1
|
Kyuno D, Asano H, Okumura R, Takasawa K, Takasawa A, Konno T, Nakamori Y, Magara K, Ono Y, Imamura M, Kimura Y, Kojima T, Osanai M. The Role of Claudin-1 in Enhancing Pancreatic Cancer Aggressiveness and Drug Resistance via Metabolic Pathway Modulation. Cancers (Basel) 2025; 17:1469. [PMID: 40361399 DOI: 10.3390/cancers17091469] [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: 04/08/2025] [Revised: 04/17/2025] [Accepted: 04/24/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND/OBJECTIVES Pancreatic ductal adenocarcinoma is a lethal malignancy, necessitating an understanding of its molecular mechanisms for the development of new therapeutic strategies. The tight junction protein claudin-1, known to influence cellular functions in various cancers and is considered a therapeutic target, remains unclear in pancreatic cancer. METHODS This study assessed claudin-1 expression in resected pancreatic cancer samples, public databases, and pancreatic cancer cell lines. Claudin-1 knockout with CRISPR/Cas9 on poorly differentiated pancreatic cancer cell lines and a proteome analysis were performed to investigate the intracellular mechanisms of claudin-1. RESULTS Claudin-1 was markedly overexpressed in pancreatic ductal adenocarcinoma and intraepithelial neoplasia compared to normal ducts, and high claudin-1 levels were an independent predictor of poor prognosis. Claudin-1 knockout diminished cell proliferation, migration, invasion, and chemoresistance in pancreatic ductal adenocarcinoma. Proteome analysis revealed the significant downregulation of aldo-keto reductase family proteins (AKR1C2, AKR1C3, and AKR1B1) in claudin-1 knockout cells, which are linked to metabolic pathways. Aldo-keto reductase knockdown reduced chemoresistance, proliferation, and invasion in these cell lines. CONCLUSIONS These findings indicate that the abnormal expression of claudin-1 promotes tumor progression and drug resistance through its interaction with aldo-keto reductase proteins, highlighting claudin-1 and aldo-keto reductase family proteins as potential biomarkers and therapeutic targets for pancreatic cancer.
Collapse
Affiliation(s)
- Daisuke Kyuno
- Department of Pathology, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
- Department of Surgery, Division of Gastroenterological Surgery, Sapporo Medical University, Sapporo 060-8556, Japan
| | - Hinae Asano
- Department of Pathology, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| | - Reona Okumura
- Department of Pathology, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| | - Kumi Takasawa
- Division of Tumor Pathology, Department of Pathology, Asahikawa Medical University, Asahikawa 078-8510, Japan
| | - Akira Takasawa
- Division of Tumor Pathology, Department of Pathology, Asahikawa Medical University, Asahikawa 078-8510, Japan
| | - Takumi Konno
- Department of Cell Science, Institute of Cancer Research, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| | - Yuna Nakamori
- Department of Pathology, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| | - Kazufumi Magara
- Department of Pathology, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| | - Yusuke Ono
- Department of Pathology, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| | - Masafumi Imamura
- Department of Surgery, Division of Gastroenterological Surgery, Sapporo Medical University, Sapporo 060-8556, Japan
| | - Yasutoshi Kimura
- Department of Surgery, Division of Gastroenterological Surgery, Sapporo Medical University, Sapporo 060-8556, Japan
| | - Takashi Kojima
- Department of Cell Science, Institute of Cancer Research, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| | - Makoto Osanai
- Department of Pathology, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| |
Collapse
|
2
|
Yuan H, Zhang Y, Liu F, Wu Y, Huang X, Liu X, Jiang L, Xiao B, Zhu Y, Chen Q, Wu P, Jiang K. Exploring the biological mechanism and clinical value of perineural invasion in pancreatic cancer. Cancer Lett 2025; 613:217515. [PMID: 39892698 DOI: 10.1016/j.canlet.2025.217515] [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: 10/28/2024] [Revised: 01/30/2025] [Accepted: 01/30/2025] [Indexed: 02/04/2025]
Abstract
Pancreatic cancer (PC) is an extremely aggressive malignancy, with a 5-year survival rate of only 13 %. Perineural invasion (PNI) is a hallmark pathological feature of PC and is observed in almost all cases. Accordingly, PC ranks highly among solid tumors in terms of PNI incidence. The interaction between PC and the nervous system plays a pivotal role in tumor growth and metastasis. In PC, PNI is a key driver of local tumor progression, distant metastasis, and poor prognosis. Clarification of tumor-nerve crosstalk and the underlying molecular mechanisms is needed to facilitate the development of new therapeutic strategies to slow PC progression and alleviate PNI-associated symptoms. In this review, we present a comprehensive overview of the manifestations and characteristics of PNI in PC, summarize the molecular networks that regulate PNI, examine the relationship between PNI and the tumor microenvironment, and discuss the current research challenges and future directions in this critical area.
Collapse
Affiliation(s)
- Hao Yuan
- Pancreas Centre, First Affiliated Hospital, Nanjing Medical University, Nanjing, China; Pancreas Institute, Nanjing Medical University, Nanjing, China
| | - Yufeng Zhang
- Pancreas Centre, First Affiliated Hospital, Nanjing Medical University, Nanjing, China; Pancreas Institute, Nanjing Medical University, Nanjing, China
| | - Fengyuan Liu
- Pancreas Centre, First Affiliated Hospital, Nanjing Medical University, Nanjing, China; Pancreas Institute, Nanjing Medical University, Nanjing, China
| | - Yang Wu
- Pancreas Centre, First Affiliated Hospital, Nanjing Medical University, Nanjing, China; Pancreas Institute, Nanjing Medical University, Nanjing, China
| | - Xumin Huang
- Pancreas Centre, First Affiliated Hospital, Nanjing Medical University, Nanjing, China; Pancreas Institute, Nanjing Medical University, Nanjing, China
| | - Xinjian Liu
- Department of Pathogen Biology, Nanjing Medical University, Nanjing, China
| | - Luyang Jiang
- Pancreas Centre, First Affiliated Hospital, Nanjing Medical University, Nanjing, China; Pancreas Institute, Nanjing Medical University, Nanjing, China
| | - Bin Xiao
- Pancreas Centre, First Affiliated Hospital, Nanjing Medical University, Nanjing, China; Pancreas Institute, Nanjing Medical University, Nanjing, China
| | - Yi Zhu
- Pancreas Centre, First Affiliated Hospital, Nanjing Medical University, Nanjing, China; Pancreas Institute, Nanjing Medical University, Nanjing, China; Department of General Surgery, First Affiliated Hospital, Nanjing Medical University, Nanjing, China.
| | - Qun Chen
- Pancreas Centre, First Affiliated Hospital, Nanjing Medical University, Nanjing, China; Pancreas Institute, Nanjing Medical University, Nanjing, China.
| | - Pengfei Wu
- Pancreas Centre, First Affiliated Hospital, Nanjing Medical University, Nanjing, China; Pancreas Institute, Nanjing Medical University, Nanjing, China.
| | - Kuirong Jiang
- Pancreas Centre, First Affiliated Hospital, Nanjing Medical University, Nanjing, China; Pancreas Institute, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
3
|
Lin X, He J, Liu F, Li L, Sun L, Niu L, Xi H, Zhan Y, Liu X, Hu P. β‑adrenergic receptor activation promotes the proliferation of HepG2 cells via the ERK1/2/CREB pathways. Oncol Lett 2023; 26:519. [PMID: 37927415 PMCID: PMC10623085 DOI: 10.3892/ol.2023.14106] [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: 02/15/2023] [Accepted: 09/12/2023] [Indexed: 11/07/2023] Open
Abstract
Primary liver cancer is one of the most frequently diagnosed malignant tumors seen in clinics, and typically exhibits aggressive invasive behaviors, a poor prognosis, and is associated with high mortality rates. Long-term stress exposure causes norepinephrine (NE) release and activates the β-Adrenergic receptor (β-AR), which in turn exacerbates the occurrence and development of different types of cancers; however, the molecular mechanisms of β-AR in liver cancer are not fully understood. In the present study, reverse transcription (RT)-PCR and RT-quantitative PCR showed that β-AR expression was upregulated in human liver cancer cells (HepG2) compared with normal liver cells (LO2). Moreover, NE treatment promoted the growth of HepG2 cells, which could be blocked by propranolol, a β-AR antagonist. Notably, NE had no significant effect on the migration and epithelial-mesenchymal transition in HepG2 cells. Further experiments revealed that NE increased the phosphorylation levels of the extracellular signal-regulated kinase 1/2 (ERK1/2) and cyclic adenosine monophosphate response element-binding protein (CREB), while inhibition of ERK1/2 and CREB activation significantly blocked NE-induced cell proliferation. In summary, the findings of the present study suggested that β-adrenergic receptor activation promoted the proliferation of HepG2 cells through ERK1/2/CREB signaling pathways.
Collapse
Affiliation(s)
- Xingcheng Lin
- Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi 330001, P.R. China
| | - Jingjing He
- Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi 330001, P.R. China
| | - Fuhong Liu
- Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi 330001, P.R. China
| | - Lehui Li
- Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi 330001, P.R. China
| | - Longhua Sun
- Department of Respiratory, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Liyan Niu
- Huan Kui College, Nanchang University, Nanchang, Jiangxi 330001, P.R. China
| | - Haolin Xi
- Queen Mary School, Nanchang University, Nanchang, Jiangxi 330001, P.R. China
| | - Yuan Zhan
- Department of Pathology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Xiaohua Liu
- Department of Nursing, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Ping Hu
- Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi 330001, P.R. China
| |
Collapse
|
4
|
Zhao Z, Hao Z, Zhang Z, Zhan X. Bioinformatics Analysis Reveals the Vital Role of AKR1B1 in Immune Infiltration and Clinical Outcomes of Gastric Cancer. DNA Cell Biol 2023. [PMID: 37285280 DOI: 10.1089/dna.2022.0644] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023] Open
Abstract
Infiltrated immune cells are an important constitute of tumor microenvironment, which exert complex effects on gastric cancer (GC) pathogenesis and progression. By using weighted gene co-expression network analysis, integrating the data from The Cancer Genome Atlas-stomach adenocarcinoma and GSE62254, we identify Aldo-Keto Reductase Family 1 Member B (AKR1B1) as a hub gene for immune regulation in GC. Notably, AKR1B1 is associated with higher immune infiltration and worse histologic grade of GC. In addition, AKR1B1 is an independent factor for predicting the survival rate of GC patients. In vitro experiments further demonstrated that AKR1B1-overexpressed THP-1-derived macrophages promoted the proliferation and migration of GC cells. Taken together, AKR1B1 plays an important role in GC progression by regulating immune microenvironment, which could be a biomarker for predicting GC prognosis as well as a potential therapeutic target for GC treatment.
Collapse
Affiliation(s)
- Zhiyue Zhao
- Department of Oncology, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai, China
- Department of Oncology, Jinling Hospital, Medical School of Nanjing University, Nanjing University, Nanjing, China
| | - Zhibin Hao
- Department of Oncology, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai, China
| | - Zheng Zhang
- Department of Orthopedics, Changzheng Hospital, Second Military Medical University (Naval Medical University), Shanghai, China
- Department of Orthopedic Rehabilitation, Qingdao Special Servicemen Recuperation Center of PLA Navy, Qingdao, China
| | - Xianbao Zhan
- Department of Oncology, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai, China
| |
Collapse
|
5
|
Emerging Roles of the Nervous System in Gastrointestinal Cancer Development. Cancers (Basel) 2022; 14:cancers14153722. [PMID: 35954387 PMCID: PMC9367305 DOI: 10.3390/cancers14153722] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/23/2022] [Accepted: 07/27/2022] [Indexed: 12/10/2022] Open
Abstract
Simple Summary Nerve–cancer cross-talk has increasingly become a focus of the oncology field, particularly in gastrointestinal (GI) cancers. The indispensable roles of the nervous system in GI tumorigenesis and malignancy have been dissected by epidemiological, experimental animal and mechanistic data. Herein, we review and integrate recent discoveries linking the nervous system to GI cancer initiation and progression, and focus on the molecular mechanisms by which nerves and neural receptor pathways drive GI malignancy. Abstract Our understanding of the fascinating connection between nervous system and gastrointestinal (GI) tumorigenesis has expanded greatly in recent years. Recent studies revealed that neurogenesis plays an active part in GI tumor initiation and progression. Tumor-driven neurogenesis, as well as neurite outgrowth of the pre-existing peripheral nervous system (PNS), may fuel GI tumor progression via facilitating cancer cell proliferation, chemoresistance, invasion and immune escape. Neurotransmitters and neuropeptides drive the activation of various oncogenic pathways downstream of neural receptors within cancer cells, underscoring the importance of neural signaling pathways in GI tumor malignancy. In addition, neural infiltration also plays an integral role in tumor microenvironments, and contributes to an environment in favor of tumor angiogenesis, immune evasion and invasion. Blockade of tumor innervation via denervation or pharmacological agents may serve as a promising therapeutic strategy against GI tumors. In this review, we summarize recent findings linking the nervous system to GI tumor progression, set the spotlight on the molecular mechanisms by which neural signaling fuels cancer aggressiveness, and highlight the importance of targeting neural mechanisms in GI tumor therapy.
Collapse
|
6
|
Ketavarapu V, Ravikanth V, Sasikala M, Rao GV, Devi CV, Sripadi P, Bethu MS, Amanchy R, Murthy HVV, Pandol SJ, Reddy DN. Integration of metabolites from meta-analysis with transcriptome reveals enhanced SPHK1 in PDAC with a background of pancreatitis. BMC Cancer 2022; 22:792. [PMID: 35854233 PMCID: PMC9295503 DOI: 10.1186/s12885-022-09816-6] [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: 03/23/2022] [Accepted: 06/22/2022] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Pathophysiology of transformation of inflammatory lesions in chronic pancreatitis (CP) to pancreatic ductal adenocarcinoma (PDAC) is not clear. METHODS We conducted a systematic review, meta-analysis of circulating metabolites, integrated this data with transcriptome analysis of human pancreatic tissues and validated using immunohistochemistry. Our aim was to establish biomarker signatures for early malignant transformation in patients with underlying CP and identify therapeutic targets. RESULTS Analysis of 19 studies revealed AUC of 0.86 (95% CI 0.81-0.91, P < 0.0001) for all the altered metabolites (n = 88). Among them, lipids showed higher differentiating efficacy between PDAC and CP; P-value (< 0.0001). Pathway enrichment analysis identified sphingomyelin metabolism (impact value-0.29, FDR of 0.45) and TCA cycle (impact value-0.18, FDR of 0.06) to be prominent pathways in differentiating PDAC from CP. Mapping circulating metabolites to corresponding genes revealed 517 altered genes. Integration of these genes with transcriptome data of CP and PDAC with a background of CP (PDAC-CP) identified three upregulated genes; PIGC, PPIB, PKM and three downregulated genes; AZGP1, EGLN1, GNMT. Comparison of CP to PDAC-CP and PDAC-CP to PDAC identified upregulation of SPHK1, a known oncogene. CONCLUSIONS Our analysis suggests plausible role for SPHK1 in development of pancreatic adenocarcinoma in long standing CP patients. SPHK1 could be further explored as diagnostic and potential therapeutic target.
Collapse
Affiliation(s)
- Vijayasarathy Ketavarapu
- grid.410866.d0000 0004 1803 177XAsian Healthcare Foundation, Asian Institute of Gastroenterology, Mindspace Rd, Gachibowli, Hyderabad, Telangana 500032 India
| | - Vishnubhotla Ravikanth
- grid.410866.d0000 0004 1803 177XAsian Healthcare Foundation, Asian Institute of Gastroenterology, Mindspace Rd, Gachibowli, Hyderabad, Telangana 500032 India
| | - Mitnala Sasikala
- grid.410866.d0000 0004 1803 177XAsian Healthcare Foundation, Asian Institute of Gastroenterology, Mindspace Rd, Gachibowli, Hyderabad, Telangana 500032 India
| | - G. V. Rao
- grid.410866.d0000 0004 1803 177XAIG Hospitals, Mindspace Rd, Gachibowli, Hyderabad, Telangana 500032 India
| | - Ch. Venkataramana Devi
- grid.412419.b0000 0001 1456 3750Department of Biochemistry, University College of Science, Osmania University, Hyderabad, 500 007 India
| | - Prabhakar Sripadi
- grid.417636.10000 0004 0636 1405Centre for Mass Spectrometry, Analytical & Structural Chemistry Department, CSIR-Indian Institute of Chemical Technology, Tarnaka, Hyderabad, 500 007 India
| | - Murali Satyanarayana Bethu
- grid.410865.eDivision of Applied Biology, CSIR-IICT (Indian Institute of Chemical Technology), Ministry of Science and Technology (GOI), Hyderabad, Telangana 500007 India ,grid.240614.50000 0001 2181 8635Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Elm &Carlton Streets, Buffalo, New York, 14221 USA
| | - Ramars Amanchy
- grid.410865.eDivision of Applied Biology, CSIR-IICT (Indian Institute of Chemical Technology), Ministry of Science and Technology (GOI), Hyderabad, Telangana 500007 India
| | - H. V. V. Murthy
- grid.410866.d0000 0004 1803 177XAsian Healthcare Foundation, Asian Institute of Gastroenterology, Mindspace Rd, Gachibowli, Hyderabad, Telangana 500032 India
| | - Stephen J. Pandol
- grid.50956.3f0000 0001 2152 9905Department of Medicine, Division of Digestive and Liver Diseases, Cedars-Sinai Medical Center, Los Angeles, CA USA
| | - D. Nageshwar Reddy
- grid.410866.d0000 0004 1803 177XAIG Hospitals, Mindspace Rd, Gachibowli, Hyderabad, Telangana 500032 India
| |
Collapse
|
7
|
Zhao H, Dong X, Huang T, Li X. A Potential Prognostic Biomarker for Glioma: Aldo-Keto Reductase Family 1 Member B1. COMPUTATIONAL INTELLIGENCE AND NEUROSCIENCE 2022; 2022:9979200. [PMID: 35341178 PMCID: PMC8956411 DOI: 10.1155/2022/9979200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 02/23/2022] [Indexed: 11/24/2022]
Abstract
Aldo-keto reductase family 1 member B1 (AKR1B1) plays a vital role in tumor development and is involved in the tumor immune process. However, its role in glioma cell is poorly studied. This study's aim was to assess the role of AKR1B1 in glioma through bioinformatics analysis. The AKR1B1 expression data and corresponding clinical data of glioma were collected from the Cancer Genome Atlas (TCGA) database. The R packages were used for data integration, extraction, analysis, and visualization. According to the median value of the risk score, all patients were divided into high-risk and low-risk groups to draw the Kaplan-Meier (KM) survival curves and to explore the level of immune infiltration. The expression of AKR1B1 was significantly elevated in glioma tissues compared to normal tissues (P < 0.001). The high expression of AKR1B1 was significantly associated with WHO grade (P < 0.001), IDH status (P < 0.001), 1p/19q codeletion (P < 0.001), primary therapy outcome (P = 0.004), and age (P < 0.05). Kaplan-Meier survival analysis found that OS (HR = 3.75, P < 0.001), DSS (HR = 3.85, P < 0.001), and PFI (HR = 2.76, P < 0.001) were lower in patients with glioma with high AKR1B1 expression than in the group with low AKR1B1 expression. Based on GESA, six pathways (including interferon gamma signaling, signaling by interleukins, cell cycle checkpoints, cytokine receptor interaction, cell adhesion molecules (CAMs), and cell surface interactions) at the vascular wall were identified as significantly different between the two groups. Moreover, highly expressed AKR1B1 was associated with immune cell infiltration. AKR1B1 plays a key role in glioma progression and prognosis and, therefore, serves as a potential biomarker for prediction of patients' survival.
Collapse
Affiliation(s)
- Hulin Zhao
- Department of Neurosurgery, Chinese PLA General Hospital, Beijing 100853, China
| | - Xuetao Dong
- Department of Neurosurgery, Chuiyangliu Hospital Affiliated To Tsinghua University, Beijing, China
| | - Tianxiang Huang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Xueji Li
- Department of Neurosurgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
8
|
Bose C, Hindle A, Lee J, Kopel J, Tonk S, Palade PT, Singhal SS, Awasthi S, Singh SP. Anticancer Activity of Ω-6 Fatty Acids through Increased 4-HNE in Breast Cancer Cells. Cancers (Basel) 2021; 13:cancers13246377. [PMID: 34944997 PMCID: PMC8699056 DOI: 10.3390/cancers13246377] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/13/2021] [Accepted: 12/15/2021] [Indexed: 12/14/2022] Open
Abstract
Simple Summary Epidemiological evidence suggests that breast cancer risk is lowered by Ω-3 and increased by Ω-6 polyunsaturated fatty acids (PUFAs). Paradoxically, the Ω-6 PUFA metabolite 4-hydroxynonenal (4-HNE) inhibits cancer cell growth. This duality prompted us to study whether arachidonic acid (AA) would enhance doxorubicin (dox) cytotoxicity towards breast cancer cells. We found that supplementing AA or inhibiting 4-HNE metabolism potentiated doxorubicin (dox) toxicity toward Her2-dependent breast cancer but spared myocardial cells. Our results suggest that Ω-6 PUFAs could improve outcomes of dox chemotherapy in Her2-overexpressing breast cancer. Abstract Her2-amplified breast cancers resistant to available Her2-targeted therapeutics continue to be a challenge in breast cancer therapy. Dox is the mainstay of chemotherapy of all types of breast cancer, but its usefulness is limited by cumulative cardiotoxicity. Because oxidative stress caused by dox generates the pro-apoptotic Ω-6 PUFA metabolite 4-hydroxynonenal (4-HNE), we surmised that Ω-6 PUFAs would increase the effectiveness of dox chemotherapy. Since the mercapturic acid pathway enzyme RALBP1 (also known as RLIP76 or Rlip) that limits cellular accumulation of 4-HNE also mediates dox resistance, the combination of Ω-6 PUFAs and Rlip depletion could synergistically improve the efficacy of dox. Thus, we studied the effects of the Ω-6 PUFA arachidonic acid (AA) and Rlip knockdown on the antineoplastic activity of dox towards Her2-amplified breast cancer cell lines SK-BR-3, which is sensitive to Her2 inhibitors, and AU565, which is resistant. AA increased lipid peroxidation, 4-HNE generation, apoptosis, cellular dox concentration and dox cytotoxicity in both cell lines while sparing cultured immortalized cardiomyocyte cells. The known functions of Rlip including clathrin-dependent endocytosis and dox efflux were inhibited by AA. Our results support a model in which 4-HNE generated by AA overwhelms the capacity of Rlip to defend against apoptosis caused by dox or 4-HNE. We propose that Ω-6 PUFA supplementation could improve the efficacy of dox or Rlip inhibitors for treating Her2-amplified breast cancer.
Collapse
Affiliation(s)
- Chhanda Bose
- Department of Internal Medicine, Division of Hematology and Oncology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (C.B.); (A.H.); (J.L.); (J.K.); (S.T.)
| | - Ashly Hindle
- Department of Internal Medicine, Division of Hematology and Oncology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (C.B.); (A.H.); (J.L.); (J.K.); (S.T.)
| | - Jihyun Lee
- Department of Internal Medicine, Division of Hematology and Oncology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (C.B.); (A.H.); (J.L.); (J.K.); (S.T.)
| | - Jonathan Kopel
- Department of Internal Medicine, Division of Hematology and Oncology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (C.B.); (A.H.); (J.L.); (J.K.); (S.T.)
| | - Sahil Tonk
- Department of Internal Medicine, Division of Hematology and Oncology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (C.B.); (A.H.); (J.L.); (J.K.); (S.T.)
| | - Philip T. Palade
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
| | - Sharad S. Singhal
- Department of Medical Oncology and Therapeutic Research, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA;
| | - Sanjay Awasthi
- Department of Internal Medicine, Division of Hematology and Oncology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (C.B.); (A.H.); (J.L.); (J.K.); (S.T.)
- Medical Oncology Service, Doctors Hospital, 16 Middle Rd., George Town, Grand Cayman KY1-1104, Cayman Islands, UK
- Correspondence: (S.A.); (S.P.S.); Tel.: +1-305-949-6066 (S.A.); +1-806-743-1540 (S.P.S.)
| | - Sharda P. Singh
- Department of Internal Medicine, Division of Hematology and Oncology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (C.B.); (A.H.); (J.L.); (J.K.); (S.T.)
- Correspondence: (S.A.); (S.P.S.); Tel.: +1-305-949-6066 (S.A.); +1-806-743-1540 (S.P.S.)
| |
Collapse
|
9
|
Zhou W, Wu C, Zhao C, Huang Z, Lu S, Fan X, Tan Y, Stalin A, You R, Liu X, Zhang J, Wu Z, Wu J. An Advanced Systems Pharmacology Strategy Reveals AKR1B1, MMP2, PTGER3 as Key Genes in the Competing Endogenous RNA Network of Compound Kushen Injection Treating Gastric Carcinoma by Integrated Bioinformatics and Experimental Verification. Front Cell Dev Biol 2021; 9:742421. [PMID: 34646828 PMCID: PMC8502965 DOI: 10.3389/fcell.2021.742421] [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: 07/16/2021] [Accepted: 09/06/2021] [Indexed: 12/24/2022] Open
Abstract
Gastric carcinoma (GC) is a severe tumor of the digestive tract with high morbidity and mortality and poor prognosis, for which novel treatment options are urgently needed. Compound Kushen injection (CKI), a classical injection of Chinese medicine, has been widely used to treat various tumors in clinical practice for decades. In recent years, a growing number of studies have confirmed that CKI has a beneficial therapeutic effect on GC, However, there are few reports on the potential molecular mechanism of action. Here, using systems pharmacology combined with proteomics analysis as a core concept, we identified the ceRNA network, key targets and signaling pathways regulated by CKI in the treatment of GC. To further explore the role of these key targets in the development of GC, we performed a meta-analysis to compare the expression differences between GC and normal gastric mucosa tissues. Functional enrichment analysis was further used to understand the biological pathways significantly regulated by the key genes. In addition, we determined the significance of the key genes in the prognosis of GC by survival analysis and immune infiltration analysis. Finally, molecular docking simulation was performed to verify the combination of CKI components and key targets. The anti-gastric cancer effect of CKI and its key targets was verified by in vivo and in vitro experiments. The analysis of ceRNA network of CKI on GC revealed that the potential molecular mechanism of CKI can regulate PI3K/AKT and Toll-like receptor signaling pathways by interfering with hub genes such as AKR1B1, MMP2 and PTGERR3. In conclusion, this study not only partially highlighted the molecular mechanism of CKI in GC therapy but also provided a novel and advanced systems pharmacology strategy to explore the mechanisms of traditional Chinese medicine formulations.
Collapse
Affiliation(s)
- Wei Zhou
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China.,China-Japan Friendship Hospital, Beijing, China
| | - Chao Wu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Chongjun Zhao
- Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Zhihong Huang
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Shan Lu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaotian Fan
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yingying Tan
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Antony Stalin
- State Key Laboratory of Subtropical Silviculture, Department of Traditional Chinese Medicine, Zhejiang A&F University, Hangzhou, China
| | - Rongli You
- Shanxi Zhendong Pharmaceutical Co., Ltd., Shanxi, China
| | - Xinkui Liu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Jingyuan Zhang
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Zhishan Wu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Jiarui Wu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
10
|
The Role of the FOXO1/β 2-AR/p-NF-κB p65 Pathway in the Development of Endometrial Stromal Cells in Pregnant Mice under Restraint Stress. Int J Mol Sci 2021; 22:ijms22031478. [PMID: 33540675 PMCID: PMC7867244 DOI: 10.3390/ijms22031478] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 01/23/2021] [Accepted: 01/28/2021] [Indexed: 12/13/2022] Open
Abstract
Restraint stress causes various maternal diseases during pregnancy. β2-Adrenergic receptor (β2-AR) and Forkhead transcription factor class O 1 (FOXO1) are critical factors not only in stress, but also in reproduction. However, the role of FOXO1 in restraint stress, causing changes in the β2-AR pathway in pregnant mice, has been unclear. The aim of this research was to investigate the β2-AR pathway of restraint stress and its impact on the oxidative stress of the maternal uterus. In the study, maternal mice were treated with restraint stress by being restrained in a transparent and ventilated device before sacrifice on Pregnancy Day 5 (P5), Pregnancy Day 10 (P10), Pregnancy Day 15 (P15), and Pregnancy Day 20 (P20) as well as on Non-Pregnancy Day 5 (NP5). Restraint stress augmented blood corticosterone (CORT), norepinephrine (NE), and blood glucose levels, while oestradiol (E2) levels decreased. Moreover, restraint stress increased the mRNA levels of the FOXO family, β2-AR, and even the protein levels of FOXO1 and β2-AR in the uterus and ovaries. Furthermore, restraint stress increased uterine oxidative stress level. In vitro, the protein levels of FOXO1 were also obviously increased when β2-AR was activated in endometrial stromal cells (ESCs). In addition, phosphorylated-nuclear factor kappa-B p65 (p-NF-κB p65) and its target genes decreased significantly when FOXO1 was inhibited. Overall, it can be said that the β2-AR/FOXO1/p-NF-κB p65 pathway was activated when pregnant mice were under restraint stress. This study provides a scientific basis for the origin of psychological stress in pregnant women.
Collapse
|
11
|
Dragomir MP, Moisoiu V, Manaila R, Pardini B, Knutsen E, Anfossi S, Amit M, Calin GA. A Holistic Perspective: Exosomes Shuttle between Nerves and Immune Cells in the Tumor Microenvironment. J Clin Med 2020; 9:jcm9113529. [PMID: 33142779 PMCID: PMC7693842 DOI: 10.3390/jcm9113529] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 10/27/2020] [Indexed: 02/06/2023] Open
Abstract
One of the limitations of cancer research has been the restricted focus on tumor cells and the omission of other non-malignant cells that are constitutive elements of this systemic disease. Current research is focused on the bidirectional communication between tumor cells and other components of the tumor microenvironment (TME), such as immune and endothelial cells, and nerves. A major success of this bidirectional approach has been the development of immunotherapy. Recently, a more complex landscape involving a multi-lateral communication between the non-malignant components of the TME started to emerge. A prime example is the interplay between immune and endothelial cells, which led to the approval of anti-vascular endothelial growth factor-therapy combined with immune checkpoint inhibitors and classical chemotherapy in non-small cell lung cancer. Hence, a paradigm shift approach is to characterize the crosstalk between different non-malignant components of the TME and understand their role in tumorigenesis. In this perspective, we discuss the interplay between nerves and immune cells within the TME. In particular, we focus on exosomes and microRNAs as a systemic, rapid and dynamic communication channel between tumor cells, nerves and immune cells contributing to cancer progression. Finally, we discuss how combinatorial therapies blocking this tumorigenic cross-talk could lead to improved outcomes for cancer patients.
Collapse
Affiliation(s)
- Mihnea P. Dragomir
- Department of Surgery, Fundeni Clinical Hospital, Carol Davila University of Medicine and Pharmacy, 022328 Bucharest, Romania
- Institute of Pathology, Charité University Hospital, 10117 Berlin, Germany
- Correspondence: (M.P.D.); (G.A.C.)
| | - Vlad Moisoiu
- Faculty of Physics, Babeş-Bolyai University, 400084 Cluj-Napoca, Romania;
| | - Roxana Manaila
- Clinical Institute of Urology and Renal Transplantation, 400006 Cluj-Napoca, Romania;
| | - Barbara Pardini
- Italian Institute for Genomic Medicine (IIGM), 10060 Candiolo, Italy;
- Candiolo Cancer Institute, FPO-IRCCS, 10060 Candiolo, Italy
| | - Erik Knutsen
- Department of Medical Biology, Faculty of Health Sciences, UiT—The Arctic University of Norway, N-9037 Tromsø, Norway;
| | - Simone Anfossi
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Moran Amit
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - George A. Calin
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
- The Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Correspondence: (M.P.D.); (G.A.C.)
| |
Collapse
|
12
|
Targeting β2-Adrenergic Receptors Shows Therapeutical Benefits in Clear Cell Renal Cell Carcinoma from Von Hippel-Lindau Disease. J Clin Med 2020; 9:jcm9092740. [PMID: 32854260 PMCID: PMC7563408 DOI: 10.3390/jcm9092740] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 08/19/2020] [Accepted: 08/21/2020] [Indexed: 12/15/2022] Open
Abstract
Von Hippel–Lindau (VHL), is a rare autosomal dominant inherited cancer in which the lack of VHL protein triggers the development of multisystemic tumors such us retinal hemangioblastomas (HB), CNS-HB, and clear cell renal cell carcinoma (ccRCC). ccRCC ranks third in terms of incidence and first in cause of death. Standard systemic therapies for VHL-ccRCC have shown limited response, with recurrent surgeries being the only effective treatment. Targeting of β2-adrenergic receptor (ADRB) has shown therapeutic antitumor benefits on VHL-retinal HB (clinical trial) and VHL-CNS HB (in vitro). Therefore, the in vitro and in vivo antitumor benefits of propranolol (ADRB-1,2 antagonist) and ICI-118,551 (ADRB-2 antagonist) on VHL−/− ccRCC primary cultures and 786-O tumor cell lines have been addressed. Propranolol and ICI-118,551 activated apoptosis inhibited gene and protein expression of HIF-2α, CAIX, and VEGF, and impaired partially the nuclear internalization of HIF-2α and NFĸB/p65. Moreover, propranolol and ICI-118,551 reduced tumor growth on two in vivo xenografts. Finally, ccRCC patients receiving propranolol as off-label treatment have shown a positive therapeutic response for two years on average. In summary, propranolol and ICI-118,551 have shown antitumor benefits in VHL-derived ccRCC, and since ccRCCs comprise 63% of the total RCCs, targeting ADRB2 becomes a promising drug for VHL and other non-VHL tumors.
Collapse
|
13
|
Ji J, Xu MX, Qian TY, Zhu SZ, Jiang F, Liu ZX, Xu WS, Zhou J, Xiao MB. The AKR1B1 inhibitor epalrestat suppresses the progression of cervical cancer. Mol Biol Rep 2020; 47:6091-6103. [PMID: 32761301 DOI: 10.1007/s11033-020-05685-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 07/26/2020] [Indexed: 12/24/2022]
Abstract
Cervical cancer is the leading cause of cancer-related death among women worldwide. Identifying an effective treatment with fewer side effects is imperative, because all of the current treatments have unique disadvantages. Aldo-keto reductase family 1 member B1 (AKR1B1) is highly expressed in various cancers and is associated with tumor development, but has not been studied in cervical cancer. In the current study, we used CRISPR/Cas9 technology to establish a stable HeLa cell line with AKR1B1 knockout. In vitro, AKR1B1 knockout inhibited the proliferation, migration and invasion of HeLa cells, providing evidence that AKR1B1 is an innovative therapeutic target. Notably, the clinically used epalrestat, an inhibitor of aldose reductases, including AKR1B1, had the same effect as AKR1B1 knockout on HeLa cells. This result suggests that epalrestat could be used in the clinical treatment of cervical cancer, a prospect that undoubtedly requires further research. Moreover, aiming to determine the underlying regulatory mechanism of AKR1B1, we screened a series of differentially regulated genes (DEGs) by RNA sequencing and verified selected DEGs by quantitative RT-PCR. In addition, gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses of the DEGs revealed a correlation between AKR1B1 and cancer. In summary, epalrestat inhibits the progression of cervical cancer by inhibiting AKR1B1, and thus may be a new drug for the clinical treatment of cervical cancer.
Collapse
Affiliation(s)
- Jie Ji
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
- Department of Gastroenterology and Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, No. 20 Xisi Road, Nantong, 226001, People's Republic of China
- Medical College, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Min-Xue Xu
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
- Department of Gastroenterology and Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, No. 20 Xisi Road, Nantong, 226001, People's Republic of China
- Medical College, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Tian-Yang Qian
- Chinese Medicine 193, First Clinical Medical School, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China
| | - Sheng-Ze Zhu
- Medical College, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Feng Jiang
- Department of Gastroenterology and Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, No. 20 Xisi Road, Nantong, 226001, People's Republic of China
| | - Zhao-Xiu Liu
- Department of Gastroenterology and Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, No. 20 Xisi Road, Nantong, 226001, People's Republic of China
| | - Wei-Song Xu
- Department of Gastroenterology, Second People's Hospital of Nantong, Nantong, 226001, Jiangsu, People's Republic of China
| | - Juan Zhou
- Department of Respiratory Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, People's Republic of China.
| | - Ming-Bing Xiao
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, People's Republic of China.
- Department of Gastroenterology and Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, No. 20 Xisi Road, Nantong, 226001, People's Republic of China.
| |
Collapse
|
14
|
Luo L, Chen C, He H, Cai M, Ling C. Silencing of Long Non-Coding RNA (LncRNA) Non-Coding RNA Activated by DNA Damage (NORAD) Inhibits Proliferation, Invasion, Migration, and Promotes Apoptosis of Glioma Cells via Downregulating the Expression of AKR1B1. Med Sci Monit 2020; 26:e922659. [PMID: 32778640 PMCID: PMC7392058 DOI: 10.12659/msm.922659] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Background We aimed to investigate the functions of long non-coding RNA (lncRNA) non-coding RNA activated by DNA damage (NORAD) in glioma and identify the potential mechanisms. Material/Methods The expression of NORAD and AKR1B1 in human glioma cell lines were examined using reverse transcription-quantitative polymerase chain reaction (RT-qPCR). Then, cell proliferation, invasion, and migration were tested by Cell Counting Kit-8 (CCK-8), colony formation assay, Transwell, and scratch wound healing assay after NORAD silencing. Meanwhile, western blotting was utilized to measure the expression of migration-related proteins. Apoptosis of glioma cells was detected using flow cytometry and apoptosis-related proteins expression was determined. Moreover, the correlation between NORAD and AKR1B1 was verified by RNA-binding protein immunoprecipitation (RIP assay). After co-transfection with AKR1B1 overexpressed plasmid and NORAD siRNA, cell proliferation, invasion, migration, and apoptosis were examined again. Furthermore, the expression of proteins in extracellular signal-regulated kinase (ERK) signaling was tested using western blotting. Results The results revealed that NORAD and AKR1B1 were highly expressed in glioma cells. NORAD silencing inhibited proliferation, invasion and migration but promoted apoptosis of glioma cells, accompanied by the expression changes of migration- and apoptosis-related proteins. However, after co-transfection with AKR1B1 pcDNA3.1 in NORAD silencing cells, the effects of NORAD silencing on proliferation, invasion, migration, and apoptosis were attenuated. Consistently, the expression of phosphorylated ERK (p-ERK) was decreased after NORAD silencing, which were reversed following AKR1B1 overexpression. Conclusions These findings demonstrated that NORAD silencing suppressed proliferation, invasion, and migration and boosted apoptosis of glioma cells via downregulating the AKR1B1 expression, which may provide a potential therapeutic target for glioma treatment.
Collapse
Affiliation(s)
- Lun Luo
- Department of Neurosurgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China (mainland)
| | - Chuan Chen
- Department of Neurosurgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China (mainland)
| | - Haiyong He
- Department of Neurosurgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China (mainland)
| | - Meiqin Cai
- Department of Neurosurgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China (mainland)
| | - Cong Ling
- Department of Neurosurgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China (mainland)
| |
Collapse
|
15
|
Khayami R, Hashemi SR, Kerachian MA. Role of aldo-keto reductase family 1 member B1 (AKR1B1) in the cancer process and its therapeutic potential. J Cell Mol Med 2020; 24:8890-8902. [PMID: 32633024 PMCID: PMC7417692 DOI: 10.1111/jcmm.15581] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 05/21/2020] [Accepted: 05/27/2020] [Indexed: 02/06/2023] Open
Abstract
The role of aldo‐keto reductase family 1 member B1 (AKR1B1) in cancer is not totally clear but growing evidence is suggesting to have a great impact on cancer progression. AKR1B1 could participate in a complicated network of signalling pathways, proteins and miRNAs such as mir‐21 mediating mechanisms like inflammatory responses, cell cycle, epithelial to mesenchymal transition, cell survival and apoptosis. AKR1B1 has been shown to be mostly overexpressed in cancer. This overexpression has been associated with inflammatory mediators including nuclear factor kappa‐light‐chain‐enhancer of activated B cells (NFκB), cell cycle mediators such as cyclins and cyclin‐dependent kinases (CDKs), survival proteins and pathways like mammalian target of rapamycin (mTOR) and protein kinase B (PKB) or AKT, and other regulatory factors in response to reactive oxygen species (ROS) and prostaglandin synthesis. In addition, inhibition of AKR1B1 has been shown to mostly have anti‐cancer effects. Several studies have also suggested that AKR1B1 inhibition as an adjuvant therapy could render tumour cells more sensitive to anti‐cancer therapy or alleviate the adverse effects of therapy. AKR1B1 could also be considered as a potential cancer diagnostic biomarker since its promoter has shown high levels of methylation. Although pre‐clinical investigations on the role of AKR1B1 in cancer and the application of its inhibitors have shown promising results, the lack of clinical studies on AKR1B1 inhibitors has hampered the use of these drugs to treat cancer. Thus, there is a need to conduct more clinical studies on the application of AKR1B1 inhibitors as adjuvant therapy on different cancers.
Collapse
Affiliation(s)
- Reza Khayami
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyyed Reza Hashemi
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Amin Kerachian
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Cancer Genetics Research Unit, Reza Radiotherapy and Oncology Center, Mashhad, Iran
| |
Collapse
|
16
|
Li X, Yang J, Gu X, Xu J, Li H, Qian J, Chen L. The Expression and Clinical Significance of Aldo-Keto Reductase 1 Member B1 in Gastric Carcinoma. DNA Cell Biol 2020; 39:1322-1327. [PMID: 32412859 DOI: 10.1089/dna.2020.5550] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
To study the expression of aldo-keto reductase 1 member B1 (AKR1B1) in gastric carcinoma (GC), the correlation between AKR1B1 and the clinicopathological characteristics of GC patients, and provide reference for the diagnosis and prognosis of GC patients. One hundred thirty-six patients with GC were collected, and the expression level of AKR1B1 in GC and adjacent tissues was detected by immunohistochemistry assays. The clinicopathological features and prognosis of GC patients were collected to analyze the relationship with AKR1B1 expression. The positive expression of AKR1B1 in GC tissues was significantly higher than that of adjacent nontumor tissues. The difference of AKR1B1 expression between GC tissues and paired adjacent nontumor tissues was statistically significant (p < 0.001). AKR1B1 was closely related to tumor size, regional lymph node (N), metastases (M), and tumor-node-metastasis (TNM) stage (p < 0.05). The overall survival of patients with low expression of AKR1B1 was significantly better than that of patients with high expression of AKR1B1 by Kaplan-Meier survival analysis (p < 0.001). AKR1B1 plays an important role in the occurrence and development of GC, and it has a certain reference value for the prognosis of GC patients.
Collapse
Affiliation(s)
- Xin Li
- Department of Oncology, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine (TCM), Shanghai, China
| | - Jinzu Yang
- Department of Oncology, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine (TCM), Shanghai, China
| | - Xiaoqiang Gu
- Department of Oncology, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine (TCM), Shanghai, China
| | - Jiahua Xu
- Department of Oncology, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine (TCM), Shanghai, China
| | - Hongwei Li
- Department of Oncology, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine (TCM), Shanghai, China
| | - Jianxin Qian
- Department of Oncology, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine (TCM), Shanghai, China
| | - Ling Chen
- Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine Affiliated to Shanghai University of Traditional Chinese Medicine (TCM), Shanghai, China
| |
Collapse
|
17
|
Ghali GZ, Ghali MGZ. β adrenergic receptor modulated signaling in glioma models: promoting β adrenergic receptor-β arrestin scaffold-mediated activation of extracellular-regulated kinase 1/2 may prove to be a panacea in the treatment of intracranial and spinal malignancy and extra-neuraxial carcinoma. Mol Biol Rep 2020; 47:4631-4650. [PMID: 32303958 PMCID: PMC7165076 DOI: 10.1007/s11033-020-05427-1] [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: 02/25/2020] [Accepted: 04/03/2020] [Indexed: 12/03/2022]
Abstract
Neoplastically transformed astrocytes express functionally active cell surface β adrenergic receptors (βARs). Treatment of glioma models in vitro and in vivo with β adrenergic agonists variably amplifies or attenuates cellular proliferation. In the majority of in vivo models, β adrenergic agonists generally reduce cellular proliferation. However, treatment with β adrenergic agonists consistently reduces tumor cell invasive potential, angiogenesis, and metastasis. β adrenergic agonists induced decreases of invasive potential are chiefly mediated through reductions in the expression of matrix metalloproteinases types 2 and 9. Treatment with β adrenergic agonists also clearly reduce tumoral neoangiogenesis, which may represent a putatively useful mechanism to adjuvantly amplify the effects of bevacizumab. Bevacizumab is a monoclonal antibody targeting the vascular endothelial growth factor receptor. We may accordingly designate βagonists to represent an enhancer of bevacizumab. The antiangiogenic effects of β adrenergic agonists may thus effectively render an otherwise borderline effective therapy to generate significant enhancement in clinical outcomes. β adrenergic agonists upregulate expression of the major histocompatibility class II DR alpha gene, effectively potentiating the immunogenicity of tumor cells to tumor surveillance mechanisms. Authors have also demonstrated crossmodal modulation of signaling events downstream from the β adrenergic cell surface receptor and microtubular polymerization and depolymerization. Complex effects and desensitization mechanisms of the β adrenergic signaling may putatively represent promising therapeutic targets. Constant stimulation of the β adrenergic receptor induces its phosphorylation by β adrenergic receptor kinase (βARK), rendering it a suitable substrate for alternate binding by β arrestins 1 or 2. The binding of a β arrestin to βARK phosphorylated βAR promotes receptor mediated internalization and downregulation of cell surface receptor and contemporaneously generates a cell surface scaffold at the βAR. The scaffold mediated activation of extracellular regulated kinase 1/2, compared with protein kinase A mediated activation, preferentially favors cytosolic retention of ERK1/2 and blunting of nuclear translocation and ensuant pro-transcriptional activity. Thus, βAR desensitization and consequent scaffold assembly effectively retains the cytosolic homeostatic functions of ERK1/2 while inhibiting its pro-proliferative effects. We suggest these mechanisms specifically will prove quite promising in developing primary and adjuvant therapies mitigating glioma growth, angiogenesis, invasive potential, and angiogenesis. We suggest generating compounds and targeted mutations of the β adrenergic receptor favoring β arrestin binding and scaffold facilitated activation of ERK1/2 may hold potential promise and therapeutic benefit in adjuvantly treating most or all cancers. We hope our discussion will generate fruitful research endeavors seeking to exploit these mechanisms.
Collapse
Affiliation(s)
- George Zaki Ghali
- United States Environmental Protection Agency, Arlington, VA, USA.,Emeritus Professor, Department of Toxicology, Purdue University, West Lafayette, IN, USA
| | - Michael George Zaki Ghali
- Department of Neurological Surgery, University of California, San Francisco, 505 Parnassus Avenue, Box-0112, San Francisco, CA, 94143, USA. .,Department of Neurological Surgery, Karolinska Institutet, Nobels väg 6, Solna and Alfred Nobels Allé 8, Huddinge, SE-171 77, Stockholm, Sweden.
| |
Collapse
|
18
|
Xu K, Huang Y, Zhou T, Wang C, Chi Q, Shi J, Zhu P, Dong N. Nobiletin exhibits potent inhibition on tumor necrosis factor alpha‐induced calcification of human aortic valve interstitial cells via targeting ABCG2 and AKR1B1. Phytother Res 2019; 33:1717-1725. [PMID: 31016813 DOI: 10.1002/ptr.6360] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 02/25/2019] [Accepted: 03/06/2019] [Indexed: 01/28/2023]
Affiliation(s)
- Kang Xu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical CollegeHuazhong University of Science and Technology Wuhan China
| | - Yuming Huang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical CollegeHuazhong University of Science and Technology Wuhan China
| | - Tingwen Zhou
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical CollegeHuazhong University of Science and Technology Wuhan China
| | - Chunli Wang
- National Innovation and Attracting Talents “111” base, Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of BioengineeringChongqing University Chongqing China
| | - Qingjia Chi
- Department of Mechanics and Engineering Structure, Hubei Key Laboratory of Theory and Application of Advanced Materials MechanicsWuhan University of Technology Wuhan China
| | - Jiawei Shi
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical CollegeHuazhong University of Science and Technology Wuhan China
| | - Peng Zhu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical CollegeHuazhong University of Science and Technology Wuhan China
| | - Nianguo Dong
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical CollegeHuazhong University of Science and Technology Wuhan China
| |
Collapse
|