1
|
Kawade G, Kurata M, Matsuki Y, Fukuda S, Onishi I, Kinowaki Y, Watabe S, Ishibashi S, Ikeda M, Yamamoto M, Ohashi K, Kitagawa M, Yamamoto K. Mediation of Ferroptosis Suppressor Protein 1 Expression via 4-Hydroxy-2-Nonenal Accumulation Contributes to Acquisition of Resistance to Apoptosis and Ferroptosis in Diffuse Large B-Cell Lymphoma. J Transl Med 2024; 104:102027. [PMID: 38311062 DOI: 10.1016/j.labinv.2024.102027] [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: 06/02/2023] [Revised: 01/14/2024] [Accepted: 01/25/2024] [Indexed: 02/06/2024] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL) is the most common non-Hodgkin lymphoma. New therapeutic strategies are needed for the treatment of refractory DLBCL. 4-Hydroxy-2-nonenal (4-HNE) is a cytotoxic lipid peroxidation marker, which alters intracellular signaling and induces genetic mutations. Lipid peroxidation is associated with nonapoptotic cell death, called ferroptosis. However, the relationship between 4-HNE accumulation and feroptotic regulators in DLBCL has not been fully evaluated. Here, we aimed to evaluate the accumulation of lipid peroxide and the expression of ferroptosis suppressor protein 1 (FSP1) in DLBCL using immunohistochemistry. We found a significant increase in the expression of FSP1 in cases with nuclear 4-HNE accumulation (P = .021). Both nuclear and cytoplasmic 4-HNE accumulation and FSP1 positivity were independent predictors of worse prognosis. In vitro exposure to 4-HNE resulted in its concentration- and time-dependent intracellular accumulation and increased expression of FSP1. Furthermore, short-term (0.25 and 1.0 μM) or long-term (0.25 μM) exposure to 4-HNE induced resistance to not only apoptosis but also ferroptosis. Taken together, regulation of FSP1 through 4-HNE accumulation may attenuate resistance to cell death in treatment-resistant DLBCL and might help develop novel therapeutic strategies for refractory DLBCL.
Collapse
Affiliation(s)
- Genji Kawade
- Department of Comprehensive Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan; Department of Human Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Morito Kurata
- Department of Comprehensive Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yuko Matsuki
- Department of Comprehensive Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Sho Fukuda
- Department of Comprehensive Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Iichiroh Onishi
- Department of Comprehensive Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yuko Kinowaki
- Department of Comprehensive Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Shiori Watabe
- Department of Comprehensive Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Sachiko Ishibashi
- Department of Comprehensive Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Masumi Ikeda
- Department of Comprehensive Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Masahide Yamamoto
- Department of Hematology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kenichi Ohashi
- Department of Human Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Masanobu Kitagawa
- Department of Comprehensive Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kouhei Yamamoto
- Department of Comprehensive Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan; Department of Human Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan.
| |
Collapse
|
2
|
Fan Y, Li X, Li J, Xiong X, Yin S, Fu W, Wang P, Liu J, Xiong Y. Differential metabolites screening in yak (Bos grunniens) seminal plasma after cryopreservation and the evaluation of the effect of galactose on post-thaw sperm motility. Theriogenology 2024; 215:249-258. [PMID: 38103402 DOI: 10.1016/j.theriogenology.2023.12.011] [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: 08/27/2023] [Revised: 12/07/2023] [Accepted: 12/08/2023] [Indexed: 12/19/2023]
Abstract
Sperm survival and activity depend on the provision of energy and nutrients from seminal plasma (SP). This study aimed to investigate the variations of metabolites within SP before and after freezing and subsequently explore the potential regulatory mechanisms affecting yak sperm cryodamage due to changes in metabolites in the SP. Untargeted metabolomics analysis was performed to screen for differential metabolites, followed by KEGG analysis to identify enriched signaling pathways. The combinatorial analysis of metabolomics and sperm proteomics revealed the influence of key SP metabolites on sperm proteins. Subsequently, the relevant differentially expressed proteins were verified by Western blot analysis. Finally, the mechanism underlying the positive effect of galactose on sperm motility was determined by assessing the change in ATP content in sperm before and after freezing and thawing. The data showed that a total of 425 and 269 metabolites were identified in the positive and negative ion modes, respectively. Freezing and thawing resulted in the up-regulation of 70 metabolites and the down-regulation of 29 metabolites in SP. The primary impact of freezing and thawing was observed in carbohydrate metabolism, including pyruvate metabolism, pentose phosphate pathway, galactose metabolism, the TCA cycle, and butanoate metabolism. In the combined analysis and Western blot results, a significant positive correlation was observed between galactose and Aldo-keto reductase family 1 member B1 (AKR1B1) (P < 0.05), which has the ability to convert galactose into galactol. Furthermore, the addition of galactose to thawed semen improved sperm motility by increasing AKR1B1 protein in sperm and was associated with the content of ATP. These data identify differential metabolites between fresh and frozen-thawed SP and suggest that galactose is a valuable additive for cryopreserved sperm, providing a theoretical basis for further exploration of the refrigerant formula for yak sperm cryopreservation.
Collapse
Affiliation(s)
- Yilin Fan
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Reservation and Utilization, Ministry of Education and Sichuan Province, Southwest Minzu University, Chengdu, 610041, China; Key Laboratory of Animal Science of National Ethnic Affairs Commission of China, Southwest Minzu University, Chengdu, 610041, China
| | - Xiaowei Li
- Longri Breeding Stock Farm of Sichuan Province, Dujiangyan, 611800, China
| | - Jian Li
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Reservation and Utilization, Ministry of Education and Sichuan Province, Southwest Minzu University, Chengdu, 610041, China; Key Laboratory of Animal Science of National Ethnic Affairs Commission of China, Southwest Minzu University, Chengdu, 610041, China.
| | - Xianrong Xiong
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Reservation and Utilization, Ministry of Education and Sichuan Province, Southwest Minzu University, Chengdu, 610041, China; Key Laboratory of Animal Science of National Ethnic Affairs Commission of China, Southwest Minzu University, Chengdu, 610041, China
| | - Shi Yin
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Reservation and Utilization, Ministry of Education and Sichuan Province, Southwest Minzu University, Chengdu, 610041, China; Key Laboratory of Animal Science of National Ethnic Affairs Commission of China, Southwest Minzu University, Chengdu, 610041, China
| | - Wei Fu
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Reservation and Utilization, Ministry of Education and Sichuan Province, Southwest Minzu University, Chengdu, 610041, China; Key Laboratory of Animal Science of National Ethnic Affairs Commission of China, Southwest Minzu University, Chengdu, 610041, China
| | - Peng Wang
- Sichuan Ganzi Tibetan Autonomous Prefecture, Institute of Animal Husbandry Science, Kangding, 626000, China
| | - Jun Liu
- Sichuan Ganzi Tibetan Autonomous Prefecture, Institute of Animal Husbandry Science, Kangding, 626000, China
| | - Yan Xiong
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Reservation and Utilization, Ministry of Education and Sichuan Province, Southwest Minzu University, Chengdu, 610041, China; Key Laboratory of Animal Science of National Ethnic Affairs Commission of China, Southwest Minzu University, Chengdu, 610041, China.
| |
Collapse
|
3
|
Zhang Y, Gao S, Xia S, Yang H, Bao X, Zhang Q, Xu Y. Linarin ameliorates ischemia-reperfusion injury by the inhibition of endoplasmic reticulum stress targeting AKR1B1. Brain Res Bull 2024; 207:110868. [PMID: 38181967 DOI: 10.1016/j.brainresbull.2024.110868] [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: 09/29/2023] [Revised: 12/18/2023] [Accepted: 01/01/2024] [Indexed: 01/07/2024]
Abstract
Due to various factors, there is still a lack of effective neuroprotective agents for ischemic stroke in clinical practice. Neuroinflammation and neuronal apoptosis mediated by endoplasmic reticulum stress are some of the important pathological mechanisms in ischemic stroke. Linarin has been reported to have anti-inflammation, antioxidant, and anti-apoptotic effects in myocardial ischemia, osteoarthritis, and kidney disease. Whether it exerts neuroprotective functions in ischemic stroke has not been investigated. The results showed that linarin could reduce the infarct volume in cerebral ischemia animal models, improve the neurological function scores and suppress the expression of inflammatory factors mediating the NF-κB. Meanwhile, it could protect the neurons from OGD/R-induced-apoptosis, which was related to the PERK-eIF2α pathway. Our results suggested linarin could inhibit neuronal inflammation and apoptosis induced by endoplasmic reticulum stress. Furthermore, the neuroprotective effect of linarin may be related to the inhibition of AKR1B1. Our study offers new insight into protecting against ischemia-reperfusion injury by linarin treatment in stroke.
Collapse
Affiliation(s)
- Yuqian Zhang
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210008, China
| | - Shenghan Gao
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China; Department of Neurology, Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing 210008, China
| | - Shengnan Xia
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Haiyan Yang
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Xinyu Bao
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Qingxiu Zhang
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210008, China; Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China; Department of Neurology, Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing 210008, China.
| | - Yun Xu
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210008, China; Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China; Department of Neurology, Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing 210008, China; Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing 210008, China; Jiangsu Provincial Key Discipline of Neurology, Nanjing 210008, China; Nanjing Neurology Medical Center, Nanjing 210008, China.
| |
Collapse
|
4
|
Kang YL, Kim J, Kwak SB, Kim YS, Huh J, Park JW. The polyol pathway and nuclear ketohexokinase A signaling drive hyperglycemia-induced metastasis of gastric cancer. Exp Mol Med 2024; 56:220-234. [PMID: 38200154 PMCID: PMC10834943 DOI: 10.1038/s12276-023-01153-3] [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: 01/10/2023] [Revised: 10/20/2023] [Accepted: 11/07/2023] [Indexed: 01/12/2024] Open
Abstract
Diabetes might be associated with increased cancer risk, with several studies reporting hyperglycemia as a primary oncogenic stimulant. Since glucose metabolism is linked to numerous metabolic pathways, it is difficult to specify the mechanisms underlying hyperglycemia-induced cancer progression. Here, we focused on the polyol pathway, which is dramatically activated under hyperglycemia and causes diabetic complications. We investigated whether polyol pathway-derived fructose facilitates hyperglycemia-induced gastric cancer metastasis. We performed bioinformatics analysis of gastric cancer datasets and immunohistochemical analyses of gastric cancer specimens, followed by transcriptomic and proteomic analyses to evaluate phenotypic changes in gastric cancer cells. Consequently, we found a clinical association between the polyol pathway and gastric cancer progression. In gastric cancer cell lines, hyperglycemia enhanced cell migration and invasion, cytoskeletal rearrangement, and epithelial-mesenchymal transition (EMT). The hyperglycemia-induced acquisition of metastatic potential was mediated by increased fructose derived from the polyol pathway, which stimulated the nuclear ketohexokinase-A (KHK-A) signaling pathway, thereby inducing EMT by repressing the CDH1 gene. In two different xenograft models of cancer metastasis, gastric cancers overexpressing AKR1B1 were found to be highly metastatic in diabetic mice, but these effects of AKR1B1 were attenuated by KHK-A knockdown. In conclusion, hyperglycemia induces fructose formation through the polyol pathway, which in turn stimulates the KHK-A signaling pathway, driving gastric cancer metastasis by inducing EMT. Thus, the polyol and KHK-A signaling pathways could be potential therapeutic targets to decrease the metastatic risk in gastric cancer patients with diabetes.
Collapse
Affiliation(s)
- Ye-Lim Kang
- Department of Biomedical Science, BK21-Plus Education Program, Seoul National University College of Medicine, Daehak-ro, Jongno-gu, Seoul, 03080, Korea
- Department of Pharmacology, Seoul National University College of Medicine, Daehak-ro, Jongno-gu, Seoul, 03080, Korea
| | - Jiyoung Kim
- Department of Biomedical Science, BK21-Plus Education Program, Seoul National University College of Medicine, Daehak-ro, Jongno-gu, Seoul, 03080, Korea
- Department of Pharmacology, Seoul National University College of Medicine, Daehak-ro, Jongno-gu, Seoul, 03080, Korea
| | - Su-Bin Kwak
- Department of Biomedical Science, BK21-Plus Education Program, Seoul National University College of Medicine, Daehak-ro, Jongno-gu, Seoul, 03080, Korea
- Department of Pharmacology, Seoul National University College of Medicine, Daehak-ro, Jongno-gu, Seoul, 03080, Korea
| | - Yi-Sook Kim
- Department of Biomedical Science, BK21-Plus Education Program, Seoul National University College of Medicine, Daehak-ro, Jongno-gu, Seoul, 03080, Korea
- Department of Pharmacology, Seoul National University College of Medicine, Daehak-ro, Jongno-gu, Seoul, 03080, Korea
| | - June Huh
- Department of Chemical and Biological Engineering, Korea University, Anam-ro, Seongbuk-gu, Seoul, 02841, Korea
| | - Jong-Wan Park
- Department of Biomedical Science, BK21-Plus Education Program, Seoul National University College of Medicine, Daehak-ro, Jongno-gu, Seoul, 03080, Korea.
- Department of Pharmacology, Seoul National University College of Medicine, Daehak-ro, Jongno-gu, Seoul, 03080, Korea.
- Cancer Research Institute and Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Daehak-ro, Jongno-gu, Seoul, 03080, Korea.
| |
Collapse
|
5
|
Gupta JK. The Role of Aldose Reductase in Polyol Pathway: An Emerging Pharmacological Target in Diabetic Complications and Associated Morbidities. Curr Pharm Biotechnol 2024; 25:1073-1081. [PMID: 37649296 DOI: 10.2174/1389201025666230830125147] [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: 03/09/2023] [Revised: 07/09/2023] [Accepted: 08/07/2023] [Indexed: 09/01/2023]
Abstract
The expression of aldose reductase leads to a variety of biological and pathological effects. It is a multifunctional enzyme which has a tendency to reduce aldehydes to the corresponding sugar.alcohol. In diabetic conditions, the aldose reductase enzyme converts glucose into sorbitol using nicotinamide adenine dinucleotide phosphate as a cofactor. It is a key enzyme in polyol pathway which is a surrogate course of glucose metabolism. The polyol pathway has a significant impact on the aetiology of complications in individuals with end-stage diabetes. The exorbitant level of sorbitol leads to the accumulation of intracellular reactive oxygen species in diabetic heart, neurons, kidneys, eyes and other vasculatures, leading to many complications and pathogenesis. Recently, the pathophysiological role of aldose reductase has been explored with multifarious perspectives. Research on aldose reductase suggest that besides implying in diabetic complications, the enzyme also turns down the lipid-derived aldehydes as well as their glutathione conjugates. Although aldose reductase has certain lucrative role in detoxification of toxic lipid aldehydes, its overexpression leads to intracellular accumulation of sorbitol which is involved in secondary diabetic complications, such as neuropathy, cataractogenesis, nephropathy, retinopathy and cardiovascular pathogenesis. Osmotic upset and oxidative stress are produced by aldose reductase via the polyol pathway. The inhibition of aldose reductase alters the activation of transcription factors like NF-ƙB. Moreover, in many preclinical studies, aldose reductase inhibitors have been observed to reduce inflammation-related impediments, such as asthma, sepsis and colon cancer, in diabetic subjects. Targeting aldose reductase can bestow a novel cognizance for this primordial enzyme as an ingenious strategy to prevent diabetic complications and associated morbidities. In this review article, the significance of aldose reductase is briefly discussed along with their prospective applications in other afflictions.
Collapse
Affiliation(s)
- Jeetendra Kumar Gupta
- Department of Pharmacology, Institute of Pharmaceutical Research, GLA University Mathura, Uttar Pradesh, India
| |
Collapse
|
6
|
Liu L, Zhu L, Cheng Z, Sun Y, Zhou Y, Cao J. Aberrant expression of AKR1B1 indicates poor prognosis and promotes gastric cancer progression by regulating the AKT-mTOR pathway. Aging (Albany NY) 2023; 15:9661-9675. [PMID: 37751590 PMCID: PMC10564407 DOI: 10.18632/aging.205041] [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: 05/23/2023] [Accepted: 08/21/2023] [Indexed: 09/28/2023]
Abstract
Gastric cancer (GC) is a common malignant tumor in the digestive tract and a major cause of global cancer death. Due to the limited access to early screening, many patients are diagnosed with advanced GC. Therefore, postoperative radiotherapy and chemotherapy possess limited efficacy in treating GC. AKR1B1 has been associated with tumorigenesis and metastasis across various tumors, becoming a potential therapeutic target for GC. However, its role and mechanism in GC remain unclear. In this study, AKR1B1 was elevated in GC tissue, depicting a poor prognosis. AKR1B1 is closely related to age, vascular and neural invasion, lymph node metastasis, and the TNM stage of GC. The developed survival prediction model suggested that AKR1B1 expression level is crucial in the prognosis of GC patients. Moreover, the expression level of AKR1B1 in GC tissues is closely associated with the AKT-mTOR pathway. In vitro and in vivo assays functional assays helped determine the oncogenic role of AKR1B1. Additionally, the knockdown of AKR1B1 expression level in GC cell lines could effectively suppress the AKT-mTOR pathway and inhibit the proliferation and migration of tumor cells. In conclusion, this study provides a theoretical basis to establish the potential association and regulatory mechanism of AKR1B1 while offering a new strategy for GC-targeted therapy.
Collapse
Affiliation(s)
- Luojie Liu
- Department of Gastroenterology, Changshu Hospital Affiliated to Soochow University, Suzhou 215501, China
| | - Lihua Zhu
- Department of Gastroenterology, Changshu Hospital Affiliated to Soochow University, Suzhou 215501, China
| | - Zhengwu Cheng
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wannan Medical College, Wuhu 241000, China
| | - Yibin Sun
- Department of Gastroenterology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu 215005, China
| | - Yuqing Zhou
- Department of Gastroenterology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu 215005, China
| | - Jiwei Cao
- Department of General Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu 215005, China
| |
Collapse
|
7
|
Radenkovic S, Ligezka AN, Mokashi SS, Driesen K, Dukes-Rimsky L, Preston G, Owuocha LF, Sabbagh L, Mousa J, Lam C, Edmondson A, Larson A, Schultz M, Vermeersch P, Cassiman D, Witters P, Beamer LJ, Kozicz T, Flanagan-Steet H, Ghesquière B, Morava E. Tracer metabolomics reveals the role of aldose reductase in glycosylation. Cell Rep Med 2023; 4:101056. [PMID: 37257447 PMCID: PMC10313913 DOI: 10.1016/j.xcrm.2023.101056] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 03/14/2023] [Accepted: 05/04/2023] [Indexed: 06/02/2023]
Abstract
Abnormal polyol metabolism is predominantly associated with diabetes, where excess glucose is converted to sorbitol by aldose reductase (AR). Recently, abnormal polyol metabolism has been implicated in phosphomannomutase 2 congenital disorder of glycosylation (PMM2-CDG) and an AR inhibitor, epalrestat, proposed as a potential therapy. Considering that the PMM2 enzyme is not directly involved in polyol metabolism, the increased polyol production and epalrestat's therapeutic mechanism in PMM2-CDG remained elusive. PMM2-CDG, caused by PMM2 deficiency, presents with depleted GDP-mannose and abnormal glycosylation. Here, we show that, apart from glycosylation abnormalities, PMM2 deficiency affects intracellular glucose flux, resulting in polyol increase. Targeting AR with epalrestat decreases polyols and increases GDP-mannose both in patient-derived fibroblasts and in pmm2 mutant zebrafish. Using tracer studies, we demonstrate that AR inhibition diverts glucose flux away from polyol production toward the synthesis of sugar nucleotides, and ultimately glycosylation. Finally, PMM2-CDG individuals treated with epalrestat show a clinical and biochemical improvement.
Collapse
Affiliation(s)
- Silvia Radenkovic
- Department of Clinical Genomics, Mayo Clinic, Rochester, MN 55905, USA; Metabolomics Expertise Center, Center for Cancer Biology, VIB, 3000 Leuven, Belgium; Laboratory of Applied Mass Spectrometry, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium; Laboratory of Hepatology, Department of CHROMETA, KU Leuven, 3000 Leuven, Belgium.
| | - Anna N Ligezka
- Department of Clinical Genomics, Mayo Clinic, Rochester, MN 55905, USA; Department of Medical Diagnostics, Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland
| | - Sneha S Mokashi
- JC Self Research Institute, Greenwood Genetic Center, Greenwood, SC 29646, USA
| | - Karen Driesen
- Metabolomics Expertise Center, Center for Cancer Biology, VIB, 3000 Leuven, Belgium; Laboratory of Applied Mass Spectrometry, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium; Department of Development and Regeneration, Faculty of Medicine, KU Leuven, Leuven, Belgium
| | - Lynn Dukes-Rimsky
- JC Self Research Institute, Greenwood Genetic Center, Greenwood, SC 29646, USA
| | - Graeme Preston
- Department of Clinical Genomics, Mayo Clinic, Rochester, MN 55905, USA
| | - Luckio F Owuocha
- Department of Biochemistry, 117 Schweitzer Hall, University of Missouri, Columbia, MO 65211, USA
| | - Leila Sabbagh
- Department of Clinical Genomics, Mayo Clinic, Rochester, MN 55905, USA
| | - Jehan Mousa
- Department of Clinical Genomics, Mayo Clinic, Rochester, MN 55905, USA
| | - Christina Lam
- Division of Genetic Medicine, Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA; Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Andrew Edmondson
- Section of Biochemical Genetics, Division of Human Genetics, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Austin Larson
- Section of Clinical Genetics and Metabolism, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA
| | - Matthew Schultz
- Biochemical Genetics Laboratory, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | | | - David Cassiman
- Laboratory of Hepatology, Department of CHROMETA, KU Leuven, 3000 Leuven, Belgium; Metabolic Center, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Peter Witters
- Metabolic Center, University Hospitals Leuven, 3000 Leuven, Belgium; Department of Development and Regeneration, Faculty of Medicine, KU Leuven, Leuven, Belgium
| | - Lesa J Beamer
- Department of Biochemistry, 117 Schweitzer Hall, University of Missouri, Columbia, MO 65211, USA
| | - Tamas Kozicz
- Department of Clinical Genomics, Mayo Clinic, Rochester, MN 55905, USA; Biochemical Genetics Laboratory, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA; Department of Anatomy and Department of Genetics, University of Pecs Medical School, Pecs, Hungary
| | | | - Bart Ghesquière
- Metabolomics Expertise Center, Center for Cancer Biology, VIB, 3000 Leuven, Belgium; Laboratory of Applied Mass Spectrometry, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Eva Morava
- Department of Clinical Genomics, Mayo Clinic, Rochester, MN 55905, USA; Biochemical Genetics Laboratory, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA; Metabolic Center, University Hospitals Leuven, 3000 Leuven, Belgium; Department of Anatomy and Department of Genetics, University of Pecs Medical School, Pecs, Hungary.
| |
Collapse
|
8
|
Syamprasad NP, Jain S, Rajdev B, Prasad N, Kallipalli R, Naidu VGM. Aldose reductase and cancer metabolism: The master regulator in the limelight. Biochem Pharmacol 2023; 211:115528. [PMID: 37011733 DOI: 10.1016/j.bcp.2023.115528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 03/25/2023] [Accepted: 03/27/2023] [Indexed: 04/03/2023]
Abstract
It is strongly established that metabolic reprogramming mediates the initiation, progression, and metastasis of a variety of cancers. However, there is no common biomarker identified to link the dysregulated metabolism and cancer progression. Recent studies strongly advise the involvement of aldose reductase (AR) in cancer metabolism. AR-mediated glucose metabolism creates a Warburg-like effect and an acidic tumour microenvironment in cancer cells. Moreover, AR overexpression is associated with the impairment of mitochondria and the accumulation of free fatty acids in cancer cells. Further, AR-mediated reduction of lipid aldehydes and chemotherapeutics are involved in the activation of factors promoting proliferation and chemo-resistance. In this review, we have delineated the possible mechanisms by which AR modulates cellular metabolism for cancer proliferation and survival. An in-depth understanding of cancer metabolism and the role of AR might lead to the use of AR inhibitors as metabolic modulating agents for the therapy of cancer.
Collapse
Affiliation(s)
- N P Syamprasad
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research Guwahati, Sila Village, Changsari, Assam 781101, India
| | - Siddhi Jain
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research Guwahati, Sila Village, Changsari, Assam 781101, India
| | - Bishal Rajdev
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research Guwahati, Sila Village, Changsari, Assam 781101, India
| | - Neethu Prasad
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research Guwahati, Sila Village, Changsari, Assam 781101, India
| | - Ravindra Kallipalli
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research Guwahati, Sila Village, Changsari, Assam 781101, India
| | - V G M Naidu
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research Guwahati, Sila Village, Changsari, Assam 781101, India.
| |
Collapse
|
9
|
Sardelli G, Scali V, Signore G, Balestri F, Cappiello M, Mura U, Del Corso A, Moschini R. Response of a Human Lens Epithelial Cell Line to Hyperglycemic and Oxidative Stress: The Role of Aldose Reductase. Antioxidants (Basel) 2023; 12:antiox12040829. [PMID: 37107204 PMCID: PMC10135174 DOI: 10.3390/antiox12040829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/23/2023] [Accepted: 03/25/2023] [Indexed: 03/31/2023] Open
Abstract
A common feature of different types of diabetes is the high blood glucose levels, which are known to induce a series of metabolic alterations, leading to damaging events in different tissues. Among these alterations, both increased polyol pathway flux and oxidative stress are considered to play relevant roles in the response of different cells. In this work, the effect on a human lens epithelial cell line of stress conditions, consisting of exposure to either high glucose levels or to the lipid peroxidation product 4-hydroxy-2-nonenal, is reported. The occurrence of osmotic imbalance, alterations of glutathione levels, and expression of inflammatory markers was monitored. A common feature of the two stress conditions was the expression of COX-2, which, only in the case of hyperglycemic stress, occurred through NF-κB activation. In our cell model, aldose reductase activity, which is confirmed as the only activity responsible for the osmotic imbalance occurring in hyperglycemic conditions, seemed to have no role in controlling the onset of the inflammatory phenomena. However, it played a relevant role in cellular detoxification against lipid peroxidation products. These results, in confirming the multifactorial nature of the inflammatory phenomena, highlight the dual role of aldose reductase as having both damaging but also protecting activity, depending on stress conditions.
Collapse
Affiliation(s)
- Gemma Sardelli
- Biochemistry Unit, Department of Biology, University of Pisa, 56123 Pisa, Italy
| | - Viola Scali
- Biochemistry Unit, Department of Biology, University of Pisa, 56123 Pisa, Italy
| | - Giovanni Signore
- Biochemistry Unit, Department of Biology, University of Pisa, 56123 Pisa, Italy
- Interdepartmental Research Center Nutrafood “Nutraceuticals and Food for Health”, University of Pisa, 56124 Pisa, Italy
| | - Francesco Balestri
- Biochemistry Unit, Department of Biology, University of Pisa, 56123 Pisa, Italy
- Interdepartmental Research Center Nutrafood “Nutraceuticals and Food for Health”, University of Pisa, 56124 Pisa, Italy
| | - Mario Cappiello
- Biochemistry Unit, Department of Biology, University of Pisa, 56123 Pisa, Italy
- Interdepartmental Research Center Nutrafood “Nutraceuticals and Food for Health”, University of Pisa, 56124 Pisa, Italy
| | - Umberto Mura
- Biochemistry Unit, Department of Biology, University of Pisa, 56123 Pisa, Italy
| | - Antonella Del Corso
- Biochemistry Unit, Department of Biology, University of Pisa, 56123 Pisa, Italy
- Interdepartmental Research Center Nutrafood “Nutraceuticals and Food for Health”, University of Pisa, 56124 Pisa, Italy
- Correspondence: ; Tel.: +39-050-2211450
| | - Roberta Moschini
- Biochemistry Unit, Department of Biology, University of Pisa, 56123 Pisa, Italy
- Interdepartmental Research Center Nutrafood “Nutraceuticals and Food for Health”, University of Pisa, 56124 Pisa, Italy
| |
Collapse
|
10
|
Jaganjac M, Zarkovic N. Lipid Peroxidation Linking Diabetes and Cancer: The Importance of 4-Hydroxynonenal. Antioxid Redox Signal 2022; 37:1222-1233. [PMID: 36242098 DOI: 10.1089/ars.2022.0146] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Significance: It is commonly believed that diabetes mellitus may be associated with cancer. Hence, diabetic patients are at higher risk for hepatocellular carcinoma, pancreatic cancer, colorectal cancer, and breast cancer, but the mechanisms that may link these two severe diseases are not well understood. Recent Advances: A number of factors have been suggested to promote tumorigenesis in diabetic patients, including insulin resistance, hyperglycemia, dyslipidemia, inflammation, and elevated insulin-like growth factor-1 (IGF-1), which may also promote pro-oxidants, and thereby alter redox homeostasis. The consequent oxidative stress associated with lipid peroxidation appears to be a possible pathogenic link between cancer and diabetes. Critical Issues: Having summarized the above aspects of diabetes and cancer pathology, we propose that the major bioactive product of oxidative degradation of polyunsaturated fatty acids (PUFAs), the reactive aldehyde 4-hydroxynonenal (4-HNE), which is also considered a second messenger of free radicals, may be the key pathogenic factor linking diabetes and cancer. Future Directions: Because the bioactivities of 4-HNE are cell-type and concentration-dependent, are often associated with inflammation, and are involved in signaling processes that regulate antioxidant activities, proliferation, differentiation, and apoptosis, we believe that further research in this direction could reveal options for better control of diabetes and cancer. Controlling the production of 4-HNE to avoid its cytotoxicity to normal but not cancer cells while preventing its diabetogenic activities could be an important aspect of modern integrative biomedicine. Antioxid. Redox Signal. 37, 1222-1233.
Collapse
Affiliation(s)
- Morana Jaganjac
- Laboratory for Oxidative Stress, Division of Molecular Medicine, Ruder Boskovic Institute, Zagreb, Croatia
| | - Neven Zarkovic
- Laboratory for Oxidative Stress, Division of Molecular Medicine, Ruder Boskovic Institute, Zagreb, Croatia
| |
Collapse
|
11
|
Novel Pyridinium Based Ionic Liquid Promoter for Aqueous Knoevenagel Condensation: Green and Efficient Synthesis of New Derivatives with Their Anticancer Evaluation. Molecules 2022; 27:molecules27092940. [PMID: 35566291 PMCID: PMC9105511 DOI: 10.3390/molecules27092940] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 04/30/2022] [Accepted: 05/01/2022] [Indexed: 11/22/2022] Open
Abstract
Herein, a distinctive dihydroxy ionic liquid ([Py-2OH]OAc) was straightforwardly assembled from the sonication of pyridine with 2-chloropropane-1,3-diol by employing sodium acetate as an ion exchanger. The efficiency of the ([Py-2OH]OAc as a promoter for the sono-synthesis of a novel library of condensed products through DABCO-catalyzed Knoevenagel condensation process of adequate active cyclic methylenes and ninhydrin was next investigated using ultimate greener conditions. All of the reactions studied went cleanly and smoothly, and the resulting Knoevenagel condensation compounds were recovered in high yields without detecting the aldol intermediates in the end products. Compared to traditional strategies, the suggested approach has numerous advantages including mild reaction conditions with no by-products, eco-friendly solvent, outstanding performance in many green metrics, and usability in gram-scale synthesis. The reusability of the ionic liquid was also studied, with an overall retrieved yield of around 97% for seven consecutive runs without any substantial reduction in the performance. The novel obtained compounds were further assessed for their in vitro antitumor potential toward three human tumor cell lines: Colo-205 (colon cancer), MCF-7 (breast cancer), and A549 (lung cancer) by employing the MTT assay, and the findings were evaluated with the reference Doxorubicin. The results demonstrated that the majority of the developed products had potent activities at very low doses. Compounds comprising rhodanine (5) or chromane (12) moieties exhibited the most promising cytotoxic effects toward three cell lines, particularly rhodanine carboxylic acid derivative (5c), showing superior cytotoxic effects against the investigated cell lines compared to the reference drug. Furthermore, automated docking simulation studies were also performed to support the results obtained.
Collapse
|
12
|
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: 2.0] [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
|
13
|
Singh M, Kapoor A, Bhatnagar A. Physiological and Pathological Roles of Aldose Reductase. Metabolites 2021; 11:655. [PMID: 34677370 PMCID: PMC8541668 DOI: 10.3390/metabo11100655] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/17/2021] [Accepted: 09/17/2021] [Indexed: 12/15/2022] Open
Abstract
Aldose reductase (AR) is an aldo-keto reductase that catalyzes the first step in the polyol pathway which converts glucose to sorbitol. Under normal glucose homeostasis the pathway represents a minor route of glucose metabolism that operates in parallel with glycolysis. However, during hyperglycemia the flux of glucose via the polyol pathway increases significantly, leading to excessive formation of sorbitol. The polyol pathway-driven accumulation of osmotically active sorbitol has been implicated in the development of secondary diabetic complications such as retinopathy, nephropathy, and neuropathy. Based on the notion that inhibition of AR could prevent these complications a range of AR inhibitors have been developed and tested; however, their clinical efficacy has been found to be marginal at best. Moreover, recent work has shown that AR participates in the detoxification of aldehydes that are derived from lipid peroxidation and their glutathione conjugates. Although in some contexts this antioxidant function of AR helps protect against tissue injury and dysfunction, the metabolic transformation of the glutathione conjugates of lipid peroxidation-derived aldehydes could also lead to the generation of reactive metabolites that can stimulate mitogenic or inflammatory signaling events. Thus, inhibition of AR could have both salutary and injurious outcomes. Nevertheless, accumulating evidence suggests that inhibition of AR could modify the effects of cardiovascular disease, asthma, neuropathy, sepsis, and cancer; therefore, additional work is required to selectively target AR inhibitors to specific disease states. Despite past challenges, we opine that a more gainful consideration of therapeutic modulation of AR activity awaits clearer identification of the specific role(s) of the AR enzyme in health and disease.
Collapse
Affiliation(s)
- Mahavir Singh
- Eye and Vision Science Laboratory, Department of Physiology, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Aniruddh Kapoor
- Internal Medicine—Critical Care, School of Medicine, Saint Louis University, St. Louis, MO 63141, USA;
| | - Aruni Bhatnagar
- Christina Lee Brown Envirome Institute, School of Medicine, University of Louisville, Louisville, KY 40202, USA;
| |
Collapse
|
14
|
Xu Z, Qu H, Ren Y, Gong Z, Ri HJ, Chen X. An Update on the Potential Roles of E2F Family Members in Colorectal Cancer. Cancer Manag Res 2021; 13:5509-5521. [PMID: 34276228 PMCID: PMC8277564 DOI: 10.2147/cmar.s320193] [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: 05/14/2021] [Accepted: 07/01/2021] [Indexed: 12/24/2022] Open
Abstract
Colorectal cancer (CRC) is a major health burden worldwide, and thus, optimised diagnosis and treatments are imperative. E2F transcription factors (E2Fs) are a family of transcription factors consisting of eight genes, contributing to the oncogenesis and development of CRC. Importantly, E2Fs control not only the cell cycle but also apoptosis, senescence, DNA damage response, and drug resistance by interacting with multiple signaling pathways. However, the specific functions and intricate machinery of these eight E2Fs in human CRC remain unclear in many respects. Evidence on E2Fs and CRC has been scattered on the related regulatory genes, microRNAs (miRNAs), and competing endogenous RNAs (ceRNAs). Accordingly, some drugs targeting E2Fs have been transferred from preclinical to clinical application. Herein, we have systemically reviewed the current literature on the roles of various E2Fs in CRC with the purpose of providing possible clinical implications for patient diagnosis and prognosis and future treatment strategy design, thereby furthering the understanding of the E2Fs.
Collapse
Affiliation(s)
- ZhaoHui Xu
- Department of Hernia and Colorectal Surgery, The Second Hospital of Dalian Medical University, Dalian, 116023, People's Republic of China
| | - Hui Qu
- Department of Hernia and Colorectal Surgery, The Second Hospital of Dalian Medical University, Dalian, 116023, People's Republic of China
| | - YanYing Ren
- Department of Hernia and Colorectal Surgery, The Second Hospital of Dalian Medical University, Dalian, 116023, People's Republic of China
| | - ZeZhong Gong
- Department of Hernia and Colorectal Surgery, The Second Hospital of Dalian Medical University, Dalian, 116023, People's Republic of China
| | - Hyok Ju Ri
- Department of Hernia and Colorectal Surgery, The Second Hospital of Dalian Medical University, Dalian, 116023, People's Republic of China
| | - Xin Chen
- Department of Hernia and Colorectal Surgery, The Second Hospital of Dalian Medical University, Dalian, 116023, People's Republic of China
| |
Collapse
|
15
|
Sonowal H, Ramana KV. Development of Aldose Reductase Inhibitors for the Treatment of Inflammatory Disorders and Cancer: Current Drug Design Strategies and Future Directions. Curr Med Chem 2021; 28:3683-3712. [PMID: 33109031 DOI: 10.2174/0929867327666201027152737] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 09/22/2020] [Accepted: 09/24/2020] [Indexed: 11/22/2022]
Abstract
Aldose Reductase (AR) is an enzyme that converts glucose to sorbitol during the polyol pathway of glucose metabolism. AR has been shown to be involved in the development of secondary diabetic complications due to its involvement in causing osmotic as well as oxidative stress. Various AR inhibitors have been tested for their use to treat secondary diabetic complications, such as retinopathy, neuropathy, and nephropathy in clinical studies. Recent studies also suggest the potential role of AR in mediating various inflammatory complications. Therefore, the studies on the development and potential use of AR inhibitors to treat inflammatory complications and cancer besides diabetes are currently on the rise. Further, genetic mutagenesis studies, computer modeling, and molecular dynamics studies have helped design novel and potent AR inhibitors. This review discussed the potential new therapeutic use of AR inhibitors in targeting inflammatory disorders and cancer besides diabetic complications. Further, we summarized studies on how AR inhibitors have been designed and developed for therapeutic purposes in the last few decades.
Collapse
Affiliation(s)
- Himangshu Sonowal
- Moores Cancer Center, University of California San Diego, La Jolla, California 92037, United States
| | - Kota V Ramana
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, United States
| |
Collapse
|
16
|
Aldo Keto Reductases AKR1B1 and AKR1B10 in Cancer: Molecular Mechanisms and Signaling Networks. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1347:65-82. [PMID: 33945128 DOI: 10.1007/5584_2021_634] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Deregulation of metabolic pathways has increasingly been appreciated as a major driver of cancer in recent years. The principal cancer-associated alterations in metabolism include abnormal uptake of glucose and amino acids and the preferential use of metabolic pathways for the production of biomass and nicotinamide adenine dinucleotide phosphate (NADPH). Aldo-keto reductases (AKRs) are NADPH dependent cytosolic enzymes that can catalyze the reduction of carbonyl groups to primary and secondary alcohols using electrons from NADPH. Aldose reductase, also known as AKR1B1, catalyzes the conversion of excess glucose to sorbitol and has been studied extensively for its role in a number of diabetic pathologies. In recent years, however, high expression of the AKR1B and AKR1C family of enzymes has been strongly associated with worse outcomes in different cancer types. This review provides an overview of the catalysis-dependent and independent data emerging on the molecular mechanisms of the functions of AKRBs in different tumor models with an emphasis of the role of these enzymes in chemoresistance, inflammation, oxidative stress and epithelial-to-mesenchymal transition.
Collapse
|
17
|
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: 42] [Impact Index Per Article: 10.5] [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
|
18
|
Khedr MA, Al-Wabli RI, Almutairi MS, Zaghary WA. QSAR-based rational discovery of novel substituted-4'-iminospiro[indoline-3,3'-[1,2,5]thiadiazolidinyl]-2-one 1',1'-dioxide with potent in vitro anticancer activity. BMC Chem 2019; 13:3. [PMID: 31355364 PMCID: PMC6659568 DOI: 10.1186/s13065-019-0520-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 01/16/2019] [Indexed: 11/10/2022] Open
Abstract
Recent studies have suggested that aldose reductase inhibition preferentially inhibits the growth of cancer cells. However, the investigations of this issue are not many. Novel nine substituted- 4'-iminospiro[indoline-3,3'-[1,2,5] thiadiazolidinyl]-2-one 1',1'-dioxide derivatives were designed by both isosteric replacement of the imidazolidine-2,5-dione moiety in spirohydantoin scaffold and conformational rigidification approaches. A QSAR with high predictive power (r2 = 0.99) was created from a series of potent aldose reductase inhibitors and was used to predict the activity of our new compounds. Compound 5 showed the best docking score (- 33.24 kcal/mol) with the least RMSD value (< 1.5) obtained by molecular dynamic simulations over 20 ns. All compounds showed promising anticancer activities especially compound 5 that achieved the highest inhibitory activities with IC50; 0.013, 0.031, 0.064, and 0.048 mmol/L against breast, colon, prostate, and lung cell lines respectively. The discovery of this lead compound confirmed the rational design. Further investigations may be required for optimization of this compound.
Collapse
Affiliation(s)
- Mohammed A. Khedr
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Helwan University, Ein Helwan, Cairo, 11790 Egypt
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al Hasa, 31982 Saudi Arabia
| | - Reem I. Al-Wabli
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, 11451 Saudi Arabia
| | - Maha S. Almutairi
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, 11451 Saudi Arabia
| | - Wafaa A. Zaghary
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Helwan University, Ein Helwan, Cairo, 11790 Egypt
| |
Collapse
|
19
|
Saxena A, Tammali R, Ramana KV, Srivastava SK. Aldose Reductase Inhibitor, Fidarestat Prevents High-fat Diet-induced Intestinal Polyps in Apc Min/+ Mice. Curr Cancer Drug Targets 2019; 18:905-911. [PMID: 28786349 DOI: 10.2174/1568009617666170808105633] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 06/08/2017] [Accepted: 06/29/2017] [Indexed: 12/12/2022]
Abstract
BACKGROUND Recent epidemiological and experimental studies have shown that obesity is a major risk factor for Colorectal Cancer (CRC). Regular intake of high fat-containing diet can promote obesity and metabolic syndrome by increasing the insulin resistance and inflammatory response which contribute to carcinogenesis. Previously, we have shown that inhibition of polyol pathway enzyme aldose reductase (AR) prevents carcinogens- and inflammatory growth factorsinduced CRC. However, the effect of AR inhibition on a high-fat diet (HFD)-induced formation of intestinal polyps in Apc-deficient Min (multiple intestinal neoplasia; ApcMin/+) mice is not known. METHODS We examined the effect of AR inhibitor, fidarestat on the HFD-induced formation of preneoplastic intestinal polyps in ApcMin/+ mice which is an excellent model of colon cancer. RESULTS APCMin/+ mice fed for 12 weeks of HFD caused a significant increase in the formation of polyps in the small and large intestines and fidarestat given along with the HFD prevented the number of intestinal polyps. Fidarestat also decreased the size of the polyps in the intestines of HFDtreated APC Min mice. Further, the expression levels of beta-catenin, PCNA, PKC-β2, P-AKT, Pp65, COX-2, and iNOS in the small and large intestines of HFD-treated mice significantly increased, and AR inhibitor prevented it. CONCLUSION Our results thus suggest that fidarestat could be used as a potential chemopreventive drug for intestinal cancers due to APC gene mutations.
Collapse
Affiliation(s)
- Ashish Saxena
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Ravinder Tammali
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Kota V Ramana
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Satish K Srivastava
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas 77555, United States
| |
Collapse
|
20
|
Karpisheh V, Nikkhoo A, Hojjat-Farsangi M, Namdar A, Azizi G, Ghalamfarsa G, Sabz G, Yousefi M, Yousefi B, Jadidi-Niaragh F. Prostaglandin E2 as a potent therapeutic target for treatment of colon cancer. Prostaglandins Other Lipid Mediat 2019; 144:106338. [PMID: 31100474 DOI: 10.1016/j.prostaglandins.2019.106338] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 03/30/2019] [Accepted: 05/10/2019] [Indexed: 02/07/2023]
Abstract
Although colon cancer is one of the most important triggers of cancer related mortality, a few therapeutic options exist for this disease, including combination chemotherapy, anti-EGFR and anti-angiogenic agents. However, none of these therapeutics are fully effective for complete remission, and this issue needs further investigations, particularly in the patients with advanced disease. It has been shown that colon carcinogenesis process is associated with upregulation of prostaglandin (PG) levels. Moreover, conversion of pre-malignant cells to malignant was also related with increased generation of PGs in susceptible subjects. Among the prostanoids, PGE2 is the most important produced member which generated in high levels by colon tumor cells. Generation of PGE2 by action of cyclooxygenase (COX)-2 can promote growth and development, resistance to apoptosis, proliferation, invasion and metastasis, angiogenesis and drug resistance in colon cancer. Increased levels of PGE2 and COX-2 in colon cancer is reported by various investigators which was associated with disease progression. It is suggested that there is a positive feedback loop between COX-2 and PGE2, in which function of COX-2 induces generation of PGE2, and upregulation of PGE2 increases the expression of COX-2 in colon cancer. Although an existence of this feedback loop is well-documented, its precise mechanism, signaling pathways, and the particular E-type prostanoid (EP) receptor mediating this feedback are elusive. Therefore, it seems that targeting COX-2/PGE2/EP receptors may be supposed as a potent therapeutic strategy for treatment of colon cancer. In this review, we try to clarify the role of PGE2 in cancer progression and its targeting for treatment of colon cancer.
Collapse
Affiliation(s)
- Vahid Karpisheh
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Afshin Nikkhoo
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Hojjat-Farsangi
- Bioclinicum, Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden; The Persian Gulf Marine Biotechnology Medicine Research Center, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Afshin Namdar
- Department of Dentistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, T6G 2E1 Canada
| | - Gholamreza Azizi
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Ghasem Ghalamfarsa
- Cellular and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Gholamabas Sabz
- Cellular and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Mehdi Yousefi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Bahman Yousefi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Farhad Jadidi-Niaragh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
21
|
Banala VT, Urandur S, Sharma S, Sharma M, Shukla RP, Marwaha D, Gautam S, Dwivedi M, Mishra PR. Targeted co-delivery of the aldose reductase inhibitor epalrestat and chemotherapeutic doxorubicin via a redox-sensitive prodrug approach promotes synergistic tumor suppression. Biomater Sci 2019; 7:2889-2906. [DOI: 10.1039/c9bm00221a] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Redox responsive epalrestat prodrug micelles facilitate synergistic concentrations of doxorubicin with an advantage of CD44 down-regulation and reduced cardiotoxicity.
Collapse
Affiliation(s)
- Venkatesh Teja Banala
- Division of Pharmaceutics and Pharmacokinetics
- CSIR-Central Drug Research Institute Lucknow
- India
| | - Sandeep Urandur
- Division of Pharmaceutics and Pharmacokinetics
- CSIR-Central Drug Research Institute Lucknow
- India
| | - Shweta Sharma
- Division of Pharmaceutics and Pharmacokinetics
- CSIR-Central Drug Research Institute Lucknow
- India
| | - Madhu Sharma
- Division of Pharmaceutics and Pharmacokinetics
- CSIR-Central Drug Research Institute Lucknow
- India
| | - Ravi P. Shukla
- Division of Pharmaceutics and Pharmacokinetics
- CSIR-Central Drug Research Institute Lucknow
- India
| | - Disha Marwaha
- Division of Pharmaceutics and Pharmacokinetics
- CSIR-Central Drug Research Institute Lucknow
- India
| | - Shalini Gautam
- Division of Pharmaceutics and Pharmacokinetics
- CSIR-Central Drug Research Institute Lucknow
- India
| | - Monika Dwivedi
- Division of Pharmaceutics and Pharmacokinetics
- CSIR-Central Drug Research Institute Lucknow
- India
| | - Prabhat Ranjan Mishra
- Division of Pharmaceutics and Pharmacokinetics
- CSIR-Central Drug Research Institute Lucknow
- India
| |
Collapse
|
22
|
Regulation of aldose reductase activity by tubulin and phenolic acid derivates. Arch Biochem Biophys 2018; 654:19-26. [DOI: 10.1016/j.abb.2018.07.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 06/29/2018] [Accepted: 07/11/2018] [Indexed: 12/26/2022]
|
23
|
Shukla K, Sonowal H, Saxena A, Ramana KV, Srivastava SK. Aldose reductase inhibitor, fidarestat regulates mitochondrial biogenesis via Nrf2/HO-1/AMPK pathway in colon cancer cells. Cancer Lett 2017; 411:57-63. [PMID: 28986187 PMCID: PMC5693654 DOI: 10.1016/j.canlet.2017.09.031] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 09/18/2017] [Accepted: 09/21/2017] [Indexed: 12/25/2022]
Abstract
Although we have shown earlier that aldose reductase (AR) inhibitors prevent colorectal cancer cell (CRC) growth in culture as well as in nude mice xenografts, the mechanism(s) is not well understood. In this study, we have investigated how AR inhibition prevents CRC growth by regulating the mitochondrial biogenesis via Nrf2/HO-1 pathway. Incubation of CRC cells such as SW-480, HT29, and HCT116 with AR inhibitor, fidarestat that non-covalently binds to the enzyme, increases the expression of Nrf2. Further, fidarestat augmented the EGF-induced expression of Nrf2 in CRC cells. Fidarestat also increased the Nrf2 -DNA binding activity as well as expression of HO-1 and NQO1 and activation of SOD and catalase in SW480 cells. Similarly, in nude mice xenograft tumor tissues, Nrf2 and HO-1 levels were significantly higher in fidarestat-treated mice compared to controls. Further, stimulation of CRC cells with EGF in the presence of fidarestat increased the mRNA levels of PGC-1α, Nrf1 and TFAM and protein levels of PGC-1α, TFAM and COX-IV and decreased the mitochondrial DNA damage as measured by 8-hydroxy-2'-deoxyguanosine levels. AR inhibitor also modulated the phosphorylations of AMPK and mTOR and expression of p53 in EGF-treated cells. Collectively, our results indicate that AR inhibitor prevents CRC growth by increasing mitochondrial biogenesis via increasing the expression of Nrf2/HO-1/AMPK/p53 and decreasing the mitochondrial DNA damage.
Collapse
Affiliation(s)
- Kirtikar Shukla
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Himangshu Sonowal
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Ashish Saxena
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Kota V Ramana
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Satish K Srivastava
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA.
| |
Collapse
|
24
|
Awasthi YC, Ramana KV, Chaudhary P, Srivastava SK, Awasthi S. Regulatory roles of glutathione-S-transferases and 4-hydroxynonenal in stress-mediated signaling and toxicity. Free Radic Biol Med 2017; 111:235-243. [PMID: 27794453 PMCID: PMC5643026 DOI: 10.1016/j.freeradbiomed.2016.10.493] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 10/20/2016] [Accepted: 10/22/2016] [Indexed: 12/17/2022]
Abstract
Glutathione-S-Transferases (GSTs) have primarily been thought to be xenobiotic metabolizing enzymes that protect cells from toxic drugs and environmental electrophiles. However, in last three decades, these enzymes have emerged as the regulators of oxidative stress-induced signaling and toxicity. 4-Hydroxy-trans 2-nonenal (HNE) an end-product of lipid peroxidation, has been shown to be a major determinant of oxidative stress-induced signaling and toxicity. HNE is involved in signaling pathways, including apoptosis, proliferation, modulation of gene expression, activation of transcription factors/repressors, cell cycle arrest, and differentiation. In this article, available evidence for a major role of GSTs in the regulation of HNE-mediated cell signaling processes through modulation of the intracellular levels of HNE is discussed.
Collapse
Affiliation(s)
- Yogesh C Awasthi
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA.
| | - Kota V Ramana
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA.
| | - Pankaj Chaudhary
- Department of Molecular and Medical Genetics, University of North Texas Health Science Center, Fort worth, TX 76107, USA
| | - Satish K Srivastava
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Sanjay Awasthi
- Department of Internal Medicine-Oncology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| |
Collapse
|
25
|
Guéraud F. 4-Hydroxynonenal metabolites and adducts in pre-carcinogenic conditions and cancer. Free Radic Biol Med 2017; 111:196-208. [PMID: 28065782 DOI: 10.1016/j.freeradbiomed.2016.12.025] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 12/14/2016] [Accepted: 12/17/2016] [Indexed: 12/22/2022]
Abstract
4-hydroxy-2-nonenal (HNE) is an amazing reactive compound, originating from lipid peroxidation within cells but also in food and considered as a "second messenger" of oxidative stress. Due to its chemical features, HNE is able to make covalent links with DNA, proteins and lipids. The aim of this review is to give a comprehensive summary of the chemical properties of HNE and of the consequences of its reactivity in relation to cancer development. The formation of exocyclic etheno-and propano-adducts and genotoxic effects are addressed. The adduction to cellular proteins and the repercussions on the regulation of cell signaling pathways involved in cancer development are reviewed, notably on the Nrf2/Keap1/ARE pathway. The metabolic pathways leading to the inactivation/elimination or, on the contrary, to the bioactivation of HNE are considered. A special focus is given on the link between HNE and colorectal cancer development, due to its occurrence in foodstuffs and in the digestive lumen, during digestion.
Collapse
Affiliation(s)
- Françoise Guéraud
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRA, ENVT, INP-Purpan, UPS, Toulouse, France.
| |
Collapse
|
26
|
Gasparovic AC, Milkovic L, Sunjic SB, Zarkovic N. Cancer growth regulation by 4-hydroxynonenal. Free Radic Biol Med 2017; 111:226-234. [PMID: 28131901 DOI: 10.1016/j.freeradbiomed.2017.01.030] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 01/16/2017] [Accepted: 01/18/2017] [Indexed: 02/07/2023]
Abstract
While reactive oxygen species (ROS) gain their carcinogenic effects by DNA mutations, if generated in the vicinity of genome, lipid peroxidation products, notably 4-hydroxynonenal (HNE), have much more complex modes of activities. Namely, while ROS are short living and have short efficiency distance range (in nm or µm) HNE has strong binding affinity for proteins, thus forming relatively stable adducts. Hence, HNE can diffuse from the site or origin changing structure and function of respective proteins. Consequently HNE can influence proliferation, differentiation and apoptosis of cancer cells on one hand, while on the other it can affect genome functionality, too. Although HNE is considered to be important factor of carcinogenesis due to its ability to covalently bind to DNA, it might also be cytotoxic for cancer cells, as well as it can modulate their growth. In addition to direct cytotoxicity, HNE is also involved in activity mechanisms by which several cytostatic drugs and radiotherapy exhibit their anticancer effects. Complementary to that, the metabolic pathway for HNE detoxification through RLIP76, which is enhanced in cancer, may be a target for anti-cancer treatments. In addition, some cancer cells can undergo apoptosis or necrosis, if exposed to supraphysiological HNE levels in the cancer microenvironment, especially if challenged additionally by pro-oxidative cytostatics and/or inflammation. These findings could explain previously observed disappearance of HNE from invading cancer cells, which is associated with the increase of HNE in non-malignant cells close to invading cancer utilizing cardiolipin as the source of cancer-inhibiting HNE.
Collapse
Affiliation(s)
| | | | | | - Neven Zarkovic
- Rudjer Boskovic Institute, Bijenicka 54, Zagreb, Croatia.
| |
Collapse
|
27
|
Taskoparan B, Seza EG, Demirkol S, Tuncer S, Stefek M, Gure AO, Banerjee S. Opposing roles of the aldo-keto reductases AKR1B1 and AKR1B10 in colorectal cancer. Cell Oncol (Dordr) 2017; 40:563-578. [PMID: 28929377 DOI: 10.1007/s13402-017-0351-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/31/2017] [Indexed: 12/12/2022] Open
Abstract
PURPOSE Aldo-keto reductases (including AKR1B1 and AKR1B10) constitute a family of oxidoreductases that have been implicated in the pathophysiology of diabetes and cancer, including colorectal cancer (CRC). Available data indicate that, despite their similarities in structure and enzymatic functions, their roles in CRC may be divergent. Here, we aimed to determine the expression and functional implications of AKR1B1 and AKR1B10 in CRC. METHODS AKR1B1 and AKR1B10 gene expression levels were analyzed using publicly available microarray data and ex vivo CRC-derived cDNA samples. Gene Set Enrichment Analysis (GSEA), The Cancer Genome Atlas (TCGA) RNA-seq data and The Cancer Proteome Atlas (TCPA) proteome data were analyzed to determine the effect of high and low AKR1B1 and AKR1B10 expression levels in CRC patients. Proliferation, cell cycle progression, cellular motility, adhesion and inflammation were determined in CRC-derived cell lines in which these genes were either exogenously overexpressed or silenced. RESULTS We found that the expression of AKR1B1 was unaltered, whereas that of AKR1B10 was decreased in primary CRCs. GSEA revealed that, while high AKR1B1 expression was associated with increased cell cycle progression, cellular motility and inflammation, high AKR1B10 expression was associated with a weak inflammatory phenotype. Functional studies carried out in CRC-derived cell lines confirmed these data. Microarray data analysis indicated that high expression levels of AKR1B1 and AKR1B10 were significantly associated with shorter and longer disease-free survival rates, respectively. A combined gene expression signature of AKR1B10 (low) and AKR1B1 (high) showed a better prognostic stratification of CRC patients independent of confounding factors. CONCLUSIONS Despite their similarities, the expression levels and functions of AKR1B1 and AKR1B10 are highly divergent in CRC, and they may have prognostic implications.
Collapse
Affiliation(s)
- Betul Taskoparan
- Department of Biological Sciences, Orta Doğu Teknik Üniversitesi (ODTU/METU), Ankara, Turkey
| | - Esin Gulce Seza
- Department of Biological Sciences, Orta Doğu Teknik Üniversitesi (ODTU/METU), Ankara, Turkey
| | - Secil Demirkol
- Department of Molecular Biology and Genetics, Bilkent Üniversitesi, Ankara, Turkey
| | - Sinem Tuncer
- Department of Biological Sciences, Orta Doğu Teknik Üniversitesi (ODTU/METU), Ankara, Turkey
| | - Milan Stefek
- Department of Biochemical Pharmacology, Institute of Experimental Pharmacology and Toxicology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Ali Osmay Gure
- Department of Molecular Biology and Genetics, Bilkent Üniversitesi, Ankara, Turkey
| | - Sreeparna Banerjee
- Department of Biological Sciences, Orta Doğu Teknik Üniversitesi (ODTU/METU), Ankara, Turkey.
| |
Collapse
|
28
|
Blockage of NOX2/MAPK/NF- κB Pathway Protects Photoreceptors against Glucose Deprivation-Induced Cell Death. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:5093473. [PMID: 29085555 PMCID: PMC5611869 DOI: 10.1155/2017/5093473] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2017] [Revised: 07/08/2017] [Accepted: 07/18/2017] [Indexed: 01/08/2023]
Abstract
Acute energy failure is one of the critical factors contributing to the pathogenic mechanisms of retinal ischemia. Our previous study demonstrated that glucose deprivation can lead to a caspase-dependent cell death of photoreceptors. The aim of this study was to decipher the upstream signal pathway in glucose deprivation- (GD-) induced cell death. We mimicked acute energy failure by using glucose deprivation in photoreceptor cells (661W cells). GD-induced oxidative stress was evaluated by measuring ROS with the DCFH-DA assay and HO-1 expression by Western blot analysis. The activation of NOX2/MAPK/NF-κB signal was assessed by Western blot and immunohistochemical assays. The roles of these signals in GD-induced cell death were measured by using their specific inhibitors. Inhibition of Rac-1 and NOX2 suppressed GD-induced oxidative stress and protected photoreceptors against GD-induced cell death. NOX2 was an upstream signal in the caspase-dependent cell death cascade, yet the downstream MAPK pathways were activated and blocking MAPK signals rescued 661W cells from GD-induced death. In addition, GD caused the activation of NF-κB signal and inhibiting NF-κB significantly protected 661W cells. These observations may provide insights for treating retinal ischemic diseases and protecting retinal neurons from ischemia-induced cell death.
Collapse
|
29
|
Dutta S, Kharkar PS, Sahu NU, Khanna A. Molecular docking prediction and in vitro studies elucidate anti-cancer activity of phytoestrogens. Life Sci 2017; 185:73-84. [PMID: 28720470 DOI: 10.1016/j.lfs.2017.07.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2017] [Revised: 07/02/2017] [Accepted: 07/13/2017] [Indexed: 02/08/2023]
|
30
|
Liu B, Fu XQ, Li T, Su T, Guo H, Zhu PL, Tse AKW, Liu SM, Yu ZL. Computational and experimental prediction of molecules involved in the anti-melanoma action of berberine. JOURNAL OF ETHNOPHARMACOLOGY 2017; 208:225-235. [PMID: 28729227 DOI: 10.1016/j.jep.2017.07.023] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 05/07/2017] [Accepted: 07/15/2017] [Indexed: 06/07/2023]
Abstract
ETHNOPHARMACOLOGIC RELEVANCE Berberine (BBR) is a naturally occurring alkaloid compound that can be found in Chinese medicinal herbs such as Rhizoma Coptidis and Phellodendri Cortex. These BBR containing herbs are commonly used by Chinese medicine doctors to treat cancers including melanoma. In this study, we explored proteins potentially involved in the anti-melanoma effects of BBR using computational and experimental approaches. MATERIALS AND METHODS Target proteins of BBR were predicted using the reverse pharmacophore screening, molecular docking and molecular dynamics. Anti-melanoma activities of BBR in melanoma cells were examined by MTT and EdU proliferation assays. Effects of BBR on activities of target proteins in melanoma cells were examined by Western blotting or fluorescence assay. RESULTS Ten proteins implicated in cancer and with high fit-score in the reverse pharmacophore screening were selected as potential targets of BBR. Molecular docking and molecular dynamics revealed that BBR could stably bind to four of the ten proteins, namely 3-phosphoinositide-dependent protein kinase 1 (PDK1), glucocorticoid receptor (GR), p38 mitogen-activated protein kinase (p38) and dihydroorotate dehydrogenase (DHODH). Cellular experiments showed that BBR inhibited cell proliferation, increased the phosphorylation of GR and p38, and inhibited the activity of DHODH in A375 human melanoma cells. CONCLUSIONS These findings suggest that p38, GR and DHODH are potentially involved in the anti-melanoma action of BBR. This study provided a chemical and pharmacological justification for the clinical use of BBR-containing herbs in melanoma treatment.
Collapse
Affiliation(s)
- Bin Liu
- Center for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; Consun Chinese Medicines Research Centre for Renal Diseases, Hong Kong Baptist University, Hong Kong, China; HKBU Shenzhen Research Institute and Continuing Education, Shenzhen, China; Guangzhou Institute of Cardiovascular Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiu-Qiong Fu
- Center for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; Consun Chinese Medicines Research Centre for Renal Diseases, Hong Kong Baptist University, Hong Kong, China; HKBU Shenzhen Research Institute and Continuing Education, Shenzhen, China
| | - Ting Li
- Center for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; Consun Chinese Medicines Research Centre for Renal Diseases, Hong Kong Baptist University, Hong Kong, China; HKBU Shenzhen Research Institute and Continuing Education, Shenzhen, China
| | - Tao Su
- Center for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; Consun Chinese Medicines Research Centre for Renal Diseases, Hong Kong Baptist University, Hong Kong, China; HKBU Shenzhen Research Institute and Continuing Education, Shenzhen, China
| | - Hui Guo
- Center for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; Consun Chinese Medicines Research Centre for Renal Diseases, Hong Kong Baptist University, Hong Kong, China; HKBU Shenzhen Research Institute and Continuing Education, Shenzhen, China
| | - Pei-Li Zhu
- Center for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; Consun Chinese Medicines Research Centre for Renal Diseases, Hong Kong Baptist University, Hong Kong, China; HKBU Shenzhen Research Institute and Continuing Education, Shenzhen, China
| | - Anfernee Kai-Wing Tse
- Center for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; Consun Chinese Medicines Research Centre for Renal Diseases, Hong Kong Baptist University, Hong Kong, China; HKBU Shenzhen Research Institute and Continuing Education, Shenzhen, China
| | - Shi-Ming Liu
- Guangzhou Institute of Cardiovascular Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| | - Zhi-Ling Yu
- Center for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; Consun Chinese Medicines Research Centre for Renal Diseases, Hong Kong Baptist University, Hong Kong, China; HKBU Shenzhen Research Institute and Continuing Education, Shenzhen, China.
| |
Collapse
|
31
|
Patras L, Sylvester B, Luput L, Sesarman A, Licarete E, Porfire A, Muntean D, Drotar DM, Rusu AD, Nagy AL, Catoi C, Tomuta I, Vlase L, Banciu M, Achim M. Liposomal prednisolone phosphate potentiates the antitumor activity of liposomal 5-fluorouracil in C26 murine colon carcinoma in vivo. Cancer Biol Ther 2017; 18:616-626. [PMID: 28696813 DOI: 10.1080/15384047.2017.1345392] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The antitumor efficacy of 5-fluorouracil (5-FU) in advanced colorectal cancer (CRC) is hindered not only by the low therapeutic index, but also by tumor cell resistance to this cytotoxic drug. Therefore, to enhance the 5-FU antitumor activity, the present research used a novel tumor-targeted therapy based on the co-administration of 5-FU encapsulated in long-circulating liposomes (LCL-5-FU) together with liposomal prednisolone phosphate (LCL-PLP), a formulation with known anti-angiogenic actions on C26 murine colon carcinoma cells. Thus, we assessed the in vivo effects of the combined liposomal drug therapy on C26 carcinoma growth as well as on the production of molecular markers with key roles in tumor development such as angiogenic, inflammatory, and oxidative stress molecules. To get further insight into the polarization state of tumor microenvironment after the treatment, we determined the IL-10/IL-12p70 ratio in tumors. Our results showed that combined liposomal drug therapy inhibited almost totally tumor growth and was superior as antitumor activity to both single liposomal drug therapies tested. The antitumor efficacy of the combined therapy was mainly related to the anti-angiogenic and anti-inflammatory actions on C26 carcinoma milieu, being favored by its controlling effect on intratumor oxidative stress and the skewing of polarization of tumor microenvironmental cells toward their antineoplastic phenotypes. Thus, our study unveils a promising treatment strategy for CRC that should be furthermore considered.
Collapse
Affiliation(s)
- Laura Patras
- a Department of Molecular Biology and Biotechnology , Faculty of Biology and Geology, Babes-Bolyai University , Cluj-Napoca , Romania.,b Molecular Biology Centre , Institute for Interdisciplinary Research in Bio-Nano-Sciences, Babes-Bolyai University , Cluj-Napoca , Romania
| | - Bianca Sylvester
- c Department of Pharmaceutical Technology and Biopharmaceutics , Faculty of Pharmacy, Iuliu Hatieganu University of Medicine and Pharmacy , Cluj-Napoca , Romania
| | - Lavinia Luput
- a Department of Molecular Biology and Biotechnology , Faculty of Biology and Geology, Babes-Bolyai University , Cluj-Napoca , Romania.,b Molecular Biology Centre , Institute for Interdisciplinary Research in Bio-Nano-Sciences, Babes-Bolyai University , Cluj-Napoca , Romania
| | - Alina Sesarman
- a Department of Molecular Biology and Biotechnology , Faculty of Biology and Geology, Babes-Bolyai University , Cluj-Napoca , Romania.,b Molecular Biology Centre , Institute for Interdisciplinary Research in Bio-Nano-Sciences, Babes-Bolyai University , Cluj-Napoca , Romania
| | - Emilia Licarete
- a Department of Molecular Biology and Biotechnology , Faculty of Biology and Geology, Babes-Bolyai University , Cluj-Napoca , Romania.,b Molecular Biology Centre , Institute for Interdisciplinary Research in Bio-Nano-Sciences, Babes-Bolyai University , Cluj-Napoca , Romania
| | - Alina Porfire
- c Department of Pharmaceutical Technology and Biopharmaceutics , Faculty of Pharmacy, Iuliu Hatieganu University of Medicine and Pharmacy , Cluj-Napoca , Romania
| | - Dana Muntean
- c Department of Pharmaceutical Technology and Biopharmaceutics , Faculty of Pharmacy, Iuliu Hatieganu University of Medicine and Pharmacy , Cluj-Napoca , Romania
| | - Denise Minerva Drotar
- a Department of Molecular Biology and Biotechnology , Faculty of Biology and Geology, Babes-Bolyai University , Cluj-Napoca , Romania.,b Molecular Biology Centre , Institute for Interdisciplinary Research in Bio-Nano-Sciences, Babes-Bolyai University , Cluj-Napoca , Romania
| | - Alexandra Doina Rusu
- a Department of Molecular Biology and Biotechnology , Faculty of Biology and Geology, Babes-Bolyai University , Cluj-Napoca , Romania.,b Molecular Biology Centre , Institute for Interdisciplinary Research in Bio-Nano-Sciences, Babes-Bolyai University , Cluj-Napoca , Romania
| | - Andras-Laszlo Nagy
- d Department of Veterinary Toxicology , University of Agricultural Sciences and Veterinary Medicine , Cluj-Napoca , Romania
| | - Cornel Catoi
- d Department of Veterinary Toxicology , University of Agricultural Sciences and Veterinary Medicine , Cluj-Napoca , Romania
| | - Ioan Tomuta
- c Department of Pharmaceutical Technology and Biopharmaceutics , Faculty of Pharmacy, Iuliu Hatieganu University of Medicine and Pharmacy , Cluj-Napoca , Romania
| | - Laurian Vlase
- c Department of Pharmaceutical Technology and Biopharmaceutics , Faculty of Pharmacy, Iuliu Hatieganu University of Medicine and Pharmacy , Cluj-Napoca , Romania
| | - Manuela Banciu
- a Department of Molecular Biology and Biotechnology , Faculty of Biology and Geology, Babes-Bolyai University , Cluj-Napoca , Romania.,b Molecular Biology Centre , Institute for Interdisciplinary Research in Bio-Nano-Sciences, Babes-Bolyai University , Cluj-Napoca , Romania
| | - Marcela Achim
- c Department of Pharmaceutical Technology and Biopharmaceutics , Faculty of Pharmacy, Iuliu Hatieganu University of Medicine and Pharmacy , Cluj-Napoca , Romania
| |
Collapse
|
32
|
Wang X, Yang Y, Huycke MM. Microbiome-driven carcinogenesis in colorectal cancer: Models and mechanisms. Free Radic Biol Med 2017; 105:3-15. [PMID: 27810411 DOI: 10.1016/j.freeradbiomed.2016.10.504] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 10/19/2016] [Accepted: 10/25/2016] [Indexed: 02/07/2023]
Abstract
Colorectal cancer (CRC) is a leading cause of cancer death and archetype for cancer as a genetic disease. However, the mechanisms for genetic change and their interactions with environmental risk factors have been difficult to unravel. New hypotheses, models, and methods are being used to investigate a complex web of risk factors that includes the intestinal microbiome. Recent research has clarified how the microbiome can generate genomic change in CRC. Several phenotypes among a small group of selected commensals have helped us better understand how mutations and chromosomal instability (CIN) are induced in CRC (e.g., toxin production, metabolite formation, radical generation, and immune modulation leading to a bystander effect). This review discusses recent hypotheses, models, and mechanisms by which the intestinal microbiome contributes to the initiation and progression of sporadic and colitis-associated forms of CRC. Overall, it appears the microbiome can initiate and/or promote CRC at all stages of tumorigenesis by acting as an inducer of DNA damage and CIN, regulating cell growth and death, generating epigenetic changes, and modulating host immune responses. Understanding how the microbiome interacts with other risk factors to define colorectal carcinogenesis will ultimately lead to more accurate risk prediction. A deeper understanding of CRC etiology will also help identify new targets for prevention and treatment and help accelerate the decline in mortality for this common cancer.
Collapse
Affiliation(s)
- Xingmin Wang
- Department of Radiation Oncology, University of Oklahoma Health Sciences Center, USA; Muchmore Laboratories for Infectious Diseases Research, Oklahoma City VA Health Care System, USA
| | - Yonghong Yang
- Gansu Province Children's Hospital, Lanzhou, China; Key Laboratory of Gastrointestinal Cancer, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Mark M Huycke
- Muchmore Laboratories for Infectious Diseases Research, Oklahoma City VA Health Care System, USA; Department of Internal Medicine, PO Box 26901, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73126-0901, USA.
| |
Collapse
|
33
|
Wu X, Li X, Fu Q, Cao Q, Chen X, Wang M, Yu J, Long J, Yao J, Liu H, Wang D, Liao R, Dong C. AKR1B1 promotes basal-like breast cancer progression by a positive feedback loop that activates the EMT program. J Exp Med 2017; 214:1065-1079. [PMID: 28270406 PMCID: PMC5379972 DOI: 10.1084/jem.20160903] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 11/03/2016] [Accepted: 12/20/2016] [Indexed: 01/04/2023] Open
Abstract
The treatment of BLBC represents an unmet medical need. Wu et al. show that AKR1B1 facilitates BLBC progression through a positive feedback loop that activates the EMT program, suggesting that inhibition of AKR1B1 has the potential to become a valuable therapeutic strategy for BLBC. Basal-like breast cancer (BLBC) is associated with high-grade, distant metastasis and poor prognosis. Elucidating the determinants of aggressiveness in BLBC may facilitate the development of novel interventions for this challenging disease. In this study, we show that aldo-keto reductase 1 member B1 (AKR1B1) overexpression highly correlates with BLBC and predicts poor prognosis in breast cancer patients. Mechanistically, Twist2 transcriptionally induces AKR1B1 expression, leading to nuclear factor κB (NF-κB) activation. In turn, NF-κB up-regulates Twist2 expression, thereby fulfilling a positive feedback loop that activates the epithelial–mesenchymal transition program and enhances cancer stem cell (CSC)–like properties in BLBC. AKR1B1 expression promotes, whereas AKR1B1 knockdown inhibits, tumorigenicity and metastasis. Importantly, epalrestat, an AKR1B1 inhibitor that has been approved for the treatment of diabetic complications, significantly suppresses CSC properties, tumorigenicity, and metastasis of BLBC cells. Together, our study identifies AKR1B1 as a key modulator of tumor aggressiveness and suggests that pharmacologic inhibition of AKR1B1 has the potential to become a valuable therapeutic strategy for BLBC.
Collapse
Affiliation(s)
- Xuebiao Wu
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, Hangzhou 310058, China.,Zhejiang Key Laboratory for Disease Proteomics, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xiaoli Li
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, Hangzhou 310058, China.,Zhejiang Key Laboratory for Disease Proteomics, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Qiang Fu
- Department of Immunology, Binzhou Medical University, Yantai 264003, China
| | - Qianhua Cao
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, Hangzhou 310058, China.,Zhejiang Key Laboratory for Disease Proteomics, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xingyu Chen
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, Hangzhou 310058, China.,Zhejiang Key Laboratory for Disease Proteomics, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Mengjie Wang
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, Hangzhou 310058, China.,Zhejiang Key Laboratory for Disease Proteomics, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Jie Yu
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, Hangzhou 310058, China.,Zhejiang Key Laboratory for Disease Proteomics, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Jingpei Long
- Department of Breast Surgery, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Jun Yao
- Department of Neuro-oncology, M.D. Anderson Cancer Center, The University of Texas, Houston, TX 77030
| | - Huixin Liu
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, Hangzhou 310058, China.,Zhejiang Key Laboratory for Disease Proteomics, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Danping Wang
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, Hangzhou 310058, China.,Zhejiang Key Laboratory for Disease Proteomics, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Ruocen Liao
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, Hangzhou 310058, China.,Zhejiang Key Laboratory for Disease Proteomics, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Chenfang Dong
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, Hangzhou 310058, China .,Zhejiang Key Laboratory for Disease Proteomics, Zhejiang University School of Medicine, Hangzhou 310058, China
| |
Collapse
|
34
|
Rotondo R, Moschini R, Renzone G, Tuccinardi T, Balestri F, Cappiello M, Scaloni A, Mura U, Del-Corso A. Human carbonyl reductase 1 as efficient catalyst for the reduction of glutathionylated aldehydes derived from lipid peroxidation. Free Radic Biol Med 2016; 99:323-332. [PMID: 27562619 DOI: 10.1016/j.freeradbiomed.2016.08.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 08/11/2016] [Accepted: 08/12/2016] [Indexed: 12/14/2022]
Abstract
Human recombinant carbonyl reductase 1 (E.C. 1.1.1.184, hCBR1) is shown to efficiently act as aldehyde reductase on glutathionylated alkanals, namely 3-glutathionyl-4-hydroxynonanal (GSHNE), 3-glutathionyl-nonanal, 3-glutathionyl-hexanal and 3-glutathionyl-propanal. The presence of the glutathionyl moiety appears as a necessary requirement for the susceptibility of these compounds to the NADPH-dependent reduction by hCBR1. In fact the corresponding alkanals and alkenals, and the cysteinyl and γ-glutamyl-cysteinyl alkanals adducts were either ineffective or very poorly active as CBR1 substrates. Mass spectrometry analysis reveals the ability of hCBR1 to reduce GSHNE to the corresponding GS-dihydroxynonane (GSDHN) and at the same time to catalyze the oxidation of the hemiacetal form of GSHNE, generating the 3-glutathionylnonanoic-δ-lactone. These data are indicative of the ability of the enzyme to catalyze a disproportion reaction of the substrate through the redox recycle of the pyridine cofactor. A rationale for the observed preferential activity of hCBR1 on different GSHNE diastereoisomers is given by molecular modelling. These results evidence the potential of hCBR1 acting on GSHNE to accomplish a dual role, both in terms of HNE detoxification and, through the production of GSDHN, in terms of involvement into the signalling cascade of the cellular inflammatory response.
Collapse
Affiliation(s)
- Rossella Rotondo
- University of Pisa, Department of Biology, Biochemistry Unit, via S. Zeno, 51, Pisa, Italy
| | - Roberta Moschini
- University of Pisa, Department of Biology, Biochemistry Unit, via S. Zeno, 51, Pisa, Italy
| | - Giovanni Renzone
- Proteomics & Mass Spectrometry Laboratory, ISPAAM-CNR, via Argine, 1085, Napoli, Italy
| | | | - Francesco Balestri
- University of Pisa, Department of Biology, Biochemistry Unit, via S. Zeno, 51, Pisa, Italy
| | - Mario Cappiello
- University of Pisa, Department of Biology, Biochemistry Unit, via S. Zeno, 51, Pisa, Italy
| | - Andrea Scaloni
- Proteomics & Mass Spectrometry Laboratory, ISPAAM-CNR, via Argine, 1085, Napoli, Italy
| | - Umberto Mura
- University of Pisa, Department of Biology, Biochemistry Unit, via S. Zeno, 51, Pisa, Italy
| | - Antonella Del-Corso
- University of Pisa, Department of Biology, Biochemistry Unit, via S. Zeno, 51, Pisa, Italy.
| |
Collapse
|
35
|
Horiyama S, Hatai M, Takahashi Y, Date S, Masujima T, Honda C, Ichikawa A, Yoshikawa N, Nakamura K, Kunitomo M, Takayama M. Intracellular Metabolism of α,β-Unsaturated Carbonyl Compounds, Acrolein, Crotonaldehyde and Methyl Vinyl Ketone, Active Toxicants in Cigarette Smoke: Participation of Glutathione Conjugation Ability and Aldehyde–Ketone Sensitive Reductase Activity. Chem Pharm Bull (Tokyo) 2016; 64:585-93. [DOI: 10.1248/cpb.c15-00986] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Shizuyo Horiyama
- School of Pharmacy and Pharmaceutical Sciences, Mukogawa Women’s University
| | - Mayuko Hatai
- School of Pharmacy and Pharmaceutical Sciences, Mukogawa Women’s University
| | - Yuta Takahashi
- School of Pharmacy and Pharmaceutical Sciences, Mukogawa Women’s University
| | - Sachiko Date
- Riken Quantitative Biology Center, OLABB, Osaka University
| | | | - Chie Honda
- School of Pharmacy and Pharmaceutical Sciences, Mukogawa Women’s University
| | - Atsushi Ichikawa
- School of Pharmacy and Pharmaceutical Sciences, Mukogawa Women’s University
| | - Noriko Yoshikawa
- School of Pharmacy and Pharmaceutical Sciences, Mukogawa Women’s University
| | - Kazuki Nakamura
- School of Pharmacy and Pharmaceutical Sciences, Mukogawa Women’s University
| | - Masaru Kunitomo
- School of Pharmacy and Pharmaceutical Sciences, Mukogawa Women’s University
| | - Mitsuo Takayama
- International Graduate School of Arts and Sciences, Yokohama City University
| |
Collapse
|
36
|
Ruiz FX, Cousido-Siah A, Porté S, Domínguez M, Crespo I, Rechlin C, Mitschler A, de Lera ÁR, Martín MJ, de la Fuente JÁ, Klebe G, Parés X, Farrés J, Podjarny A. Structural Determinants of the Selectivity of 3-Benzyluracil-1-acetic Acids toward Human Enzymes Aldose Reductase and AKR1B10. ChemMedChem 2015; 10:1989-2003. [PMID: 26549844 DOI: 10.1002/cmdc.201500393] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Indexed: 12/15/2022]
Abstract
The human enzymes aldose reductase (AR) and AKR1B10 have been thoroughly explored in terms of their roles in diabetes, inflammatory disorders, and cancer. In this study we identified two new lead compounds, 2-(3-(4-chloro-3-nitrobenzyl)-2,4-dioxo-3,4-dihydropyrimidin-1(2H)-yl)acetic acid (JF0048, 3) and 2-(2,4-dioxo-3-(2,3,4,5-tetrabromo-6-methoxybenzyl)-3,4-dihydropyrimidin-1(2H)-yl)acetic acid (JF0049, 4), which selectively target these enzymes. Although 3 and 4 share the 3-benzyluracil-1-acetic acid scaffold, they have different substituents in their aryl moieties. Inhibition studies along with thermodynamic and structural characterizations of both enzymes revealed that the chloronitrobenzyl moiety of compound 3 can open the AR specificity pocket but not that of the AKR1B10 cognate. In contrast, the larger atoms at the ortho and/or meta positions of compound 4 prevent the AR specificity pocket from opening due to steric hindrance and provide a tighter fit to the AKR1B10 inhibitor binding pocket, probably enhanced by the displacement of a disordered water molecule trapped in a hydrophobic subpocket, creating an enthalpic signature. Furthermore, this selectivity also occurs in the cell, which enables the development of a more efficient drug design strategy: compound 3 prevents sorbitol accumulation in human retinal ARPE-19 cells, whereas 4 stops proliferation in human lung cancer NCI-H460 cells.
Collapse
Affiliation(s)
- Francesc X Ruiz
- Department of Integrative Biology, Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS, INSERM, UdS, rue Laurent Fries, 67404, Illkirch CEDEX, France. .,Center for Advanced Biotechnology and Medicine, Department of Chemistry and Chemical Biology, Rutgers University, 08854-5627, Piscataway, NJ, (USA).
| | - Alexandra Cousido-Siah
- Department of Integrative Biology, Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS, INSERM, UdS, rue Laurent Fries, 67404, Illkirch CEDEX, France
| | - Sergio Porté
- Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, 08193, Bellaterra, Barcelona, Spain
| | - Marta Domínguez
- Departmento de Química Orgánica and Centro de Investigaciones Biomédicas (CINBIO), Universidade de Vigo, 363100, Vigo, Spain
| | - Isidro Crespo
- Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, 08193, Bellaterra, Barcelona, Spain
| | - Chris Rechlin
- Institute of Pharmaceutical Chemistry, University of Marburg, Marbacher Weg 6, 35032, Marburg, Germany
| | - André Mitschler
- Department of Integrative Biology, Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS, INSERM, UdS, rue Laurent Fries, 67404, Illkirch CEDEX, France
| | - Ángel R de Lera
- Departmento de Química Orgánica and Centro de Investigaciones Biomédicas (CINBIO), Universidade de Vigo, 363100, Vigo, Spain
| | - María Jesús Martín
- Biomar Microbial Technologies S.A., Parque Tecnológico de León, 24009, León, Spain
| | | | - Gerhard Klebe
- Institute of Pharmaceutical Chemistry, University of Marburg, Marbacher Weg 6, 35032, Marburg, Germany
| | - Xavier Parés
- Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, 08193, Bellaterra, Barcelona, Spain
| | - Jaume Farrés
- Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, 08193, Bellaterra, Barcelona, Spain
| | - Alberto Podjarny
- Department of Integrative Biology, Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS, INSERM, UdS, rue Laurent Fries, 67404, Illkirch CEDEX, France.
| |
Collapse
|
37
|
Schaur RJ, Siems W, Bresgen N, Eckl PM. 4-Hydroxy-nonenal-A Bioactive Lipid Peroxidation Product. Biomolecules 2015; 5:2247-337. [PMID: 26437435 PMCID: PMC4693237 DOI: 10.3390/biom5042247] [Citation(s) in RCA: 141] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Revised: 07/24/2015] [Accepted: 07/29/2015] [Indexed: 12/23/2022] Open
Abstract
This review on recent research advances of the lipid peroxidation product 4-hydroxy-nonenal (HNE) has four major topics: I. the formation of HNE in various organs and tissues, II. the diverse biochemical reactions with Michael adduct formation as the most prominent one, III. the endogenous targets of HNE, primarily peptides and proteins (here the mechanisms of covalent adduct formation are described and the (patho-) physiological consequences discussed), and IV. the metabolism of HNE leading to a great number of degradation products, some of which are excreted in urine and may serve as non-invasive biomarkers of oxidative stress.
Collapse
Affiliation(s)
- Rudolf J Schaur
- Institute of Molecular Biosciences, University of Graz, Heinrichstrasse 33a, 8010 Graz, Austria.
| | - Werner Siems
- Institute for Medical Education, KortexMed GmbH, Hindenburgring 12a, 38667 Bad Harzburg, Germany.
| | - Nikolaus Bresgen
- Division of Genetics, Department of Cell Biology, University of Salzburg, Hellbrunnerstasse 34, 5020 Salzburg, Austria.
| | - Peter M Eckl
- Division of Genetics, Department of Cell Biology, University of Salzburg, Hellbrunnerstasse 34, 5020 Salzburg, Austria.
| |
Collapse
|
38
|
Singhal SS, Singhal J, Figarola JL, Riggs A, Horne D, Awasthi S. 2′-Hydroxyflavanone: A promising molecule for kidney cancer prevention. Biochem Pharmacol 2015; 96:151-8. [DOI: 10.1016/j.bcp.2015.04.022] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 04/27/2015] [Indexed: 12/18/2022]
|
39
|
Balestri F, Cappiello M, Moschini R, Rotondo R, Abate M, Del-Corso A, Mura U. Modulation of aldose reductase activity by aldose hemiacetals. Biochim Biophys Acta Gen Subj 2015. [PMID: 26215787 DOI: 10.1016/j.bbagen.2015.07.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Glucose is considered as one of the main sources of cell damage related to aldose reductase (AR) action in hyperglycemic conditions and a worldwide effort is posed in searching for specific inhibitors of the enzyme. This AR substrate has often been reported as generating non-hyperbolic kinetics, mimicking a negative cooperative behavior. This feature was explained by the simultaneous action of two enzyme forms acting on the same substrate. METHODS The reduction of different aldoses and other classical AR substrates was studied using pure preparations of bovine lens and human recombinant AR. RESULTS The apparent cooperative behavior of AR acting on glucose and other hexoses and pentoses, but not on tethroses, glyceraldehyde, 4-hydroxynonenal and 4-nitrobenzaldehyde, is generated by a partial nonclassical competitive inhibition exerted by the aldose hemiacetal on the reduction of the free aldehyde. A kinetic model is proposed and kinetic parameters are determined for the reduction of l-idose. CONCLUSIONS Due to the unavoidable presence of the hemiacetal, glucose reduction by AR occurs under different conditions with respect to other relevant AR-substrates, such as alkanals and alkenals, coming from membrane lipid peroxidation. This may have implications in searching for AR inhibitors. The emerging kinetic parameters for the aldoses free aldehyde indicate the remarkable ability of the enzyme to interact and reduce highly hydrophilic and bulky substrates. GENERAL SIGNIFICANCE The discovery of aldose reductase modulation by hemiacetals offers a new perspective in searching for aldose reductase inhibitors to be developed as drugs counteracting the onset of diabetic complications.
Collapse
Affiliation(s)
- Francesco Balestri
- University of Pisa, Department of Biology, Biochemistry Unit, via San Zeno, 51, Pisa 56127, Italy
| | - Mario Cappiello
- University of Pisa, Department of Biology, Biochemistry Unit, via San Zeno, 51, Pisa 56127, Italy; Interdepartmental Research Center Nutrafood "Nutraceuticals and Food for Health", University of Pisa, Pisa, Italy
| | - Roberta Moschini
- University of Pisa, Department of Biology, Biochemistry Unit, via San Zeno, 51, Pisa 56127, Italy; Interdepartmental Research Center Nutrafood "Nutraceuticals and Food for Health", University of Pisa, Pisa, Italy
| | - Rossella Rotondo
- University of Pisa, Department of Biology, Biochemistry Unit, via San Zeno, 51, Pisa 56127, Italy
| | - Marco Abate
- University of Pisa, Department of Mathematics, via Buonarroti, 2, Pisa 56127, Italy
| | - Antonella Del-Corso
- University of Pisa, Department of Biology, Biochemistry Unit, via San Zeno, 51, Pisa 56127, Italy; Interdepartmental Research Center Nutrafood "Nutraceuticals and Food for Health", University of Pisa, Pisa, Italy.
| | - Umberto Mura
- University of Pisa, Department of Biology, Biochemistry Unit, via San Zeno, 51, Pisa 56127, Italy; Interdepartmental Research Center Nutrafood "Nutraceuticals and Food for Health", University of Pisa, Pisa, Italy
| |
Collapse
|
40
|
Walia S, Kamal R, Kanwar SS, Dhawan DK. Cyclooxygenase as a Target in Chemoprevention by Probiotics During 1,2-Dimethylhydrazine Induced Colon Carcinogenesis in Rats. Nutr Cancer 2015; 67:603-11. [DOI: 10.1080/01635581.2015.1011788] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
41
|
Shoeb M, Ansari NH, Srivastava SK, Ramana KV. 4-Hydroxynonenal in the pathogenesis and progression of human diseases. Curr Med Chem 2014; 21:230-7. [PMID: 23848536 DOI: 10.2174/09298673113209990181] [Citation(s) in RCA: 116] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2012] [Revised: 06/18/2013] [Accepted: 07/05/2013] [Indexed: 02/06/2023]
Abstract
Metastable aldehydes produced by lipid peroxidation act as 'toxic second messengers' that extend the injurious potential of free radicals. 4-hydroxy 2-nonenal (HNE), a highly toxic and most abundant stable end product of lipid peroxidation, has been implicated in the tissue damage, dysfunction, injury associated with aging and other pathological states such as cancer, Alzheimer, diabetes, cardiovascular and inflammatory complications. Further, HNE has been considered as a oxidative stress marker and it act as a secondary signaling molecule to regulates a number of cell signaling pathways. Biological activity of HNE depends on its intracellular concentration, which can differentially modulate cell death, growth and differentiation. Therefore, the mechanisms responsible for maintaining the intracellular levels of HNE are most important, not only in the defense against oxidative stress but also in the pathophysiology of a number of disease processes. In this review, we discussed the significance of HNE in mediating various disease processes and how regulation of its metabolism could be therapeutically effective.
Collapse
Affiliation(s)
- Mohammad Shoeb
- Department of Biochemistry and Molecular biology, University of Texas Medical Branch, Galveston, Texas -77555, USA.
| | | | | | | |
Collapse
|
42
|
Maccari R, Ottanà R. Targeting Aldose Reductase for the Treatment of Diabetes Complications and Inflammatory Diseases: New Insights and Future Directions. J Med Chem 2014; 58:2047-67. [DOI: 10.1021/jm500907a] [Citation(s) in RCA: 113] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Rosanna Maccari
- Dipartimento
di Scienze del
Farmaco e dei Prodotti per la Salute, Università degli Studi di Messina, Polo Universitario dell’Annunziata, 98168 Messina, Italy
| | - Rosaria Ottanà
- Dipartimento
di Scienze del
Farmaco e dei Prodotti per la Salute, Università degli Studi di Messina, Polo Universitario dell’Annunziata, 98168 Messina, Italy
| |
Collapse
|
43
|
Aldose reductase inhibition suppresses azoxymethane-induced colonic premalignant lesions in C57BL/KsJ-db/db mice. Cancer Lett 2014; 355:141-7. [PMID: 25218594 DOI: 10.1016/j.canlet.2014.09.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Revised: 08/29/2014] [Accepted: 09/04/2014] [Indexed: 12/31/2022]
Abstract
Type-2 diabetes and obesity-related metabolic abnormalities are major risk factors for the development of colon cancer. In the present study, we examined the effects of polyol pathway enzyme aldose reductase (AR) inhibitor, fidarestat, on the development of azoxymethane (AOM)-induced colonic premalignant lesions in C57BL/KsJ-db/db obese mice. Our results indicate that fidarestat given in the drinking water caused a significant reduction in the total number of colonic premalignant lesions in the AOM treated obese mice. Further, the expression levels of PKC-β2, AKT, COX-2 and iNOS in the colonic mucosa of AOM-treated mice were significantly decreased by fidarestat. The serum levels of IL-1α, IP-10, MIG, TNF-α and VEGF are significantly suppressed in AOM + fidarestat treated obese mice. Fidarestat also decreased the expression of COX-2, iNOS, XIAP, survivin, β-catenin and NF-κB in high glucose-treated HT29 colon cancer cells. In conclusion, our results indicate that fidarestat inhibits the development of colonic premalignant lesions in an obesity-related colon cancer and is chemopreventive to colorectal carcinogenesis in obese individuals.
Collapse
|
44
|
Chatzopoulou M, Pegklidou K, Papastavrou N, Demopoulos VJ. Development of aldose reductase inhibitors for the treatment of inflammatory disorders. Expert Opin Drug Discov 2013; 8:1365-80. [PMID: 24090200 DOI: 10.1517/17460441.2013.843524] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Accumulating evidence attributes a significant role to aldose reductase (ALR2) in the pathogenesis of several inflammatory pathologies. Aldose reductase inhibitors (ARIs) were found to attenuate reactive oxygen species (ROS) production both in vitro and in vivo. Thus, they disrupt signaling cascades that lead to the production of cytokines/chemokines, which induce and exacerbate inflammation. As a result, ARIs might hold a significant therapeutic potential as alternate anti-inflammatory drugs. AREAS COVERED The authors present a comprehensive review of the current data that support the central role of ALR2 in several inflammatory pathologies (i.e., diabetes, cancer, sepsis, asthma and ocular inflammation). Further, the authors describe the potential underlying molecular mechanisms and provide a commentary on the status of ARIs in this field. EXPERT OPINION It is important that future efforts focus on delineating all the steps of the molecular mechanism that implicates ALR2 in inflammatory pathologies. At the same time, utilizing the previous efforts in the field of ARIs, several candidates that have been proven safe in the clinic may be evaluated for their clinical significance as anti-inflammatory medication. Finally, structurally novel ARIs, designed to target specifically the proinflammatory subpocket of ALR2, should be pursued.
Collapse
Affiliation(s)
- Maria Chatzopoulou
- Aristotle University of Thessaloniki, School of Pharmacy, Department of Pharmaceutical Chemistry , 54124 Thessaloniki , Greece ;
| | | | | | | |
Collapse
|
45
|
Shoeb M, Ramana KV, Srivastava SK. Aldose reductase inhibition enhances TRAIL-induced human colon cancer cell apoptosis through AKT/FOXO3a-dependent upregulation of death receptors. Free Radic Biol Med 2013; 63:280-90. [PMID: 23732517 PMCID: PMC3729926 DOI: 10.1016/j.freeradbiomed.2013.05.039] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Revised: 05/10/2013] [Accepted: 05/24/2013] [Indexed: 12/26/2022]
Abstract
One of the major problems associated with the chemotherapy of tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) that selectively kills tumor cells is decreased drug resistance. This warranted the development of safe novel pharmacological agents that could sensitize the tumor cells to TRAIL. Herein, we examined the role of aldose reductase (AR) in sensitizing cancer cells to TRAIL and potentiating TRAIL-induced apoptosis of human colon cancer cells. We demonstrate that AR inhibition potentiates TRAIL-induced cytotoxicity in cancer cells by upregulation of both death receptor (DR)-5 and DR4. Knockdown of DR5 and DR4 significantly (>85%) reduced the sensitizing effect of the AR inhibitor fidarestat on TRAIL-induced apoptosis. Further, AR inhibition also downregulates cell survival proteins (Bcl-xL, Bcl-2, survivin, XIAP, and FLIP) and upregulates the expression of proapoptotic proteins such as Bax and alters mitochondrial membrane potential, leading to cytochrome c release, caspases-3 activation, and PARP cleavage. We found that AR inhibition regulates AKT/PI3K-dependent activation of forkhead transcription factor FOXO3a. Knockdown of FOXO3a significantly (>80%) abolished AR inhibition-induced upregulation of DR5 and DR4 and apoptosis in colon cancer cells. Overall, our results show that fidarestat potentiates TRAIL-induced apoptosis through downregulation of cell survival proteins and upregulation of death receptors via activation of the AKT/FOXO3a pathway.
Collapse
Affiliation(s)
- Mohammad Shoeb
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas, 77555
| | - Kota V Ramana
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas, 77555
| | - Satish K Srivastava
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas, 77555
| |
Collapse
|
46
|
Saxena A, Shoeb M, Ramana KV, Srivastava SK. Aldose reductase inhibition suppresses colon cancer cell viability by modulating microRNA-21 mediated programmed cell death 4 (PDCD4) expression. Eur J Cancer 2013; 49:3311-9. [PMID: 23827854 DOI: 10.1016/j.ejca.2013.05.031] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Revised: 05/02/2013] [Accepted: 05/30/2013] [Indexed: 12/11/2022]
Abstract
Inhibition of polyol pathway enzyme aldose reductase (AR) has been shown to prevent colon cancer cells growth in culture and in nude mice xenografts. However, the role of AR in the mediation of growth factor-induced colon cancer cells growth is not well understood. In this study, we have investigated how AR inhibition prevents tumour growth via regulation of microRNA (miR)-21-mediated programmed cell death 4 (PDCD4) expression in colon cancer cells in in vitro and in vivo. Treatment of colon cancer cells (HT29, SW480 and Caco-2) with epidermal growth factor (EGF) caused increased expression of miR-21 and inhibition of AR prevented it. Further, AR inhibition also increased PDCD4, a putative target of miR-21 in human colon cancer cells. Inhibition of AR also prevented EGF-induced phosphorylation of PDCD4. Treatment of HT29 cells with AR inhibitor, fidarestat, prevented the EGF-induced phosphorylation of mammalian target of rapamycin (mTOR), regulatory associated protein of mTOR (Raptor), eukaryotic initiation factor 4E (eIF4E), p70 S6 kinase (S6K) and eukaryotic initiation factor 4E (eIF4E)-binding protein 1 (4E-BP1) and increased the phosphorylation of 5' adenosine monophosphate-activated protein kinase (AMPK). Similarly, in nude mice xenograft tissues, PDCD4 and 4E-BP1 levels were significantly higher in AR inhibitor-treated mice compared to controls. Collectively, these results indicate that AR inhibition prevents growth factor-induced colon cancer growth by down-regulating miR-21 expression and increasing PDCD4 levels through the reactive oxygen species (ROS)/AMPK/mTOR/AP1/4E-BP1 pathway.
Collapse
Affiliation(s)
- Ashish Saxena
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, United States
| | | | | | | |
Collapse
|
47
|
Regulation of NF-κB-induced inflammatory signaling by lipid peroxidation-derived aldehydes. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2013; 2013:690545. [PMID: 23710287 PMCID: PMC3654319 DOI: 10.1155/2013/690545] [Citation(s) in RCA: 139] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Accepted: 03/22/2013] [Indexed: 12/28/2022]
Abstract
Oxidative stress plays a critical role in the pathophysiology of a wide range of diseases including cancer. This view has broadened significantly with the recent discoveries that reactive oxygen species initiated lipid peroxidation leads to the formation of potentially toxic lipid aldehyde species such as 4-hydroxy-trans-2-nonenal (HNE), acrolein, and malondialdehyde which activate various signaling intermediates that regulate cellular activity and dysfunction via a process called redox signaling. The lipid aldehyde species formed during synchronized enzymatic pathways result in the posttranslational modification of proteins and DNA leading to cytotoxicity and genotoxicty. Among the lipid aldehyde species, HNE has been widely accepted as a most toxic and abundant lipid aldehyde generated during lipid peroxidation. HNE and its glutathione conjugates have been shown to regulate redox-sensitive transcription factors such as NF-κB and AP-1 via signaling through various protein kinase cascades. Activation of redox-sensitive transcription factors and their nuclear localization leads to transcriptional induction of several genes responsible for cell survival, differentiation, and death. In this review, we describe the mechanisms by which the lipid aldehydes transduce activation of NF-κB signaling pathways that may help to develop therapeutic strategies for the prevention of a number of inflammatory diseases.
Collapse
|
48
|
Tang J, Du Y, Petrash JM, Sheibani N, Kern TS. Deletion of aldose reductase from mice inhibits diabetes-induced retinal capillary degeneration and superoxide generation. PLoS One 2013; 8:e62081. [PMID: 23614016 PMCID: PMC3628579 DOI: 10.1371/journal.pone.0062081] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Accepted: 03/16/2013] [Indexed: 12/12/2022] Open
Abstract
Purpose Pharmacologic inhibition of aldose reductase (AR) previously has been studied with respect to diabetic retinopathy with mixed results. Since drugs can have off-target effects, we studied the effects of AR deletion on the development and molecular abnormalities that contribute to diabetic retinopathy. Since recent data suggests an important role for leukocytes in the development of the retinopathy, we determined also if AR in leukocytes contributes to leukocyte-mediated death of retinal endothelial cells in diabetes. Methods Wild-type (WT; C57BL/6J) and AR deficient (AR−/−) mice were made diabetic with streptozotocin. Mice were sacrificed at 2 and 10 months of diabetes to evaluate retinal vascular histopathology, to quantify retinal superoxide production and biochemical and physiological abnormalities in the retina, and to assess the number of retinal endothelial cells killed by blood leukocytes in a co-culture system. Results Diabetes in WT mice developed the expected degeneration of retinal capillaries, and increased generation of superoxide by the retina. Leukocytes from diabetic WT mice also killed more retinal endothelial cells than did leukocytes from nondiabetic animals (p<0.0001). Deletion of AR largely (P<0.05) inhibited the diabetes-induced degeneration of retinal capillaries, as well as the increase in superoxide production by retina. AR-deficiency significantly inhibited the diabetes-induced increase in expression of inducible nitric oxide synthase (iNOS) in retina, but had no significant effect on expression of intercellular adhesion molecule-1 (ICAM-1), phosphorylated p38 MAPK, or killing of retinal endothelial cells by leukocytes. Conclusions AR contributes to the degeneration of retinal capillaries in diabetic mice. Deletion of the enzyme inhibits the diabetes-induced increase in expression of iNOS and of superoxide production, but does not correct a variety of other pro-inflammatory abnormalities associated with the development of diabetic retinopathy.
Collapse
Affiliation(s)
- Jie Tang
- Department of Medicine, Case Western Reserve University, Cleveland, Ohio, United States of America
- Department of Ophthalmology, Heilongjiang Province Hospital, Heilongjiang Province, Harbin, China
| | - Yunpeng Du
- Department of Medicine, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - J. Mark Petrash
- Department of Ophthalmology, University of Colorado, Denver, Colorado, United States of America
| | - Nader Sheibani
- Department of Ophthalmology, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Timothy S. Kern
- Department of Medicine, Case Western Reserve University, Cleveland, Ohio, United States of America
- Cleveland VAMC Research Service 151, Cleveland, Ohio, United States of America
- * E-mail:
| |
Collapse
|
49
|
Saxena A, Tammali R, Ramana KV, Srivastava SK. Aldose reductase inhibition prevents colon cancer growth by restoring phosphatase and tensin homolog through modulation of miR-21 and FOXO3a. Antioxid Redox Signal 2013; 18:1249-62. [PMID: 22978663 PMCID: PMC3584509 DOI: 10.1089/ars.2012.4643] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
AIMS We have shown earlier that inhibition of aldose reductase (AR), an oxidative stress-response protein, prevents colon cancer cell growth in vitro and in vivo. Changes in microribonucleic acid (miR) expression can contribute to cancer by modulating the functional expression of critical genes involved in cancer growth and metastasis. However, the molecular mechanisms by which AR regulates miR expression and their dependent mitogenic effects in cancer cells are not known. Therefore, we investigated how AR regulates growth factor-induced expression of miRs and growth of colon cancer cells. RESULTS Inhibition of AR significantly downregulated growth factor-induced miR-21 expression in human colon cancer cells, HT29, SW480, and Caco-2. Further, AR inhibition also increased phosphatase and tensin homolog (PTEN) (a direct target of miR-21) and forkhead box O3A (FOXO3a) in colon cancer cells. Our results obtained with HT29 cells ablated with FOXO3a siRNA showed increased activator protein-1 (AP-1) activation and miR-21 expression, indicating that FOXO3a represses miR-21 via AP-1 inactivation. Inhibition of AR also prevented the epidermal growth factor-induced phosphorylation of phosphatidylinositol 3-kinase (PI3K), serine/threonine kinase (AKT), c-Jun, c-Fos, PTEN, and FOXO3a, and deoxyribonucleic acid (DNA)-binding activity of AP-1. More importantly, in human colon adenocarcinoma xenograft tissues, miR-21 expression was lower, and PTEN and FOXO3a levels were significantly higher in AR inhibitor-treated mice compared to controls. INNOVATION These findings demonstrate a novel role of AR in the regulation of miR-21 and its target PTEN in growth factor-induced colon cancer cell growth. CONCLUSIONS Collectively, these results show a novel role of AR in mediation of growth factor-induced colon cancer growth by modulating miR-21, PTEN, and FOXO3a expression through reactive oxygen species (ROS)/PI3K/AKT/AP-1.
Collapse
Affiliation(s)
- Ashish Saxena
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas 77555, USA
| | | | | | | |
Collapse
|
50
|
Liu J, Zhong X, Li J, Liu B, Guo S, Chen J, Tan Q, Wang Q, Ma W, Wu Z, Wang H, Hou M, Zhang HT, Zhou Q. Screening and identification of lung cancer metastasis-related genes by suppression subtractive hybridization. Thorac Cancer 2012; 3:207-216. [PMID: 28920308 DOI: 10.1111/j.1759-7714.2011.00092.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND AND OBJECTIVE Lung cancer metastasis is a complicated process in which multiple stages and multiple genes are involved. There is an urgent need to use new molecular biology techniques to get more systematic information and have a general idea of the molecular events that take place in lung cancer metastasis. The object of this study was to construct the subtracted cDNA libraries of different metastatic potential lung cancer cell lines, NL9980 and L9981, which were established and screened from human lung large cell carcinoma cell line, WCQH-9801. METHOD The forward and reverse subtracted cDNA libraries were constructed in the large cell lung cancer cell lines NL9980 and L9981 with the same heredity background but different metastatic potential, by suppression subtractive hybridization (SSH). The positive clones were preliminarily screened by blue-white colony and precisely identified by PCR. The forward and reverse subtracted libraries were screened and identified by dot blot so as to obtain the clones corresponding to gene segments with differential expression. DNA sequencing was performed to analyze the sequences of differential expression segments, which were then searched and compared using the Basic Local Alignment Search Tool from The National Center for Biotechnology Information NCBI BLAST tools. Quantitative real-time reverse transcription polymerase chain reaction (RT-PCR) and western blotting were performed to confirm the differential expressed genes both on RNA and protein levels. RESULTS The forward and reverse subtracted cDNA libraries of the different large cell lung cancer cell lines with metastatic potential were successfully constructed. With blue-white colony and dot blot, 307 positive clones in the forward subtracted library and 78 positive clones in the reverse subtracted library were obtained. Fifty-five clones were successfully sequenced in the forward subtracted library while 31 clones were successfully sequenced in the reverse subtracted library. One new expressed sequence tag (EST) segment was identified from the reverse subtracted cDNA library and was successfully submitted to GenBank and embodied by GenBank. For the differentially expressed genes between L9981 and NL9980 screened by SSH, four genes, ANXA2, KRT18, ACTG1 was upregulated in L9981 cells compared to NL9980 cells. Annexin A2 (which was encoded by ANXA2), γ-actin (which was encoded by ACTG1), and aldose reductase (which was encoded by AKR1B1) proteins were upregulated in L9981 cells compared to NL9980 cells by western blotting. CONCLUSION The forward and reverse subtracted cDNA libraries of different metastatic potential large cell lung cancer cell lines were successfully constructed by SSH. A series of genes have been screened out to have significantly different expression levels between lung cancer cell lines NL9980 and L9981. A new EST segment that may represent a new metastasis-related gene has been identified. Consistent with the result of SSH, both quantitative real-time RT-PCR and western Blotting confirmed the upregulation of ANXA2, ACTG1 and AKR1B1 in lung cancer cell line L9981 compared with NL9980. These three genes may play important roles in lung cancer metastasis.
Collapse
Affiliation(s)
- Jiewei Liu
- The Key Laboratory of Lung Cancer Molecular Biology in Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan Province, ChinaTianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, ChinaDepartment of Thoracic Surgery, First Affiliated Hospital, Dalian Medical University, Dalian, ChinaDepartment of Respiratory Medicine, the Second Hospital Affiliated to Dalian Medical University, Dalian, ChinaSoochow University Laboratory of Cancer Molecular Genetics, Medical College of Soochow University, Sino-Singapore Industrial Park, Suzhou, China
| | - Xiaorong Zhong
- The Key Laboratory of Lung Cancer Molecular Biology in Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan Province, ChinaTianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, ChinaDepartment of Thoracic Surgery, First Affiliated Hospital, Dalian Medical University, Dalian, ChinaDepartment of Respiratory Medicine, the Second Hospital Affiliated to Dalian Medical University, Dalian, ChinaSoochow University Laboratory of Cancer Molecular Genetics, Medical College of Soochow University, Sino-Singapore Industrial Park, Suzhou, China
| | - Juan Li
- The Key Laboratory of Lung Cancer Molecular Biology in Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan Province, ChinaTianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, ChinaDepartment of Thoracic Surgery, First Affiliated Hospital, Dalian Medical University, Dalian, ChinaDepartment of Respiratory Medicine, the Second Hospital Affiliated to Dalian Medical University, Dalian, ChinaSoochow University Laboratory of Cancer Molecular Genetics, Medical College of Soochow University, Sino-Singapore Industrial Park, Suzhou, China
| | - Baoxing Liu
- The Key Laboratory of Lung Cancer Molecular Biology in Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan Province, ChinaTianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, ChinaDepartment of Thoracic Surgery, First Affiliated Hospital, Dalian Medical University, Dalian, ChinaDepartment of Respiratory Medicine, the Second Hospital Affiliated to Dalian Medical University, Dalian, ChinaSoochow University Laboratory of Cancer Molecular Genetics, Medical College of Soochow University, Sino-Singapore Industrial Park, Suzhou, China
| | - Shanxian Guo
- The Key Laboratory of Lung Cancer Molecular Biology in Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan Province, ChinaTianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, ChinaDepartment of Thoracic Surgery, First Affiliated Hospital, Dalian Medical University, Dalian, ChinaDepartment of Respiratory Medicine, the Second Hospital Affiliated to Dalian Medical University, Dalian, ChinaSoochow University Laboratory of Cancer Molecular Genetics, Medical College of Soochow University, Sino-Singapore Industrial Park, Suzhou, China
| | - Jun Chen
- The Key Laboratory of Lung Cancer Molecular Biology in Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan Province, ChinaTianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, ChinaDepartment of Thoracic Surgery, First Affiliated Hospital, Dalian Medical University, Dalian, ChinaDepartment of Respiratory Medicine, the Second Hospital Affiliated to Dalian Medical University, Dalian, ChinaSoochow University Laboratory of Cancer Molecular Genetics, Medical College of Soochow University, Sino-Singapore Industrial Park, Suzhou, China
| | - Qingwei Tan
- The Key Laboratory of Lung Cancer Molecular Biology in Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan Province, ChinaTianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, ChinaDepartment of Thoracic Surgery, First Affiliated Hospital, Dalian Medical University, Dalian, ChinaDepartment of Respiratory Medicine, the Second Hospital Affiliated to Dalian Medical University, Dalian, ChinaSoochow University Laboratory of Cancer Molecular Genetics, Medical College of Soochow University, Sino-Singapore Industrial Park, Suzhou, China
| | - Qin Wang
- The Key Laboratory of Lung Cancer Molecular Biology in Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan Province, ChinaTianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, ChinaDepartment of Thoracic Surgery, First Affiliated Hospital, Dalian Medical University, Dalian, ChinaDepartment of Respiratory Medicine, the Second Hospital Affiliated to Dalian Medical University, Dalian, ChinaSoochow University Laboratory of Cancer Molecular Genetics, Medical College of Soochow University, Sino-Singapore Industrial Park, Suzhou, China
| | - Wei Ma
- The Key Laboratory of Lung Cancer Molecular Biology in Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan Province, ChinaTianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, ChinaDepartment of Thoracic Surgery, First Affiliated Hospital, Dalian Medical University, Dalian, ChinaDepartment of Respiratory Medicine, the Second Hospital Affiliated to Dalian Medical University, Dalian, ChinaSoochow University Laboratory of Cancer Molecular Genetics, Medical College of Soochow University, Sino-Singapore Industrial Park, Suzhou, China
| | - Zhihao Wu
- The Key Laboratory of Lung Cancer Molecular Biology in Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan Province, ChinaTianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, ChinaDepartment of Thoracic Surgery, First Affiliated Hospital, Dalian Medical University, Dalian, ChinaDepartment of Respiratory Medicine, the Second Hospital Affiliated to Dalian Medical University, Dalian, ChinaSoochow University Laboratory of Cancer Molecular Genetics, Medical College of Soochow University, Sino-Singapore Industrial Park, Suzhou, China
| | - Haisu Wang
- The Key Laboratory of Lung Cancer Molecular Biology in Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan Province, ChinaTianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, ChinaDepartment of Thoracic Surgery, First Affiliated Hospital, Dalian Medical University, Dalian, ChinaDepartment of Respiratory Medicine, the Second Hospital Affiliated to Dalian Medical University, Dalian, ChinaSoochow University Laboratory of Cancer Molecular Genetics, Medical College of Soochow University, Sino-Singapore Industrial Park, Suzhou, China
| | - Mei Hou
- The Key Laboratory of Lung Cancer Molecular Biology in Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan Province, ChinaTianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, ChinaDepartment of Thoracic Surgery, First Affiliated Hospital, Dalian Medical University, Dalian, ChinaDepartment of Respiratory Medicine, the Second Hospital Affiliated to Dalian Medical University, Dalian, ChinaSoochow University Laboratory of Cancer Molecular Genetics, Medical College of Soochow University, Sino-Singapore Industrial Park, Suzhou, China
| | - Hong-Tao Zhang
- The Key Laboratory of Lung Cancer Molecular Biology in Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan Province, ChinaTianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, ChinaDepartment of Thoracic Surgery, First Affiliated Hospital, Dalian Medical University, Dalian, ChinaDepartment of Respiratory Medicine, the Second Hospital Affiliated to Dalian Medical University, Dalian, ChinaSoochow University Laboratory of Cancer Molecular Genetics, Medical College of Soochow University, Sino-Singapore Industrial Park, Suzhou, China
| | - Qinghua Zhou
- The Key Laboratory of Lung Cancer Molecular Biology in Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan Province, ChinaTianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, ChinaDepartment of Thoracic Surgery, First Affiliated Hospital, Dalian Medical University, Dalian, ChinaDepartment of Respiratory Medicine, the Second Hospital Affiliated to Dalian Medical University, Dalian, ChinaSoochow University Laboratory of Cancer Molecular Genetics, Medical College of Soochow University, Sino-Singapore Industrial Park, Suzhou, China
| |
Collapse
|