{"title":"In Silico and In Vitro Verification of the Effects of Chemotherapeutic Doxorubicin and 5-Fluorouracil in Combination With Curcumin and Vitamin C on MCF-7 Cells.","authors":"Aslı Akyüz, Duygu Yaşar Şirin","doi":"10.1002/jcb.30688","DOIUrl":"10.1002/jcb.30688","url":null,"abstract":"<p><p>Breast cancer ranks among the most prevalent cancers. Enhancing the effectiveness of chemotherapy and patient survival is the objective of many studies. In the literature, no study has investigated the combined effect of vitamin c and curcumin with chemotherapy drugs on cell viability in the MCF-7 cell line, nor the mechanism of inflammation induced by cancer drugs, both in vitro and in silico. Thus, the purpose of this study was to assess the synergistic effect of curcumin and vitamin c in combination with the chemotherapy drugs 5-fluorouracil and doxorubicin. The cytokine hub genes of the Toll-like receptor pathway for the administered drugs were identified using the Cytoscape program, and docking studies were conducted via the Cb Dock2 website. In silico analyses indicated that doxorubicin and curcumin displayed comparable characteristics, achieving the highest interaction scores (-10) with marker proteins, whereas 5-fluorouracil and vitamin c showed lower interaction scores. Cell viability was evaluated through MTT analysis and AO/PI staining, while the expression of inflammation-related markers IL-6, IL-10, and TNF-α proteins determined using the ELISA method. After 24 h, the cell viability of the chemotherapeutic drugs administered in combination with curcumin decreased by up to 28%. Subsequently, applications at 48 and 72 h were performed. These results indicate that the effect of curcumin on cell viability is significant when combined with chemotherapy drugs. In the ELISA test, a 52% expression of IL-6 was noted in MCF-7 cells treated with curcumin, whereas the IL-6 level decreased to 15% in the other experimental groups. An increase was observed in the TNF-α expression with 5-fluorouracil and doxorubicin compared to the control, while a notable decrease was recorded in the applications with vitamin c and curcumin (p < 0.05). This study demonstrates that vitamin c and curcumin exhibit a synergistic effect with chemotherapeutic agents in the inflammatory system.</p>","PeriodicalId":15219,"journal":{"name":"Journal of cellular biochemistry","volume":" ","pages":"e30688"},"PeriodicalIF":3.0,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142854239","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"FOXO3 Activates MFN2 Expression to Maintain the Autophagy Response in Cancer Cells Under Amino Acid Deprivation.","authors":"Xu Jiang, Jing Wang, Fang Ma, Yuyun Li","doi":"10.1002/jcb.30641","DOIUrl":"10.1002/jcb.30641","url":null,"abstract":"<p><p>The lack of amino acids triggers the autophagic response. Some studies have shown such starvation conditions also induce mitochondrial fusion, revealing a close correlation between the two processes. Although Mitofusin-2 (MFN2) has been demonstrated to play a role in fusion regulation, its role in the autophagic response and the variables that activate MFN2 under stress remain unknown. In this investigation, we screened and confirmed that forkhead box protein O3 (FOXO3) participates in MFN2's expression during short periods of starvation. Luciferase reporter test proved that FOXO3 facilitates MFN2's transcription by binding to its promoter region, and FOXO3 downregulation directly depresses MFN2's expression. Consequently, inhibiting the FOXO3-MFN2 axis results in the loss of mitochondrial fusion, disrupting the normal morphology of mitochondria, impairing the degradation of substrates, and reducing autophagosome accumulation, ultimately leading to the blockage of the autophagy. In conclusion, our work demonstrates that the FOXO3-MFN2 pathway is essential for adaptive changes in mitochondrial morphology and cellular autophagy response under nutritional constraints.</p>","PeriodicalId":15219,"journal":{"name":"Journal of cellular biochemistry","volume":" ","pages":"e30641"},"PeriodicalIF":3.0,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142035954","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Yilei Guo, Bibimaryam Khan, Juanjuan Shi, Yongzhong Hou
{"title":"PPARδ Antagonist Inhibited CD47 Expression and Phagocytosis.","authors":"Yilei Guo, Bibimaryam Khan, Juanjuan Shi, Yongzhong Hou","doi":"10.1002/jcb.30685","DOIUrl":"10.1002/jcb.30685","url":null,"abstract":"<p><p>Increasing evidence suggests that CD47 is highly expressed in multiple types of cancer, which could bind to SIRPα on macrophage, leading to inhibition of macrophage phagocytosis and promotion of tumor growth. However, the regulatory mechanism of CD47 gene expression is not completely clear. Our results indicated that colon cancer cells treated with GSK0660 drug, which is one of the PPARδ antagonists, significantly reduced CD47 gene and protein expression levels in a time and dose-dependent manner. CD47 reporter plasmid was constructed and dual-luciferase analysis was performed. The results suggest that GSK0660 treatment markedly reduced CD47 gene transcriptional activity. Moreover, co-cultured analysis showed that GSK0660 treatment increased phagocytosis. BALB/C mice implanted with CT-26 colon cancer cells were treated with GSK0660, and the results showed that GSK0660 significantly inhibited tumor growth. Moreover, the combination of CD47 monoclonal antibody with GSK0660 drug significantly inhibited tumor growth compared to GSK0660 or CD47 antibody treatment alone. These findings suggest that GSK0660 synergized with CD47 antibody to enhance antitumor immunotherapy.</p>","PeriodicalId":15219,"journal":{"name":"Journal of cellular biochemistry","volume":" ","pages":"e30685"},"PeriodicalIF":3.0,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142780047","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Interaction of PGC7 and HP1BP3 Maintains Meg3-DMR Methylation by Regulating Chromatin Configuration.","authors":"Yingxiang Liu, Weijie Hao, Chenyang Huang, Peiwen Feng, Hongliang Liu, Zekun Guo","doi":"10.1002/jcb.30667","DOIUrl":"10.1002/jcb.30667","url":null,"abstract":"<p><p>Genomic imprinting is essential for mammalian development. PGC7, an important maternal factor, binds to dimethylated histone H3K9 (H3K9me2), maintaining DNA methylation in zygotes and stem cells. However, the underlying molecular mechanisms of PGC7-maintained genomic imprinting in stem cells are not clear. Our previous study has identified that PGC7 interacts with HP1BP3, a novel member of the histone H1 family. In this study, we found that PGC7 interacts with the central globular domain of HP1BP3 through its C-terminal tail and that HP1BP3 is responsible for the recruitment of PGC7 at the Meg3 differentially methylated region (DMR) in the Dlk1-Dio3 imprinted domain. HP1BP3 or PGC7 depletion decreases enrichment in the Meg3-DMR, leading to DNA hypermethylation in this region. Moreover, the cooperative binding of PGC7 and HP1BP3 can antagonize the enrichment of DNMT3A in the Meg3-DMR, and the depletion of HP1BP3 or PGC7 separately induces chromosome decondensation in this region. In summary, this is the first study demonstrating that PGC7 and HP1BP3 synergistically maintain the methylation status of the Meg3-DMR by enabling a chromatin configuration that interferes with the binding of the de novo DNA methyltransferase DNMT3A.</p>","PeriodicalId":15219,"journal":{"name":"Journal of cellular biochemistry","volume":" ","pages":"e30667"},"PeriodicalIF":3.0,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142466374","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Septins: Structural Insights, Functional Dynamics, and Implications in Health and Disease.","authors":"Aurosikha Das, Ambarish Kunwar","doi":"10.1002/jcb.30660","DOIUrl":"10.1002/jcb.30660","url":null,"abstract":"<p><p>Septins are a class of proteins with diverse and vital roles in cell biology. Structurally, they form hetero-oligomeric complexes and assemble into filaments, contributing to the organization of cells. These filaments act as scaffolds, aiding in processes like membrane remodeling, cytokinesis, and cell motility. Functionally, septins are essential to cell division, playing essential roles in cytokinetic furrow formation and maintaining the structural integrity of the contractile ring. They also regulate membrane trafficking and help organize intracellular organelles. In terms of physiology, septins facilitate cell migration, phagocytosis, and immune responses by maintaining membrane integrity and influencing cytoskeletal dynamics. Septin dysfunction is associated with pathophysiological conditions. Mutations in septin genes have been linked to neurodegenerative diseases, such as hereditary spastic paraplegias, underscoring their significance in neuronal function. Septins also play a role in cancer and infectious diseases, making them potential targets for therapeutic interventions. Septins serve as pivotal components of intracellular signaling networks, engaging with diverse proteins like kinases and phosphatases. By modulating the activity of these molecules, septins regulate vital cellular pathways. This integral role in signaling makes septins central to orchestrating cellular responses to environmental stimuli. This review mainly focuses on the human septins, their structural composition, regulatory functions, and implication in pathophysiological conditions underscores their importance in fundamental cellular biology. Moreover, their potential as therapeutic targets across various diseases further emphasizes their significance.</p>","PeriodicalId":15219,"journal":{"name":"Journal of cellular biochemistry","volume":" ","pages":"e30660"},"PeriodicalIF":3.0,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142347413","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Effective Targeting of Colorectal Cancer Stem Cells by Inducing Differentiation Mediated by Low-Dose Vitamin C via β-Catenin Retention in the Cell Membrane.","authors":"Shanooja Shanavas, Utsav Sen, Rajkumar Banerjee, Sudheer Shenoy P, Bipasha Bose","doi":"10.1002/jcb.30686","DOIUrl":"10.1002/jcb.30686","url":null,"abstract":"<p><p>Cancer stem cells (CSCs) are implicated as the underlying cause of tumor recurrence due to their refractoriness to conventional therapies. Targeting CSCs through novel approaches can hinder their survival and proliferation, potentially reducing the challenges associated with tumor relapse. Our previous study demonstrated that colorectal cancer stem cells (CR-CSCs) showed sensitivity to Vitamin C (Vit C), displaying a dose-responsive effect where low doses (2-10 µM) promoted cell proliferation while high doses induced cell death. In this study, we unraveled the mechanistic effects of low doses that, although induced proliferation, remarkably facilitated stemness reduction in HT-29 cell line-derived CR-CSCs. Our findings revealed that Vit C doses of 2 and 6 µM resulted in a reduction in stemness as evidenced by a reduced CD44<sup>+</sup> cell population, representing CR-CSCs. The key finding was the remarkable increase in the expression of β-catenin protein following low-dose Vit C treatment, despite a reduction in stemness, accompanied by a mesenchymal to epithelial transition (MET). The sequestration of upregulated β-catenin via E-cadherin to the cell membrane was identified as a mechanism for reduced stemness, MET, and differentiation of CR-CSCs. Importantly, the epithelial phenotype induced by low-dose Vit C rendered CR-CSCs sensitive to conventional treatments, enhancing chemosensitivity to Cisplatin, Paclitaxel, and 5-Fluorouracil by 60%-90%. These findings suggest that low dose Vit C could serve as an adjuvant to conventional therapeutic strategies for targeting advanced colorectal cancer by sensitizing CR-CSCs to chemotherapy and potentially reducing tumor recurrence.</p>","PeriodicalId":15219,"journal":{"name":"Journal of cellular biochemistry","volume":" ","pages":"e30686"},"PeriodicalIF":3.0,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142806781","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Gengnianchun Against H<sub>2</sub>O<sub>2</sub>-Induced Oxidative Damage in KGN Cells via miR-548m/FOXO3 Signaling.","authors":"Jun Li, Yanqiu Rao, Tao Pu, Ting Xu, Wenjun Wang","doi":"10.1002/jcb.30701","DOIUrl":"https://doi.org/10.1002/jcb.30701","url":null,"abstract":"<p><p>Gengnianchun (GNC) is a traditional remedy used for diminished ovarian reserve, but its underlying mechanisms remain unclear. This study aimed to explore these mechanisms in human granulosa-like cancer (KGN) cells pretreated with medicated rat serum (MRS) before H<sub>2</sub>O<sub>2</sub> exposure. MRS pretreatment significantly alleviated H<sub>2</sub>O<sub>2</sub>-induced cell damage, including improvements in cell viability, superoxide dismutase and GSH-Px activities, and Bcl-2 expression. Conversely, H<sub>2</sub>O<sub>2</sub> treatment increased apoptosis, autophagosomes, IL-1β, TNF-α, reactive oxygen species, malondialdehyde levels, and the expression of LC-II/LC3-I, Bax, and Beclin-1. GEO database analysis revealed significant differential expression of several miRNAs, including miR-548m. qPCR confirmed that MRS upregulated miR-548m expression, which was downregulated by H<sub>2</sub>O<sub>2</sub> in a dose-dependent manner. Preincubation with MRS prevented the decline in miR-548m expression and mitigated H<sub>2</sub>O<sub>2</sub>-induced damage, including improvements in cell viability, apoptosis, autophagy, and oxidative stress. miR-548m suppressed FOXO3 3'-UTR luciferase activity, and anti-miR-548m enhanced it. Transfection with miR-548m reduced FOXO3 mRNA and protein levels, while anti-miR-548m increased them. These findings suggest that GNC protects against H<sub>2</sub>O<sub>2</sub>-induced ovarian damage by modulating the miR-548m/FOXO3 axis, triggering autophagy and apoptosis.</p>","PeriodicalId":15219,"journal":{"name":"Journal of cellular biochemistry","volume":"126 1","pages":"e30701"},"PeriodicalIF":3.0,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142949513","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Repurposing Non-Nucleosidic Reverse Transcriptase Inhibitors (NNRTIs) to Overcome EGFR T790M-Mediated Acquired Resistance in Non-Small Cell Lung Cancer.","authors":"Iqrar Ahmad, Harun M Patel","doi":"10.1002/jcb.30653","DOIUrl":"10.1002/jcb.30653","url":null,"abstract":"<p><p>This study investigates the repurposing potential of non-nucleosidic reverse transcriptase inhibitors (NNRTIs), specifically Rilpivirine and Etravirine, as L858R/T790M tyrosine kinase inhibitors for addressing acquired resistance in non-small cell lung cancer (NSCLC). Using in silico molecular docking, Rilpivirine demonstrated a docking score of -7.534 kcal/mol, comparable to established epidermal growth factor receptor tyrosine kinase inhibitors (EGFR TKIs) like Osimertinib and WZ4002. Molecular dynamics (MD) simulations over 200 ns revealed the stability of the Rilpivirine-EGFR complex, with RMSD values ranging from 2.5 to 3.5 Å. The in vitro antiproliferative assays showed that Rilpivirine had an IC<sub>50</sub> value of 2.3 µM against H1975 cells, while WZ4002 had an IC<sub>50</sub> of 0.291 µM, indicating moderate efficacy. Enzymatic assays revealed that Rilpivirine inhibited the double mutant epidermal growth factor receptor tyrosine kinase (EGFR TK) with an IC<sub>50</sub> value of 54.22 nM and spared the wild-type EGFR TK with an IC<sub>50</sub> of 22.52 nM. These findings suggest Rilpivirine's potential as a therapeutic agent for NSCLC with EGFR L858R/T790M mutations.</p>","PeriodicalId":15219,"journal":{"name":"Journal of cellular biochemistry","volume":" ","pages":"e30653"},"PeriodicalIF":3.0,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142288027","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Guoju Hong, Lin Zhou, Wei He, Qiushi Wei, Jiake Xu
{"title":"The Effects and Mechanisms of Chrysosplenetin in Targeting RANKL-Induced NF-κB Signaling and NFATc1 Activation to Protect Bone Density in Osteolytic Diseases.","authors":"Guoju Hong, Lin Zhou, Wei He, Qiushi Wei, Jiake Xu","doi":"10.1002/jcb.30670","DOIUrl":"10.1002/jcb.30670","url":null,"abstract":"<p><p>Chrysosplenetin (CHR), an O-methylated flavonol from Chamomilla recutita and Laggera pterodonta, has previously demonstrated efficacy in enhancing osteoblast differentiation for treating postmenopausal osteoporosis. This study aims to evaluate CHR's potential to inhibit osteoclastogenesis and prevent bone deterioration in both in vitro and in vivo models. Using tartaric acid-resistant acid phosphatase staining and hydroxyapatite resorption assays, we examined the impact of CHR on RANKL-induced osteoclasts derived from mouse bone marrow monocytes. Additionally, Western blot analysis and qRT-PCR were utilized to assess the protein and gene expressions within the MAPK and NF-κB signaling pathways, as well as the NFATc1 pathway. In vivo, CHR's effects were validated using micro-CT and histomorphometry in an ovariectomized mouse model, showing significant reduction in osteoclast activity and bone loss. The study confirms CHR's inhibition of osteoclastogenesis through interference with RANKL-mediated signaling pathways, suggesting its potential as a novel therapeutic agent for osteolytic conditions related to osteoclast-osteoblast dysregulation.</p>","PeriodicalId":15219,"journal":{"name":"Journal of cellular biochemistry","volume":" ","pages":"e30670"},"PeriodicalIF":3.0,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142557922","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Budheswar Dehury, Sarbani Mishra, Sunita Panda, Mahender Kumar Singh, Nischal L Simha, Sanghamitra Pati
{"title":"Structural Dynamics of Neutral Amino Acid Transporter SLC6A19 in Simple and Complex Lipid Bilayers.","authors":"Budheswar Dehury, Sarbani Mishra, Sunita Panda, Mahender Kumar Singh, Nischal L Simha, Sanghamitra Pati","doi":"10.1002/jcb.30693","DOIUrl":"10.1002/jcb.30693","url":null,"abstract":"<p><p>B0AT1 (SLC6A19) is a major sodium-coupled neutral amino acid transporter that relies on angiotensin converting enzyme 2 (ACE2) or collectrin for membrane trafficking. Despite its significant role in disorders associated with amino acid metabolism, there is a deficit of comprehensive structure-function understanding of B0AT1 in lipid environment. Herein, we have employed molecular dynamics (MD) simulations to explore the architectural characteristics of B0AT1 in two distinct environments: a simplified POPC bilayer and a complex lipid system replicating the native membrane composition. Notably, our B0AT1 analysis in terms of structural stability and regions of maximum flexibility shows consistency in both the systems with enhanced structural features in the case of complex lipid system. Our findings suggest that diacylglycerol phospholipids significantly alter the pore radius, hydrophobic index, and surface charge distribution of B0AT1, thereby affecting the flexibility of transmembrane helices TM7, TM12, and loop connecting TM7-TM8, crucial for ACE2-B0AT1 interaction. Pro41, Ser190, Arg214, Arg240, Ser413, Pro414, Cys463, and Val582 are among the most prominent lipid binding residues that might influence B0AT1 functionality. We also perceive notable lipid mediated deviation in the degree of tilt and loss of helicity in TM1 and TM6 which might affect the substrate binding sites S1 and S2 in B0AT1. Considerably, destabilization in the structure of B0AT1 in lipid environment was evident upon mutation in TM domain, associated with Hartnup disorder through various structure-based protein stability tools. Our two-tiered approach allowed us to validate the use of POPC as a baseline for initial analyses of SLC transporters. Altogether, our all-atoms MD study provides a platform for future investigations into the structure-function mechanism of B0AT1 in realistic lipid mimetic bilayers and offers a framework for developing new therapeutic agents targeting this transporter.</p>","PeriodicalId":15219,"journal":{"name":"Journal of cellular biochemistry","volume":"126 1","pages":"e30693"},"PeriodicalIF":3.0,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11696832/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142921830","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}