首页 > 最新文献

Cancer Cell International最新文献

英文 中文
Single-cell transcriptome sequencing reveals the mechanism of Realgar improvement on erythropoiesis in mice with myelodysplastic syndrome.
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2025-04-08 DOI: 10.1186/s12935-025-03768-0
Hao Xu, Kexin Hu, Yanlu Wang, Shuyang Cai, Fan Wu, Jizhang Bao, Qi Hu, Yu Guan, Yuchen Tao, Jiahui Lu

Myelodysplastic syndrome (MDS) is a malignant hematologic disorder with limited curative options, primarily reliant on hematopoietic stem cell transplantation. Anemia, a prevalent symptom of MDS, has few effective treatment strategies. Realgar, though known for its therapeutic effects on MDS, remains poorly understood in terms of its mechanism of action. In this study, both in vivo and in vitro experiments were conducted using Realgar and its primary active component, As2S2, to examine their impact on mouse erythroblasts at the single-cell level. Realgar treatment significantly altered the transcriptional profiles and cellular composition of bone marrow in mice, both in vivo and in vitro. Differentially expressed genes in erythroblasts regulated by Realgar were identified, unveiling potential regulatory functions and signaling pathways, such as heme biosynthesis, hemoglobin production, oxygen binding, IL-17 signaling, and MAPK pathways. These findings suggest that Realgar enhances the differentiation of erythroblasts in mouse bone marrow and improves overall blood cell counts. This work offers preliminary insights into Realgar's mechanisms, expands the understanding of this mineral medicine, and may inform strategies to optimize its therapeutic potential in hematologic diseases.

{"title":"Single-cell transcriptome sequencing reveals the mechanism of Realgar improvement on erythropoiesis in mice with myelodysplastic syndrome.","authors":"Hao Xu, Kexin Hu, Yanlu Wang, Shuyang Cai, Fan Wu, Jizhang Bao, Qi Hu, Yu Guan, Yuchen Tao, Jiahui Lu","doi":"10.1186/s12935-025-03768-0","DOIUrl":"https://doi.org/10.1186/s12935-025-03768-0","url":null,"abstract":"<p><p>Myelodysplastic syndrome (MDS) is a malignant hematologic disorder with limited curative options, primarily reliant on hematopoietic stem cell transplantation. Anemia, a prevalent symptom of MDS, has few effective treatment strategies. Realgar, though known for its therapeutic effects on MDS, remains poorly understood in terms of its mechanism of action. In this study, both in vivo and in vitro experiments were conducted using Realgar and its primary active component, As<sub>2</sub>S<sub>2</sub>, to examine their impact on mouse erythroblasts at the single-cell level. Realgar treatment significantly altered the transcriptional profiles and cellular composition of bone marrow in mice, both in vivo and in vitro. Differentially expressed genes in erythroblasts regulated by Realgar were identified, unveiling potential regulatory functions and signaling pathways, such as heme biosynthesis, hemoglobin production, oxygen binding, IL-17 signaling, and MAPK pathways. These findings suggest that Realgar enhances the differentiation of erythroblasts in mouse bone marrow and improves overall blood cell counts. This work offers preliminary insights into Realgar's mechanisms, expands the understanding of this mineral medicine, and may inform strategies to optimize its therapeutic potential in hematologic diseases.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":"25 1","pages":"135"},"PeriodicalIF":5.3,"publicationDate":"2025-04-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143810517","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
CD44 on cancer stem cell is a potential immunological and prognostic pan-cancer biomarker.
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2025-04-08 DOI: 10.1186/s12935-025-03748-4
Ya-Juan Zhu, Si-Ying Li, Shan-Shan Yang, Yang Du, Zhuo-Yuan Zhang, Ji-Yan Liu

Background: CD44, a widely recognized cancer stem cell marker, displayed a vital participation in the cancer immune invasion and may related with the response to the immunotherapy. However, the role of CD44 in cancer immunology is not well defined. Therefore, we intended to explore its prognostic value and potential immunological functions across 33 human cancer types.

Methods: Based on the data of patients from The Cancer Genome Atlas (TCGA), Sangerbox was used to analyze the correlations between CD44 expression and tumor-infiltrated immune cells, immune checkpoints, neoantigens, microsatellite instability (MSI), and tumor mutational burden (TMB) in human cancers. A mouse model xenografted with shRNA-CD44 MC38 was established.

Results: The elevated CD44 was associated with tumor stage and prognosis in several different cancers. GSEA results showed that upregulated CD44 involved in cancer stem cell associated process, antigen processing and presentation, and immune cells proliferation and activation. CD44 plays an essential role in the tumor immune regulation and immune checkpoints inhibitor response. The correlation of CD44 gene expression and infiltration levels of immune cells varied across different cancer types. Notably, the upregulation of CD44 expression is positively correlated with regulatory CD4 T cells, macrophages M1 and M2 in several analyzed cancers. Furthermore, we verified the effect of CD44 on tumor growth and immune microenvironment in mouse xenografted with shRNA-CD44 MC38. Moreover, DNA methylation existed in CD44 expression and associated with dysfunctional T-cell phenotypes via different mechanisms, thus resulting in tissue-dependent prognoses.

Conclusion: CD44 is both a cancer stem cell marker and a potential prognostic and immunological biomarker in various malignant tumors. Moreover, CD44 could be a novel target for immune-based therapy.

{"title":"CD44 on cancer stem cell is a potential immunological and prognostic pan-cancer biomarker.","authors":"Ya-Juan Zhu, Si-Ying Li, Shan-Shan Yang, Yang Du, Zhuo-Yuan Zhang, Ji-Yan Liu","doi":"10.1186/s12935-025-03748-4","DOIUrl":"https://doi.org/10.1186/s12935-025-03748-4","url":null,"abstract":"<p><strong>Background: </strong>CD44, a widely recognized cancer stem cell marker, displayed a vital participation in the cancer immune invasion and may related with the response to the immunotherapy. However, the role of CD44 in cancer immunology is not well defined. Therefore, we intended to explore its prognostic value and potential immunological functions across 33 human cancer types.</p><p><strong>Methods: </strong>Based on the data of patients from The Cancer Genome Atlas (TCGA), Sangerbox was used to analyze the correlations between CD44 expression and tumor-infiltrated immune cells, immune checkpoints, neoantigens, microsatellite instability (MSI), and tumor mutational burden (TMB) in human cancers. A mouse model xenografted with shRNA-CD44 MC38 was established.</p><p><strong>Results: </strong>The elevated CD44 was associated with tumor stage and prognosis in several different cancers. GSEA results showed that upregulated CD44 involved in cancer stem cell associated process, antigen processing and presentation, and immune cells proliferation and activation. CD44 plays an essential role in the tumor immune regulation and immune checkpoints inhibitor response. The correlation of CD44 gene expression and infiltration levels of immune cells varied across different cancer types. Notably, the upregulation of CD44 expression is positively correlated with regulatory CD4 T cells, macrophages M1 and M2 in several analyzed cancers. Furthermore, we verified the effect of CD44 on tumor growth and immune microenvironment in mouse xenografted with shRNA-CD44 MC38. Moreover, DNA methylation existed in CD44 expression and associated with dysfunctional T-cell phenotypes via different mechanisms, thus resulting in tissue-dependent prognoses.</p><p><strong>Conclusion: </strong>CD44 is both a cancer stem cell marker and a potential prognostic and immunological biomarker in various malignant tumors. Moreover, CD44 could be a novel target for immune-based therapy.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":"25 1","pages":"134"},"PeriodicalIF":5.3,"publicationDate":"2025-04-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143810514","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
CXCL2: a key player in the tumor microenvironment and inflammatory diseases.
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2025-04-07 DOI: 10.1186/s12935-025-03765-3
Yuanhao Lv, Caizheng Chen, Miaomiao Han, Chenfei Tian, Fuyang Song, Sijia Feng, Miaoming Xu, Ziyin Zhao, Hongyan Zhou, Wei Su, Jiateng Zhong

CXCL2 (C-X-C Motif Chemokine Ligand 2), a constituent of the C-X-C chemokine subfamily, serves as a powerful chemotactic factor for neutrophils, facilitating leukocyte recruitment and movement while initiating an inflammatory response. Recent investigations have demonstrated the pivotal involvement of CXCL2 in carcinogenesis. Within the tumor microenvironment, CXCL2 modulates cellular activity primarily via its interaction with the CXCR2 receptor. The activation of signaling pathways, including ERK/MAPK, NF-κB/MAPK, PI3K/AKT, and JAK/STAT3, highlights CXCL2's inclination to promote tumorigenesis. Furthermore, the role of CXCL2 encompasses inflammatory conditions like lung inflammation, atherosclerosis, and obesity. This article examines the structural characteristics, biological roles, and molecular foundation of CXCL2 in carcinogenesis and inflammatory disorders.

CXCL2(C-X-C Motif 趋化因子配体 2)是 C-X-C 趋化因子亚家族的一个成员,是中性粒细胞的强大趋化因子,可促进白细胞的募集和移动,同时引发炎症反应。最近的研究表明,CXCL2 在致癌过程中起着关键作用。在肿瘤微环境中,CXCL2 主要通过与 CXCR2 受体的相互作用来调节细胞活性。ERK/MAPK、NF-κB/MAPK、PI3K/AKT 和 JAK/STAT3 等信号通路的激活,凸显了 CXCL2 促进肿瘤发生的倾向。此外,CXCL2 的作用还包括肺部炎症、动脉粥样硬化和肥胖等炎症。本文探讨了 CXCL2 在致癌和炎症性疾病中的结构特征、生物学作用和分子基础。
{"title":"CXCL2: a key player in the tumor microenvironment and inflammatory diseases.","authors":"Yuanhao Lv, Caizheng Chen, Miaomiao Han, Chenfei Tian, Fuyang Song, Sijia Feng, Miaoming Xu, Ziyin Zhao, Hongyan Zhou, Wei Su, Jiateng Zhong","doi":"10.1186/s12935-025-03765-3","DOIUrl":"10.1186/s12935-025-03765-3","url":null,"abstract":"<p><p>CXCL2 (C-X-C Motif Chemokine Ligand 2), a constituent of the C-X-C chemokine subfamily, serves as a powerful chemotactic factor for neutrophils, facilitating leukocyte recruitment and movement while initiating an inflammatory response. Recent investigations have demonstrated the pivotal involvement of CXCL2 in carcinogenesis. Within the tumor microenvironment, CXCL2 modulates cellular activity primarily via its interaction with the CXCR2 receptor. The activation of signaling pathways, including ERK/MAPK, NF-κB/MAPK, PI3K/AKT, and JAK/STAT3, highlights CXCL2's inclination to promote tumorigenesis. Furthermore, the role of CXCL2 encompasses inflammatory conditions like lung inflammation, atherosclerosis, and obesity. This article examines the structural characteristics, biological roles, and molecular foundation of CXCL2 in carcinogenesis and inflammatory disorders.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":"25 1","pages":"133"},"PeriodicalIF":5.3,"publicationDate":"2025-04-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143802547","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Brachyury promotes proliferation and migration of colorectal cancer cells by targeting MMP14.
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2025-04-07 DOI: 10.1186/s12935-025-03726-w
Ming Chen, Huiheng Qu, Xiao Liang, Ying Huang, Zhengjie Yang, Pei Lu, Keqin Shi, Peng Chen, Yanjing Zhang, Hui Zhou, Jiazeng Xia, Jun Shen

Background: The incidence and mortality rates of colorectal cancer (CRC) are rising, and it is the second most common cause of cancer-related deaths worldwide. Although the transcription factor, Brachyury is intricately linked with various clinical malignancies, the mechanisms by which it influences CRC cell proliferation and migration are inadequately understood.

Methods: Tissue microarray was used to evaluate Brachyury expression in CRC and adjacent normal tissues. The effects of Brachyury on HCT116 and SW480 CRC cells were also examined in vitro, including using Cell Counting Kit-8, colony formation, and transwell assays, and in vivo through subcutaneous tumorigenesis assays in a nude mouse xenograft model. Chromatin immunoprecipitation was used to evaluate Brachyury binding to the MMP14 promoter and its impact on MMP14 expression. Rescue experiments were used to elucidate MMP14's role in mediating Brachyury's effect on CRC cell behavior.

Results: Brachyury expression was significantly higher in CRC tissues than in adjacent normal tissues, and it promotes CRC oncogenesis in vitro and in vivo. Rescue experiments established MMP14 as a direct, downstream Brachyury target, affirming that MMP14 enhanced Brachyury-driven CRC cell proliferation.

Conclusion: Our findings highlight targeting the Brachyury-MMP14 axis as a potential novel approach for CRC clinical therapy.

{"title":"Brachyury promotes proliferation and migration of colorectal cancer cells by targeting MMP14.","authors":"Ming Chen, Huiheng Qu, Xiao Liang, Ying Huang, Zhengjie Yang, Pei Lu, Keqin Shi, Peng Chen, Yanjing Zhang, Hui Zhou, Jiazeng Xia, Jun Shen","doi":"10.1186/s12935-025-03726-w","DOIUrl":"10.1186/s12935-025-03726-w","url":null,"abstract":"<p><strong>Background: </strong>The incidence and mortality rates of colorectal cancer (CRC) are rising, and it is the second most common cause of cancer-related deaths worldwide. Although the transcription factor, Brachyury is intricately linked with various clinical malignancies, the mechanisms by which it influences CRC cell proliferation and migration are inadequately understood.</p><p><strong>Methods: </strong>Tissue microarray was used to evaluate Brachyury expression in CRC and adjacent normal tissues. The effects of Brachyury on HCT116 and SW480 CRC cells were also examined in vitro, including using Cell Counting Kit-8, colony formation, and transwell assays, and in vivo through subcutaneous tumorigenesis assays in a nude mouse xenograft model. Chromatin immunoprecipitation was used to evaluate Brachyury binding to the MMP14 promoter and its impact on MMP14 expression. Rescue experiments were used to elucidate MMP14's role in mediating Brachyury's effect on CRC cell behavior.</p><p><strong>Results: </strong>Brachyury expression was significantly higher in CRC tissues than in adjacent normal tissues, and it promotes CRC oncogenesis in vitro and in vivo. Rescue experiments established MMP14 as a direct, downstream Brachyury target, affirming that MMP14 enhanced Brachyury-driven CRC cell proliferation.</p><p><strong>Conclusion: </strong>Our findings highlight targeting the Brachyury-MMP14 axis as a potential novel approach for CRC clinical therapy.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":"25 1","pages":"132"},"PeriodicalIF":5.3,"publicationDate":"2025-04-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143802545","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Long noncoding RNAs in acute myeloid leukemia: biomarkers, prognostic indicators, and treatment potential.
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2025-04-05 DOI: 10.1186/s12935-025-03763-5
Maryam Farajzadeh, Mehrdad Fathi, Pooya Jalali, Armin Mahmoudsalehi Kheshti, Shahla Khodayari, Mohammad Hojjat-Farsangi, Farhad Jadidi

Long noncoding RNAs (lncRNAs) have been recognized as significant modulators of gene expression and are essential for various biological functions, even though they don't appear to have the ability to encode proteins. Originally considered dark matter, lncRNAs have been recognized as being dysregulated and contributing to the onset, progression, and resistance to treatment of acute myeloid leukemia (AML). AML is a prevalent type of leukemia characterized by the disruption of myeloid cell differentiation, leading to an increased number of immature myeloid progenitor cells. Currently, the need for novel biomarkers and treatment targets to enhance therapeutic alternatives has led to a focus on lncRNAs as possible indicators for prognostic, therapeutic, and diagnostic systems in various human cancers, including AML. Recent research has recognized a limited set of lncRNAs as possible prognostic biomarkers or diagnoses in AML. This review evaluates the key research that highlights the significance of lncRNAs in AML and discusses their roles and impacts on the disease. Furthermore, we intend to underscore the importance of lncRNAs as new and trustworthy markers for the diagnosis, prediction, drug resistance, and targets for treatment in AML.

{"title":"Long noncoding RNAs in acute myeloid leukemia: biomarkers, prognostic indicators, and treatment potential.","authors":"Maryam Farajzadeh, Mehrdad Fathi, Pooya Jalali, Armin Mahmoudsalehi Kheshti, Shahla Khodayari, Mohammad Hojjat-Farsangi, Farhad Jadidi","doi":"10.1186/s12935-025-03763-5","DOIUrl":"10.1186/s12935-025-03763-5","url":null,"abstract":"<p><p>Long noncoding RNAs (lncRNAs) have been recognized as significant modulators of gene expression and are essential for various biological functions, even though they don't appear to have the ability to encode proteins. Originally considered dark matter, lncRNAs have been recognized as being dysregulated and contributing to the onset, progression, and resistance to treatment of acute myeloid leukemia (AML). AML is a prevalent type of leukemia characterized by the disruption of myeloid cell differentiation, leading to an increased number of immature myeloid progenitor cells. Currently, the need for novel biomarkers and treatment targets to enhance therapeutic alternatives has led to a focus on lncRNAs as possible indicators for prognostic, therapeutic, and diagnostic systems in various human cancers, including AML. Recent research has recognized a limited set of lncRNAs as possible prognostic biomarkers or diagnoses in AML. This review evaluates the key research that highlights the significance of lncRNAs in AML and discusses their roles and impacts on the disease. Furthermore, we intend to underscore the importance of lncRNAs as new and trustworthy markers for the diagnosis, prediction, drug resistance, and targets for treatment in AML.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":"25 1","pages":"131"},"PeriodicalIF":5.3,"publicationDate":"2025-04-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11972515/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143787966","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Ginger extract inhibits c-MET activation and suppresses osteosarcoma in vitro and in vivo.
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2025-04-04 DOI: 10.1186/s12935-025-03759-1
Ruoping Yanzhang, Mingyang Yan, Zhaojie Yang, Huijun Zhang, Yin Yu, Xiangping Li, Ruifang Shen, Xiao Chu, Siyuan Han, Ziliang Zhang, Junyan Teng, Hao Li, Tao Li, Guoguo Jin, Zhiping Guo

Background: Osteosarcoma (OS) as an invasive and lethal malignancy showing a low 5-year survival rate requires novel therapeutic targets and their suppressors to improve prevention and treatment strategies.

Methods: Our research served to clarify the therapeutic potential of ginger extract and its underlying antineoplastic mechanisms in OS. In vitro studies were used to detect the anti-proliferation ability of ginger extract towards OS cells. Patient-derived xenograft (PDX) was performed to confirm whether ginger extract suppressed tumor growth. Cancer Heat Shock Protein (HSP) database was utilized to identify the potential target of ginger extract, which was subsequently validated through a computational docking model screening method, molecular dynamics simulations and pull-down assay. Analysis of the Gene Expression Omnibus (GEO) database revealed the c-MET expression among OS samples as well as the potential mechanism. Immunohistochemistry (IHC) staining corroborated the c-MET expression level among OS tissues relative to the controls. Functional studies involving c-MET knockdown among OS cell lines were produced to elucidate the functional role of c-MET in OS cellular processes.

Results: In vitro studies demonstrated that ginger extract administration impeded OS cell progress while inducing apoptosis and inhibiting migration. Moreover, in vivo tests unveiled that ginger extract prominently inhibited patient-derived xenograft (PDX) tumor development. Cancer HSP database analysis recognized c-MET as an underlying target of ginger extract, which was subsequently validated through a computational docking model screening, molecular dynamics simulations and pull-down assay. Analysis of the Gene Expression Omnibus (GEO) database combined with immunohistochemistry (IHC) staining corroborated the c-MET overexpression among OS tissues in contrast with the controls. Next, our study confirmed the significant suppression of cell progress and anchorage-independent growth, while concomitantly inducing apoptosis after c-MET knockdown, underscoring its prospect for a therapeutic target.

Conclusion: Collectively, our findings show that c-MET is a prospective therapeutic target for OS. Ginger extract, a natural c-MET inhibitor, exhibits potent antineoplastic effects by suppressing OS growth both in vitro and in vivo, highlighting its prospect for a new therapeutic agent of this aggressive malignancy.

{"title":"Ginger extract inhibits c-MET activation and suppresses osteosarcoma in vitro and in vivo.","authors":"Ruoping Yanzhang, Mingyang Yan, Zhaojie Yang, Huijun Zhang, Yin Yu, Xiangping Li, Ruifang Shen, Xiao Chu, Siyuan Han, Ziliang Zhang, Junyan Teng, Hao Li, Tao Li, Guoguo Jin, Zhiping Guo","doi":"10.1186/s12935-025-03759-1","DOIUrl":"10.1186/s12935-025-03759-1","url":null,"abstract":"<p><strong>Background: </strong>Osteosarcoma (OS) as an invasive and lethal malignancy showing a low 5-year survival rate requires novel therapeutic targets and their suppressors to improve prevention and treatment strategies.</p><p><strong>Methods: </strong>Our research served to clarify the therapeutic potential of ginger extract and its underlying antineoplastic mechanisms in OS. In vitro studies were used to detect the anti-proliferation ability of ginger extract towards OS cells. Patient-derived xenograft (PDX) was performed to confirm whether ginger extract suppressed tumor growth. Cancer Heat Shock Protein (HSP) database was utilized to identify the potential target of ginger extract, which was subsequently validated through a computational docking model screening method, molecular dynamics simulations and pull-down assay. Analysis of the Gene Expression Omnibus (GEO) database revealed the c-MET expression among OS samples as well as the potential mechanism. Immunohistochemistry (IHC) staining corroborated the c-MET expression level among OS tissues relative to the controls. Functional studies involving c-MET knockdown among OS cell lines were produced to elucidate the functional role of c-MET in OS cellular processes.</p><p><strong>Results: </strong>In vitro studies demonstrated that ginger extract administration impeded OS cell progress while inducing apoptosis and inhibiting migration. Moreover, in vivo tests unveiled that ginger extract prominently inhibited patient-derived xenograft (PDX) tumor development. Cancer HSP database analysis recognized c-MET as an underlying target of ginger extract, which was subsequently validated through a computational docking model screening, molecular dynamics simulations and pull-down assay. Analysis of the Gene Expression Omnibus (GEO) database combined with immunohistochemistry (IHC) staining corroborated the c-MET overexpression among OS tissues in contrast with the controls. Next, our study confirmed the significant suppression of cell progress and anchorage-independent growth, while concomitantly inducing apoptosis after c-MET knockdown, underscoring its prospect for a therapeutic target.</p><p><strong>Conclusion: </strong>Collectively, our findings show that c-MET is a prospective therapeutic target for OS. Ginger extract, a natural c-MET inhibitor, exhibits potent antineoplastic effects by suppressing OS growth both in vitro and in vivo, highlighting its prospect for a new therapeutic agent of this aggressive malignancy.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":"25 1","pages":"130"},"PeriodicalIF":5.3,"publicationDate":"2025-04-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11971884/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143787962","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Heme oxygenase 1 confers gilteritinib resistance in FLT3-ITD acute myeloid leukemia in a STAT6-dependent manner.
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2025-04-04 DOI: 10.1186/s12935-025-03757-3
Tianzhuo Zhang, Danna Wei, Yun Zhan, Zhengmei Long, Tingting Lu, Peng Zhao, Rui Gao, Qian Kang, Luxin Zhang, Min Liu, Xueying Yang, Jishi Wang

Background: Acute myeloid leukemia (AML) is the most common type of acute leukemia in adults. We previously discovered that heme oxygenase 1 (HO1) is crucial for chemoresistance in AML, but the detailed molecular mechanism of that remains unclear.

Methods: RNA sequencing was conducted to assess transcriptomic changes in three pairs of AML cells after regulating the expression of HO1. The molecular mechanism by which HO1 induces gilteritinib resistance in FLT3-ITD (FMS-like tyrosine kinase 3 (FLT3) internal tandem duplication (ITD)) AML was evaluated by quantitative real-time PCR (qRT-PCR), CCK-8, flow cytometry, and western blotting. FLT3-ITD AML mouse models were established to investigate the effects of HO1 expression on gilteritinib resistance in vivo.

Results: In these three pairs of AML cells, we discovered that HO1-mediated drug resistance is connected to the interleukin-4-mediated signaling pathway (specifically STAT6) only in MV4-11 cells with the FLT3-ITD mutation. Further findings revealed that HO1 overexpression confers gilteritinib resistance in FLT3-ITD AML cell lines and primary individual specimens. While suppression of HO1 sensitized FLT3-ITD AML cell lines and primary individual specimens to gilteritinib. Mechanistically, western blotting and flow cytometry confirmed that HO1-mediated gilteritinib resistance is related to STAT6 phosphorylation in FLT3-ITD AML cell lines and primary individual specimens. Moreover, tumor-bearing mice were employed to determine that HO1 overexpression conferred gilteritinib resistance in vivo.

Conclusions: Collectively, these studies illustrate that HO1 may act as a successful treatment target for gilteritinib-resistant FLT3-ITD AML patients.

{"title":"Heme oxygenase 1 confers gilteritinib resistance in FLT3-ITD acute myeloid leukemia in a STAT6-dependent manner.","authors":"Tianzhuo Zhang, Danna Wei, Yun Zhan, Zhengmei Long, Tingting Lu, Peng Zhao, Rui Gao, Qian Kang, Luxin Zhang, Min Liu, Xueying Yang, Jishi Wang","doi":"10.1186/s12935-025-03757-3","DOIUrl":"10.1186/s12935-025-03757-3","url":null,"abstract":"<p><strong>Background: </strong>Acute myeloid leukemia (AML) is the most common type of acute leukemia in adults. We previously discovered that heme oxygenase 1 (HO1) is crucial for chemoresistance in AML, but the detailed molecular mechanism of that remains unclear.</p><p><strong>Methods: </strong>RNA sequencing was conducted to assess transcriptomic changes in three pairs of AML cells after regulating the expression of HO1. The molecular mechanism by which HO1 induces gilteritinib resistance in FLT3-ITD (FMS-like tyrosine kinase 3 (FLT3) internal tandem duplication (ITD)) AML was evaluated by quantitative real-time PCR (qRT-PCR), CCK-8, flow cytometry, and western blotting. FLT3-ITD AML mouse models were established to investigate the effects of HO1 expression on gilteritinib resistance in vivo.</p><p><strong>Results: </strong>In these three pairs of AML cells, we discovered that HO1-mediated drug resistance is connected to the interleukin-4-mediated signaling pathway (specifically STAT6) only in MV4-11 cells with the FLT3-ITD mutation. Further findings revealed that HO1 overexpression confers gilteritinib resistance in FLT3-ITD AML cell lines and primary individual specimens. While suppression of HO1 sensitized FLT3-ITD AML cell lines and primary individual specimens to gilteritinib. Mechanistically, western blotting and flow cytometry confirmed that HO1-mediated gilteritinib resistance is related to STAT6 phosphorylation in FLT3-ITD AML cell lines and primary individual specimens. Moreover, tumor-bearing mice were employed to determine that HO1 overexpression conferred gilteritinib resistance in vivo.</p><p><strong>Conclusions: </strong>Collectively, these studies illustrate that HO1 may act as a successful treatment target for gilteritinib-resistant FLT3-ITD AML patients.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":"25 1","pages":"129"},"PeriodicalIF":5.3,"publicationDate":"2025-04-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11969713/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143787964","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Pan-cancer analysis identifies CLEC12A as a potential biomarker and therapeutic target for lung adenocarcinoma.
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2025-04-03 DOI: 10.1186/s12935-025-03755-5
Desheng Zhou, Yachao Cui, Tianxiang Liang, Zhenpeng Wu, Haiping Yan, Yingchang Li, Wenguang Yin, Yunen Lin, Qiang You

C-type lectin domain family 12 member A (CLEC12A) is a type II transmembrane glycoprotein widely expressed in innate immune cells, where it plays a crucial role in immune modulation and has been implicated in cancer progression. However, its precise function in oncogenesis and immune infiltration remains incompletely understood. To investigate this, we utilized multiple databases to assess the mRNA and protein expression levels of CLEC12A across normal tissues and a broad spectrum of cancers. We also evaluated its prognostic and diagnostic significance in pan-cancer contexts. Furthermore, the relationship between CLEC12A expression and immune cell infiltration, immune checkpoints, and immune predictors was explored. In addition, Weighted Gene Co-Expression Network Analysis (WGCNA) and differential expression analysis were performed to examine the biological relevance of CLEC12A in lung adenocarcinoma (LUAD). We also leveraged various databases to predict CLEC12A's response to immunotherapy and drug sensitivity. Finally, in vitro experiments validated the functional role of CLEC12A in LUAD. Our comprehensive pan-cancer analysis revealed that CLEC12A exhibited distinct expression patterns across different cancer types, suggesting its potential as both a diagnostic and prognostic biomarker. Notably, CLEC12A expression was strongly correlated with immune cell infiltration, immune checkpoints, and immune predictors. Functional enrichment analysis highlighted that increased CLEC12A expression in LUAD was associated with a variety of immune-related biological processes and pathways. Moreover, CLEC12A showed significant predictive value for immunotherapy outcomes, and several drugs targeting CLEC12A were identified. In vitro experiments further demonstrated that CLEC12A overexpression inhibited the proliferation, migration, and invasion of LUAD cells. Taken together, our findings position CLEC12A as a promising candidate for cancer detection, prognosis, and as a therapeutic target, particularly in LUAD, where it may serve as a potential target for both immunotherapy and targeted therapy.

{"title":"Pan-cancer analysis identifies CLEC12A as a potential biomarker and therapeutic target for lung adenocarcinoma.","authors":"Desheng Zhou, Yachao Cui, Tianxiang Liang, Zhenpeng Wu, Haiping Yan, Yingchang Li, Wenguang Yin, Yunen Lin, Qiang You","doi":"10.1186/s12935-025-03755-5","DOIUrl":"10.1186/s12935-025-03755-5","url":null,"abstract":"<p><p>C-type lectin domain family 12 member A (CLEC12A) is a type II transmembrane glycoprotein widely expressed in innate immune cells, where it plays a crucial role in immune modulation and has been implicated in cancer progression. However, its precise function in oncogenesis and immune infiltration remains incompletely understood. To investigate this, we utilized multiple databases to assess the mRNA and protein expression levels of CLEC12A across normal tissues and a broad spectrum of cancers. We also evaluated its prognostic and diagnostic significance in pan-cancer contexts. Furthermore, the relationship between CLEC12A expression and immune cell infiltration, immune checkpoints, and immune predictors was explored. In addition, Weighted Gene Co-Expression Network Analysis (WGCNA) and differential expression analysis were performed to examine the biological relevance of CLEC12A in lung adenocarcinoma (LUAD). We also leveraged various databases to predict CLEC12A's response to immunotherapy and drug sensitivity. Finally, in vitro experiments validated the functional role of CLEC12A in LUAD. Our comprehensive pan-cancer analysis revealed that CLEC12A exhibited distinct expression patterns across different cancer types, suggesting its potential as both a diagnostic and prognostic biomarker. Notably, CLEC12A expression was strongly correlated with immune cell infiltration, immune checkpoints, and immune predictors. Functional enrichment analysis highlighted that increased CLEC12A expression in LUAD was associated with a variety of immune-related biological processes and pathways. Moreover, CLEC12A showed significant predictive value for immunotherapy outcomes, and several drugs targeting CLEC12A were identified. In vitro experiments further demonstrated that CLEC12A overexpression inhibited the proliferation, migration, and invasion of LUAD cells. Taken together, our findings position CLEC12A as a promising candidate for cancer detection, prognosis, and as a therapeutic target, particularly in LUAD, where it may serve as a potential target for both immunotherapy and targeted therapy.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":"25 1","pages":"128"},"PeriodicalIF":5.3,"publicationDate":"2025-04-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11967068/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143779219","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
STAT3/TGFBI signaling promotes the temozolomide resistance of glioblastoma through upregulating glycolysis by inducing cellular senescence.
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2025-04-03 DOI: 10.1186/s12935-025-03770-6
Yanbin Zhang, Xiaohua Xiao, Ge Yang, Xiaobing Jiang, Shujie Jiao, Yingli Nie, Tao Zhang

Glioblastoma (GBM) is the most lethal type of brain tumor. Recent studies have indicated that cellular senescence-targeted therapy is a promising approach for cancer treatment. However, the underlying mechanisms remain to be clarified. In this study, 101 unique combinations of 10 machine learning algorithms were used to construct prognostic models based on cellular senescence-related genes (CSRGs). We developed the CSRG signature (CSRGS) using machine learning models that exhibited optimal performance. GBM samples were stratified into high- and low-CSRGS groups based on CSRGS scores. Patients in the high-CSRGS group exhibited a worse prognosis, higher immune infiltration, and increased sensitivity to immune checkpoint blockade therapy. Furthermore, senescence-related pathways were significantly correlated with glycolysis, indicating upregulated glycolytic metabolism in senescent GBM cells. We identified TGFBI as a key regulator that played vital roles in both glycolysis and cellular senescence in GBM. TGFBI was overexpressed in GBM samples compared to normal brain tissues, and its knockdown via shRNA inhibited cellular senescence, glycolysis, and temozolomide resistance. Chromatin immunoprecipitation (ChIP) and luciferase reporter assays confirmed that TGFBI is a direct STAT3 target and is required for the STAT3-induced promotion of cellular senescence, glycolysis, and drug resistance. The STAT3-TGFBI axis could be a potential target for senescence-targeted GBM therapy.

{"title":"STAT3/TGFBI signaling promotes the temozolomide resistance of glioblastoma through upregulating glycolysis by inducing cellular senescence.","authors":"Yanbin Zhang, Xiaohua Xiao, Ge Yang, Xiaobing Jiang, Shujie Jiao, Yingli Nie, Tao Zhang","doi":"10.1186/s12935-025-03770-6","DOIUrl":"10.1186/s12935-025-03770-6","url":null,"abstract":"<p><p>Glioblastoma (GBM) is the most lethal type of brain tumor. Recent studies have indicated that cellular senescence-targeted therapy is a promising approach for cancer treatment. However, the underlying mechanisms remain to be clarified. In this study, 101 unique combinations of 10 machine learning algorithms were used to construct prognostic models based on cellular senescence-related genes (CSRGs). We developed the CSRG signature (CSRGS) using machine learning models that exhibited optimal performance. GBM samples were stratified into high- and low-CSRGS groups based on CSRGS scores. Patients in the high-CSRGS group exhibited a worse prognosis, higher immune infiltration, and increased sensitivity to immune checkpoint blockade therapy. Furthermore, senescence-related pathways were significantly correlated with glycolysis, indicating upregulated glycolytic metabolism in senescent GBM cells. We identified TGFBI as a key regulator that played vital roles in both glycolysis and cellular senescence in GBM. TGFBI was overexpressed in GBM samples compared to normal brain tissues, and its knockdown via shRNA inhibited cellular senescence, glycolysis, and temozolomide resistance. Chromatin immunoprecipitation (ChIP) and luciferase reporter assays confirmed that TGFBI is a direct STAT3 target and is required for the STAT3-induced promotion of cellular senescence, glycolysis, and drug resistance. The STAT3-TGFBI axis could be a potential target for senescence-targeted GBM therapy.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":"25 1","pages":"127"},"PeriodicalIF":5.3,"publicationDate":"2025-04-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11967127/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143779220","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
EIF2S2 as a prognostic marker and therapeutic target in glioblastoma: insights into its role and downstream mechanisms.
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2025-04-02 DOI: 10.1186/s12935-025-03762-6
Bo Fan, Qing Pan, Xiaokai Yuan, Wei Du, Zhongjie Yan

Glioblastoma (GBM) the most common and most aggressive primary brain tumor has a five-year survival rate of less than 5%. The onset of GBM is very complicated and has always been the focus of researchers. This study analyzed data from 637 GBM and 20 normal tissues from The Cancer Genome Atlas (TCGA), and patients were categorized into high and low EIF2S2 expression groups. The Overall survival and disease-free survival of GBM patients in low expression of EIF2S2 group were significantly higher than those in high expression of EIF2S2 group (p < 0.001), and the expression level of EIF2S2 was significantly correlated with tumor grade (p < 0.001) and tumor recurrence (p < 0.001). The study designed three different short hairpin RNA (shRNA) sequence vectors, identifying shEIF2S2-1 as the most effective. This vector significantly reduced EIF2S2 expression, cell proliferation, and migration while increasing apoptosis in SHG-44 and U251 cells (p < 0.01). By detecting SHG-44 cells infected with shEIF2S2 vector and shCtrl with human whole gene expression chip, we identified WNT5A that is a downstream target gene of EIF2S2. Interfering with WNT5A and overexpressing EIF2S2 in SHG-44 and U251 cells revealed that EIF2S2 regulates WNT5A expression. This regulation led to an increased apoptosis rate (p < 0.05) and a significant reduction in cell migration (p < 0.05) in both the EIF2S2 overexpression and shWNT5A interference groups, confirming that WNT5A is a downstream regulatory target of EIF2S2. This study revealed the key role of EIF2S2 in GBM and its potential molecular mechanism.

{"title":"EIF2S2 as a prognostic marker and therapeutic target in glioblastoma: insights into its role and downstream mechanisms.","authors":"Bo Fan, Qing Pan, Xiaokai Yuan, Wei Du, Zhongjie Yan","doi":"10.1186/s12935-025-03762-6","DOIUrl":"10.1186/s12935-025-03762-6","url":null,"abstract":"<p><p>Glioblastoma (GBM) the most common and most aggressive primary brain tumor has a five-year survival rate of less than 5%. The onset of GBM is very complicated and has always been the focus of researchers. This study analyzed data from 637 GBM and 20 normal tissues from The Cancer Genome Atlas (TCGA), and patients were categorized into high and low EIF2S2 expression groups. The Overall survival and disease-free survival of GBM patients in low expression of EIF2S2 group were significantly higher than those in high expression of EIF2S2 group (p < 0.001), and the expression level of EIF2S2 was significantly correlated with tumor grade (p < 0.001) and tumor recurrence (p < 0.001). The study designed three different short hairpin RNA (shRNA) sequence vectors, identifying shEIF2S2-1 as the most effective. This vector significantly reduced EIF2S2 expression, cell proliferation, and migration while increasing apoptosis in SHG-44 and U251 cells (p < 0.01). By detecting SHG-44 cells infected with shEIF2S2 vector and shCtrl with human whole gene expression chip, we identified WNT5A that is a downstream target gene of EIF2S2. Interfering with WNT5A and overexpressing EIF2S2 in SHG-44 and U251 cells revealed that EIF2S2 regulates WNT5A expression. This regulation led to an increased apoptosis rate (p < 0.05) and a significant reduction in cell migration (p < 0.05) in both the EIF2S2 overexpression and shWNT5A interference groups, confirming that WNT5A is a downstream regulatory target of EIF2S2. This study revealed the key role of EIF2S2 in GBM and its potential molecular mechanism.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":"25 1","pages":"126"},"PeriodicalIF":5.3,"publicationDate":"2025-04-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11967041/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143771506","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Cancer Cell International
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1