1
|
Chen B, Wang D, Xu Y, Guo Q, Pan J, Yu S, Fang Y, Xiao S, Ruan Y, Yang S, Lin M, Hong J, Zhan Z, Lin S. 5-Hydroxymethylcytosines in circulating cell-free DNA as a diagnostic biomarker for nasopharyngeal carcinoma. Eur J Cancer 2024; 210:114294. [PMID: 39213787 DOI: 10.1016/j.ejca.2024.114294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 07/07/2024] [Accepted: 08/12/2024] [Indexed: 09/04/2024]
Abstract
OBJECTIVE To evaluate the diagnostic value of 5-hydroxymethylcytosines (5hmC) in circulating cell-free DNA (cfDNA) for nasopharyngeal carcinoma (NPC) and to develop a diagnostic model. METHODS Genome-wide 5hmC profiles in cfDNA from 174 NPC patients and 146 non-cancer individuals were analyzed using the 5hmC-Seal technique. A cfDNA 5hmC-based diagnostic model to identify NPC patients was developed using least absolute shrinkage and selection operator (LASSO) logistic regression, and performance was evaluated with receiver operating characteristic (ROC) curves and confusion matrices. RESULTS The 5hmC-Seal data from patients with NPC showed a different genome-wide distribution than non-tumor samples. Our initial analysis revealed a 12-gene-based 5hmC marker panel to be an accurate diagnostic model effectively distinguishing between NPC samples and non-cancerous samples (training set: area under curve (AUC)= 0.97 [95 % CI: 0.94-0.99]; and test set: AUC= 0.93 [95 % CI: 0.88-0.98]) superior to EBV DNA testing. The diagnostic score performed well in differentiating the non-cancer subjects from early-stage NPC (training set: AUC=0.99 [95 % CI: 0.98-1]; test set: AUC=0.98 [95 % CI: 0.95-1]), and advanced-stage NPC (training set: AUC=0.96 [95 % CI: 0.93-0.99]; test set: AUC=0.93 [95 % CI: 0.88-0.98]). Notably, in EBV-negative patients, the diagnostic scores showed excellent capacity for distinguishing EBV-negative patients with NPC from non-cancer subjects in both the training set (AUC= 0.94 [95 % CI: 0.88-1]) and test set (AUC=0.91 [95 % CI: 0.81-1]). CONCLUSION 5hmC modifications in cfDNA are promising noninvasive biomarkers for NPC, offering high sensitivity and specificity, particularly for early-stage and EBV-negative NPC.
Collapse
Affiliation(s)
- Bijuan Chen
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian 350014, China
| | - Di Wang
- Department of Molecular Pathology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian 350014, China
| | - Yun Xu
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian 350014, China
| | - Qiaojuan Guo
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian 350014, China
| | - Jianji Pan
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian 350014, China
| | - Sisi Yu
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian 350014, China
| | - Yunxiang Fang
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian 350014, China
| | - Shuxiang Xiao
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian 350014, China
| | - Yuanyuan Ruan
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian 350014, China
| | - Shanshan Yang
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian 350014, China
| | - Mingan Lin
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian 350014, China
| | - Jinsheng Hong
- Department of Radiotherapy, Cancer Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China; Department of Radiotherapy, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China; Key Laboratory of Radiation Biology of Fujian Higher Education Institutions, The First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China.
| | - Zhouwei Zhan
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian 350014, China.
| | - Shaojun Lin
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian 350014, China.
| |
Collapse
|
2
|
Yuan SHC, Wu CC, Wang YC, Chan XY, Chu HW, Yang Y, Liu HP. AGR2-mediated unconventional secretion of 14-3-3ε and α-actinin-4, responsive to ER stress and autophagy, drives chemotaxis in canine mammary tumor cells. Cell Mol Biol Lett 2024; 29:84. [PMID: 38822246 PMCID: PMC11140979 DOI: 10.1186/s11658-024-00601-w] [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: 12/25/2023] [Accepted: 05/21/2024] [Indexed: 06/02/2024] Open
Abstract
BACKGROUND Canine mammary tumors (CMTs) in intact female dogs provide a natural model for investigating metastatic human cancers. Our prior research identified elevated expression of Anterior Gradient 2 (AGR2), a protein disulfide isomerase (PDI) primarily found in the endoplasmic reticulum (ER), in CMT tissues, highly associated with CMT progression. We further demonstrated that increased AGR2 expression actively influences the extracellular microenvironment, promoting chemotaxis in CMT cells. Unraveling the underlying mechanisms is crucial for assessing the potential of therapeutically targeting AGR2 as a strategy to inhibit a pro-metastatic microenvironment and impede tumor metastasis. METHODS To identify the AGR2-modulated secretome, we employed proteomics analysis of the conditioned media (CM) from two CMT cell lines ectopically expressing AGR2, compared with corresponding vector-expressing controls. AGR2-regulated release of 14-3-3ε (gene: YWHAE) and α-actinin 4 (gene: ACTN4) was validated through ectopic expression, knockdown, and knockout of the AGR2 gene in CMT cells. Extracellular vesicles derived from CMT cells were isolated using either differential ultracentrifugation or size exclusion chromatography. The roles of 14-3-3ε and α-actinin 4 in the chemotaxis driven by the AGR2-modulated CM were investigated through gene knockdown, antibody-mediated interference, and recombinant protein supplement. Furthermore, the clinical relevance of the release of 14-3-3ε and α-actinin 4 was assessed using CMT tissue-immersed saline and sera from CMT-afflicted dogs. RESULTS Proteomics analysis of the AGR2-modulated secretome revealed increased abundance in 14-3-3ε and α-actinin 4. Ectopic expression of AGR2 significantly increased the release of 14-3-3ε and α-actinin 4 in the CM. Conversely, knockdown or knockout of AGR2 expression remarkably reduced their release. Silencing 14-3-3ε or α-actinin 4 expression diminished the chemotaxis driven by AGR2-modulated CM. Furthermore, AGR2 controls the release of 14-3-3ε and α-actinin 4 primarily via non-vesicular routes, responding to the endoplasmic reticulum (ER) stress and autophagy activation. Knockout of AGR2 resulted in increased α-actinin 4 accumulation and impaired 14-3-3ε translocation in autophagosomes. Depletion of extracellular 14-3-3ε or α-actinin 4 reduced the chemotaxis driven by AGR2-modulated CM, whereas supplement with recombinant 14-3-3ε in the CM enhanced the CM-driven chemotaxis. Notably, elevated levels of 14-3-3ε or α-actinin 4 were observed in CMT tissue-immersed saline compared with paired non-tumor samples and in the sera of CMT dogs compared with healthy dogs. CONCLUSION This study elucidates AGR2's pivotal role in orchestrating unconventional secretion of 14-3-3ε and α-actinin 4 from CMT cells, thereby contributing to paracrine-mediated chemotaxis. The insight into the intricate interplay between AGR2-involved ER stress, autophagy, and unconventional secretion provides a foundation for refining strategies aimed at impeding metastasis in both canine mammary tumors and potentially human cancers.
Collapse
Affiliation(s)
- Stephen Hsien-Chi Yuan
- Department of Veterinary Medicine, College of Veterinary Medicine, National Chung Hsing University, Taichung, 40227, Taiwan
| | - Chih-Ching Wu
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Otolaryngology-Head and Neck Surgery, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Molecular Medicine Research Center, Chang Gung University, Taoyuan, Taiwan
- Department of Medical Biotechnology and Laboratory Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yu-Chih Wang
- Graduate Institute of Veterinary Pathology, College of Veterinary Medicine, National Chung Hsing University, Taichung, 40227, Taiwan
| | - Xiu-Ya Chan
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Hao-Wei Chu
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Youngsen Yang
- Department of Oncology, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Hao-Ping Liu
- Department of Veterinary Medicine, College of Veterinary Medicine, National Chung Hsing University, Taichung, 40227, Taiwan.
- Biotechnology Center, National Chung Hsing University, Taichung, 40227, Taiwan.
| |
Collapse
|
3
|
Zhou M, Gan XL, Ren YX, Chen QX, Yang YZ, Weng ZJ, Zhang XF, Guan JX, Tang LY, Ren ZF. AGR2 and FOXA1 as prognostic markers in ER-positive breast cancer. BMC Cancer 2023; 23:743. [PMID: 37568077 PMCID: PMC10416444 DOI: 10.1186/s12885-023-10964-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 05/16/2023] [Indexed: 08/13/2023] Open
Abstract
BACKGROUND The prognostic role of either forkhead box A1 (FOXA1) or anterior gradient 2 (AGR2) in breast cancer has been found separately. Considering that there were interplays between them depending on ER status, we aimed to assess the statistical interaction between AGR2 and FOXA1 on breast cancer prognosis and examine the prognostic role of the combination of them by ER status. METHODS AGR2 and FOXA1 expression in tumor tissues were evaluated with tissue microarrays by immunohistochemistry in 915 breast cancer patients with follow up data. The expression levels of these two markers were treated as binary variables, and many different cutoff values were tried for each marker. Survival and Cox proportional hazard analyses were used to evaluate the relationship between AGR2, FOXA1 and prognosis, and the statistical interaction between them on the prognosis was assessed on multiplicative scale. RESULTS Statistical interaction between AGR2 and FOXA1 on the PFS was significant with all the cutoff points in ER-positive breast cancer patients but not ER-negative ones. Among ER-positive patients, the poor prognostic role of the high level of FOXA1 was significant only in patients with the low level of AGR2, and vice versa. When AGR2 and FOXA1 were considered together, patients with low levels of both markers had significantly longer PFS compared with all other groups. CONCLUSIONS There was a statistical interaction between AGR2 and FOXA1 on the prognosis of ER-positive breast cancer. The combination of AGR2 and FOXA1 was a more useful marker for the prognosis of ER-positive breast cancer patients.
Collapse
Affiliation(s)
- Meng Zhou
- School of Public Health, Sun Yat-Sen University, 74 Zhongshan 2Nd Rd, Guangzhou, 510080, China
| | - Xing-Li Gan
- School of Public Health, Sun Yat-Sen University, 74 Zhongshan 2Nd Rd, Guangzhou, 510080, China
| | - Yue-Xiang Ren
- The Third Affiliated Hospital, Sun Yat-Sen University, 600 Tianhe Rd, Guangzhou, 510630, China.
| | - Qian-Xin Chen
- School of Public Health, Sun Yat-Sen University, 74 Zhongshan 2Nd Rd, Guangzhou, 510080, China
| | | | - Zi-Jin Weng
- The Third Affiliated Hospital, Sun Yat-Sen University, 600 Tianhe Rd, Guangzhou, 510630, China
| | - Xiao-Fang Zhang
- The Third Affiliated Hospital, Sun Yat-Sen University, 600 Tianhe Rd, Guangzhou, 510630, China
| | - Jie-Xia Guan
- The Third Affiliated Hospital, Sun Yat-Sen University, 600 Tianhe Rd, Guangzhou, 510630, China
| | - Lu-Ying Tang
- The Third Affiliated Hospital, Sun Yat-Sen University, 600 Tianhe Rd, Guangzhou, 510630, China.
| | - Ze-Fang Ren
- School of Public Health, Sun Yat-Sen University, 74 Zhongshan 2Nd Rd, Guangzhou, 510080, China.
| |
Collapse
|
4
|
Ly TTG, Yun J, Ha JS, Kim YJ, Jang WB, Van Le TH, Rethineswaran VK, Choi J, Kim JH, Min SH, Lee DH, Yang JS, Chung JS, Kwon SM. Inhibitory Effect of Etravirine, a Non-Nucleoside Reverse Transcriptase Inhibitor, via Anterior Gradient Protein 2 Homolog Degradation against Ovarian Cancer Metastasis. Int J Mol Sci 2022; 23:944. [PMID: 35055132 PMCID: PMC8777939 DOI: 10.3390/ijms23020944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 01/11/2022] [Accepted: 01/12/2022] [Indexed: 02/01/2023] Open
Abstract
Anterior gradient protein 2 homolog (AGR2), an endoplasmic reticulum protein, is secreted in the tumor microenvironment. AGR2 is a member of the disulfide isomerase family, is highly expressed in multiple cancers, and promotes cancer metastasis. In this study, we found that etravirine, which is a non-nucleoside reverse transcriptase inhibitor, could induce AGR2 degradation via autophagy. Moreover, etravirine diminished proliferation, migration, and invasion in vitro. Moreover, in an orthotopic xenograft mouse model, the combination of etravirine and paclitaxel significantly suppressed cancer progression and metastasis. This drug may be a promising therapeutic agent for the treatment of ovarian cancer.
Collapse
Affiliation(s)
- Thanh Truong Giang Ly
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan 50612, Korea; (T.T.G.L.); (J.Y.); (J.-S.H.); (Y.-J.K.); (W.-B.J.); (T.H.V.L.); (V.K.R.); (J.C.)
- Convergence Stem Cell Research Center, Pusan National University, Yangsan 50612, Korea;
| | - Jisoo Yun
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan 50612, Korea; (T.T.G.L.); (J.Y.); (J.-S.H.); (Y.-J.K.); (W.-B.J.); (T.H.V.L.); (V.K.R.); (J.C.)
- Convergence Stem Cell Research Center, Pusan National University, Yangsan 50612, Korea;
| | - Jong-Seong Ha
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan 50612, Korea; (T.T.G.L.); (J.Y.); (J.-S.H.); (Y.-J.K.); (W.-B.J.); (T.H.V.L.); (V.K.R.); (J.C.)
- Convergence Stem Cell Research Center, Pusan National University, Yangsan 50612, Korea;
| | - Yeon-Ju Kim
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan 50612, Korea; (T.T.G.L.); (J.Y.); (J.-S.H.); (Y.-J.K.); (W.-B.J.); (T.H.V.L.); (V.K.R.); (J.C.)
- Convergence Stem Cell Research Center, Pusan National University, Yangsan 50612, Korea;
| | - Woong-Bi Jang
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan 50612, Korea; (T.T.G.L.); (J.Y.); (J.-S.H.); (Y.-J.K.); (W.-B.J.); (T.H.V.L.); (V.K.R.); (J.C.)
- Convergence Stem Cell Research Center, Pusan National University, Yangsan 50612, Korea;
| | - Thi Hong Van Le
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan 50612, Korea; (T.T.G.L.); (J.Y.); (J.-S.H.); (Y.-J.K.); (W.-B.J.); (T.H.V.L.); (V.K.R.); (J.C.)
- Convergence Stem Cell Research Center, Pusan National University, Yangsan 50612, Korea;
| | - Vinoth Kumar Rethineswaran
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan 50612, Korea; (T.T.G.L.); (J.Y.); (J.-S.H.); (Y.-J.K.); (W.-B.J.); (T.H.V.L.); (V.K.R.); (J.C.)
- Convergence Stem Cell Research Center, Pusan National University, Yangsan 50612, Korea;
| | - Jaewoo Choi
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan 50612, Korea; (T.T.G.L.); (J.Y.); (J.-S.H.); (Y.-J.K.); (W.-B.J.); (T.H.V.L.); (V.K.R.); (J.C.)
- Convergence Stem Cell Research Center, Pusan National University, Yangsan 50612, Korea;
| | - Jae-Ho Kim
- Convergence Stem Cell Research Center, Pusan National University, Yangsan 50612, Korea;
| | - Sang-Hyun Min
- New Drug Development Center, Deagu Gyeongbuk Medical Innovation Foundation, Deagu 41061, Korea;
| | - Dong-Hyung Lee
- Department of Obstetrics and Gynecology, Pusan National University Yangsan Hospital, Yangsan 50612, Korea; (D.-H.L.); (J.-S.Y.)
| | - Ju-Seok Yang
- Department of Obstetrics and Gynecology, Pusan National University Yangsan Hospital, Yangsan 50612, Korea; (D.-H.L.); (J.-S.Y.)
| | - Joo-Seop Chung
- Department of Hematology-Oncology, Pusan National University Hospital Medical Research Institute, Busan 49241, Korea
| | - Sang-Mo Kwon
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan 50612, Korea; (T.T.G.L.); (J.Y.); (J.-S.H.); (Y.-J.K.); (W.-B.J.); (T.H.V.L.); (V.K.R.); (J.C.)
- Convergence Stem Cell Research Center, Pusan National University, Yangsan 50612, Korea;
| |
Collapse
|
5
|
Yuan SHC, Chang SC, Huang Y, Liu HP. Serum Level of Tumor-Overexpressed AGR2 Is Significantly Associated with Unfavorable Prognosis of Canine Malignant Mammary Tumors. Animals (Basel) 2021; 11:ani11102923. [PMID: 34679944 PMCID: PMC8532596 DOI: 10.3390/ani11102923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/21/2021] [Accepted: 10/07/2021] [Indexed: 11/25/2022] Open
Abstract
Simple Summary Canine malignant mammary tumor (MMT) is a prevalent malignancy in intact female dogs. A current lack of easily accessible tumor biomarkers hinders a timely assessment of the disease outcome. This study reveals that anterior gradient protein 2 (AGR2) is overexpressed in canine MMT tissues, and elevated levels of extracellular AGR2 in sera of MMT dogs are significantly associated with progression and remote metastasis of MMT and an unfavorable overall survival of the patients. Hence, serum eAGR2 level is significantly associated with an adverse outcome of MMT dogs and holds a predictive potential in MMT prognosis. Abstract Canine malignant mammary tumors (MMTs) are prevalent malignancy in intact female dogs with a high incidence of metastasis and recurrence. A current lack of easily accessible tumor biomarkers hinders a timely assessment of the disease outcome. We previously identified anterior gradient protein 2 (AGR2) with higher protein abundance in canine MMT tissues compared with normal counterparts. AGR2 is an endoplasmic reticulum-resident protein disulfide isomerase involved in the regulation of protein processing and also exists extracellularly via secretion to exert pro-oncogenic functions. In the present study, we validated overexpression of AGR2 in canine MMT tissues from 45 dogs using immunohistochemistry and immunoblotting, and assessed serum AGR2 levels in 81 dogs with MMTs and 21 benign cases using a competitive enzyme-linked immunosorbent assay (ELISA). Our data revealed that serum eAGR2 levels are significantly correlated with MMT progression (p = 0.0007) and remote tumor metastasis (p = 0.002). Moreover, elevated levels of serum eAGR2 are associated with an unfavorable overall survival of MMT dogs in later stage (p = 0.0158). Area under the time-dependent ROC curve (AUC) of serum eAGR2 level as a prognostic indicator was 0.839. Collectively, this study uncovered that serum eAGR2 level is significantly associated with an adverse outcome of MMT dogs and holds a predictive potential in MMT prognosis.
Collapse
Affiliation(s)
- Stephen Hsien-Chi Yuan
- Department of Veterinary Medicine, College of Veterinary Medicine, National Chung Hsing University, Taichung 40227, Taiwan; (S.H.-C.Y.); (S.-C.C.)
| | - Shih-Chieh Chang
- Department of Veterinary Medicine, College of Veterinary Medicine, National Chung Hsing University, Taichung 40227, Taiwan; (S.H.-C.Y.); (S.-C.C.)
| | - Yenlin Huang
- Department of Pathology, Chang Gung Memorial Hospital, Taoyuan 33378, Taiwan;
| | - Hao-Ping Liu
- Department of Veterinary Medicine, College of Veterinary Medicine, National Chung Hsing University, Taichung 40227, Taiwan; (S.H.-C.Y.); (S.-C.C.)
- Correspondence: ; Tel.: +886-4-22840368 (ext. 51)
| |
Collapse
|
6
|
de Moraes CL, Cruz E Melo N, Valoyes MAV, Naves do Amaral W. AGR2 and AGR3 play an important role in the clinical characterization and prognosis of basal like breast cancer. Clin Breast Cancer 2021; 22:e242-e252. [PMID: 34462207 DOI: 10.1016/j.clbc.2021.07.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 07/10/2021] [Accepted: 07/18/2021] [Indexed: 12/30/2022]
Abstract
INTRODUCTION Basal-like Breast Cancer (BLBC) represents an important molecular subtype of breast cancer characterized by an aggressive behavior, molecular pathology poorly understood and a limited treatment. OBJECTIVE We aim to search for molecular differences between non-BLBC and BLBC tumors in order to propose possible diagnostic and prognostic biomarkers using databases. Metodology: Microarray processed data were downloaded from GEO database considering non-BLBC and BLBC. Enrichment analysis was evaluated using GO consortium and Ingenuity, protein-protein interaction, gene Ontology and co-expression analysis using STRING. Gene expression data was extracted using TCGA, METABRIC and Breast Cancer Gene-Expression Miner v4.2 databases. The Survival was evaluated using The Kaplan-Meier plotter. RESULTS Were identified 58 upregulated and 58 downregulated genes enriched in signaling pathways like PDGF, Angiogenesis, Integrin and WNT. AGR2 and AGR3 expression were reduced in BLBC in relation to non-BLBC tumors, patients aged ≤51 years, and with negativity of ER, PR and HER-2 and nodal status. Low expression of AGR2 and AGR3 were associated with worse OS and RFS for all breast cancer cases. But according to the molecular stratification, low AGR2 conferred worst OS in luminal A, worst RFS in BLBC and good OS and RFS in luminal B. High AGR3 conferred worse OS and RFS in BLBC, but low AGR3 attributed worse OS in luminal A. CONCLUSION AGR2 and AGR3 expression were able to differentiate non-BLBC from BLBC. Downregulation of AGR2 and AGR3 was associated with BLBC clinical phenotype. Furthermore, both genes behave different when considering prognosis and molecular stratification.
Collapse
Affiliation(s)
- Carolina Leão de Moraes
- Department of Gynaecology and Obstetrics, Faculty of Medicine, Federal University of Goiás, Goiânia, Brazil.
| | - Natália Cruz E Melo
- Department of Gynaecology and Obstetrics, Faculty of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - Maira Andrea Valoyes Valoyes
- Discipline of Oncology, Department of Radiology and Oncology, Faculty of Medicine, University of Sao Paulo, Sao Paulo, Brazil; Laboratory of Molecular Genetics, Center for Translational Research in Oncology (LIM24), Cancer Institute of Sao Paulo, Sao Paulo, Brazil
| | - Waldemar Naves do Amaral
- Department of Gynaecology and Obstetrics, Faculty of Medicine, Federal University of Goiás, Goiânia, Brazil; Graduate Program in Health Sciences, Faculty of Medicine, Federal University of Goiás, Goiânia, Brazil
| |
Collapse
|
7
|
Moidu NA, A Rahman NS, Syafruddin SE, Low TY, Mohtar MA. Secretion of pro-oncogenic AGR2 protein in cancer. Heliyon 2020; 6:e05000. [PMID: 33005802 PMCID: PMC7519367 DOI: 10.1016/j.heliyon.2020.e05000] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 08/25/2020] [Accepted: 09/17/2020] [Indexed: 12/12/2022] Open
Abstract
Anterior gradient-2 (AGR2) protein mediates the formation, breakage and isomerization of disulphide bonds during protein maturation in the endoplasmic reticulum (ER) and contributes to the homoeostasis of the secretory pathway. AGR2 promotes tumour development and metastasis and its elevated expression is almost completely restricted to malignant tumours. Interestingly, this supposedly ER-resident protein can be localised to other compartments of cancer cells and can also be secreted into the extracellular milieu. There are emerging evidences that describe the gain-of-function activities of the extracellular AGR2, particularly in cancer development. Here, we reviewed studies detailing the expression, pathological and physiological roles associated with AGR2 and compared the duality of localization, intracellular and extracellular, with special emphasis on the later. We also discussed the possible mechanisms of AGR2 secretion as well as deliberating the functional impacts of AGR2 in cancer settings. Last, we deliberate the current therapeutic strategies and posit the potential use AGR2, as a prognosis and diagnosis marker in cancer.
Collapse
Affiliation(s)
- Nurshahirah Ashikin Moidu
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia, Cheras 56000 Kuala Lumpur, Malaysia
| | - Nisa Syakila A Rahman
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia, Cheras 56000 Kuala Lumpur, Malaysia
| | - Saiful Effendi Syafruddin
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia, Cheras 56000 Kuala Lumpur, Malaysia
| | - Teck Yew Low
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia, Cheras 56000 Kuala Lumpur, Malaysia
| | - M Aiman Mohtar
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia, Cheras 56000 Kuala Lumpur, Malaysia
| |
Collapse
|
8
|
AGR2-induced glucose metabolism facilitated the progression of endometrial carcinoma via enhancing the MUC1/HIF-1α pathway. Hum Cell 2020; 33:790-800. [PMID: 32304027 DOI: 10.1007/s13577-020-00356-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 04/01/2020] [Indexed: 01/08/2023]
Abstract
Anterior gradient 2 (AGR2) was proved to modulate cancer progression. However, the role of AGR2 on endometrial cancer was not established. Here, we investigated the effects of AGR2 expression on endometrial cancer and explored the regulation mechanism. In the study, we found that AGR2 was overexpressed in tumor tissues of 30 endometrial cancer patients. A high level of AGR2 promoted endometrial cancer cells proliferation, migration and invasion. AGR2 induced the expression of lactate dehydrogenase A (LDHA), phosphoglycerate kinase 1 (PGK1), kallikrein 2 (HK2), and enolase 1-α (ENO1), glucose uptake and lactate production. AGR2 could bind to MUC1 and induce MUC1 and hypoxia-inducible factor 1α (HIF-1α). The inhibition effects of AGR2 knockdown on cells proliferation, migration and invasion ability were abolished by the overexpression of MUC1. Besides, the overexpression of MUC1 also reversed the inhibition effects of AGR2 knockdown on the expression of LDHA, HK2, PGK1 and ENO1, glucose uptake and lactate production. AGR2 knockdown inhibited tumor growth, the levels of Ki-67, MUC1, HIF-1α and glycolysis. In conclusion, AGR2 was overexpressed in endometrial cancer and AGR2-induced glucose metabolism facilitated the progression of endometrial carcinoma via the MUC1/HIF-1α pathway. AGR2 may be an effective therapeutic target for endometrial carcinoma.
Collapse
|
9
|
Alsereihi R, Schulten HJ, Bakhashab S, Saini K, Al-Hejin AM, Hussein D. Leveraging the Role of the Metastatic Associated Protein Anterior Gradient Homologue 2 in Unfolded Protein Degradation: A Novel Therapeutic Biomarker for Cancer. Cancers (Basel) 2019; 11:cancers11070890. [PMID: 31247903 PMCID: PMC6678570 DOI: 10.3390/cancers11070890] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Revised: 06/18/2019] [Accepted: 06/21/2019] [Indexed: 12/15/2022] Open
Abstract
Effective diagnostic, prognostic and therapeutic biomarkers can help in tracking disease progress, predict patients’ survival, and considerably affect the drive for successful clinical management. The present review aims to determine how the metastatic-linked protein anterior gradient homologue 2 (AGR2) operates to affect cancer progression, and to identify associated potential diagnostic, prognostic and therapeutic biomarkers, particularly in central nervous system (CNS) tumors. Studies that show a high expression level of AGR2, and associate the protein expression with the resilience to chemotherapeutic treatments or with poor cancer survival, are reported. The primary protein structures of the seven variants of AGR2, including their functional domains, are summarized. Based on experiments in various biological models, this review shows an orchestra of multiple molecules that regulate AGR2 expression, including a feedback loop with p53. The AGR2-associated molecular functions and pathways including genomic integrity, proliferation, apoptosis, angiogenesis, adhesion, migration, stemness, and inflammation, are detailed. In addition, the mechanisms that can enable the rampant oncogenic effects of AGR2 are clarified. The different strategies used to therapeutically target AGR2-positive cancer cells are evaluated in light of the current evidence. Moreover, novel associated pathways and clinically relevant deregulated genes in AGR2 high CNS tumors are identified using a meta-analysis approach.
Collapse
Affiliation(s)
- Reem Alsereihi
- Neurooncology Translational Group, King Fahd Medical Research Center, King Abdulaziz University, P.O. Box 80216, Jeddah, 21589, Saudi Arabia.
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, P.O. Box 80203, Jeddah 21589, Saudi Arabia.
| | - Hans-Juergen Schulten
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, P.O. Box 80216, Jeddah 21589, Saudi Arabia.
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| | - Sherin Bakhashab
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, P.O. Box 80216, Jeddah 21589, Saudi Arabia.
- Biochemistry Department, King Abdulaziz University, P.O. Box 80218, Jeddah 21589, Saudi Arabia.
| | - Kulvinder Saini
- School of Biotechnology, Eternal University, Baru Sahib-173101, Himachal Pradesh, India.
| | - Ahmed M Al-Hejin
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, P.O. Box 80203, Jeddah 21589, Saudi Arabia.
- Microbiology Unit, King Fahad Medical Research Center, King Abdulaziz University, P.O. Box 80216, Jeddah 21589, Saudi Arabia.
| | - Deema Hussein
- Neurooncology Translational Group, King Fahd Medical Research Center, King Abdulaziz University, P.O. Box 80216, Jeddah, 21589, Saudi Arabia.
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| |
Collapse
|
10
|
Wen W, Mai SJ, Lin HX, Zhang MY, Huang JL, Hua X, Lin C, Long ZQ, Lu ZJ, Sun XQ, Liu SL, Yang Q, Zhu Q, Wang HY, Guo L. Identification of two microRNA signatures in whole blood as novel biomarkers for diagnosis of nasopharyngeal carcinoma. J Transl Med 2019; 17:186. [PMID: 31159814 PMCID: PMC6547589 DOI: 10.1186/s12967-019-1923-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Accepted: 05/15/2019] [Indexed: 12/13/2022] Open
Abstract
Background Early diagnosis is critical to reduce the mortality caused by nasopharyngeal carcinoma (NPC). MicroRNAs (miRNAs) are dysregulated and play important roles in carcinogenesis. Therefore, this study aimed to identify diagnostically relevant circulating miRNA signatures in patients with NPC. Methods Total RNA was extracted from whole blood samples obtained from 120 patients with NPC, 30 patients with head-neck tumors (HNT), and 30 healthy subjects (HSs), and examined by using a custom microarray. The expression levels of four miRNAs identified by using the microarray were validated with quantitative real-time reverse transcription polymerase chain reaction. The 120 patients with NPC and 30 HSs were randomly assigned to training group-1 and validation group-1, respectively. By using significance analysis of microarray (SAM), the specific miRNA expression profiles in whole blood from patients with NPC are obtained. By using lasso regression and adaptive boosting, a diagnostic signature was identified in training group-1, and its accuracy was verified in validation group-1. By using the same methods, another signature to distinguish patients with NPC from those with HNT and HSs was identified in training group-2 and confirmed in validation group-2. Results There were 117 differentially expressed miRNAs (upregulated and downregulated fold change ≥ 1.5) between the patients with NPC and HSs, among which an 8-miRNA signature was identified with 96.43% sensitivity and 100% specificity [area under the curve (AUC) = 0.995] to diagnose NPC in training group-1 and 86.11% sensitivity and 88.89% specificity (AUC = 0.941) in validation group-1. Compared with traditional Epstein–Barr virus (EBV) seromarkers, this signature was more specific for NPC. Furthermore, a 16-miRNA signature to differentiate NPC from HNT and HS (HNT-HS) was established from 164 differentially expressed miRNAs, which diagnosed NPC and HNT-HS with 100% accuracy (AUC = 1.000) in training group-2 and 87.04% (AUC = 0.924) in validation group-2. Conclusions The present study identified two miRNA signatures for the highly accurate diagnosis and differential diagnosis of patients with NPC from HSs and patients with HNT. The identified miRNAs might represent novel serological biomarkers and potential therapeutic targets for NPC. Electronic supplementary material The online version of this article (10.1186/s12967-019-1923-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Wen Wen
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China.,Department of Radiotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Shi-Juan Mai
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Huan-Xin Lin
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China.,Department of Radiotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Mei-Yin Zhang
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Jia-Ling Huang
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Xin Hua
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China.,Department of Radiotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Chao Lin
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China.,Department of Nasopharyngeal Carcinoma, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Zhi-Qing Long
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China.,Department of Radiotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Zi-Jian Lu
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China.,Department of Nasopharyngeal Carcinoma, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Xiao-Qing Sun
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China.,Department of Medical Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Sai-Lan Liu
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China.,Department of Nasopharyngeal Carcinoma, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Qi Yang
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China.,Department of Radiotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Qian Zhu
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Hui-Yun Wang
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China.
| | - Ling Guo
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China. .,Department of Nasopharyngeal Carcinoma, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China.
| |
Collapse
|
11
|
Anterior Gradient-2 monoclonal antibody inhibits lung cancer growth and metastasis by upregulating p53 pathway and without exerting any toxicological effects: A preclinical study. Cancer Lett 2019; 449:125-134. [PMID: 30685412 DOI: 10.1016/j.canlet.2019.01.025] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 12/27/2018] [Accepted: 01/21/2019] [Indexed: 11/24/2022]
Abstract
Increased drug resistance and acute side effects on normal organs are the major disadvantages of traditional cancer chemotherapy and radiotherapy. This has increased the focus on targeted therapeutic strategies such as monoclonal antibody-based cancer therapies. The major advantage of antibody-based therapies is the specific inhibition of cancer-related targets, with reduced off-target side effects. Anterior gradient-2 (AGR2) is a prometastatic and proangiogenic tumor marker that is overexpressed in multiple cancers. Therefore, anti-AGR2 antibodies may be potential therapeutic agents for treating different cancers. In the present study, we examined a novel anti-AGR2 monoclonal antibody mAb18A4 and found that this antibody inhibited lung cancer progression and metastasis without exerting any adverse side effects on the major organs and blood in mice. Moreover, we found that mAb18A4 activated p53 pathway and attenuated ERK1/2-MAPK pathway. Furthermore, mAb18A4-treated cancer cell lines showed attenuated proliferation and colony formation, enhanced apoptosis, increased p53 expression, and reduced phosphorylated ERK1/2 expression. Treatment with mAb18A4 significantly reduced tumor size and suppressed tumor metastasis in and increased the survival of different xenograft tumor models. In addition, mAb18A4 potently suppressed AGR2-induced angiogenesis. Results of pharmacokinetic and toxicological analyses confirmed the safety of mAb18A4 as an antitumor treatment.
Collapse
|
12
|
Zhu Z, Ni H, You B, Shi S, Shan Y, Bao L, Duan B, You Y. Elevated EGFL6 modulates cell metastasis and growth via AKT pathway in nasopharyngeal carcinoma. Cancer Med 2018; 7:6281-6289. [PMID: 30444069 PMCID: PMC6308048 DOI: 10.1002/cam4.1883] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 09/20/2018] [Accepted: 10/29/2018] [Indexed: 11/08/2022] Open
Abstract
Epidermal growth factor-like domain multiple 6 (EGFL6) is a secreted protein, regulates maintenance and metastasis of cancer cells. Nevertheless, how EGFL6 participates in the progression and tumorigenesis of nasopharyngeal carcinoma (NPC) remains unclear. In our study, EGFL6 was detected highly expressed in 20 NPC tissues compared with normal tissues by IHC assay. Then, the level of EGFL6 in NPC serum and NPC cells was explored through enzyme-linked immunosorbent assay and western blot, the results consistent with IHC. More interestingly, EGFL6 accelerated the migration and growth of NPC in vitro assays. Considering the mechanism of migration, NPC cells were cultured with AKT activator, revealing EGFL6 facilitated the progression of NPC via AKT. Moreover, the same effect of EGFL6 in promoting NPC growth was proved in nude mice. Furthermore, heat-shock zebrafish model was established with EGFL6 overexpression. Then, CNE2 cells were injected into the model and cells mass was observed, showing that EGFL6 enhanced the migration and metastasis of NPC. Currently, as the prognosis of NPC is severely affected by distant metastasis, it might be a new therapeutic target toward EGFL6. Taken together, our results suggested that EGFL6 acts as a potential positive regulator in the migration and proliferation of NPC.
Collapse
Affiliation(s)
- Ziyu Zhu
- Department of Otorhinolaryngology Head and Neck surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Haosheng Ni
- Department of Otorhinolaryngology Head and Neck surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Bo You
- Department of Otorhinolaryngology Head and Neck surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Si Shi
- Department of Otorhinolaryngology Head and Neck surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Ying Shan
- Department of Otorhinolaryngology Head and Neck surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Lili Bao
- Department of Otorhinolaryngology Head and Neck surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Bingyue Duan
- Department of Otorhinolaryngology Head and Neck surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Yiwen You
- Department of Otorhinolaryngology Head and Neck surgery, Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|