameliorated the lethal dermatitis caused by loss of Caspase-8 in keratinocytes, providing direct genetic evidence for STING involvement in the observed skin pathology.</p><p>Genetic and biochemical evidence indicates that STING agonists enhance TNF-induced cell death in a RIPK3- and MLKL-dependent manner. ZBP1 is essential for the sensitization mediated by STING, and its deletion can completely block STING-induced necroptosis. Genetic and biochemical analysis further shows that STING upregulates ZBP1 and promotes formation of a FADD- and TNFR1-independent ZBP1–RIPK1–RIPK3 complex, thereby triggering necroptosis. Mechanistically, STING upregulates Zbp1 expression via downstream interferon-stimulated gene (ISG) signaling and promotes cytosolic Z-DNA accumulation, which binds to the Zα domains of ZBP1 to initiate activation. ZBP1 then interacts with RIPK1 and RIPK3 through its RHIM domains, forming a FADD- and TNFR1-independent complex that drives necroptosis.</p><p>In vivo experiments showed that <i>Casp8</i><sup><i>E-KO</i></sup> mice exhibited severe dermatitis and early death, while simultaneous knockout of STING or TNFR1 could notably delay the progression of inflammation and improve survival, suggesting that STING plays a key role in the pathogenesis of skin inflammation. Histological and molecular analysis further confirmed that STING deficiency specifically
{"title":"The Sentinel's Betrayal: A STING-Driven Necroptotic Axis in Caspase-8-Deficient Pathology and STING-Associated Vasculopathy With Onset in Infancy (SAVI)","authors":"Xin He, Bin Wang, Long Zhang","doi":"10.1002/mef2.70037","DOIUrl":"https://doi.org/10.1002/mef2.70037","url":null,"abstract":"<p>In a recent publication in Nature, Kelepouras et al. [<span>1</span>] reveal a new function of STING in regulating programmed cell death. The study identifies that the absence of <i>Caspase-8</i> leads to abnormal activation of the cGAS–STING signaling pathway, which induces the upregulation of ZBP1 and MLKL and licenses necroptosis independently of TNFR1 and FADD. Crucially, the authors also demonstrate that aberrant activation of STING, whether caused by Caspase-8 loss or by gain-of-function mutation in <i>Sting1</i>, invariably drives inflammatory necroptosis [<span>1</span>].</p><p>Necroptosis is a programmed cell death pathway executed by RIPK3 and mixed lineage kinase domain-like (MLKL), that has been shown to be involved in various inflammatory diseases and tissue injuries [<span>2</span>]. Caspase-8 is a key negative regulator of necroptosis Its deficiency permits aberrant activation of the pathway and causes lethal inflammation in development or in a tissue-selective manner. Deletion of TNFR1 can modestly delay dermatitis by inhibiting canonical necrosome assembly. These findings indicate that necroptosis can be activated independently of TNF-induced, FADD-mediated recruitment of RIPK1 and RIPK3 [<span>3</span>]. However, the upstream signal source and the main regulatory factors remain unclear at this point. STING as a core factor in cellular DNA recognition and the interferon pathway, has traditionally been considered to mainly participate in antiviral and innate immunity, but whether it can be associated with necroptosis has no direct evidence in the past. Deletion of Sting ameliorated the lethal dermatitis caused by loss of Caspase-8 in keratinocytes, providing direct genetic evidence for STING involvement in the observed skin pathology.</p><p>Genetic and biochemical evidence indicates that STING agonists enhance TNF-induced cell death in a RIPK3- and MLKL-dependent manner. ZBP1 is essential for the sensitization mediated by STING, and its deletion can completely block STING-induced necroptosis. Genetic and biochemical analysis further shows that STING upregulates ZBP1 and promotes formation of a FADD- and TNFR1-independent ZBP1–RIPK1–RIPK3 complex, thereby triggering necroptosis. Mechanistically, STING upregulates Zbp1 expression via downstream interferon-stimulated gene (ISG) signaling and promotes cytosolic Z-DNA accumulation, which binds to the Zα domains of ZBP1 to initiate activation. ZBP1 then interacts with RIPK1 and RIPK3 through its RHIM domains, forming a FADD- and TNFR1-independent complex that drives necroptosis.</p><p>In vivo experiments showed that <i>Casp8</i><sup><i>E-KO</i></sup> mice exhibited severe dermatitis and early death, while simultaneous knockout of STING or TNFR1 could notably delay the progression of inflammation and improve survival, suggesting that STING plays a key role in the pathogenesis of skin inflammation. Histological and molecular analysis further confirmed that STING deficiency specifically","PeriodicalId":74135,"journal":{"name":"MedComm - Future medicine","volume":"4 4","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-11-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mef2.70037","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145469711","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Li Qiu, Danqing Huang, Yuening Zhang, Yingying Zhou, Ming Luo, Chengdong Zhang, Ying Huang, Mingyuan Zou, Wenlong Lu, Hui Liu, Shaowei Liu, Haoyang Huang, Kaiwen Ye, Yuan Hui, Cheng Tang, Zilong Yan, Xi Zhong, Zhiguo Luo, Hongxin Huang, Ming Zhou, Guangshuai Jia, Qibin Leng, Jun Liu
Tumor immunogenicity determines their response to immune checkpoint inhibitors (ICIs), but the mechanisms governing pancancer immunogenicity remain incompletely understood. A further critical barrier to developing reliable predictive biomarkers is data set shift, which undermines model generalizability. Here, we address these challenges by developing a novel adversarial validation (AV)-integrated machine learning framework, focusing on immunogenic cell death (ICD)-related gene signatures (ICDRSs). We designed three AV-based strategies to mitigate data set shift and validate the efficacies across multiple machine learning algorithms. Using dual-modal data from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO), four optimal AV-based classifiers (e.g., GradientBoosting, XGBoost, LGBM, and CatBoost) were screened, which effectively reduced inter-cohort shift, enhancing both accuracy and robustness of downstream analysis. We identified novel risk/protective ICDRSs that strongly predicted patient survival and tumor immunogenicity across cancers. High-risk ICDRSs correlated with immune-exclusive microenvironments marked by impaired antigen presentation and aberrant tumor-associated macrophage development, as revealed by single-cell RNA sequencing. Validation across 13 ICI-treated cohorts revealed the capacity of ICDRSs for anti-PD-1 nonresponse. Mechanistically, risk ICDRSs were linked to CD47-SIRPA-mediated immune evasion and proliferative macrophage subsets with terminal dysfunction. This study advances understanding of tumor immunogenicity, provides novel biomarker development tools, and supports personalized cancer immunotherapy decision-making.
{"title":"A Machine Learning-Optimized Immunogenic Cell Death Signature Reveals Tumor Immunogenicity and the Immunotherapy Response of Pancancer","authors":"Li Qiu, Danqing Huang, Yuening Zhang, Yingying Zhou, Ming Luo, Chengdong Zhang, Ying Huang, Mingyuan Zou, Wenlong Lu, Hui Liu, Shaowei Liu, Haoyang Huang, Kaiwen Ye, Yuan Hui, Cheng Tang, Zilong Yan, Xi Zhong, Zhiguo Luo, Hongxin Huang, Ming Zhou, Guangshuai Jia, Qibin Leng, Jun Liu","doi":"10.1002/mef2.70035","DOIUrl":"https://doi.org/10.1002/mef2.70035","url":null,"abstract":"<p>Tumor immunogenicity determines their response to immune checkpoint inhibitors (ICIs), but the mechanisms governing pancancer immunogenicity remain incompletely understood. A further critical barrier to developing reliable predictive biomarkers is data set shift, which undermines model generalizability. Here, we address these challenges by developing a novel adversarial validation (AV)-integrated machine learning framework, focusing on immunogenic cell death (ICD)-related gene signatures (ICDRSs). We designed three AV-based strategies to mitigate data set shift and validate the efficacies across multiple machine learning algorithms. Using dual-modal data from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO), four optimal AV-based classifiers (e.g., GradientBoosting, XGBoost, LGBM, and CatBoost) were screened, which effectively reduced inter-cohort shift, enhancing both accuracy and robustness of downstream analysis. We identified novel risk/protective ICDRSs that strongly predicted patient survival and tumor immunogenicity across cancers. High-risk ICDRSs correlated with immune-exclusive microenvironments marked by impaired antigen presentation and aberrant tumor-associated macrophage development, as revealed by single-cell RNA sequencing. Validation across 13 ICI-treated cohorts revealed the capacity of ICDRSs for anti-PD-1 nonresponse. Mechanistically, risk ICDRSs were linked to CD47-SIRPA-mediated immune evasion and proliferative macrophage subsets with terminal dysfunction. This study advances understanding of tumor immunogenicity, provides novel biomarker development tools, and supports personalized cancer immunotherapy decision-making.</p>","PeriodicalId":74135,"journal":{"name":"MedComm - Future medicine","volume":"4 4","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-10-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mef2.70035","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145296960","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}