1
|
Wu A, Li H, Gao M, Liang J, Huang J, Farrés J, Cao D, Li G. The pan-cancer landscape of aldo-keto reductase1B10 reveals that its expression is diminished in gastric cancer. Front Immunol 2024; 15:1488042. [PMID: 39712017 PMCID: PMC11659136 DOI: 10.3389/fimmu.2024.1488042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 11/18/2024] [Indexed: 12/24/2024] Open
Abstract
Introduction Aldo-keto reductase 1B10 (AKR1B10) is a multifunctional enzyme, which is important in cancer development and progression, but the landscape of AKR1B10 in pan-cancers and in tumor microenvironment is unclear. Method This study integrated the sequencing data of 33 cancer types, including gastric cancer, from TCGA project to explored the expression pattern and genetic and epigenetic alterations of AKR1B10. The association of AKR1B10 expression with clinical progression of cancers was evaluated by Kaplan-Meier analysis; the potential role of AKR1B10 in tumor microenvironment (TME) and immune-related gene expression were analyzed by PURITY, ESTIMATE, TIMER and CIBERSORT algorithms. The expression of AKR1B10 and immune cell markers in gastric cancer were evaluated with multiplex immunofluorescence staining. Result Results indicated that AKR1B10 was highly expressed in the gastrointestinal tract in health donors, but the expression of AKR1B10 was significantly changed in most of cancer types, which may be ascribed to DNA methylation in its promoter. The AKR1B10 expression in cancers and its value in disease progression was bidirectional and functionally enriched in metabolism in pan-cancers. In tumor microenvironment, AKR1B10 was significantly correlated with immune cell infiltrations and immune gene expression. In the stomach, along with the diminishing of AKR1B10 expression, CD68+ macrophage increased and CD19+ B cell decreased in gastric cancer. Discussion These data indicates that AKR1B10 may be an important factor in the development and progression and a potential therapeutic target for multiple cancers, but plays as a protector in the gastric tissues.
Collapse
Affiliation(s)
- Anqi Wu
- Department of Clinical Research Center, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
- Hunan Province Key Laboratory of Basic and Clinical Pharmacological Research on Gastrointestinal Tumors, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Hao Li
- Department of Pathology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Mengnan Gao
- Department of Gastroenterology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Juan Liang
- Department of Gastroenterology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Jiaqi Huang
- Department of Gastroenterology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Jaume Farrés
- Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Deliang Cao
- Hunan Province Key Laboratory of Cancer Cellular and Molecular Pathology, Hengyang Medical School, Cancer Research Institute, University of South China, Hengyang, China
| | - Guoqing Li
- Hunan Province Key Laboratory of Basic and Clinical Pharmacological Research on Gastrointestinal Tumors, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
- Department of Gastroenterology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| |
Collapse
|
2
|
Guo M, Wang T, Ge W, Ren C, Ko BCB, Zeng X, Cao D. Role of AKR1B10 in inflammatory diseases. Scand J Immunol 2024; 100:e13390. [PMID: 38769661 DOI: 10.1111/sji.13390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 05/01/2024] [Accepted: 05/05/2024] [Indexed: 05/22/2024]
Abstract
Inflammation is an important pathophysiological process in many diseases; it has beneficial and harmful effects. When exposed to various stimuli, the body triggers an inflammatory response to eliminate invaded pathogens and damaged tissues to maintain homeostasis. However, uncontrollable persistent or excessive inflammatory responses may damage tissues and induce various diseases, such as metabolic diseases (e.g. diabetes), autoimmune diseases, nervous system-related diseases, digestive system-related diseases, and even tumours. Aldo-keto reductase 1B10 (AKR1B10) is an important player in the development and progression of multiple diseases, such as tumours and inflammatory diseases. AKR1B10 is upregulated in solid tumours, such as hepatocellular carcinoma (HCC), non-small cell lung carcinoma, and breast cancer, and is a reliable serum marker. However, information on the role of AKR1B10 in inflammation is limited. In this study, we summarized the role of AKR1B10 in inflammatory diseases, including its expression, functional contribution to inflammatory responses, and regulation of signalling pathways related to inflammation. We also discussed the role of AKR1B10 in glucose and lipid metabolism and oxidative stress. This study provides novel information and increases the understanding of clinical inflammatory diseases.
Collapse
Affiliation(s)
- Min Guo
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Tao Wang
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Wenjun Ge
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Chenran Ren
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Ben Chi-Bun Ko
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong, China
| | - Xi Zeng
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Deliang Cao
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| |
Collapse
|
3
|
Chen Q, Wang T, Wu X, Yuan H, Wei Y, Xiao Y. The role of the cytochrome P450 superfamily in the skin. Expert Rev Mol Med 2024; 26:e15. [PMID: 38621674 PMCID: PMC11140544 DOI: 10.1017/erm.2024.5] [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: 09/13/2023] [Revised: 12/10/2023] [Accepted: 02/22/2024] [Indexed: 04/17/2024]
Abstract
In mammals, the skin acts as a barrier to prevent harmful environmental stimuli from entering the circulation. CYP450s are involved in drug biotransformation, exogenous and endogenous substrate metabolism, and maintaining the normal physiological function of the skin, as well as facilitating homeostasis of the internal environment. The expression pattern of CYP450s in the skin is tissue-specific and thus differs from the liver and other organs. The development of skin topical medications, and knowledge of the toxicity and side effects of these medications require a detailed understanding of the expression and function of skin-specific CYP450s. Thus, we summarized the expression of CYP450s in the skin, their function in endogenous metabolic physiology, aberrant CYP450 expression in skin diseases and the influence of environmental variables and medications. This information will serve as a crucial foundation for future studies on the skin, as well as for the design and development of new drugs for skin diseases including topical medications.
Collapse
Affiliation(s)
- Qianqian Chen
- School of Pharmacy, Jiangsu University, Zhenjiang, China
| | - Tuan Wang
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
- Affiliated Hangzhou First People's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xia Wu
- Dermatology Department, Sir Run Run Shaw Hospital, Hangzhou, China
| | - Huipu Yuan
- Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yuan Wei
- School of Pharmacy, Jiangsu University, Zhenjiang, China
| | - Ying Xiao
- Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
4
|
Ghaffarinia A, Ayaydin F, Póliska S, Manczinger M, Bolla BS, Flink LB, Balogh F, Veréb Z, Bozó R, Szabó K, Bata-Csörgő Z, Kemény L. Psoriatic Resolved Skin Epidermal Keratinocytes Retain Disease-Residual Transcriptomic and Epigenomic Profiles. Int J Mol Sci 2023; 24:ijms24054556. [PMID: 36901987 PMCID: PMC10002496 DOI: 10.3390/ijms24054556] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 02/17/2023] [Accepted: 02/21/2023] [Indexed: 03/02/2023] Open
Abstract
The disease-residual transcriptomic profile (DRTP) within psoriatic healed/resolved skin and epidermal tissue-resident memory T (TRM) cells have been proposed to be crucial for the recurrence of old lesions. However, it is unclear whether epidermal keratinocytes are involved in disease recurrence. There is increasing evidence regarding the importance of epigenetic mechanisms in the pathogenesis of psoriasis. Nonetheless, the epigenetic changes that contribute to the recurrence of psoriasis remain unknown. The aim of this study was to elucidate the role of keratinocytes in psoriasis relapse. The epigenetic marks 5-methylcytosine (5-mC) and 5-hydroxymethylcytosine (5-hmC) were visualized using immunofluorescence staining, and RNA sequencing was performed on paired never-lesional and resolved epidermal and dermal compartments of skin from psoriasis patients. We observed diminished 5-mC and 5-hmC amounts and decreased mRNA expression of the ten-eleven translocation (TET) 3 enzyme in the resolved epidermis. SAMHD1, C10orf99, and AKR1B10: the highly dysregulated genes in resolved epidermis are known to be associated with pathogenesis of psoriasis, and the DRTP was enriched in WNT, TNF, and mTOR signaling pathways. Our results suggest that epigenetic changes detected in epidermal keratinocytes of resolved skin may be responsible for the DRTP in the same regions. Thus, the DRTP of keratinocytes may contribute to site-specific local relapse.
Collapse
Affiliation(s)
- Ameneh Ghaffarinia
- HCEMM-USZ Skin Research Group, H-6720 Szeged, Hungary
- Department of Dermatology and Allergology, Albert Szent-Györgyi Medical School, University of Szeged, H-6720 Szeged, Hungary
| | - Ferhan Ayaydin
- HCEMM-USZ, Functional Cell Biology and Immunology, Advanced Core Facility, H-6728 Szeged, Hungary
- Laboratory of Cellular Imaging, Biological Research Centre, Eötvös Loránd Research Network, H-6726 Szeged, Hungary
- Institute of Plant Biology, Biological Research Centre, H-6726 Szeged, Hungary
| | - Szilárd Póliska
- Genomic Medicine and Bioinformatics Core Facility, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| | - Máté Manczinger
- Department of Dermatology and Allergology, Albert Szent-Györgyi Medical School, University of Szeged, H-6720 Szeged, Hungary
- Systems Immunology Research Group, Institute of Biochemistry, Biological Research Centre, ELKH, H-6726 Szeged, Hungary
- HCEMM-Systems Immunology Research Group, H-6726 Szeged, Hungary
| | - Beáta Szilvia Bolla
- HCEMM-USZ Skin Research Group, H-6720 Szeged, Hungary
- Department of Dermatology and Allergology, Albert Szent-Györgyi Medical School, University of Szeged, H-6720 Szeged, Hungary
| | - Lili Borbála Flink
- HCEMM-USZ Skin Research Group, H-6720 Szeged, Hungary
- Department of Dermatology and Allergology, Albert Szent-Györgyi Medical School, University of Szeged, H-6720 Szeged, Hungary
| | - Fanni Balogh
- HCEMM-USZ Skin Research Group, H-6720 Szeged, Hungary
- Department of Dermatology and Allergology, Albert Szent-Györgyi Medical School, University of Szeged, H-6720 Szeged, Hungary
- ELKH-SZTE Dermatological Research Group, Department of Dermatology and Allergology, University of Szeged, H-6720 Szeged, Hungary
| | - Zoltán Veréb
- Regenerative Medicine and Cellular Pharmacology Laboratory (HECRIN), Department of Dermatology and Allergology, University of Szeged, H-6720 Szeged, Hungary
- Research Institute of Translational Biomedicine, Department of Dermatology and Allergology, University of Szeged, H-6720 Szeged, Hungary
| | - Renáta Bozó
- HCEMM-USZ Skin Research Group, H-6720 Szeged, Hungary
- Department of Dermatology and Allergology, Albert Szent-Györgyi Medical School, University of Szeged, H-6720 Szeged, Hungary
- ELKH-SZTE Dermatological Research Group, Department of Dermatology and Allergology, University of Szeged, H-6720 Szeged, Hungary
| | - Kornélia Szabó
- HCEMM-USZ Skin Research Group, H-6720 Szeged, Hungary
- Department of Dermatology and Allergology, Albert Szent-Györgyi Medical School, University of Szeged, H-6720 Szeged, Hungary
- ELKH-SZTE Dermatological Research Group, Department of Dermatology and Allergology, University of Szeged, H-6720 Szeged, Hungary
| | - Zsuzsanna Bata-Csörgő
- HCEMM-USZ Skin Research Group, H-6720 Szeged, Hungary
- Department of Dermatology and Allergology, Albert Szent-Györgyi Medical School, University of Szeged, H-6720 Szeged, Hungary
- ELKH-SZTE Dermatological Research Group, Department of Dermatology and Allergology, University of Szeged, H-6720 Szeged, Hungary
| | - Lajos Kemény
- HCEMM-USZ Skin Research Group, H-6720 Szeged, Hungary
- Department of Dermatology and Allergology, Albert Szent-Györgyi Medical School, University of Szeged, H-6720 Szeged, Hungary
- ELKH-SZTE Dermatological Research Group, Department of Dermatology and Allergology, University of Szeged, H-6720 Szeged, Hungary
- Correspondence:
| |
Collapse
|
5
|
Zhai Z, Yamauchi T, Shangraw S, Hou V, Matsumoto A, Fujita M. Ethanol Metabolism and Melanoma. Cancers (Basel) 2023; 15:1258. [PMID: 36831600 PMCID: PMC9954650 DOI: 10.3390/cancers15041258] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/11/2023] [Accepted: 02/13/2023] [Indexed: 02/18/2023] Open
Abstract
Malignant melanoma is the deadliest form of skin cancer. Despite significant efforts in sun protection education, melanoma incidence is still rising globally, drawing attention to other socioenvironmental risk factors for melanoma. Ethanol and acetaldehyde (AcAH) are ubiquitous in our diets, medicines, alcoholic beverages, and the environment. In the liver, ethanol is primarily oxidized to AcAH, a toxic intermediate capable of inducing tumors by forming adducts with proteins and DNA. Once in the blood, ethanol and AcAH can reach the skin. Although, like the liver, the skin has metabolic mechanisms to detoxify ethanol and AcAH, the risk of ethanol/AcAH-associated skin diseases increases when the metabolic enzymes become dysfunctional in the skin. This review highlights the evidence linking cutaneous ethanol metabolism and melanoma. We summarize various sources of skin ethanol and AcAH and describe how the reduced activity of each alcohol metabolizing enzyme affects the sensitivity threshold to ethanol/AcAH toxicity. Data from the Gene Expression Omnibus database also show that three ethanol metabolizing enzymes (alcohol dehydrogenase 1B, P450 2E1, and catalase) and an AcAH metabolizing enzyme (aldehyde dehydrogenase 2) are significantly reduced in melanoma tissues.
Collapse
Affiliation(s)
- Zili Zhai
- Department of Dermatology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Takeshi Yamauchi
- Department of Dermatology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Sarah Shangraw
- Department of Dermatology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Vincent Hou
- Department of Dermatology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Akiko Matsumoto
- Department of Social Medicine, School of Medicine, Saga University, Saga 849-8501, Japan
| | - Mayumi Fujita
- Department of Dermatology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Veterans Affairs Medical Center, VA Eastern Colorado Health Care System, Aurora, CO 80045, USA
| |
Collapse
|
6
|
Wang J, Yang J. Identification of significant genes with a poor prognosis in skin cutaneous malignant melanoma based on a bioinformatics analysis. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:448. [PMID: 35571409 PMCID: PMC9096380 DOI: 10.21037/atm-22-1163] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 04/02/2022] [Indexed: 01/04/2023]
Abstract
Background Skin cutaneous malignant melanoma (SKCM) is a deadly mutated malignancy that arises from melanocytes in the basal layer of the skin. This study sought to identify effective treatment targets that could serve as prospective therapeutic targets to improve patient outcomes. Methods The GSE83583, GSE111766, and GSE104849 data sets from the GPL10558 platform in the Gene Expression Omnibus (GEO) were used in this study. The candidate genes were identified using the GEO2R tool and a Venn diagram. The Gene Ontology (GO) enrichment and Kyoto Encyclopedia of Gene and Genome (KEGG) preliminary analyses of the differentially expressed genes (DEGs) were conducted using the Database for Annotation, Visualization and Integrated Discovery, and R software. The protein-protein interaction (PPI) network was examined using Cytoscape software. The survminer package was used to examine the overall survival of patients with the identified genes. The Human Protein Atlas (HPA) was used to verify the protein levels of significant genes with poor prognosis. The highly expressed genes in the melanoma tissues were visualized using the ggplot2 package. Results In total, 160 DEGs from 124 melanoma tissues and 9 normal melanocyte tissues were examined in this study. Cytoscape displayed 19 central nodes from the 160 DEGs. The re-analysis showed that the cytochrome P450 family 1 subfamily B member 1 (CYP1B1) and protein kinase C beta (PRKCB) were significantly enriched in the micro ribonucleic acids (RNAs) in cancer. Conclusions CYP1B1 and PRKCB were overexpressed in and correlated with the poor prognosis of SKCM. Our findings might help explore the prognosis and diagnostic markers of SKCM.
Collapse
Affiliation(s)
- Jin Wang
- Department of Bone and Soft Tissue Tumor, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Department of Dermatology, Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, China.,Graduate School, Tianjin Medical University, Tianjin, China
| | - Jilong Yang
- Department of Bone and Soft Tissue Tumor, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| |
Collapse
|
7
|
Downregulation of CYP39A1 Serves as a Novel Biomarker in Hepatocellular Carcinoma with Worse Clinical Outcome. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2021:5175581. [PMID: 35003516 PMCID: PMC8741352 DOI: 10.1155/2021/5175581] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 11/19/2021] [Accepted: 12/03/2021] [Indexed: 12/14/2022]
Abstract
Background CYP39A1 is a poorly characterized metabolic enzyme that has been investigated in a few tumors. However, the role of CYP39A1 in hepatocellular carcinoma (HCC) has not yet been clarified. In this study, the expression and clinical significance of CYP39A1 in HCC were explored. Methods CYP39A1 protein expression was detected in Akt/c-Met-induced HCC mice and 14 paired fresh HCC samples as well as another 159 HCC and matched noncancerous tissues. Meanwhile, the mRNA expression was analyzed by GEO and TCGA analysis and validated in 14 paired fresh HCC tissues. Furthermore, the relationships between CYP39A1 expression and clinicopathologic features as well as prognosis were analyzed. HCC cell growth changes were analyzed by cell viability assays after CYP39A1 overexpression and then validated after CYP39A1 knockout by DepMap database analysis. Results CYP39A1 protein expression was lower expressed in HCC mouse models, and its mRNA and protein expression were also downregulated in HCC compared with noncancerous liver tissues. Higher CYP39A1 expression was associated with well differentiation. Moreover, survival analysis indicated that lower CYP39A1 expression was associated with poorer overall survival. In addition, HepG2 and SMMC-7721 cell viability were inhibited after CYP39A1 overexpression. Genome-wide CRISPR/Cas9 proliferation screening indicated that knockout of CYP39A1 could promote HCC cell growth. Likewise, p-NF-κB and Nrf2 were suppressed after CYP39A1 overexpression. It is worth mentioning that total bile acid, total bilirubin, and direct bilirubin were significantly increased in the patients with low CYP39A1 expression. Conclusions Downregulation of CYP39A1 is associated with HCC carcinogenesis, tumor differentiation, and poor overall survival, suggesting that CYP39A1 may serve as a tumor suppressor gene and novel biomarker for HCC patients.
Collapse
|
8
|
Endo S, Matsunaga T, Nishinaka T. The Role of AKR1B10 in Physiology and Pathophysiology. Metabolites 2021; 11:332. [PMID: 34063865 PMCID: PMC8224097 DOI: 10.3390/metabo11060332] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/17/2021] [Accepted: 05/19/2021] [Indexed: 12/16/2022] Open
Abstract
AKR1B10 is a human nicotinamide adenine dinucleotide phosphate (NADPH)-dependent reductase belonging to the aldo-keto reductase (AKR) 1B subfamily. It catalyzes the reduction of aldehydes, some ketones and quinones, and interacts with acetyl-CoA carboxylase and heat shock protein 90α. The enzyme is highly expressed in epithelial cells of the stomach and intestine, but down-regulated in gastrointestinal cancers and inflammatory bowel diseases. In contrast, AKR1B10 expression is low in other tissues, where the enzyme is upregulated in cancers, as well as in non-alcoholic fatty liver disease and several skin diseases. In addition, the enzyme's expression is elevated in cancer cells resistant to clinical anti-cancer drugs. Thus, growing evidence supports AKR1B10 as a potential target for diagnosing and treating these diseases. Herein, we reviewed the literature on the roles of AKR1B10 in a healthy gastrointestinal tract, the development and progression of cancers and acquired chemoresistance, in addition to its gene regulation, functions, and inhibitors.
Collapse
Affiliation(s)
- Satoshi Endo
- Laboratory of Biochemistry, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Toshiyuki Matsunaga
- Education Center of Green Pharmaceutical Sciences, Gifu Pharmaceutical University, Gifu 502-8585, Japan;
| | - Toru Nishinaka
- Laboratory of Biochemistry, Faculty of Pharmacy, Osaka Ohtani University, Tondabayashi 584-8540, Osaka, Japan;
| |
Collapse
|
9
|
Fonseca-Gomes J, Loureiro JA, Tanqueiro SR, Mouro FM, Ruivo P, Carvalho T, Sebastião AM, Diógenes MJ, Pereira MC. In vivo Bio-Distribution and Toxicity Evaluation of Polymeric and Lipid-Based Nanoparticles: A Potential Approach for Chronic Diseases Treatment. Int J Nanomedicine 2020; 15:8609-8621. [PMID: 33177821 PMCID: PMC7652571 DOI: 10.2147/ijn.s267007] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 09/12/2020] [Indexed: 12/15/2022] Open
Abstract
INTRODUCTION Nanoparticles (NPs), as drug delivery systems, appear to be a promising tool for prolonged therapeutic strategies as they allow a controlled drug release over time. However, most of the studies found in the literature simply contemplate the use of a single or low number of dosages with low NPs concentrations. In the context of chronic diseases, like Alzheimer's disease, cancer or human immunodeficiency virus (HIV), where the therapeutic scheme is also chronic, studies with numerous repeated dosages are often neglected. METHODS We screened different NPs, polymeric and lipid-based, in a repeated-dose toxicity study, to evaluate the safety and tissue distribution of promising nanocarriers to be used in the treatment of long-lasting diseases. RESULTS After administrating 24 high concentrated doses of the selected NPs intraperitoneally (i.p.) (3 times a week for 2 months), animals have presented NPs accumulation in different tissues. However, neither toxicity, bodyweight changes nor clinical signs of disease were observed. DISCUSSION This work demonstrates no general adverse effects upon the studied NPs repeated-dose exposure, indicating the most promising NPs to be used in the different therapeutic circumstances, which may be useful in chronic diseases treatment.
Collapse
Affiliation(s)
- João Fonseca-Gomes
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa1649-028, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa1649-028, Portugal
| | - Joana A Loureiro
- LEPABE – Laboratory for Process Engineering, Environment, Biotechnology & Energy, Faculty of Engineering, University of Porto, Porto4200-465, Portugal
| | - Sara R Tanqueiro
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa1649-028, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa1649-028, Portugal
| | - Francisco M Mouro
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa1649-028, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa1649-028, Portugal
| | - Pedro Ruivo
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa1649-028, Portugal
| | - Tânia Carvalho
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa1649-028, Portugal
| | - Ana M Sebastião
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa1649-028, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa1649-028, Portugal
| | - Maria José Diógenes
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa1649-028, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa1649-028, Portugal
| | - Maria Carmo Pereira
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa1649-028, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa1649-028, Portugal
- LEPABE – Laboratory for Process Engineering, Environment, Biotechnology & Energy, Faculty of Engineering, University of Porto, Porto4200-465, Portugal
| |
Collapse
|
10
|
Liu Y, He S, Chen Y, Liu Y, Feng F, Liu W, Guo Q, Zhao L, Sun H. Overview of AKR1C3: Inhibitor Achievements and Disease Insights. J Med Chem 2020; 63:11305-11329. [PMID: 32463235 DOI: 10.1021/acs.jmedchem.9b02138] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Human aldo-keto reductase family 1 member C3 (AKR1C3) is known as a hormone activity regulator and prostaglandin F (PGF) synthase that regulates the occupancy of hormone receptors and cell proliferation. Because of the overexpression in metabolic diseases and various hormone-dependent and -independent carcinomas, as well as the emergence of clinical drug resistance, an increasing number of studies have investigated AKR1C3 inhibitors. Here, we briefly review the physiological and pathological function of AKR1C3 and then summarize the recent development of selective AKR1C3 inhibitors. We propose our viewpoints on the current problems associated with AKR1C3 inhibitors with the aim of providing a reference for future drug discovery and potential therapeutic perspectives on novel, potent, selective AKR1C3 inhibitors.
Collapse
Affiliation(s)
- Yang Liu
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Siyu He
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Ying Chen
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Yijun Liu
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Feng Feng
- Jiangsu Food and Pharmaceuticals Science College, Institute of Food and Pharmaceuticals Research, Huaian 223005, People's Republic of China.,Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Wenyuan Liu
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Qinglong Guo
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Li Zhao
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Haopeng Sun
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| |
Collapse
|
11
|
Wilkin AM, Sullivan R, Trinh T, Edson M, Kozlowski B, Meckling KA. Differential effects of the 1,25D3-MARRS receptor (ERp57/PDIA3) on murine mammary gland development depend on the vitamin D3 dose. Steroids 2020; 158:108621. [PMID: 32119872 DOI: 10.1016/j.steroids.2020.108621] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 02/23/2020] [Accepted: 02/26/2020] [Indexed: 12/19/2022]
Abstract
1,25 dihydroxyvitamin D3 (1,25D3) is the most potent biologically active form of vitamin D3. Its actions on the mammary gland include cell growth inhibition and anti-cancer effects. This study's purpose was to explore the role of the 1,25D3-membrane associated rapid response steroid (MARRS) receptor in the mammary gland using a tissue-specific knockout mouse model and a vitamin D3 dietary intervention. Three genotype groups were created using the Cre/loxp system to knock-down (+/-) and knockout (-/-) the MARRS receptor in epithelial cells of mammary glands (MG). Abdominal MGs were collected from 6-week old female mice (n = 94) on diets of 10,000 IU/kg (excess), 1,000 IU/kg (sufficient) or 0 IU/kg (deficient) of D3. There was a significant interaction between genotype and diet regarding number of terminal end buds (TEBs) (p = 0.001) and ductal coverage of the fat pad (p = 0.03). MARRS -/- mice on the sufficient diet had significantly fewer TEBs (p = 0.001) compared to MARRS +/+ on the same diet, but the opposite effect was seen in mice on the excess diet. There were no effects of genotype on TEBs when animals were vitamin D3 deficient. These results suggest that there is an effect of MARRS on mammary gland development that is dependent on 25(OH)D status, specifically, altering the number of highly proliferative TEBs. Increased numbers of TEBs have been correlated with increased breast cancer risk later in life. Therefore the results of this study warrant further examination of 25(OH)D status and recommendations in adolescent humans to reduce dietary effects on future breast cancer risk.
Collapse
Affiliation(s)
- Allison M Wilkin
- Department of Human Health and Nutritional Sciences, University of Guelph, 50 Stone Rd. E. Guelph, ON N1G 2W1, Canada.
| | - Robert Sullivan
- Department of Human Health and Nutritional Sciences, University of Guelph, 50 Stone Rd. E. Guelph, ON N1G 2W1, Canada.
| | - Thao Trinh
- Department of Human Health and Nutritional Sciences, University of Guelph, 50 Stone Rd. E. Guelph, ON N1G 2W1, Canada.
| | - Michael Edson
- Department of Human Health and Nutritional Sciences, University of Guelph, 50 Stone Rd. E. Guelph, ON N1G 2W1, Canada.
| | - Benjamin Kozlowski
- Department of Human Health and Nutritional Sciences, University of Guelph, 50 Stone Rd. E. Guelph, ON N1G 2W1, Canada.
| | - Kelly A Meckling
- Department of Human Health and Nutritional Sciences, University of Guelph, 50 Stone Rd. E. Guelph, ON N1G 2W1, Canada.
| |
Collapse
|
12
|
Review-Current Concepts in Inflammatory Skin Diseases Evolved by Transcriptome Analysis: In-Depth Analysis of Atopic Dermatitis and Psoriasis. Int J Mol Sci 2020; 21:ijms21030699. [PMID: 31973112 PMCID: PMC7037913 DOI: 10.3390/ijms21030699] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 01/14/2020] [Accepted: 01/16/2020] [Indexed: 12/14/2022] Open
Abstract
During the last decades, high-throughput assessment of gene expression in patient tissues using microarray technology or RNA-Seq took center stage in clinical research. Insights into the diversity and frequency of transcripts in healthy and diseased conditions provide valuable information on the cellular status in the respective tissues. Growing with the technique, the bioinformatic analysis toolkit reveals biologically relevant pathways which assist in understanding basic pathophysiological mechanisms. Conventional classification systems of inflammatory skin diseases rely on descriptive assessments by pathologists. In contrast to this, molecular profiling may uncover previously unknown disease classifying features. Thereby, treatments and prognostics of patients may be improved. Furthermore, disease models in basic research in comparison to the human disease can be directly validated. The aim of this article is not only to provide the reader with information on the opportunities of these techniques, but to outline potential pitfalls and technical limitations as well. Major published findings are briefly discussed to provide a broad overview on the current findings in transcriptomics in inflammatory skin diseases.
Collapse
|
13
|
Rendic SP, Peter Guengerich F. Human cytochrome P450 enzymes 5-51 as targets of drugs and natural and environmental compounds: mechanisms, induction, and inhibition - toxic effects and benefits. Drug Metab Rev 2019; 50:256-342. [PMID: 30717606 DOI: 10.1080/03602532.2018.1483401] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cytochrome P450 (P450, CYP) enzymes have long been of interest due to their roles in the metabolism of drugs, pesticides, pro-carcinogens, and other xenobiotic chemicals. They have also been of interest due to their very critical roles in the biosynthesis and metabolism of steroids, vitamins, and certain eicosanoids. This review covers the 22 (of the total of 57) human P450s in Families 5-51 and their substrate selectivity. Furthermore, included is information and references regarding inducibility, inhibition, and (in some cases) stimulation by chemicals. We update and discuss important aspects of each of these 22 P450s and questions that remain open.
Collapse
Affiliation(s)
| | - F Peter Guengerich
- b Department of Biochemistry , Vanderbilt University School of Medicine , Nashville , TN , USA
| |
Collapse
|
14
|
Cruciani S, Santaniello S, Garroni G, Fadda A, Balzano F, Bellu E, Sarais G, Fais G, Mulas M, Maioli M. Myrtus Polyphenols, from Antioxidants to Anti-Inflammatory Molecules: Exploring a Network Involving Cytochromes P450 and Vitamin D. Molecules 2019; 24:E1515. [PMID: 30999678 PMCID: PMC6515124 DOI: 10.3390/molecules24081515] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 04/05/2019] [Accepted: 04/15/2019] [Indexed: 12/11/2022] Open
Abstract
Inflammatory response represents one of the main mechanisms of healing and tissue function restoration. On the other hand, chronic inflammation leads to excessive secretion of pro-inflammatory cytokines involved in the onset of several diseases. Oxidative stress condition may contribute in worsening inflammatory state fall, increasing reactive oxygen species (ROS) production and cytokines release. Polyphenols can counteract inflammation and oxidative stress, modulating the release of toxic molecules and interacting with physiological defenses, such as cytochromes p450 enzymes. In this paper, we aimed at evaluating the anti-inflammatory properties of different concentrations of Myrtus communis L. pulp and seeds extracts, derived from liquor industrial production, on human fibroblasts. We determined ROS production after oxidative stress induction by H2O2 treatment, and the gene expression of different proinflammatory cytokines. We also analyzed the expression of CYP3A4 and CYP27B1 genes, in order to evaluate the capability of Myrtus polyphenols to influence the metabolic regulation of other molecules, including drugs, ROS, and vitamin D. Our results showed that Myrtus extracts exert a synergic effect with vitamin D in reducing inflammation and ROS production, protecting cells from oxidative stress damages. Moreover, the extracts modulate CYPs expression, preventing chronic inflammation and suggesting their use in development of new therapeutic formulations.
Collapse
Affiliation(s)
- Sara Cruciani
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy.
| | - Sara Santaniello
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy.
| | - Giuseppe Garroni
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy.
| | - Angela Fadda
- Institute of Sciences of Food Production (ISPA), Consiglio Nazionale delle Ricerche (CNR), traversa la Crucca, 3, 07100 Sassari, Italy.
| | - Francesca Balzano
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy.
| | - Emanuela Bellu
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy.
| | - Giorgia Sarais
- Department of Life and Environmental Sciences, University of Cagliari, Via Ospedale 72, 09124 Cagliari, Italy.
| | - Giacomo Fais
- Department of Life and Environmental Sciences, University of Cagliari, Via Ospedale 72, 09124 Cagliari, Italy.
| | - Maurizio Mulas
- Department of Agriculture, University of Sassari, Via De Nicola 9, I-07100 Sassari, Italy.
| | - Margherita Maioli
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy.
- Center for Developmental Biology and Reprogramming- CEDEBIOR, Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy.
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche (CNR), 09042 Monserrato, Cagliari, Italy.
| |
Collapse
|
15
|
Sun HY, Yan YJ, Li YH, Lv L. Reversing effects of ginsenosides on LPS-induced hepatic CYP3A11/3A4 dysfunction through the pregnane X receptor. JOURNAL OF ETHNOPHARMACOLOGY 2019; 229:246-255. [PMID: 30342195 DOI: 10.1016/j.jep.2018.09.041] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 09/28/2018] [Accepted: 09/29/2018] [Indexed: 06/08/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ginseng (Panax ginseng C. A. Meyer), a traditional Chinese medicine, is widely used in the adjunctive therapy of the liver diseases. AIM OF THE STUDY Ginsenosides are one kind of the main active ingredients in ginseng. Although hepatoprotective mechanisms of ginsenosides, such as anti-oxidation, anti-inflammation and anti-apoptosis, have been well studies, little is known about the effect of ginsenosides on drug metabolism in liver. Since CYP3A11/3A4 is a major enzyme catalyzing the drug metabolism in liver, an investigation of the enzyme's expression during the progression of a liver disease will gain valuable information about the hepatic drug metabolism. The purpose of this study was to determine the effect of ginsenosides on the expression of hepatic CYP3A11/3A4 in the lipopolysaccharides (LPS) injured human HepG2 cells and mice. We hypothesize that ginsenosides are important to stabilize CYP3A11/3A4 expression in an injured liver. MATERIALS AND METHODS In this study, LPS was intraperitoneally intermittently injected to induce the liver injury in mice. Ginsenosides were intragastrically administered to mice for 7 days to treat the liver injury. Serum biochemical analysis and histopathological study were taken to evaluate the hepatoprotective effect of ginsenosides. The effect of ginsenosides was also evaluated in human HepG2 cells in the presence and absence of LPS. Real-time PCR and western blotting method were used to detect the mRNA and protein levels of CYP3A11/3A4 in mouse liver tissue and human HepG2 cells. The reporter gene-transfected cells were used to identify upstream targets in HepG2 cells. RESULTS LPS injection in mice resulted in the up-regulation of pro-inflammatory cytokines such as IL-1β, IL-6 and TNF-α in liver, up-regulation of hepatic enzymes such as Tbil, ALT, AST and ALP in serum, and down-regulation of CYP3A11/3A4 expression in liver. Ginsenosides treatment reversed the up-regulation of pro-inflammatory cytokines and serum hepatic enzymes elicited by LPS. Pathological results suggest that ginsenosides reduced liver damage. Moreover, ginsenosides reversed the decrease of CYP3A11/3A4 expression in the liver of LPS-injured mouse and in LPS-treated HepG2 cells. To further investigate the regulatory mechanisms, we found that ginsenosides enhanced the rifampicin-induced pregnane X receptor (PXR) transactivation of the CYP3A4 promoter. Treatment of hPXR-over-expressed cells with ginsenosides increased the rifampicin-inducible expression of CYP3A4 in a concentration-dependent manner. CONCLUSION Ginsenosides reverse the effects of LPS-induced hepatic CYP3A11/3A4 dysfunction, suggesting that the stabilization of the CYP3A11/3A4 expression in an injured liver appears a novel hepatoprotective mechanism of ginsenosides.
Collapse
Affiliation(s)
- Hai-Yan Sun
- Department of Applied Biotechnology, Shenzhen Polytechnic, Shenzhen, Guangdong 518055, People's Republic of China.
| | - Yi-Jing Yan
- Department of Applied Biotechnology, Shenzhen Polytechnic, Shenzhen, Guangdong 518055, People's Republic of China
| | - Yan-Hui Li
- Department of Applied Biotechnology, Shenzhen Polytechnic, Shenzhen, Guangdong 518055, People's Republic of China
| | - Le Lv
- Department of Applied Biotechnology, Shenzhen Polytechnic, Shenzhen, Guangdong 518055, People's Republic of China
| |
Collapse
|
16
|
Wu W, Fan H, Jiang Y, Liao L, Li L, Zhao J, Zhang H, Shrestha C, Xie Z. Regulation of 25-hydroxyvitamin D-1-hydroxylase and 24-hydroxylase in keratinocytes by PTH and FGF23. Exp Dermatol 2018; 27:1201-1209. [PMID: 30066343 DOI: 10.1111/exd.13760] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 06/26/2018] [Accepted: 07/19/2018] [Indexed: 01/05/2023]
Affiliation(s)
- Wenlin Wu
- Department of Endocrinology and Metabolism; The Second Xiang-Ya Hospital; Central South University; Changsha China
| | - Hong Fan
- Department of Endocrinology and Metabolism; The Peace Hospital Attached to Chang-Zhi Medical College; Chang-Zhi China
| | - Yi Jiang
- Department of Pathology; The Second Xiang-Ya Hospital; Central South University; Changsha China
| | - Liyan Liao
- Department of Pathology; The Second Xiang-Ya Hospital; Central South University; Changsha China
| | - Lusha Li
- Department of Endocrinology and Metabolism; The Second Xiang-Ya Hospital; Central South University; Changsha China
| | - Juan Zhao
- Department of Endocrinology and Metabolism; The Second Xiang-Ya Hospital; Central South University; Changsha China
| | - Huiling Zhang
- Department of Endocrinology and Metabolism; The Second Xiang-Ya Hospital; Central South University; Changsha China
| | - Chandrama Shrestha
- Department of Endocrinology and Metabolism; The Second Xiang-Ya Hospital; Central South University; Changsha China
| | - Zhongjian Xie
- Department of Endocrinology and Metabolism; The Second Xiang-Ya Hospital; Central South University; Changsha China
| |
Collapse
|
17
|
Annalora AJ, O'Neil S, Bushman JD, Summerton JE, Marcus CB, Iversen PL. A k-mer based transcriptomics approach for antisense drug discovery targeting the Ewing's family of tumors. Oncotarget 2018; 9:30568-30586. [PMID: 30093970 PMCID: PMC6078127 DOI: 10.18632/oncotarget.25736] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 06/19/2018] [Indexed: 12/22/2022] Open
Abstract
Ewing’s sarcoma treatment failures are associated with high mortality indicating a need for new therapeutic approaches. We used a k-mer counting approach to identify cancer-specific mRNA transcripts in 3 Ewing’s Family Tumor (EFT) cell lines not found in the normal human transcriptome. Phosphorodiamidate morpholino oligomers targeting six EFT-specific transcripts were evaluated for cytotoxicity in TC-32 and CHLA-10 EFT lines and in HEK293 renal epithelial control cells. Average morpholino efficacy (EC50) was 0.66 ± 0.13 in TC-32, 0.25 ± 0.14 in CHLA-10 and 3.07 ± 5.02 µM in HEK293 control cells (ANOVA p < 0.01). Synergy was observed for a cocktail of 12 morpholinos at low dose (0.3 µM) in TC-32 cells, but not in CHLA-10 cells. Paired synergy was also observed in both EFT cell lines when the PHGDH pre-mRNA transcript was targeted in combination with XAGE1B or CYP4F22 transcripts. Antagonism was observed when CCND1 was targeted with XAGE1B or CYP4F22, or when IGFBP-2 was targeted with CCND1 or RBM11. This transcriptome profiling approach is highly effective for cancer drug discovery, as it identified new EWS-specific target genes (e.g. CYP4F22, RBM11 and IGBP-2), and predicted effective antisense agents (EC50 < 1 µM) that demonstrate both synergy and antagonism in combination therapy.
Collapse
Affiliation(s)
- Andrew J Annalora
- Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR 97331, USA
| | - Shawn O'Neil
- Center for Genome Research and Biocomputing, Oregon State University, Corvallis, OR 97331, USA
| | | | | | - Craig B Marcus
- Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR 97331, USA
| | - Patrick L Iversen
- Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR 97331, USA.,LS Pharma, LLC, Grand Junction, CO 81507, USA
| |
Collapse
|
18
|
Bikle DD, Patzek S, Wang Y. Physiologic and pathophysiologic roles of extra renal CYP27b1: Case report and review. Bone Rep 2018; 8:255-267. [PMID: 29963603 PMCID: PMC6021194 DOI: 10.1016/j.bonr.2018.02.004] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 02/06/2018] [Accepted: 02/23/2018] [Indexed: 01/13/2023] Open
Abstract
Although the kidney was initially thought to be the sole organ responsible for the production of 1,25(OH)2D via the enzyme CYP27b1, it is now appreciated that the expression of CYP27b1 in tissues other than the kidney is wide spread. However, the kidney is the major source for circulating 1,25(OH)2D. Only in certain granulomatous diseases such as sarcoidosis does the extra renal tissue produce sufficient 1,25(OH)2D to contribute to the circulating levels, generally associated with hypercalcemia, as illustrated by the case report preceding the review. Therefore the expression of CYP27b1 outside the kidney under normal circumstances begs the question why, and in particular whether the extra renal production of 1,25(OH)2D has physiologic importance. In this chapter this question will be discussed. First we discuss the sites for extra renal 1,25(OH)2D production. This is followed by a discussion of the regulation of CYP27b1 expression and activity in extra renal tissues, pointing out that such regulation is tissue specific and different from that of CYP27b1 in the kidney. Finally the physiologic significance of extra renal 1,25(OH)2D3 production is examined, with special focus on the role of CYP27b1 in regulation of cellular proliferation and differentiation, hormone secretion, and immune function. At this point the data do not clearly demonstrate an essential role for CYP27b1 expression in any tissue outside the kidney, but several examples pointing in this direction are provided. With the availability of the mouse enabling tissue specific deletion of CYP27b1, the role of extra renal CYP27b1 expression in normal and pathologic states can now be addressed definitively.
Collapse
Affiliation(s)
- Daniel D Bikle
- Department of Medicine, Endocrine Research Unit, Veterans Affairs Medical Center, University of California San Francisco, United States
| | - Sophie Patzek
- Department of Medicine, Endocrine Research Unit, Veterans Affairs Medical Center, University of California San Francisco, United States
| | - Yongmei Wang
- Department of Medicine, Endocrine Research Unit, Veterans Affairs Medical Center, University of California San Francisco, United States
| |
Collapse
|
19
|
Abstract
PURPOSE OF REVIEW In addition to the actions of the endocrine hormone, 1alpha,25-dihydroxyvitamin D (1,25(OH)2D) in stimulating intestinal calcium absorption, the regulation of bone mineral metabolism by 1,25(OH)2D is also considered an important contributor to calcium homeostasis. However, recent evidence suggest that 1,25(OH)2D acting either via endocrine or autocrine pathways plays varied roles in bone, which suggests that vitamin D contributes to the maintenance of bone mineral in addition to its catabolic roles. This review highlights the contrasting evidence for the direct action for vitamin D metabolism and activity in bone. RECENT FINDINGS Numerous cells within bone express vitamin D receptor (VDR), synthesise and catabolise 1,25(OH)2D via 25-hydroxyvitamin D 1alpha-hydroxylase (CYP27B1), and 25-hydroxyvitamin D 24-hydroxylase (CYP24A1) enzymes, respectively. Recent evidence suggests that all three genes are required to regulate processes of bone resorption, mineralization and fracture repair. The actions of vitamin D in bone appear to negatively or positively regulate bone mineral depending on the physiological and pathological circumstances, suggesting that vitamin D plays pleiotropic roles in bone.
Collapse
Affiliation(s)
- Paul H Anderson
- Musculoskeletal Biology Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, 5001, Australia.
| |
Collapse
|
20
|
Qixing M, Juqing X, Yajing W, Gaochao D, Wenjie X, Run S, Anpeng W, Lin X, Feng J, Jun W. The expression levels of CYP3A4 and CYP3A5 serve as potential prognostic biomarkers in lung adenocarcinoma. Tumour Biol 2017; 39:1010428317698340. [PMID: 28381170 DOI: 10.1177/1010428317698340] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Lung adenocarcinoma remains to be a high-mortality disease with few effective prognostic biomarkers. Novel biomarkers are urgently demanded to supplement the current prognostic biomarkers. Here, we explored the prognostic value of CYP3A4 and CYP3A5 in lung adenocarcinoma. The tissue microarray was made up of lung adenocarcinoma samples and corresponding normal lung tissues from Nanjing Medical University affiliated Cancer Hospital Tissue Bank. The expression of CYP3A4, together with CYP3A5, was detected by the chip data from Gene Expression Omnibus datasets and immunohistochemistry staining of the tissue microarray. Then, we assessed the relationships between CYP3A4 or CYP3A5 expression level and clinicopathological factors to estimate the clinical significance. Kaplan-Meier curves were applied to analyze the prognosis. Univariate and multivariate Cox analyses were subsequently applied to identify the independent prognostic factors. Immunohistochemistry staining results showed that by comparison with matched normal tissues, CYP3A4 was frequently hyper-expressed in lung adenocarcinoma tissues while CYP3A5 was hypo-expressed, which was consistent with the Gene Expression Omnibus analysis. Kaplan-Meier analysis indicated that high-CYP3A4 or low-CYP3A5 expression level predicted poor survival in lung adenocarcinoma patients. Multivariate Cox analysis found that hypo-expression of CYP3A5 was an independent prognostic factor. Further study revealed that combination of these two markers exhibited a more powerful predictor of poor prognosis, which could target to more accurate survival of lung adenocarcinoma. Our findings indicate that combination of CYP3A4 and CYP3A5 may serve as a novel prognostic biomarker in lung adenocarcinoma, which contribute to the precision of predicting the survival in lung adenocarcinoma.
Collapse
Affiliation(s)
- Mao Qixing
- 1 Department of Thoracic Surgery, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing, China.,2 The Fourth Clinical College of Nanjing Medical University, Nanjing, China.,3 Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Cancer Institute of Jiangsu Province, Nanjing, China
| | - Xu Juqing
- 4 Department of Oncology, Jiangsu Province Geriatric Hospital, Nanjing, China
| | - Wang Yajing
- 5 Nanjing Medical University, Nanjing, China
| | - Dong Gaochao
- 1 Department of Thoracic Surgery, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing, China.,3 Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Cancer Institute of Jiangsu Province, Nanjing, China
| | - Xia Wenjie
- 1 Department of Thoracic Surgery, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing, China.,2 The Fourth Clinical College of Nanjing Medical University, Nanjing, China.,3 Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Cancer Institute of Jiangsu Province, Nanjing, China
| | - Shi Run
- 1 Department of Thoracic Surgery, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing, China.,2 The Fourth Clinical College of Nanjing Medical University, Nanjing, China.,3 Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Cancer Institute of Jiangsu Province, Nanjing, China
| | - Wang Anpeng
- 1 Department of Thoracic Surgery, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing, China.,2 The Fourth Clinical College of Nanjing Medical University, Nanjing, China.,3 Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Cancer Institute of Jiangsu Province, Nanjing, China
| | - Xu Lin
- 1 Department of Thoracic Surgery, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing, China.,3 Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Cancer Institute of Jiangsu Province, Nanjing, China
| | - Jiang Feng
- 1 Department of Thoracic Surgery, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing, China.,3 Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Cancer Institute of Jiangsu Province, Nanjing, China
| | - Wang Jun
- 4 Department of Oncology, Jiangsu Province Geriatric Hospital, Nanjing, China
| |
Collapse
|
21
|
Nishinaka T, Miura T, Shimizu K, Terada T. Identification and characterization of functional antioxidant response elements in the promoter of the aldo-keto reductase AKR1B10 gene. Chem Biol Interact 2017; 276:160-166. [PMID: 28219640 DOI: 10.1016/j.cbi.2017.02.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 12/08/2016] [Accepted: 02/14/2017] [Indexed: 12/16/2022]
Abstract
AKR1B10 is a human-type aldo-keto reductase. The up-regulation of AKR1B10 has been associated with various cancers including non-small cell lung carcinoma, viral and bacterial infections, and skin diseases. However, the mechanisms underlying AKR1B10 gene regulation are not fully understood. We previously indicated the involvement of the transcription factor Nrf2 in AKR1B10 gene regulation. There are at least five potential Nrf2-responsive consensus sequences, so-called antioxidant response elements (AREs), and several ARE-like sequences in the 5'-flanking region up to -3282 bp of the AKR1B10 gene. In the present study, we attempted to identify functional AREs by luciferase reporter analyses using various mutants for each ARE. And we found that only those between -530 and -520 bp (ARE-A), which is the closest location to the translation start site, were functional among the five ARE consensus sites examined. Furthermore, ARE-A functioned co-operatively with the neighboring AP-1 site. Since the AP-1 site resembles ARE, the tandem arrangement of these two elements may be essential for augmented responsiveness to Nrf2 and plays an important role in AKR1B10 gene regulation by various Nrf2-mediating stimuli.
Collapse
Affiliation(s)
- Toru Nishinaka
- Laboratory of Biochemistry, Faculty of Pharmacy, Osaka Ohtani University, 3-11-1 Nishikiori-kita, Tondabayashi, Osaka, 584-8540, Japan.
| | - Takeshi Miura
- Pharmaceutical Education Support Center, School of Pharmacy and Pharmaceutical Sciences, Mukogawa Women's University, 11-68 Koshien, 9-Bancho, Nishinomiya, Hyogo, 663-8179, Japan
| | - Kahori Shimizu
- Laboratory of Biochemistry, Faculty of Pharmacy, Osaka Ohtani University, 3-11-1 Nishikiori-kita, Tondabayashi, Osaka, 584-8540, Japan
| | - Tomoyuki Terada
- Laboratory of Biochemistry, Faculty of Pharmacy, Osaka Ohtani University, 3-11-1 Nishikiori-kita, Tondabayashi, Osaka, 584-8540, Japan
| |
Collapse
|