{"title":"A Neuroendocrine Differentiation-related Molecular Model for Prognosis Prediction in Prostate Cancer Patients.","authors":"Yong Wei, Jiang-Bo Sun, Qian-Ren-Shun Qiu, Yu-Xuan Zhao, Qing-Shui Zheng, Xiong-Lin Sun, Ning Xu, Xue-Yi Xue","doi":"10.2174/0109298673362568250505074520","DOIUrl":"https://doi.org/10.2174/0109298673362568250505074520","url":null,"abstract":"<p><strong>Purpose: </strong>The purpose of this study is to construct and validate a neuroendocrine differentiation-related molecular model for predicting prognosis in patients with prostate cancer (PCa).</p><p><strong>Materials and methods: </strong>Transcriptome data for PCa were collected from the Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA) websites. Differentially expressed neuroendocrine differentiation related genes (NDGs) were identified. By utilizing multivariate Cox analysis, a neuroendocrine differentiation-related molecular model for predicting prognosis was constructed and validated. The study investigated the novel model's association with the tumor immune microenvironment, clinicopathological characteristics, tumor stemness, and anticancer treatment sensitivity. Additionally, preliminary experimental verifications of Diencephalon / Mesencephalon Homeobox 1 (DMBX1) were conducted.</p><p><strong>Results: </strong>Finally, we identified a total of 19 differentially expressed NDGs. A neuroendocrine differentiation-related molecular model was established and successfully validated both internally and externally. The high-risk group exhibited significantly poorer biochemical recurrence-free (BCRF) in the training, testing, and validating cohorts. The areas under the receiver operating characteristic curves for the training, testing, and validating cohorts were 0.825, 0.719, and 0.729, respectively. The tumor immune microenvironment, clinicopathological features, tumor stemness, and anti-cancer drug sensitivity was significantly different between high and low-risk patients. Preliminary experiments revealed that higher expression of DMBX1 significantly enhanced the proliferation, migration, and neuroendocrine differentiation of PCa cells.</p><p><strong>Conclusion: </strong>This research developed a unique neuroendocrine differentiation-related molecular model that is highly suitable for predicting BCR-free survival (BCRFS). High DMBX1 expression may promote the development and neuroendocrine differentiation of prostate cancer.</p>","PeriodicalId":10984,"journal":{"name":"Current medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":3.5,"publicationDate":"2025-05-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144076471","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}
{"title":"Next-generation Approaches in Targeting Polycystic Ovarian Syndrome: Innovative Strategies.","authors":"Pavithra Lakshmi Narayanan, Subalakshmi Sugumar, Rapura Rushendran, Chitra Vellapandian","doi":"10.2174/0109298673368951250404170052","DOIUrl":"https://doi.org/10.2174/0109298673368951250404170052","url":null,"abstract":"<p><p>Polycystic Ovary Syndrome (PCOS) is a complex endocrine disorder that affects millions of women worldwide and is characterized by ovarian dysfunction, hyperandrogenism, and metabolic abnormalities. The traditional diagnostic and therapeutic approaches often fail to address the multifaceted nature of PCOS. Recent advancements in next-generation sequencing (NGS), bioinformatics, and precision medicine have paved the way for innovative research and therapeutic strategies that promise to revolutionize PCOS management. This review focuses on exploring the genetic and molecular mechanisms of PCOS using innovative methodologies, such as genome-wide association studies (GWAS), transcriptomics, and computational approaches. The integration of big data analytics and machine learning algorithms enhances the predictive accuracy of PCOS diagnoses and treatment outcomes. In addition, the emergence of personalized medicine has enabled tailored therapeutic interventions based on individual genetic profiles and phenotypic expression. Furthermore, we explored the development of novel pharmacological agents and combinational therapies to enhance the understanding of PCOS pathophysiology. These approaches also focus on reducing inflammation, improving insulin sensitivity, and optimizing hormonal balance to achieve optimal health outcomes. The potential of digital health tools, including mobile applications and wearable technologies, to support self-monitoring and patient engagement in PCOS management is also highlighted. In conclusion, the integration of next-generation technologies and innovative research is necessary to transform the field of PCOS diagnosis and treatment, offering hope for more effective and individualized care. These underscore the importance of continued investment in advanced research methodologies and adoption of personalized therapeutic strategies to address the complexities of PCOS.</p>","PeriodicalId":10984,"journal":{"name":"Current medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":3.5,"publicationDate":"2025-05-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143978713","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}
{"title":"Prognostic Implications and Therapeutic Potential of MXD Genes in Gastric Cancer.","authors":"Dongbing Li, Guizhen Lyu","doi":"10.2174/0109298673358646250421023339","DOIUrl":"https://doi.org/10.2174/0109298673358646250421023339","url":null,"abstract":"<p><strong>Background: </strong>MAX dimerization (MXD) genes play integral roles in various types of tumors. The expression patterns, prognostic value, potential mechanisms, and roles in immunotherapy of MXD genes in gastric cancer (GC) remain not fully elucidated.</p><p><strong>Objective: </strong>We aimed to explore the role of MXDs in GC.</p><p><strong>Methods: </strong>The Wilcoxon rank sum test and t-test were employed to evaluate the differential expression of the MXD gene family members in GC tissues compared to non-paired normal gastric tissues. cBioPortal was utilized for examining genetic alterations within the MXD gene family. R software, specifically version 3.6.3, was used to scrutinize the expression patterns of MXD genes in GC, their correlation with clinical parameters, and to generate a correlation heat map. The survival package (v3.2-10) and the Cox regression model were implemented to evaluate the prognostic significance of the MXD gene family. The pROC package (v1.17.0.1) was applied to assess the diagnostic potential of the MXD gene family. R software (v3.6.3) was also used to explore potential regulatory networks involving members of the MXD gene family and related genes. The GSVA package (v1.34.0) was leveraged to investigate the link between the expression of the MXD gene family and immune cell infiltration. Visualization was facilitated by the ggplot2 (v3.3.3), survminer (v0.4.9), and clusterProfiler (v3.14.3) packages. Gene Set Cancer Analysis (GSCA) was employed to determine the sensitivity of the MXD gene family's expression to drugs from the GDSC database. The expression levels of MXD genes were validated across various cell lines using quantitative real-time PCR (qRT-PCR).</p><p><strong>Results: </strong>MXD1 was significantly upregulated in GC, while MXD3 and MXD4 were significantly downregulated. Significant correlations were identified between the expression levels of MXD3 and the T stage (p = 0.041) and age (p = 0.001) of GC patients. Additionally, a notable association was observed between MXD4 expression and the histologic grade (p = 0.006) in GC patients. Low MXD3 expression was associated with a poor prognosis in GC. Low MXD3 expression was an independent prognostic factor for poor outcomes in GC patients. MXD3 demonstrated some accuracy in predicting tumor and normal tissue outcomes (AUC = 0.884). MXDs mediate gastric carcinogenesis and progression by regulating immune cells and pathways, including endocytosis, cell cycle, and apoptosis. The expression of the MXD gene family was associated with immune cell infiltration and drug sensitivity. MXD3 and MXD4 expression levels were significantly downregulated in GC cell lines, while MXD1 expression was significantly upregulated.</p><p><strong>Conclusion: </strong>The MXD gene family may serve as novel biomarkers of poor prognosis and as potential immunotherapeutic targets for GC.</p>","PeriodicalId":10984,"journal":{"name":"Current medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":3.5,"publicationDate":"2025-05-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143985728","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}
{"title":"Binding Specificity and Local Frustration in Structure-based Drug Discovery.","authors":"Zhiqiang Yan, Yuqing Li, Ying Cao, Xuetao Tao, Jin Wang, Yongsheng Jiang","doi":"10.2174/0109298673376099250428054846","DOIUrl":"https://doi.org/10.2174/0109298673376099250428054846","url":null,"abstract":"<p><p>Evolution has optimized proteins to balance stability and function by reducing unfavorable energy states, leading to regions of flexibility and frustration on protein surfaces. These locally frustrated regions correspond to functionally important areas, such as active sites and regions for ligand binding and conformational plasticity. Typical strategies of structure-based drug discovery primarily concentrate on enhancing the binding affinity during compound screening and target identification. However, this often overlooks the binding specificity, which is critical for distinguishing specific binding partners from competing ones and avoiding off-target effects. According to the energy landscape theory, optimization of the intrinsic binding specificity involves globally minimizing the frustrations existing in the biomolecular interactions. Recent studies have demonstrated that identifying local frustrations provides a promising approach for screening more specific compounds binding with targets, and quantifying binding specificity complements typical strategies that focus on binding affinity only. This review explores the principles and strategies of computationally quantifying the binding specificity and local frustrations and discusses their applications in structure-based drug discovery. Moreover, given the advancements of artificial intelligence in protein science, this review aims to motivate the integration of AI and available approaches in quantifying the binding specificity and local frustration. We expect that an AI-powered prediction model will accelerate the drug discovery process and improve the success rate of hit compounds.</p>","PeriodicalId":10984,"journal":{"name":"Current medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":3.5,"publicationDate":"2025-05-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143973177","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}
Zhong Zhao, Wenli Chen, Hao Tang, Xiaoyan Wang, Wenjiao Huang, Subash C B Gopinath, Shu Yang
{"title":"Nanomaterial Enhances the Performance of Amyloid-beta Biosensing for Alzheimer's Disease Diagnosis.","authors":"Zhong Zhao, Wenli Chen, Hao Tang, Xiaoyan Wang, Wenjiao Huang, Subash C B Gopinath, Shu Yang","doi":"10.2174/0109298673363138250417001048","DOIUrl":"https://doi.org/10.2174/0109298673363138250417001048","url":null,"abstract":"<p><strong>Background: </strong>Highly sensitive, accurate, and low-cost detection systems are gaining interest for early intervention in the progression of Alzheimer's disease (AD). Amyloid-beta (Aβ), a peptide highly involved in the progression of AD, is found in abundance in patients with severe AD.</p><p><strong>Objective: </strong>This research focused on developing an Aβ oligomer (AβO) biosensor using a single-walled carbon nanotube-modified (SWCN) interdigitated electrode (IDE) sensor.</p><p><strong>Methods: </strong>The SWCN was functionalized onto the sensor surface through an amine linker, followed by the attachment of an aptamer-gold nanoparticle (GNP) complex, which was used to capture the AβO.</p><p><strong>Results: </strong>The GNP-aptamer was saturated at 500 nM on the SWCN surface, and AβO was detected using a sandwich consisting of aptamer-AβO-antibody. The SWCN modification increased the number of aptamer attachment sites on the IDE, while the aptamer and antibody conjugation with GNP enhanced AβO interaction. This sandwich assay detected AβO at concentrations as low as 10 fM, with a linear regression coefficient [y = 2.9189x - 2.076; R2 = 0.9544]. Furthermore, AβO-spiked artificial CSF was detected without interference, as confirmed by the increased current responses. No significant changes were recorded with control proteins, including α-synuclein, IgG antibody, and a complementary aptamer, indicating specific AβO detection.</p><p><strong>Conclusion: </strong>This SWCN modified IDE-based sandwich detects AβO at its lower level and contributes to the early diagnosis of AD.</p>","PeriodicalId":10984,"journal":{"name":"Current medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":3.5,"publicationDate":"2025-05-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143989786","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}
Daniela Teixeira Rodrigues, Eugênia Carla Kuhn, Jullia Dalbianco Godoy de Oliveira, Natália da Silva Silva Jardim, Daniel Balbé Nunes, Rafael Santos da Silva, Oscar Endrigo Dorneles Rodrigues, Custer Deocaris, João Batista Teixeira da Rocha, Daiana Silva Ávila
{"title":"In Vivo Safety Assessment of AZT-derived Organochalcogen Compounds with Promising Antiviral Effects against SARS-Cov-2.","authors":"Daniela Teixeira Rodrigues, Eugênia Carla Kuhn, Jullia Dalbianco Godoy de Oliveira, Natália da Silva Silva Jardim, Daniel Balbé Nunes, Rafael Santos da Silva, Oscar Endrigo Dorneles Rodrigues, Custer Deocaris, João Batista Teixeira da Rocha, Daiana Silva Ávila","doi":"10.2174/0109298673367163250417065816","DOIUrl":"https://doi.org/10.2174/0109298673367163250417065816","url":null,"abstract":"<p><strong>Background: </strong>Developing new COVID-19 antivirals requires understanding viral proteins, oxidative stress, and drug repositioning. Safety assessments of organochalcogen molecules derived from AZT in Caenorhabditis elegans offer promising prospects for new treatments.</p><p><strong>Objective: </strong>In this work, we evaluated the safety and antioxidant effect of eight organochalcogen AZT-derivatives using the free-living nematode C. elegans through chronic exposure [48h]. In addition, we used in silico computational modelling analyses to predict protein targets for these compounds.</p><p><strong>Methods: </strong>This study used survival, litter size, brood size as toxicological and safety parameters, subcellular localization of DAF-16, expression of SOD-3 and GST-4, and ROS levels to evaluate the antioxidant effects and target prediction by similarity set approach [SEA], protein-protein interaction [PPI] network analysis, and comparative phylogenetic analysis to predict protein targets for these compounds.</p><p><strong>Results: </strong>The molecules were safe at concentrations of 1-500 μM. AZT, R3a, and R3f promoted DAF-16 nuclear translocation without affecting SOD-3 levels. R3f reduced GST-4 levels, while R3a increased ROS levels. In silico analyses identified 16 human protein targets of AZT and its derivatives, linked to nucleotide metabolism, DNA replication, and anti-inflammatory pathways, showing high homology to C. elegans.</p><p><strong>Conclusion: </strong>We hypothesize that Se and Te atom insertion may alter pharmacological properties by modulating DAF-16, GST-4, and ROS-related pathways. in silico data suggest these derivatives are promising for antiviral activity, targeting nucleotide metabolism and DNA replication while also potentially modulating the anti-inflammatory response, an appealing feature for COVID-19 treatment.</p>","PeriodicalId":10984,"journal":{"name":"Current medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":3.5,"publicationDate":"2025-05-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143984814","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}
Densy Davis, Daniela Trisciuzzi, Rajalakshmi Sreekumar, Maria Binu Jacob, Krishnadas Madhu, Nicola Gambacorta, Marco Catto, Della Grace Thomas Parambi, Orazio Nicolotti, Bijo Mathew
{"title":"Key Chemotypes for the Rational Design of Dual AChE/BACE-1 Inhibitors.","authors":"Densy Davis, Daniela Trisciuzzi, Rajalakshmi Sreekumar, Maria Binu Jacob, Krishnadas Madhu, Nicola Gambacorta, Marco Catto, Della Grace Thomas Parambi, Orazio Nicolotti, Bijo Mathew","doi":"10.2174/0109298673350086250310080327","DOIUrl":"https://doi.org/10.2174/0109298673350086250310080327","url":null,"abstract":"<p><p>A classical one-drug-one-target approach is ineffective against diseases with a multi-factorial pathogenesis, such as Alzheimer's [AD]. On the other hand, multi-target approaches can provide a higher level of pharmacological interference which can better affect the disease network. Acetylcholinesterase [AChE], beta-site amyloid precursor protein cleaving enzyme 1 [β-secretase, BACE-1], glycogen synthase kinase 3 beta [GSK- 3β], monoamine oxidases [MAOs], metal ions in the brain, N-methyl-D-aspartate [NMDA] receptor, 5-hydroxytryptamine [5-HT] receptors, the third subtype of histamine receptor [H3 receptor], and phosphodiesterases [PDEs] are the main major targets of this network whose connection are still far from being fully understood. Aware of this limitation, we herein focus on the main chemotypes employed for AChE/BACE-1 targeting. These include mostly bioactive compounds based on chalcones, triazines, triazoles, piperidines, and flavonoids.</p>","PeriodicalId":10984,"journal":{"name":"Current medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":3.5,"publicationDate":"2025-05-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143987602","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}
{"title":"Milk Thistle (Silybum marianum): Potential Role in Cancer Prevention.","authors":"Geir Bjørklund, Olha Storchylo, Monica Butnariu, Maryam Dadar, Salvatore Chirumbolo","doi":"10.2174/0109298673371391250415105506","DOIUrl":"https://doi.org/10.2174/0109298673371391250415105506","url":null,"abstract":"<p><p>Milk thistle compounds have recently gained attention for their potential role in cancer prevention and treatment. Despite most evidence reporting this property refers to in vitro and animal studies, Milk thistle flavonoids may provide insightful suggestions about novel chemopreventive agents. This narrative review provides an overview of the current understanding of milk thistle's effects on cancer cells and highlights possible mechanisms of action. The active compounds in milk thistle mainly exhibit antioxidant and anti-inflammatory effects, which protect cells and enhance their survival responses, even inhibiting cancer development. In addition, the compounds possess immunomodulatory properties crucial in preventing cancer progression. Another important mechanism is the induction of apoptosis, promoting cancer cell death and inhibiting tumour growth. These compounds inhibit angiogenesis, preventing tumour growth and spread. Due to their potential to inhibit cancer progression, they modulate cell signalling pathways, including the MAPK and PI3K/Akt pathways, which are involved in cell growth and survival. Although current research is promising, it is crucial to address the current gaps in knowledge about milk thistle compounds in cancer prevention and treatment. Future studies should focus on rigorous clinical trials, dose optimization, mechanistic investigations, combination therapy approaches, and personalized medicine to maximize their potential. Basic experimental evidence can provide new clues to establish clinical trials to improve cancer care and reassure patients and healthcare professionals.</p>","PeriodicalId":10984,"journal":{"name":"Current medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":3.5,"publicationDate":"2025-05-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143957325","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}
Keping Feng, Qiaoman Fei, Na Huang, Ke Du, Chengbo Zhang, Yudan Fan, Ying Zhou, Yaping Zhao, Pengfei Liu, Zongfang Li
{"title":"Icaritin Attenuates HSC Activation by Down-regulating the HIF-1α and TGF-β/Smad Signaling Pathways to Ameliorate Liver Fibrosis.","authors":"Keping Feng, Qiaoman Fei, Na Huang, Ke Du, Chengbo Zhang, Yudan Fan, Ying Zhou, Yaping Zhao, Pengfei Liu, Zongfang Li","doi":"10.2174/0109298673362768250417052953","DOIUrl":"https://doi.org/10.2174/0109298673362768250417052953","url":null,"abstract":"<p><strong>Introduction: </strong>Icaritin is a bioactive flavonol isolated from the Chinese medicinal herb Epimedium. The comprehensive understanding of antifibrotic effects and associated molecular mechanisms of icaritin remains incomplete. This study aims to explore the protective effects of icaritin against liver fibrosis and to further elucidate the mechanisms involved.</p><p><strong>Methods: </strong>Human hepatic stellate LX-2 cells stimulated with TGF-β1 and a carbon tetrachloride (CCl4)-induced liver fibrosis mouse model were employed. in vitro assays were carried out to evaluate collagen type I (COl I) and α-smooth muscle actin (α-SMA) expression, while in vivo studies assessed fibrosis alleviation. Molecular mechanisms were explored via analysis of TGF-β1, phosphorylated Smad2/3, and HIF-1α protein levels using Western blotting.</p><p><strong>Results: </strong>Icaritin suppressed TGF-β1-induced COl I and α-SMA expression in LX-2 cells and ameliorated liver fibrosis in CCl4-treated mice. Mechanistically, it significantly reduced TGF-β1 levels, inhibited Smad2/3 phosphorylation, and downregulated HIF-1α protein expression in LX-2 cells.</p><p><strong>Conclusion: </strong>Icaritin attenuated experimental liver fibrosis through the inhibition of the TGF-β/Smad and HIF-1α signaling pathways, highlighting its therapeutic potential for fibrotic liver diseases.</p>","PeriodicalId":10984,"journal":{"name":"Current medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":3.5,"publicationDate":"2025-05-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143988055","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}