1
|
Heurtier V, de Nonneville A. [Capivasertib in combination with fulvestrant in locally advanced or metastatic RH+ HER2- breast cancer with PIK3CA, AKT1 or PTEN alteration, after first-line hormonal therapy]. Bull Cancer 2024; 111:997-998. [PMID: 39317592 DOI: 10.1016/j.bulcan.2024.08.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Accepted: 08/10/2024] [Indexed: 09/26/2024]
Affiliation(s)
- Victor Heurtier
- Unité de génétique clinique, institut Curie, Sorbonne université, Paris, France.
| | | |
Collapse
|
2
|
Fu R, Chen Y, Zhao J, Xie X. The signature of SARS-CoV-2-related genes predicts the immune therapeutic response and prognosis in breast cancer. BMC Med Genomics 2024; 17:260. [PMID: 39482662 PMCID: PMC11526603 DOI: 10.1186/s12920-024-02032-0] [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: 01/30/2024] [Accepted: 10/15/2024] [Indexed: 11/03/2024] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is an exceptionally contagious single-stranded RNA virus with a strong positive contagion. The COVID-19 pandemic refers to the swift worldwide dissemination of SARS-CoV-2 infection, which began in late 2019. The COVID-19 epidemic has disrupted many cancer treatments. A few reports indicate that the prevalence of SARS-CoV-2 has disrupted the treatment of breast cancer patients (BCs). However, the role of SARS-CoV-2 in the occurrence and prognosis of BC has not been elucidated. Here, we applied bioinformatics to construct a prognostic signature of SARS-CoV-2-related genes (SCRGs). Specifically, weighted gene co-expression network analysis (WGCNA) was utilized to extract co-expressed genes of differentially expressed genes (DEGs) in breast cancer and SCRGs. Then, we utilized the least absolute shrinkage and selection operator (LASSO) algorithm and univariate regression analysis to screen out three hub genes (DCTPP1, CLIP4 and ANO6) and constructed a risk score model. We further analyzed tumor immune invasion, HLA-related genes, immune checkpoint inhibitors (ICIs), and sensitivity to anticancer drugs in different SARS-CoV-2 related risk subgroups. In addition, we have developed a nomination map to expand clinical applicability. The results of our study indicate that BCs with a high-risk score are linked to negative outcomes, lower immune scores, and reduced responsiveness to anticancer medications. This suggests that the SARS-CoV-2 related signature could be used to guide prognosis assessment and treatment decisions for BCs.
Collapse
Affiliation(s)
- Ruizhi Fu
- Department of General Surgery, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515000, China
| | - Yequn Chen
- Department of Cardiology, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515000, China
| | - Jiajing Zhao
- Department of General Surgery, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515000, China.
| | - Xiaojun Xie
- Department of General Surgery, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515000, China.
| |
Collapse
|
3
|
Zhu S, Yu D, Wang X, Wang X. Predict the Drug-Drug Interaction of a Novel PI3Kα/δ Inhibitor, TQ-B3525, and Its Two Metabolites Using Physiologically Based Pharmacokinetic Modeling. J Clin Pharmacol 2024. [PMID: 39105511 DOI: 10.1002/jcph.6111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 07/22/2024] [Indexed: 08/07/2024]
Abstract
A novel dual PI3K α/δ inhibitor, TQ-B3525, has been developed for the targeted treatment of lymphoma and solid tumors. TQ-B3525 is primarily metabolized by CYP3A4 and FOM3, while also serving as a substrate for the P-glycoprotein transporter. The aim of this study was to anticipate the drug-drug interaction (DDI) of TQ-B3525 and its two metabolites with CYP3A4 enzyme potent inducer (rifampicin) and CYP3A4/P-gp inhibitor (itraconazole) utilizing a physiologically based pharmacokinetic (PBPK) modeling approach. Clinical data from healthy and cancer patient adults were employed to construct and evaluate the PBPK model for TQ-B3525, M3, and M8-3. Models involving rifampicin combined with midazolam, itraconazole combined with midazolam or digoxin were utilized to showcase the robustness of evaluating DDI effects. The simulated drug exposure of TQ-B3525, M3, and M8-3 in healthy and patient adults were consistent with clinical data, and the mean fold error values were within the acceptable ranges. The simulated results of positive substrates correspond to those reported in the literature. Co-administration with rifampicin reduces Cmax and AUC of TQ-B3525 to 76.1% and 46.0%, while increasing the levels of M3 and M8-3. With itraconazole, Cmax and AUC of TQ-B3525 rise to 131% and 204%, but decrease substantially for M3 and M8-3. PBPK model simulation results showed that the systemic exposure of TQ-B3525 was significantly affected when co-administered with CYP3A4/P-gp inducers and inhibitors. This indicates that the combination with strong inducers and inhibitors should be carefully avoided or adjust the dosage of TQ-B3525 in clinic.
Collapse
Affiliation(s)
- Shixing Zhu
- Clinical Medicine Department, Chia Tai Tianqing Pharmaceutical Group Co., Ltd., Nanjing, China
| | - Ding Yu
- Clinical Medicine Department, Chia Tai Tianqing Pharmaceutical Group Co., Ltd., Nanjing, China
| | - Xunqiang Wang
- Clinical Medicine Department, Chia Tai Tianqing Pharmaceutical Group Co., Ltd., Nanjing, China
| | - Xin Wang
- Clinical Medicine Department, Chia Tai Tianqing Pharmaceutical Group Co., Ltd., Nanjing, China
| |
Collapse
|
4
|
Chen Y, Wang X, Na X, Zhang Y, Cai L, Song J, Yang C. DMF-DM-seq: Digital-Microfluidics Enabled Dual-Modality Sequencing of Single-Cell mRNA and microRNA with High Integration, Sensitivity, and Automation. Anal Chem 2024; 96:12916-12926. [PMID: 39038243 DOI: 10.1021/acs.analchem.4c03378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/24/2024]
Abstract
Multimodal measurement of single cells provides deep insights into the intricate relationships between individual molecular layers and the regulatory mechanisms underlying intercellular variations. Here, we reported DMF-DM-seq, a highly integrated, sensitive, and automated platform for single-cell mRNA and microRNA (miRNA) co-sequencing based on digital microfluidics. This platform first integrates the processes of single-cell isolation, lysis, component separation, and simultaneous sequencing library preparation of mRNA and miRNA within a single DMF device. Compared with the current half-cell measuring strategy, DMF-DM-seq enables complete separation of single-cell mRNA and miRNA via a magnetic field application, resulting in a higher miRNA detection ability. DMF-DM-seq revealed differential expression patterns of single cells of noncancerous breast cells and noninvasive and aggressive breast cancer cells at both mRNA and miRNA levels. The results demonstrated the anticorrelated relationship between miRNA and their mRNA targets. Further, we unravel the tumor growth and metastasis-associated biological processes enriched by miRNA-targeted genes, along with important miRNA-interaction networks involved in significant signaling pathways. We also deconstruct the miRNA regulatory mechanisms underlying different signaling pathways across different breast cell types. In summary, DMF-DM-seq offers a powerful tool for a comprehensive study of the expression heterogeneity of single-cell mRNA and miRNA, which will be widely applied in basic and clinical research.
Collapse
Affiliation(s)
- Yingwen Chen
- State Key Laboratory for Physical Chemistry of Solid Surfaces, Key Laboratory for Chemical Biology of Fujian Province, Key Laboratory of Analytical Chemistry, and Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Xuanqun Wang
- State Key Laboratory for Physical Chemistry of Solid Surfaces, Key Laboratory for Chemical Biology of Fujian Province, Key Laboratory of Analytical Chemistry, and Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Xing Na
- State Key Laboratory for Physical Chemistry of Solid Surfaces, Key Laboratory for Chemical Biology of Fujian Province, Key Laboratory of Analytical Chemistry, and Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Yingkun Zhang
- State Key Laboratory for Physical Chemistry of Solid Surfaces, Key Laboratory for Chemical Biology of Fujian Province, Key Laboratory of Analytical Chemistry, and Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Linfeng Cai
- State Key Laboratory for Physical Chemistry of Solid Surfaces, Key Laboratory for Chemical Biology of Fujian Province, Key Laboratory of Analytical Chemistry, and Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Jia Song
- Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Chaoyong Yang
- State Key Laboratory for Physical Chemistry of Solid Surfaces, Key Laboratory for Chemical Biology of Fujian Province, Key Laboratory of Analytical Chemistry, and Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, No. 55, Zhenhai Road, Siming District, Xiamen 361003, China
| |
Collapse
|
5
|
Pesini C, Artal L, Paúl Bernal J, Sánchez Martinez D, Pardo J, Ramírez-Labrada A. In-depth analysis of the interplay between oncogenic mutations and NK cell-mediated cancer surveillance in solid tumors. Oncoimmunology 2024; 13:2379062. [PMID: 39036370 PMCID: PMC11259085 DOI: 10.1080/2162402x.2024.2379062] [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: 05/06/2024] [Accepted: 07/08/2024] [Indexed: 07/23/2024] Open
Abstract
Natural killer (NK) cells play a crucial role in antitumoral and antiviral responses. Yet, cancer cells can alter themselves or the microenvironment through the secretion of cytokines or other factors, hindering NK cell activation and promoting a less cytotoxic phenotype. These resistance mechanisms, often referred to as the "hallmarks of cancer" are significantly influenced by the activation of oncogenes, impacting most, if not all, of the described hallmarks. Along with oncogenes, other types of genes, the tumor suppressor genes are frequently mutated or modified during cancer. Traditionally, these genes have been associated with uncontrollable tumor growth and apoptosis resistance. Recent evidence suggests oncogenic mutations extend beyond modulating cell death/proliferation programs, influencing cancer immunosurveillance. While T cells have been more studied, the results obtained highlight NK cells as emerging key protagonists for enhancing tumor cell elimination by modulating oncogenic activity. A few recent studies highlight the crucial role of oncogenic mutations in NK cell-mediated cancer recognition, impacting angiogenesis, stress ligands, and signaling balance within the tumor microenvironment. This review will critically examine recent discoveries correlating oncogenic mutations to NK cell-mediated cancer immunosurveillance, a relatively underexplored area, particularly in the era dominated by immune checkpoint inhibitors and CAR-T cells. Building on these insights, we will explore opportunities to improve NK cell-based immunotherapies, which are increasingly recognized as promising alternatives for treating low-antigenic tumors, offering significant advantages in terms of safety and manufacturing suitability.
Collapse
Affiliation(s)
- Cecilia Pesini
- Aragón Health Research Institute (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), Zaragoza, Spain
- Center for Biomedical Research in the Network of Infectious Diseases (CIBERINFEC), Carlos III Health Institute, Zaragoza, Spain
- Department of Microbiology, Radiology, Pediatry and Public Health, University of Zaragoza, Zaragoza, Spain
| | - Laura Artal
- Aragón Health Research Institute (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), Zaragoza, Spain
- Institute of Carbochemistry (ICB-CSIC), Zaragoza, Spain
| | - Jorge Paúl Bernal
- Aragón Health Research Institute (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), Zaragoza, Spain
| | - Diego Sánchez Martinez
- Aragón Health Research Institute (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), Zaragoza, Spain
- Aragón I + D Foundation (ARAID), Government of Aragon, Zaragoza, Spain
| | - Julián Pardo
- Aragón Health Research Institute (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), Zaragoza, Spain
- Center for Biomedical Research in the Network of Infectious Diseases (CIBERINFEC), Carlos III Health Institute, Zaragoza, Spain
- Department of Microbiology, Radiology, Pediatry and Public Health, University of Zaragoza, Zaragoza, Spain
| | - Ariel Ramírez-Labrada
- Aragón Health Research Institute (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), Zaragoza, Spain
- Center for Biomedical Research in the Network of Infectious Diseases (CIBERINFEC), Carlos III Health Institute, Zaragoza, Spain
| |
Collapse
|
6
|
Chen Y, Zhu H, Luo Y, Tong S, Liu Y. EZH2: The roles in targeted therapy and mechanisms of resistance in breast cancer. Biomed Pharmacother 2024; 175:116624. [PMID: 38670045 DOI: 10.1016/j.biopha.2024.116624] [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: 01/28/2024] [Revised: 04/04/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
Drug resistance presents a formidable challenge in the realm of breast cancer therapy. Accumulating evidence suggests that enhancer of zeste homolog 2 (EZH2), a component of the polycomb repressive complex 2 (PRC2), may serve as a key regulator in controlling drug resistance. EZH2 overexpression has been observed in breast cancer and many other malignancies, showing a strong correlation with poor outcomes. This review aims to summarize the mechanisms by which EZH2 regulates drug resistance, with a specific focus on breast cancer, in order to provide a comprehensive understanding of the underlying molecular processes. Additionally, we will discuss the current strategies and outcomes of targeting EZH2 using both single agents and combination therapies, with the goal of offering improved guidance for the clinical treatment of breast cancer patients who have developed drug resistance.
Collapse
Affiliation(s)
- Yun Chen
- Department of Clinical Pharmacy, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, PR China; Clinical Pharmacy Innovation Institute, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, PR China.
| | - Hongyan Zhu
- Department of Clinical Pharmacy, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, PR China; Clinical Pharmacy Innovation Institute, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, PR China.
| | - Yi Luo
- Clinical Pharmacy Innovation Institute, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, PR China; Biotheus Inc., Guangdong Province, Zhuhai 519080, PR China.
| | - Shuangmei Tong
- Department of Clinical Pharmacy, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, PR China; Clinical Pharmacy Innovation Institute, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, PR China.
| | - Yan Liu
- Department of Clinical Pharmacy, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, PR China; Clinical Pharmacy Innovation Institute, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, PR China.
| |
Collapse
|
7
|
André C, Bertaut A, Ladoire S, Desmoulins I, Jankowski C, Beltjens F, Charon-Barra C, Bergeron A, Richard C, Boidot R, Arnould L. HER2-Low Luminal Breast Carcinoma Is Not a Homogenous Clinicopathological and Molecular Entity. Cancers (Basel) 2024; 16:2009. [PMID: 38893129 PMCID: PMC11171142 DOI: 10.3390/cancers16112009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 05/21/2024] [Accepted: 05/22/2024] [Indexed: 06/21/2024] Open
Abstract
BACKGROUND With the development of some new antibody-drug conjugates, the HER2 classification of breast carcinomas now includes the HER2-low (H2L) category: IHC 1+, 2+ non-amplified by ISH, and double-equivocal carcinomas, mostly luminal, expressing hormone receptors (HR+). METHODS We analyzed mutational status and transcriptomic activities of three HER2 effector pathways: PI3K-AKT, MAPK, and JAK-STAT, in association with clinicopathologic features, in 62 H2L carcinomas compared to 43 HER2-positive and 20 HER2-negative carcinomas, all HR+. RESULTS H2L carcinomas had significantly lower histoprognostic grades and mitotic and Ki67 proliferation indexes than HER2-positive carcinomas. Their PIK3CA mutation rates were close to those of HER2-negative and significantly higher than in HER2-positive carcinomas, contrary to TP53 mutations. At the transcriptomic level, we identified three distinct groups which did not reflect the new HER2 classification. H2L and HER2-negative carcinomas shared most of clinicopathological and molecular characteristics, except HER2 membrane expression (mRNA levels). The presence of a mutation in a signaling pathway had a strong pathway activation effect. PIK3CA mutations were more prevalent in H2L carcinomas, leading to a strong activation of the PI3K-AKT signaling pathway even in the absence of HER2 overexpression/amplification. CONCLUSION PIK3CA mutations may explain the failure of conventional anti-HER2 treatments, suggesting that new antibody-drug conjugates may be more effective.
Collapse
Affiliation(s)
- Céline André
- Unit of Pathology, Department of Tumor Biology and Pathology, Georges-François Leclerc Cancer Center, 21000 Dijon, France; (F.B.); (C.C.-B.); (A.B.); (L.A.)
- Unit of Pathology, University Hospital Center, 21000 Dijon, France
| | - Aurélie Bertaut
- Unit of Methodology and Biostatistics, Georges-François Leclerc Cancer Center, 21000 Dijon, France;
| | - Sylvain Ladoire
- Department of Medical Oncology, Georges-François Leclerc Cancer Center, 21000 Dijon, France; (S.L.); (I.D.)
- Unit 1231 (INSERM U1231), National Institute of Health and Medical Research, 21000 Dijon, France
- Department of Medicine, University of Burgundy Franche-Comté, 21000 Dijon, France
| | - Isabelle Desmoulins
- Department of Medical Oncology, Georges-François Leclerc Cancer Center, 21000 Dijon, France; (S.L.); (I.D.)
| | - Clémentine Jankowski
- Department of Surgery, Georges-François Leclerc Cancer Center, 21000 Dijon, France;
| | - Françoise Beltjens
- Unit of Pathology, Department of Tumor Biology and Pathology, Georges-François Leclerc Cancer Center, 21000 Dijon, France; (F.B.); (C.C.-B.); (A.B.); (L.A.)
| | - Céline Charon-Barra
- Unit of Pathology, Department of Tumor Biology and Pathology, Georges-François Leclerc Cancer Center, 21000 Dijon, France; (F.B.); (C.C.-B.); (A.B.); (L.A.)
| | - Anthony Bergeron
- Unit of Pathology, Department of Tumor Biology and Pathology, Georges-François Leclerc Cancer Center, 21000 Dijon, France; (F.B.); (C.C.-B.); (A.B.); (L.A.)
| | - Corentin Richard
- Unit of Molecular Pathology, Department of Tumor Biology and Pathology, Georges-François Leclerc Cancer Center, 21000 Dijon, France; (C.R.); (R.B.)
| | - Romain Boidot
- Unit of Molecular Pathology, Department of Tumor Biology and Pathology, Georges-François Leclerc Cancer Center, 21000 Dijon, France; (C.R.); (R.B.)
| | - Laurent Arnould
- Unit of Pathology, Department of Tumor Biology and Pathology, Georges-François Leclerc Cancer Center, 21000 Dijon, France; (F.B.); (C.C.-B.); (A.B.); (L.A.)
- Unit 1231 (INSERM U1231), National Institute of Health and Medical Research, 21000 Dijon, France
| |
Collapse
|
8
|
Zhang X, Yang F, Huang Z, Liu X, Xia G, Huang J, Yang Y, Li J, Huang J, Liu Y, Zhou T, Qi W, Gao G, Yang X. Macrophages Promote Subtype Conversion and Endocrine Resistance in Breast Cancer. Cancers (Basel) 2024; 16:678. [PMID: 38339428 PMCID: PMC10854660 DOI: 10.3390/cancers16030678] [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: 12/30/2023] [Revised: 01/25/2024] [Accepted: 01/29/2024] [Indexed: 02/12/2024] Open
Abstract
BACKGROUND The progression of tumors from less aggressive subtypes to more aggressive states during metastasis poses challenges for treatment strategies. Previous studies have revealed the molecular subtype conversion between primary and metastatic tumors in breast cancer (BC). However, the subtype conversion during lymph node metastasis (LNM) and the underlying mechanism remains unclear. METHODS We compared clinical subtypes in paired primary tumors and positive lymph nodes (PLNs) in BC patients and further validated them in the mouse model. Bioinformatics analysis and macrophage-conditioned medium treatment were performed to investigate the role of macrophages in subtype conversion. RESULTS During LNM, hormone receptors (HRs) were down-regulated, while HER2 was up-regulated, leading to the transformation of luminal A tumors towards luminal B tumors and from luminal B subtype towards HER2-enriched (HER2-E) subtype. The mouse model demonstrated the elevated levels of HER2 in PLN while retaining luminal characteristics. Among the various cells in the tumor microenvironment (TME), macrophages were the most clinically relevant in terms of prognosis. The treatment of a macrophage-conditioned medium further confirmed the downregulation of HR expression and upregulation of HER2 expression, inducing tamoxifen resistance. Through bioinformatics analysis, MNX1 was identified as a potential transcription factor governing the expression of HR and HER2. CONCLUSION Our study revealed the HER2-E subtype conversion during LNM in BC. Macrophages were the crucial cell type in TME, inducing the downregulation of HR and upregulation of HER2, probably via MNX1. Targeting macrophages or MNX1 may provide new avenues for endocrine therapy and targeted treatment of BC patients with LNM.
Collapse
Affiliation(s)
- Xiaoyan Zhang
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; (X.Z.); (F.Y.); (Z.H.); (X.L.); (G.X.); (J.H.); (Y.Y.); (J.L.); (J.H.); (Y.L.); (T.Z.); (W.Q.)
| | - Fengyu Yang
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; (X.Z.); (F.Y.); (Z.H.); (X.L.); (G.X.); (J.H.); (Y.Y.); (J.L.); (J.H.); (Y.L.); (T.Z.); (W.Q.)
| | - Zhijian Huang
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; (X.Z.); (F.Y.); (Z.H.); (X.L.); (G.X.); (J.H.); (Y.Y.); (J.L.); (J.H.); (Y.L.); (T.Z.); (W.Q.)
- Department of Pathology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, China
| | - Xiaojun Liu
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; (X.Z.); (F.Y.); (Z.H.); (X.L.); (G.X.); (J.H.); (Y.Y.); (J.L.); (J.H.); (Y.L.); (T.Z.); (W.Q.)
| | - Gan Xia
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; (X.Z.); (F.Y.); (Z.H.); (X.L.); (G.X.); (J.H.); (Y.Y.); (J.L.); (J.H.); (Y.L.); (T.Z.); (W.Q.)
| | - Jieye Huang
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; (X.Z.); (F.Y.); (Z.H.); (X.L.); (G.X.); (J.H.); (Y.Y.); (J.L.); (J.H.); (Y.L.); (T.Z.); (W.Q.)
| | - Yang Yang
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; (X.Z.); (F.Y.); (Z.H.); (X.L.); (G.X.); (J.H.); (Y.Y.); (J.L.); (J.H.); (Y.L.); (T.Z.); (W.Q.)
| | - Junchen Li
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; (X.Z.); (F.Y.); (Z.H.); (X.L.); (G.X.); (J.H.); (Y.Y.); (J.L.); (J.H.); (Y.L.); (T.Z.); (W.Q.)
| | - Jin Huang
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; (X.Z.); (F.Y.); (Z.H.); (X.L.); (G.X.); (J.H.); (Y.Y.); (J.L.); (J.H.); (Y.L.); (T.Z.); (W.Q.)
| | - Yuxin Liu
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; (X.Z.); (F.Y.); (Z.H.); (X.L.); (G.X.); (J.H.); (Y.Y.); (J.L.); (J.H.); (Y.L.); (T.Z.); (W.Q.)
| | - Ti Zhou
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; (X.Z.); (F.Y.); (Z.H.); (X.L.); (G.X.); (J.H.); (Y.Y.); (J.L.); (J.H.); (Y.L.); (T.Z.); (W.Q.)
| | - Weiwei Qi
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; (X.Z.); (F.Y.); (Z.H.); (X.L.); (G.X.); (J.H.); (Y.Y.); (J.L.); (J.H.); (Y.L.); (T.Z.); (W.Q.)
| | - Guoquan Gao
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; (X.Z.); (F.Y.); (Z.H.); (X.L.); (G.X.); (J.H.); (Y.Y.); (J.L.); (J.H.); (Y.L.); (T.Z.); (W.Q.)
- Department of Internal Medicine, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou 510700, China
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Xia Yang
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; (X.Z.); (F.Y.); (Z.H.); (X.L.); (G.X.); (J.H.); (Y.Y.); (J.L.); (J.H.); (Y.L.); (T.Z.); (W.Q.)
- Department of Internal Medicine, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou 510700, China
- Guangdong Engineering & Technology Research Center for Gene Manipulation and Biomacromolecular Products, Sun Yat-sen University, Guangzhou 510080, China
| |
Collapse
|
9
|
Zhou YF, Li JT, Zheng QL, Ren KL, Yi CC. METTL3-mediated m 6 A methylation of TRAF5 inhibits lung adenocarcinoma cell metastasis via activation of the PI3K/AKT/NF-κB signaling pathway. Kaohsiung J Med Sci 2024; 40:150-160. [PMID: 38088510 DOI: 10.1002/kjm2.12791] [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: 04/10/2023] [Revised: 10/23/2023] [Accepted: 11/07/2023] [Indexed: 02/03/2024] Open
Abstract
Tumor necrosis factor receptor-associated factor 5 (TRAF5) has been implicated in the pathogenesis of human malignancies. This work aimed to clarify the role of TRAF5 in lung adenocarcinoma (LUAD) progression. Herein, we uncovered that TRAF5 level was reduced in LUAD tissues. Low TRAF5 expression correlated with dismal prognosis in LUAD patients. Moreover, upregulated TRAF5 impeded cell viability, migration, and invasion, induced apoptosis in vitro, as well as impaired tumorigenicity in vivo. However, depletion of TRAF5 revealed opposing results. Moreover, TRAF5 was identified as the downstream target of methyltransferase-like 3 (METTL3)-elicited N6 -methyladenosine (m6 A) modification. METTL3 stabilized TRAF5 mRNA and positively modulated TRAF5 level. Further, TRAF5 depletion relieved the repressive phenotype caused by METTL3 addition. In addition, it was manifested that the METTL3/TRAF5 axis served as an inhibitor in LUAD through the PI3K/AKT/Nuclear Factor-Kappa B (NF-κB) signaling. Collectively, we propose that METTL3-mediated TRAF5 m6 A modification exerted as a vital tumor inhibitory function in LUAD development. The METTL3/TRAF5 axis may be a critical effector of LUAD progression.
Collapse
Affiliation(s)
- Yu-Fei Zhou
- Department of Thoracic Surgery, The People's Hospital of Dongying (Dongying Hospital of Shandong Provincial Hospital Group), Dongying, China
| | - Jiang-Tao Li
- Department of Thoracic Surgery, The People's Hospital of Laiyang, Yantai, China
| | - Qing-Lin Zheng
- Department of Thoracic Surgery, The People's Hospital of Dongying (Dongying Hospital of Shandong Provincial Hospital Group), Dongying, China
| | - Kun-Lun Ren
- Department of Thoracic Surgery, The People's Hospital of Dongying (Dongying Hospital of Shandong Provincial Hospital Group), Dongying, China
| | - Cheng-Cheng Yi
- Department of Thoracic Surgery, The People's Hospital of Dongying (Dongying Hospital of Shandong Provincial Hospital Group), Dongying, China
| |
Collapse
|
10
|
Elmi M, Dass JH, Dass CR. The Various Roles of PEDF in Cancer. Cancers (Basel) 2024; 16:510. [PMID: 38339261 PMCID: PMC10854708 DOI: 10.3390/cancers16030510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 01/16/2024] [Accepted: 01/23/2024] [Indexed: 02/12/2024] Open
Abstract
Pigment epithelium-derived factor (PEDF) is a natural immunomodulator, anti-inflammatory, anti-angiogenic, anti-tumour growth and anti-metastasis factor, which can enhance tumour response to PEDF but can also conversely have pro-cancerous effects. Inflammation is a major cause of cancer, and it has been proven that PEDF has anti-inflammatory properties. PEDF's functional activity can be investigated through measuring metastatic and metabolic biomarkers that will be discussed in this review.
Collapse
Affiliation(s)
- Mitra Elmi
- Curtin Medical School, Curtin University, Bentley, WA 6102, Australia; (M.E.); (J.H.D.)
- Curtin Health Innovation Research Institute, Curtin Medical School, Curtin University, Bentley, WA 6102, Australia
| | - Joshua H. Dass
- Curtin Medical School, Curtin University, Bentley, WA 6102, Australia; (M.E.); (J.H.D.)
- Sir Charles Gairdner Hospital, Nedlands, WA 6009, Australia
| | - Crispin R. Dass
- Curtin Medical School, Curtin University, Bentley, WA 6102, Australia; (M.E.); (J.H.D.)
- Curtin Health Innovation Research Institute, Curtin Medical School, Curtin University, Bentley, WA 6102, Australia
| |
Collapse
|
11
|
Sajjadi E, Frascarelli C, Venetis K, Bonizzi G, Ivanova M, Vago G, Guerini-Rocco E, Fusco N. Computational pathology to improve biomarker testing in breast cancer: how close are we? Eur J Cancer Prev 2023; 32:460-467. [PMID: 37038997 DOI: 10.1097/cej.0000000000000804] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2023]
Abstract
The recent advancements in breast cancer precision medicine have highlighted the urgency for the precise and reproducible characterization of clinically actionable biomarkers. Despite numerous standardization efforts, biomarker testing by conventional methodologies is challenged by several issues such as high inter-observer variabilities, the spatial heterogeneity of biomarkers expression, and technological heterogeneity. In this respect, artificial intelligence-based digital pathology approaches are being increasingly recognized as promising methods for biomarker testing and subsequently improved clinical management. Here, we provide an overview on the most recent advances for artificial intelligence-assisted biomarkers testing in breast cancer, with a particular focus on tumor-infiltrating lymphocytes, programmed death-ligand 1, phosphatidylinositol-3 kinase catalytic alpha, and estrogen receptor 1. Challenges and solutions for this integrative analysis in pathology laboratories are also provided.
Collapse
Affiliation(s)
- Elham Sajjadi
- Department of Oncology and Hemato-Oncology, University of Milan
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Chiara Frascarelli
- Department of Oncology and Hemato-Oncology, University of Milan
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | | | - Giuseppina Bonizzi
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Mariia Ivanova
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Gianluca Vago
- Department of Oncology and Hemato-Oncology, University of Milan
| | - Elena Guerini-Rocco
- Department of Oncology and Hemato-Oncology, University of Milan
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Nicola Fusco
- Department of Oncology and Hemato-Oncology, University of Milan
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| |
Collapse
|
12
|
Baljon KJ, Ramaiah P, Saleh EAM, Al-Dolaimy F, Al-Dami FH, Gandla K, Alkhafaji AT, Abbas AHR, Alsaalamy AH, Bisht YS. LncRNA PVT1: as a therapeutic target for breast cancer. Pathol Res Pract 2023; 248:154675. [PMID: 37531833 DOI: 10.1016/j.prp.2023.154675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 07/03/2023] [Accepted: 07/04/2023] [Indexed: 08/04/2023]
Abstract
A significant number of women are identified with breast cancer (BC) every year, making it among the most prevalent malignancies and one of the leading causes of mortality globally. Despite significant progress in understanding BC pathogenesis and treatment options, there is still a need to identify new therapeutic targets and develop more effective treatments. LncRNAs have been discovered as biomarkers and a promising target for various cancers, including BC. PVT1 is a particular one of these lncRNAs, and research has indicated that it has a significant impact on the appearance and progression of BC.PVT1 is an attractive therapeutic target for BC due to its role in promoting cancer cell growth, metastasis and invasion. In addition to its potential as a treatment strategy, PVT1 may also have diagnostic value in BC. In this article, we will discuss targeting PVT1 as a treatment strategy for BC.
Collapse
Affiliation(s)
| | | | - Ebraheem Abdu Musad Saleh
- Department of Chemistry,College of Arts and Science, Prince Sattam Bin Abdulaziz University, Wadi Al-Dawasir 11991, Saudi Arabia.
| | | | - Farqad Hassan Al-Dami
- Department of Medical Laboratory Techniques, Altoosi University College, Najaf, Iraq
| | - Kumaraswamy Gandla
- Department of Pharmaceutical Analysis, Chaitanya Deemed to be University, Hanamkonda, India.
| | | | - Ahmed Hussien R Abbas
- College of technical engineering, the Islamic University, Najaf, Iraq; College of technical engineering, the Islamic University of Al Diwaniyah, Iraq; College of technical engineering, the Islamic University of Babylon, Iraq
| | - Ali Hashiem Alsaalamy
- College of technical engineering, Imam Ja'afar Al-Sadiq University, Al-Muthanna 66002, Iraq
| | - Yashwant Singh Bisht
- Uttaranchal Institute of Technology, Uttaranchal University, Dehradun 248007, India
| |
Collapse
|
13
|
Patellongi I, Amiruddin A, Massi MN, Islam AA, Pratama MY, Sutandyo N, Latar NH, Faruk M. Circulating miR-221/222 expression as microRNA biomarker predicting tamoxifen treatment outcome: a case-control study. Ann Med Surg (Lond) 2023; 85:3806-3815. [PMID: 37554919 PMCID: PMC10406100 DOI: 10.1097/ms9.0000000000001061] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 07/02/2023] [Indexed: 08/10/2023] Open
Abstract
The high mortality rate in breast cancer (BC) patients is generally due to metastases resistant to systemic therapy. Two causes of systemic therapy resistance in BC patients are circulating miRNAs-221 and miR-222, leading to improved BC cell proliferation, survival, and reduced cell apoptosis. This study investigated the miRNA expression changes associated with cancer cell resistance to tamoxifen therapy and is expected to be clinically meaningful before providing endocrine therapy to luminal-type BC patients who express them. Methods This case-control research included individuals with the luminal subtype of BC who had received tamoxifen medication for around one year. Furthermore, the case group contained 15 individuals with local recurrence or metastases, while the control group comprised 19 patients without local recurrence or metastases. Plasma miR-221/222 quantification was performed with real-time PCR using transcript-specific primers. Results A significant difference was found in circulating miR-221 expression between cases and controls (P=0.005) but not in miR-222 expression (P=0.070). There were no significant differences between miR-221/222 expression, progesterone receptor, Ki67 protein levels, lymphovascular invasion, and stage. However, receiver operator characteristic curve analyses showed miR-221/222 expressions predictive of tamoxifen resistance (P=0.030) with a sensitivity of 60.00 and a specificity of 83.33%. Conclusion The use of circulating miR-221/222 expression can predict relapse as well as resistance to tamoxifen treatment in BC patients, and their testing is recommended for luminal subtype BC patients who will undergo tamoxifen therapy to determine their risk of tamoxifen resistance early, increasing treatment effectiveness.
Collapse
Affiliation(s)
| | | | | | | | | | - Noorwati Sutandyo
- Department of Medical Hematology-Oncology, Dharmais Hospital National Cancer Center, Jakarta, Indonesia
| | - Nani H.M. Latar
- Endocrine and Breast Surgery Unit, Department of Surgery, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Center, Kuala Lumpur, Malaysia
| | - Muhammad Faruk
- Department of Surgery, Faculty of Medicine, Universitas Hasanuddin, Makassar
| |
Collapse
|
14
|
Mlinarić M, Lučić I, Milković L, da Silva IV, Tartaro Bujak I, Musani V, Soveral G, Čipak Gašparović A. AQP3-Dependent PI3K/Akt Modulation in Breast Cancer Cells. Int J Mol Sci 2023; 24:ijms24098133. [PMID: 37175840 PMCID: PMC10179317 DOI: 10.3390/ijms24098133] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 04/27/2023] [Accepted: 04/28/2023] [Indexed: 05/15/2023] Open
Abstract
Aquaporin 3 (AQP3) is a peroxiporin, a membrane protein that channels hydrogen peroxide in addition to water and glycerol. AQP3 expression also correlates with tumor progression and malignancy and is, therefore, a potential target in breast cancer therapy. In addition, epithelial growth factor receptor (EGFR) plays an important role in breast cancer. Therefore, we investigated whether disruption of the lipid raft harboring EGFR could affect AQP3 expression, and conversely, whether AQP3 silencing would affect the EGFR/phosphoinositide-3-kinase (PI3K)/Protein kinase B (PKB or Akt) signaling pathway in breast cancer cell lines with different malignant capacities. We evaluated H2O2 uptake, cell migratory capacity, and expression of PI3K, pAkt/Akt in three breast cancer cell lines, MCF7, SkBr3, and SUM159PT, and in the nontumorigenic breast epithelial cell line MCF10A. Our results show different responses between the tested cell lines, especially when compared to the nontumorigenic cell line. Neither lipid raft disruption nor EGF stimuli had an effect on PI3K/Akt pathway in MCF10A cell line. AQP3-silencing in SkBr3 and SUM159PT showed that AQP3 can modulate PI3K/Akt activation in these cells. Interestingly, SUM159PT cells increase nuclear factor-E2-related factor 2 (NRF2) in response to lipid raft disruption and EGF stimuli, suggesting an oxidative-dependent response to these treatments. These results suggest that in breast cancer cell lines, AQP3 is not directly related to PI3K/Akt pathway but rather in a cell-line-dependent manner.
Collapse
Affiliation(s)
- Monika Mlinarić
- Division of Molecular Medicine, Ruđer Bošković Institute, 10000 Zagreb, Croatia
| | - Ivan Lučić
- Division of Molecular Medicine, Ruđer Bošković Institute, 10000 Zagreb, Croatia
| | - Lidija Milković
- Division of Molecular Medicine, Ruđer Bošković Institute, 10000 Zagreb, Croatia
| | - Inês V da Silva
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal
| | - Ivana Tartaro Bujak
- Division of Molecular Medicine, Ruđer Bošković Institute, 10000 Zagreb, Croatia
| | - Vesna Musani
- Division of Molecular Medicine, Ruđer Bošković Institute, 10000 Zagreb, Croatia
| | - Graça Soveral
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal
| | | |
Collapse
|
15
|
Hayama S, Nakamura R, Ishige T, Sangai T, Sakakibara M, Fujimoto H, Ishigami E, Masuda T, Nakagawa A, Teranaka R, Ota S, Itoga S, Yamamoto N, Nagashima T, Otsuka M. The impact of PIK3CA mutations and PTEN expression on the effect of neoadjuvant therapy for postmenopausal luminal breast cancer patients. BMC Cancer 2023; 23:384. [PMID: 37106324 PMCID: PMC10134571 DOI: 10.1186/s12885-023-10853-y] [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: 10/27/2022] [Accepted: 04/16/2023] [Indexed: 04/29/2023] Open
Abstract
BACKGROUND There is pressing needs to find the biomarker in the selection of neoadjuvant therapy in postmenopausal luminal breast cancer patients. We examined the hypothesis that PIK3CA mutations and low phosphatase and tensin homolog (PTEN) expression affect the response to neoadjuvant therapy and prognosis in postmenopausal luminal breast cancer patients. METHODS Postmenopausal patients with estrogen receptor-positive, human epidermal growth factor receptor 2-negative breast cancer, up to stage II, who underwent neoadjuvant chemotherapy (NAC; n = 60) or neoadjuvant endocrine therapy (NAE; n = 55) were selected. PIK3CA exon 9 and exon 20 mutations were screened by high resolution melting analysis and confirmed by Sanger sequence. PTEN expression was evaluated by immunohistochemistry. The relationships among PIK3CA mutations, PTEN expression, clinicopathological features, the pathological effect of neoadjuvant therapy, recurrence-free survival (RFS) and overall survival were analyzed. RESULTS Among 115 patients, PIK3CA mutations and low PTEN expression before treatment were detected in 35 patients (30.4%) and in 28 patients (24.3%), respectively. In the NAC group, tumor with PIK3CA mutations showed significantly poorer response than tumor with PIK3CA wild-type (p = 0.03). On the other hand, in the NAE group, there was no significant difference in pathological therapeutic effect between tumor with PIK3CA mutations and tumor with PIK3CA wild-type (p = 0.54). In the NAC group, the log-rank test showed no difference in RFS between patients with PIK3CA mutations and PIK3CA wild-type (p = 0.43), but patients with low PTEN expression showed significantly worse RFS compared to patients with high PTEN expression (5 year RFS 0.64 vs. 0.87, p = 0.01). In the Cox proportional hazards model for RFS, PTEN expression, progesterone receptor, and pathological therapeutic effect were predictive factors for time to recurrence (All p < 0.05). CONCLUSIONS PIK3CA mutations are associated with resistance to NAC but do not affect the response to NAE. Low PTEN expression does not affect response to either NAC or NAE but correlates with shorter RFS in patients who received NAC. These biomarkers will be further evaluated for clinical use to treat postmenopausal luminal breast cancer patients.
Collapse
Affiliation(s)
- Shouko Hayama
- Department of General Surgery, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-Ku, Chiba-Shi, Chiba, 260-8677, Japan
- Department of Breast Surgery, Chiba Cancer Center, 666-2 Nitona, Chuo-Ku, Chiba-Shi, Chiba, 260-8717, Japan
| | - Rikiya Nakamura
- Department of Breast Surgery, Chiba Cancer Center, 666-2 Nitona, Chuo-Ku, Chiba-Shi, Chiba, 260-8717, Japan
| | - Takayuki Ishige
- Department of Molecular Diagnosis, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-Ku, Chiba-Shi, Chiba, 260-8677, Japan
| | - Takafumi Sangai
- Department of Breast and Thyroid Surgery, Kitasato University School of Medicine, 1-15-1 Kitazato Minami-Ku, Sagamihara, Kanagawa, 252-0374, Japan.
| | - Masahiro Sakakibara
- Departments of Breast Surgery, Toho University Sakura Medical Center, 564-1 Shimoshizu, Sakura, Chiba, 285-8741, Japan
| | - Hiroshi Fujimoto
- Department of General Surgery, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-Ku, Chiba-Shi, Chiba, 260-8677, Japan
| | - Emi Ishigami
- Department of General Surgery, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-Ku, Chiba-Shi, Chiba, 260-8677, Japan
| | - Takahito Masuda
- Department of General Surgery, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-Ku, Chiba-Shi, Chiba, 260-8677, Japan
| | - Ayako Nakagawa
- Department of General Surgery, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-Ku, Chiba-Shi, Chiba, 260-8677, Japan
| | - Ryotaro Teranaka
- Department of General Surgery, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-Ku, Chiba-Shi, Chiba, 260-8677, Japan
| | - Satoshi Ota
- Department of Pathology, Chiba University Hospital, 1-8-1 Inohana, Chuo-Ku, Chiba-Shi, Chiba, 260-8677, Japan
| | - Sakae Itoga
- Department of Applied Genomics, Kazusa DNA Research Institute, 2-6-7 Kazusa-Kamatari, Kisarazu Chiba, 292-0818, Japan
| | - Naohito Yamamoto
- Department of Breast Surgery, Chiba Cancer Center, 666-2 Nitona, Chuo-Ku, Chiba-Shi, Chiba, 260-8717, Japan
| | - Takeshi Nagashima
- Department of General Surgery, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-Ku, Chiba-Shi, Chiba, 260-8677, Japan
| | - Masayuki Otsuka
- Department of General Surgery, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-Ku, Chiba-Shi, Chiba, 260-8677, Japan
| |
Collapse
|
16
|
Mishra AP, Singh P, Yadav S, Nigam M, Seidel V, Rodrigues CF. Role of the Dietary Phytochemical Curcumin in Targeting Cancer Cell Signalling Pathways. PLANTS (BASEL, SWITZERLAND) 2023; 12:plants12091782. [PMID: 37176840 PMCID: PMC10180989 DOI: 10.3390/plants12091782] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 04/19/2023] [Accepted: 04/22/2023] [Indexed: 05/15/2023]
Abstract
The diarylheptanoid curcumin [(1E,6E)-1,7-bis(4-hydroxy-3-methoxyphenyl)hepta-1,6-diene-3,5-dione] is one of the phenolic pigments responsible for the yellow colour of turmeric (Curcuma longa L.). This phytochemical has gained much attention in recent years due to its therapeutic potential in cancer. A range of drug delivery approaches have been developed to optimise the pharmacokinetic profile of curcumin and ensure that it reaches its target sites. Curcumin exhibits numerous biological effects, including anti-inflammatory, cardioprotective, antidiabetic, and anti-aging activities. It has also been extensively studied for its role as a cancer chemopreventive and anticancer agent. This review focusses on the role of curcumin in targeting the cell signalling pathways involved in cancer, particularly via modulation of growth factors, transcription factors, kinases and other enzymes, pro-inflammatory cytokines, and pro-apoptotic and anti-apoptotic proteins. It is hoped that this study will help future work on the potential of curcumin to fight cancer.
Collapse
Affiliation(s)
- Abhay Prakash Mishra
- Department of Pharmacology, Faculty of Health Science, University of Free State, Bloemfontein 9300, South Africa
| | - Pratichi Singh
- Department of Biosciences, School of Basic and Applied Sciences, Galgotias University, Greater Noida 203201, Uttar Pradesh, India
| | - Shikha Yadav
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida 203201, Uttar Pradesh, India
| | - Manisha Nigam
- Department of Biochemistry, H. N. B. Garhwal University, Srinagar Garhwal 246174, Uttarakhand, India
| | - Veronique Seidel
- Natural Products Research Laboratory, Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | - Celia Fortuna Rodrigues
- LEPABE-Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- ALiCE-Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- TOXRUN-Toxicology Research Unit, Cooperativa de Ensino Superior Politécnico e Universitário-CESPU, 4585-116 Gandra PRD, Portugal
| |
Collapse
|
17
|
Obaidat D, Giordo R, Kleinbrink EL, Banisad E, Grossman LI, Arshad R, Stark A, Maroun MC, Lipovich L, Fernandez-Madrid F. Non-coding regions of nuclear-DNA-encoded mitochondrial genes and intergenic sequences are targeted by autoantibodies in breast cancer. Front Genet 2023; 13:970619. [PMID: 37082114 PMCID: PMC10111166 DOI: 10.3389/fgene.2022.970619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 11/15/2022] [Indexed: 03/31/2023] Open
Abstract
Autoantibodies against mitochondrial-derived antigens play a key role in chronic tissue inflammation in autoimmune disorders and cancers. Here, we identify autoreactive nuclear genomic DNA (nDNA)-encoded mitochondrial gene products (GAPDH, PKM2, GSTP1, SPATA5, MFF, TSPOAP1, PHB2, COA4, and HAGH) recognized by breast cancer (BC) patients’ sera as nonself, supporting a direct relationship of mitochondrial autoimmunity to breast carcinogenesis. Autoreactivity of multiple nDNA-encoded mitochondrial gene products was mapped to protein-coding regions, 3’ untranslated regions (UTRs), as well as introns. In addition, autoantibodies in BC sera targeted intergenic sequences that may be parts of long non-coding RNA (lncRNA) genes, including LINC02381 and other putative lncRNA neighbors of the protein-coding genes ERCC4, CXCL13, SOX3, PCDH1, EDDM3B, and GRB2. Increasing evidence indicates that lncRNAs play a key role in carcinogenesis. Consistent with this, our findings suggest that lncRNAs, as well as mRNAs of nDNA-encoded mitochondrial genes, mechanistically contribute to BC progression. This work supports a new paradigm of breast carcinogenesis based on a globally dysfunctional genome with altered function of multiple mitochondrial and non-mitochondrial oncogenic pathways caused by the effects of autoreactivity-induced dysregulation of multiple genes and their products. This autoimmunity-based model of carcinogenesis will open novel avenues for BC treatment.
Collapse
Affiliation(s)
- Deya Obaidat
- Department of Internal Medicine, Division of Rheumatology, Wayne State University School of Medicine, Detroit, MI, United States
| | - Roberta Giordo
- Department of Basic Medical Sciences, College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
| | - Erica L. Kleinbrink
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, United States
- Quantitative Life Sciences, McGill University, Montreal, QC, Canada
| | - Emilia Banisad
- Department of Internal Medicine, Division of Rheumatology, Wayne State University School of Medicine, Detroit, MI, United States
| | - Lawrence I. Grossman
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, United States
| | - Rooshan Arshad
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, United States
| | - Azadeh Stark
- Department of Pathology, Henry Ford Health System, Detroit, MI, United States
| | - Marie-Claire Maroun
- Department of Internal Medicine, Division of Rheumatology, Wayne State University School of Medicine, Detroit, MI, United States
| | - Leonard Lipovich
- Department of Basic Medical Sciences, College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
- Shenzhen Huayuan Biotechnology Co. Ltd, Shenzhen Huayuan Biological Science Research Institute, Shenzhen, Guangdong, China
- *Correspondence: Leonard Lipovich, ; Félix Fernandez-Madrid,
| | - Félix Fernandez-Madrid
- Department of Internal Medicine, Division of Rheumatology, Wayne State University School of Medicine, Detroit, MI, United States
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, United States
- Karmanos Cancer Institute, Wayne State University, Detroit, MI, United States
- *Correspondence: Leonard Lipovich, ; Félix Fernandez-Madrid,
| |
Collapse
|
18
|
Bredin HK, Krakstad C, Hoivik EA. PIK3CA mutations and their impact on survival outcomes of patients with endometrial cancer: A systematic review and meta-analysis. PLoS One 2023; 18:e0283203. [PMID: 36943861 PMCID: PMC10030019 DOI: 10.1371/journal.pone.0283203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 03/03/2023] [Indexed: 03/23/2023] Open
Abstract
Several studies have highlighted the frequent alterations of the PI3K pathway in endometrial cancer leading to increased signaling activation with potential for targeted treatment. The objective of this meta-study was to evaluate how PIK3CA exon 9/20 mutations affect survival in endometrial cancer patients, based on available literature. Topic-based search strategies were applied to databases including CENTRAL, MEDLINE, Embase, Web of Science and COSMIC. All studies assessing the impact of mutations in exon 9 and exon 20 of PIK3CA on survival rates of endometrial cancer patients were selected for inclusion. Statistical meta-analysis was performed with the 'meta' package in RStudio. Overall, 7 of 612 screened articles were included in the present study, comprising 1098 women with endometrial cancer. Meta-analysis revealed a tendency of impaired survival for patients with PIK3CA exon 9 and/or exon 20 mutations (RR 1.28; 95% CI 0.84, 1.94; p = 0.25). This tendency was consistent in subgroup analyses stratified by histologic type or -grade, with the most prominent effect in low-grade endometrial cancers (RR 2.04; 95% CI 0.90, 4.62; p = 0.09). In summary, these results suggest that PIK3CA mutations negatively influence survival outcomes of patients with endometrial cancer, including those with low-grade tumors.
Collapse
Affiliation(s)
- Hanna K. Bredin
- Department of Clinical Science, Centre for Cancer Biomarkers, University of Bergen, Bergen, Norway
- Department of Obstetrics and Gynecology, Haukeland University Hospital, Bergen, Norway
| | - Camilla Krakstad
- Department of Clinical Science, Centre for Cancer Biomarkers, University of Bergen, Bergen, Norway
- Department of Obstetrics and Gynecology, Haukeland University Hospital, Bergen, Norway
| | - Erling A. Hoivik
- Department of Clinical Science, Centre for Cancer Biomarkers, University of Bergen, Bergen, Norway
- Department of Obstetrics and Gynecology, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
19
|
R HC, Kumar S U, R G, Naayanan PJ, Sathiyarajeswaren P, Devi MSS, K SS, Doss C GP. An integrated investigation of structural and pathway alteration caused by PIK3CA and TP53 mutations identified in cfDNA of metastatic breast cancer. J Cell Biochem 2023; 124:188-204. [PMID: 36563059 DOI: 10.1002/jcb.30354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/12/2022] [Accepted: 11/24/2022] [Indexed: 12/24/2022]
Abstract
In peripheral blood, cell-free DNA (cfDNA) contains circulating tumor DNA (ctDNA), which indicates molecular abnormalities in metastatic breast tumor tissue. The sequencing of cfDNA of Metastatic Breast Cancer (MBC) patients allows assessment of therapy response and noninvasive treatment. In the proposed study, clinically significant alterations in PIK3CA and TP53 genes associated with MBC resulting in a missense substitution of His1047Arg and Arg282Trp from an next-generation sequencing-based multi-gene panel were reported in a cfDNA of a patient with MBC. To investigate the impact of the reported mutation, we used molecular docking, molecular dynamics simulation, network analysis, and pathway analysis. Molecular Docking analysis determined the distinct binding pattern revealing H1047R-ATP complex has a higher number of Hydrogen bonds (H-bonds) and binding affinity with a slight difference compared to the PIK3CA-ATP complex. Following, molecular dynamics simulation for 200 ns, of which H1047R-ATP complex resulted in the instability of PIK3CA. Similarly, for TP53 mutant R282W, the zinc-free state (apo) and zinc-bounded (holo) complexes were investigated for conformational change between apo and holo complexes, of which the holo complex mutant R282W was unstable. To validate the conformational change of PIK3CA and TP53, 80% mutation of H1047R in the kinase domain of p110α expressed ubiquitously in PIK3CA protein that alters PI3K pathway, while R282W mutation in DNA binding helix (H2) region of P53 protein inhibits the transcription factor in P53 pathway causing MBC. According to our findings, the extrinsic (hypoxia, oxidative stress, and acidosis); intrinsic factors (MYC amplification) in PIK3CA and TP53 mutations will provide potential insights for developing novel therapeutic methods for MBC therapy.
Collapse
Affiliation(s)
- Hephzibah Cathryn R
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - Udhaya Kumar S
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - Gnanasambandan R
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | | | | | - M S Shree Devi
- Siddha Central Research Institute (CCRS), Chennai, Tamil Nadu, India
| | - Satish Srinivas K
- Department of Radiation Oncology, Sri Ramachandra Institute of Higher Education and Research, Porur, Chennai, India
| | - George Priya Doss C
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| |
Collapse
|
20
|
Cerma K, Piacentini F, Moscetti L, Barbolini M, Canino F, Tornincasa A, Caggia F, Cerri S, Molinaro A, Dominici M, Omarini C. Targeting PI3K/AKT/mTOR Pathway in Breast Cancer: From Biology to Clinical Challenges. Biomedicines 2023; 11:biomedicines11010109. [PMID: 36672617 PMCID: PMC9855880 DOI: 10.3390/biomedicines11010109] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 12/16/2022] [Accepted: 12/21/2022] [Indexed: 01/04/2023] Open
Abstract
Breast cancer (BC) is the most common women cancer and cause of cancer death. Despite decades of scientific progress in BC treatments, the clinical benefit of new drugs is modest in several cases. The phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) pathway mutations are frequent in BC (20-40%) and are significant causes of aggressive tumor behavior, as well as treatment resistance. Improving knowledge of the PI3K/AKT/mTOR pathway is an urgent need. This review aims to highlight the central role of PI3K-mTORC1/C2 mutations in the different BC subtypes, in terms of clinical outcomes and treatment efficacy. The broad base of knowledge in tumor biology is a key point for personalized BC therapy in the precision medicine era.
Collapse
Affiliation(s)
- Krisida Cerma
- Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena, 41122 Modena, Italy
| | - Federico Piacentini
- Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena, 41122 Modena, Italy
- Division of Medical Oncology, University Hospital of Modena, 41122 Modena, Italy
- GOIRC (Gruppo Oncologico Italiano di Ricerca Clinica), 43126 Parma, Italy
| | - Luca Moscetti
- Division of Medical Oncology, University Hospital of Modena, 41122 Modena, Italy
- GOIRC (Gruppo Oncologico Italiano di Ricerca Clinica), 43126 Parma, Italy
| | - Monica Barbolini
- Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena, 41122 Modena, Italy
- Division of Medical Oncology, University Hospital of Modena, 41122 Modena, Italy
| | - Fabio Canino
- Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena, 41122 Modena, Italy
| | - Antonio Tornincasa
- Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena, 41122 Modena, Italy
| | - Federica Caggia
- Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena, 41122 Modena, Italy
| | - Sara Cerri
- Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena, 41122 Modena, Italy
| | - Alessia Molinaro
- Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena, 41122 Modena, Italy
| | - Massimo Dominici
- Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena, 41122 Modena, Italy
- Division of Medical Oncology, University Hospital of Modena, 41122 Modena, Italy
| | - Claudia Omarini
- Division of Medical Oncology, University Hospital of Modena, 41122 Modena, Italy
- GOIRC (Gruppo Oncologico Italiano di Ricerca Clinica), 43126 Parma, Italy
- Correspondence: ; Tel.: +39-059-422-2845
| |
Collapse
|
21
|
Li J, Zhang H, Gao F. Identification of miRNA biomarkers for breast cancer by combining ensemble regularized multinomial logistic regression and Cox regression. BMC Bioinformatics 2022; 23:434. [PMID: 36258162 PMCID: PMC9580207 DOI: 10.1186/s12859-022-04982-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Accepted: 10/05/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Breast cancer is one of the most common cancers in women. It is necessary to classify breast cancer subtypes because different subtypes need specific treatment. Identifying biomarkers and classifying breast cancer subtypes is essential for developing appropriate treatment methods for patients. MiRNAs can be easily detected in tumor biopsy and play an inhibitory or promoting role in breast cancer, which are considered promising biomarkers for distinguishing subtypes. RESULTS A new method combing ensemble regularized multinomial logistic regression and Cox regression was proposed for identifying miRNA biomarkers in breast cancer. After adopting stratified sampling and bootstrap sampling, the most suitable sample subset for miRNA feature screening was determined via ensemble 100 regularized multinomial logistic regression models. 124 miRNAs that participated in the classification of at least 3 subtypes and appeared at least 50 times in 100 integrations were screened as features. 22 miRNAs from the proposed feature set were further identified as the biomarkers for breast cancer by using Cox regression based on survival analysis. The accuracy of 5 methods on the proposed feature set was significantly higher than on the other two feature sets. The results of 7 biological analyses illustrated the rationality of the identified biomarkers. CONCLUSIONS The screened features can better distinguish breast cancer subtypes. Notably, the genes and proteins related to the proposed 22 miRNAs were considered oncogenes or inhibitors of breast cancer. 9 of the 22 miRNAs have been proved to be markers of breast cancer. Therefore, our results can be considered in future related research.
Collapse
Affiliation(s)
- Juntao Li
- College of Mathematics and Information Science, Henan Normal University, Xinxiang, China
| | - Hongmei Zhang
- College of Mathematics and Information Science, Henan Normal University, Xinxiang, China
| | - Fugen Gao
- College of Mathematics and Information Science, Henan Normal University, Xinxiang, China
| |
Collapse
|
22
|
Tert-butylhydroquinone protects the retina from oxidative stress in STZ-induced diabetic rats via the PI3K/Akt/eNOS pathway. Eur J Pharmacol 2022; 935:175297. [PMID: 36174669 DOI: 10.1016/j.ejphar.2022.175297] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 09/19/2022] [Accepted: 09/20/2022] [Indexed: 02/07/2023]
Abstract
This study aims to investigate whether tert-butylhydroquinone protects the retina from oxidative stress in STZ-induced experimental diabetic rats through the activation of phosphinositide 3-kinase (PI3K)/Akt/endothelial nitric oxide synthase (eNOS) pathway.In vitro, NO, reactive oxygen species(ROS), eNOS, p-eNOS Ser1179, Akt, p-Akt Ser473 and L-NAME protein expression was analyzed within rMC-1 cells cultivated within normal control(NC), high glucose (HG) and HG-containing tert-butyl hydroquinone (tBHQ) (5 μM) medium. We confirmed tBHQ's protection through administering inhibitors of PI3K and Akt. In vivo, tBHQ was administered at a ratio of 1% (w/w) to diabetic rats was induced through an STZ injection (65 mg/kg) for a 3-month period, and the retinal expression of eNOS, p-eNOS Ser1179, Akt, and p-Akt Ser473 proteins was measured using Western blotting (WB) assay. We also utilized the TUNEL kit for detecting retinal cell apoptosis. The changes of retinal morphology and visual function were measured by performing hematoxylin-eosin staining (HE staining) and electroretinograms. In vitro, ROS levels were increased in the high glucose group, NO levels were decreased, and the relative expression of Akt/p-Akt Ser473 and eNOs/p-eNOS Ser1179 was reduced. tBHQ abolished these changes, and these effects were suppressed by specific inhibitors. In vivo, tBHQ upregulated retinal protein expression in STZ-induced diabetic rats, reduced retinal apoptotic cell numbers, and partially prevented abnormalities in retinal function and structure caused by diabetes. tBHQ alleviates oxidative stress during diabetic retinopathy by upregulating the PI3K/Akt/eNOS pathway and partially restoring the structure and function of the retina. It may play a role in delaying vision loss caused by diabetic retinopathy.
Collapse
|
23
|
Mathur R, Jha NK, Saini G, Jha SK, Shukla SP, Filipejová Z, Kesari KK, Iqbal D, Nand P, Upadhye VJ, Jha AK, Roychoudhury S, Slama P. Epigenetic factors in breast cancer therapy. Front Genet 2022; 13:886487. [PMID: 36212140 PMCID: PMC9539821 DOI: 10.3389/fgene.2022.886487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 07/20/2022] [Indexed: 11/17/2022] Open
Abstract
Epigenetic modifications are inherited differences in cellular phenotypes, such as cell gene expression alterations, that occur during somatic cell divisions (also, in rare circumstances, in germ line transmission), but no alterations to the DNA sequence are involved. Histone alterations, polycomb/trithorax associated proteins, short non-coding or short RNAs, long non—coding RNAs (lncRNAs), & DNA methylation are just a few biological processes involved in epigenetic events. These various modifications are intricately linked. The transcriptional potential of genes is closely conditioned by epigenetic control, which is crucial in normal growth and development. Epigenetic mechanisms transmit genomic adaptation to an environment, resulting in a specific phenotype. The purpose of this systematic review is to glance at the roles of Estrogen signalling, polycomb/trithorax associated proteins, DNA methylation in breast cancer progression, as well as epigenetic mechanisms in breast cancer therapy, with an emphasis on functionality, regulatory factors, therapeutic value, and future challenges.
Collapse
Affiliation(s)
- Runjhun Mathur
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, India
- Dr. A.P.J Abdul Kalam Technical University, Lucknow, India
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, India
- Department of Biotechnology, School of Applied and Life Sciences (SALS), Uttaranchal University, Dehradun, India
- Department of Biotechnology Engineering and Food Technology, Chandigarh University, Mohali, India
| | - Gaurav Saini
- Department of Civil Engineering, Netaji Subhas University of Technology, Delhi, India
| | - Saurabh Kumar Jha
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, India
- Department of Biotechnology Engineering and Food Technology, Chandigarh University, Mohali, India
| | - Sheo Prasad Shukla
- Department of Civil Engineering, Rajkiya Engineering College, Banda, India
| | - Zita Filipejová
- Small Animal Clinic, University of Veterinary Sciences Brno, Brno, Czechia
| | | | - Danish Iqbal
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Al Majma'ah, Saudi Arabia
- Health and Basic Sciences Research Center, Majmaah University, Al Majma'ah, Saudi Arabia
| | - Parma Nand
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, India
| | - Vijay Jagdish Upadhye
- Center of Research for Development (CR4D), Parul Institute of Applied Sciences (PIAS), Parul University, Vadodara, Gujarat
| | - Abhimanyu Kumar Jha
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, India
- *Correspondence: Abhimanyu Kumar Jha, ; Shubhadeep Roychoudhury,
| | - Shubhadeep Roychoudhury
- Department of Life Science and Bioinformatics, Assam University, Silchar, India
- *Correspondence: Abhimanyu Kumar Jha, ; Shubhadeep Roychoudhury,
| | - Petr Slama
- Department of Animal Morphology, Physiology, and Genetics, Faculty of AgriSciences, Mendel University in Brno, Brno, Czechia
| |
Collapse
|
24
|
Liu Z, Meng D, Wang J, Cao H, Feng P, Wu S, Wang N, Dang C, Hou P, Xia P. GASP1 enhances malignant phenotypes of breast cancer cells and decreases their response to paclitaxel by forming a vicious cycle with IGF1/IGF1R signaling pathway. Cell Death Dis 2022; 13:751. [PMID: 36042202 PMCID: PMC9427794 DOI: 10.1038/s41419-022-05198-6] [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: 05/05/2022] [Revised: 08/16/2022] [Accepted: 08/17/2022] [Indexed: 01/21/2023]
Abstract
There is a potential correlation between G-protein-coupled receptor-associated sorting protein 1 (GASP1) and breast tumorigenesis. However, its biological function and underlying molecular mechanism in breast cancer have not been clearly delineated. Here, we demonstrated that GASP1 was highly expressed in breast cancers, and patients harboring altered GASP1 showed a worse prognosis than those with wild-type GASP1. Functional studies showed that GASP1 knockout significantly suppressed malignant properties of breast cancer cells, such as inhibition of cell proliferation, colony formation, migration, invasion and xenograft tumor growth in nude mice as well as induction of G1-phase cell cycle arrest, and vice versa. Mechanistically, GASP1 inhibited proteasomal degradation of insulin-like growth factor 1 receptor (IGF1R) by competitively binding to IGF1R with ubiquitin E3 ligase MDM2, thereby activating its downstream signaling pathways such as NF-κB, PI3K/AKT, and MAPK/ERK pathways given their critical roles in breast tumorigenesis and progression. IGF1, in turn, stimulated GASP1 expression by activating the PI3K/AKT pathway, forming a vicious cycle propelling the malignant progression of breast cancer. Besides, we found that GASP1 knockout obviously improved the response of breast cancer cells to paclitaxel. Collectively, this study demonstrates that GASP1 enhances malignant behaviors of breast cancer cells and decreases their cellular response to paclitaxel by interacting with and stabilizing IGF1R, and suggests that it may serve as a valuable prognostic factor and potential therapeutic target in breast cancer.
Collapse
Affiliation(s)
- Zhao Liu
- grid.452438.c0000 0004 1760 8119Department of Surgical Oncology, The First Affiliated Hospital of Xi’an Jiaotong University, 710061 Xi’an, People’s Republic of China ,grid.452438.c0000 0004 1760 8119Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi’an Jiaotong University, 710061 Xi’an, People’s Republic of China
| | - Du Meng
- grid.452438.c0000 0004 1760 8119Department of Radio Oncology, The First Affiliated Hospital of Xi’an Jiaotong University, 710061 Xi’an, People’s Republic of China
| | - Jianling Wang
- grid.452438.c0000 0004 1760 8119Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi’an Jiaotong University, 710061 Xi’an, People’s Republic of China
| | - Hongxin Cao
- grid.452438.c0000 0004 1760 8119Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi’an Jiaotong University, 710061 Xi’an, People’s Republic of China
| | - Peng Feng
- grid.452438.c0000 0004 1760 8119Department of Surgical Oncology, The First Affiliated Hospital of Xi’an Jiaotong University, 710061 Xi’an, People’s Republic of China
| | - Siyu Wu
- grid.452438.c0000 0004 1760 8119Department of Surgical Oncology, The First Affiliated Hospital of Xi’an Jiaotong University, 710061 Xi’an, People’s Republic of China
| | - Na Wang
- grid.478124.c0000 0004 1773 123XDepartment of Endocrinology, Xi’an Central Hospital, 710003 Xi’an, People’s Republic of China
| | - Chengxue Dang
- grid.452438.c0000 0004 1760 8119Department of Surgical Oncology, The First Affiliated Hospital of Xi’an Jiaotong University, 710061 Xi’an, People’s Republic of China
| | - Peng Hou
- grid.452438.c0000 0004 1760 8119Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi’an Jiaotong University, 710061 Xi’an, People’s Republic of China ,grid.452438.c0000 0004 1760 8119Department of Endocrinology, The First Affiliated Hospital of Xi’an Jiaotong University, 710061 Xi’an, People’s Republic of China
| | - Peng Xia
- grid.452438.c0000 0004 1760 8119Department of Surgical Oncology, The First Affiliated Hospital of Xi’an Jiaotong University, 710061 Xi’an, People’s Republic of China
| |
Collapse
|
25
|
Khan SU, Fatima K, Malik F. Understanding the cell survival mechanism of anoikis-resistant cancer cells during different steps of metastasis. Clin Exp Metastasis 2022; 39:715-726. [PMID: 35829806 DOI: 10.1007/s10585-022-10172-9] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Accepted: 05/25/2022] [Indexed: 11/26/2022]
Abstract
Anchorage-independent survival of cancer cells is associated with metastasis as it enables cells to travel to secondary target sites. Tissue integrity is generally maintained by detachment-induced cell death called 'anoikis', but cancer cells undergoing the multistep metastatic process show resistance to anoikis. Anoikis resistance enables these cells to survive through the extracellular matrix (ECM) deprived phase, which starts when cancer cells detach and move into the circulation till cells reach to the secondary target site. Comprehensive analysis of the molecular and functional biology of anoikis resistance in cancer cells will provide crucial details about cancer metastasis, enabling us to identify novel therapeutic targets against cancer cell dissemination and ultimately secondary tumor formation. This review broadly summarizes recent advances in the understanding of cellular and molecular events leading to anoikis and anoikis resistance. It further elaborates more about the signaling cross-talk in anoikis resistance and its regulation during metastasis.
Collapse
Affiliation(s)
- Sameer Ullah Khan
- Department of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Sanat Nagar, 190005, Srinagar, Jammu and Kashmir, India
- Academy of Scientific and Innovative Research (AcSIR), 201002, Ghaziabad, India
| | - Kaneez Fatima
- Department of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Sanat Nagar, 190005, Srinagar, Jammu and Kashmir, India
- Academy of Scientific and Innovative Research (AcSIR), 201002, Ghaziabad, India
| | - Fayaz Malik
- Department of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Sanat Nagar, 190005, Srinagar, Jammu and Kashmir, India.
| |
Collapse
|
26
|
Vathiotis IA, Trontzas I, Gavrielatou N, Gomatou G, Syrigos NK, Kotteas EA. Immune Checkpoint Blockade in Hormone Receptor-Positive Breast Cancer: Resistance Mechanisms and Future Perspectives. Clin Breast Cancer 2022; 22:642-649. [PMID: 35906130 DOI: 10.1016/j.clbc.2022.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 05/21/2022] [Accepted: 06/29/2022] [Indexed: 11/03/2022]
Abstract
Anti-programmed cell death protein 1 immunotherapy has been incorporated in the treatment algorithm of triple-negative breast cancer (TNBC). However, clinical trial results for patients with hormone receptor (HR)-positive disease appear less compelling. HR-positive tumors exhibit lower levels of programmed death-ligand 1 expression in comparison with their triple-negative counterparts. Moreover, signaling through estrogen receptor alters the immune microenvironment, rendering such tumors immunologically "cold." To explain differential responses to immune checkpoint blockade, this review interrogates differences between HR-positive and TNBC. Starting from distinct genomic features, we further present disparities concerning the tumor microenvironment and finally, we summarize early-phase clinical trial results on promising novel immunotherapy combinations.
Collapse
Affiliation(s)
- Ioannis A Vathiotis
- Department of Medicine, School of Medicine, National and Kapodistrian University of Athens, Athens, Attica, Greece; Department of Pathology, Yale University School of Medicine, New Haven, CT.
| | - Ioannis Trontzas
- Department of Medicine, School of Medicine, National and Kapodistrian University of Athens, Athens, Attica, Greece
| | - Niki Gavrielatou
- Department of Pathology, Yale University School of Medicine, New Haven, CT
| | - Georgia Gomatou
- Department of Medicine, School of Medicine, National and Kapodistrian University of Athens, Athens, Attica, Greece
| | - Nikolaos K Syrigos
- Department of Medicine, School of Medicine, National and Kapodistrian University of Athens, Athens, Attica, Greece
| | - Elias A Kotteas
- Department of Medicine, School of Medicine, National and Kapodistrian University of Athens, Athens, Attica, Greece
| |
Collapse
|
27
|
Shen WQ, Guo Y, Ru WE, Li C, Zhang GC, Liao N, Du GQ. Using an Improved Residual Network to Identify PIK3CA Mutation Status in Breast Cancer on Ultrasound Image. Front Oncol 2022; 12:850515. [PMID: 35719907 PMCID: PMC9204315 DOI: 10.3389/fonc.2022.850515] [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: 01/07/2022] [Accepted: 04/11/2022] [Indexed: 11/30/2022] Open
Abstract
Background The detection of phosphatidylinositol-3 kinase catalytic alpha (PIK3CA) gene mutations in breast cancer is a key step to design personalizing an optimal treatment strategy. Traditional genetic testing methods are invasive and time-consuming. It is urgent to find a non-invasive method to estimate the PIK3CA mutation status. Ultrasound (US), one of the most common methods for breast cancer screening, has the advantages of being non-invasive, fast imaging, and inexpensive. In this study, we propose to develop a deep convolutional neural network (DCNN) to identify PIK3CA mutations in breast cancer based on US images. Materials and Methods We retrospectively collected 312 patients with pathologically confirmed breast cancer who underwent genetic testing. All US images (n=800) of breast cancer patients were collected and divided into the training set (n=600) and test set (n=200). A DCNN-Improved Residual Network (ImResNet) was designed to identify the PIK3CA mutations. We also compared the ImResNet model with the original ResNet50 model, classical machine learning models, and other deep learning models. Results The proposed ImResNet model has the ability to identify PIK3CA mutations in breast cancer based on US images. Notably, our ImResNet model outperforms the original ResNet50, DenseNet201, Xception, MobileNetv2, and two machine learning models (SVM and KNN), with an average area under the curve (AUC) of 0.775. Moreover, the overall accuracy, average precision, recall rate, and F1-score of the ImResNet model achieved 74.50%, 74.17%, 73.35%, and 73.76%, respectively. All of these measures were significantly higher than other models. Conclusion The ImResNet model gives an encouraging performance in predicting PIK3CA mutations based on breast US images, providing a new method for noninvasive gene prediction. In addition, this model could provide the basis for clinical adjustments and precision treatment.
Collapse
Affiliation(s)
- Wen-Qian Shen
- Department of Ultrasound, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,Department of Ultrasound, The Second Affifiliated Hospital of Harbin Medical University, Harbin, China
| | - Yanhui Guo
- Department of Computer Science, University of Illinois Springfield, Springfield, IL, United States
| | - Wan-Er Ru
- Department of Ultrasound, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,College of Medicine, Shantou University, Shantou, China
| | - Cheukfai Li
- Department of Breast Cancer, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Guo-Chun Zhang
- Department of Breast Cancer, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Ning Liao
- Department of Breast Cancer, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Guo-Qing Du
- Department of Ultrasound, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| |
Collapse
|
28
|
Dong M, Shan B, Han X, Zhao X, Wang F, Zhu L, Ou Q, Ma X, Pan Y. Baseline Mutations and Up-Regulation of PI3K-AKT Pathway Serve as Potential Indicators of Lack of Response to Neoadjuvant Chemotherapy in Stage II/III Breast Cancer. Front Oncol 2022; 11:784985. [PMID: 35480699 PMCID: PMC9036956 DOI: 10.3389/fonc.2021.784985] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 12/28/2021] [Indexed: 12/19/2022] Open
Abstract
Background Neoadjuvant chemotherapy (NAC) has been expanded to hormone receptor (HR) positive breast cancer (BC) patients with operable disease, to increase the likelihood of breast-conserving surgery. Genomic profiling at baseline would reveal NAC response relevant genomic features and signaling pathways, guiding clinical NAC utilization based on patients’ genomic characteristics. Methods We prospectively studied stage II/III BC patients who were eligible for breast-conserving surgery. Patients received epirubicin and cyclophosphamide for 4 cycles, followed by another 4-cycle docetaxel, and human epidermal growth factor receptor (HER2) positive patients were additionally treated with herceptin when using docetaxel (EC-T(H)). NAC responses were evaluated as pathologic complete response (pCR) or non-pathologic complete response (non-pCR). Genomic features related to NAC responses were identified by profiling baseline tumor tissues sampled one day before NAC, using whole-exome sequencing. Differentially expressed genes and up-/down-regulated pathways were investigated by performing RNA-sequencing. Results A total of 25 stage II/III BC patients were enrolled, including 5 patients ultimately evaluated as pCR and 20 patients evaluated as non-pCR. PIK3CA (48%) and TP53 (40%) mutations were enriched in patients not achieving pCR. Mutated phosphatidylinositol-3-kinase-AKT (PI3K-AKT) pathway and homologous recombinational repair pathway were also more frequently observed in patients evaluated as non-pCR. Significant arm-level amplifications (8q24.23 and 17q12) and deletions (1p32.2, 4p14, 7q11.23, 10q21.3, 11q23.3, etc.) were identified among patients not achieving pCR, while patients achieving pCR displayed no significant copy number alterations. Significantly up-regulated expression of PI3K-AKT pathway genes was also detected among patients failed to achieve pCR, compared to patients achieving pCR. Conclusion Compared to BC patients achieving pCR to NAC, aberrant activation of PI3K-AKT pathway genes were more frequently observed in patients not achieving pCR, consistent with the significant up-regulation of PI3K-AKT pathway gene expression in the non-pCR subgroup. Together, these findings indicate that upregulated PI3K-AKT pathway serves as a potential indicator of lack of response to NAC in stage II/III BC patients, and other effective therapeutic options are urgently needed for those resistant patients.
Collapse
Affiliation(s)
- Menghao Dong
- Department of Medical Oncology, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Benjie Shan
- Department of Medical Oncology, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Xinghua Han
- Department of Medical Oncology, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Xiaotian Zhao
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, China
| | - Fufeng Wang
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, China
| | - Liuqing Zhu
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, China
| | - Qiuxiang Ou
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, China
| | - Xiaopeng Ma
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yueyin Pan
- Department of Medical Oncology, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
29
|
Chen SY, Hsu YH, Wang SY, Chen YY, Hong CJ, Yen GC. Lucidone inhibits autophagy and MDR1 via HMGB1/RAGE/PI3K/Akt signaling pathway in pancreatic cancer cells. Phytother Res 2022; 36:1664-1677. [PMID: 35224793 DOI: 10.1002/ptr.7385] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 12/22/2021] [Accepted: 12/24/2021] [Indexed: 12/12/2022]
Abstract
Gemcitabine (GEM) drug resistance remains a difficult challenge in pancreatic ductal adenocarcinoma (PDAC) treatment. Therefore, identifying a safe and effective treatment strategy for PDAC is urgent. Lucidone is a natural compound extracted from the fruits of Lindera erythrocarpa Makino. However, the role of lucidone in PDAC inhibition remains unclear. In addition, high-mobility group box 1 (HMGB1) and receptor for advanced glycation end products (RAGE) are involved in multidrug resistance protein 1 (MDR1) regulation and GEM resistance. Thus, this study aimed to explore the function of lucidone in tumor cytotoxicity and chemosensitivity through the suppression of RAGE-initiated signaling in PDAC cells. The data showed that lucidone significantly promoted apoptotic cell death and inhibited the expression of autophagic proteins (Atg5, Beclin-1, LC3-II, and Vps34) and MDR1 by inhibiting the HMGB1/RAGE/PI3K/Akt axis in both MIA Paca-2 cells and MIA Paca-2GEMR cells (GEM-resistant cells). Notably, convincing data were also obtained in experiments involving RAGE-specific siRNA transfection. In addition, remarkable cell proliferation was observed after treatment with lucidone combined with GEM, particularly in MIA Paca-2GEMR cells, indicating that lucidone treatment enhanced chemosensitivity. Collectively, this study provided the underlying mechanism by which lucidone treatment inhibited HMGB1/RAGE-initiated PI3K/Akt/MDR1 signaling and consequently enhanced chemosensitivity in PDAC.
Collapse
Affiliation(s)
- Sheng-Yi Chen
- Department of Food Science and Biotechnology, National Chung Hsing University, Taichung, Taiwan
| | - Yi-Hao Hsu
- Department of Food Science and Biotechnology, National Chung Hsing University, Taichung, Taiwan
| | - Sheng-Yang Wang
- Department of Forestry, National Chung Hsing University, Taichung, Taiwan
| | - Ying-Yin Chen
- Department of Food Science and Biotechnology, National Chung Hsing University, Taichung, Taiwan
| | - Cheng-Jie Hong
- Department of Food Science and Biotechnology, National Chung Hsing University, Taichung, Taiwan
| | - Gow-Chin Yen
- Department of Food Science and Biotechnology, National Chung Hsing University, Taichung, Taiwan
| |
Collapse
|
30
|
Vadukoot AK, Mottemmal S, Vekaria PH. Curcumin as a Potential Therapeutic Agent in Certain Cancer Types. Cureus 2022; 14:e22825. [PMID: 35399416 PMCID: PMC8980239 DOI: 10.7759/cureus.22825] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/03/2022] [Indexed: 12/17/2022] Open
Abstract
Cancer is a devastating disease condition and is the second most common etiology of death globally. After decades of research in the field of hematological malignancies and cellular therapeutics, we are still looking for therapeutic agents with the most efficacies and least toxicities. Curcumin is one of the cancer therapeutic agents that is derived from the Curcuma longa (turmeric) plant, and still in vitro and in vivo research is going on to find its beneficial effects on various cancers. Due to its potency to affect multiple targets of different cellular pathways, it is considered a promising agent to tackle various cancers alone or in combination with the existing chemotherapies. This review covers basic properties, mechanism of action, potential targets (molecules and cell-signaling pathways) of curcumin, as well as its effect on various solid and hematological malignancies.
Collapse
|
31
|
Shi D, Li H, Zhang Z, He Y, Chen M, Sun L, Zhao P. Cryptotanshinone inhibits proliferation and induces apoptosis of breast cancer MCF-7 cells via GPER mediated PI3K/AKT signaling pathway. PLoS One 2022; 17:e0262389. [PMID: 35061800 PMCID: PMC8782479 DOI: 10.1371/journal.pone.0262389] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 12/23/2021] [Indexed: 12/15/2022] Open
Abstract
G protein-coupled estrogen receptor (GPER) was reported to be a potential target in the breast cancer therapy. This study aimed to illuminate the function of GPER and its mediated PI3K/AKT pathway in cryptotanshinone (CPT) inducing cell apoptosis and antiproliferation effect on GPER positive breast cancer MCF-7 cells. Cell proliferation was tested by MTT assay. Apoptosis rates were tested by Annexin V-FITC/PI double staining and the cell cycle was researched by flow cytometry. Autodock vina was applied to make molecular docking between CPT or estradiol and GPER. siRNA technique and GPER specific agonist G-1 or antagonist G-15 were applied to verify the mediated function of GPER. Apoptosis and cell cycle related proteins, as well as the key proteins on PI3K/AKT signaling pathway were detected by western blot. The results indicated that CPT could exert antiproliferation effects by arresting cell cycle in G2/M phase and downregulating the expression of cyclin D, cyclin B and cyclin A. Besides, apoptosis induced by CPT was observed. CPT might be a novel GPER binding compounds. Significantly, suppression of PI3K/AKT signal transduction by CPT was further increased by G-1 and decreased by G-15. The study revealed that the effect of antiproliferation and apoptosis treating with CPT on MCF-7 cells might be through the downregulation of PI3K/AKT pathway mediated by activated GPER.
Collapse
Affiliation(s)
- Danning Shi
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Hongbo Li
- Department of Gynecology, Affiliated Hospital of Shaanxi University of Chinese Medicine, Shaanxi, 712000, China
| | - Zeye Zhang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yueshuang He
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Meng Chen
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Liping Sun
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Piwen Zhao
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, China
- * E-mail:
| |
Collapse
|
32
|
Anti-Cancer Effects of α-Cubebenoate Derived from Schisandra chinensis in CT26 Colon Cancer Cells. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27030737. [PMID: 35164001 PMCID: PMC8839175 DOI: 10.3390/molecules27030737] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 01/18/2022] [Accepted: 01/20/2022] [Indexed: 11/26/2022]
Abstract
α-Cubebenoate derived from Schisandra chinensis has been reported to possess anti-allergic, anti-obesity, and anti-inflammatory effects and to exhibit anti-septic activity, but its anti-cancer effects have not been investigated. To examine the anti-cancer activity of α-cubebenoate, we investigated its effects on the proliferation, apoptosis, and metastasis of CT26 cells. The viabilities of CT26 cells (a murine colorectal carcinoma cell line) and HCT116 cells (a human colon cancer cell line) were remarkably and dose-dependently diminished by α-cubebenoate, whereas the viability of CCD-18Co cells (a normal human fibroblast cell line) were unaffected. Furthermore, α-cubebenoate treatment increased the number of apoptotic CT26 cells as compared with Vehicle-treated cells and increased Bax, Bcl-2, Cas-3, and Cleaved Cas-3 protein levels by activating the MAP kinase signaling pathway. α-Cubebenoate also suppressed CT26 migration by regulating the PI3K/AKT signaling pathway. Furthermore, similar reductions were observed in the expression levels of some migration-related proteins including VEGFA, MMP2, and MMP9. Furthermore, reduced VEGFA expression was found to be accompanied by the phosphorylations of FAK and MLC in the downstream signaling pathway of adhesion protein. The results of the present study provide novel evidence that α-cubebenoate can stimulate apoptosis and inhibit metastasis by regulating the MAPK, PI3K/AKT, and FAK/MLC signaling pathways.
Collapse
|
33
|
Ziasarabi P, Sahebkar A, Ghasemi F. Evaluation of the Effects of Nanomicellar Curcumin, Berberine, and Their Combination with 5-Fluorouracil on Breast Cancer Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1328:21-35. [PMID: 34981469 DOI: 10.1007/978-3-030-73234-9_3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
INTRODUCTION Breast cancer is one of the main challenging areas in cancer treatment. Natural compounds such as curcumin and berberine have been approved with anticancer effects and are more favorable to people. Here, we investigated the potential synergistic anticancer effects of these two compounds in combination with the standard cancer drug 5-FU on the growth of MCF-7 breast cancer cells. MATERIALS AND METHODS This study tested the effects of six different treatments on cancer cell growth: A) control; B) curcumin; C) berberine; D) 5-FU; E) curcumin + berberine; and F) curcumin + berberine + 5-FU. The IC50 concentration of each treatment on cancer cell growth was determined using the MTT assay. Invasiveness of cells grown in 3D culture was analyzed using the transwell chamber technique. Expression levels of genes involved in cancer cell growth and survival (WNT1, APC, AXIN1, CTNNB1, TCF, MTOR, AKT1, MAPK1, PTEN, BIRC5, CCNG1) were evaluated by real-time PCR. RESULTS There was a reduction in cancer cell growth and invasion, and an increase in cellular decomposition across all treatment groups compared to the control with the strongest effects seen in the combined curcumin/berberine/5-FU group. The expression levels of all tested genes were altered in all treatment groups compared to the control, with that of WNT1, CTNNB1, TCF, MTOR, AKT1, BIRC5, and CCND1 showing the most robust changes in the combined curcumin/berberine/5-FU treatment. CONCLUSIONS All treatment groups had anti-growth, anti-invasion, and pro-apoptotic effects on MCF-7 breast cancer cells in culture. In addition, all treatment groups showed changes in the expression of the genes involved in cancer cell growth and survival with the strongest effects found for the curcumin/berberine/5-FU combination. Therefore, curcumin and berberine may improve the anticancer effects of chemotherapy and these natural compounds should undergo further testing as potential adjuvants.
Collapse
Affiliation(s)
- Parisa Ziasarabi
- Laboratorio de Psicobiología, Campus Santiago Ramón y Cajal, University of Sevilla, Sevilla, Spain
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Faezeh Ghasemi
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran.
| |
Collapse
|
34
|
Allouchery V, Perdrix A, Calbrix C, Berghian A, Lequesne J, Fontanilles M, Leheurteur M, Etancelin P, Sarafan-Vasseur N, Di Fiore F, Clatot F. Circulating PIK3CA mutation detection at diagnosis in non-metastatic inflammatory breast cancer patients. Sci Rep 2021; 11:24041. [PMID: 34911971 PMCID: PMC8674263 DOI: 10.1038/s41598-021-02643-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 11/11/2021] [Indexed: 01/04/2023] Open
Abstract
Inflammatory breast cancer (IBC) is an aggressive BC subtype with poor outcomes. A targetable somatic PIK3CA mutation is reported in 30% of IBC, allowing for treatment by PI3Kα-specific inhibitors, such as alpelisib. The aim of this study was to evaluate the detection rate of circulating PIK3CA mutation in locally-advanced IBC (LAIBC) patients harbouring a PIK3CA mutation on initial biopsy. This monocentric retrospective study was based on available stored plasma samples and tumour biopsies at diagnosis from all LAIBC patients treated with neo-adjuvant chemotherapy (NCT) between 2008 and 2018 at the Centre Henri Becquerel. PIK3CA mutations (E542K, E545K, H1047R/L) were assessed by droplet digital PCR (ddPCR) in plasma samples and tumoral tissue at diagnosis. A total of 55 patients were included. Overall, 14/55 patients (25%) had a PIK3CA mutation identified on baseline biopsy (H1047R = 8; H1047L = 3; E545K = 2; E542K = 1). Among them, 11 (79%) patients had enough DNA for circulating DNA analyses, and corresponding circulating PIK3CA mutations were found in 6/11 (55%). Among the 41 patients without PIK3CA mutations on biopsy, 32 (78%) had enough DNA for circulating DNA analysis, and no circulating PIK3CA mutation was identified. Our results revealed no prognostic or predictive value of PIK3CA mutations at the diagnosis of non-metastatic IBC but highlighted the prognostic value of the cfDNA rate at diagnosis. Our study showed that a corresponding circulating PIK3CA mutation was identified in 55% of LAIBC patients with PIK3CA-mutated tumours, while no circulating mutation was found among patients with PI3KCA wild-type tumours.
Collapse
Affiliation(s)
- Violette Allouchery
- Department of Medical Oncology, Centre Henri Becquerel, 1 Rue d'Amiens, 76038, Rouen Cedex 1, France.
| | - Anne Perdrix
- IRON Group, Inserm U1245, UNIROUEN, Rouen University Hospital, Normandy Centre for Genomic and Personalized Medicine, Normandie Université, Rouen, France.,Department of Bio-Pathology, Centre Henri Becquerel, Rouen, France
| | - Céline Calbrix
- IRON Group, Inserm U1245, UNIROUEN, Rouen University Hospital, Normandy Centre for Genomic and Personalized Medicine, Normandie Université, Rouen, France.,Department of Bio-Pathology, Centre Henri Becquerel, Rouen, France
| | - Anca Berghian
- Department of Bio-Pathology, Centre Henri Becquerel, Rouen, France
| | - Justine Lequesne
- Department of Biostatistics, Rouen University Hospital, Rouen, France
| | - Maxime Fontanilles
- Department of Medical Oncology, Centre Henri Becquerel, 1 Rue d'Amiens, 76038, Rouen Cedex 1, France.,IRON Group, Inserm U1245, UNIROUEN, Rouen University Hospital, Normandy Centre for Genomic and Personalized Medicine, Normandie Université, Rouen, France
| | - Marianne Leheurteur
- Department of Medical Oncology, Centre Henri Becquerel, 1 Rue d'Amiens, 76038, Rouen Cedex 1, France
| | | | - Nasrin Sarafan-Vasseur
- IRON Group, Inserm U1245, UNIROUEN, Rouen University Hospital, Normandy Centre for Genomic and Personalized Medicine, Normandie Université, Rouen, France.,Department of Bio-Pathology, Centre Henri Becquerel, Rouen, France
| | - Frédéric Di Fiore
- Department of Medical Oncology, Centre Henri Becquerel, 1 Rue d'Amiens, 76038, Rouen Cedex 1, France.,IRON Group, Inserm U1245, UNIROUEN, Rouen University Hospital, Normandy Centre for Genomic and Personalized Medicine, Normandie Université, Rouen, France.,Department of Gastroenterology, Rouen University Hospital, Rouen, France
| | - Florian Clatot
- Department of Medical Oncology, Centre Henri Becquerel, 1 Rue d'Amiens, 76038, Rouen Cedex 1, France.,IRON Group, Inserm U1245, UNIROUEN, Rouen University Hospital, Normandy Centre for Genomic and Personalized Medicine, Normandie Université, Rouen, France
| |
Collapse
|
35
|
Shrihastini V, Muthuramalingam P, Adarshan S, Sujitha M, Chen JT, Shin H, Ramesh M. Plant Derived Bioactive Compounds, Their Anti-Cancer Effects and In Silico Approaches as an Alternative Target Treatment Strategy for Breast Cancer: An Updated Overview. Cancers (Basel) 2021; 13:cancers13246222. [PMID: 34944840 PMCID: PMC8699774 DOI: 10.3390/cancers13246222] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 12/08/2021] [Accepted: 12/09/2021] [Indexed: 12/13/2022] Open
Abstract
Cancer is one of the most common malignant diseases that occur worldwide, among which breast cancer is the second leading cause of death in women. The subtypes are associated with differences in the outcome and were selected for treatments according to the estrogen receptor, progesterone receptor, and human epidermal growth factor receptor. Triple-negative breast cancer, one of the subtypes of breast cancer, is difficult to treat and can even lead to death. If breast cancer is not treated during the initial stages, it may spread to nearby organs, a process called metastasis, through the blood or lymph system. For in vitro studies, MCF-7, MDA-MB-231, MDA-MB-468, and T47B are the most commonly used breast cancer cell lines. Clinically, chemotherapy and radiotherapy are usually expensive and can also cause side effects. To overcome these issues, medicinal plants could be the best alternative for chemotherapeutic drugs with fewer side effects and cost-effectiveness. Furthermore, the genes involved in breast cancer can be regulated and synergized with signaling molecules to suppress the proliferation of breast cancer cells. In addition, nanoparticles encapsulating (nano-encapsulation) medicinal plant extracts showed a significant reduction in the apoptotic and cytotoxic activities of breast cancer cells. This present review mainly speculates an overview of the native medicinal plant derived anti-cancerous compounds with its efficiency, types and pathways involved in breast cancer along with its genes, the mechanism of breast cancer brain metastasis, chemoresistivity and its mechanism, bioinformatics approaches which could be an effective alternative for drug discovery.
Collapse
Affiliation(s)
- Vijayakumar Shrihastini
- Department of Biotechnology, Sri Shakthi Institute of Engineering and Technology, Coimbatore 641062, Tamil Nadu, India; (V.S.); (M.S.)
| | - Pandiyan Muthuramalingam
- Department of Biotechnology, Sri Shakthi Institute of Engineering and Technology, Coimbatore 641062, Tamil Nadu, India; (V.S.); (M.S.)
- Correspondence: (P.M.); (J.-T.C.)
| | - Sivakumar Adarshan
- Department of Biotechnology, Science Campus, Alagappa University, Karaikudi 630003, Tamil Nadu, India; (S.A.); (M.R.)
| | - Mariappan Sujitha
- Department of Biotechnology, Sri Shakthi Institute of Engineering and Technology, Coimbatore 641062, Tamil Nadu, India; (V.S.); (M.S.)
| | - Jen-Tsung Chen
- Department of Life Sciences, National University of Kaohsiung, Kaohsiung 811, Taiwan
- Correspondence: (P.M.); (J.-T.C.)
| | - Hyunsuk Shin
- Department of Horticultural Sciences, Gyeongsang National University, Jinju 52725, Korea;
| | - Manikandan Ramesh
- Department of Biotechnology, Science Campus, Alagappa University, Karaikudi 630003, Tamil Nadu, India; (S.A.); (M.R.)
| |
Collapse
|
36
|
The Discovery of New Drug-Target Interactions for Breast Cancer Treatment. Molecules 2021; 26:molecules26247474. [PMID: 34946556 PMCID: PMC8704452 DOI: 10.3390/molecules26247474] [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] [Received: 11/22/2021] [Revised: 12/07/2021] [Accepted: 12/07/2021] [Indexed: 01/09/2023] Open
Abstract
Drug–target interaction (DTIs) prediction plays a vital role in probing new targets for breast cancer research. Considering the multifaceted challenges associated with experimental methods identifying DTIs, the in silico prediction of such interactions merits exploration. In this study, we develop a feature-based method to infer unknown DTIs, called PsePDC-DTIs, which fuses information regarding protein sequences extracted by pseudo-position specific scoring matrix (PsePSSM), detrended cross-correlation analysis coefficient (DCCA coefficient), and an FP2 format molecular fingerprint descriptor of drug compounds. In addition, the synthetic minority oversampling technique (SMOTE) is employed for dealing with the imbalanced data after Lasso dimensionality reduction. Then, the processed feature vectors are put into a random forest classifier to perform DTIs predictions on four gold standard datasets, including nuclear receptors (NR), G-protein-coupled receptors (GPCR), ion channels (IC), and enzymes (E). Furthermore, we explore new targets for breast cancer treatment using its risk genes identified from large-scale genome-wide genetic studies using PsePDC-DTIs. Through five-fold cross-validation, the average values of accuracy in NR, GPCR, IC, and E datasets are 95.28%, 96.19%, 96.74%, and 98.22%, respectively. The PsePDC-DTIs model provides us with 10 potential DTIs for breast cancer treatment, among which erlotinib (DB00530) and FGFR2 (hsa2263), caffeine (DB00201) and KCNN4 (hsa3783), as well as afatinib (DB08916) and FGFR2 (hsa2263) are found with direct or inferred evidence. The PsePDC-DTIs model has achieved good prediction results, establishing the validity and superiority of the proposed method.
Collapse
|
37
|
Jiang H, Li M, Du K, Ma C, Cheng Y, Wang S, Nie X, Fu C, He Y. Traditional Chinese Medicine for adjuvant treatment of breast cancer: Taohong Siwu Decoction. Chin Med 2021; 16:129. [PMID: 34857023 PMCID: PMC8638166 DOI: 10.1186/s13020-021-00539-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 11/17/2021] [Indexed: 02/06/2023] Open
Abstract
The high incidence of breast cancer is the greastest threat to women' health all over the world. Among them, HER-2 positive breast cancer has the characteristics of high malignancy, easy recurrence and metastasis, and poor prognosis. Traditional Chinese medicine (TCM) has a rich theoretical basis and clinical application for breast cancer. TCM believes that blood stasis syndrome is one of the important pathogenesis of breast formation and development. Taohong Siwu Decoction (TSHWD) is based on the "First Prescription of Gynecology" Siwu Decoction. It is widely used in various blood stasis and blood deficiency syndromes, mainly in gynecological blood stasis. Clinical studies have found that THSWD can treat breast cancer by reducing blood vessel and lymphangiogenesis with auxiliary chemotherapy. In this study, we aim to explore the material basis and mechanism of THSWD in the treatment of HER-2 positive breast cancer through literature review and network pharmacology studies. Through a literature review of the traditional application, chemical composition of Chinese herbal medicine of THSWD, as well as its clinical reports and pharmacological research on breast cancer treatment. Meanwhile, we conducted "component-pathway-target" network through network pharmacology reveals the main material basis, possible targets and pathways of THSWD in inhibiting HER-2 positive breast cancer. Literature review and network pharmacology research results had predicted that, baicalein, kaempferol, caffeic acid, amygdalin, quercetin, ferulic acid, gallic acid, catalpol, hydroxysafflor yellow A, paeoniflorin in THSWD are the main effective chemical composition. THSWD regulates 386 protein targets and 166 pathways related to breast cancer. The molecular mechanism is mainly to improve the microenvironment of tumor cells, regulate the process of tumor cell EMT, and inhibit tumor cell proliferation and metastasis. This study revealed the mechanism of action of THSWD in the treatment of HER-2 positive breast cancer through literature review and network pharmacology studies, providing a scientific basis for clinical application.
Collapse
Affiliation(s)
- Huajuan Jiang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy College, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Wenjiang District, Chengdu, 611137, Sichuan, China
| | - Minmin Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy College, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Wenjiang District, Chengdu, 611137, Sichuan, China
| | - Kequn Du
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy College, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Wenjiang District, Chengdu, 611137, Sichuan, China
| | - Chuan Ma
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy College, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Wenjiang District, Chengdu, 611137, Sichuan, China
| | - Yanfen Cheng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy College, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Wenjiang District, Chengdu, 611137, Sichuan, China
| | - Shengju Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy College, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Wenjiang District, Chengdu, 611137, Sichuan, China
| | - Xin Nie
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy College, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Wenjiang District, Chengdu, 611137, Sichuan, China
| | - Chaomei Fu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy College, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Wenjiang District, Chengdu, 611137, Sichuan, China
| | - Yao He
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy College, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Wenjiang District, Chengdu, 611137, Sichuan, China.
- Guizhou Yibai Pharmaceutical Co. Ltd, Guiyang, 550008, Guizhou, China.
| |
Collapse
|
38
|
Chen J, Wang M, Wang H, Long M. Zearalenone promotes apoptosis of mouse Leydig cells by targeting phosphatase and tensin homolog and thus inhibiting the PI3K/AKT signal pathway. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:67779-67787. [PMID: 34264493 DOI: 10.1007/s11356-021-15282-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 06/29/2021] [Indexed: 06/13/2023]
Abstract
Zearalenone (ZEA) is a mycotoxin with estrogenic activity whose main effect is to impair the reproductive systems of animals. It leads to reproductive disorders in livestock and thus causes serious losses to agriculture and animal husbandry. This study aims to examine whether ZEA induces toxicity in Leydig cells through the PI3K/AKT signaling pathway and also to investigate the role played by the upstream phosphatase and tensin homolog (PTEN) gene. An adenovirus vector model was constructed to interfere with the PTEN gene to investigate whether ZEA promotes the apoptosis of TM3 cells through the PI3K/AKT pathway. Apoptosis was detected cytometrically and the protein expression levels of PTEN, AKT, p-AKT, Bax, and Bcl-2 were evaluated via western blot analysis. The results show that ZEA induces apoptosis of TM3 cells. PTEN expression is significantly increased (P < 0.01), Bax expression is increased (P < 0.05), AKT and p-AKT expression of anti-apoptotic protein is significantly decreased (P < 0.01), and Bcl-2 protein expression is decreased (P < 0.05) in the ZEA group compared with the control group. In the shRNA+ZEA group, the expression levels of PTEN and Bax proteins are significantly decreased (P < 0.01), AKT protein is significantly increased (P < 0.01), and p-AKT protein is increased (P < 0.05) compared with the ZEA group. This study thus demonstrates that ZEA promotes apoptosis of TM3 cells by targeting PTEN and thus inhibiting the PI3K/AKT signal pathway.
Collapse
Affiliation(s)
- Jia Chen
- College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China
| | - Mingyang Wang
- College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China
| | - Hanli Wang
- College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China
| | - Miao Long
- College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China.
| |
Collapse
|
39
|
Li Y, Zhan Z, Yin X, Fu S, Deng X. Targeted Therapeutic Strategies for Triple-Negative Breast Cancer. Front Oncol 2021; 11:731535. [PMID: 34778045 PMCID: PMC8581040 DOI: 10.3389/fonc.2021.731535] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Accepted: 10/12/2021] [Indexed: 12/13/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer, which is characterized by the absence of estrogen receptor (ER) and progesterone receptor (PR) expression and the absence of human epidermal growth factor receptor 2 (HER2) expression/amplification. Conventional chemotherapy is the mainstay of systemic treatment for TNBC. However, lack of molecular targeted therapies and poor prognosis of TNBC patients have prompted a great effort to discover effective targets for improving the clinical outcomes. For now, poly (ADP-ribose) polymerase (PARP) inhibitors (PARPi's) and immune checkpoint inhibitors have been approved for the treatment of TNBC. Moreover, agents that target signal transduction, angiogenesis, epigenetic modifications, and cell cycle are under active preclinical or clinical investigations. In this review, we highlight the current major developments in targeted therapies of TNBC, with some descriptions about their (dis)advantages and future perspectives.
Collapse
Affiliation(s)
- Ying Li
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Department of Pathophysiology, Hunan Normal University School of Medicine, Changsha, China.,Key Laboratory of Translational Cancer Stem Cell Research, Hunan Normal University, Changsha, China
| | - Zhijun Zhan
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Department of Pathophysiology, Hunan Normal University School of Medicine, Changsha, China.,Key Laboratory of Translational Cancer Stem Cell Research, Hunan Normal University, Changsha, China
| | - Xuemin Yin
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Department of Pathophysiology, Hunan Normal University School of Medicine, Changsha, China.,Key Laboratory of Translational Cancer Stem Cell Research, Hunan Normal University, Changsha, China
| | - Shujun Fu
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Department of Pathophysiology, Hunan Normal University School of Medicine, Changsha, China.,Key Laboratory of Translational Cancer Stem Cell Research, Hunan Normal University, Changsha, China
| | - Xiyun Deng
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Department of Pathophysiology, Hunan Normal University School of Medicine, Changsha, China.,Key Laboratory of Translational Cancer Stem Cell Research, Hunan Normal University, Changsha, China
| |
Collapse
|
40
|
Van Herck Y, Feyaerts A, Alibhai S, Papamichael D, Decoster L, Lambrechts Y, Pinchuk M, Bechter O, Herrera-Caceres J, Bibeau F, Desmedt C, Hatse S, Wildiers H. Is cancer biology different in older patients? THE LANCET HEALTHY LONGEVITY 2021; 2:e663-e677. [DOI: 10.1016/s2666-7568(21)00179-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 07/14/2021] [Accepted: 07/15/2021] [Indexed: 12/13/2022]
|
41
|
Acharya S, Chatterjee S, Chaudhuri S, Singh MK, Bhattacharya D, Bhattacharjee M, Ghosh A, Chaudhuri S. Akt Phosphorylation Orchestrates T11TS Mediated Cell Cycle Arrest in Glioma Cells. Cancer Invest 2021; 39:854-870. [PMID: 34569407 DOI: 10.1080/07357907.2021.1986060] [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] [Indexed: 10/20/2022]
Abstract
The novel anti-neoplastic glycopeptide T11TS retards glioma both in in-vitro clinical samples and in-vivo models. This study investigates the correlation between altering the glioma microenvironment with glioma arrest and death. Flow cytometry, immunoblotting, ELISA, and co-immunoprecipitation were employed to investigate glioma cell arrest and death. Results include a decline in phosphorylation of Akt and attenuation of p21 phosphorylation (Thr145,Ser146) and disassociation of p-Akt-Mdm2 and p-Akt-BAD facilitating death by Akt>BAD. T11TS influence phosphorylation patterns in two focal axes Akt>p21 and Akt>Mdm2>p53. The current article provides crucial insight in deciphering the mechanism of T11TS induced glioma cell arrest and death.
Collapse
Affiliation(s)
- Sagar Acharya
- Department of Zoology, Vidyasagar University, Paschim Medinipur, Midnapore, India
| | | | | | - Manoj Kumar Singh
- Department of Laboratory Medicine, School of Tropical Medicine, Kolkata, India
| | - Debanjan Bhattacharya
- Department of Neurology and Rehabilitation Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | | | - Anirban Ghosh
- Department of Zoology, Netaji Subhas Open University, Kolkata, India
| | - Swapna Chaudhuri
- Department of Laboratory Medicine, School of Tropical Medicine, Kolkata, India.,Chittaranjan National Cancer Institute, Kolkata, India
| |
Collapse
|
42
|
Kou L, Wei S, Kou P. Current Progress and Perspectives on Using Gold Compounds for the Modulation of Tumor Cell Metabolism. Front Chem 2021; 9:733463. [PMID: 34434922 PMCID: PMC8382570 DOI: 10.3389/fchem.2021.733463] [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] [Received: 06/30/2021] [Accepted: 07/26/2021] [Indexed: 01/14/2023] Open
Abstract
Altered cellular metabolism, which is essential for the growth and survival of tumor cells in a specific microenvironment, is one of the hallmarks of cancer. Among the most significant changes in the metabolic pattern of tumor cells is the shift from oxidative phosphorylation to aerobic glycolysis for glucose utilization. Tumor cells also exhibit changes in patterns of protein and nucleic acid metabolism. Recently, gold compounds have been shown to target several metabolic pathways and a number of metabolites in tumor cells. In this review, we summarize how gold compounds modulate glucose, protein, and nucleic acid metabolism in tumor cells, resulting in anti-tumor effects. We also discuss the rationale underlying the anti-tumor effects of these gold compounds and highlight how to effectively utilize against various types of tumors.
Collapse
Affiliation(s)
- Leiya Kou
- The First Clinical College, Hubei University of Chinese Medicine, Wuhan, China
| | - Shuang Wei
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology, Wuhan, China
| | - Pei Kou
- The First Clinical College, Hubei University of Chinese Medicine, Wuhan, China.,Department of Medical Record, Wuhan No. 1 Hospital, Wuhan, China
| |
Collapse
|
43
|
Zoi V, Galani V, Lianos GD, Voulgaris S, Kyritsis AP, Alexiou GA. The Role of Curcumin in Cancer Treatment. Biomedicines 2021; 9:biomedicines9091086. [PMID: 34572272 PMCID: PMC8464730 DOI: 10.3390/biomedicines9091086] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 08/19/2021] [Accepted: 08/23/2021] [Indexed: 12/24/2022] Open
Abstract
Curcumin is a polyphenol extracted from the rhizomes of the turmeric plant, Curcuma longa which has anti-inflammatory, and anticancer properties. Chronic inflammation is associated with the development of cancer. Curcumin acts on the regulation of various immune modulators, including cytokines, cyclooxygenase-2 (COX-2), and reactive oxygen species (ROS), which partly explains its anticancer effects. It also takes part in the downregulation of growth factors, protein kinases, oncogenic molecules and various signaling pathways, such as nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), c-Jun N-terminal kinase (JNK) and signal transducer and activator of transcription 3 (STAT3) signaling. Clinical trials of curcumin have been completed or are ongoing for various types of cancer. This review presents the molecular mechanisms of curcumin in different types of cancer and the evidence from the most recent clinical trials.
Collapse
Affiliation(s)
- Vasiliki Zoi
- Neurosurgical Institute, School of Medicine, University of Ioannina, 45500 Ioannina, Greece; (V.Z.); (S.V.); (A.P.K.)
- Department of Anatomy Histology-Embryology, School of Medicine, University of Ioannina, 45500 Ioannina, Greece;
| | - Vasiliki Galani
- Department of Anatomy Histology-Embryology, School of Medicine, University of Ioannina, 45500 Ioannina, Greece;
| | - Georgios D. Lianos
- Department of Surgery, University Hospital of Ioannina, 45500 Ioannina, Greece;
| | - Spyridon Voulgaris
- Neurosurgical Institute, School of Medicine, University of Ioannina, 45500 Ioannina, Greece; (V.Z.); (S.V.); (A.P.K.)
- Department of Neurosurgery, School of Medicine Ioannina, University of Ioannina, 45500 Ioannina, Greece
| | - Athanasios P. Kyritsis
- Neurosurgical Institute, School of Medicine, University of Ioannina, 45500 Ioannina, Greece; (V.Z.); (S.V.); (A.P.K.)
| | - George A. Alexiou
- Neurosurgical Institute, School of Medicine, University of Ioannina, 45500 Ioannina, Greece; (V.Z.); (S.V.); (A.P.K.)
- Department of Neurosurgery, School of Medicine Ioannina, University of Ioannina, 45500 Ioannina, Greece
- Correspondence:
| |
Collapse
|
44
|
Ren L, Zhou H, Lei L, Zhang Y, Cai H, Wang X. Long non-coding RNA FOXD3 antisense RNA 1 augments anti-estrogen resistance in breast cancer cells through the microRNA-363/ trefoil factor 1/ phosphatidylinositol 3-kinase/protein kinase B axis. Bioengineered 2021; 12:5266-5278. [PMID: 34424807 PMCID: PMC8806484 DOI: 10.1080/21655979.2021.1962694] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Long non-coding RNA (lncRNA) FOXD3 antisense RNA 1 (FOXD3-AS1) has been reported to participate in multiple processes that contribute toward the development of cancer. The present study aimed to explore the effect of lncRNA FOXD3-AS1 on anti-estrogen resistance in breast cancer (BC) cells. FOXD3-AS1 was found to be highly expressed in BC cell lines. Moreover, FOXD3-AS1 was highly expressed in estrogen receptor-negative (ER-) cells compared to the ER-positive (ER+) cells. FOXD3-AS1 overexpression in T47D and MCF-7 (ER+) cells enhanced the resistance of cells to tamoxifen (TMX), whereas FOX3-AS1 downregulation reduced the TMX resistance in MDA-MB-231 (ER-) cells. Similar results were reproduced in vivo that FOXD3-AS1 inhibition reduced the growth of xenograft tumors formed by MDA-MB-231 cells following TMX treatment whereas FOXD3-AS1 overexpression in T47D cells facilitated tumor growth. The bioinformatic analysis and luciferase assays indicated that FOXD3-AS1 sponged microRNA-363 (miR-363) to restore expression of trefoil factor 1 (TFF1) mRNA. Overexpression of miR-363 reduced T47D cell proliferation induced by FOXD3-AS1, whereas overexpression of TFF1 restored growth of MDA-MB-231 cells reduced after FOXD3-AS1 silencing. The phosphorylation of phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) was increased by FOXD3-AS1 but attenuated by miR-363. Inhibition of PI3K/Akt blocked the role of FOXD3-AS1 and reduced the TMX resistance in T47D and MCF-7 cells. Taken together, the present study suggested that FOXD3-AS1 sponges miR-363 to upregulate TFF1 expression, leading to PI3K/Akt signaling activation and anti-estrogen resistance in BC cells.
Collapse
Affiliation(s)
- Lili Ren
- Department of Integration of Traditional Chinese and Western Medicine, the Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, P.R. China
| | - Huanhuan Zhou
- Department of Medical Oncology, the Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, P.R. China
| | - Lei Lei
- Department of Medical Oncology, the Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, P.R. China
| | - Yongjun Zhang
- Department of Integration of Traditional Chinese and Western Medicine, the Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, P.R. China
| | - Hu Cai
- Department of Integration of Traditional Chinese and Western Medicine, the Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, P.R. China
| | - Xiaojia Wang
- Department of Medical Oncology, the Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, P.R. China
| |
Collapse
|
45
|
Liu N, Wang X, Li X, Lv X, Xie H, Guo Z, Wang J, Dou G, Du Y, Song D. Identification of effective natural PIK3CA H1047R inhibitors by computational study. Aging (Albany NY) 2021; 13:20246-20257. [PMID: 34415239 PMCID: PMC8436935 DOI: 10.18632/aging.203409] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 08/03/2021] [Indexed: 12/20/2022]
Abstract
Triple-negative breast cancer (TNBC) is a highly aggressive subtype of breast cancer with a poor prognosis and a high recurrence rate. PIK3CA gene is frequently mutated in breast cancer, with PIK3CA H1047R as the hotspot mutation reported in TNBC. We used the ZINC database to screen natural compounds that could be structurally modified to develop drugs targeting the PIK3CA H1047R mutant protein in the PI3K pathway. The LibDock module showed that 2,749 compounds could strongly bind to the PIK3CA H1047R protein. Ultimately, the top 20 natural ligands with high LibDock scores were used for further analyses including assessment of ADME (absorption, distribution, metabolism, and excretion), toxicity, stability, and binding affinity. ZINC000004098448 and ZINC000014715656 were selected as the safest drug candidates with strong binding affinity to PIK3CA H1047R, no hepatotoxicity, less carcinogenicity, better plasma protein binding (PPB) properties, and enhanced intestinal permeability and absorption than the two reference drugs, PKI-402 and wortmannin. Moreover, their lower potential energies than those of PIK3CA H1047R confirmed the stability of the ligand-receptor complex under physiological conditions. ZINC000004098448 and ZINC000014715656 are thus safe and stable leads for designing drugs against PIK3CA H1047R as part of a targeted therapeutic approach for patients with TNBC.
Collapse
Affiliation(s)
- Naimeng Liu
- Department of Breast Surgery, The First Hospital of Jilin University, Changchun, China
| | - Xinhui Wang
- Department of Oncology, First People's Hospital of Xinxiang, Xinxiang, China
| | - Xuan Li
- Department of Obstetrics and Gynecology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Xiaye Lv
- Department of Hematology, Honghui Hospital, Xi'an Jiao Tong University, Xi'an, China
| | - Haoqun Xie
- Clinical College, Jilin University, Changchun, China
| | - Zhen Guo
- Clinical College, Jilin University, Changchun, China
| | - Jing Wang
- Clinical College, Jilin University, Changchun, China
| | - Gaojing Dou
- Department of Breast Surgery, The First Hospital of Jilin University, Changchun, China.,Clinical College, Jilin University, Changchun, China
| | - Ye Du
- Department of Breast Surgery, The First Hospital of Jilin University, Changchun, China
| | - Dong Song
- Department of Breast Surgery, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
46
|
Jiang Q, Zheng N, Bu L, Zhang X, Zhang X, Wu Y, Su Y, Wang L, Zhang X, Ren S, Dai X, Wu D, Xie W, Wei W, Zhu Y, Guo J. SPOP-mediated ubiquitination and degradation of PDK1 suppresses AKT kinase activity and oncogenic functions. Mol Cancer 2021; 20:100. [PMID: 34353330 PMCID: PMC8340461 DOI: 10.1186/s12943-021-01397-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 07/24/2021] [Indexed: 12/25/2022] Open
Abstract
Background 3-phosphoinositide-dependent protein kinase-1 (PDK1) acts as a master kinase of protein kinase A, G, and C family (AGC) kinase to predominantly govern cell survival, proliferation, and metabolic homeostasis. Although the regulations to PDK1 downstream substrates such as protein kinase B (AKT) and ribosomal protein S6 kinase beta (S6K) have been well established, the upstream regulators of PDK1, especially its degrader, has not been defined yet. Method A clustered regularly interspaced short palindromic repeats (CRISPR)-based E3 ligase screening approach was employed to identify the E3 ubiquitin ligase for degrading PDK1. Western blotting, immunoprecipitation assays and immunofluorescence (IF) staining were performed to detect the interaction or location of PDK1 with speckle-type POZ protein (SPOP). Immunohistochemistry (IHC) staining was used to study the expression of PDK1 and SPOP in prostate cancer tissues. In vivo and in vitro ubiquitination assays were performed to measure the ubiquitination conjugation of PDK1 by SPOP. In vitro kinase assays and mass spectrometry approach were carried out to identify casein kinase 1 (CK1) and glycogen synthase kinase 3 (GSK3)-mediated PDK1 phosphorylation. The biological effects of PDK1 mutations and correlation with SPOP mutations were performed with colony formation, soft agar assays and in vivo xenograft mouse models. Results We identified that PDK1 underwent SPOP-mediated ubiquitination and subsequent proteasome-dependent degradation. Specifically, SPOP directly bound PDK1 by the consensus degron in a CK1/GSK3β-mediated phosphorylation dependent manner. Pathologically, prostate cancer patients associated mutations of SPOP impaired PDK1 degradation and thus activated the AKT kinase, resulting in tumor malignancies. Meanwhile, mutations that occurred around or within the PDK1 degron, by either blocking SPOP to bind the degron or inhibiting CK1 or GSK3β-mediated PDK1 phosphorylation, could markedly evade SPOP-mediated PDK1 degradation, and played potently oncogenic roles via activating the AKT kinase. Conclusions Our results not only reveal a physiological regulation of PDK1 by E3 ligase SPOP, but also highlight the oncogenic roles of loss-of-function mutations of SPOP or gain-of-function mutations of PDK1 in tumorigenesis through activating the AKT kinase. Supplementary Information The online version contains supplementary material available at 10.1186/s12943-021-01397-5.
Collapse
Affiliation(s)
- Qiwei Jiang
- Institute of Precision Medicine, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510275, Guangdong, China
| | - Nana Zheng
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Lang Bu
- Institute of Precision Medicine, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510275, Guangdong, China
| | - Xiaomei Zhang
- Institute of Precision Medicine, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510275, Guangdong, China
| | - Xiaoling Zhang
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Jilin, Changchun, China.,National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Jilin, Changchun, China
| | - Yuanzhong Wu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, Guangdong, China
| | - Yaqing Su
- Institute of Precision Medicine, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510275, Guangdong, China
| | - Lei Wang
- Institute of Precision Medicine, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510275, Guangdong, China
| | - Xiaomin Zhang
- Department of Urology, Shanghai Changhai Hospital, Shanghai, 200433, China
| | - Shancheng Ren
- Department of Urology, Shanghai Changhai Hospital, Shanghai, 200433, China
| | - Xiangpeng Dai
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Jilin, Changchun, China.,National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Jilin, Changchun, China
| | - Depei Wu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Wei Xie
- Institute of Precision Medicine, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510275, Guangdong, China
| | - Wenyi Wei
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA.
| | - Yasheng Zhu
- Department of Urology, Shanghai Changhai Hospital, Shanghai, 200433, China.
| | - Jianping Guo
- Institute of Precision Medicine, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510275, Guangdong, China.
| |
Collapse
|
47
|
Mir H, Kapur N, Gales DN, Sharma PK, Oprea-Ilies G, Johnson AT, Singh R, Singh S. CXCR6-CXCL16 Axis Promotes Breast Cancer by Inducing Oncogenic Signaling. Cancers (Basel) 2021; 13:cancers13143568. [PMID: 34298782 PMCID: PMC8306453 DOI: 10.3390/cancers13143568] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 07/12/2021] [Accepted: 07/14/2021] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Breast cancer (BrCa) is the second leading cause of cancer-related deaths in American women, and its incidence is on the rise. Insufficient understanding of the mechanisms leading to BrCa limits the effectiveness of the treatment. In this article, we show the importance of a chemokine axis-CXCR6/CXCL16 in supporting BrCa progression. We have delineated BrCa-promoting mechanisms induced by this chemokine axis at the molecular level. This work projects the therapeutic significance of CXCR6/CXCL16 signaling for the treatment of BrCa. Abstract Precise mechanisms underlying breast cancer (BrCa) metastasis are undefined, which becomes a challenge for effective treatments. Chemokine signaling instigates the trafficking of cancer cells in addition to leukocytes. This study aimed to ascertain the clinical and biological significance of the CXCR6/CXCL16 signaling axis in the pathobiology of BrCa. Our data show a higher expression of CXCR6 in BrCa cell lines and tissues. Stage-III BrCa tissues express significantly higher CXCR6 compared to stage-II tissues. The ligand, CXCL16, could remain tethered to the cell surface, and, after proteolytic shedding of the ectodomain, the N-terminal fragment is released, converting it to its oncogenic, soluble form. Like CXCR6, N-terminal CXCL16 and ADAM-10 were significantly higher in stage-III than stage-II, but no significant difference was observed in the C-terminal fragment of CXCL16. Further, stimulation of the CXCR6/CXCL16 axis activated Src, FAK, ERK1/2, and PI3K signaling pathways, as per antibody microarray analysis, which also underlie CXCL16-induced F-actin polymerization. The CXCR6/CXCL16 axis induces cytoskeleton rearrangement facilitating migration and invasion and supports BrCa cell survival by activating the PI3K/Akt pathway. This study highlights the significance of the CXCR6/CXCL16 axis and ADAM10 as potential therapeutic targets for advanced-stage BrCa.
Collapse
Affiliation(s)
- Hina Mir
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA 30310, USA; (H.M.); (N.K.); (D.N.G.); (R.S.)
- Cancer Health Equity Institute, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Neeraj Kapur
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA 30310, USA; (H.M.); (N.K.); (D.N.G.); (R.S.)
- Cancer Health Equity Institute, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Dominique N. Gales
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA 30310, USA; (H.M.); (N.K.); (D.N.G.); (R.S.)
- Cancer Health Equity Institute, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Praveen K. Sharma
- Centre for Life Sciences, Central University of Jharkhand, Jharkhand 835205, India;
| | - Gabriela Oprea-Ilies
- Department of Pathology & Laboratory Medicine, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA;
| | - Anita T. Johnson
- Comprehensive Cancer Care Network, Cancer Treatment Center of America, Atlanta, GA 30265, USA;
| | - Rajesh Singh
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA 30310, USA; (H.M.); (N.K.); (D.N.G.); (R.S.)
- Cancer Health Equity Institute, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Shailesh Singh
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA 30310, USA; (H.M.); (N.K.); (D.N.G.); (R.S.)
- Cancer Health Equity Institute, Morehouse School of Medicine, Atlanta, GA 30310, USA
- Cell and Molecular Biology Program, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
- Correspondence: ; Tel.: +1-404-756-5718
| |
Collapse
|
48
|
Liegmann AS, Heselmeyer-Haddad K, Lischka A, Hirsch D, Chen WD, Torres I, Gemoll T, Rody A, Thorns C, Gertz EM, Alkemade H, Hu Y, Habermann JK, Ried T. Single Cell Genetic Profiling of Tumors of Breast Cancer Patients Aged 50 Years and Older Reveals Enormous Intratumor Heterogeneity Independent of Individual Prognosis. Cancers (Basel) 2021; 13:3366. [PMID: 34282768 PMCID: PMC8267950 DOI: 10.3390/cancers13133366] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/25/2021] [Accepted: 06/30/2021] [Indexed: 02/07/2023] Open
Abstract
PURPOSE Older breast cancer patients are underrepresented in cancer research even though the majority (81.4%) of women dying of breast cancer are 55 years and older. Here we study a common phenomenon observed in breast cancer which is a large inter- and intratumor heterogeneity; this poses a tremendous clinical challenge, for example with respect to treatment stratification. To further elucidate genomic instability and tumor heterogeneity in older patients, we analyzed the genetic aberration profiles of 39 breast cancer patients aged 50 years and older (median 67 years) with either short (median 2.4 years) or long survival (median 19 years). The analysis was based on copy number enumeration of eight breast cancer-associated genes using multiplex interphase fluorescence in situ hybridization (miFISH) of single cells, and by targeted next-generation sequencing of 563 cancer-related genes. RESULTS We detected enormous inter- and intratumor heterogeneity, yet maintenance of common cancer gene mutations and breast cancer specific chromosomal gains and losses. The gain of COX2 was most common (72%), followed by MYC (69%); losses were most prevalent for CDH1 (74%) and TP53 (69%). The degree of intratumor heterogeneity did not correlate with disease outcome. Comparing the miFISH results of diploid with aneuploid tumor samples significant differences were found: aneuploid tumors showed significantly higher average signal numbers, copy number alterations (CNAs) and instability indices. Mutations in PIKC3A were mostly restricted to luminal A tumors. Furthermore, a significant co-occurrence of CNAs of DBC2/MYC, HER2/DBC2 and HER2/TP53 and mutual exclusivity of CNAs of HER2 and PIK3CA mutations and CNAs of CCND1 and PIK3CA mutations were revealed. CONCLUSION Our results provide a comprehensive picture of genome instability profiles with a large variety of inter- and intratumor heterogeneity in breast cancer patients aged 50 years and older. In most cases, the distribution of chromosomal aneuploidies was consistent with previous results; however, striking exceptions, such as tumors driven by exclusive loss of chromosomes, were identified.
Collapse
Affiliation(s)
- Anna-Sophie Liegmann
- Section of Translational Surgical Oncology and Biobanking, Department of Surgery, University of Lübeck and University Medical Center Schleswig-Holstein, 23562 Lübeck, Germany; (A.-S.L.); (A.L.); (T.G.); (H.A.)
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (K.H.-H.); (D.H.); (W.-D.C.); (I.T.); (Y.H.)
| | - Kerstin Heselmeyer-Haddad
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (K.H.-H.); (D.H.); (W.-D.C.); (I.T.); (Y.H.)
| | - Annette Lischka
- Section of Translational Surgical Oncology and Biobanking, Department of Surgery, University of Lübeck and University Medical Center Schleswig-Holstein, 23562 Lübeck, Germany; (A.-S.L.); (A.L.); (T.G.); (H.A.)
| | - Daniela Hirsch
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (K.H.-H.); (D.H.); (W.-D.C.); (I.T.); (Y.H.)
- Institute of Pathology, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Wei-Dong Chen
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (K.H.-H.); (D.H.); (W.-D.C.); (I.T.); (Y.H.)
| | - Irianna Torres
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (K.H.-H.); (D.H.); (W.-D.C.); (I.T.); (Y.H.)
| | - Timo Gemoll
- Section of Translational Surgical Oncology and Biobanking, Department of Surgery, University of Lübeck and University Medical Center Schleswig-Holstein, 23562 Lübeck, Germany; (A.-S.L.); (A.L.); (T.G.); (H.A.)
| | - Achim Rody
- Department of Gynecology and Obstetrics, Campus Lübeck, University Hospital of Schleswig-Holstein, 23562 Lübeck, Germany;
| | - Christoph Thorns
- Institute of Pathology, Marienkrankenhaus Hamburg, 22087 Hamburg, Germany;
- Institute of Pathology, University of Lübeck and University Medical Center Schleswig-Holstein, 23562 Lübeck, Germany
| | - Edward Michael Gertz
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA;
| | - Hendrik Alkemade
- Section of Translational Surgical Oncology and Biobanking, Department of Surgery, University of Lübeck and University Medical Center Schleswig-Holstein, 23562 Lübeck, Germany; (A.-S.L.); (A.L.); (T.G.); (H.A.)
| | - Yue Hu
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (K.H.-H.); (D.H.); (W.-D.C.); (I.T.); (Y.H.)
| | - Jens K. Habermann
- Section of Translational Surgical Oncology and Biobanking, Department of Surgery, University of Lübeck and University Medical Center Schleswig-Holstein, 23562 Lübeck, Germany; (A.-S.L.); (A.L.); (T.G.); (H.A.)
- Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska Institute, 171 77 Stockholm, Sweden
| | - Thomas Ried
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (K.H.-H.); (D.H.); (W.-D.C.); (I.T.); (Y.H.)
| |
Collapse
|
49
|
Albogami SM, Asiri Y, Asiri A, Alnefaie AA, Alnefaie S. Effects of neoadjuvant therapies on genetic regulation of targeted pathways in ER+ primary ductal breast carcinoma: A meta-analysis of microarray datasets. Saudi Pharm J 2021; 29:656-669. [PMID: 34400859 PMCID: PMC8347676 DOI: 10.1016/j.jsps.2021.04.027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 04/24/2021] [Indexed: 12/11/2022] Open
Abstract
Breast cancer arises as a result of multiple interactions between environmental and genetic factors. Conventionally, breast cancer is treated based on histopathological and clinical features. DNA technologies like the human genome microarray are now partially integrated into clinical practice and are used for developing new "personalized medicines" and "pharmacogenetics" for improving the efficiency and safety of cancer medications. We investigated the effects of four established therapies-for ER+ ductal breast cancer-on the differential gene expression. The therapies included single agent tamoxifen, two-agent docetaxel and capecitabine, or combined three-agents CAF (cyclophosphamide, doxorubicin, and fluorouracil) and CMF (cyclophosphamide, methotrexate, and fluorouracil). Genevestigator 8.1.0 was used to compare five datasets from patients with infiltrating ductal carcinoma, untreated or treated with selected drugs, to those from the healthy control. We identified 74 differentially expressed genes involved in three pathways, i.e., apoptosis (extrinsic and intrinsic), oxidative signaling, and PI3K/Akt signaling. The treatments affected the expression of apoptotic genes (TNFRSF10B [TRAIL], FAS, CASP3/6/7/8, PMAIP1 [NOXA], BNIP3L, BNIP3, BCL2A1, and BCL2), the oxidative stress-related genes (NOX4, XDH, MAOA, GSR, GPX3, and SOD3), and the PI3K/Akt pathway gene (ERBB2 [HER2]). Breast cancer treatments are complex with varying drug responses and efficacy among patients. This necessitates identifying novel biomarkers for predicting the drug response, using available data and new technologies. GSR, NOX4, CASP3, and ERBB2 are potential biomarkers for predicting the treatment response in primary ER+ ductal breast carcinoma.
Collapse
Key Words
- BC, breast cancer
- Bax, Bcl-2-associated X
- Bcl2, B-cell lymphoma 2
- CAF, cyclophosphamide, doxorubicin, and fluorouracil
- CASP3
- CMF, cyclophosphamide, methotrexate, and fluorouracil
- Chemotherapy
- DC, docetaxel and capecitabine
- ER+ ductal carcinoma
- ER, estrogen receptor
- ERBB2 (HER2)
- FC, fold-change
- FU, fluorouracil
- GSR
- H2O2, hydrogen peroxide
- HER2, human epidermal growth factor 2
- IGF-1, insulin-like growth factor-1
- NOX4
- OH●, hydroxyl radical
- PI3K/Akt, phosphatidylinositol 3-kinase/protein kinase B
- PM, personalized medicine
- PR, progesterone receptor
- PRISMA, Preferred Reporting Items for Systematic Reviews and Meta-Analyses
- ROS, reactive oxygen species
- TGF-α/β, transforming growth factor alpha/beta
- TMX, tamoxifen
- TS, thymidylate synthase
Collapse
Affiliation(s)
- Sarah M. Albogami
- Department of Biotechnology, College of Science, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Yousif Asiri
- Department of Clinical Pharmacy, College of Pharmacy, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Abdulaziz Asiri
- Pharmaceutical Care Division, King Faisal Specialist Hospital and Research Center, P.O. Box 3354, MBC#11, Riyadh 11211, Saudi Arabia
| | - Alaa A. Alnefaie
- International Medical Center Hospital, P.O. Box 953, Jeddah 21423, Saudi Arabia
| | - Sahar Alnefaie
- Department of Surgery, College of Medicine, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| |
Collapse
|
50
|
Li X, Zhou J, Xiao M, Zhao L, Zhao Y, Wang S, Gao S, Zhuang Y, Niu Y, Li S, Li X, Zhu Y, Zhang M, Tang J. Uncovering the Subtype-Specific Molecular Characteristics of Breast Cancer by Multiomics Analysis of Prognosis-Associated Genes, Driver Genes, Signaling Pathways, and Immune Activity. Front Cell Dev Biol 2021; 9:689028. [PMID: 34277633 PMCID: PMC8280810 DOI: 10.3389/fcell.2021.689028] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 05/28/2021] [Indexed: 01/04/2023] Open
Abstract
Breast cancer is a heterogeneous malignant disease with different prognoses and has been divided into four molecular subtypes. It is believed that molecular events occurring in breast stem/progenitor cells contribute to the carcinogenesis and development of different breast cancer subtypes. However, these subtype-specific molecular characteristics are largely unknown. In this study, we employed 1217 breast cancer samples from The Cancer Genome Atlas (TCGA) database for a multiomics analysis of the molecular characteristics of different breast cancer subtypes based on PAM50 algorithms. We detected the expression changes of subtype-specific genes and revealed that the expression of particular subtype-specific genes significantly affected prognosis. We also investigated the mutations and copy number variations (CNVs) of breast cancer driver genes and the representative genes of ten signaling pathways in different subtypes and revealed several subtype-specifically altered genes. Moreover, we detected the infiltration of various immune cells in different subtypes of breast cancer and showed that the infiltration levels of major immune cell types are different among these subtypes. Additionally, we investigated the factors affecting the immune infiltration level and the immune cytolytic activity in different breast cancer subtypes, namely, the mutation burden, genome instability and cancer-associated fibroblast (CAF) infiltration. This study may shed light on the molecular events contributing to carcinogenesis and development and provide potential markers and targets for the clinical diagnosis and treatment of different breast cancer subtypes.
Collapse
Affiliation(s)
- Xinhui Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Jian Zhou
- School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Mingming Xiao
- Department of Pathology, The People's Hospital of Liaoning Province, Shenyang, China
| | - Lingyu Zhao
- Department of Pathology, Harbin Medical University, Harbin, China
| | - Yan Zhao
- Department of Oncology, Chifeng City Hospital, Chifeng, China
| | - Shuoshuo Wang
- Department of Pathology, Harbin Medical University, Harbin, China
| | - Shuangshu Gao
- Department of Pathology, Harbin Medical University, Harbin, China
| | - Yuan Zhuang
- Department of Pathology, Harbin Medical University, Harbin, China
| | - Yi Niu
- Department of Oncology, Chifeng City Hospital, Chifeng, China
| | - Shijun Li
- Department of Pathology, Chifeng City Hospital, Chifeng, China
| | - Xiaobo Li
- Department of Pathology, Harbin Medical University, Harbin, China
| | - Yuanyuan Zhu
- Department of Pathology, Harbin Medical University, Harbin, China
| | - Minghui Zhang
- Department of Oncology, Chifeng City Hospital, Chifeng, China
| | - Jing Tang
- Department of Pathology, Harbin Medical University, Harbin, China
| |
Collapse
|