1
|
Peng W, Li GF, Lin GW, Cheng XX, Zuo XY, Lin QH, Liu SQ, Li DJ, Lin DC, Yin JQ, Luo CL, Zhang YY, Xie XB, Bei JX. Identification of novel germline mutations in FUT7 and EXT1 linked with hereditary multiple exostoses. Oncogene 2024:10.1038/s41388-024-03254-3. [PMID: 39690272 DOI: 10.1038/s41388-024-03254-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 11/29/2024] [Accepted: 12/05/2024] [Indexed: 12/19/2024]
Abstract
Hereditary multiple exostoses (HME) is an autosomal dominant skeletal disorder primarily linked with mutations in Exostosin-1 (EXT1) and Exostosin-2 (EXT2) genes. However, not all HME cases can be explained by these mutations, and its pathogenic mechanisms are not fully understood. Herein, utilizing whole-exome sequencing and genetic screening with a family trio design, we identify two novel rare mutations co-segregating with HME in a Chinese family, including a nonsense mutation (c.204G>A, p.Trp68*) in EXT1 and a missense mutation (c.893T>G, p.Phe298Cys) in FUT7. Functional assays reveal that the FUT7 mutation affects the cellular localization of FUT7 protein and regulates cell proliferation. Notably, the simultaneous loss of fut7 and ext1 in a zebrafish model results in severe chondrodysplasia, indicating a functional link between FUT7 and EXT1 in chondrocyte regulation. Additionally, we unveil that FUT7 p.Phe298Cys reduces EXT1 expression through IL6/STAT3/SLUG axis at the transcription level and through ubiquitination-related proteasomal degradation at the protein level. Together, our findings not only identify novel germline mutations in FUT7 and EXT1 genes, but also highlight the critical interaction between these genes, suggesting a potential 'second-hit' mechanism over EXT1 mutations in HME pathogenesis. This insight enhances our understanding of the mechanisms underlying HME and opens new avenues for potential therapeutic interventions.
Collapse
Affiliation(s)
- Wan Peng
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, 510095, P. R. China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
- Blood Tranfusion Department, Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, 510440, P. R. China
| | - Gao-Fei Li
- Innovation Centre of Ministry of Education for Development and Diseases, School of medicine, South China University of Technology, Guangzhou, 510006, P. R. China
| | - Guo-Wang Lin
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Xi-Xi Cheng
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Xiao-Yu Zuo
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Qiao-Hong Lin
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Shu-Qiang Liu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - De-Jun Li
- Prenatal Diagnosis Center, Reproductive Medicine Center, The First Hospital of Jilin University, Changchun, 130021, P. R. China
| | - Dao-Chao Lin
- Department of Orthopaedics, Shulan (Hangzhou) Hospital, Hangzhou, 311000, P. R. China
| | - Jun-Qiang Yin
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, P. R. China
| | - Chun-Ling Luo
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Yi-Yue Zhang
- Innovation Centre of Ministry of Education for Development and Diseases, School of medicine, South China University of Technology, Guangzhou, 510006, P. R. China
| | - Xian-Biao Xie
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, P. R. China.
| | - Jin-Xin Bei
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, 510095, P. R. China.
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China.
- Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China.
- Department of Medical Oncology, National Cancer Centre of Singapore, Singapore, Singapore.
| |
Collapse
|
2
|
YANG YUSHI, HU CHUJIAO, LEI SHAN, BAO XIN, ZENG ZHIRUI, CAO WENPENG. Using Multi-Omics Analysis to Explore Diagnostic Tool and Optimize Drug Therapy Selection for Patients with Glioma Based on Cross-Talk Gene Signature. Oncol Res 2024; 32:1921-1934. [PMID: 39574472 PMCID: PMC11576925 DOI: 10.32604/or.2024.046191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 02/28/2024] [Indexed: 11/24/2024] Open
Abstract
Background The heterogeneity of prognosis and treatment benefits among patients with gliomas is due to tumor microenvironment characteristics. However, biomarkers that reflect microenvironmental characteristics and predict the prognosis of gliomas are limited. Therefore, we aimed to develop a model that can effectively predict prognosis, differentiate microenvironment signatures, and optimize drug selection for patients with glioma. Materials and Methods The CIBERSORT algorithm, bulk sequencing analysis, and single-cell RNA (scRNA) analysis were employed to identify significant cross-talk genes between M2 macrophages and cancer cells in glioma tissues. A predictive model was constructed based on cross-talk gene expression, and its effect on prognosis, recurrence prediction, and microenvironment characteristics was validated in multiple cohorts. The effect of the predictive model on drug selection was evaluated using the OncoPredict algorithm and relevant cellular biology experiments. Results A high abundance of M2 macrophages in glioma tissues indicates poor prognosis, and cross-talk between macrophages and cancer cells plays a crucial role in shaping the tumor microenvironment. Eight genes involved in the cross-talk between macrophages and cancer cells were identified. Among them, periostin (POSTN), chitinase 3 like 1 (CHI3L1), serum amyloid A1 (SAA1), and matrix metallopeptidase 9 (MMP9) were selected to construct a predictive model. The developed model demonstrated significant efficacy in distinguishing patient prognosis, recurrent cases, and characteristics of high inflammation, hypoxia, and immunosuppression. Furthermore, this model can serve as a valuable tool for guiding the use of trametinib. Conclusions In summary, this study provides a comprehensive understanding of the interplay between M2 macrophages and cancer cells in glioma; utilizes a cross-talk gene signature to develop a predictive model that can predict the differentiation of patient prognosis, recurrence instances, and microenvironment characteristics; and aids in optimizing the application of trametinib in glioma patients.
Collapse
Affiliation(s)
- YUSHI YANG
- Department of Pathology and Pathophysiology, School of Basic Medicine, Guizhou Medical University, Guiyang, 550025, China
| | - CHUJIAO HU
- Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D; State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China
| | - SHAN LEI
- Department of Physiology, School of Basic Medicine, Guizhou Medical University, Guiyang, 550025, China
| | - XIN BAO
- Department of Physiology, School of Basic Medicine, Guizhou Medical University, Guiyang, 550025, China
| | - ZHIRUI ZENG
- Department of Physiology, School of Basic Medicine, Guizhou Medical University, Guiyang, 550025, China
| | - WENPENG CAO
- Department of Anatomy, School of Basic Medicine, Guizhou Medical University, Key Laboratory of Human Brain Bank for Functions and Diseases of Department of Education of Guizhou Province, Guizhou Medical University, Guiyang, 550025, China
| |
Collapse
|
3
|
Kociper B, Škorja Milić N, Ogrizek I, Miš K, Pirkmajer S. Inhibition of the ubiquitin-proteasome system reduces the abundance of pyruvate dehydrogenase kinase 1 in cultured myotubes. J Muscle Res Cell Motil 2024; 45:155-169. [PMID: 39080182 DOI: 10.1007/s10974-024-09679-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 07/05/2024] [Indexed: 08/11/2024]
Abstract
Pyruvate dehydrogenase kinase (PDK), which phosphorylates the pyruvate dehydrogenase complex, regulates glucose metabolism in skeletal muscle. PDK1, an isozyme whose expression is controlled by hypoxia-inducible factor-1α (HIF-1α), is thought to play a role in muscle adaptation to hypoxia. While transcriptional upregulation of PDK1 by HIF-1α is well characterised, mechanisms controlling proteolysis of PDK1 in skeletal muscle have not been thoroughly investigated. Proteasome inhibitor MG132 paradoxically reduced the abundance of PDK1 in human cancer cells and rat L6 myotubes, suggesting that MG132 might direct PDK1 towards autophagic degradation. The objectives of our current study were to determine (1) whether MG132 suppresses PDK1 levels in primary human myotubes, (2) whether chloroquine, an inhibitor of autophagy, prevents MG132-induced suppression of PDK1 in L6 myotubes, and (3) whether PYR-41, an inhibitor of ubiquitination, suppresses PDK1 in L6 myotubes. Using qPCR and/or immunoblotting, we found that despite markedly upregulating HIF-1α protein, MG132 did not alter the PDK1 expression in cultured primary human myotubes, while it suppressed both PDK1 mRNA and protein in L6 myotubes. The PDK1 levels in L6 myotubes were suppressed also during co-treatment with chloroquine and MG132. PYR-41 markedly increased the abundance of HIF-1α in primary human and L6 myotubes, while reducing the abundance of PDK1. In L6 myotubes treated with PYR-41, chloroquine increased the abundance of the epidermal growth factor receptor, but did not prevent the suppression of PDK1. Collectively, our results suggest that cultured myotubes degrade PDK1 via a pathway that cannot be inhibited by MG132, PYR-41, and/or chloroquine.
Collapse
Affiliation(s)
- Blaž Kociper
- Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloška 4, Ljubljana, 1000, Slovenia
| | - Nives Škorja Milić
- Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloška 4, Ljubljana, 1000, Slovenia
| | - Ivana Ogrizek
- Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloška 4, Ljubljana, 1000, Slovenia
| | - Katarina Miš
- Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloška 4, Ljubljana, 1000, Slovenia
| | - Sergej Pirkmajer
- Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloška 4, Ljubljana, 1000, Slovenia.
| |
Collapse
|
4
|
Huang Y, Yang X, Wei M, Yang X, Yuan Z, Huang J, Wei J, Tian L. FUT11 expression in gastric cancer: its prognostic significance and role in immune regulation. Discov Oncol 2024; 15:250. [PMID: 38941002 PMCID: PMC11213843 DOI: 10.1007/s12672-024-01120-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 06/21/2024] [Indexed: 06/29/2024] Open
Abstract
BACKGROUND Gastric cancer (GC) is a malignant digestive tract tumor with a high recurrence rate and poor prognosis. Fucosylation is important in tumor glycosylation, in which the key enzyme is fucosyltransferase (FUT). FUT11 is a member of the fucosyltransferase family and has been closely associated with the development of multiple cancers. However, the specific relationship between FUT11 and GC prognosis and its molecular mechanism has not been fully studied. This study explored FUT11 expression, clinical correlation, and its role in GC occurrence and development to deepen understanding of its function. METHODS FUT11 expression in 33 cancers was preliminarily analyzed using the Tumor Immunoassay Resource (TIMER2.0) database. FUT11 expression in GC was evaluated using The Cancer Genome Atlas stomach adenocarcinoma (TCGA-STAD) and Gene Expression Profiling Interactive Analysis (GEPIA2) data and verified using the Gene Expression Omnibus (GEO) GSE65801 dataset. Furthermore, we studied the survival prognosis of FUT11 in GC and analyzed its effect on the survival rate of patients with GC using the KM-plotter. We also performed COX regression analysis on TCGA GC clinical data and analyzed FUT11 expression in the pathway using the STRING and LinkedOmics databases. Moreover, the relationship between FUT11 and GC immune infiltration level was examined, and the Kaplan-Meier survival analysis diagram was constructed. The FUT11 genetic variation information was retrieved using cBioPortal, and its drug sensitivity was analyzed using CellMiner. Finally, differential FUT11 expression in GC tissues was verified using immunohistochemistry. RESULTS The data mining and analysis demonstrated that FUT11 expression was abnormally elevated in GC tissues and correlated with poor patient prognosis. The FUT11 expression level was an independent prognostic factor for GC. The difference in FUT11 expression level resulted in different degrees of immune cell infiltration in the patients with GC, which might regulate the tumor microenvironment. FUT11 affected GC development by participating in cancer pathways such as PI3K-AKT, neuroactive ligand-receptor, and MAPK. Immunohistochemical staining revealed that FUT11 was highly expressed in GC. CONCLUSIONS This study revealed that FUT11 expression is significantly increased in GC tissues. This increase is associated with poor prognosis and might affect immune regulation. FUT11 might have immunological and targeted therapeutic value, providing a new approach to GC treatment.
Collapse
Affiliation(s)
- Yanqing Huang
- The First Clinical Medical College, Guangxi Medical University, Nanning, 530021, Guangxi, China
- Gastrointestine & Gland Surgery Division I, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Xiaoying Yang
- The First Clinical Medical College, Guangxi Medical University, Nanning, 530021, Guangxi, China
- Gastrointestine & Gland Surgery Division I, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Mengda Wei
- The First Clinical Medical College, Guangxi Medical University, Nanning, 530021, Guangxi, China
- Gastrointestine & Gland Surgery Division I, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Xi Yang
- The First Clinical Medical College, Guangxi Medical University, Nanning, 530021, Guangxi, China
- Gastrointestine & Gland Surgery Division I, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Zhenmin Yuan
- The First Clinical Medical College, Guangxi Medical University, Nanning, 530021, Guangxi, China
- Gastrointestine & Gland Surgery Division I, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Junjie Huang
- The First Clinical Medical College, Guangxi Medical University, Nanning, 530021, Guangxi, China
- Gastrointestine & Gland Surgery Division I, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Junren Wei
- The First Clinical Medical College, Guangxi Medical University, Nanning, 530021, Guangxi, China
- Gastrointestine & Gland Surgery Division I, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Lei Tian
- The First Clinical Medical College, Guangxi Medical University, Nanning, 530021, Guangxi, China.
- Gastrointestine & Gland Surgery Division I, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China.
| |
Collapse
|
5
|
Shu ML, Xia JK, Yan J, Feng YJ, Qian CJ, Teng XS, Yao J. Circ_0002395 promotes aerobic glycolysis and proliferation in pancreatic adenocarcinoma cells via miR-548c-3p/PDK1 axis. J Bioenerg Biomembr 2024; 56:55-71. [PMID: 38041751 DOI: 10.1007/s10863-023-09995-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 11/12/2023] [Indexed: 12/03/2023]
Abstract
Circular RNAs (circRNAs) showing unusual expressions have been discovered in pancreatic adenocarcinoma (PAAD). However, the functions and underlying mechanisms of these circRNAs still remain largely unclear. Our current study discovered a notable increase in the expression of circRNA hsa_circ_0002395 (circ_0002395) in both PAAD tissues and cell lines. This up-regulation of circ_0002395 was found to be associated with larger tumor sizes and lymph node metastasis. Furthermore, our findings showed that circ_0002395 facilitated aerobic glycolysis and cell proliferation in PAAD cells by regulating the miR-548c-3p/PDK1 axis. Mechanistically, we identified circ_0002395 as a competing endogenous RNA (ceRNA) that sponged miR-548c-3p, thereby promoting PDK1 expression and aerobic glycolysis, and ultimately resulting in the enhancement of cell proliferation. Our findings found that circ_0002395 promoted proliferation of PAAD cells by enhancing PDK1 expression and aerobic glycolysis by sponging miR-548c-3p.
Collapse
Affiliation(s)
- Meng-Lu Shu
- Early Gastrointestinal Cancer Research Center, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, 318000, Zhejiang Province, China
- School of Medicine, Taizhou University, Taizhou, 318000, Zhejiang Province, China
| | - Jun-Kai Xia
- Early Gastrointestinal Cancer Research Center, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, 318000, Zhejiang Province, China
- School of Medicine, Taizhou University, Taizhou, 318000, Zhejiang Province, China
| | - Jing Yan
- Early Gastrointestinal Cancer Research Center, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, 318000, Zhejiang Province, China
- School of Medicine, Taizhou University, Taizhou, 318000, Zhejiang Province, China
| | - Yu-Jie Feng
- Early Gastrointestinal Cancer Research Center, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, 318000, Zhejiang Province, China
- School of Medicine, Taizhou University, Taizhou, 318000, Zhejiang Province, China
| | - Cui-Juan Qian
- Early Gastrointestinal Cancer Research Center, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, 318000, Zhejiang Province, China.
- School of Medicine, Taizhou University, Taizhou, 318000, Zhejiang Province, China.
| | - Xiao-Sheng Teng
- Early Gastrointestinal Cancer Research Center, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, 318000, Zhejiang Province, China.
- Department of Gastroenterology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, 318000, Zhejiang Province, China.
| | - Jun Yao
- School of Medicine, Taizhou University, Taizhou, 318000, Zhejiang Province, China.
- Department of Gastroenterology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, 318000, Zhejiang Province, China.
| |
Collapse
|
6
|
Cao W, Dai S, Ruan W, Long T, Zeng Z, Lei S. Pancreatic stellate cell-derived exosomal tRF-19-PNR8YPJZ promotes proliferation and mobility of pancreatic cancer through AXIN2. J Cell Mol Med 2023; 27:2533-2546. [PMID: 37488774 PMCID: PMC10468654 DOI: 10.1111/jcmm.17852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 07/05/2023] [Accepted: 07/08/2023] [Indexed: 07/26/2023] Open
Abstract
The pancreatic stellate cells (PSCs) play an important role in the development of pancreatic cancer (PC) through mechanisms that remain unclear. Exosomes secreted from PSCs act as mediators for communication in PC. This study aimed to explore the role of PSC-derived exosomal small RNAs derived from tRNAs (tDRs) in PC cells. Exosomes from PSCs were extracted and used to detect their effects on PC cell proliferation, migration and invasion. Exosomal tDRs profiling was performed to identify PSC-derived exosomal tDRs. ISH and qRT-PCR were used to examine the tRF-19-PNR8YPJZ levels and clinical value in clinical samples. The biological function of exosomal tRF-19-PNR8YPJZ was determined using the CCK-8, clone formation, wound healing and transwell assays, subcutaneous tumour formation and lung metastatic models. The relationship between the selected exosomal tRF-19-PNR8YPJZ and AXIN2 was determined by RNA sequencing, luciferase reporter assay. PSC-derived exosomes promoted the proliferation, migration, and invasion of PC cells. Novel and abundant tDRs are found to be differentially expressed in PANC-1 cells after treatment with PSC-derived exosomes, such as tRF-19-PNR8YPJZ. PC tissue samples showed markedly higher levels of tRF-19-PNR8YPJZ than normal controls. Patients with PC exhibiting high tRF-19-PNR8YPJZ expression had a highly lymph node invasion, metastasis, perineural invasion, advanced clinical stage and poor overall survival. Exosomal tRF-19-PNR8YPJZ from PSCs targeted AXIN2 in PC cells and decreased its expression, thus activating the Wnt pathway and promoting proliferation and metastasis. Exosomal tRF-19-PNR8YPJZ from PSCs promoted proliferation and metastasis in PC cells via AXIN2.
Collapse
Affiliation(s)
- Wenpeng Cao
- Department of Anatomy, School of Basic MedicineGuizhou Medical UniversityGuiyangChina
| | - Shisi Dai
- Department of Anatomy, School of Basic MedicineGuizhou Medical UniversityGuiyangChina
- Department of Anatomy, School of Basic MedicineGuizhou Nursing Vocational collegeGuiyangChina
| | - Wanyuan Ruan
- School of Clinical MedicineGuizhou Medical UniversityGuiyangChina
| | - Tingting Long
- Department of Anatomy, School of Basic MedicineGuizhou Medical UniversityGuiyangChina
| | - Zhirui Zeng
- Department of Physiology, School of Basic MedicineGuizhou Medical UniversityGuiyangChina
| | - Shan Lei
- Department of Physiology, School of Basic MedicineGuizhou Medical UniversityGuiyangChina
| |
Collapse
|
7
|
Zhang W, Wang H, Wang H, Xu C, Zhao R, Yao J, Zhai C, Han W, Pan H, Sheng J. Integrated Analysis Identifies DPP7 as a Prognostic Biomarker in Colorectal Cancer. Cancers (Basel) 2023; 15:3954. [PMID: 37568770 PMCID: PMC10416901 DOI: 10.3390/cancers15153954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 07/30/2023] [Accepted: 08/02/2023] [Indexed: 08/13/2023] Open
Abstract
Colorectal cancer has a poor prognosis and is prone to recurrence and metastasis. DPP7, a prolyl peptidase, is reported to regulate lymphocyte quiescence. However, the correlation of DPP7 with prognosis in CRC remains unclear. With publicly available cohorts, the Wilcoxon rank-sum test and logistic regression were employed to analyze the relationship between DPP7 expression and the clinicopathological features of CRC patients. Specific pathways of differentially expressed genes were determined through biofunctional analysis and gene set enrichment analysis (GSEA). qPCR and immunohistochemical staining were used to determine DPP7 expression levels in surgical specimens. The public dataset and analysis of the biospecimens of CRC patients revealed that DPP7, in the CRC samples, was expressed significantly higher than in non-tumor tissues. Moreover, increased DPP7 was significantly associated with a higher N stage, lymphatic invasion, and shorter overall survival. Functionally, DPP7 is involved in neuroactive ligand-receptor interaction and olfactory transduction signaling. We identified a series of targeted drugs and small-molecule drugs with responses to DPP7. To conclude, DPP7 is a valuable diagnostic and prognostic biomarker for CRC and considered as a new therapeutic target.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China; (W.Z.); (H.W.); (H.W.); (R.Z.); (J.Y.); (C.Z.); (W.H.); (H.P.)
| | - Haidong Wang
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China; (W.Z.); (H.W.); (H.W.); (R.Z.); (J.Y.); (C.Z.); (W.H.); (H.P.)
| | - Huadi Wang
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China; (W.Z.); (H.W.); (H.W.); (R.Z.); (J.Y.); (C.Z.); (W.H.); (H.P.)
| | - Chuchu Xu
- Department of Gastrointestinal Surgery, Shaoxing People’s Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing 312000, China;
| | - Rongjie Zhao
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China; (W.Z.); (H.W.); (H.W.); (R.Z.); (J.Y.); (C.Z.); (W.H.); (H.P.)
| | - Junlin Yao
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China; (W.Z.); (H.W.); (H.W.); (R.Z.); (J.Y.); (C.Z.); (W.H.); (H.P.)
| | - Chongya Zhai
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China; (W.Z.); (H.W.); (H.W.); (R.Z.); (J.Y.); (C.Z.); (W.H.); (H.P.)
| | - Weidong Han
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China; (W.Z.); (H.W.); (H.W.); (R.Z.); (J.Y.); (C.Z.); (W.H.); (H.P.)
| | - Hongming Pan
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China; (W.Z.); (H.W.); (H.W.); (R.Z.); (J.Y.); (C.Z.); (W.H.); (H.P.)
| | - Jin Sheng
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China; (W.Z.); (H.W.); (H.W.); (R.Z.); (J.Y.); (C.Z.); (W.H.); (H.P.)
| |
Collapse
|
8
|
Wu J, Li S, Huang Y, Zeng Z, Mei T, Wang S, Wang W, Zhang F. MRI features of pituitary adenoma apoplexy and their relationship with hypoxia, proliferation, and pathology. JOURNAL OF CLINICAL ULTRASOUND : JCU 2023. [PMID: 37235536 DOI: 10.1002/jcu.23492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 04/20/2023] [Accepted: 05/12/2023] [Indexed: 05/28/2023]
Abstract
OBJECTIVE We aim to study the MRI features of pituitary adenoma (PA) apoplexy and their relationship with hypoxia, proliferation, and pathology. METHODS Sixty-seven patients with MRI signs of PA apoplexy were selected. According to the MRI signs, they were divided into the parenchymal group and the cystic group. The parenchymal group had a low signal area on T2WI without cyst >2 mm and this area was not significantly enhanced on the corresponding TW1 enhancement. The cystic group had a cyst >2 mm on T2WI, and the cyst showed liquid stratification on T2WI or high signal on T1WI. The relative T1WI (rT1WI) enhancement value and relative T2WI (rT2WI) value of non-apoplexy areas were measured. Protein levels of hypoxia-inducible factor-1 (HIF-1α), pyruvate dehydrogenase kinase 1 (PDK1), and Ki67 were detected with immunohistochemistry and Western blot. Nuclear morphology was observed with HE staining. RESULTS The rT1WI enhancement average value, rT2WI average value, Ki67 protein expression level, and the number of abnormal nuclear morphology of non-apoplexy lesions in the parenchymal group were significantly lower than those in the cystic group. The protein expression levels of HIF-1α and PDK1 in the parenchymal group were significantly higher than those in the cystic group. HIF-1α protein was positively correlated with PDK1 but negatively correlated with Ki67. CONCLUSION When there is PA apoplexy, the ischemia and hypoxia of the cystic group are lesser than those of the parenchymal group, but the proliferation is stronger.
Collapse
Affiliation(s)
- Jianwu Wu
- Department of Neurosurgery, 900 Hospital of the Joint Logistics Team, Fuzhou, People's Republic of China
| | - Songyuan Li
- Department of Neurosurgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, People's Republic of China
| | - Yinxing Huang
- Department of Neurosurgery, 900 Hospital of the Joint Logistics Team, Fuzhou, People's Republic of China
| | - Zihuan Zeng
- Department of Neurosurgery, 900 Hospital of the Joint Logistics Team, Fuzhou, People's Republic of China
| | - Tao Mei
- Department of Neurosurgery, Shenzhen University General Hospital, Shenzhen, People's Republic of China
| | - Shousen Wang
- Department of Neurosurgery, 900 Hospital of the Joint Logistics Team, Fuzhou, People's Republic of China
| | - Wei Wang
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Fangfang Zhang
- Department of Endocrinology, Affiliated Fuzhou First Hospital of Fujian Medical University, Fuzhou, People's Republic of China
| |
Collapse
|
9
|
Comprehensive Analysis Identified Glycosyltransferase Signature to Predict Glioma Prognosis and TAM Phenotype. BIOMED RESEARCH INTERNATIONAL 2023; 2023:6082635. [PMID: 36685667 PMCID: PMC9859707 DOI: 10.1155/2023/6082635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/09/2022] [Accepted: 11/23/2022] [Indexed: 01/15/2023]
Abstract
Glycosylation is the most common posttranslational modification of proteins. Glycosyltransferase gene differential expression dictates the glycosylation model and is epigenetically regulating glioma progression and immunity. This study is aimed at identifying the glycosyltransferase gene signature to predict the prognosis and immune characteristics of glioma. The glycosyltransferase gene signature of glioma was identified in the TCGA database and validated in the CGGA database. Glioma patients were then divided into high- and low-risk groups based on risk scores to compare survival differences and predictive capacity. Subsequently, validation of glycosyltransferase gene signature merits by comparing with other signatures and utility in clinical judgment. The immune cell infiltration, immune pathways, and immune checkpoint expression level were also analyzed and compared in the high- and low-risk groups. Finally, the signature and its gene function were tested in our cohort and in vitro experiments. Eight glycosyltransferase genes were identified to establish the glycosyltransferase signature to predict the prognosis of glioma patients. The survival time was shorter in the high-risk group compared to the low-risk group based on glycosyltransferase signature and was confirmed in an independent external cohort. The glycosyltransferase signature displayed outstanding predictive capacity than other signatures in the TCGA and CGGA database cohorts. Furthermore, patients in the high-risk group were positively correlated with TAM infiltration, immune checkpoint expression level, and protumor immune pathways in TCGA cohorts. Validation of clinical tissue specimens revealed that the high-risk group was closely associated with infiltration of M2 TAMs. High-risk genes in the signature promote glioma proliferation, invasion, and macrophage recruitment in an in vitro validation of U87 and U251 cell lines. This carefully constructed that glycosyltransferase signature can predict the prognosis and immune profile of gliomas and help us evaluate subsequent macrophage-targeted therapies as well as other immune microenvironment modulation therapeutic strategies.
Collapse
|
10
|
Abou Khouzam R, Sharda M, Rao SP, Kyerewah-Kersi SM, Zeinelabdin NA, Mahmood AS, Nawafleh H, Khan MS, Venkatesh GH, Chouaib S. Chronic hypoxia is associated with transcriptomic reprogramming and increased genomic instability in cancer cells. Front Cell Dev Biol 2023; 11:1095419. [PMID: 36968212 PMCID: PMC10033758 DOI: 10.3389/fcell.2023.1095419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 02/23/2023] [Indexed: 03/29/2023] Open
Abstract
Hypoxia afflicts the microenvironment of solid tumors fueling malignancy. We investigated the impact of long hypoxia exposure on transcriptional remodeling, tumor mutational burden (TMB), and genomic instability of cancer cells that were grouped based on their inherent sensitivity or resistance to hypoxia. A hypoxia score was used as a metric to distinguish between the most hypoxia-sensitive (hypoxia high (HH)), and most resistant (hypoxia low (HL)) cancer cells. By applying whole exome sequencing and microarray analysis, we showed that the HH group was indeed more sensitive to hypoxia, having significantly higher TMB (p = 0.03) and copy number losses (p = 0.03), as well as a trend of higher transcriptional response. Globally cells adapted by decreasing expression of genes involved in metabolism, proliferation, and protein maturation, and increasing alternative splicing. They accumulated mutations, especially frameshift insertions, and harbored increased copy number alterations, indicating increased genomic instability. Cells showing highest TMB simultaneously experienced a significant downregulation of DNA replication and repair and chromosomal maintenance pathways. A sixteen-gene common response to chronic hypoxia was put forth, including genes regulating angiogenesis and proliferation. Our findings show that chronic hypoxia enables survival of tumor cells by metabolic reprogramming, modulating proliferation, and increasing genomic instability. They additionally highlight key adaptive pathways that can potentially be targeted to prevent cancer cells residing in chronically hypoxic tumor areas from thriving.
Collapse
Affiliation(s)
- Raefa Abou Khouzam
- Thumbay Research Institute for Precision Medicine, Gulf Medical University, Ajman, United Arab Emirates
| | - Mohak Sharda
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore, Karnataka, India
- School of Life Science, The University of Trans-Disciplinary Health Sciences & Technology (TDU), Bangalore, India
| | - Shyama Prasad Rao
- Center for Bioinformatics, NITTE deemed to be University, Mangaluru, India
| | | | - Nagwa Ahmed Zeinelabdin
- Thumbay Research Institute for Precision Medicine, Gulf Medical University, Ajman, United Arab Emirates
| | - Ayda Shah Mahmood
- Thumbay Research Institute for Precision Medicine, Gulf Medical University, Ajman, United Arab Emirates
| | - Husam Nawafleh
- Thumbay Research Institute for Precision Medicine, Gulf Medical University, Ajman, United Arab Emirates
| | - Munazza Samar Khan
- Thumbay Research Institute for Precision Medicine, Gulf Medical University, Ajman, United Arab Emirates
| | - Goutham Hassan Venkatesh
- Thumbay Research Institute for Precision Medicine, Gulf Medical University, Ajman, United Arab Emirates
| | - Salem Chouaib
- Thumbay Research Institute for Precision Medicine, Gulf Medical University, Ajman, United Arab Emirates
- INSERM UMR 1186, Integrative Tumor Immunology and Cancer Immunotherapy, Gustave Roussy, EPHE, Faculty De médecine University Paris-Sud, University Paris-Saclay, Villejuif, France
- *Correspondence: Salem Chouaib, ,
| |
Collapse
|
11
|
Zeng Z, Lei S, Wang J, Yang Y, Lan J, Tian Q, Chen T, Hao X. A novel hypoxia-driven gene signature that can predict the prognosis of hepatocellular carcinoma. Bioengineered 2022; 13:12193-12210. [PMID: 35549979 PMCID: PMC9276011 DOI: 10.1080/21655979.2022.2073943] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Hypoxia environment exists in already started hepatocellular carcinoma (HCC) and promotes its progression by driving changes in the gene expression profiles of cells. However, the status of hypoxia-driven genes in HCC is largely unknown. In the present study, 368 HCC tissues from The Cancer Genome Atlas were divided into high and low hypoxia groups according to their hypoxia signatures. A total of 1,142 differentially expressed genes (DEGs) were identified between the two groups, and 34 of these DEGs were highly expressed in HCC tissues compared with adjacent tissues, especially in HCC tissues from patients with stage III-IV HCC. After constructing a protein-protein interaction network and applying the least absolute shrinkage and selection operator Cox regression method for 34 DEGs, a three-gene signature (complement factor H related 3 [CFHR3], egl-9 family hypoxia inducible factor 3 [EGLN3], and chromogranin A [CHGA]) was constructed and had prognostic value to predicted outcome of patients with HCC. This three-gene signature was suitable for classifying patients with HCC in the International Cancer Genome Consortium. CFHR3 shows remarkable diagnostic value in HCC. Hypoxia decreased CFHR3 expression, but increased HCC cell proliferation and motility. Overexpression of CFHR3 in HCC cells under hypoxia reversed the stimulatory effects of hypoxia and suppressed cell proliferation and metastasis in vivo. In conclusion, we identified a novel hypoxia-driven gene signature (CFHR3, EGLN3, and CHGA) for reliable prognostic prediction of HCC, and demonstrated that overexpression of CFHR3 may be a potential strategy to overcome hypoxia and treat HCC.
Collapse
Affiliation(s)
- Zhirui Zeng
- Guizhou Provincial Key Laboratory of Pathogenesis & Drug Research on Common Chronic Diseases, Department of Physiology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China.,State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, China.,Key Laboratory of Chemistry for Natural Products of Guizhou Province, Chinese Academy of Sciences, Guiyang, China
| | - Shan Lei
- Guizhou Provincial Key Laboratory of Pathogenesis & Drug Research on Common Chronic Diseases, Department of Physiology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
| | - Jingya Wang
- Guizhou Provincial Key Laboratory of Pathogenesis & Drug Research on Common Chronic Diseases, Department of Physiology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
| | - Yushi Yang
- Department of Pathology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Jinzhi Lan
- Guizhou Provincial Key Laboratory of Pathogenesis & Drug Research on Common Chronic Diseases, Department of Physiology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
| | - Qianting Tian
- Guizhou Provincial Key Laboratory of Pathogenesis & Drug Research on Common Chronic Diseases, Department of Physiology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
| | - Tengxiang Chen
- Guizhou Provincial Key Laboratory of Pathogenesis & Drug Research on Common Chronic Diseases, Department of Physiology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China.,Precision Medicine Research Institute of Guizhou Medical University, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Xiaojiang Hao
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, China.,Key Laboratory of Chemistry for Natural Products of Guizhou Province, Chinese Academy of Sciences, Guiyang, China
| |
Collapse
|
12
|
Cao W, Lei S, Zeng Z, Xiao C, Sun B, Xie P, Li Y, Luo D, Yu W. Transformer 2 alpha homolog is a downstream gene of hypoxia-inducible factor 1 subunit alpha and is involved in the progression of pancreatic cancer. Bioengineered 2022; 13:13238-13251. [PMID: 35635094 PMCID: PMC9275993 DOI: 10.1080/21655979.2022.2079243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 05/11/2022] [Accepted: 05/13/2022] [Indexed: 11/06/2022] Open
Abstract
Intratumoral hypoxia is a common feature of pancreatic cancer (PC) and also plays a role in its progression. However, hypoxia-regulated signatures in PC are still not completely understood. This study aimed to identify core hypoxia-associated genes and determine their underlying molecular mechanisms in PC cells. Transformer 2 alpha homolog (TRA2A) was found to be an important hypoxia-associated gene, which was upregulated in PC tissues and in PC cells cultured under hypoxia. High TRA2A expression was associated with advanced stage, poor differentiation, and lymph node metastasis. Under normoxic and hypoxic conditions, knockdown of TRA2A both markedly suppressed PC cell proliferation and motility in vitro and in vivo, as well as activation of the AKT pathway. Hypoxia-inducible factor 1 subunit alpha (HIF1α) upregulated the transcription of TRA2A by directly binding to its promoter. TRA2A showed a co-expression relationship with HIF1α in PC tissues. Overexpression of TRA2A alleviated the pro-inhibitive functions of HIF1α-inhibition on PC cell proliferation and motility under hypoxia. In conclusion, TRA2A is a crucial downstream gene of HIF1α that accelerates the proliferation and motility of PC cells. TRA2A may be a novel and practical molecular target for investigating the hypoxic response of PC cells.Abbreviations: TRA2A, transformer 2A protein; PC, pancreatic cancer; HIF1α, hypoxia-inducible factor 1-alpha; GEO, Gene Expression Omnibus; IHC, immunohistochemical staining.
Collapse
Affiliation(s)
- Wenpeng Cao
- Department of Anatomy, School of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Shan Lei
- Department of Physiology, School of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Zhirui Zeng
- Department of Physiology, School of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Chaolun Xiao
- Department of Anatomy, School of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Baofei Sun
- Department of Anatomy, School of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Peng Xie
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education, School of Basic Medical, Guizhou Medical University, Guiyang, Guizhou, China
- Key Laboratory of Medical Molecular Biology, School of Basic Medical, Guizhou Medical University, Guiyang, Guizhou, China
| | - Yumei Li
- Department of Anatomy, School of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Daopeng Luo
- Department of Anatomy, School of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Wenfeng Yu
- Department of Anatomy, School of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, China
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education, School of Basic Medical, Guizhou Medical University, Guiyang, Guizhou, China
- Key Laboratory of Medical Molecular Biology, School of Basic Medical, Guizhou Medical University, Guiyang, Guizhou, China
| |
Collapse
|
13
|
Cao W, Tian R, Pan R, Sun B, Xiao C, Chen Y, Zeng Z, Lei S. Terpinen-4-ol inhibits the proliferation and mobility of pancreatic cancer cells by downregulating Rho-associated coiled-coil containing protein kinase 2. Bioengineered 2022; 13:8643-8656. [PMID: 35322742 PMCID: PMC9161900 DOI: 10.1080/21655979.2022.2054205] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Terpinen-4-ol (T4O), a compound isolated from the seeds of turmeric, has exhibited anti-malignancy, anti-aging, and anti-inflammatory properties in previous studies. However, the specific effects and molecular mechanisms of T4O on pancreatic cancer (PC) cells remain largely unknown. In this study, we demonstrated that T4O markedly suppressed PC cell proliferation and colony formation in vitro and induced apoptosis. Similarly, T4O significantly inhibited the migration and invasion of PC cells in vitro. Through RNA sequencing, 858 differentially expressed genes (DEGs) were identified, which were enriched in the Rhodopsin (RHO)/ Ras homolog family member A (RHOA) signaling pathway. Rho-associated coiled-coil containing protein kinase 2 (ROCK2), a DEG enriched in the RHO/RHOA signaling pathway, was considered as a key target of T4O in PC cells; it was significantly reduced after T4O treatment, highly expressed in PC tissues, and negatively associated with patient outcome. Overexpression of ROCK2 significantly reduced the inhibitory effects of T4O on PC cell proliferation and mobility. Moreover, T4O inhibited cell proliferation in vivo and decreased the Ki-67, cell nuclear antigen, EMT markers, and ROCK2 expression. In conclusion, we consider that T4O can suppress the malignant biological behavior of PC by reducing the expression of ROCK2, thus contributing to PC therapy.
Collapse
Affiliation(s)
- Wenpeng Cao
- Department of Anatomy, School of Basic Medicine, Guizhou Medical University, Guiyang, China
| | - Ruhua Tian
- Department of Physiology, School of Basic Medicine, Guizhou Medical University, Guiyang, China
| | - Runsang Pan
- Department of Pathophysiology, School of Basic Medicine, Guizhou Medical University, Guiyang, China
| | - Baofei Sun
- Department of Anatomy, School of Basic Medicine, Guizhou Medical University, Guiyang, China
| | - Chaolun Xiao
- Department of Anatomy, School of Basic Medicine, Guizhou Medical University, Guiyang, China
| | - Yunhua Chen
- Department of Anatomy, School of Basic Medicine, Guizhou Medical University, Guiyang, China
| | - Zhirui Zeng
- Department of Physiology, School of Basic Medicine, Guizhou Medical University, Guiyang, China
| | - Shan Lei
- Department of Physiology, School of Basic Medicine, Guizhou Medical University, Guiyang, China
| |
Collapse
|
14
|
Rosa-Fernandes L, Oba-Shinjo SM, Macedo-da-Silva J, Marie SKN, Palmisano G. Aberrant Protein Glycosylation in Brain Cancers, with Emphasis on Glioblastoma. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1382:39-70. [DOI: 10.1007/978-3-031-05460-0_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
|