643 results on '"Xin Yuan Guan"'
Search Results
2. GPRC5A promotes lung colonization of esophageal squamous cell carcinoma
- Author
-
Hongyu Zhou, Licheng Tan, Baifeng Zhang, Dora Lai Wan Kwong, Ching Ngar Wong, Yu Zhang, Beibei Ru, Yingchen Lyu, Kin To Hugo Siu, Jie Luo, Yuma Yang, Qin Liu, Yixin Chen, Weiguang Zhang, Chaohui He, Peng Jiang, Yanru Qin, Beilei Liu, and Xin-Yuan Guan
- Subjects
Science - Abstract
Abstract Emerging evidence suggests that cancer cells may disseminate early, prior to the formation of traditional macro-metastases. However, the mechanisms underlying the seeding and transition of early disseminated cancer cells (DCCs) into metastatic tumors remain poorly understood. Through single-cell RNA sequencing, we show that early lung DCCs from esophageal squamous cell carcinoma (ESCC) exhibit a trophoblast-like ‘tumor implantation’ phenotype, which enhances their dissemination and supports metastatic growth. Notably, ESCC cells overexpressing GPRC5A demonstrate improved implantation and persistence, resulting in macro-metastases in the lungs. Clinically, elevated GPRC5A level is associated with poorer outcomes in a cohort of 148 ESCC patients. Mechanistically, GPRC5A is found to potentially interact with WWP1, facilitating the polyubiquitination and degradation of LATS1, thereby activating YAP1 signaling pathways essential for metastasis. Importantly, targeting YAP1 axis with CA3 or TED-347 significantly diminishes early implantation and macro-metastases. Thus, the GPRC5A/WWP1/LATS1/YAP1 pathway represents a crucial target for therapeutic intervention in ESCC lung metastases.
- Published
- 2024
- Full Text
- View/download PDF
3. Exploring treatment options in cancer: tumor treatment strategies
- Author
-
Beilei Liu, Hongyu Zhou, Licheng Tan, Kin To Hugo Siu, and Xin-Yuan Guan
- Subjects
Medicine ,Biology (General) ,QH301-705.5 - Abstract
Abstract Traditional therapeutic approaches such as chemotherapy and radiation therapy have burdened cancer patients with onerous physical and psychological challenges. Encouragingly, the landscape of tumor treatment has undergone a comprehensive and remarkable transformation. Emerging as fervently pursued modalities are small molecule targeted agents, antibody-drug conjugates (ADCs), cell-based therapies, and gene therapy. These cutting-edge treatment modalities not only afford personalized and precise tumor targeting, but also provide patients with enhanced therapeutic comfort and the potential to impede disease progression. Nonetheless, it is acknowledged that these therapeutic strategies still harbour untapped potential for further advancement. Gaining a comprehensive understanding of the merits and limitations of these treatment modalities holds the promise of offering novel perspectives for clinical practice and foundational research endeavours. In this review, we discussed the different treatment modalities, including small molecule targeted drugs, peptide drugs, antibody drugs, cell therapy, and gene therapy. It will provide a detailed explanation of each method, addressing their status of development, clinical challenges, and potential solutions. The aim is to assist clinicians and researchers in gaining a deeper understanding of these diverse treatment options, enabling them to carry out effective treatment and advance their research more efficiently.
- Published
- 2024
- Full Text
- View/download PDF
4. HSPA4 upregulation induces immune evasion via ALKBH5/CD58 axis in gastric cancer
- Author
-
Daqin Suo, Xiaoling Gao, Qingyun Chen, Tingting Zeng, Jiarong Zhan, Guanghui Li, Yinli Zheng, Senlin Zhu, Jingping Yun, Xin-Yuan Guan, and Yan Li
- Subjects
HSPA4 ,ALKBH5 ,CD58 ,Immunotherapy ,Gastric cancer ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Introduction Gastric cancer (GC) is one of the leading causes of cancer-related death worldwide. Recently, targeted therapies including PD1 (programmed cell death 1) antibodies have been used in advanced GC patients. However, identifying new biomarker for immunotherapy is still urgently needed. The objective of this study is to unveil the immune evasion mechanism of GC cells and identify new biomarkers for immune checkpoint blockade therapy in patients with GC. Methods Coimmunoprecipitation and meRIP were performed to investigate the mechanism of immune evasion of GC cells. Cocuture system was established to evaluate the cytotoxicity of cocultured CD8+ T cells. The clinical significance of HSPA4 upregulation was analyzed by multiplex fluorescent immunohistochemistry staining in GC tumor tissues. Results Histone acetylation causes HSPA4 upregulation in GC tumor tissues. HSPA4 upregulation increases the protein stability of m6A demethylase ALKBH5. ALKBH5 decreases CD58 in GC cells through m6A methylation regulation. The cytotoxicity of CD8+ T cells are impaired and PD1/PDL1 axis is activated when CD8+ T cells are cocultured with HSPA4 overexpressed GC cells. HSPA4 upregulation is associated with worse 5-year overall survival of GC patients receiving only surgery. It is an independent prognosis factor for worse survival of GC patients. In GC patients receiving the combined chemotherapy with anti-PD1 immunotherapy, HSPA4 upregulation is observed in responders compared with non-responders. Conclusion HSPA4 upregulation causes the decrease of CD58 in GC cells via HSPA4/ALKBH5/CD58 axis, followed by PD1/PDL1 activation and impairment of CD8+ T cell’s cytotoxicity, finally induces immune evasion of GC cells. HSPA4 upregulation is associated with worse overall survival of GC patients with only surgery. Meanwhile, HSPA4 upregulation predicts for better response in GC patients receiving the combined immunotherapy.
- Published
- 2024
- Full Text
- View/download PDF
5. Identification and Characterization of Metastasis‐Initiating Cells in ESCC in a Multi‐Timepoint Pulmonary Metastasis Mouse Model
- Author
-
Ching Ngar Wong, Yu Zhang, Beibei Ru, Songna Wang, Hongyu Zhou, Jiarun Lin, Yingchen Lyu, Yanru Qin, Peng Jiang, Victor Ho‐Fun Lee, and Xin‐Yuan Guan
- Subjects
early cancer metastasis ,early metastasis microenvironment ,esophageal squamous cell carcinoma ,metastasis biomarkers ,metastasis‐initiating cells ,multiplex staining ,Science - Abstract
Abstract Metastasis is the biggest obstacle to esophageal squamous cell carcinoma (ESCC) treatment. Single‐cell RNA sequencing analyses are applied to investigate lung metastatic ESCC cells isolated from pulmonary metastasis mouse model at multiple timepoints to characterize early metastatic microenvironment. A small population of parental KYSE30 cell line (Cluster S) resembling metastasis‐initiating cells (MICs) is identified because they survive and colonize at lung metastatic sites. Differential expression profile comparisons between Cluster S and other subpopulations identified a panel of 7 metastasis‐initiating signature genes (MIS), including CD44 and TACSTD2, to represent MICs in ESCC. Functional studies demonstrated MICs (CD44high) exhibited significantly enhanced cell survival (resistances to oxidative stress and apoptosis), migration, invasion, stemness, and in vivo lung metastasis capabilities, while bioinformatics analyses revealed enhanced organ development, stress responses, and neuron development, potentially remodel early metastasis microenvironment. Meanwhile, early metastasizing cells demonstrate quasi‐epithelial‐mesenchymal phenotype to support both invasion and anchorage. Multiplex immunohistochemistry (mIHC) staining of 4 MISs (CD44, S100A14, RHOD, and TACSTD2) in ESCC clinical samples demonstrated differential MIS expression scores (dMISs) predict lymph node metastasis, overall survival, and risk of carcinothrombosis.
- Published
- 2024
- Full Text
- View/download PDF
6. Fibroblasts in metastatic lymph nodes confer cisplatin resistance to ESCC tumor cells via PI16
- Author
-
Lily Liang, Xu Zhang, Xiaodong Su, Tingting Zeng, Daqin Suo, Jingping Yun, Xin Wang, Xin-Yuan Guan, and Yan Li
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Although many studies have compared tumor fibroblasts (T-Fbs) and nontumor fibroblasts (N-Fbs), less is understood about the stromal contribution of metastatic lymph node fibroblasts (LN-Fbs) to the evolving microenvironment. Here, we explored the characteristics of LN-Fbs in esophageal squamous cell carcinoma (ESCC) and the interactions between fibroblasts and ESCC tumor cells in metastatic lymph nodes. Fibroblasts were isolated from tumor, nontumor and metastatic lymph node tissues from different patients with ESCC. Transcriptome sequencing was performed on the fibroblasts. Tumor growth and drug-resistance assays were carried out, and characteristics of T-Fbs, N-Fbs and LN-Fbs were determined. Liquid chromatography-tandem mass spectrometry (LC-MS/MS) was used to assay the culture medium of fibroblasts. The results demonstrated that fibroblasts derived from different tissues had different characteristics. Coculture with LN-Fbs conditioned medium inhibited ESCC tumor cell growth and induced chemoresistance in ESCC cells. LN-Fbs induced chemoresistance to cisplatin in ESCC cells by secreting PI16. Coculture with LN-Fbs conditioned medium decreased cisplatin-induced apoptosis in ESCC cells by regulating the p38 and JNK cell signaling pathways. Survival analyses showed that patients with high PI16 expression in Fbs of lymph nodes exhibited worse overall survival. We also examined PI16 expression in interstitial tissues in ESCC tumor samples of patients receiving platinum-based therapy postsurgery and found that high PI16 expression in tumor interstitial tissues was an independent prognostic factor for ESCC patients. In addition, an in vivo assay demonstrated that PI16 knockdown increased the sensitivity of ESCC cells to cisplatin. Our results suggest that fibroblasts in metastatic lymph nodes decrease apoptosis of ESCC cells via PI16, thereby providing a cisplatin-resistance niche and supporting ESCC tumor cells to survive in metastatic lymph nodes. PI16 is also a potential target for effectively blocking the chemoresistance niche signaling circuit in response to cisplatin.
- Published
- 2023
- Full Text
- View/download PDF
7. SPINK1-induced tumor plasticity provides a therapeutic window for chemotherapy in hepatocellular carcinoma
- Author
-
Ki-Fong Man, Lei Zhou, Huajian Yu, Ka-Hei Lam, Wei Cheng, Jun Yu, Terence K. Lee, Jing-Ping Yun, Xin-Yuan Guan, Ming Liu, and Stephanie Ma
- Subjects
Science - Abstract
Abstract Tumor lineage plasticity, considered a hallmark of cancer, denotes the phenomenon in which tumor cells co-opt developmental pathways to attain cellular plasticity, enabling them to evade targeted therapeutic interventions. However, the underlying molecular events remain largely elusive. Our recent study identified CD133/Prom1 in hepatocellular carcinoma (HCC) tumors to mark proliferative tumor-propagating cells with cancer stem cell-like properties, that follow a dedifferentiation trajectory towards a more embryonic state. Here we show SPINK1 to strongly associate with CD133 + HCC, and tumor dedifferentiation. Enhanced transcriptional activity of SPINK1 is mediated by promoter binding of ELF3, which like CD133, is found to increase following 5-FU and cisplatin treatment; while targeted depletion of CD133 will reduce both ELF3 and SPINK1. Functionally, SPINK1 overexpression promotes tumor initiation, self-renewal, and chemoresistance by driving a deregulated EGFR-ERK-CDK4/6-E2F2 signaling axis to induce dedifferentiation of HCC cells into their ancestral lineages. Depleting SPINK1 function by neutralizing antibody treatment or in vivo lentivirus-mediated Spink1 knockdown dampens HCC cancer growth and their ability to resist chemotherapy. Targeting oncofetal SPINK1 may represent a promising therapeutic option for HCC treatment.
- Published
- 2023
- Full Text
- View/download PDF
8. PPM1G promotes cell proliferation via modulating mutant GOF p53 protein expression in hepatocellular carcinoma
- Author
-
Wen Hu, Shao-Lin Ma, Liang Qiong, Yu Du, Li-Ping Gong, Yu-Hang Pan, Li-Ping Sun, Jing-Yun Wen, Jian-Ning Chen, Xin-Yuan Guan, and Chun-Kui Shao
- Subjects
Biochemistry ,Genetics ,Molecular genetics ,Science - Abstract
Summary: The serine/threonine protein phosphatase family involves series of cellular processes, such as pre-mRNA splicing. The function of one of its members, protein phosphatase, Mg2+/Mn2+ dependent 1G (PPM1G), remains unclear in hepatocellular carcinoma (HCC). Our results demonstrated that PPM1G was significantly overexpressed in HCC cells and tumor tissues compared with the normal liver tissues at both protein and RNA levels. High PPM1G expression is associated with shorter overall survival (p
- Published
- 2024
- Full Text
- View/download PDF
9. Loss of ESRP2 Activates TAK1‐MAPK Signaling through the Fetal RNA‐Splicing Program to Promote Hepatocellular Carcinoma Progression
- Author
-
Qian Yan, Xiaona Fang, Xiaoxia Liu, Sai Guo, Siqi Chen, Min Luo, Ping Lan, and Xin‐Yuan Guan
- Subjects
epithelial splicing regulatory protein 2 ,fetal reprogramming ,hepatocellular carcinoma ,RNA splicing ,TAK1/MAPK activation ,Science - Abstract
Abstract Tumors usually display fetal‐like characteristics, and many oncofetal proteins have been identified. However, fetal‐like reprogramming of RNA splicing in hepatocellular carcinoma (HCC) is poorly understood. Here, it is demonstrated that the expression of epithelial splicing regulatory protein 2 (ESRP2), an RNA splicing factor, is suppressed in fetal hepatocytes and HCC, in parallel with tumor progression. By combining RNA‐Seq with splicing analysis, it is identified that ESRP2 controls the fetal‐to‐adult switch of multiple splice isoforms in HCC. Functionally, ESRP2 suppressed cell proliferation and migration by specifically switching the alternative splicing (AS) of the TAK1 gene and restraining the expression of the fetal and oncogenic isoform, TAK1_ΔE12. Notably, aberrant TAK1 splicing led to the activation of p38MAPK signaling and predicted poor prognosis in HCC patients. Further investigation revealed that TAK1_ΔE12 protein interacted closely with TAB3 and formed liquid condensation in HCC cells, resulting in p38MAPK activation, enhanced cell migration, and accelerated tumorigenesis. Loss of ESRP2 sensitized HCC cells to TAK1 kinase inhibitor (TAK1i), promoting pyroptotic cell death and CD8+ T cell infiltration. Combining TAK1i with immune checkpoint therapy achieved potent tumor regression in mice. Overall, the findings reveal a previously unexplored onco‐fetal reprogramming of RNA splicing and provide novel therapeutic avenues for HCC.
- Published
- 2024
- Full Text
- View/download PDF
10. Functions of double‐negative B cells in autoimmune diseases, infections, and cancers
- Author
-
Michael King Yung Chung, Lanqi Gong, Dora Lai‐Wan Kwong, Victor Ho‐Fun Lee, Ann Wing‐Mui Lee, Xin‐Yuan Guan, Ngar‐Woon Kam, and Wei Dai
- Subjects
autoimmune disease ,cancer immunosuppression ,COVID‐19 ,double‐negative B cells ,tumor microenvironment ,Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Abstract
Abstract Most mature B cells can be divided into four subtypes based on the expression of the surface markers IgD and CD27: IgD+CD27− naïve B cells, IgD+CD27+ unswitched memory B cells, IgD−CD27+ switched memory B cells, and IgD−CD27− double‐negative (DN) B cells. Despite their small population size in normal peripheral blood, DN B cells play integral roles in various diseases. For example, they generate autoimmunity in autoimmune conditions, while these cells may generate both autoimmune and antipathogenic responses in COVID‐19, or act in a purely antipathogenic capacity in malaria. Recently, DN B cells have been identified in nasopharyngeal carcinoma and non‐small‐cell lung cancers, where they may play an immunosuppressive role. The distinct functions that DN B cells play in different diseases suggest that they are a heterogeneous B‐cell population. Therefore, further study of the mechanisms underlying the involvement of DN B cells in these diseases is essential for understanding their pathogenesis and the development of therapeutic strategies. Further research is thus warranted to characterize the DN B‐cell population in detail.
- Published
- 2023
- Full Text
- View/download PDF
11. Nasopharyngeal carcinoma cells promote regulatory T cell development and suppressive activity via CD70-CD27 interaction
- Author
-
Lanqi Gong, Jie Luo, Yu Zhang, Yuma Yang, Shanshan Li, Xiaona Fang, Baifeng Zhang, Jiao Huang, Larry Ka-Yue Chow, Dittman Chung, Jinlin Huang, Cuicui Huang, Qin Liu, Lu Bai, Yuen Chak Tiu, Pingan Wu, Yan Wang, George Sai-Wah Tsao, Dora Lai-wan Kwong, Anne Wing-Mui Lee, Wei Dai, and Xin-Yuan Guan
- Subjects
Science - Abstract
Response rates to immunotherapies in patients with nasopharyngeal carcinoma (NPC) are still limited. Here the authors show that tumor-restricted CD70 correlates with regulatory T cell abundance and suppressive activity in NPC and that CD70 blockade improves response to anti-PD1 in preclinical models.
- Published
- 2023
- Full Text
- View/download PDF
12. Integrated analysis of racial disparities in genomic architecture identifies a trans‐ancestry prognostic subtype in bladder cancer
- Author
-
Baifeng Zhang, Peilin Jia, Jiayin Wang, Guangsheng Pei, Changxi Wang, Shimei Pei, Xiangchun Li, Zhongming Zhao, Xin Yi, Xin‐yuan Guan, and Yi Huang
- Subjects
bladder cancer ,genomic architectures ,tumor heterogeneity ,tumor microenvironment ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
The incidence of bladder cancer and patient survival vary greatly among different populations, but the influence of the associated molecular features and evolutionary processes on its clinical treatment and prognostication remains unknown. Here, we analyze the genomic architectures of 505 bladder cancer patients from Asian/Black/White populations. We identify a previously unknown association between AHNAK mutations and activity of the APOBEC‐a mutational signature, the activity of which varied substantially across populations. All significantly mutated genes but only half of arm‐level somatic copy number alterations (SCNAs) are enriched with clonal events, indicating large‐scale SCNAs as rich sources of bladder cancer clonal diversities. The prevalence of TP53 and ATM clonal mutations as well as the associated burden of SCNAs is significantly higher in Whites/Blacks than in Asians. We identify a trans‐ancestry prognostic subtype of bladder cancer characterized by enrichment of non‐muscle‐invasive patients and muscle‐invasive patients with good prognosis, increased CREBBP/FGFR3/HRAS/NFE2L2 mutations, decreased intra‐tumor heterogeneity and genome instability, and an activated tumor microenvironment.
- Published
- 2023
- Full Text
- View/download PDF
13. 964 Single-cell spatial characterization of microenvironmental impacts on immunotherapy discrepancies between adult and pediatric nasopharyngeal carcinoma patients
- Author
-
Peng Jiang, Yu Zhang, Xin-Yuan Guan, Lanqi Gong, Grace Guan, and Beibei Ru
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2023
- Full Text
- View/download PDF
14. The Origin, Differentiation, and Functions of Cancer-Associated Fibroblasts in Gastrointestinal CancerSummary
- Author
-
Jiao Huang, Wai-Ying Tsang, Zhi-Hong Li, and Xin-Yuan Guan
- Subjects
Tumor Microenvironment ,Cancer-Associated Fibroblasts ,Gastrointestinal Tumors ,Single-Cell Technologies ,Diseases of the digestive system. Gastroenterology ,RC799-869 - Abstract
Emerging evidence has shown the importance of the tumor microenvironment in tumorigenesis and progression. Cancer-associated fibroblasts (CAFs) are one of the most infiltrated stroma cells of the tumor microenvironment in gastrointestinal tumors. CAFs play crucial roles in tumor development and therapeutic response by biologically secreting soluble factors or structurally remodeling the extracellular matrix. Conceivably, CAFs may become excellent targets for tumor prevention and treatment. However, the limited knowledge of the heterogeneity of CAFs represents a huge challenge for clinically targeting CAFs. In this review, we summarize the newest understanding of gastrointestinal CAFs, with a special focus on their origin, differentiation, and function. We also discuss the current understanding of CAF subpopulations as shown by single-cell technologies.
- Published
- 2023
- Full Text
- View/download PDF
15. Blockade of NMT1 enzymatic activity inhibits N-myristoylation of VILIP3 protein and suppresses liver cancer progression
- Author
-
Xiang-Peng Tan, Yan He, Jing Yang, Xian Wei, You-Long Fan, Guo-Geng Zhang, Yi-Dong Zhu, Zheng-Qiu Li, Hua-Xin Liao, Da-Jiang Qin, Xin-Yuan Guan, and Bin Li
- Subjects
Medicine ,Biology (General) ,QH301-705.5 - Abstract
Abstract Hepatocellular carcinoma (HCC) is one of the most common malignant tumors. Identification of the underlying mechanism of HCC progression and exploration of new therapeutic drugs are urgently needed. Here, a compound library consisting of 419 FDA-approved drugs was taken to screen potential anticancer drugs. A series of functional assays showed that desloratadine, an antiallergic drug, can repress proliferation in HCC cell lines, cell-derived xenograft (CDX), patient-derived organoid (PDO) and patient-derived xenograft (PDX) models. N-myristoyl transferase 1 (NMT1) was identified as a target protein of desloratadine by drug affinity responsive target stability (DARTS) and surface plasmon resonance (SPR) assays. Upregulation of NMT1 expression enhanced but NMT1 knockdown suppressed tumor growth in vitro and in vivo. Metabolic labeling and mass spectrometry analyses revealed that Visinin-like protein 3 (VILIP3) was a new substrate of NMT1 in protein N-myristoylation modification, and high NMT1 or VILIP3 expression was associated with advanced stages and poor survival in HCC. Mechanistically, desloratadine binds to Asn-246 in NMT1 and inhibits its enzymatic activity, disrupting the NMT1-mediated myristoylation of the VILIP3 protein and subsequent NFκB/Bcl-2 signaling. Conclusively, this study demonstrates that desloratadine may be a novel anticancer drug and that NMT1-mediated myristoylation contributes to HCC progression and is a potential biomarker and therapeutic target in HCC.
- Published
- 2023
- Full Text
- View/download PDF
16. The current status of tumor microenvironment and cancer stem cells in sorafenib resistance of hepatocellular carcinoma
- Author
-
Siqi Chen, Yaqing Du, Xin-Yuan Guan, and Qian Yan
- Subjects
hepatocellular carcinoma ,sorafenib resistance ,tumor microenvironment ,cancer stem cells ,hypoxia ,immune microenvironment ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Hepatocellular carcinoma (HCC) is a heterogeneous and aggressive liver cancer that presents limited treatment options. Despite being the standard therapy for advanced HCC, sorafenib frequently encounters resistance, emphasizing the need to uncover the underlying mechanisms and develop effective treatments. This comprehensive review highlights the crucial interplay between the tumor microenvironment, cancer stem cells (CSCs), and epithelial-mesenchymal transition (EMT) in the context of sorafenib resistance. The tumor microenvironment, encompassing hypoxia, immune cells, stromal cells, and exosomes, exerts a significant impact on HCC progression and therapy response. Hypoxic conditions and immune cell infiltration create an immunosuppressive milieu, shielding tumor cells from immune surveillance and hindering therapeutic efficacy. Additionally, the presence of CSCs emerges as a prominent contributor to sorafenib resistance, with CD133+ CSCs implicated in drug resistance and tumor initiation. Moreover, CSCs undergo EMT, a process intimately linked to tumor progression, CSC activation, and further promotion of sorafenib resistance, metastasis, and tumor-initiating capacity. Elucidating the correlation between the tumor microenvironment, CSCs, and sorafenib resistance holds paramount importance in the quest to develop reliable biomarkers capable of predicting therapeutic response. Novel therapeutic strategies must consider the influence of the tumor microenvironment and CSC activation to effectively overcome sorafenib resistance in HCC.
- Published
- 2023
- Full Text
- View/download PDF
17. Optimal time for early therapeutic response prediction in nasopharyngeal carcinoma with functional magnetic resonance imaging
- Author
-
Alan W.L. Mui, Anne W.M. Lee, Wai-Tong Ng, Victor H.F. Lee, Varut Vardhanabhuti, Shei-Yee Man, Daniel T.T. Chua, and Xin-Yuan Guan
- Subjects
DCE-MRI ,DW-MRI ,MRS ,ADC ,Nasopharyngeal Carcinoma ,Medical physics. Medical radiology. Nuclear medicine ,R895-920 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Background and Purpose: Physiological changes in tumour occur much earlier than morphological changes. They can potentially be used as biomarkers for therapeutic response prediction. This study aimed to investigate the optimal time for early therapeutic response prediction with multi-parametric magnetic resonance imaging (MRI) in patients with nasopharyngeal carcinoma (NPC) receiving concurrent chemo-radiotherapy (CCRT). Material and Methods: Twenty-seven NPC patients were divided into the responder (N = 23) and the poor-responder (N = 4) groups by their primary tumour post-treatment shrinkages. Single-voxel proton MR spectroscopy (1H-MRS), diffusion-weighted (DW) and dynamic contrast-enhanced (DCE) MRI were scanned at baseline, weekly during CCRT and post-CCRT. The median choline peak in 1H-MRS, the median apparent diffusion coefficient (ADC) in DW-MRI, the median influx rate constant (Ktrans), reflux rate constant (Kep), volume of extravascular-extracellular space per unit volume (Ve), and initial area under the time-intensity curve for the first 60 s (iAUC60) in DCE-MRI were compared between the two groups with the Mann-Whitney tests for any significant difference at different time points. Results: In DW-MRI, the percentage increase in ADC from baseline to week-1 for the responders (median = 11.39%, IQR = 18.13%) was higher than the poor-responders (median = 4.91%, IQR = 7.86%) (p = 0.027). In DCE-MRI, the iAUC60 on week-2 was found significantly higher in the poor-responders (median = 0.398, IQR = 0.051) than the responders (median = 0.192, IQR = 0.111) (p = 0.012). No significant difference was found in median choline peaks in 1H-MRS at all time points. Conclusion: Early perfusion and diffusion changes occurred in primary tumours of NPC patients treated with CCRT. The DW-MRI on week-1 and the DCE-MRI on week-2 were the optimal time points for early therapeutic response prediction.
- Published
- 2023
- Full Text
- View/download PDF
18. Long non-coding RNA NEAT1 mediated RPRD1B stability facilitates fatty acid metabolism and lymph node metastasis via c-Jun/c-Fos/SREBP1 axis in gastric cancer
- Author
-
Yongxu Jia, Qian Yan, Yinli Zheng, Lei Li, Baifeng Zhang, Zhiwei Chang, Zehua Wang, Hong Tang, Yanru Qin, and Xin-Yuan Guan
- Subjects
RPRD1B ,Lymph node metastasis ,Fatty acid metabolism, c-Jun/c-Fos/SREBP1 axis ,NEAT1 ,hnRNPA2B1 ,TRIM25 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Lymph node metastasis is one of most common determinants of the stage and prognosis of gastric cancer (GC). However, the key molecular events and mechanisms mediating lymph node metastasis remain elusive. Methods RNA sequencing was used to identify driver genes responsible for lymph node metastasis in four cases of gastric primary tumors, metastatic lesions of lymph nodes and matched normal gastric epithelial tissue. qRT–PCR and IHC were applied to examine RPRD1B expression. Metastatic functions were evaluated in vitro and in vivo. RNA-seq was used to identify target genes. ChIP, EMSA and dual luciferase reporter assays were conducted to identify the binding sites of target genes. Co-IP, RIP, MeRIP, RNA-FISH and ubiquitin assays were applied to explore the underlying mechanisms. Results The top 8 target genes (RPRD1B, MAP4K4, MCM2, TOPBP1, FRMD8, KBTBD2, ADAM10 and CXCR4) that were significantly upregulated in metastatic lymph nodes of individuals with GC were screened. The transcriptional cofactor RPRD1B (regulation of nuclear pre-mRNA domain containing 1B) was selected for further characterization. The clinical analysis showed that RPRD1B was significantly overexpressed in metastatic lymph nodes and associated with poor outcomes in patients with GC. The Mettl3-induced m6A modification was involved in the upregulation of RPRD1B. Functionally, RPRD1B promoted lymph node metastasis capabilities in vitro and in vivo. Mechanistic studies indicated that RPRD1B increased fatty acid uptake and synthesis by transcriptionally upregulating c-Jun/c-Fos and activating the c-Jun/c-Fos/SREBP1 axis. In addition, NEAT1 was upregulated significantly by c-Jun/c-Fos in RPRD1B-overexpressing cells. NEAT1, in turn, increased the stability of the RPRD1B mRNA by recruiting the m6A “reader” protein hnRNPA2B1 and reduced the degradation of the RPRD1B protein by inhibiting TRIM25-mediated ubiquitination. Notably, this functional circuitry was disrupted by an inhibitor of c-Jun/c-Fos/AP1 proteins (SR11302) and small interfering RNAs targeting NEAT1, leading to a preferential impairment of lymph node metastasis. Conclusions Based on these findings, RPRD1B facilitated FA metabolism and assisted primary tumor implantation in lymph nodes via the c-Jun/c-Fos/SREBP1 axis, which was enhanced by a NEAT1-mediated positive feedback loop, serving as a potential therapeutic target for GC treatment.
- Published
- 2022
- Full Text
- View/download PDF
19. QSOX1 facilitates dormant esophageal cancer stem cells to evade immune elimination via PD-L1 upregulation and CD8 T cell exclusion.
- Author
-
Jia-Ru Wei, Baifeng Zhang, Yu Zhang, Wo-Ming Chen, Xiao-Ping Zhang, Ting-Ting Zeng, Yan Li, Ying-Hui Zhu, Xin-Yuan Guan, and Lei Li
- Abstract
Dormant cancer stem cells (DCSCs) exhibit characteristics of chemotherapy resistance and immune escape, and they are a crucial source of tumor recurrence and metastasis. However, the underlying mechanisms remain unrevealed. We demonstrate that enriched Gzmk+ CD8+ T cells within the niche of esophageal DCSCs restrict the outgrowth of tumor mass. Nonetheless, DCSCs can escape immune elimination by enhancing PD-L1 signaling, thereby maintaining immune equilibrium. Quiescent fibroblast-derived quiescin sulfhydryl oxidase 1 (QSOX1) promotes the expression of PD-L1 and its own expression in DCSCs by elevating the level of reactive oxygen species. Additionally, high QSOX1 in the dormant tumor niche contributes to the exclusion of CD8+ T cells. Conversely, blocking QSOX1 with Ebselen in combination with anti-PD-1 and chemotherapy can effectively eradicate residual DCSCs by reducing PD-L1 expression and promoting CD8+ T cell infiltration. Clinically, high expression of QSOX1 predicts a poor response to anti-PD-1 treatment in patients with esophageal cancer. Thus, our findings reveal a mechanism whereby QSOX1 promotes PD-L1 upregulation and T cell exclusion, facilitating the immune escape of DCSCs, and QSOX1 inhibition, combined with immunotherapy and chemotherapy, represents a promising therapeutic approach for eliminating DCSCs and preventing recurrence. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
20. ARHGAP15 promotes metastatic colonization in gastric cancer by suppressing RAC1-ROS pathway.
- Author
-
Fei-Fei Zhang, Chen Jiang, Dong-Ping Jiang, Yu-Zhu Cui, Xin-Yue Wang, Liang-Zhan Sun, Miao Chen, Ka-On Lam, Sha-Yi Wu, Krista Verhoeft, Dora Lai-Wan Kwong, and Xin-Yuan Guan
- Subjects
Genetics ,QH426-470 - Abstract
The molecular mechanism of tumor metastasis, especially how metastatic tumor cells colonize in a distant site, remains poorly understood. Here we reported that ARHGAP15, a Rho GTPase activating protein, enhanced gastric cancer (GC) metastatic colonization, which was quite different from its reported role as a tumor suppressor gene in other cancers. It was upregulated in metastatic lymph nodes and significantly associated with a poor prognosis. Ectopic expression of ARHGAP15 promoted metastatic colonization of gastric cancer cells in murine lungs and lymph nodes in vivo or protected cells from oxidative-related death in vitro. However, genetic downregulation of ARHGAP15 had the opposite effect. Mechanistically, ARHGAP15 inactivated RAC1 and then decreased intracellular accumulation of reactive oxygen species (ROS), thus enhancing the antioxidant capacity of colonizing tumor cells under oxidative stress. This phenotype could be phenocopied by inhibition of RAC1 or rescued by the introduction of constitutively active RAC1 into cells. Taken together, these findings suggested a novel role of ARHGAP15 in promoting gastric cancer metastasis by quenching ROS through inhibiting RAC1 and its potential value for prognosis estimation and targeted therapy.
- Published
- 2023
- Full Text
- View/download PDF
21. CircLONP2 enhances colorectal carcinoma invasion and metastasis through modulating the maturation and exosomal dissemination of microRNA-17
- Author
-
Kai Han, Feng-Wei Wang, Chen-Hui Cao, Han Ling, Jie-Wei Chen, Ri-Xin Chen, Zi-Hao Feng, Jie Luo, Xiao-Han Jin, Jin-Ling Duan, Shu-Man Li, Ning-Fang Ma, Jing-Ping Yun, Xin-Yuan Guan, Zhi-Zhong Pan, Ping Lan, Rui-Hua Xu, and Dan Xie
- Subjects
Colorectal carcinoma ,circLONP2 ,Metastasis ,microRNA-17 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Metastasis causes the vast majority of colorectal carcinoma (CRC)-related deaths. However, little is known about the specific traits and underlying mechanisms of metastasis-initiating cells in primary CRC. And whether or not circular RNAs (circRNAs) take part in this particular event remain not adequately stated yet. Methods A screening method based on Transwell assay was first applied to build CRC subgroups with different metastatic potential. High throughput RNA sequencing was used to find out novel metastatic drivers in CRC metastasis-initiating step. A series of in vitro and in vivo assays were further applied to elucidate the functions and underlying molecular mechanisms of circRNAs in CRC metastasis. Results A circRNA consisting of exon 8–11 of LONP2, termed as circLONP2, was upregulated in metastasis-initiating CRC subgroups. Aberrant higher expression of circLONP2 was observed in primary CRC tissues with established metastasis, and along the invasive margin in metastatic site. High expression of circLONP2 predicted unfavorable overall survival. Functional studies revealed that circLONP2 could enhance the invasiveness of CRC cells in vitro, and targeting circLONP2 through anti-sense oligonucleotide (ASO) dramatically reduced the penetrance of metastasis to foreign organs in vivo. Mechanically, circLONP2 directly interacted with and promoted the processing of primary microRNA-17 (pri-miR-17), through recruiting DiGeorge syndrome critical region gene 8 (DGCR8) and Drosha complex in DDX1-dependent manner. Meanwhile, upregulated mature miR-17-5p could be assembled into exosomes and internalized by neighboring cells to enhance their aggressiveness. Conclusions Our data indicate that circLONP2 acts as key metastasis-initiating molecule during CRC progression through modulating the intracellular maturation and intercellular transfer of miR-17, resulting in dissemination of metastasis-initiating ability in primary site and acceleration of metastasis formation in foreign organs. circLONP2 could serve as an effective prognostic predictor and/or novel anti-metastasis therapeutic target in CRC treatment.
- Published
- 2020
- Full Text
- View/download PDF
22. Potential synthetic lethality for breast cancer: A selective sirtuin 2 inhibitor combined with a multiple kinase inhibitor sorafenib
- Author
-
Hua-Li Wang, Xue Ma, Xin-Yuan Guan, Chen Song, Guo-Bo Li, Ya-Mei Yu, and Ling-Ling Yang
- Subjects
SIRT2 ,Sorafenib ,Synthetic lethality ,Breast cancer ,Combination therapy ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Sorafenib is a clinically useful multiple kinase inhibitor for the treatment of kidney cancer, liver cancer and acute myelocytic leukemia, while it has shown weak efficacy in suppressing breast cancer. Since sirtuin2 (SIRT2) is an important epigenetic regulator and associated with several cancer types including breast cancer, development and evaluation of new SIRT2 inhibitors to probe their therapeutic potentials is currently desirable. A highly selective SIRT2 inhibitor named I was previously developed by us, which showed activity to inhibit non-small cell lung cancer cell lines in vitro. We herein report expanded screening of I and its structurally similar inactive compound II against other cancer cell lines, and found that I had a wide spectrum of anticancer activity while II had no such effects. The I-sorafenib combination treatment exerted obvious synergistic reduction on cell viability of MCF-7 cells. We observed that the combination treatment could suppress cell proliferation, survival and migration, arrest cell cycle at G0/G1 phase, and induce apoptosis in MCF-7 cells, when compared with the single treatment. In vivo studies revealed that the combination treatment showed stronger tumor growth inhibition (87%), comparing with I-(42.8%) or sorafenib-solely-treated groups (61.1%) in MCF-7 xenograft model. In conclusion, this work clearly revealed a potential synthetic lethality effect for I combined with sorafenib, and will probably offer a new strategy at least for breast cancer treatment.
- Published
- 2022
- Full Text
- View/download PDF
23. Development of an oncogenic dedifferentiation SOX signature with prognostic significance in hepatocellular carcinoma
- Author
-
Mei-Mei Li, Yun-Qiang Tang, Yuan-Feng Gong, Wei Cheng, Hao-Long Li, Fan-En Kong, Wen-Jie Zhu, Shan-Shan Liu, Li Huang, Xin-Yuan Guan, Ning-Fang Ma, and Ming Liu
- Subjects
Oncogenic dedifferentiation ,Prognostic value ,Stem cell-like properties ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Gradual loss of terminal differentiation markers and gain of stem cell-like properties is a major hall mark of cancer malignant progression. The stem cell pluripotent transcriptional factor SOX family play critical roles in governing tumor plasticity and lineage specification. This study aims to establish a novel SOX signature to monitor the extent of tumor dedifferentiation and predict prognostic significance in hepatocellular carcinoma (HCC). Methods The RNA-seq data from The Cancer Genome Atlas (TCGA) LIHC project were chronologically divided into the training (n = 188) and testing cohort (n = 189). LIRI-JP project from International Cancer Genome Consortium (ICGC) data portal was used as an independent validation cohort (n = 232). Kaplan-Meier and multivariable Cox analyses were used to examine the clinical significance and prognostic value of the signature genes. Results The SOX gene family members were found to be aberrantly expressed in clinical HCC patients. A five-gene SOX signature with prognostic value was established in the training cohort. The SOX signature genes were found to be closely associated with tumor grade and tumor stage. Liver cancer dedifferentiation markers (AFP, CD133, EPCAM, and KRT19) were found to be progressively increased while hepatocyte terminal differentiation markers (ALB, G6PC, CYP3A4, and HNF4A) were progressively decreased from HCC patients with low SOX signature scores to patients with high SOX signature scores. Kaplan-Meier survival analysis further indicated that the newly established SOX signature could robustly predict patient overall survival in both training, testing, and independent validation cohort. Conclusions An oncogenic dedifferentiation SOX signature presents a great potential in predicting prognostic significance in HCC, and might provide novel biomarkers for precision oncology further in the clinic.
- Published
- 2019
- Full Text
- View/download PDF
24. SOX9 is a dose-dependent metastatic fate determinant in melanoma
- Author
-
Xintao Yang, Rui Liang, Chunxi Liu, Jessica Aijia Liu, May Pui Lai Cheung, Xuelai Liu, On Ying Man, Xin-Yuan Guan, Hong Lok Lung, and Martin Cheung
- Subjects
SOX9 ,SOX10 ,NEDD9 ,RHOA ,RAC1 ,Melanoma ,Metastatic ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background In this research, we aimed to resolve contradictory results whether SOX9 plays a positive or negative role in melanoma progression and determine whether SOX9 and its closely related member SOX10 share the same or distinct targets in mediating their functions in melanoma. Methods Immunofluorescence, TCGA database and qPCR were used to analyze the correlation between the expression patterns and levels of SOX9, SOX10 and NEDD9 in melanoma patient samples. AlamarBlue, transwell invasion and colony formation assays in melanoma cell lines were conducted to investigate the epistatic relationship between SOX10 and NEDD9, as well as the effects of graded SOX9 expression levels. Lung metastasis was determined by tail vein injection assay. Live cell imaging was conducted to monitor dynamics of melanoma migratory behavior. RHOA and RAC1 activation assays measured the activity of Rho GTPases. Results High SOX9 expression was predominantly detected in patients with distant melanoma metastases whereas SOX10 was present in the different stages of melanoma. Both SOX9 and SOX10 exhibited distinct but overlapping expression patterns with metastatic marker NEDD9. Accordingly, SOX10 was required for NEDD9 expression, which partly mediated its oncogenic functions in melanoma cells. Compensatory upregulation of SOX9 expression in SOX10-inhibited melanoma cells reduced growth and migratory capacity, partly due to elevated expression of cyclin-dependent kinase inhibitor p21 and lack of NEDD9 induction. Conversely, opposite phenomenon was observed when SOX9 expression was further elevated to a range of high SOX9 expression levels in metastatic melanoma specimens, and that high levels of SOX9 can restore melanoma progression in the absence of SOX10 both in vitro and in vivo. In addition, overexpression of SOX9 can also promote invasiveness of the parental melanoma cells by modulating the expression of various matrix metalloproteinases. SOX10 or high SOX9 expression regulates melanoma mesenchymal migration through the NEDD9-mediated focal adhesion dynamics and Rho GTPase signaling. Conclusions These results unravel NEDD9 as a common target for SOX10 or high SOX9 to partly mediate their oncogenic events, and most importantly, reconcile previous discrepancies that suboptimal level of SOX9 expression is anti-metastatic whereas high level of SOX9 is metastatic in a heterogeneous population of melanoma.
- Published
- 2019
- Full Text
- View/download PDF
25. HKDC1 upregulation promotes glycolysis and disease progression, and confers chemoresistance onto gastric cancer
- Author
-
Mei‐Qian Wang, Yi‐Ru Chen, Hui‐Wen Xu, Jia‐Rong Zhan, Da‐Qin Suo, Ji‐Jin Wang, Yuan‐Zhen Ma, Xin‐Yuan Guan, Yan Li, and Sen‐Lin Zhu
- Subjects
Cancer Research ,Oncology ,General Medicine - Abstract
There is increasing evidence that hexokinase is involved in cell proliferation and migration. However, the function of the hexokinase domain containing protein-1 (HKDC1) in gastric cancer (GC) remains unclear. Immunohistochemistry analysis and big data mining were used to evaluate the correlation between HKDC1 expression and clinical features in GC. In addition, the biological function and molecular mechanism of HKDC1 in GC were studied by in vitro and in vivo assays. Our study indicated that HKDC1 expression was upregulated in GC tissues compared with adjacent nontumor tissues. High expression of HKDC1 was associated with worse prognosis. Functional experiments demonstrated that HKDC1 upregulation promoted glycolysis, cell proliferation, and tumorigenesis. In addition, HKDC1 could enhance GC invasion and metastasis by inducing epithelial-mesenchymal transition (EMT). Abrogation of HKDC1 could effectively attenuate its oncogenic and metastatic function. Moreover, HKDC1 promoted GC proliferation and migration in vivo. HKDC1 overexpression conferred chemoresistance to cisplatin, oxaliplatin, and 5-fluorouracil (5-Fu) onto GC cells. Furthermore, nuclear factor kappa-B (NF-κB) inhibitor PS-341 could attenuate tumorigenesis, metastasis, and drug resistance ability induced by HKDC1 overexpression in GC cells. Our results highlight a critical role of HKDC1 in promoting glycolysis, tumorigenesis, and EMT of GC cells via activating the NF-κB pathway. In addition, HKDC1-mediated drug resistance was associated with DNA damage repair, which further activated NF-κB signaling. HKDC1 upregulation may be used as a potential indicator for choosing an effective chemotherapy regimen for GC patients undergoing chemotherapy.
- Published
- 2023
- Full Text
- View/download PDF
26. Down‐regulation of TROP‐2 Predicts Poor Prognosis of Hepatocellular Carcinoma Patients
- Author
-
Sarah T. K. Sin, Yan Li, Ming Liu, Yun‐Fei Yuan, Stephanie Ma, and Xin‐Yuan Guan
- Subjects
Diseases of the digestive system. Gastroenterology ,RC799-869 - Abstract
Hepatocellular carcinoma (HCC) is one of the most common and lethal cancer types worldwide, especially in Asian countries. Genetic alterations, including hyperactivation of oncogenes and loss of expression of tumor suppressor genes, greatly contribute to the initiation and progression of HCC. Here we report that down‐regulation of trophoblast cell surface antigen 2 (TROP‐2) was frequently detected in HCC. Transcriptome sequencing of non‐tumor and HCC patient samples revealed down‐regulation of TROP‐2 in tumor tissues. Immunohistochemical staining showed nearly undetectable levels of TROP‐2 in HCC tissues but distinct and strong staining of TROP‐2 in adjacent non‐tumor tissues. The frequent down‐regulation of TROP‐2 expression was further confirmed in an in‐house cohort of 205 pairs of HCC patient samples and in the Cancer Genome Atlas (TCGA) databases. Furthermore, the down‐regulation of TROP‐2 was associated with poor overall survival of HCC patients, severe adjacent organ invasion, and poor differentiation of HCC. Using bisulfite genomic sequencing and methylation‐specific polymerase chain reaction analyses, we show that higher levels of promoter methylation were detected in the DNA samples of HCC tissues (low TROP‐2 expression) than that of the non‐tumor tissues (high TROP‐2 expression). Conclusion: Taken together, our data suggest that promoter hypermethylation contributes to the frequent down‐regulation of TROP‐2 in HCC, and that TROP‐2 down‐regulation predicts poor prognosis of HCC patients.
- Published
- 2018
- Full Text
- View/download PDF
27. Supplementary Table S1 from Aldehyde Dehydrogenase Discriminates the CD133 Liver Cancer Stem Cell Populations
- Author
-
Xin-Yuan Guan, Bo-Jian Zheng, Jana Yim-Hung Wo, Kwan Ho Tang, Terence Kin-Wah Lee, Kwok Wah Chan, and Stephanie Ma
- Abstract
Supplementary Table S1 from Aldehyde Dehydrogenase Discriminates the CD133 Liver Cancer Stem Cell Populations
- Published
- 2023
- Full Text
- View/download PDF
28. Data from Aldehyde Dehydrogenase Discriminates the CD133 Liver Cancer Stem Cell Populations
- Author
-
Xin-Yuan Guan, Bo-Jian Zheng, Jana Yim-Hung Wo, Kwan Ho Tang, Terence Kin-Wah Lee, Kwok Wah Chan, and Stephanie Ma
- Abstract
Recent efforts in our study of cancer stem cells (CSC) in hepatocellular carcinoma (HCC) have led to the identification of CD133 as a prominent HCC CSC marker. Findings were based on experiments done on cell lines and xenograft tumors where expression of CD133 was detected at levels as high as 65%. Based on the CSC theory, CSCs are believed to represent only a minority number of the tumor mass. This is indicative that our previously characterized CD133+ HCC CSC population is still heterogeneous, consisting of perhaps subsets of cells with differing tumorigenic potential. We hypothesized that it is possible to further enrich the CSC population by means of additional differentially expressed markers. Using a two-dimensional PAGE approach, we compared protein profiles between CD133+ and CD133− subpopulations isolated from Huh7 and PLC8024 and identified aldehyde dehydrogenase 1A1 as one of the proteins that are preferentially expressed in the CD133+ subfraction. Analysis of the expression of several different ALDH isoforms and ALDH enzymatic activity in liver cell lines found ALDH to be positively correlated with CD133 expression. Dual-color flow cytometry analysis found the majority of ALDH+ to be CD133+, yet not all CD133+ HCC cells were ALDH+. Subsequent studies on purified subpopulations found CD133+ALDH+ cells to be significantly more tumorigenic than their CD133−ALDH+ or CD133−ALDH− counterparts, both in vitro and in vivo. These data, combined with those from our previous work, reveal the existence of a hierarchical organization in HCC bearing tumorigenic potential in the order of CD133+ALDH+ > CD133+ALDH− > CD133−ALDH−. ALDH, expressed along CD133, can more specifically characterize the tumorigenic liver CSC population. (Mol Cancer Res 2008;6(7):1146–53)
- Published
- 2023
- Full Text
- View/download PDF
29. A T cell resilience model associated with response to immunotherapy in multiple tumor types
- Author
-
Yu Zhang, Trang Vu, Douglas C. Palmer, Rigel J. Kishton, Lanqi Gong, Jiao Huang, Thanh Nguyen, Zuojia Chen, Cari Smith, Ferenc Livák, Rohit Paul, Chi-Ping Day, Chuan Wu, Glenn Merlino, Kenneth Aldape, Xin-yuan Guan, and Peng Jiang
- Subjects
Mice ,Receptors, Chimeric Antigen ,Animals ,Humans ,Membrane Proteins ,Immunotherapy ,General Medicine ,CD8-Positive T-Lymphocytes ,Carrier Proteins ,Immunotherapy, Adoptive ,Melanoma ,Article ,General Biochemistry, Genetics and Molecular Biology - Abstract
Despite breakthroughs in cancer immunotherapy, most tumor-reactive T cells cannot persist in solid tumors due to an immunosuppressive environment. We developed Tres (tumor-resilient T cell, https://resilience.ccr.cancer.gov/), a computational model utilizing single-cell transcriptomic data to identify signatures of T cells that are resilient to immunosuppressive signals, such as transforming growth factor-β1, tumor necrosis factor-related apoptosis-inducing ligand and prostaglandin E2. Tres reliably predicts clinical responses to immunotherapy in melanoma, lung cancer, triple-negative breast cancer and B cell malignancies using bulk T cell transcriptomic data from pre-treatment tumors from patients who received immune-checkpoint inhibitors (n = 38), infusion products for chimeric antigen receptor T cell therapies (n = 34) and pre-manufacture samples for chimeric antigen receptor T cell or tumor-infiltrating lymphocyte therapies (n = 84). Further, Tres identified FIBP, whose functions are largely unknown, as the top negative marker of tumor-resilient T cells across many solid tumor types. FIBP knockouts in murine and human donor CD8(+) T cells significantly enhanced T cell-mediated cancer killing in in vitro co-cultures. Further, Fibp knockout in murine T cells potentiated the in vivo efficacy of adoptive cell transfer in the B16 tumor model. Fibp knockout T cells exhibit reduced cholesterol metabolism, which inhibits effector T cell function. These results demonstrate the utility of Tres in identifying biomarkers of T cell effectiveness and potential therapeutic targets for immunotherapies in solid tumors.
- Published
- 2022
- Full Text
- View/download PDF
30. Targeting MFGE8 secreted by cancer-associated fibroblasts blocks angiogenesis and metastasis in esophageal squamous cell carcinoma.
- Author
-
Beilei Liu, Baifeng Zhang, Jiali Qi, Hongyu Zhou, Licheng Tan, Jinlin Huang, Jiao Huang, Xiaona Fang, Lanqi Gong, Jie Luo, Shan Liu, Li Fu, Fei Ling, Stephianie Ma, Dora Lai-Wan Kwong, Xin Wang, and Xin-Yuan Guan
- Subjects
SQUAMOUS cell carcinoma ,NEOVASCULARIZATION ,FIBROBLASTS ,CANCER invasiveness ,METASTASIS - Abstract
Cancer-associated fibroblasts (CAFs) play vital roles in establishing a suitable tumor microenvironment. In this study, RNA sequencing data revealed that CAFs could promote cell proliferation, angiogenesis, and ECM reconstitution by binding to integrin families and activating PI3K/AKT pathways in esophageal squamous cell carcinoma (ESCC). The secretions of CAFs play an important role in regulating these biological activities. Among these secretions, we found that MFGE8 is specifically secreted by CAFs in ESCC. Additionally, the secreted MFGE8 protein is essential in CAF-regulated vascularization, tumor proliferation, drug resistance, and metastasis. By binding to Integrin aVß3/aVß5 receptors, MFGE8 promotes tumor progression by activating both the PI3K/AKT and ERK/AKT pathways. Interestingly, the biological function of MFGE8 secreted by CAFs fully demonstrated the major role of CAFs in ESCC and its mode of mechanism, showing that MFGE8 could be a driver factor of CAFs in remodeling the tumor environment. In vivo treatment targeting CAFs-secreting MFGE8 or its receptor produced significant inhibitory effects on ESCC growth and metastasis, which provides an approach for the treatment of ESCC. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
31. Data from Characterization of Tumor-Suppressive Function of SOX6 in Human Esophageal Squamous Cell Carcinoma
- Author
-
Xin-Yuan Guan, Li Fu, Dora L. Kwong, Yan Li, Leilei Chen, Jiaoyu Ai, Yinghui Zhu, Haibo Liu, Fajun Xie, Hong Tang, and Yan-Ru Qin
- Abstract
Purpose: By using cDNA microarray analysis, we identified a transcriptional factor, SOX6, was frequently downregulated in esophageal squamous cell carcinoma (ESCC). The aim of this study is to investigate the role of SOX6 in human esophageal cancer development, and to examine the prevalence and clinical significance of SOX6 downregulation in ESCC.Experimental Design: Expressions of SOX6 mRNA in 50 ESCCs and SOX6 protein in 300 ESCCs were investigated by semiquantitative RT-PCR and immunohistochemistry, respectively. The tumor-suppressive function of SOX6 was characterized by cell growth, foci formation, wound-healing and cell invasive assays, and tumor xenograft experiment. Western blot analysis was applied to detect protein expression levels.Results: SOX6 was frequently downregulated in primary ESCCs in both mRNA level (29/50, 58%) and protein level (149/219, 68.0%), which was significantly associated with the poor differentiation (P = 0.029), lymph node metastases (P = 0.014), advanced TNM stage (P = 0.000), and disease-specific survival (P < 0.001). Multivariate analysis indicated that the downregulation of SOX6 (P = 0.000) was a significant independent prognostic factors for ESCC. Functional studies showed that SOX6 was able to suppress both in vitro and in vivo tumorigenic ability of ESCC cells. The tumor-suppressive mechanism of SOX6 was associated with its role in G1/S cell-cycle arrest by upregulating expressions of p53 and p21WAF1/CIP1 and downregulating expressions of cyclin D1/CDK4, cyclin A, and β-catenin.Conclusions: We provided the first evidence that SOX6 is a novel tumor-suppressor gene in ESCC development and is a potential prognostic marker in ESCC. Clin Cancer Res; 17(1); 46–55. ©2010 AACR.
- Published
- 2023
- Full Text
- View/download PDF
32. Supplemental Table 2 from The RARS–MAD1L1 Fusion Gene Induces Cancer Stem Cell–like Properties and Therapeutic Resistance in Nasopharyngeal Carcinoma
- Author
-
Mu-Sheng Zeng, Xing Zhang, Jennifer R. Grandis, Yun-Fei Xia, Fei Han, Ming-Yuan Chen, Hai-Qiang Mai, Ling Guo, Vivian W.Y. Lui, Jia-Jie Hao, Xin-Yuan Guan, Zi-Feng Wang, Li-Juan Hu, Li-Hua Xu, Ai-Jun Zhou, Yan-min Liu, Li Yuan, Ze-Dong Hu, Zhi-Rui Lin, Zhi-Hua Liu, and Qian Zhong
- Abstract
Cell Line authentication
- Published
- 2023
- Full Text
- View/download PDF
33. supplemental figure legend from The RARS–MAD1L1 Fusion Gene Induces Cancer Stem Cell–like Properties and Therapeutic Resistance in Nasopharyngeal Carcinoma
- Author
-
Mu-Sheng Zeng, Xing Zhang, Jennifer R. Grandis, Yun-Fei Xia, Fei Han, Ming-Yuan Chen, Hai-Qiang Mai, Ling Guo, Vivian W.Y. Lui, Jia-Jie Hao, Xin-Yuan Guan, Zi-Feng Wang, Li-Juan Hu, Li-Hua Xu, Ai-Jun Zhou, Yan-min Liu, Li Yuan, Ze-Dong Hu, Zhi-Rui Lin, Zhi-Hua Liu, and Qian Zhong
- Abstract
supplemental figure legend
- Published
- 2023
- Full Text
- View/download PDF
34. Supplementary Information from TP53INP1 Downregulation Activates a p73-Dependent DUSP10/ERK Signaling Pathway to Promote Metastasis of Hepatocellular Carcinoma
- Author
-
Stephanie Ma, Alice Carrier, Nelson J Dusetti, Terence K Lee, Dan Xie, Yunfei Yuan, Nikki P Lee, Xin-Yuan Guan, Man Tong, Stella Chai, Lok-Hei Chan, and Kai-Yu Ng
- Abstract
Supplementary Tables 1-3 contains primer sequences for qPCR, luciferase reporter and ChIP assays. Figure Legends for Supplementary Figures 1-4 are also included.
- Published
- 2023
- Full Text
- View/download PDF
35. Figure S2 from Epstein–Barr Virus miRNA BART2-5p Promotes Metastasis of Nasopharyngeal Carcinoma by Suppressing RND3
- Author
-
Xin-Yuan Guan, Dora L.W. Kwong, Jing-Ping Yun, Mu-Sheng Zeng, Sai Wah Tsao, Jiabin Lu, Tingting Zeng, Maria Li Lung, Yan-Qun Xiang, Lei Li, and Chen Jiang
- Abstract
Supplementary Figure 2 (a) The four potential binding sites (position 931,1497,1522,1635) on the 3'UTR were predicted by the PITA analysis. Bases labeled blue were the mutated nucleotides of potential binding sites. (b) Downregulated RND3 expression in BART2 expressing CNE2 cells was revealed by western blot. (c) RIP assay was performed with CNE2 cells with Vector or BART2 expression. The RT-PCR revealed that RND3 3'UTR was enriched on the RISC complex in BART2 expressing cells, but not in cells carrying vector. Input: cell lysate before immunoprecipitation. NC: precipitates pulled down by isotype control mouse IgG. Ago2: precipitates pulled down by anti-Ago2 antibody. Ago2 is core component of RISC. (d) Quality control of RIP assay. Western Blot confirmed that Ago2 was successfully and specifically pulled down by anti-Ago2 antibody. Light and heavy chains of primary antibody were also detected. (e) RND3 expression was inhibited in lung metastatic lesions formed by BART2 expressing cells. (f) Knock down efficiency of RND3 by shRNA was determined by western blot. (Left) Migration and invasion assays were performed to evaluate cell motility after knock down RND3. Migrated or invaded cells were counted and statistically compared in the bar chart. (Middle and Right) (g) Successful reconstitution of RND3 was confirmed by western blot. (Left) Cell migration and invasion was evaluated by transwell assay. Migrated or invaded cells were counted and statistically compared in the bar chart. (Middle and Right) (h) HONE-1 EBVï¼^+)and HONE-1 cells were transfected with Control mimic/siRNA targeting BART2-5p or infected by lentivirus to establish stable cell lines with Vector/RND3 expression. Migration assays were performed 8h after transient transfection or once Blasticidin selection finished. Migrated cells were counted under microscope for following statistical analysis. (i) HONE-1 EBVï¼^+)and HONE-1 cells were transfected with Control mimic/siRNA targeting BART2-5p or infected by lentivirus to establish stable cell lines with Vector/RND3 expression. Cell invasiveness was evaluated 8h after transient transfection or once Blasticidin selection finished. Invaded cells were counted under microscope for subsequent statistical analysis.
- Published
- 2023
- Full Text
- View/download PDF
36. Suppl Figure 1 from FSTL1 Promotes Metastasis and Chemoresistance in Esophageal Squamous Cell Carcinoma through NFκB–BMP Signaling Cross-talk
- Author
-
Stephanie Ma, Xin-Yuan Guan, Wen Ning, Kwok Wah Chan, Yan-Ru Qin, Annie L. Cheung, Nikki P. Lee, Simon Law, Xiao-Yan Ming, Terence K. Lee, Man Tong, Tin Lok Wong, Kai Yu Ng, and Marco Chi-Chung Lau
- Abstract
Representative immunohistochemical staining of high, medium and low FSTL1 in non-neoplastic squamous epithelium (non-tumor) and poorly differentiated ESCC tissue from 6 different patients.
- Published
- 2023
- Full Text
- View/download PDF
37. Data from ORAI2 Promotes Gastric Cancer Tumorigenicity and Metastasis through PI3K/Akt Signaling and MAPK-Dependent Focal Adhesion Disassembly
- Author
-
Ka-On Lam, Xin-Yuan Guan, Xiang Lin, Yanru Qin, Yan Li, Krista R. Verhoeft, Ying Tang, Ying Wang, Liang-Zhan Sun, Yu-Zhu Cui, Yongxu Jia, Zhaojie Lv, Feifei Zhang, Jiao Huang, Miao Chen, and Shayi Wu
- Abstract
The ubiquitous second messenger Ca2+ has long been recognized as a key regulator in cell migration. Locally confined Ca2+, in particular, is essential for building front-to-rear Ca2+ gradient, which serves to maintain the morphologic polarity required in directionally migrating cells. However, little is known about the source of the Ca2+ and the mechanism by which they crosstalk between different signaling pathways in cancer cells. Here, we report that calcium release–activated calcium modulator 2 (ORAI2), a poorly characterized store-operated calcium (SOC) channel subunit, predominantly upregulated in the lymph node metastasis of gastric cancer, supports cell proliferation and migration. Clinical data reveal that a high frequency of ORAI2-positive cells in gastric cancer tissues significantly correlated with poor differentiation, invasion, lymph node metastasis, and worse prognosis. Gain- and loss-of-function showed that ORAI2 promotes cell motility, tumor formation, and metastasis in both gastric cancer cell lines and mice. Mechanistically, ORAI2 mediated SOC activity and regulated tumorigenic properties through the activation of the PI3K/Akt signaling pathways. Moreover, ORAI2 enhanced the metastatic ability of gastric cancer cells by inducing FAK-mediated MAPK/ERK activation and promoted focal adhesion disassembly at rear-edge of the cell. Collectively, our results demonstrate that ORAI2 is a novel gene that plays an important role in the tumorigenicity and metastasis of gastric cancer.Significance:These findings describe the critical role of ORAI2 in gastric cancer cell migration and tumor metastasis and uncover the translational potential to advance drug discovery along the ORAI2 signaling pathway.
- Published
- 2023
- Full Text
- View/download PDF
38. Supplementary Tables from Epstein–Barr Virus miRNA BART2-5p Promotes Metastasis of Nasopharyngeal Carcinoma by Suppressing RND3
- Author
-
Xin-Yuan Guan, Dora L.W. Kwong, Jing-Ping Yun, Mu-Sheng Zeng, Sai Wah Tsao, Jiabin Lu, Tingting Zeng, Maria Li Lung, Yan-Qun Xiang, Lei Li, and Chen Jiang
- Abstract
Supplementary tables include: Table.S1 Primers Table.S2 Clinical data of NPC patients Table.S3 Antibody list Table.S4 Multivariate analysis of Progression Free Time in nasopharyngeal carcinoma patients Table.S5 Association of expression level of Bart 2-5p with patients' clinical features in nasopharyngeal carcinoma Table.S6 Summary of metastatic status in spleen injection model
- Published
- 2023
- Full Text
- View/download PDF
39. Data from Epstein–Barr Virus miRNA BART2-5p Promotes Metastasis of Nasopharyngeal Carcinoma by Suppressing RND3
- Author
-
Xin-Yuan Guan, Dora L.W. Kwong, Jing-Ping Yun, Mu-Sheng Zeng, Sai Wah Tsao, Jiabin Lu, Tingting Zeng, Maria Li Lung, Yan-Qun Xiang, Lei Li, and Chen Jiang
- Abstract
Nasopharyngeal carcinoma is an Epstein–Barr virus (EBV)-related malignancy. Recently, we found that the EBV-encoded miRNA BART2-5p was increased in the serum of patients with preclinical nasopharyngeal carcinoma and that the copy number positively correlated with disease progression. In this study, we established its role in nasopharyngeal carcinoma progression and explored underlying mechanisms and clinical significance. BART2-5p was an independent unfavorable prognostic factor for progression-free survival and its circulating abundance positively associated with distant metastasis. Ectopic expression of BART2-5p promoted migration and invasion of EBV-negative nasopharyngeal carcinoma cells, whereas genetic downregulation of BART2-5p in EBV-positive nasopharyngeal carcinoma cells decreased aggressiveness. Mechanistically, BART2-5p targeted RND3, a negative regulator of Rho signaling. Downregulation of RND3 phenocopied the effect of BART2-5p and reconstitution of RND3 rescued the phenotype. By suppressing RND3, BART2-5p activated Rho signaling to enhance cell motility. These findings suggest a novel role for EBV miRNA BART2-5p in promoting nasopharyngeal carcinoma metastasis and its potential value as a prognostic indicator or therapeutic target.Significance:This study shows that EBV-encoded BART2-5p miRNA suppresses expression of the RND3 Rho family GTPase, consequently promoting ROCK signaling, cell motility, and metastatic behavior of NPC cells.
- Published
- 2023
- Full Text
- View/download PDF
40. Supplementary Figure Legends from Epstein–Barr Virus miRNA BART2-5p Promotes Metastasis of Nasopharyngeal Carcinoma by Suppressing RND3
- Author
-
Xin-Yuan Guan, Dora L.W. Kwong, Jing-Ping Yun, Mu-Sheng Zeng, Sai Wah Tsao, Jiabin Lu, Tingting Zeng, Maria Li Lung, Yan-Qun Xiang, Lei Li, and Chen Jiang
- Abstract
Supplementary Figure Legends
- Published
- 2023
- Full Text
- View/download PDF
41. Supplementary Video 2 from ORAI2 Promotes Gastric Cancer Tumorigenicity and Metastasis through PI3K/Akt Signaling and MAPK-Dependent Focal Adhesion Disassembly
- Author
-
Ka-On Lam, Xin-Yuan Guan, Xiang Lin, Yanru Qin, Yan Li, Krista R. Verhoeft, Ying Tang, Ying Wang, Liang-Zhan Sun, Yu-Zhu Cui, Yongxu Jia, Zhaojie Lv, Feifei Zhang, Jiao Huang, Miao Chen, and Shayi Wu
- Abstract
SUN216-Sh2
- Published
- 2023
- Full Text
- View/download PDF
42. Suppl Figure 3 from FSTL1 Promotes Metastasis and Chemoresistance in Esophageal Squamous Cell Carcinoma through NFκB–BMP Signaling Cross-talk
- Author
-
Stephanie Ma, Xin-Yuan Guan, Wen Ning, Kwok Wah Chan, Yan-Ru Qin, Annie L. Cheung, Nikki P. Lee, Simon Law, Xiao-Yan Ming, Terence K. Lee, Man Tong, Tin Lok Wong, Kai Yu Ng, and Marco Chi-Chung Lau
- Abstract
FSTL1(+) cells in ESCC clinical samples that received (with treatment, n = 22) or did not receive (without treatment, n = 15) chemotherapy treatment prior to resection, as detected by IHC analyses.
- Published
- 2023
- Full Text
- View/download PDF
43. Figure S2 from TP53INP1 Downregulation Activates a p73-Dependent DUSP10/ERK Signaling Pathway to Promote Metastasis of Hepatocellular Carcinoma
- Author
-
Stephanie Ma, Alice Carrier, Nelson J Dusetti, Terence K Lee, Dan Xie, Yunfei Yuan, Nikki P Lee, Xin-Yuan Guan, Man Tong, Stella Chai, Lok-Hei Chan, and Kai-Yu Ng
- Abstract
Effects of TP53INP1 shRNAs on migration and invasion was rescued by TP53INP1 overexpression, ruling out potential off-target effects of the knockdown shRNAs.
- Published
- 2023
- Full Text
- View/download PDF
44. Supplementary Table and Figures from ORAI2 Promotes Gastric Cancer Tumorigenicity and Metastasis through PI3K/Akt Signaling and MAPK-Dependent Focal Adhesion Disassembly
- Author
-
Ka-On Lam, Xin-Yuan Guan, Xiang Lin, Yanru Qin, Yan Li, Krista R. Verhoeft, Ying Tang, Ying Wang, Liang-Zhan Sun, Yu-Zhu Cui, Yongxu Jia, Zhaojie Lv, Feifei Zhang, Jiao Huang, Miao Chen, and Shayi Wu
- Abstract
Table and Figures
- Published
- 2023
- Full Text
- View/download PDF
45. Supplementary Data from COOH-Terminal Truncated HBV X Protein Plays Key Role in Hepatocarcinogenesis
- Author
-
Xin-Yuan Guan, Yan An Su, Mengsu Yang, Li Fu, Chi-Hung Tzang, Xueyan Bai, Jackie Fung, Meng-Chao Wu, Yi Wang, Jun Yang, Jun Wu, Dan Xie, Liang Hu, Sze Hang Lau, and Ning-Fang Ma
- Abstract
Supplementary Data from COOH-Terminal Truncated HBV X Protein Plays Key Role in Hepatocarcinogenesis
- Published
- 2023
- Full Text
- View/download PDF
46. Supplementary Materials and Methods from ORAI2 Promotes Gastric Cancer Tumorigenicity and Metastasis through PI3K/Akt Signaling and MAPK-Dependent Focal Adhesion Disassembly
- Author
-
Ka-On Lam, Xin-Yuan Guan, Xiang Lin, Yanru Qin, Yan Li, Krista R. Verhoeft, Ying Tang, Ying Wang, Liang-Zhan Sun, Yu-Zhu Cui, Yongxu Jia, Zhaojie Lv, Feifei Zhang, Jiao Huang, Miao Chen, and Shayi Wu
- Abstract
Supplementary Materials and Methods
- Published
- 2023
- Full Text
- View/download PDF
47. Data from COOH-Terminal Truncated HBV X Protein Plays Key Role in Hepatocarcinogenesis
- Author
-
Xin-Yuan Guan, Yan An Su, Mengsu Yang, Li Fu, Chi-Hung Tzang, Xueyan Bai, Jackie Fung, Meng-Chao Wu, Yi Wang, Jun Yang, Jun Wu, Dan Xie, Liang Hu, Sze Hang Lau, and Ning-Fang Ma
- Abstract
Purpose: X protein (HBx), a product of hepatitis B virus, has been closely associated with the development of hepatocellular carcinoma (HCC). Based on observations that the COOH-terminal truncated HBx was frequently detected in HCC, the aim of this study is to evaluate the function of COOH-terminal truncated HBx in hepatocarcinogenesis.Experimental Design: Expression pattern of HBx was analyzed by immunohistochemistry on tissue microarray containing 194 pairs of HCCs and their matched nontumor liver tissues. MIHA and HepG2 cells transfected with full-length (X2) and COOH-terminal truncated HBx (X1) were tested for their ability to grow in soft agar and form tumors in vivo. Proliferation and apoptosis were assessed using 2,3-bis[2-methoxy-4-nitro-5-sulfophenyl]-2H-tetrazolium-5-carboxanilide inner salt and terminal deoxyribonucleotidyl transferase-mediated dUTP nick-end labeling assays, respectively. To gain additional insight, the expression profile of HepG2-X2 and HepG2-X1 were compared using cDNA microarray.Results: COOH-terminal truncated HBx was frequently detected in HCCs (79.3%, n = 111), and our in vitro and in vivo studies showed that the truncated rather than the full-length HBx could effectively transform immortalized liver cell line MIHA. Interestingly, expression profiling revealed differential expression of key genes implicated in the control of cell cycle and apoptosis.Conclusions: These findings strongly suggest that the COOH-terminal truncated HBx plays a critical role in the HCC carcinogenesis via the activation of cell proliferation.
- Published
- 2023
- Full Text
- View/download PDF
48. Supplementary Video 1 from ORAI2 Promotes Gastric Cancer Tumorigenicity and Metastasis through PI3K/Akt Signaling and MAPK-Dependent Focal Adhesion Disassembly
- Author
-
Ka-On Lam, Xin-Yuan Guan, Xiang Lin, Yanru Qin, Yan Li, Krista R. Verhoeft, Ying Tang, Ying Wang, Liang-Zhan Sun, Yu-Zhu Cui, Yongxu Jia, Zhaojie Lv, Feifei Zhang, Jiao Huang, Miao Chen, and Shayi Wu
- Abstract
SUN216-Ctl
- Published
- 2023
- Full Text
- View/download PDF
49. Supplementary Video S4 from MAP9 Loss Triggers Chromosomal Instability, Initiates Colorectal Tumorigenesis, and Is Associated with Poor Survival of Patients with Colorectal Cancer
- Author
-
Jun Yu, Jerry W. Shay, Housheng H. He, Sunny H. Wong, Hong Wei, Xin Yuan Guan, Guoping Wang, Yanquan Zhang, Shanshan Gao, Yong Zeng, Wen Deng, Yunfei Zhou, Jianning Zhai, Chi Chun Wong, Liuyang Zhao, Chuangen Li, Junzhe Huang, and Shiyan Wang
- Abstract
Supplementary Video S4
- Published
- 2023
- Full Text
- View/download PDF
50. supplementary information-R from PDSS2 Deficiency Induces Hepatocarcinogenesis by Decreasing Mitochondrial Respiration and Reprogramming Glucose Metabolism
- Author
-
Xin-Yuan Guan, Zongwei Cai, Jinjun Li, Peng Huang, Yunfei Yuan, Dan Xie, Xiaoshi Zhang, Wen Deng, Song Gao, Yinghui Zhu, Ying Zhou, Tingting Zeng, Xiaojiao Ban, Yumin Hu, Zhi Tang, Lei Li, Shuhai Lin, and Yan Li
- Abstract
supplementary information (word)
- Published
- 2023
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.