1
|
Liang G, He J, Chen T, Zhang L, Yu K, Shen W. Identification of ALDH7A1 as a DNA-methylation-driven gene in lung squamous cell carcinoma. Ann Med 2025; 57:2442529. [PMID: 39711312 DOI: 10.1080/07853890.2024.2442529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 10/17/2024] [Accepted: 10/29/2024] [Indexed: 12/24/2024] Open
Abstract
BACKGROUND Deoxyribose nucleic acid (DNA) methylation is an important epigenetic modification that plays an important role in the occurrence and development of tumors. Identifying key methylation-driven genes that affect the prognosis of lung squamous cell carcinoma (LUSC) can provide direction for targeted therapy research. METHODS AND RESULTS Methylation and RNA-seq data were downloaded from The Cancer Genome Atlas (TCGA). The MethylMix package was used to integrate and analyze the methylation and gene expression data from TCGA, and the LUSC dataset (GSE37745) was downloaded from GEO for validation. Forty-five DNA-methylation-driven genes (MDGs) were obtained, and 3 genes (TRIM61, SMIM22, and ALDH7A1) were significantly associated with survival by using univariate and multivariate Cox regression. A risk model was constructed. KM analysis showed that patients with high-risk scores had poor survival. A nomination plot for prognosis prediction of LUSC patients was constructed, which showed a good predictive efficiency for tumor prognosis. The high expression of ALDH7A1 was an independent risk factor for poor prognosis in LUSC. The expression of ALDH7A1 in LUSC was negatively correlated with its methylation status (COR = -0.655). GSEA analysis showed that high expression of ALDH7A1 could activate multiple signaling pathways (JAK-STAT signaling pathway and mTOR signaling pathway). In vitro cell experiments confirmed that in LUSC, silencing ALDH7A1 could inhibit tumor progression, while overexpression of ALDH7A1 could promote tumor progression. CONCLUSION Our results indicated that ALDH7A1, a newly discovered MDG in LUSC, could act as an independent prognostic factor for OS in LUSC, with the potential to become a potential target for LUSC diagnosis and treatment. High expression of ALDH7A1 in LUSC could promote the occurrence and development of tumors. Signaling pathways, such as JAK-STAT and mTOR signaling pathways, might regulate the high expression of ALDH7A1.
Collapse
Affiliation(s)
- Gaofeng Liang
- Department of Thoracic Surgery, The Affiliated LiHuiLi Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Jinxian He
- Department of Thoracic Surgery, The Affiliated LiHuiLi Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Tian Chen
- Department of Radiation Oncology, The Affiliated LiHuiLi Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Liang Zhang
- cDepartment of Respiratory Medicine, The Affiliated LiHuiLi Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Kaizhong Yu
- Department of Thoracic Surgery, The Affiliated LiHuiLi Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Weiyu Shen
- Department of Thoracic Surgery, The Affiliated LiHuiLi Hospital of Ningbo University, Ningbo, Zhejiang, China
| |
Collapse
|
2
|
Nguyen AL, Facey COB, Boman BM. The Significance of Aldehyde Dehydrogenase 1 in Cancers. Int J Mol Sci 2024; 26:251. [PMID: 39796106 PMCID: PMC11720537 DOI: 10.3390/ijms26010251] [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: 11/14/2024] [Revised: 12/16/2024] [Accepted: 12/26/2024] [Indexed: 01/13/2025] Open
Abstract
The goal of this paper is to discuss the role of ALDH isozymes in different cancers, review advances in ALDH1-targeting cancer therapies, and explore a mechanism that explains how ALDH expression becomes elevated during cancer development. ALDH is often overexpressed in cancer, and each isoform has a unique expression pattern and a distinct role in different cancers. The abnormal expression of ALDHs in different cancer types (breast, colorectal, lung, gastric, cervical, melanoma, prostate, and renal) is presented and correlated with patient prognosis. ALDH plays a significant role in various cellular functions, such as metabolism, oxidative stress response, detoxification, and cellular differentiation. Among the ALDH families, ALDH1 has gained considerable attention as a cancer stem cell (CSC) marker due to its significant role in the maintenance of stemness and the differentiation of stem cells (SCs), along with its involvement in tumorigenesis. A description of the cellular mechanisms and physiology of ALDH1 that underlies cancer development is provided. Moreover, current advances in ALDH1-targeting cancer therapies are discussed.
Collapse
Affiliation(s)
- Anh L. Nguyen
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA;
- Center for Translational Cancer Research, Helen F. Graham Cancer Center and Research Institute, 4701 Ogletown-Stanton Road, Newark, DE 19713, USA;
| | - Caroline O. B. Facey
- Center for Translational Cancer Research, Helen F. Graham Cancer Center and Research Institute, 4701 Ogletown-Stanton Road, Newark, DE 19713, USA;
| | - Bruce M. Boman
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA;
- Center for Translational Cancer Research, Helen F. Graham Cancer Center and Research Institute, 4701 Ogletown-Stanton Road, Newark, DE 19713, USA;
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
3
|
Mohamed MA, Elsaman T, Mohamed MS, Eltayib EM. Computational investigations of flavonoids as ALDH isoform inhibitors for treatment of cancer. SAR AND QSAR IN ENVIRONMENTAL RESEARCH 2024; 35:837-875. [PMID: 39503629 DOI: 10.1080/1062936x.2024.2415593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 10/05/2024] [Indexed: 11/08/2024]
Abstract
Human aldehyde dehydrogenases (ALDHs) are a group of 19 isoforms often overexpressed in cancer stem cells (CSCs). These enzymes play critical roles in CSC protection, maintenance, cancer progression, therapeutic resistance, and poor prognosis. Thus, targeting ALDH isoforms offers potential for innovative cancer treatments. Flavonoids, known for their ability to affect multiple cancer-related pathways, have shown anticancer activity by downregulating specific ALDH isoforms. This study aimed to evaluate 830 flavonoids from the PubChem database against five ALDH isoforms (ALDH1A1, ALDH1A2, ALDH1A3, ALDH2, ALDH3A1) using computational methods to identify potent inhibitors. Extra precision (XP) Glide docking and MM-GBSA free binding energy calculations identified several flavonoids with high binding affinities. MD simulation highlighted flavonoids 1, 2, 18, 27, and 42 as potential specific inhibitors for each isoform, respectively. Flavonoid 10 showed high binding affinities for ALDH1A2, ALDH1A3, and ALDH3A1, emerging as a potential multi-ALDH inhibitor. ADMET property evaluation indicated that the promising hits have acceptable drug-like profiles, but further optimization is needed to enhance their therapeutic efficacy and reduce toxicity, making them more effective ALDH inhibitors for future cancer treatment.
Collapse
Affiliation(s)
- M A Mohamed
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University, Sakaka, Kingdom of Saudi Arabia
| | - T Elsaman
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University, Sakaka, Kingdom of Saudi Arabia
| | - M S Mohamed
- Department of Pharmaceutics, College of Pharmacy, Jouf University, Sakaka, Kingdom of Saudi Arabia
| | - E M Eltayib
- Department of Pharmaceutics, College of Pharmacy, Jouf University, Sakaka, Kingdom of Saudi Arabia
| |
Collapse
|
4
|
Kim JH, Lee J, Lee KW, Xiong H, Li M, Kim JS. Trapped in Cells: A Selective Accumulation Approach for Type-I Photodynamic Ablation of Cancer Stem-like Cells. JACS AU 2024; 4:3657-3667. [PMID: 39328753 PMCID: PMC11423316 DOI: 10.1021/jacsau.4c00642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 08/15/2024] [Accepted: 08/16/2024] [Indexed: 09/28/2024]
Abstract
Aldehyde dehydrogenase (ALDH) is an enzyme responsible for converting aldehyde functional groups into carboxylate metabolites. Elevated ALDH activity is a characteristic feature of cancer stem-like cells (CSCs). As a novel approach to target the CSC trait of overexpressing ALDH, we aimed to utilize ALDH activity for the selective accumulation of a photosensitizer in ALDHHigh CSCs. A novel ALDH substrate photosensitizer, SCHO, with thionylated coumarin and N-ethyl-4-(aminomethyl)benzaldehyde was developed to achieve this goal. Our study demonstrated the efficient metabolism of the aldehyde unit of SCHO into carboxylate, leading to its accumulation in ALDHHigh MDA-MB-231 cells. Importantly, we established the selectivity of SCHO as an ALDHHigh cell photosensitizer as it is not a substrate for ABC transporters. SCHO-based photodynamic therapy triggers apoptosis and pyroptosis in MDA-MB-231 cells and further reduces the characteristics of CSCs. Our study presents a novel strategy to target CSCs by exploiting their cellular metabolism to enhance photosensitizer accumulation, highlighting the potential of photodynamic therapy as a powerful tool for eliminating ALDHHigh CSCs.
Collapse
Affiliation(s)
- Ji Hyeon Kim
- Department
of Chemistry, Korea University, Seoul 02841, Korea
- Department
of Chemical and Systems Biology, Chem-H
and Stanford Cancer Institute, Stanford School of Medicine, Stanford
University, Stanford, California 94305, United States
| | - Jieun Lee
- Department
of Chemistry, Korea University, Seoul 02841, Korea
| | - Kyung-Woo Lee
- Department
of Chemistry, Korea University, Seoul 02841, Korea
| | - Hao Xiong
- Department
of Chemistry, Korea University, Seoul 02841, Korea
| | - Mingle Li
- College
of Materials Science and Engineering, Shenzhen
University, Shenzhen 518060, China
| | - Jong Seung Kim
- Department
of Chemistry, Korea University, Seoul 02841, Korea
| |
Collapse
|
5
|
Duan JJ, Cai J, Gao L, Yu SC. ALDEFLUOR activity, ALDH isoforms, and their clinical significance in cancers. J Enzyme Inhib Med Chem 2023; 38:2166035. [PMID: 36651035 PMCID: PMC9858439 DOI: 10.1080/14756366.2023.2166035] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
High aldehyde dehydrogenase (ALDH) activity is a metabolic feature of adult stem cells and various cancer stem cells (CSCs). The ALDEFLUOR system is currently the most commonly used method for evaluating ALDH enzyme activity in viable cells. This system is applied extensively in the isolation of normal stem cells and CSCs from heterogeneous cell populations. For many years, ALDH1A1 has been considered the most important subtype among the 19 ALDH family members in determining ALDEFLUOR activity. However, in recent years, studies of many types of normal and tumour tissues have demonstrated that other ALDH subtypes can also significantly influence ALDEFLUOR activity. In this article, we briefly review the relationships between various members of the ALDH family and ALDEFLUOR activity. The clinical significance of these ALDH isoforms in different cancers and possible directions for future studies are also summarised.
Collapse
Affiliation(s)
- Jiang-Jie Duan
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital; Third Military Medical University (Army Medical University), Chongqing, China,International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing, China,Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Chongqing, China,Ministry of Education, Key Laboratory of Cancer Immunopathology, Chongqing, China
| | - Jiao Cai
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital; Third Military Medical University (Army Medical University), Chongqing, China,International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing, China,Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Chongqing, China,Ministry of Education, Key Laboratory of Cancer Immunopathology, Chongqing, China
| | - Lei Gao
- Department of Hematology, Xinqiao Hospital; Third Medical University (Army Medical University), Chongqing, China
| | - Shi-Cang Yu
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital; Third Military Medical University (Army Medical University), Chongqing, China,International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing, China,Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Chongqing, China,Ministry of Education, Key Laboratory of Cancer Immunopathology, Chongqing, China,Jin-feng Laboratory, Chongqing, China,CONTACT Shi-Cang Yu Department of Stem Cell and Regenerative Medicine, Third Military Medical University (Army Medical University), Chongqing400038, China
| |
Collapse
|
6
|
Xanthis V, Mantso T, Dimtsi A, Pappa A, Fadouloglou VE. Human Aldehyde Dehydrogenases: A Superfamily of Similar Yet Different Proteins Highly Related to Cancer. Cancers (Basel) 2023; 15:4419. [PMID: 37686694 PMCID: PMC10650815 DOI: 10.3390/cancers15174419] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/30/2023] [Accepted: 09/01/2023] [Indexed: 09/10/2023] Open
Abstract
The superfamily of human aldehyde dehydrogenases (hALDHs) consists of 19 isoenzymes which are critical for several physiological and biosynthetic processes and play a major role in the organism's detoxification via the NAD(P) dependent oxidation of numerous endogenous and exogenous aldehyde substrates to their corresponding carboxylic acids. Over the last decades, ALDHs have been the subject of several studies as it was revealed that their differential expression patterns in various cancer types are associated either with carcinogenesis or promotion of cell survival. Here, we attempt to provide a thorough review of hALDHs' diverse functions and 3D structures with particular emphasis on their role in cancer pathology and resistance to chemotherapy. We are especially interested in findings regarding the association of structural features and their changes with effects on enzymes' functionalities. Moreover, we provide an updated outline of the hALDHs inhibitors utilized in experimental or clinical settings for cancer therapy. Overall, this review aims to provide a better understanding of the impact of ALDHs in cancer pathology and therapy from a structural perspective.
Collapse
Affiliation(s)
| | | | | | | | - Vasiliki E. Fadouloglou
- Department of Molecular Biology & Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| |
Collapse
|
7
|
Yang H, Su M, Liu M, Sheng Y, Zhu L, Yang L, Mu R, Zou J, Liu X, Liu L. Hepatic retinaldehyde deficiency is involved in diabetes deterioration by enhancing PCK1- and G6PC-mediated gluconeogenesis. Acta Pharm Sin B 2023; 13:3728-3743. [PMID: 37719384 PMCID: PMC10501888 DOI: 10.1016/j.apsb.2023.06.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 03/14/2023] [Accepted: 05/06/2023] [Indexed: 09/19/2023] Open
Abstract
Type 2 diabetes (T2D) is often accompanied with an induction of retinaldehyde dehydrogenase 1 (RALDH1 or ALDH1A1) expression and a consequent decrease in hepatic retinaldehyde (Rald) levels. However, the role of hepatic Rald deficiency in T2D progression remains unclear. In this study, we demonstrated that reversing T2D-mediated hepatic Rald deficiency by Rald or citral treatments, or liver-specific Raldh1 silencing substantially lowered fasting glycemia levels, inhibited hepatic glucogenesis, and downregulated phosphoenolpyruvate carboxykinase 1 (PCK1) and glucose-6-phosphatase (G6PC) expression in diabetic db/db mice. Fasting glycemia and Pck1/G6pc mRNA expression levels were strongly negatively correlated with hepatic Rald levels, indicating the involvement of hepatic Rald depletion in T2D deterioration. A similar result that liver-specific Raldh1 silencing improved glucose metabolism was also observed in high-fat diet-fed mice. In primary human hepatocytes and oleic acid-treated HepG2 cells, Rald or Rald + RALDH1 silencing resulted in decreased glucose production and downregulated PCK1/G6PC mRNA and protein expression. Mechanistically, Rald downregulated direct repeat 1-mediated PCK1 and G6PC expression by antagonizing retinoid X receptor α, as confirmed by luciferase reporter assays and molecular docking. These results highlight the link between hepatic Rald deficiency, glucose dyshomeostasis, and the progression of T2D, whilst also suggesting RALDH1 as a potential therapeutic target for T2D.
Collapse
Affiliation(s)
- Hanyu Yang
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Mengxiang Su
- Department of Pharmaceutical Analysis, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Ming Liu
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Yun Sheng
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Liang Zhu
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Lu Yang
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Ruijing Mu
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Jianjun Zou
- Department of Clinical Pharmacology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Xiaodong Liu
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Li Liu
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
8
|
Zhou X, Mitra R, Hou F, Zhou S, Wang L, Jiang W. Genomic Landscape and Potential Regulation of RNA Editing in Drug Resistance. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2207357. [PMID: 36912579 PMCID: PMC10190536 DOI: 10.1002/advs.202207357] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/31/2023] [Indexed: 05/18/2023]
Abstract
Adenosine-to-inosine RNA editing critically affects the response of cancer therapies. However, comprehensive identification of drug resistance-related RNA editing events and systematic understanding of how RNA editing mediates anticancer drug resistance remain unclear. Here, 7157 differential editing sites (DESs) are identified from 98 127 informative RNA editing sites in tumor tissues, many of which are validated in cancer cell lines. Diverse editing patterns of DESs are discovered in resistant samples, which could not be fully explained by adenosine deaminase acting on RNA enzymes. Some RNA-binding proteins are identified that potentially regulate these editing events. Notably, the DESs are significantly enriched in 3'-untranslated regions (3'-UTRs). The impact of DESs in 3'-UTR on the microRNA (miRNA) regulations is explored, and some triplets (DES, miRNA, and gene) that may contribute to drug resistance are identified. In addition, it is determined that the functions of genes enriched with DESs are associated with drug resistance, such as apoptosis, drug metabolism, and DNA synthesis involved in DNA repair. An online resource (http://www.jianglab.cn/REDR/) to support convenient retrieval of DESs is also built. The findings reveal the landscape and potential regulatory mechanism of RNA editing in drug resistance, providing new therapeutic targets for reversing drug resistance.
Collapse
Affiliation(s)
- Xu Zhou
- Department of Biomedical EngineeringNanjing University of Aeronautics and AstronauticsNanjing211106P. R. China
| | - Ramkrishna Mitra
- Department of PharmacologyPhysiology, and Cancer BiologySidney Kimmel Cancer CenterThomas Jefferson UniversityPhiladelphiaPennsylvania19107USA
| | - Fei Hou
- Department of Biomedical EngineeringNanjing University of Aeronautics and AstronauticsNanjing211106P. R. China
| | - Shunheng Zhou
- Department of Biomedical EngineeringNanjing University of Aeronautics and AstronauticsNanjing211106P. R. China
| | - Lihong Wang
- Department of PathophysiologySchool of MedicineSoutheast UniversityNanjing210009P. R. China
| | - Wei Jiang
- Department of Biomedical EngineeringNanjing University of Aeronautics and AstronauticsNanjing211106P. R. China
| |
Collapse
|
9
|
Karaaslanli A, Saha S, Maiti T, Aviyente S. Kernelized multiview signed graph learning for single-cell RNA sequencing data. BMC Bioinformatics 2023; 24:127. [PMID: 37016281 PMCID: PMC10071725 DOI: 10.1186/s12859-023-05250-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Accepted: 03/22/2023] [Indexed: 04/06/2023] Open
Abstract
BACKGROUND Characterizing the topology of gene regulatory networks (GRNs) is a fundamental problem in systems biology. The advent of single cell technologies has made it possible to construct GRNs at finer resolutions than bulk and microarray datasets. However, cellular heterogeneity and sparsity of the single cell datasets render void the application of regular Gaussian assumptions for constructing GRNs. Additionally, most GRN reconstruction approaches estimate a single network for the entire data. This could cause potential loss of information when single cell datasets are generated from multiple treatment conditions/disease states. RESULTS To better characterize single cell GRNs under different but related conditions, we propose the joint estimation of multiple networks using multiple signed graph learning (scMSGL). The proposed method is based on recently developed graph signal processing (GSP) based graph learning, where GRNs and gene expressions are modeled as signed graphs and graph signals, respectively. scMSGL learns multiple GRNs by optimizing the total variation of gene expressions with respect to GRNs while ensuring that the learned GRNs are similar to each other through regularization with respect to a learned signed consensus graph. We further kernelize scMSGL with the kernel selected to suit the structure of single cell data. CONCLUSIONS scMSGL is shown to have superior performance over existing state of the art methods in GRN recovery on simulated datasets. Furthermore, scMSGL successfully identifies well-established regulators in a mouse embryonic stem cell differentiation study and a cancer clinical study of medulloblastoma.
Collapse
Affiliation(s)
- Abdullah Karaaslanli
- Department of Electrical and Computer Engineering, Michigan State University, East Lansing, MI, USA.
| | - Satabdi Saha
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Tapabrata Maiti
- Department of Statistics and Probability, Michigan State University, East Lansing, MI, USA
| | - Selin Aviyente
- Department of Electrical and Computer Engineering, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
10
|
The Expanding Role of Cancer Stem Cell Marker ALDH1A3 in Cancer and Beyond. Cancers (Basel) 2023; 15:cancers15020492. [PMID: 36672441 PMCID: PMC9857290 DOI: 10.3390/cancers15020492] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/08/2023] [Accepted: 01/11/2023] [Indexed: 01/15/2023] Open
Abstract
Aldehyde dehydrogenase 1A3 (ALDH1A3) is one of 19 ALDH enzymes expressed in humans, and it is critical in the production of hormone receptor ligand retinoic acid (RA). We review the role of ALDH1A3 in normal physiology, its identification as a cancer stem cell marker, and its modes of action in cancer and other diseases. ALDH1A3 is often over-expressed in cancer and promotes tumor growth, metastasis, and chemoresistance by altering gene expression, cell signaling pathways, and glycometabolism. The increased levels of ALDH1A3 in cancer occur due to genetic amplification, epigenetic modifications, post-transcriptional regulation, and post-translational modification. Finally, we review the potential of targeting ALDH1A3, with both general ALDH inhibitors and small molecules specifically designed to inhibit ALDH1A3 activity.
Collapse
|
11
|
Dhanyamraju PK, Schell TD, Amin S, Robertson GP. Drug-Tolerant Persister Cells in Cancer Therapy Resistance. Cancer Res 2022; 82:2503-2514. [PMID: 35584245 PMCID: PMC9296591 DOI: 10.1158/0008-5472.can-21-3844] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 03/15/2022] [Accepted: 05/09/2022] [Indexed: 01/21/2023]
Abstract
One of the current stumbling blocks in our fight against cancer is the development of acquired resistance to therapy, which is attributable to approximately 90% of cancer-related deaths. Undercutting this process during treatment could significantly improve cancer management. In many cases, drug resistance is mediated by a drug-tolerant persister (DTP) cell subpopulation present in tumors, often referred to as persister cells. This review provides a summary of currently known persister cell subpopulations and approaches to target them. A specific DTP cell subpopulation with elevated levels of aldehyde dehydrogenase (ALDH) activity has stem cell-like characteristics and a high level of plasticity, enabling them to switch rapidly between high and low ALDH activity. Further studies are required to fully elucidate the functions of ALDH-high DTP cells, how they withstand drug concentrations that kill other cells, and how they rapidly adapt under levels of high cellular stress and eventually lead to more aggressive, recurrent, and drug-resistant cancer. Furthermore, this review addresses the processes used by the ALDH-high persister cell subpopulation to enable cancer progression, the ALDH isoforms important in these processes, interactions of ALDH-high DTPs with the tumor microenvironment, and approaches to therapeutically modulate this subpopulation in order to more effectively manage cancer.
Collapse
Affiliation(s)
- Pavan Kumar Dhanyamraju
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA 17033
| | - Todd D Schell
- Departments of Microbiology and Immunology, The Pennsylvania State University College of Medicine, Hershey, PA 17033
| | - Shantu Amin
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA 17033
| | - Gavin P Robertson
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA 17033
- Department of Pathology, The Pennsylvania State University College of Medicine, Hershey, PA 17033
- Department of Dermatology, The Pennsylvania State University College of Medicine, Hershey, PA 17033
- Department of Surgery, The Pennsylvania State University College of Medicine, Hershey, PA 17033
- The Penn State Melanoma and Skin Cancer Center, The Pennsylvania State University College of Medicine, Hershey, PA 17033
- Penn State Melanoma Therapeutics Program, The Pennsylvania State University College of Medicine, Hershey, PA 17033
| |
Collapse
|
12
|
Rezayatmand H, Razmkhah M, Razeghian-Jahromi I. Drug resistance in cancer therapy: the Pandora's Box of cancer stem cells. Stem Cell Res Ther 2022; 13:181. [PMID: 35505363 PMCID: PMC9066908 DOI: 10.1186/s13287-022-02856-6] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 04/14/2022] [Indexed: 12/18/2022] Open
Abstract
Drug resistance is the main culprit of failure in cancer therapy that may lead to cancer relapse. This resistance mostly originates from rare, but impactful presence of cancer stem cells (CSCs). Ability to self-renewal and differentiation into heterogeneous cancer cells, and harboring morphologically and phenotypically distinct cells are prominent features of CSCs. Also, CSCs substantially contribute to metastatic dissemination. They possess several mechanisms that help them to survive even after exposure to chemotherapy drugs. Although chemotherapy is able to destroy the bulk of tumor cells, CSCs are left almost intact, and make tumor entity resistant to treatment. Eradication of a tumor mass needs complete removal of tumor cells as well as CSCs. Therefore, it is important to elucidate key features underlying drug resistance raised by CSCs in order to apply effective treatment strategies. However, the challenging point that threatens safety and specificity of chemotherapy is the common characteristics between CSCs and normal peers such as signaling pathways and markers. In the present study, we tried to present a comprehensive appraisal on CSCs, mechanisms of their drug resistance, and recent therapeutic methods targeting this type of noxious cells.
Collapse
Affiliation(s)
| | - Mahboobeh Razmkhah
- Shiraz Institute for Cancer Research, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Iman Razeghian-Jahromi
- Cardiovascular Research Center, Shiraz University of Medical Sciences, 3rd Floor, Mohammad Rasoolallah Research Tower, Namazi Hospital, Shiraz, Iran.
| |
Collapse
|
13
|
Zanoni M, Bravaccini S, Fabbri F, Arienti C. Emerging Roles of Aldehyde Dehydrogenase Isoforms in Anti-cancer Therapy Resistance. Front Med (Lausanne) 2022; 9:795762. [PMID: 35299840 PMCID: PMC8920988 DOI: 10.3389/fmed.2022.795762] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 02/10/2022] [Indexed: 12/19/2022] Open
Abstract
Aldehyde dehydrogenases (ALDHs) are a family of detoxifying enzymes often upregulated in cancer cells and associated with therapeutic resistance. In humans, the ALDH family comprises 19 isoenzymes active in the majority of mammalian tissues. Each ALDH isoform has a specific differential expression pattern and most of them have individual functional roles in cancer. ALDHs are overexpressed in subpopulations of cancer cells with stem-like features, where they are involved in several processes including cellular proliferation, differentiation, detoxification and survival, participating in lipids and amino acid metabolism and retinoic acid synthesis. In particular, ALDH enzymes protect cancer cells by metabolizing toxic aldehydes in less reactive and more soluble carboxylic acids. High metabolic activity as well as conventional anticancer therapies contribute to aldehyde accumulation, leading to DNA double strand breaks (DSB) through the generation of reactive oxygen species (ROS) and lipid peroxidation. ALDH overexpression is crucial not only for the survival of cancer stem cells but can also affect immune cells of the tumour microenvironment (TME). The reduction of ROS amount and the increase in retinoic acid signaling impairs immunogenic cell death (ICD) inducing the activation and stability of immunosuppressive regulatory T cells (Tregs). Dissecting the role of ALDH specific isoforms in the TME can open new scenarios in the cancer treatment. In this review, we summarize the current knowledge about the role of ALDH isoforms in solid tumors, in particular in association with therapy-resistance.
Collapse
Affiliation(s)
- Michele Zanoni
- Biosciences Laboratory,IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | | | | | - Chiara Arienti
- Biosciences Laboratory,IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| |
Collapse
|
14
|
Ryan SL, Dave KA, Beard S, Gyimesi M, McTaggart M, Sahin KB, Molloy C, Gandhi NS, Boittier E, O'Leary CG, Shah ET, Bolderson E, Baird AM, Richard DJ, O'Byrne KJ, Adams MN. Identification of Proteins Deregulated by Platinum-Based Chemotherapy as Novel Biomarkers and Therapeutic Targets in Non-Small Cell Lung Cancer. Front Oncol 2021; 11:615967. [PMID: 33777753 PMCID: PMC7991912 DOI: 10.3389/fonc.2021.615967] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 01/19/2021] [Indexed: 12/24/2022] Open
Abstract
Platinum-based chemotherapy remains the cornerstone of treatment for most people with non-small cell lung cancer (NSCLC), either as adjuvant therapy in combination with a second cytotoxic agent or in combination with immunotherapy. Resistance to therapy, either in the form of primary refractory disease or evolutionary resistance, remains a significant issue in the treatment of NSCLC. Hence, predictive biomarkers and novel combinational strategies are required to improve the effectiveness and durability of treatment response 6for people with NSCLC. The aim of this study was to identify novel biomarkers and/or druggable proteins from deregulated protein networks within non-oncogene driven disease that are involved in the cellular response to cisplatin. Following exposure of NSCLC cells to cisplatin, in vitro quantitative mass spectrometry was applied to identify altered protein response networks. A total of 65 proteins were significantly deregulated following cisplatin exposure. These proteins were assessed to determine if they are druggable targets using novel machine learning approaches and to identify whether these proteins might serve as prognosticators of platinum therapy. Our data demonstrate novel candidates and drug-like molecules warranting further investigation to improve response to platinum agents in NSCLC.
Collapse
Affiliation(s)
- Sarah-Louise Ryan
- Faculty of Health, School of Biomedical Sciences, Institute of Health and Biomedical Innovation, Translational Research Institute, Queensland University of Technology, Woolloongabba, QLD, Australia
| | - Keyur A Dave
- Faculty of Health, School of Biomedical Sciences, Institute of Health and Biomedical Innovation, Translational Research Institute, Queensland University of Technology, Woolloongabba, QLD, Australia
| | - Sam Beard
- Faculty of Health, School of Biomedical Sciences, Institute of Health and Biomedical Innovation, Translational Research Institute, Queensland University of Technology, Woolloongabba, QLD, Australia
| | - Martina Gyimesi
- Faculty of Health, School of Biomedical Sciences, Institute of Health and Biomedical Innovation, Translational Research Institute, Queensland University of Technology, Woolloongabba, QLD, Australia
| | - Matthew McTaggart
- Faculty of Health, School of Biomedical Sciences, Institute of Health and Biomedical Innovation, Translational Research Institute, Queensland University of Technology, Woolloongabba, QLD, Australia
| | - Katherine B Sahin
- Faculty of Health, School of Biomedical Sciences, Institute of Health and Biomedical Innovation, Translational Research Institute, Queensland University of Technology, Woolloongabba, QLD, Australia
| | - Christopher Molloy
- Faculty of Health, School of Biomedical Sciences, Institute of Health and Biomedical Innovation, Translational Research Institute, Queensland University of Technology, Woolloongabba, QLD, Australia
| | - Neha S Gandhi
- Faculty of Science and Engineering, School of Chemistry and Physics, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD, Australia
| | - Eric Boittier
- Faculty of Health, School of Biomedical Sciences, Institute of Health and Biomedical Innovation, Translational Research Institute, Queensland University of Technology, Woolloongabba, QLD, Australia
| | - Connor G O'Leary
- Faculty of Health, School of Biomedical Sciences, Institute of Health and Biomedical Innovation, Translational Research Institute, Queensland University of Technology, Woolloongabba, QLD, Australia.,Cancer Services, Princess Alexandra Hospital, Woolloongabba, QLD, Australia
| | - Esha T Shah
- Faculty of Health, School of Biomedical Sciences, Institute of Health and Biomedical Innovation, Translational Research Institute, Queensland University of Technology, Woolloongabba, QLD, Australia
| | - Emma Bolderson
- Faculty of Health, School of Biomedical Sciences, Institute of Health and Biomedical Innovation, Translational Research Institute, Queensland University of Technology, Woolloongabba, QLD, Australia
| | - Anne-Marie Baird
- Department of Clinical Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland.,Thoracic Oncology Research Group, Labmed Directorate, St. James's Hospital, Dublin, Ireland
| | - Derek J Richard
- Faculty of Health, School of Biomedical Sciences, Institute of Health and Biomedical Innovation, Translational Research Institute, Queensland University of Technology, Woolloongabba, QLD, Australia
| | - Kenneth J O'Byrne
- Faculty of Health, School of Biomedical Sciences, Institute of Health and Biomedical Innovation, Translational Research Institute, Queensland University of Technology, Woolloongabba, QLD, Australia.,Cancer Services, Princess Alexandra Hospital, Woolloongabba, QLD, Australia.,Thoracic Oncology Research Group, Labmed Directorate, St. James's Hospital, Dublin, Ireland
| | - Mark N Adams
- Faculty of Health, School of Biomedical Sciences, Institute of Health and Biomedical Innovation, Translational Research Institute, Queensland University of Technology, Woolloongabba, QLD, Australia
| |
Collapse
|
15
|
Verma M, Khan MIK, Kadumuri RV, Chakrapani B, Awasthi S, Mahesh A, Govindaraju G, Chavali PL, Rajavelu A, Chavali S, Dhayalan A. PRMT3 interacts with ALDH1A1 and regulates gene-expression by inhibiting retinoic acid signaling. Commun Biol 2021; 4:109. [PMID: 33495566 PMCID: PMC7835222 DOI: 10.1038/s42003-020-01644-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Accepted: 12/15/2020] [Indexed: 12/23/2022] Open
Abstract
Protein arginine methyltransferase 3 (PRMT3) regulates protein functions by introducing asymmetric dimethylation marks at the arginine residues in proteins. However, very little is known about the interaction partners of PRMT3 and their functional outcomes. Using yeast-two hybrid screening, we identified Retinal dehydrogenase 1 (ALDH1A1) as a potential interaction partner of PRMT3 and confirmed this interaction using different methods. ALDH1A1 regulates variety of cellular processes by catalyzing the conversion of retinaldehyde to retinoic acid. By molecular docking and site-directed mutagenesis, we identified the specific residues in the catalytic domain of PRMT3 that facilitate interaction with the C-terminal region of ALDH1A1. PRMT3 inhibits the enzymatic activity of ALDH1A1 and negatively regulates the expression of retinoic acid responsive genes in a methyltransferase activity independent manner. Our findings show that in addition to regulating protein functions by introducing methylation modifications, PRMT3 could also regulate global gene expression through protein-protein interactions. Here, the authors demonstrate that protein arginine methyltransferase 3 (PRMT3) interacts with and inhibits the retinal dehydrogenase ALDH1A1, negatively regulating the expression of retinoic acid responsive genes. This study shows that PRMT3 affects diverse biological processes not only by globally regulating protein function through methylation but also by regulating gene expression.
Collapse
Affiliation(s)
- Mamta Verma
- Department of Biotechnology, Pondicherry University, Puducherry, 605014, India
| | - Mohd Imran K Khan
- Department of Biotechnology, Pondicherry University, Puducherry, 605014, India
| | - Rajashekar Varma Kadumuri
- Department of Biology, Indian Institute of Science Education and Research (IISER) Tirupati, Tirupati, Andhra Pradesh, 517507, India
| | - Baskar Chakrapani
- Department of Biotechnology, Pondicherry University, Puducherry, 605014, India
| | - Sharad Awasthi
- Department of Biotechnology, Pondicherry University, Puducherry, 605014, India
| | - Arun Mahesh
- Department of Biotechnology, Pondicherry University, Puducherry, 605014, India
| | - Gayathri Govindaraju
- Interdisciplinary Biology, Rajiv Gandhi Centre for Biotechnology, Trivandrum, Kerala, 695014, India
| | - Pavithra L Chavali
- CSIR-Centre for Cellular & Molecular Biology, Hyderabad, Telangana, 500007, India
| | - Arumugam Rajavelu
- Interdisciplinary Biology, Rajiv Gandhi Centre for Biotechnology, Trivandrum, Kerala, 695014, India
| | - Sreenivas Chavali
- Department of Biology, Indian Institute of Science Education and Research (IISER) Tirupati, Tirupati, Andhra Pradesh, 517507, India.
| | - Arunkumar Dhayalan
- Department of Biotechnology, Pondicherry University, Puducherry, 605014, India.
| |
Collapse
|
16
|
Koenders STA, van Rooden EJ, van den Elst H, Florea BI, Overkleeft HS, van der Stelt M. STA-55, an Easily Accessible, Broad-Spectrum, Activity-Based Aldehyde Dehydrogenase Probe. Chembiochem 2020; 21:1911-1917. [PMID: 31985142 DOI: 10.1002/cbic.201900771] [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: 12/19/2019] [Indexed: 12/22/2022]
Abstract
Aldehyde dehydrogenases (ALDHs) convert aldehydes into carboxylic acids and are often upregulated in cancer. They have been linked to therapy resistance and are therefore potential therapeutic targets. However, only a few selective and potent inhibitors are currently available for this group of enzymes. Competitive activity-based protein profiling (ABPP) would aid the development and validation of new selective inhibitors. Herein, a broad-spectrum activity-based probe that reports on several ALDHs is presented. This probe was used in a competitive ABPP protocol against three ALDH inhibitors in lung cancer cells to determine their selectivity profiles and establish their target engagement.
Collapse
Affiliation(s)
- Sebastiaan T A Koenders
- Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Eva J van Rooden
- Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Hans van den Elst
- Department of Bio-Organic Synthesis, Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Bogdan I Florea
- Department of Bio-Organic Synthesis, Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Herman S Overkleeft
- Department of Bio-Organic Synthesis, Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Mario van der Stelt
- Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| |
Collapse
|
17
|
Dinavahi SS, Gowda R, Gowda K, Bazewicz CG, Chirasani VR, Battu MB, Berg A, Dokholyan NV, Amin S, Robertson GP. Development of a Novel Multi-Isoform ALDH Inhibitor Effective as an Antimelanoma Agent. Mol Cancer Ther 2020; 19:447-459. [PMID: 31754071 PMCID: PMC10763724 DOI: 10.1158/1535-7163.mct-19-0360] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 08/22/2019] [Accepted: 11/13/2019] [Indexed: 11/16/2022]
Abstract
The aldehyde dehydrogenases (ALDH) are a major family of detoxifying enzymes that contribute to cancer progression and therapy resistance. ALDH overexpression is associated with a poor prognosis in many cancer types. The use of multi-ALDH isoform or isoform-specific ALDH inhibitors as anticancer agents is currently hindered by the lack of viable candidates. Most multi-ALDH isoform inhibitors lack bioavailability and are nonspecific or toxic, whereas most isoform-specific inhibitors are not effective as monotherapy due to the overlapping functions of ALDH family members. The present study details the development of a novel, potent, multi-isoform ALDH inhibitor, called KS100. The rationale for drug development was that inhibition of multiple ALDH isoforms might be more efficacious for cancer compared with isoform-specific inhibition. Enzymatic IC50s of KS100 were 207, 1,410, and 240 nmol/L toward ALDH1A1, 2, and 3A1, respectively. Toxicity of KS100 was mitigated by development of a nanoliposomal formulation, called NanoKS100. NanoKS100 had a loading efficiency of approximately 69% and was stable long-term. NanoKS100 was 5-fold more selective for killing melanoma cells compared with normal human fibroblasts. NanoKS100 administered intravenously at a submaximal dose (3-fold lower) was effective at inhibiting xenografted melanoma tumor growth by approximately 65% without organ-related toxicity. Mechanistically, inhibition by KS100 significantly reduced total cellular ALDH activity to increase reactive oxygen species generation, lipid peroxidation, and accumulation of toxic aldehydes leading to apoptosis and autophagy. Collectively, these data suggest the successful preclinical development of a nontoxic, bioavailable, nanoliposomal formulation containing a novel multi-ALDH isoform inhibitor effective in the treatment of cancer.
Collapse
Affiliation(s)
- Saketh S Dinavahi
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
- The Melanoma and Skin Cancer Center, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
- The Melanoma Therapeutics Program, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Raghavendra Gowda
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
- The Melanoma and Skin Cancer Center, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
- The Melanoma Therapeutics Program, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
- Foreman Foundation for Melanoma Research, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Krishne Gowda
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Christopher G Bazewicz
- The Melanoma and Skin Cancer Center, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
- The Melanoma Therapeutics Program, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
- Department of Dermatology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Venkat R Chirasani
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Madhu Babu Battu
- Laboratory of Molecular Cell Biology, Centre for DNA Fingerprinting and Diagnostics, Uppal, Hyderabad, India
| | - Arthur Berg
- Department of Public Health Sciences, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Nikolay V Dokholyan
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Shantu Amin
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Gavin P Robertson
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania.
- The Melanoma and Skin Cancer Center, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
- The Melanoma Therapeutics Program, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
- Foreman Foundation for Melanoma Research, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
- Department of Dermatology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
- Department of Pathology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
- Department of Surgery, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| |
Collapse
|
18
|
Dinavahi SS, Gowda R, Bazewicz CG, Battu MB, Lin JM, Chitren RJ, Pandey MK, Amin S, Robertson GP, Gowda K. Design, synthesis characterization and biological evaluation of novel multi-isoform ALDH inhibitors as potential anticancer agents. Eur J Med Chem 2019; 187:111962. [PMID: 31887569 DOI: 10.1016/j.ejmech.2019.111962] [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] [Received: 09/11/2019] [Revised: 12/03/2019] [Accepted: 12/09/2019] [Indexed: 12/31/2022]
Abstract
The aldehyde dehydrogenases (ALDHs) are a family of detoxifying enzymes that are overexpressed in various cancers. Increased expression of ALDH is associated with poor prognosis, stemness, and drug resistance. Because of the critical role of ALDH in cancer stem cells, several ALDH inhibitors have been developed. Nonetheless, all these inhibitors either lack efficacy or are too toxic or have not been tested extensively. Thus, the continued development of ALDH inhibitors is warranted. In this study, we designed and synthesized potent multi-ALDH isoform inhibitors based on the isatin backbone. The early molecular docking studies and enzymatic tests revealed that 3(a-l) and 4(a-l) are the potent ALDH1A1, ALDHA2, and ALDH3A1 inhibitors. ALDH inhibitory IC50s of 3(a-l) and 4(a-l) were 230 nM to >10,000 nM for ALDH1A1, 939 nM to >10,000 nM for ALDH2 and 193 nM to >10,000 nM for ALDH3A1. The most potent compounds 3(h-l) had IC50s for killing melanoma cells ranged from 2.1 to 5.7 μM, while for colon cancer cells, it ranged from 2.5 to 5.8 μM and for multiple myeloma cells ranging from 0.3 to 4.7 μM. Toxicity studies of 3(h-l) revealed that 3h to be the least toxic multi-ALDH isoform inhibitor. Mechanistically, 3(h-l) caused increased ROS activity, lipid peroxidation, and toxic aldehyde accumulation, secondary to potent multi-ALDH isoform inhibition leading to increased apoptosis and G2/M cell cycle arrest. Together, the study details the design, synthesis, and evaluation of potent, multi-isoform ALDH inhibitors to treat cancers.
Collapse
Affiliation(s)
- Saketh S Dinavahi
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, United States; Melanoma and Skin Cancer Center, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, United States
| | - Raghavendra Gowda
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, United States; Melanoma and Skin Cancer Center, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, United States
| | - Christopher G Bazewicz
- Melanoma and Skin Cancer Center, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, United States; College of Medicine, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, United States
| | - Madhu Babu Battu
- Laboratory of Molecular Cell Biology, Centre for DNA Fingerprinting and Diagnostics, Uppal, Hyderabad, 500039, India
| | - Jyh Ming Lin
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, United States
| | - Robert J Chitren
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, 08103, United States
| | - Manoj K Pandey
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, 08103, United States
| | - Shantu Amin
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, United States; Penn State Cancer Institute, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, United States
| | - Gavin P Robertson
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, United States; Department of Pathology, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, United States; Department of Dermatology, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, United States; Department of Surgery, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, United States; Melanoma and Skin Cancer Center, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, United States
| | - Krishne Gowda
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, United States; Penn State Cancer Institute, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, United States.
| |
Collapse
|
19
|
ALDH1A2 Is a Candidate Tumor Suppressor Gene in Ovarian Cancer. Cancers (Basel) 2019; 11:cancers11101553. [PMID: 31615043 PMCID: PMC6826427 DOI: 10.3390/cancers11101553] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 09/29/2019] [Accepted: 10/10/2019] [Indexed: 12/22/2022] Open
Abstract
Aldehyde dehydrogenase 1 family member A2 (ALDH1A2) is a rate-limiting enzyme involved in cellular retinoic acid synthesis. However, its functional role in ovarian cancer remains elusive. Here, we found that ALDH1A2 was the most prominently downregulated gene among ALDH family members in ovarian cancer cells, according to complementary DNA microarray data. Low ALDH1A2 expression was associated with unfavorable prognosis and shorter disease-free and overall survival for ovarian cancer patients. Notably, hypermethylation of ALDH1A2 was significantly higher in ovarian cancer cell lines when compared to that in immortalized human ovarian surface epithelial cell lines. ALDH1A2 expression was restored in various ovarian cancer cell lines after treatment with the DNA methylation inhibitor 5-aza-2'-deoxycytidine. Furthermore, silencing DNA methyltransferase 1 (DNMT1) or 3B (DNMT3B) restored ALDH1A2 expression in ovarian cancer cell lines. Functional studies revealed that forced ALDH1A2 expression significantly impaired the proliferation of ovarian cancer cells and their invasive activity. To the best of our knowledge, this is the first study to show that ALDH1A2 expression is regulated by the epigenetic regulation of DNMTs, and subsequently that it might act as a tumor suppressor in ovarian cancer, further suggesting that enhancing ALDH1A2-linked signaling might provide new opportunities for therapeutic intervention in ovarian cancer.
Collapse
|
20
|
Dinavahi SS, Bazewicz CG, Gowda R, Robertson GP. Aldehyde Dehydrogenase Inhibitors for Cancer Therapeutics. Trends Pharmacol Sci 2019; 40:774-789. [DOI: 10.1016/j.tips.2019.08.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Revised: 07/29/2019] [Accepted: 08/08/2019] [Indexed: 02/07/2023]
|
21
|
Helsby NA, Yong M, van Kan M, de Zoysa JR, Burns KE. The importance of both CYP2C19 and CYP2B6 germline variations in cyclophosphamide pharmacokinetics and clinical outcomes. Br J Clin Pharmacol 2019; 85:1925-1934. [PMID: 31218720 DOI: 10.1111/bcp.14031] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 05/28/2019] [Accepted: 05/31/2019] [Indexed: 12/17/2022] Open
Abstract
Cyclophosphamide is an alkylating agent used in the treatment of solid and haematological malignancies and as an immunosuppressive agent. As a prodrug, it is dependent on bioactivation to the active phosphoramide mustard metabolite to elicit its therapeutic effect. This focused review will highlight the evidence for the role of germline pharmacogenetic variation in both plasma pharmacokinetics and clinical outcomes. There is a substantial indication from 13 pharmacokinetic and 17 therapeutic outcome studies, in contexts as diverse as haematological malignancy, breast cancer, systemic lupus erythematosus and myeloablation, that pharmacogenetic variation in both CYP2C19 and CYP2B6 influence the bioactivation of cyclophosphamide. An additional role for pharmacogenetic variation in ALDH1A1 has also been reported. Future studies should comprehensively assess these 3 pharmacogenes and undertake appropriate statistical analysis of gene-gene interactions to confirm these findings and may allow personalised treatment regimens.
Collapse
Affiliation(s)
- N A Helsby
- Molecular Medicine and Pathology, Faculty of Medicine and Health Sciences, University of Auckland, Auckland, New Zealand
| | - M Yong
- Molecular Medicine and Pathology, Faculty of Medicine and Health Sciences, University of Auckland, Auckland, New Zealand
| | - M van Kan
- Molecular Medicine and Pathology, Faculty of Medicine and Health Sciences, University of Auckland, Auckland, New Zealand
| | - J R de Zoysa
- Renal Service, North Shore Hospital, Waitemata District Health Board, Auckland, New Zealand.,Department of Medicine, Faculty of Medicine and Health Sciences, University of Auckland, Auckland, New Zealand
| | - K E Burns
- Molecular Medicine and Pathology, Faculty of Medicine and Health Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
22
|
Ahmed Laskar A, Younus H. Aldehyde toxicity and metabolism: the role of aldehyde dehydrogenases in detoxification, drug resistance and carcinogenesis. Drug Metab Rev 2019; 51:42-64. [DOI: 10.1080/03602532.2018.1555587] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Amaj Ahmed Laskar
- Enzymology Laboratory, Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, India
| | - Hina Younus
- Enzymology Laboratory, Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, India
| |
Collapse
|
23
|
Vassalli G. Aldehyde Dehydrogenases: Not Just Markers, but Functional Regulators of Stem Cells. Stem Cells Int 2019; 2019:3904645. [PMID: 30733805 PMCID: PMC6348814 DOI: 10.1155/2019/3904645] [Citation(s) in RCA: 206] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 11/25/2018] [Indexed: 12/26/2022] Open
Abstract
Aldehyde dehydrogenase (ALDH) is a superfamily of enzymes that detoxify a variety of endogenous and exogenous aldehydes and are required for the biosynthesis of retinoic acid (RA) and other molecular regulators of cellular function. Over the past decade, high ALDH activity has been increasingly used as a selectable marker for normal cell populations enriched in stem and progenitor cells, as well as for cell populations from cancer tissues enriched in tumor-initiating stem-like cells. Mounting evidence suggests that ALDH not only may be used as a marker for stem cells but also may well regulate cellular functions related to self-renewal, expansion, differentiation, and resistance to drugs and radiation. ALDH exerts its functional actions partly through RA biosynthesis, as all-trans RA reverses the functional effects of pharmacological inhibition or genetic suppression of ALDH activity in many cell types in vitro. There is substantial evidence to suggest that the role of ALDH as a stem cell marker comes down to the specific isoform(s) expressed in a particular tissue. Much emphasis has been placed on the ALDH1A1 and ALDH1A3 members of the ALDH1 family of cytosolic enzymes required for RA biosynthesis. ALDH1A1 and ALDH1A3 regulate cellular function in both normal stem cells and tumor-initiating stem-like cells, promoting tumor growth and resistance to drugs and radiation. An improved understanding of the molecular mechanisms by which ALDH regulates cellular function will likely open new avenues in many fields, especially in tissue regeneration and oncology.
Collapse
Affiliation(s)
- Giuseppe Vassalli
- Laboratory of Cellular and Molecular Cardiology, Cardiocentro Ticino, Lugano, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana (USI), Lugano, Switzerland
- Center for Molecular Cardiology, University of Zürich, Zürich, Switzerland
| |
Collapse
|
24
|
Toledo-Guzmán ME, Hernández MI, Gómez-Gallegos ÁA, Ortiz-Sánchez E. ALDH as a Stem Cell Marker in Solid Tumors. Curr Stem Cell Res Ther 2019; 14:375-388. [PMID: 30095061 DOI: 10.2174/1574888x13666180810120012] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Revised: 07/23/2018] [Accepted: 07/24/2018] [Indexed: 02/07/2023]
Abstract
Aldehyde dehydrogenase (ALDH) is an enzyme that participates in important cellular mechanisms as aldehyde detoxification and retinoic acid synthesis; moreover, ALDH activity is involved in drug resistance, a characteristic of cancer stem cells (CSCs). Even though ALDH is found in stem cells, CSCs and progenitor cells, this enzyme has been successfully used to identify and isolate cell populations with CSC properties from several tumor origins. ALDH is allegedly involved in cell differentiation through its product, retinoic acid. However, direct or indirect ALDH inhibition, using specific inhibitors or retinoic acid, has shown a reduction in ALDH activity, along with the loss of stem cell traits, reduction of cell proliferation, invasion, and drug sensitization. For these reasons, ALDH and retinoic acid are promising therapeutic targets. This review summarizes the current evidence for ALDH as a CSCs marker in solid tumors, as well as current knowledge about the functional roles of ALDH in CSCs. We discuss the controversy of ALDH activity to maintain CSC stemness, or conversely, to promote cell differentiation. Finally, we review the advances in using ALDH inhibitors as anti-cancer drugs.
Collapse
Affiliation(s)
- Mariel E Toledo-Guzmán
- Departamento de Bioquimica, Laboratorio de Terapia Genica, Escuela Nacional de Ciencias Biologicas, Posgrado de Biomedicina y Biotecnologia Molecular, Instituto Politecnico Nacional, Mexico City, Mexico
- Subdireccion de Investigacion Basica, Instituto Nacional de Cancerologia, Av San Fernando 22, Colonia Seccion XVI, Tlalpan 14080, Mexico City, Mexico
| | - Miguel Ibañez Hernández
- Departamento de Bioquimica, Laboratorio de Terapia Genica, Escuela Nacional de Ciencias Biologicas, Posgrado de Biomedicina y Biotecnologia Molecular, Instituto Politecnico Nacional, Mexico City, Mexico
| | - Ángel A Gómez-Gallegos
- Subdireccion de Investigacion Basica, Instituto Nacional de Cancerologia, Av San Fernando 22, Colonia Seccion XVI, Tlalpan 14080, Mexico City, Mexico
- Posgrado de Ciencias Biológicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Elizabeth Ortiz-Sánchez
- Subdireccion de Investigacion Basica, Instituto Nacional de Cancerologia, Av San Fernando 22, Colonia Seccion XVI, Tlalpan 14080, Mexico City, Mexico
| |
Collapse
|
25
|
Design, synthesis, and ex vivo evaluation of a selective inhibitor for retinaldehyde dehydrogenase enzymes. Bioorg Med Chem 2018; 26:5766-5779. [PMID: 30409702 DOI: 10.1016/j.bmc.2018.10.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 10/03/2018] [Accepted: 10/12/2018] [Indexed: 11/22/2022]
Abstract
The retinaldehyde dehydrogenase (RALDH) enzymes, RALDH1, RALDH2, and RALDH3, catalyze the irreversible oxidation of retinaldehyde to all-trans-retinoic acid (ATRA). Despite the importance of the RALDH enzymes in embryonic development, postnatal growth and differentiation, and in several disease states, there are no commercially available inhibitors that specifically target these isozymes. We report here the development and characterization of a small molecule inhibitor dichloro-all-trans-retinone (DAR) (Summers et al., 2017) that is an irreversible inhibitor of RALDH1, 2, and 3 that effectively inhibits RALDH1, 2, and 3 in the nanomolar range but has no inhibitory activity against mitochondrial ALDH2. These results provide support for the development of DAR as a specific ATRA synthesis inhibitor for a variety of experimental and clinical applications.
Collapse
|
26
|
Bazewicz CG, Dinavahi SS, Schell TD, Robertson GP. Aldehyde dehydrogenase in regulatory T-cell development, immunity and cancer. Immunology 2018; 156:47-55. [PMID: 30387499 DOI: 10.1111/imm.13016] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 10/10/2018] [Accepted: 10/26/2018] [Indexed: 12/14/2022] Open
Abstract
The role of aldehyde dehydrogenase (ALDH) in carcinogenesis and resistance to cancer therapies is well known. Mounting evidence also suggests a potentially important role for ALDH in the induction and function of regulatory T (Treg) cells. Treg cells are important cells of the immune system involved in promoting immune tolerance and preventing aberrant immune responses to beneficial or non-harmful antigens. However, Treg cells also impair tumor immunity, leading to the progression of various carcinomas. ALDH expression and the subsequent production of retinoic acid by numerous cells, including dendritic cells, macrophages, eosinophils and epithelial cells, seems important in Treg induction and function in multiple organ systems. This is particularly evident in the gastrointestinal tract, pulmonary tract and skin, which are exposed to a myriad of environmental antigens and represent interfaces between the human body and the outside world. Expression of ALDH in Treg cells themselves may also be involved in the proliferation of these cells and resistance to certain cytotoxic therapies. Hence, inhibition of ALDH expression may be useful to treat cancer. Besides the direct effect of ALDH inhibition on carcinogenesis and resistance to cancer therapies, inhibition of ALDH could potentially augment the immune response to tumor antigens by inhibiting Treg induction, function and ability to promote immune tolerance to tumor cells in multiple cancer types.
Collapse
Affiliation(s)
- Christopher G Bazewicz
- College of Medicine, The Pennsylvania State University Medical Center, Hershey, PA, USA.,The Penn State Melanoma and Skin Cancer Center, The Pennsylvania State University Medical Center, Hershey, PA, USA
| | - Saketh S Dinavahi
- The Penn State Melanoma and Skin Cancer Center, The Pennsylvania State University Medical Center, Hershey, PA, USA.,Department of Pharmacology, The Pennsylvania State University Medical Center, Hershey, PA, USA
| | - Todd D Schell
- Department of Microbiology and Immunology, The Pennsylvania State University Medical Center, Hershey, PA, USA
| | - Gavin P Robertson
- The Penn State Melanoma and Skin Cancer Center, The Pennsylvania State University Medical Center, Hershey, PA, USA.,Department of Pharmacology, The Pennsylvania State University Medical Center, Hershey, PA, USA.,Department of Pathology, The Pennsylvania State University Medical Center, Hershey, PA, USA.,Department of Dermatology, The Pennsylvania State University Medical Center, Hershey, PA, USA.,Department of Surgery, The Pennsylvania State University Medical Center, Hershey, PA, USA.,Penn State Melanoma Therapeutics Program, The Pennsylvania State University Medical Center, Hershey, PA, USA.,Foreman Foundation for Melanoma Research, The Pennsylvania State University Medical Center, Hershey, PA, USA
| |
Collapse
|
27
|
Hirashima K, Yue F, Kobayashi M, Uchida Y, Nakamura S, Tomotsune D, Matsumoto K, Takizawa-Shirasawa S, Yokoyama T, Kanno H, Sasaki K. Cell biological profiling of reprogrammed cancer stem cell-like colon cancer cells maintained in culture. Cell Tissue Res 2018; 375:697-707. [PMID: 30284085 DOI: 10.1007/s00441-018-2933-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Accepted: 09/18/2018] [Indexed: 12/11/2022]
Abstract
Cancer stem cells (CSCs) are specific targets for therapeutic applications, but the rarity of CSCs within tumors makes the isolation of CSCs difficult. To overcome these problems, we generated CSCs in vitro using established reprogramming techniques. We transduced four previously established reprogramming factors, Oct3/4, Sox2, Klf4, and L-myc, into the colon cancer cell lines LoVo and OUMS-23, and investigated the biological characteristics of these lines. Tra-1-60+ cells were obtained from reprogrammed induced pluripotent stem (iPS) cell-like colonies and showed CSC properties, including colony formation, maintenance of colonies by repeated passages, and feeder cell dependency, as well as increased expressions of CSC markers such as CD133 and ALDH1. The CSC-like cells showed increased chemoresistance to 5-fluorouracil and elevated tumorigenicity upon transplantation into kidneys of immune-deficient mice. These tumors shifted to a poorly differentiated stage with many atypical cells, cytoplasmic mucin, and focal papillary components, with demonstrated dedifferentiation. The principal component analysis from DNA microarrays showed that though both cell lines moved to iPS cells after reprogramming, they were not completely identical to iPS cells. Significantly elevated gene expression of Decorin and CD90 was observed in CSC-like cells. Together, these results show that reprogramming of cancer cells produced not pluripotent stem cells but CSC-like cells, and these findings will provide biological information about genuine CSCs and help establish new CSC-targeted therapies.
Collapse
Affiliation(s)
- Kanji Hirashima
- Department of Anatomy and Organ Technology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, 390-8621, Japan.
| | - Fengming Yue
- Department of Anatomy and Organ Technology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, 390-8621, Japan
| | - Mikiko Kobayashi
- Department of Pathology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, 390-8621, Japan
| | - Yuriko Uchida
- Department of Anatomy and Organ Technology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, 390-8621, Japan
| | - Shunsuke Nakamura
- Department of Anatomy and Organ Technology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, 390-8621, Japan
| | - Daihachiro Tomotsune
- Department of Anatomy and Organ Technology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, 390-8621, Japan.,Department of Biotechnology and Biomedical Engineering, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano, 390-8621, Japan
| | - Ken Matsumoto
- Nissui Pharmaceutical Co., Ltd., 1075-2 Hokunanmoro, Yuki, Ibaraki, 307-0036, Japan
| | | | - Tadayuki Yokoyama
- Bourbon Corporation, 4-2-14 Matsunami, Kashiwazaki, Niigata, 945-8611, Japan
| | - Hiroyuki Kanno
- Department of Pathology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, 390-8621, Japan
| | - Katsunori Sasaki
- Department of Anatomy and Organ Technology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, 390-8621, Japan.,Department of Biotechnology and Biomedical Engineering, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano, 390-8621, Japan
| |
Collapse
|
28
|
Counihan JL, Wiggenhorn AL, Anderson KE, Nomura DK. Chemoproteomics-Enabled Covalent Ligand Screening Reveals ALDH3A1 as a Lung Cancer Therapy Target. ACS Chem Biol 2018; 13:1970-1977. [PMID: 30004670 DOI: 10.1021/acschembio.8b00381] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Chemical genetics is a powerful approach for identifying therapeutically active small molecules, but identifying the mechanisms of action underlying hit compounds remains challenging. Chemoproteomic platforms have arisen to tackle this challenge and enable rapid mechanistic deconvolution of small-molecule screening hits. Here, we have screened a cysteine-reactive covalent ligand library to identify hit compounds that impair cell survival and proliferation in nonsmall cell lung carcinoma cells, but not in primary human bronchial epithelial cells. Through this screen, we identified a covalent ligand hit, DKM 3-42, which impaired both in situ and in vivo lung cancer pathogenicity. We used activity-based protein profiling to discover that the primary target of DKM 3-42 was the catalytic cysteine in aldehyde dehydrogenase 3A1 (ALDH3A1). We performed further chemoproteomics-enabled covalent ligand screening directly against ALDH3A1, and identified a more potent and selective lead covalent ligand, EN40, which inhibits ALDH3A1 activity and impairs lung cancer pathogenicity. We show here that ALDH3A1 represents a potentially novel therapeutic target for lung cancers that express ALDH3A1 and put forth two selective ALDH3A1 inhibitors. Overall, we show the utility of combining chemical genetics screening of covalent ligand libraries with chemoproteomic approaches to rapidly identify anticancer leads and targets.
Collapse
|
29
|
Transcriptome analysis and prognosis of ALDH isoforms in human cancer. Sci Rep 2018; 8:2713. [PMID: 29426835 PMCID: PMC5807355 DOI: 10.1038/s41598-018-21123-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 01/30/2018] [Indexed: 12/12/2022] Open
Abstract
Overexpression of ALDH is associated with cancer stem-like features and poor cancer prognosis. High ALDH activity has been observed in cancer stem-like cells. There are a total of 19 human ALDH isoforms, all of which are associated with reducing oxidative stress and protecting cells from damage. However, it is unknown whether all ALDHs are associated with poor cancer prognosis and which ones play a significant role in cancer progression. In this study, we used RNA sequencing data from The Cancer Genome Atlas (TCGA) to evaluate the differential expression of 19 ALDH isoforms in 5 common human cancers. The 19 ALDH genes were analyzed with an integrating meta-analysis of cancer prognosis. Genotyping and next-generation RNA sequencing for 30 pairwise samples of head and neck squamous cell carcinoma were performed and compared with the TCGA cohort. The analysis showed that each ALDH isoform had a specific differential expression pattern, most of which were related to prognosis in human cancer. A lower expression of ALDH2 in the tumor was observed, which was independent from the ALDH2 rs671 SNP variant and the expression of other mitochondria-associated protein coding genes. This study provides new insight into the association between ALDH expression and cancer prognosis.
Collapse
|
30
|
Pass HI, Lavilla C, Canino C, Goparaju C, Preiss J, Noreen S, Blandino G, Cioce M. Inhibition of the colony-stimulating-factor-1 receptor affects the resistance of lung cancer cells to cisplatin. Oncotarget 2018; 7:56408-56421. [PMID: 27486763 PMCID: PMC5302923 DOI: 10.18632/oncotarget.10895] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 06/30/2016] [Indexed: 02/07/2023] Open
Abstract
In the present work we show that multiple lung cancer cell lines contain cisplatin resistant cell subpopulations expressing the Colony-Stimulating-Factor-Receptor-1 (CSF-1R) and surviving chemotherapy-induced stress. By exploiting siRNA-mediated knock down in vitro and the use of an investigational CSF-1R TKI (JNJ-40346527) in vitro and in vivo, we show that expression and function of the receptor are required for the clonogenicity and chemoresistance of the cell lines. Thus, inhibition of the kinase activity of the receptor reduced the levels of EMT-associated genes, stem cell markers and chemoresistance genes. Additionally, the number of high aldehyde dehydrogenase (ALDH) expressing cells was reduced, consequent to the lack of cisplatin-induced increase of ALDH isoforms. This affected the collective chemoresistance of the treated cultures. Treatment of tumor bearing mice with JNJ-40346527, at pharmacologically relevant doses, produced strong chemo-sensitizing effects in vivo. These anticancer effects correlated with a reduced number of CSF-1Rpos cells, in tumors excised from the treated mice. Depletion of the CD45pos cells within the treated tumors did not, apparently, play a major role in mediating the therapeutic response to the TKI. Thus, lung cancer cells express a functional CSF-1 and CSF-1R duo which mediates pro-tumorigenic effects in vivo and in vitro and can be targeted in a therapeutically relevant way. These observations complement the already known role for the CSF-1R at mediating the pro-tumorigenic properties of tumor-infiltrating immune components.
Collapse
Affiliation(s)
- Harvey I Pass
- Division of Thoracic Surgery, Department of Cardiothoracic Surgery, Langone Medical Center, New York University, New York, USA
| | - Carmencita Lavilla
- New York University Langone Medical Center, New York University, New York, USA
| | - Claudia Canino
- Division of Thoracic Surgery, Department of Cardiothoracic Surgery, Langone Medical Center, New York University, New York, USA.,University Campus Biomedico, Rome, Italy
| | - Chandra Goparaju
- Division of Thoracic Surgery, Department of Cardiothoracic Surgery, Langone Medical Center, New York University, New York, USA
| | - Jordan Preiss
- Division of Thoracic Surgery, Department of Cardiothoracic Surgery, Langone Medical Center, New York University, New York, USA
| | - Samrah Noreen
- New York University Langone Medical Center, New York University, New York, USA
| | - Giovanni Blandino
- Translational Oncogenomics Unit, Italian National Cancer Institute 'Regina Elena', Rome, Italy.,Department of Oncology, Juravinski Cancer Center-McMaster University, Hamilton, Ontario, Canada
| | - Mario Cioce
- Division of Thoracic Surgery, Department of Cardiothoracic Surgery, Langone Medical Center, New York University, New York, USA.,Translational Oncogenomics Unit, Italian National Cancer Institute 'Regina Elena', Rome, Italy
| |
Collapse
|
31
|
Chlapek P, Slavikova V, Mazanek P, Sterba J, Veselska R. Why Differentiation Therapy Sometimes Fails: Molecular Mechanisms of Resistance to Retinoids. Int J Mol Sci 2018; 19:ijms19010132. [PMID: 29301374 PMCID: PMC5796081 DOI: 10.3390/ijms19010132] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 12/28/2017] [Accepted: 12/29/2017] [Indexed: 12/12/2022] Open
Abstract
Retinoids represent a popular group of differentiation inducers that are successfully used in oncology for treatment of acute promyelocytic leukemia in adults and of neuroblastoma in children. The therapeutic potential of retinoids is based on their key role in the regulation of cell differentiation, growth, and apoptosis, which provides a basis for their use both in cancer therapy and chemoprevention. Nevertheless, patients treated with retinoids often exhibit or develop resistance to this therapy. Although resistance to retinoids is commonly categorized as either acquired or intrinsic, resistance as a single phenotypic feature is usually based on the same mechanisms that are closely related or combined in both of these types. In this review, we summarize the most common changes in retinoid metabolism and action that may affect the sensitivity of a tumor cell to treatment with retinoids. The availability of retinoids can be regulated by alterations in retinol metabolism or in retinoid intracellular transport, by degradation of retinoids or by their efflux from the cell. Retinoid effects on gene expression can be regulated via retinoid receptors or via other molecules in the transcriptional complex. Finally, the role of small-molecular-weight inhibitors of altered cell signaling pathways in overcoming the resistance to retinoids is also suggested.
Collapse
Affiliation(s)
- Petr Chlapek
- Laboratory of Tumor Biology, Department of Experimental Biology, Faculty of Science, Masaryk University, 61137 Brno, Czech Republic.
- International Clinical Research Center, St. Anne's University Hospital, 65691 Brno, Czech Republic.
| | - Viera Slavikova
- Laboratory of Tumor Biology, Department of Experimental Biology, Faculty of Science, Masaryk University, 61137 Brno, Czech Republic.
- International Clinical Research Center, St. Anne's University Hospital, 65691 Brno, Czech Republic.
| | - Pavel Mazanek
- Department of Pediatric Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic.
| | - Jaroslav Sterba
- International Clinical Research Center, St. Anne's University Hospital, 65691 Brno, Czech Republic.
- Department of Pediatric Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic.
| | - Renata Veselska
- Laboratory of Tumor Biology, Department of Experimental Biology, Faculty of Science, Masaryk University, 61137 Brno, Czech Republic.
- International Clinical Research Center, St. Anne's University Hospital, 65691 Brno, Czech Republic.
- Department of Pediatric Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic.
| |
Collapse
|
32
|
Prieto-Vila M, Takahashi RU, Usuba W, Kohama I, Ochiya T. Drug Resistance Driven by Cancer Stem Cells and Their Niche. Int J Mol Sci 2017; 18:ijms18122574. [PMID: 29194401 PMCID: PMC5751177 DOI: 10.3390/ijms18122574] [Citation(s) in RCA: 351] [Impact Index Per Article: 43.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 11/22/2017] [Accepted: 11/24/2017] [Indexed: 12/11/2022] Open
Abstract
Drug resistance represents one of the greatest challenges in cancer treatment. Cancer stem cells (CSCs), a subset of cells within the tumor with the potential for self-renewal, differentiation and tumorigenicity, are thought to be the major cause of cancer therapy failure due to their considerable chemo- and radioresistance, resulting in tumor recurrence and eventually metastasis. CSCs are situated in a specialized microenvironment termed the niche, mainly composed of fibroblasts and endothelial, mesenchymal and immune cells, which also play pivotal roles in drug resistance. These neighboring cells promote the molecular signaling pathways required for CSC maintenance and survival and also trigger endogenous drug resistance in CSCs. In addition, tumor niche components such as the extracellular matrix also physically shelter CSCs from therapeutic agents. Interestingly, CSCs contribute directly to the niche in a bilateral feedback loop manner. Here, we review the recent advances in the study of CSCs, the niche and especially their collective contribution to resistance, since increasingly studies suggest that this interaction should be considered as a target for therapeutic strategies.
Collapse
Affiliation(s)
- Marta Prieto-Vila
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, Tokyo 104-0045, Japan.
| | - Ryou-U Takahashi
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, Tokyo 104-0045, Japan.
| | - Wataru Usuba
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, Tokyo 104-0045, Japan.
| | - Isaku Kohama
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, Tokyo 104-0045, Japan.
| | - Takahiro Ochiya
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, Tokyo 104-0045, Japan.
| |
Collapse
|
33
|
Croker AK, Rodriguez-Torres M, Xia Y, Pardhan S, Leong HS, Lewis JD, Allan AL. Differential Functional Roles of ALDH1A1 and ALDH1A3 in Mediating Metastatic Behavior and Therapy Resistance of Human Breast Cancer Cells. Int J Mol Sci 2017; 18:ijms18102039. [PMID: 28937653 PMCID: PMC5666721 DOI: 10.3390/ijms18102039] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 09/18/2017] [Accepted: 09/18/2017] [Indexed: 12/31/2022] Open
Abstract
Previous studies indicate that breast cancer cells with high aldehyde dehydrogenase (ALDH) activity and CD44 expression (ALDHhiCD44+) contribute to metastasis and therapy resistance, and that ALDH1 correlates with poor outcome in breast cancer patients. The current study hypothesized that ALDH1 functionally contributes to breast cancer metastatic behavior and therapy resistance. Expression of ALDH1A1 or ALDH1A3 was knocked down in MDA-MB-468 and SUM159 human breast cancer cells using siRNA. Resulting impacts on ALDH activity (Aldefluor® assay); metastatic behavior and therapy response in vitro (proliferation/adhesion/migration/colony formation/chemotherapy and radiation) and extravasation/metastasis in vivo (chick choroiallantoic membrane assay) was assessed. Knockdown of ALDH1A3 but not ALDH1A1 in breast cancer cells decreased ALDH activity, and knockdown of ALDH1A1 reduced breast cancer cell metastatic behavior and therapy resistance relative to control (p < 0.05). In contrast, knockdown of ALDH1A3 did not alter proliferation, extravasation, or therapy resistance, but increased adhesion/migration and decreased colony formation/metastasis relative to control (p < 0.05). This is the first study to systematically examine the function of ALDH1 isozymes in individual breast cancer cell behaviors that contribute to metastasis. Our novel results indicate that ALDH1 mediates breast cancer metastatic behavior and therapy resistance, and that different enzyme isoforms within the ALDH1 family differentially impact these cell behaviors.
Collapse
Affiliation(s)
- Alysha K Croker
- London Regional Cancer Program, London Health Sciences Centre, 790 Commissioners Road East, London, ON N6A 4L6, Canada.
- Department of Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, Western University, London, ON N6A 5C1, Canada.
| | - Mauricio Rodriguez-Torres
- London Regional Cancer Program, London Health Sciences Centre, 790 Commissioners Road East, London, ON N6A 4L6, Canada.
- Department of Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, Western University, London, ON N6A 5C1, Canada.
| | - Ying Xia
- London Regional Cancer Program, London Health Sciences Centre, 790 Commissioners Road East, London, ON N6A 4L6, Canada.
| | - Siddika Pardhan
- Department of Surgery, Schulich School of Medicine & Dentistry, Western University, London, ON N6A 5C1, Canada.
| | - Hon Sing Leong
- Department of Surgery, Schulich School of Medicine & Dentistry, Western University, London, ON N6A 5C1, Canada.
| | - John D Lewis
- Department of Oncology, University of Alberta, 5-142C Katz Group Building, 114th St. and 87th Ave. S., Edmonton, AB T6G 2E1, Canada.
| | - Alison L Allan
- London Regional Cancer Program, London Health Sciences Centre, 790 Commissioners Road East, London, ON N6A 4L6, Canada.
- Department of Oncology and Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, Western University, London, ON N6A 5C1, Canada.
- Cancer Research Laboratory Program, Lawson Health Research Institute, 750 Base Line Road, Suite 300, London, ON N6C 2R5, Canada.
| |
Collapse
|
34
|
Yang C, Wang X, Liao X, Han C, Yu T, Qin W, Zhu G, Su H, Yu L, Liu X, Lu S, Chen Z, Liu Z, Huang K, Liu Z, Liang Y, Huang J, Xiao K, Peng M, Winkle CA, O'Brien SJ, Peng T. Aldehyde dehydrogenase 1 (ALDH1) isoform expression and potential clinical implications in hepatocellular carcinoma. PLoS One 2017; 12:e0182208. [PMID: 28792511 PMCID: PMC5549701 DOI: 10.1371/journal.pone.0182208] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 07/16/2017] [Indexed: 01/01/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most prevalent and life-threatening malignancies worldwide. There are few diagnostic and prognostic biomarkers and druggable targets for HCC. Aldehyde dehydrogenase 1 (ALDH1) is a marker of stem cells in a variety of cancers, but the mRNA levels and prognostic value of ALDH1 isoforms in HCC patients remain unknown. In the present study, gene ontology annotation of the ALDH1 family was performed using the Database for Annotation, Visualization and Integrated Discovery (DAVID), and the gene pathway analsis was performed using GeneMANIA software. The initial prognostic value of ALDH1 expression in 360 HCC patients was assessed using the OncoLnc database. The expression levels of ALDH1 isoforms in normal liver tissues and clinical specimens of cancer vs. normal control datasets were determined using the GTEx and Oncomine databases, respectively. We then analyzed the prognostic value of ALDH1 expression in 212 hepatitis B virus (HBV)-related HCC patients using the GEO database. We found that the ALDH1 isoform showed high aldehyde dehydrogenase activity. The ALDH1A1, ALDH1B1, and ALDH1L1 genes encoded for the ALDH1 enzyme. High ALDH1B1 expression had protective qualities in HCC patients. Moreover, HBV-related HCC patients who showed high ALDH1L1 gene expression had a better clinical outcomes. In addition, high ALDH1A1 expression was associated with a 57-month recurrence-free survival in HBV-related HCC patients. High ALDH1B1 expression was protective for HCCs with multiple nodules and high serum alpha-fetoprotein (AFP) level. Furthermore, high serum AFP levels contributed to lower ALDH1L1. ALDH1A1, ALDH1B1, and ALDH1L1, all of which were considered promising diagnostic and prognostic markers as well as potential drug targets.
Collapse
Affiliation(s)
- Cheng–kun Yang
- Department of Hepatobiliary Surgery, The first Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Province, China
| | - Xiang–kun Wang
- Department of Hepatobiliary Surgery, The first Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Province, China
| | - Xi–wen Liao
- Department of Hepatobiliary Surgery, The first Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Province, China
| | - Chuang–ye Han
- Department of Hepatobiliary Surgery, The first Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Province, China
| | - Ting–dong Yu
- Department of Hepatobiliary Surgery, The first Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Province, China
| | - Wei Qin
- Department of Hepatobiliary Surgery, The first Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Province, China
| | - Guang–zhi Zhu
- Department of Hepatobiliary Surgery, The first Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Province, China
| | - Hao Su
- Department of Hepatobiliary Surgery, The first Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Province, China
| | - Long Yu
- Department of Hepatobiliary and Pancreatic Surgery, The first Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Xiao–guang Liu
- Department of Hepatobiliary Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong Province, China
| | - Si–cong Lu
- Department of Hepatobiliary Surgery, The first Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Province, China
| | - Zhi–wei Chen
- Department of Hepatobiliary Surgery, The first Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Province, China
| | - Zhen Liu
- Department of Hepatobiliary Surgery, The first Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Province, China
| | - Ke–tuan Huang
- Department of Hepatobiliary Surgery, The first Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Province, China
| | - Zheng–tao Liu
- Department of Hepatobiliary Surgery, The first Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Province, China
| | - Yu Liang
- Department of Hepatobiliary Surgery, The first Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Province, China
| | - Jian–lu Huang
- Department of Hepatobiliary Surgery, Third Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Province, China
| | - Kai–yin Xiao
- Department of Hepatobiliary Surgery, The first Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Province, China
| | - Min–hao Peng
- Department of Hepatobiliary Surgery, The first Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Province, China
| | - Cheryl Ann Winkle
- Laboratory of Genomic Diversity, National Cancer Institute, NIH, Frederick, MD, United States of America
| | - Stephen J. O'Brien
- Laboratory of Genomic Diversity, National Cancer Institute, NIH, Frederick, MD, United States of America
| | - Tao Peng
- Department of Hepatobiliary Surgery, The first Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Province, China
| |
Collapse
|
35
|
Butler SJ, Richardson L, Farias N, Morrison J, Coomber BL. Characterization of cancer stem cell drug resistance in the human colorectal cancer cell lines HCT116 and SW480. Biochem Biophys Res Commun 2017; 490:29-35. [DOI: 10.1016/j.bbrc.2017.05.176] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 05/29/2017] [Indexed: 12/28/2022]
|
36
|
He X, Deng Y, Yue W. Investigating critical genes and gene interaction networks that mediate cyclophosphamide sensitivity in chronic myelogenous leukemia. Mol Med Rep 2017; 16:523-532. [PMID: 28560425 PMCID: PMC5482156 DOI: 10.3892/mmr.2017.6636] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 02/15/2017] [Indexed: 01/06/2023] Open
Abstract
Drug resistance is an obstacle in the treatment of chronic myelogenous leukemia (CML), and is a common reason for treatment failure or disease progression. However, the underlying mechanisms of cyclophosphamide resistance remain poorly defined. In the present study, microarray data concerning cyclophosphamide‑sensitive and ‑resistant chronic myelogenous leukemia cell lines were analyzed. A total of 258 differentially‑expressed genes (DEGs) were identified between these two groups, from which 139 DEGs were upregulated and 119 were downregulated. Several candidate genes that were associated with cyclophosphamide resistance were also identified. These DEGs were subsequently classified using Gene Ontology and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment pathway analysis. A total of 487 biological processes and 17 KEGG pathways were revealed to be enriched. Furthermore, an interaction network was established to identify the core genes that regulated cyclophosphamide resistance. Signal transducer and activator of transcription 5A (STAT5A), FYN proto‑oncogene, Src family tyrosine kinase and spleen associated tyrosine kinase were revealed to be the hub genes in multiple enriched biological processes and signaling pathways, indicating that these were involved in mediating cyclophosphamide sensitivity in CML cells. The expression levels of 5 DEGs were also confirmed in two human CML cell lines (K‑562 and KU812) by reverse transcription‑quantitative polymerase chain reaction. Furthermore, selective knockdown of STAT5A and S100 calcium binding protein A4 (S100A4) recovered cyclophosphamide sensitivity in K‑562 cells, suggesting their involvement in drug resistance. The present study identified several potential genes and pathways contributing to cyclophosphamide resistance, and confirmed the involvement of STAT5A and S100A4 in drug resistance. These results enable improved understanding of the mechanisms underlying drug resistance in CML cells.
Collapse
MESH Headings
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Cell Line, Tumor
- Computational Biology/methods
- Cyclophosphamide/pharmacology
- Cyclophosphamide/therapeutic use
- Databases, Nucleic Acid
- Drug Resistance, Neoplasm/genetics
- Epistasis, Genetic
- Gene Expression Profiling
- Gene Expression Regulation, Leukemic/drug effects
- Gene Ontology
- Gene Regulatory Networks
- Humans
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Reproducibility of Results
- S100 Calcium-Binding Protein A4/genetics
- S100 Calcium-Binding Protein A4/metabolism
- STAT5 Transcription Factor/genetics
- STAT5 Transcription Factor/metabolism
- Transcriptome
Collapse
Affiliation(s)
- Xiao He
- Blood Transfusion Department, The First People's Hospital of Yancheng City, Yancheng, Jiangsu 224006, P.R. China
| | - Yuying Deng
- Blood Transfusion Department, The First People's Hospital of Yancheng City, Yancheng, Jiangsu 224006, P.R. China
| | - Wei Yue
- Blood Transfusion Department, The First People's Hospital of Yancheng City, Yancheng, Jiangsu 224006, P.R. China
| |
Collapse
|
37
|
Kim J, Shin JH, Chen CH, Cruz L, Farnebo L, Yang J, Borges P, Kang G, Mochly-Rosen D, Sunwoo JB. Targeting aldehyde dehydrogenase activity in head and neck squamous cell carcinoma with a novel small molecule inhibitor. Oncotarget 2017; 8:52345-52356. [PMID: 28881734 PMCID: PMC5581033 DOI: 10.18632/oncotarget.17017] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 03/30/2017] [Indexed: 01/02/2023] Open
Abstract
Chemoresistant cancer cells express high levels of aldehyde dehydrogenases (ALDHs), particularly in head and neck squamous cell carcinoma (HNSCC). The ALDH family of enzymes detoxify both exogenous and endogenous aldehydes. Since many chemotherapeutic agents, such as cisplatin, result in the generation of cytotoxic aldehydes and oxidative stress, we hypothesized that cells expressing high levels of ALDH may be more chemoresistant due to their increased detoxifying capacity and that inhibitors of ALDHs may sensitize them to these drugs. Here, we show that overall ALDH activity is increased with cisplatin treatment of HNSCC and that ALDH3A1 protein expression is particularly enriched in cells treated with cisplatin. Activation of ALDH3A1 by a small molecule activator (Alda-89) increased survival of HNSCC cells treated with cisplatin. Conversely, treatment with a novel small molecule ALDH inhibitor (Aldi-6) resulted in a marked decrease in cell viability, and the combination of Aldi-6 and cisplatin resulted in a more pronounced reduction of cell viability and a greater reduction in tumor burden in vivo than what was observed with cisplatin alone. These data indicate that ALDH3A1 contributes to cisplatin resistance in HNSCC and that the targeting of ALDH, specifically, ALDH3A1, appears to be a promising strategy in this disease.
Collapse
Affiliation(s)
- Jeewon Kim
- Stanford Cancer Institute, School of Medicine, Stanford University, Stanford, CA, 94305, USA
| | - June Ho Shin
- Stanford Cancer Institute, School of Medicine, Stanford University, Stanford, CA, 94305, USA.,Division of Head and Neck Surgery, Department of Otolaryngology, Stanford University, Stanford, CA, 94305, USA
| | - Che-Hong Chen
- Department of Chemical and Systems Biology, Stanford University, School of Medicine, Stanford, CA, 94305, USA
| | - Leslie Cruz
- Department of Chemical and Systems Biology, Stanford University, School of Medicine, Stanford, CA, 94305, USA
| | - Lovisa Farnebo
- Division of Head and Neck Surgery, Department of Otolaryngology, Stanford University, Stanford, CA, 94305, USA.,Division of Otorhinolaryngology, Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, SE-58185, Linköping, Sweden
| | - Jieying Yang
- Stanford Cancer Institute, School of Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Paula Borges
- Division of Head and Neck Surgery, Department of Otolaryngology, Stanford University, Stanford, CA, 94305, USA
| | - Gugene Kang
- Department of Developmental Biology, Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Daria Mochly-Rosen
- Department of Chemical and Systems Biology, Stanford University, School of Medicine, Stanford, CA, 94305, USA
| | - John B Sunwoo
- Stanford Cancer Institute, School of Medicine, Stanford University, Stanford, CA, 94305, USA.,Division of Head and Neck Surgery, Department of Otolaryngology, Stanford University, Stanford, CA, 94305, USA
| |
Collapse
|
38
|
Buchman CD, Hurley TD. Inhibition of the Aldehyde Dehydrogenase 1/2 Family by Psoralen and Coumarin Derivatives. J Med Chem 2017; 60:2439-2455. [PMID: 28219011 PMCID: PMC5765548 DOI: 10.1021/acs.jmedchem.6b01825] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Aldehyde dehydrogenase 2 (ALDH2), one of 19 ALDH superfamily members, catalyzes the NAD+-dependent oxidation of aldehydes to their respective carboxylic acids. In this study, we further characterized the inhibition of four psoralen and coumarin derivatives toward ALDH2 and compared them to the ALDH2 inhibitor daidzin for selectivity against five ALDH1/2 isoenzymes. Compound 2 (Ki = 19 nM) binds within the aldehyde-binding site of the free enzyme species of ALDH2. Thirty-three structural analogs were examined to develop a stronger SAR profile. Seven compounds maintained or improved upon the selectivity toward one of the five ALDH1/2 isoenzymes, including compound 36, a selective inhibitor for ALDH2 (Ki = 2.4 μM), and compound 32, which was 10-fold selective for ALDH1A1 (Ki = 1.2 μM) versus ALDH1A2. Further medicinal chemistry on the compounds' basic scaffold could enhance the potency and selectivity for ALDH1A1 or ALDH2 and generate chemical probes to examine the unique and overlapping functions of the ALDH1/2 isoenzymes.
Collapse
|
39
|
In vitro activity and stability of pure human salivary aldehyde dehydrogenase. Int J Biol Macromol 2017; 96:798-806. [DOI: 10.1016/j.ijbiomac.2016.12.084] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 12/28/2016] [Accepted: 12/31/2016] [Indexed: 11/22/2022]
|
40
|
Reduced aldehyde dehydrogenase expression in preeclamptic decidual mesenchymal stem/stromal cells is restored by aldehyde dehydrogenase agonists. Sci Rep 2017; 7:42397. [PMID: 28205523 PMCID: PMC5304324 DOI: 10.1038/srep42397] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 01/09/2017] [Indexed: 01/24/2023] Open
Abstract
High resistance to oxidative stress is a common feature of mesenchymal stem/stromal cells (MSC) and is associated with higher cell survival and ability to respond to oxidative damage. Aldehyde dehydrogenase (ALDH) activity is a candidate “universal” marker for stem cells. ALDH expression was significantly lower in decidual MSC (DMSC) isolated from preeclamptic (PE) patients. ALDH gene knockdown by siRNA transfection was performed to create a cell culture model of the reduced ALDH expression detected in PE-DMSC. We showed that ALDH activity in DMSC is associated with resistance to hydrogen peroxide (H2O2)-induced toxicity. Our data provide evidence that ALDH expression in DMSC is required for cellular resistance to oxidative stress. Furthermore, candidate ALDH activators were screened and two of the compounds were effective in upregulating ALDH expression. This study provides a proof-of-principle that the restoration of ALDH activity in diseased MSC is a rational basis for a therapeutic strategy to improve MSC resistance to cytotoxic damage.
Collapse
|
41
|
Yasgar A, Titus SA, Wang Y, Danchik C, Yang SM, Vasiliou V, Jadhav A, Maloney DJ, Simeonov A, Martinez NJ. A High-Content Assay Enables the Automated Screening and Identification of Small Molecules with Specific ALDH1A1-Inhibitory Activity. PLoS One 2017; 12:e0170937. [PMID: 28129349 PMCID: PMC5271370 DOI: 10.1371/journal.pone.0170937] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Accepted: 01/12/2017] [Indexed: 12/20/2022] Open
Abstract
Aldehyde dehydrogenase enzymes (ALDHs) have a broad spectrum of biological activities through the oxidation of both endogenous and exogenous aldehydes. Increased expression of ALDH1A1 has been identified in a wide-range of human cancer stem cells and is associated with cancer relapse and poor prognosis, raising the potential of ALDH1A1 as a therapeutic target. To facilitate quantitative high-throughput screening (qHTS) campaigns for the discovery, characterization and structure-activity-relationship (SAR) studies of small molecule ALDH1A1 inhibitors with cellular activity, we show herein the miniaturization to 1536-well format and automation of a high-content cell-based ALDEFLUOR assay. We demonstrate the utility of this assay by generating dose-response curves on a comprehensive set of prior art inhibitors as well as hundreds of ALDH1A1 inhibitors synthesized in house. Finally, we established a screening paradigm using a pair of cell lines with low and high ALDH1A1 expression, respectively, to uncover novel cell-active ALDH1A1-specific inhibitors from a collection of over 1,000 small molecules.
Collapse
Affiliation(s)
- Adam Yasgar
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, United States of America
| | - Steven A. Titus
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, United States of America
| | - Yuhong Wang
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, United States of America
| | - Carina Danchik
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, United States of America
| | - Shyh-Ming Yang
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, United States of America
| | - Vasilis Vasiliou
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, CT, United States of America
| | - Ajit Jadhav
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, United States of America
| | - David J. Maloney
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, United States of America
| | - Anton Simeonov
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, United States of America
- * E-mail: (AS); (NJM)
| | - Natalia J. Martinez
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, United States of America
- * E-mail: (AS); (NJM)
| |
Collapse
|
42
|
Wei Y, Wu S, Xu W, Liang Y, Li Y, Zhao W, Wu J. Depleted aldehyde dehydrogenase 1A1 (ALDH1A1) reverses cisplatin resistance of human lung adenocarcinoma cell A549/DDP. Thorac Cancer 2017; 8:26-32. [PMID: 27813328 PMCID: PMC5217899 DOI: 10.1111/1759-7714.12400] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 09/21/2016] [Accepted: 09/21/2016] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Cisplatin is the standard first-line chemotherapeutic agent for the treatment of non-small cell lung cancer (NSCLC). However, resistance to chemotherapy has been a major obstacle in the management of NSCLC. Aldehyde dehydrogenase 1A1 (ALDH1A1) overexpression has been observed in a variety of cancers, including lung cancer. The purpose of this study was to investigate the effect of ALDH1A1 expression on cisplatin resistance and explore the mechanism responsible. METHODS Reverse transcriptase-PCR was applied to measure the messenger RNA expression of ALDH1A1, while Western blot assay was employed to evaluate the protein expression of ALDH1A1, B-cell lymphoma 2, Bcl-2-like protein 4, phospho-protein kinase B (p-AKT) and AKT. A short hairpin RNA was used to knockdown ALDH1A1 expression. A 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay was used to determine the effect of ALDH1A1 decrease on cell viability. The cell apoptotic rate was tested using flow cytometry assay. RESULTS ALDH1A1 is overexpressed in cisplatin resistant cell line A549/DDP, compared with A549. ALDH1A1 depletion significantly decreased A549/DDP proliferation, increased apoptosis, and reduced cisplatin resistance. In addition, the phosphoinositide 3-kinase (PI3K) / AKT pathway is activated in A549/DDP, and ALDH1A1 knockdown reduced the phosphorylation level of AKT. Moreover, the combination of ALDH1A1-short hairpin RNA and PI3K/AKT pathway inhibitor LY294002 markedly inhibited cell viability, enhanced apoptotic cell death, and increased cisplatin sensitivity. CONCLUSION These results suggest that ALDH1A1 depletion could reverse cisplatin resistance in human lung cancer cell line A549/DDP, and may act as a potential target for the treatment of lung cancers resistant to cisplatin.
Collapse
Affiliation(s)
- Yunyan Wei
- Department of GeriatricsThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Shuangshuang Wu
- Department of GeriatricsThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Wei Xu
- Department of GeriatricsThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Yan Liang
- Department of GeriatricsThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Yue Li
- Department of GeriatricsThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Weihong Zhao
- Department of GeriatricsThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Jianqing Wu
- Department of GeriatricsThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| |
Collapse
|
43
|
Moreb JS, Ucar-Bilyeu DA, Khan A. Use of retinoic acid/aldehyde dehydrogenase pathway as potential targeted therapy against cancer stem cells. Cancer Chemother Pharmacol 2016; 79:295-301. [PMID: 27942929 DOI: 10.1007/s00280-016-3213-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 11/29/2016] [Indexed: 12/28/2022]
Abstract
A large number of studies have investigated possible drug resistance mechanisms of cancer cells and suggested strategies to overcome it. In this review, we outline the role and function of aldehyde dehydrogenase (ALDH) activity in multiple cellular functions and in cancer stem cells (CSCs) and focus on the role of retinoic acid (RA), one of the products of ALDH isozymes. We discuss our observation that ATRA and other RAs can suppress ALDH activity and decrease different ALDH isozyme proteins and result in detrimental effects on cell proliferation, invasion and chemotherapy sensitivity. We review the known uses of different RAs in the treatment of cancers. We review the use of RAs in combination with chemo-/radiotherapy and the major signaling pathways affected in different tumor types. We provide follow-up on studies that may have used our prior observation with the aim of targeting the CSCs. We conclude with summary of the findings and potential impact of published studies on future use of RAs in the targeting of CSCs and drug resistance.
Collapse
Affiliation(s)
- Jan S Moreb
- Hematology/Oncology Division, Department of Medicine, University of Florida, 1600 SW Archer Rd, PO Box 100277, Gainesville, FL, 32610, USA.
| | | | - Abdullah Khan
- Hematology/Oncology Division, Department of Medicine, University of Florida, 1600 SW Archer Rd, PO Box 100277, Gainesville, FL, 32610, USA
| |
Collapse
|
44
|
Bogen A, Buske C, Hiddemann W, Bohlander SK, Christ O. Variable aldehyde dehydrogenase activity and effects on chemosensitivity of primitive human leukemic cells. Exp Hematol 2016; 47:54-63. [PMID: 27826122 DOI: 10.1016/j.exphem.2016.10.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 10/23/2016] [Accepted: 10/27/2016] [Indexed: 01/10/2023]
Abstract
Aldehyde dehydrogenase (ALDH) activity is an established feature of primitive normal human hematopoietic cells, in which it has been associated with a high expression of the 1A1 isoform of ALDH. High ALDH 1A1 activity has been reported to also characterize cells that propagate malignant populations arising in other tissues, but the regulation and basis of ALDH activity in primary human leukemic cells has not been well studied. We obtained samples from patients with newly diagnosed acute myeloid leukemia (AML; n = 21) and chronic myeloid leukemia (CML; n = 8) and analyzed different phenotypically and functionally defined subsets for their ALDH activity using the ALDEFLUOR® kit and expression of the ALDH1A1 gene. We detected cells with high ALDH activity (ALDHpos) in all samples from AML and CML patients. These were consistently enriched in the CD34+ population of these samples, but typically not in the CD34+CD38- subset. Leukemic cells with direct clonogenic activity in vitro or those able to repopulate the bone marrow of sublethally irradiated non-obese diabetic (NOD)/severe combined immunodeficiency (SCID) mice were both ALDHpos and ALDHneg. Interestingly, ALDH1A1 transcripts were highest in the ALDHneg leukemic cells and, in studies with leukemic cell lines, exposure to an inhibitor of ALDH activity variably affected sensitivity to daunorubicin. Cells with high ALDH activity are commonly found within the CD34+ population of primary human leukemic cells but, unlike in normal hematopoietic tissues, do not selectively or consistently comprise those with proliferative potential or other distinct functional properties.
Collapse
Affiliation(s)
- Anja Bogen
- Department of Medicine III, University of Munich, Munich, Germany
| | - Christian Buske
- CCC Ulm, Institute of Experimental Cancer Research, University Hospital Ulm, Ulm, Germany
| | | | - Stefan K Bohlander
- Department of Molecular Medicine and Pathology, The University of Auckland, Auckland, New Zealand
| | - Oliver Christ
- Department of Medicine III, University of Munich, Munich, Germany.
| |
Collapse
|
45
|
Cancer stem cell markers in prostate cancer: an immunohistochemical study of ALDH1, SOX2 and EZH2. Pathology 2016; 47:622-8. [PMID: 26517640 DOI: 10.1097/pat.0000000000000325] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The aims of this study were to investigate the immunohistochemical expression and potential prognostic significance of putative cancer stems cell markers ALDH1, EZH2 and SOX2 in prostate cancer.A total of 142 consecutive radical prostatectomies submitted to one laboratory with a diagnosis of prostatic adenocarcinoma between 2008 and 2012 were retrieved and retrospectively studied. Immunohistochemistry for the three markers was performed in each case and both univariate and multivariate analyses were undertaken to evaluate the correlation between the staining patterns and known histopathological prognostic features.ALDH1 showed a statistically significant association with tumour stage p < 0.001), extraprostatic extension (p < 0.001) and lymphovascular invasion (p = 0.001). EZH2 correlated with Gleason score (p = 0.044) and lymph node metastases (p = 0.023). SOX2 showed a statistically significant correlation with lymphovascular invasion only (p = 0.018) in both univariate and multivariate analyses.Cancer stem cell markers are variably expressed in prostate adenocarcinoma and immunohistochemical staining for ALDH1 and EZH2 may have a role in predicting tumour aggressiveness before treatment of prostate cancer.
Collapse
|
46
|
Ranji P, Salmani Kesejini T, Saeedikhoo S, Alizadeh AM. Targeting cancer stem cell-specific markers and/or associated signaling pathways for overcoming cancer drug resistance. Tumour Biol 2016; 37:13059-13075. [DOI: 10.1007/s13277-016-5294-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 08/18/2016] [Indexed: 02/07/2023] Open
|
47
|
Aldehyde dehydrogenase 3A1 promotes multi-modality resistance and alters gene expression profile in human breast adenocarcinoma MCF-7 cells. Int J Biochem Cell Biol 2016; 77:120-128. [PMID: 27276244 DOI: 10.1016/j.biocel.2016.06.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Revised: 03/21/2016] [Accepted: 06/03/2016] [Indexed: 12/20/2022]
Abstract
Aldehyde dehydrogenases participate in a variety of cellular homeostatic mechanisms like metabolism, proliferation, differentiation, apoptosis, whereas recently, they have been implicated in normal and cancer cell stemness. We explored roles for ALDH3A1 in conferring resistance to chemotherapeutics/radiation/oxidative stress and whether ectopic overexpression of ALDH3A1 could lead to alterations of gene expression profile associated with cancer stem cell-like phenotype. MCF-7 cells were stably transfected either with an empty vector (mock) or human aldehyde dehydrogenase 3A1 cDNA. The expression of aldehyde dehydrogenase 3A1 in MCF-7 cells was associated with altered cell proliferation rate and enhanced cell resistance against various chemotherapeutic drugs (4-hydroxyperoxycyclophosphamide, doxorubicin, etoposide, and 5-fluorouracil). Aldehyde dehydrogenase 3A1 expression also led to increased tolerance of MCF-7 cells to gamma radiation and hydrogen peroxide-induced stress. Furthermore, aldehyde dehydrogenase 3A1-expressing MCF-7 cells exhibited gene up-regulation of cyclins A, B1, B2, and down-regulation of cyclin D1 as well as transcription factors p21, CXR4, Notch1, SOX2, SOX4, OCT4, and JAG1. When compared to mock cells, no changes were observed in mRNA levels of ABCA2 and ABCB1 protein pumps with only a minor decrease of the ABCG2 pump in the aldehyde dehydrogenase 3A1-expressing cells. Also, the adhesion molecules EpCAM and CD49F were also found to be up-regulated in aldehyde dehydrogenase 3A1expressing cells. Taken together, ALDH3A1 confers a multi-modality resistance phenotype in MCF-7 cells associated with slower growth rate, increased clonogenic capacity, and altered gene expression profile, underlining its significance in cell homeostasis.
Collapse
|
48
|
Wu D, Mou YP, Chen K, Cai JQ, Zhou YC, Pan Y, Xu XW, Zhou W, Gao JQ, Chen DW, Zhang RC. Aldehyde dehydrogenase 3A1 is robustly upregulated in gastric cancer stem-like cells and associated with tumorigenesis. Int J Oncol 2016; 49:611-22. [DOI: 10.3892/ijo.2016.3551] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 05/16/2016] [Indexed: 11/05/2022] Open
|
49
|
Morbidelli L, Donnini S, Ziche M. Targeting endothelial cell metabolism for cardio-protection from the toxicity of antitumor agents. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2016; 2:3. [PMID: 33530139 PMCID: PMC7837145 DOI: 10.1186/s40959-016-0010-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 02/16/2016] [Indexed: 12/17/2022]
Abstract
The vascular endothelium plays a fundamental role in the maintenance of tissue homeostasis, regulating local blood flow and other physiological processes. Chemotherapeutic drugs and target therapies, including antiangiogenic drugs targeting vascular endothelial growth factor (VEGF) or its receptors, not only efficiently act against tumor growth, but may also induce endothelial dysfunction and cardiovascular toxicity. Continued research efforts aim to better understand, prevent and mitigate these chemotherapy associated cardiovascular diseases. Conventional chemotherapeutic agents, such as anthracyclines, platinum compounds, and taxanes, and newer targeted agents, such as bevacizumab, trastuzumab, and tyrosine kinase inhibitors, have known risk of cardiovascular toxicity, which can limit their effectiveness by promoting increased morbidity and/or mortality. This review describes a) the activity of anticancer agents in inducing endothelial dysfunction, b) the metabolic pathways and signalling cascades which may be targeted by protective agents able to maintain or restore endothelial cell function, such as endothelial nitric oxide synthase/fibroblast growth factor-2 (eNOS-FGF-2) pathway, and c) the drugs/strategies reported to improve endothelial function and to reduce the risks of cardiovascular diseases such as angiotensin converting enzyme inhibitors (ACEi) and beta blockers, that are fundamental therapies in chronic heart failure (HF), as well as non-standard HF treatments such ad nitric oxide donors and antioxidant strategies. There is increasing interest in whether ACEi, beta-blockers, and/or statins might prevent and/or therapeutically control cardiotoxic effects in cancer patients. Maintaining endothelial function during or following treatments with chemotherapeutic agents, without affecting anti-tumor drug-effectiveness, is essential for preserving or recovering cardiovascular homeostasis. In this respect, the early detection and immediate therapy of cardiovascular toxicity appear crucial for substantial recovery of cardiac function in cancer patients.
Collapse
Affiliation(s)
- Lucia Morbidelli
- Department of Life Sciences, University of Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Sandra Donnini
- Department of Life Sciences, University of Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Marina Ziche
- Department of Life Sciences, University of Siena, Via A. Moro 2, 53100 Siena, Italy
| |
Collapse
|
50
|
Buchman CD, Mahalingan KK, Hurley TD. Discovery of a series of aromatic lactones as ALDH1/2-directed inhibitors. Chem Biol Interact 2015; 234:38-44. [PMID: 25641190 DOI: 10.1016/j.cbi.2014.12.038] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Revised: 12/17/2014] [Accepted: 12/31/2014] [Indexed: 01/02/2023]
Abstract
In humans, the aldehyde dehydrogenase superfamily consists of 19 isoenzymes which mostly catalyze the NAD(P)(+)-dependent oxidation of aldehydes. Many of these isoenzymes have overlapping substrate specificities and therefore their potential physiological functions may overlap. Thus the development of new isoenzyme-selective probes would be able to better delineate the function of a single isoenzyme and its individual contribution to the metabolism of a particular substrate. This specific study was designed to find a novel modulator of ALDH2, a mitochondrial ALDH isoenzyme most well-known for its role in acetaldehyde oxidation. 53 compounds were initially identified to modulate the activity of ALDH2 by a high-throughput esterase screen from a library of 63,000 compounds. Of these initial 53 compounds, 12 were found to also modulate the oxidation of propionaldehyde by ALDH2. Single concentration measurements at 10μM compound were performed using ALDH1A1, ALDH1A2, ALDH1A3, ALDH2, ALDH1B1, ALDH3A1, ALDH4A1, and/or ALDH5A1 to determine the selectivity of these 12 compounds toward ALDH2. Four of the twelve compounds shared an aromatic lactone structure and were found to be potent inhibitors of the ALDH1/2 isoenzymes, but have no inhibitory effect on ALDH3A1, ALDH4A1 or ALDH5A1. Two of the aromatic lactones show selectivity within the ALDH1/2 class, and one appears to be selective for ALDH2 compared to all other isoenzymes tested.
Collapse
Affiliation(s)
- Cameron D Buchman
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202-5122, United States
| | - Krishna K Mahalingan
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202-5122, United States
| | - Thomas D Hurley
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202-5122, United States.
| |
Collapse
|