Cancer lettersPub Date : 2025-05-17DOI: 10.1016/j.canlet.2025.217794
Sugantha Priya Elayapillai , Samrita Dogra , Cole Hladik , James Lausen , Matthew Bruns , Amy Gin Gossett , Fariba Behbod , Chao Xu , Roy Zhang , Wei-Qun Ding , Bethany N. Hannafon
{"title":"Preferential release of microRNAs via extracellular vesicles is associated with ductal carcinoma in situ to invasive breast cancer progression","authors":"Sugantha Priya Elayapillai , Samrita Dogra , Cole Hladik , James Lausen , Matthew Bruns , Amy Gin Gossett , Fariba Behbod , Chao Xu , Roy Zhang , Wei-Qun Ding , Bethany N. Hannafon","doi":"10.1016/j.canlet.2025.217794","DOIUrl":"10.1016/j.canlet.2025.217794","url":null,"abstract":"<div><div>Ductal carcinoma in situ (DCIS) is a precancerous condition that increases the risk of invasive breast cancer (IBC), but not all DCIS cases progress to IBC. The molecular factors driving this transition remain unclear. Small extracellular vesicles (sEVs), or exosomes, play a role in advanced cancer progression, though their function in DCIS is poorly understood. This study explores the role of sEVs and their RNA content in DCIS progression. We found that Rab27A, a key regulator of exosome release, is upregulated in DCIS and IBC tissues compared to normal breast tissue. Inhibiting sEV release by knocking down Rab27A disrupted pro-invasive signaling and reduced invasion in a DCIS mouse model. Using the MCF10 breast cancer progression series, we observed increased microRNA (miRNA) content in sEVs as cells transitioned from normal to malignant, with the most significant differential miRNA expression seen in IBC-derived sEVs. In vivo, DCIS progression raised circulating sEV miRNA levels, which were reduced by Rab27A knockdown. Reintroducing miR-205, enriched in IBC-derived sEVs, suppressed DCIS cell proliferation, invasion, and epithelial-mesenchymal transition (EMT) markers. Co-expression of miR-205 with Rab27A knockdown also suppressed TGF-β signaling, activated MAPK p38, and induced cell cycle arrest and apoptosis. These findings show that the RNA cargo of sEVs changes during malignancy, with specific miRNAs driving DCIS progression. Re-expression of miR-205 offers a promising therapeutic approach to prevent DCIS from becoming invasive.</div></div>","PeriodicalId":9506,"journal":{"name":"Cancer letters","volume":"625 ","pages":"Article 217794"},"PeriodicalIF":9.1,"publicationDate":"2025-05-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144101418","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Cancer lettersPub Date : 2025-05-17DOI: 10.1016/j.canlet.2025.217787
Sai Liu , Dongjuan Liu , Xin Zeng , Jiongke Wang , Jiajia Liu , Junxin Cheng , Kexin Lei , Hetian Bai , Ning Ji , Min Zhou , Lu Jiang , Hongxia Dan , Jing Li , Qianming Chen
{"title":"Corrigendum to “PA28γ acts as a dual regulator of IL-6 and CCL2 and contributes to tumor angiogenesis in oral squamous cell carcinoma” [Cancer Lett. 428 (2018) 192–200]","authors":"Sai Liu , Dongjuan Liu , Xin Zeng , Jiongke Wang , Jiajia Liu , Junxin Cheng , Kexin Lei , Hetian Bai , Ning Ji , Min Zhou , Lu Jiang , Hongxia Dan , Jing Li , Qianming Chen","doi":"10.1016/j.canlet.2025.217787","DOIUrl":"10.1016/j.canlet.2025.217787","url":null,"abstract":"","PeriodicalId":9506,"journal":{"name":"Cancer letters","volume":"626 ","pages":"Article 217787"},"PeriodicalIF":9.1,"publicationDate":"2025-05-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144070606","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Cancer lettersPub Date : 2025-05-16DOI: 10.1016/j.canlet.2025.217807
Dandan Hu , Jialin Zhao , Miaoqing Wu , Ying Zhou , Beile Lyu , Chaoqun Xu , Chao Huang , Zixuan Su , Hui Zhang , Jintao Guo , Weiwei Tang , Gong Chen , Qiyuan Li
{"title":"Microbial interactions induce the mutational signature of mismatch repair deficiency in colorectal cancer and associated with EPPK1 mutations","authors":"Dandan Hu , Jialin Zhao , Miaoqing Wu , Ying Zhou , Beile Lyu , Chaoqun Xu , Chao Huang , Zixuan Su , Hui Zhang , Jintao Guo , Weiwei Tang , Gong Chen , Qiyuan Li","doi":"10.1016/j.canlet.2025.217807","DOIUrl":"10.1016/j.canlet.2025.217807","url":null,"abstract":"<div><div>To better understand the impact of microbial interactions on the clonal evolution of colorectal cancer (CRC), we conducted high-resolution profiling of the gut microbiome of 101 treatment-naïve primary CRC patients using nanopore sequencing. We performed an integrated analysis of microbiome and tumor exome data to identify symbiotic microbes that interactively influence the mutational processes and the subsequent clonality of CRC. Our results suggested that <em>Dialister pneumosintes</em> and <em>Fusobacterium animalis</em> were both associated with somatic <em>EPPK1</em> mutations and promote <strong>SBS6</strong> (mismatch repair deficiency, dMMR) activity. Notably, we showed that the symbiotic architecture of <em>Dialister pneumosintes</em> and <em>Fusobacterium animalis</em> undergoes significant changes with the mutational status of <em>EPPK1</em>. In addition, we identified specific metabolic pathways involving key metabolites that potentially mediate microbial interactions in CRC. These findings provide new insights into the interplay between the gut microbiome and the mutation landscape of colorectal cancer, thereby informing the clonal evolution of CRC and new strategies for precision medicine.</div></div>","PeriodicalId":9506,"journal":{"name":"Cancer letters","volume":"625 ","pages":"Article 217807"},"PeriodicalIF":9.1,"publicationDate":"2025-05-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144092863","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Cancer lettersPub Date : 2025-05-16DOI: 10.1016/j.canlet.2025.217804
Ludi Yang , Sipeng Zuo , Ruobing Jia , Xiang Gu , Qili Liao , Yu Hua , Shengfang Ge , Mengjia He , Jiayan Fan , Xuemei Tong , Tifei Yuan , Renbing Jia , Xianqun Fan , Peiwei Chai , Xuyang Wen
{"title":"Lactylation-boosted polycomb repression of KLF4 elicits glycolysis in retinoblastoma: A positive feedback circuit between histone modifications","authors":"Ludi Yang , Sipeng Zuo , Ruobing Jia , Xiang Gu , Qili Liao , Yu Hua , Shengfang Ge , Mengjia He , Jiayan Fan , Xuemei Tong , Tifei Yuan , Renbing Jia , Xianqun Fan , Peiwei Chai , Xuyang Wen","doi":"10.1016/j.canlet.2025.217804","DOIUrl":"10.1016/j.canlet.2025.217804","url":null,"abstract":"<div><div>The perturbation of histone modification homeostasis is a hallmark of oncogene activation and tumor suppressor gene silencing. Howbeit, the intricate interplay among diverse histone modifications in the context of tumorigenesis is not fully understood. Herein, we unveil a positive feedback mechanism involving lactylation and methylation of histones, which is instrumental in the oncogenic progression of retinoblastoma. First, we pinpointed that the selective upregulation of SUZ12 leads to the upregulation of H3K27me3 modification in retinoblastoma, which is attributed to heightened levels of histone lactylation. Notably, the targeted suppression of <em>SUZ12</em> has demonstrated significant therapeutic benefits in both <em>in vitro</em> and <em>in vivo</em> models of retinoblastoma. Furthermore, multi-omics analysis has identified Krüppel-like factor 4 (<em>KLF4</em>) as a key downstream effector of SUZ12. Mechanistically, SUZ12 is implicated in the enhancement of the H3K27me3 mark on the <em>KLF4</em> promoter, thereby repressing its transcription. Intriguingly, the downregulation of <em>KLF4</em> is associated with an upregulation of glycolysis and a concomitant accumulation of the onco-metabolite lactate, which in turn augments histone lactylation. In conclusion, we provide novel insights into the intricate interplay between lactylation and methylation of histones, shedding light on the epigenetic-metabolic reprogramming that underlies oncogene activation and tumor suppressor gene inactivation in cancer.</div></div>","PeriodicalId":9506,"journal":{"name":"Cancer letters","volume":"625 ","pages":"Article 217804"},"PeriodicalIF":9.1,"publicationDate":"2025-05-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144090303","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Cancer lettersPub Date : 2025-05-15DOI: 10.1016/j.canlet.2025.217788
Jiayi Li , Huaijin Zheng , Xiang Zhang , Boju Pan , Junliang Lu , Liangrui Zhou , Taiping Zhang , Menghua Dai , Junchao Guo , Weibin Wang , Xianlin Han , Qiang Xu , Yuze Hua , Jorg Kleeff , Huanwen Wu , Zhiyong Liang , Qiaofei Liu , Quan Liao
{"title":"Molecular profiling reveals the malignant potential in solid pseudopapillary neoplasms of the pancreas","authors":"Jiayi Li , Huaijin Zheng , Xiang Zhang , Boju Pan , Junliang Lu , Liangrui Zhou , Taiping Zhang , Menghua Dai , Junchao Guo , Weibin Wang , Xianlin Han , Qiang Xu , Yuze Hua , Jorg Kleeff , Huanwen Wu , Zhiyong Liang , Qiaofei Liu , Quan Liao","doi":"10.1016/j.canlet.2025.217788","DOIUrl":"10.1016/j.canlet.2025.217788","url":null,"abstract":"<div><div>Solid pseudopapillary neoplasm of the pancreas (SPN) is a rare tumor primarily affecting middle-aged women, typically characterized by indolent behavior but occasionally demonstrating malignant potential through invasive growth and metastasis. To elucidate the molecular mechanisms driving this heterogeneity, a multi-omics approach was applied to analyze paired metastatic lesions, primary tumors, and normal pancreatic tissues. Methylation profiling via the Illumina Infinium Methylation EPIC BeadChip identified 2425 differentially methylated positions (DMPs) in metastatic versus primary lesions, with 798 DMPs conserved across both lesion types. Tyrosine kinases and cGMP-PKG signaling pathway were the most significantly enriched KEGG pathways involved in the DMPs. Transcriptomic analysis of invasive and non-invasive SPNs using NanoString revealed 99 differentially expressed genes (DEGs). Immunohistochemical validation confirmed elevated protein expression of LY96, IFI16, and GLUD1 in invasive cases. Circulating free DNA (cfDNA) sequencing did not detect genetic mutation in non-metastatic SPN, in contrast, 42.9 % positivity of genetic mutations were detected in metastatic SPNs. Tumor microenvironment analysis by using the GEO database, 850 K methylation sequencing, NanoString transcriptome, highlighted enriched immune-suppressive stromal components in aggressive tumors. These findings establish a molecular signature linking methylation dysregulation, transcriptomic alterations, liquid biopsy, and immune evasion to SPN progression, offering potential biomarkers for risk stratification and therapeutic targeting.</div></div>","PeriodicalId":9506,"journal":{"name":"Cancer letters","volume":"625 ","pages":"Article 217788"},"PeriodicalIF":9.1,"publicationDate":"2025-05-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144072669","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Cancer lettersPub Date : 2025-05-15DOI: 10.1016/j.canlet.2025.217803
Zhe Sun , Rui Wu , Xiaohui Liang , Tiezhu Shi , Yuan Zhang , Zelin Pan , Weidong Zhang , Xin Luan
{"title":"MLCK inhibition induces synthetic lethality in MYC-driven cancer","authors":"Zhe Sun , Rui Wu , Xiaohui Liang , Tiezhu Shi , Yuan Zhang , Zelin Pan , Weidong Zhang , Xin Luan","doi":"10.1016/j.canlet.2025.217803","DOIUrl":"10.1016/j.canlet.2025.217803","url":null,"abstract":"<div><div>The dysregulation of MYC is widely implicated in human cancers, yet MYC remains an ‘undruggable’ target. Here, we performed a CRISPR-based loss-of-function screen focusing on kinases, most of which are ‘druggable,’ to identify genes essential for MYC<sup>high</sup> but not MYC<sup>low</sup> cells. Using an isogenic pair of nonmalignant cells with and without ectopic MYC expression, we uncovered novel MYC synthetic lethal (MYC-SL) interactions, including Myosin Light-Chain Kinase (MLCK) as the most potent MYC-SL target. Inhibition of MLCK induced MYC-dependent cell death, significantly suppressing tumor growth in MYC-driven xenografts, the Apc<sup>Min/+</sup> mouse model of colon cancer, and the MYC-transgenic hepatocellular carcinoma (HCC) model, without apparent toxicity. This cell death is attributed to selective DNA damage and p53-mediated apoptosis. Mechanistically, MYC activation promotes nuclear accumulation of myosin II at stalled replication forks, where it resolves replication stress and supports survival. MLCK inhibition disrupts myosin II activity, leading to unresolved replication stress, DNA damage, and activation of the p53-mediated apoptosis pathway. Our findings suggest that targeting MLCK offers a promising therapeutic strategy for MYC-driven cancers.</div></div>","PeriodicalId":9506,"journal":{"name":"Cancer letters","volume":"625 ","pages":"Article 217803"},"PeriodicalIF":9.1,"publicationDate":"2025-05-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144090213","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Cancer lettersPub Date : 2025-05-15DOI: 10.1016/j.canlet.2025.217806
Soumya Mukherjee, Emily A. Warden, Jianmin Zhang
{"title":"YAP/TAZ: An epitome of tumorigenesis","authors":"Soumya Mukherjee, Emily A. Warden, Jianmin Zhang","doi":"10.1016/j.canlet.2025.217806","DOIUrl":"10.1016/j.canlet.2025.217806","url":null,"abstract":"<div><div>Mounting evidence has demonstrated that the transcriptional coactivators Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ), are the main effectors of the Hippo signal transduction pathway that is involved in multiple layered events in tumorigenesis. The role of YAP/TAZ in cancer development is critical in a context dependent manner. Overexpression of YAP/TAZ induces cell proliferation and is elevated in various cancers and many other malignancies. On the other hand, studies have shown YAP binds p73 to activate PML transcription in response to DNA damage and generate a DNA-damage-induced feedback loop. Intriguingly, at the genomic level, YAP/TAZ genes are rarely mutated in cancer, except in specific tumors. The central role of YAP/TAZ in driving tumorigenesis is attributed through diverse mechanisms, such as regulatory kinases, cellular mechano-transduction, epigenetic modification/alterations, post-translational modifications, protein -protein interaction and nucleo-cytoplasmic export import. The complex interplay among feedback loops and crosstalk between various signaling pathways portrays the dynamic nature of YAP/TAZ. Thus, a comprehensive understanding of how posttranslational modifications and nucleo-cytoplasmic traffic of YAP/TAZ dynamically regulate and control each other holds great promise for selectively targeting YAP/TAZ import and export for drug therapy.</div></div>","PeriodicalId":9506,"journal":{"name":"Cancer letters","volume":"625 ","pages":"Article 217806"},"PeriodicalIF":9.1,"publicationDate":"2025-05-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144092864","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Cancer lettersPub Date : 2025-05-14DOI: 10.1016/j.canlet.2025.217782
Yasuaki Uemoto , Chang-Ching A Lin , Bingnan Wang , Dan Ye , Yisheng V. Fang , Emmanuel Bikorimana , Fabiana Napolitano , Maria Rosario Chica-Parrado , Cheung Li , Saurabh Mendiratta , Chuo Chen , Ariella B. Hanker , Carlos L. Arteaga
{"title":"Corrigendum to “Selective degradation of FGFR1/2 overcomes antiestrogen resistance in ER+ breast cancer with FGFR1/2 alterations” [619 1 (2025) 217668 1–13]","authors":"Yasuaki Uemoto , Chang-Ching A Lin , Bingnan Wang , Dan Ye , Yisheng V. Fang , Emmanuel Bikorimana , Fabiana Napolitano , Maria Rosario Chica-Parrado , Cheung Li , Saurabh Mendiratta , Chuo Chen , Ariella B. Hanker , Carlos L. Arteaga","doi":"10.1016/j.canlet.2025.217782","DOIUrl":"10.1016/j.canlet.2025.217782","url":null,"abstract":"","PeriodicalId":9506,"journal":{"name":"Cancer letters","volume":"626 ","pages":"Article 217782"},"PeriodicalIF":9.1,"publicationDate":"2025-05-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143943620","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Cancer lettersPub Date : 2025-05-14DOI: 10.1016/j.canlet.2025.217802
Yanhui Liu , Xiao Chen , Katharina S. Fischer , Siqi Fu , Li Yuan , Xing Hu
{"title":"Keloids revisited: Current concepts in treatment and differential diagnosis","authors":"Yanhui Liu , Xiao Chen , Katharina S. Fischer , Siqi Fu , Li Yuan , Xing Hu","doi":"10.1016/j.canlet.2025.217802","DOIUrl":"10.1016/j.canlet.2025.217802","url":null,"abstract":"<div><div>Keloid is a special type of scar considered prototypic of skin fibrosis. Unlike hypertrophic scars, keloids exceed the margins of the original wound, and exist over time without a quiescent or regressive phase. Although keloids do not metastasize, they exhibit tumor-like characteristics, and share many similarities. Large epidemiological study demonstrates that patients with keloids have a 1.49-fold higher risk for cancers. Keloids can lead to severe functional impairments and diminish quality of life which increases hidden costs for patients and medical systems. The main goals of treatments are to improve scar appearance, symptoms and patient's quality of life (QoL). However, the microenvironment, pathogenesis, formation and development of the keloid are complex, the efficacy of multiple treatments were limited. Therefore, this up-to-date review aimed to target the current concepts in keloid treatment and differential diagnosis. The goal is to provide a reference for doctors and researchers to improve the accuracy of diagnosis and facilitate the selection of personalized treatment methods for patients with keloids.</div></div>","PeriodicalId":9506,"journal":{"name":"Cancer letters","volume":"625 ","pages":"Article 217802"},"PeriodicalIF":9.1,"publicationDate":"2025-05-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144076208","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Cancer lettersPub Date : 2025-05-12DOI: 10.1016/j.canlet.2025.217793
Ruici Yang , Shanzhao Wang , Zhichao Li , Cong Yin , Wei Huang , Weiren Huang
{"title":"Patient-derived organoid co-culture systems as next-generation models for bladder cancer stem cell research","authors":"Ruici Yang , Shanzhao Wang , Zhichao Li , Cong Yin , Wei Huang , Weiren Huang","doi":"10.1016/j.canlet.2025.217793","DOIUrl":"10.1016/j.canlet.2025.217793","url":null,"abstract":"<div><div>Three-dimensional patient-derived organoids (PDOs) have emerged as a powerful model for investigating the molecular and cellular mechanisms underlying bladder cancer, particularly in the context of cancer stem cells (CSCs) and drug screening. However, a significant limitation of conventional PDOs is the absence of tumor microenvironment (TME), which includes critical stromal, immune and microbial components that influence tumor behavior and treatment response. In this review, we provide a comprehensive overview of the recent advancements in PDO co-culture systems designed to integrate TME elements. Additionally, we emphasize the role of biomedical engineering technologies, such as 3D bioprinting and organoids-on-a-chip, in enhancing the physiological relevance of these models. Furthermore, we explore how bladder PDO co-culture systems are applied in research on bladder CSC characterization, evolution and treatment responses. Finally, we discuss future directions for improving PDO systems to achieve more accurate preclinical modeling and drug discovery.</div></div>","PeriodicalId":9506,"journal":{"name":"Cancer letters","volume":"625 ","pages":"Article 217793"},"PeriodicalIF":9.1,"publicationDate":"2025-05-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144076210","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}