1
|
Paul SK, Guendouzi A, Banerjee A, Guendouzi A, Haldar R. Identification of approved drugs with ALDH1A1 inhibitory potential aimed at enhancing chemotherapy sensitivity in cancer cells: an in-silico drug repurposing approach. J Biomol Struct Dyn 2025; 43:3830-3844. [PMID: 38189344 DOI: 10.1080/07391102.2023.2300127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 12/21/2023] [Indexed: 01/09/2024]
Abstract
The aldehyde dehydrogenase 1A1 (ALDH1A1) also known as retinal dehydrogenase, is an enzyme normally involved in the cellular metabolism, development and detoxification processes in healthy cells. However, it's also considered a cancer stem cell marker and its high levels of expression in several cancers, including breast, lung, ovarian, and colon cancer have been associated with poor prognosis and resistance to chemotherapy. Given its crucial role in chemotherapy resistance by detoxification of chemotherapeutic drugs, ALDH1A1 has attracted significant research interest as a potential therapeutic target for cancer. Though a few synthetic inhibitors of ALDH1A1 have been synthesized and their efficacy has been proved in-vitro and in-vivo studies, none of them have passed clinical trials so far. In this scenario, we have performed an in-silico study to verify whether any of the already approved drugs used for various purposes has the ability to inhibit catalytic activity of ALDH1A1, so that they can be repurposed for cancer therapy. Keeping in mind the feasibility of repurposing in a larger population we have selected the approved drugs from five widely used drug categories such as antibiotic, antiviral, antifungal, anti diabetic and antihypertensive for screening. Computational techniques like molecular docking, molecular dynamics simulations and MM-PBSA binding energy calculation have been used in this study to screen the approved drugs. Based on the logical analysis of results, we propose that three drugs - telmisartan, irbesartan and maraviroc can inhibit the catalytic activity of ALDH1A1 and thus can be repurposed to increase chemotherapy sensitivity in cancer cells.
Collapse
Affiliation(s)
- Sanjay Kumar Paul
- Department of Physiology, University of Calcutta, Kolkata, India
- Department of Zoology, Rammohan College, Kolkata, West Bengal, India
| | - Abdelmadjid Guendouzi
- Center for Research in Pharmaceutical Sciences (CRSP), Constantine, Algeria
- Ecole Normale Supérieure ENS Constantine, Constantine, Algeria
| | | | | | - Rajen Haldar
- Department of Physiology, University of Calcutta, Kolkata, India
| |
Collapse
|
2
|
Zhang W, Song Z, Tian Y, Zhang R, Guo Z, Yang Y, Jiang X, Zhang R. Copper homeostasis and Cuprotosis: Exploring novel therapeutic strategies for connective tissue diseases. Int Immunopharmacol 2025; 145:113698. [PMID: 39642560 DOI: 10.1016/j.intimp.2024.113698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 11/09/2024] [Accepted: 11/20/2024] [Indexed: 12/09/2024]
Abstract
Copper is an indispensable element for human health, with its balance being critical to prevent the onset of diseases, particularly those affecting connective tissues. Imbalances in copper levels can lead to pathological alterations. Research indicates that copper supplements and chelators hold promise for the treatment of certain conditions, yet the precise mechanisms by which copper imbalances and the cell death mechanism known as cuprotosis contribute to connective tissue diseases remain elusive. This paper delves into the potential role of copper imbalance and cuprotosis in connective tissue diseases and evaluates the underlying cellular mechanisms. The goal is to offer practical insights into targeted therapies for dysregulated copper metabolism, with the aim of devising novel strategies for the treatment of connective tissue diseases.
Collapse
Affiliation(s)
- Wenlan Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Zhijie Song
- Department of Rheumatology and Immunology, Chifeng Cancer Hospital, Chifeng 024000, Inner Mongolia Autonomous Region, China
| | - Yuanyuan Tian
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Ruifeng Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Zhigang Guo
- Department of Rheumatology and Immunology, Chifeng Cancer Hospital, Chifeng 024000, Inner Mongolia Autonomous Region, China
| | - Yanmei Yang
- Department of Rheumatology and Immunology, Chifeng Cancer Hospital, Chifeng 024000, Inner Mongolia Autonomous Region, China
| | - Xijuan Jiang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Ruoyi Zhang
- Department of Rheumatology and Immunology, Chifeng Cancer Hospital, Chifeng 024000, Inner Mongolia Autonomous Region, China.
| |
Collapse
|
3
|
Tahiliani H, Dhayalan A, Li MC, Hsieh HP, Coumar MS. Aldehyde dehydrogenases as drug targets for cancer: SAR and structural biology aspects for inhibitor design. Bioorg Chem 2025; 154:108019. [PMID: 39689509 DOI: 10.1016/j.bioorg.2024.108019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 10/20/2024] [Accepted: 11/28/2024] [Indexed: 12/19/2024]
Abstract
Aldehydes are organic compounds containing a carbonyl group found exogenously or produced by normal metabolic processes and their accumulation can lead to toxicity if not cleared. Aldehyde dehydrogenases (ALDHs) are NAD(P)+-dependent enzymes that catalyze the oxidation of such aldehydes and prevent their accumulation. Along with this primary detoxification function, the known 19 human isoforms of ALDHs, which act on different substrates, are also involved in various physiological and developmental processes. Functional alterations of ALDHs via mutations or expression levels cause various disease conditions, including many different cancer types like lung, ovarian, etc. These properties make this family of enzymes an ideal therapeutic and prognostic target for drug development. However, sequence similarities between the ALDH isoforms force the need to design inhibitors for a specific isoform using the differences in the substrate-binding sites of each protein. This has resulted in developing isoform-specific inhibitors, especially for ALDH1A1, ALDH2, and ALDH3A1, which are implicated in various cancers. In this review, we briefly outline the functional roles of the different isoforms of the ALDH family members, their role in cancer and discuss the various selective inhibitors that have been developed for the ALDH1A1 and ALDH3A1 enzymes, along with a detailed examination of the respective structure-activity relationship (SAR) studies available. From the available SAR and structural biology data, insights into the functional groups and interactions necessary to develop selective inhibitors for ALDH1A1 and ALDH3A1 are highlighted, which can act as a guide for developing more potent and selective inhibitors of ALDH isoforms.
Collapse
Affiliation(s)
- Himanshu Tahiliani
- Department of Bioinformatics, School of Life Scicnces, Pondicherry University, Pondicherry 605014, India
| | - Arunkumar Dhayalan
- Department of Biotechnology, School of Life Sciences, Pondicherry University, Pondicherry 605014, India
| | - Mu-Chun Li
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 350401, Taiwan, ROC; Biomedical Translation Research Center, Academia Sinica, Taipei City 115202, Taiwan, ROC
| | - Hsing-Pang Hsieh
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 350401, Taiwan, ROC; Biomedical Translation Research Center, Academia Sinica, Taipei City 115202, Taiwan, ROC; Department of Chemistry, National Tsing Hua University, Hsinchu City 300044, Taiwan, ROC
| | - Mohane Selvaraj Coumar
- Department of Bioinformatics, School of Life Scicnces, Pondicherry University, Pondicherry 605014, India.
| |
Collapse
|
4
|
Zhang ML, Li WX, Wang XY, Chen XF, Zhang H, Meng GQ, Chen YL, Wu YL, Yang LQ, Zhang SQ, Feng KR, Niu L, Tang JF. Characterizing metabolomic and transcriptomic changes, and investigating the therapeutic mechanism of Psoralea corylifolia linn. In the treatment of kidney-yang deficiency syndrome in rats. Heliyon 2024; 10:e39006. [PMID: 39524713 PMCID: PMC11550036 DOI: 10.1016/j.heliyon.2024.e39006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 09/24/2024] [Accepted: 10/04/2024] [Indexed: 11/16/2024] Open
Abstract
Kidney-yang deficiency syndrome (KYDS) is characterized by a metabolic disorder stemming from neuroendocrine dysregulation, often associated with hepatic dysfunction. In traditional Chinese medicine, Psoralea corylifolia Linn. (BGZ) is commonly utilized for treating KYDS. However, the specific therapeutic effects of BGZ on liver function regulation remain unclear. To evaluate the protective effects of BGZ against KYDS in rats, organ index, alanine aminotransferase (ALT), aspartate aminotransferase (AST), and other biochemical indices were analyzed. Hematoxylin and eosin (HE) staining was utilized to assess liver histopathology. Additionally, transcriptomic and metabolomic analyses were conducted to identify potential biomarkers. BGZ treatment led to a significant reduction in ALT and AST levels, accompanied by improvements in liver histopathology in rats with KYDS. Moreover, BGZ induced significant alterations in 92 differentially expressed genes (DEGs) and 20 metabolites in the KYDS rat model. The comprehensive examination of metabolites and DEGs identified potential mechanisms underlying the therapeutic effects of BGZ, highlighting the neuroactive ligand-receptor interaction, cAMP signaling pathway, calcium signaling pathway, and cytokine-cytokine receptor interaction as key mechanisms. Validation of key targets within the cAMP pathway was substantiated through enzyme-linked immunosorbent assay and real-time quantitative polymerase chain reaction. The cAMP pathway emerges as a plausible mechanism through which BGZ exerts protective effects against KYDS. The findings of this study contribute to an improved understanding of the therapeutic actions of BGZ and establish a groundwork for further research into the complex pathways involved, as well as the potential for drug-targeted therapies for KYDS.
Collapse
Affiliation(s)
- Ming-Liang Zhang
- Department of Pharmacy, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, 450000, China
- Henan Province Engineering Research Center for Clinical Application, Evaluation and Transformation of Traditional Chinese Medicine, Zhengzhou, 450000, China
- Henan Provincial Key Laboratory for Clinical Pharmacy of Traditional Chinese Medicine, Zhengzhou, 450000, China
| | - Wei-Xia Li
- Department of Pharmacy, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, 450000, China
- Henan Province Engineering Research Center for Clinical Application, Evaluation and Transformation of Traditional Chinese Medicine, Zhengzhou, 450000, China
- Henan Provincial Key Laboratory for Clinical Pharmacy of Traditional Chinese Medicine, Zhengzhou, 450000, China
- Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Xiao-Yan Wang
- Department of Pharmacy, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, 450000, China
- Henan Province Engineering Research Center for Clinical Application, Evaluation and Transformation of Traditional Chinese Medicine, Zhengzhou, 450000, China
- Henan Provincial Key Laboratory for Clinical Pharmacy of Traditional Chinese Medicine, Zhengzhou, 450000, China
| | - Xiao-Fei Chen
- Department of Pharmacy, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, 450000, China
- Henan Province Engineering Research Center for Clinical Application, Evaluation and Transformation of Traditional Chinese Medicine, Zhengzhou, 450000, China
- Henan Provincial Key Laboratory for Clinical Pharmacy of Traditional Chinese Medicine, Zhengzhou, 450000, China
| | - Hui Zhang
- Department of Pharmacy, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, 450000, China
- Henan Province Engineering Research Center for Clinical Application, Evaluation and Transformation of Traditional Chinese Medicine, Zhengzhou, 450000, China
- Henan Provincial Key Laboratory for Clinical Pharmacy of Traditional Chinese Medicine, Zhengzhou, 450000, China
| | - Gao-Quan Meng
- Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Yu-Long Chen
- Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Ya-Li Wu
- Department of Pharmacy, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, 450000, China
- Henan Province Engineering Research Center for Clinical Application, Evaluation and Transformation of Traditional Chinese Medicine, Zhengzhou, 450000, China
- Henan Provincial Key Laboratory for Clinical Pharmacy of Traditional Chinese Medicine, Zhengzhou, 450000, China
| | - Liu-Qing Yang
- Department of Pharmacy, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, 450000, China
- Henan Province Engineering Research Center for Clinical Application, Evaluation and Transformation of Traditional Chinese Medicine, Zhengzhou, 450000, China
- Henan Provincial Key Laboratory for Clinical Pharmacy of Traditional Chinese Medicine, Zhengzhou, 450000, China
| | - Shu-Qi Zhang
- Department of Pharmacy, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, 450000, China
- Henan Province Engineering Research Center for Clinical Application, Evaluation and Transformation of Traditional Chinese Medicine, Zhengzhou, 450000, China
- Henan Provincial Key Laboratory for Clinical Pharmacy of Traditional Chinese Medicine, Zhengzhou, 450000, China
| | - Ke-Ran Feng
- Department of Pharmacy, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, 450000, China
- Henan Province Engineering Research Center for Clinical Application, Evaluation and Transformation of Traditional Chinese Medicine, Zhengzhou, 450000, China
- Henan Provincial Key Laboratory for Clinical Pharmacy of Traditional Chinese Medicine, Zhengzhou, 450000, China
| | - Lu Niu
- Department of Pharmacy, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, 450000, China
- Henan Province Engineering Research Center for Clinical Application, Evaluation and Transformation of Traditional Chinese Medicine, Zhengzhou, 450000, China
- Henan Provincial Key Laboratory for Clinical Pharmacy of Traditional Chinese Medicine, Zhengzhou, 450000, China
| | - Jin-Fa Tang
- Department of Pharmacy, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, 450000, China
- Henan Province Engineering Research Center for Clinical Application, Evaluation and Transformation of Traditional Chinese Medicine, Zhengzhou, 450000, China
- Henan Provincial Key Laboratory for Clinical Pharmacy of Traditional Chinese Medicine, Zhengzhou, 450000, China
- Henan University of Traditional Chinese Medicine, Zhengzhou, China
| |
Collapse
|
5
|
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
|
6
|
Ren Z, Liu N, Jia H, Sun M, Ma S, Zhao B, Chen Y, Miao X, Cao Z, Dong J. Discovery of Aldehyde Dehydrogenase as a Potential Fungicide Target and Screening of its Natural Inhibitors against Fusarium verticillioides. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:19424-19435. [PMID: 39172074 DOI: 10.1021/acs.jafc.4c05553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/23/2024]
Abstract
Fusarium verticillioides is the primary pathogen causing ear rot and stalk rot in corn (Zea mays). It not only affects yields but also produces mycotoxins endangering both human and animal health. Aldehyde dehydrogenase (ALDH) is essential for the oxidation of aldehydes in living organisms, making it a potential target for human drug design. However, there are limited reports on its function in plant pathogenic fungus. In this study, we analyzed the expression levels and gene knockout mutants, revealing that ALDH genes FvALDH-43 and FvALDH-96 in F. verticillioides played significant roles in pathogenicity and resistance to low-temperature stress by affecting antioxidant capacity. Virtual screening for natural product inhibitors and molecular docking were performed targeting FvALDH-43 and FvALDH-96. Following the biological activity analysis, three natural flavonoid compounds featuring a 2-hydroxyphenol chromene were identified. Among these, Taxifolin exhibited the highest biological activity and low toxicity. Both in vitro and in vivo biological evaluations confirmed that Taxifolin targeted ALDH and inhibited its activity. These findings indicate that aldehyde dehydrogenase may serve as a promising target for the design of novel fungicides.
Collapse
Affiliation(s)
- Zhiguo Ren
- State Key Laboratory of North China Crop Improvement and regulation/Hebei Key Laboratory of Plant Physiology and Molecular Pathology, Baoding 071000, China
- College of Plant Protection, Hebei Agricultural University, Baoding 071000, China
| | - Ning Liu
- State Key Laboratory of North China Crop Improvement and regulation/Hebei Key Laboratory of Plant Physiology and Molecular Pathology, Baoding 071000, China
- College of Plant Protection, Hebei Agricultural University, Baoding 071000, China
| | - Hui Jia
- State Key Laboratory of North China Crop Improvement and regulation/Hebei Key Laboratory of Plant Physiology and Molecular Pathology, Baoding 071000, China
- College of Plant Protection, Hebei Agricultural University, Baoding 071000, China
| | - Manli Sun
- State Key Laboratory of North China Crop Improvement and regulation/Hebei Key Laboratory of Plant Physiology and Molecular Pathology, Baoding 071000, China
- College of Plant Protection, Hebei Agricultural University, Baoding 071000, China
| | - Shujie Ma
- State Key Laboratory of North China Crop Improvement and regulation/Hebei Key Laboratory of Plant Physiology and Molecular Pathology, Baoding 071000, China
- College of Plant Protection, Hebei Agricultural University, Baoding 071000, China
| | - Bin Zhao
- State Key Laboratory of North China Crop Improvement and regulation/Hebei Key Laboratory of Plant Physiology and Molecular Pathology, Baoding 071000, China
- College of Plant Protection, Hebei Agricultural University, Baoding 071000, China
| | - Yue Chen
- State Key Laboratory of North China Crop Improvement and regulation/Hebei Key Laboratory of Plant Physiology and Molecular Pathology, Baoding 071000, China
| | - Xiaoyang Miao
- Hebei Peiran's Century Nutritional Foods Co., Ltd., Cangzhou 061000, China
| | - Zhiyan Cao
- State Key Laboratory of North China Crop Improvement and regulation/Hebei Key Laboratory of Plant Physiology and Molecular Pathology, Baoding 071000, China
- College of Plant Protection, Hebei Agricultural University, Baoding 071000, China
| | - Jingao Dong
- State Key Laboratory of North China Crop Improvement and regulation/Hebei Key Laboratory of Plant Physiology and Molecular Pathology, Baoding 071000, China
- College of Plant Protection, Hebei Agricultural University, Baoding 071000, China
| |
Collapse
|
7
|
Takahashi C, Chtcherbinine M, Huddle BC, Wilson MW, Emmel T, Hohlman RM, McGonigal S, Buckanovich RJ, Larsen SD, Hurley TD. Development of substituted benzimidazoles as inhibitors of human aldehyde dehydrogenase 1A isoenzymes. Chem Biol Interact 2024; 391:110910. [PMID: 38364885 PMCID: PMC11062403 DOI: 10.1016/j.cbi.2024.110910] [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: 11/02/2023] [Revised: 02/01/2024] [Accepted: 02/07/2024] [Indexed: 02/18/2024]
Abstract
Aldehyde dehydrogenase 1A (ALDH1A) isoforms may be a useful target for overcoming chemotherapy resistance in high-grade serous ovarian cancer (HGSOC) and other solid tumor cancers. However, as different cancers express different ALDH1A isoforms, isoform selective inhibitors may have a limited therapeutic scope. Furthermore, resistance to an ALDH1A isoform selective inhibitor could arise via induction of expression of other ALDH1A isoforms. As such, we have focused on the development of pan-ALDH1A inhibitors, rather than on ALDH1A isoform selective compounds. Herein, we report the development of a new group of pan-ALDH1A inhibitors to assess whether broad spectrum ALDH1A inhibition is an effective adjunct to chemotherapy in HGSOC. Optimization of the CM10 scaffold, aided by ALDH1A1 crystal structures, led to improved biochemical potencies, improved cellular efficacy as demonstrated by reduction in ALDEFLUOR signal in HGSOC cells, and substantial improvements in liver microsomal stability. Based on this work we identified two compounds 17 and 25 suitable for future in vivo proof of concept experiments.
Collapse
Affiliation(s)
- Cyrus Takahashi
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Mikhail Chtcherbinine
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Brandt C Huddle
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Michael W Wilson
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Timothy Emmel
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Robert M Hohlman
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Stacy McGonigal
- Department of Obstetrics, Gynecology, and Reproductive Sciences, the Magee-Women's Research Institute, Pittsburgh, PA 15213, USA
| | - Ronald J Buckanovich
- Department of Obstetrics, Gynecology, and Reproductive Sciences, the Magee-Women's Research Institute, Pittsburgh, PA 15213, USA; Division of Hematology-Oncology, Departments of Internal Medicine and Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh Medical Center and the Magee-Women's Research Institute, Pittsburgh, PA, 15213, USA
| | - Scott D Larsen
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Thomas D Hurley
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| |
Collapse
|
8
|
Tripathi N, Bhardwaj N, Kumar S, Jain SK. Phytochemical and Pharmacological Aspects of Psoralen - A Bioactive Furanocoumarin from Psoralea corylifolia Linn. Chem Biodivers 2023; 20:e202300867. [PMID: 37752710 DOI: 10.1002/cbdv.202300867] [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: 06/14/2023] [Revised: 09/25/2023] [Accepted: 09/26/2023] [Indexed: 09/28/2023]
Abstract
Since long ago, medicinal plants have played a vital role in drug discovery. Being blessed and rich in chemovars with diverse scaffolds, they have unique characteristics of evolving based on the need. The World Health Organization also mentions that medicinal plants remain at the center for meeting primary healthcare needs as the population relies on them. The plant-derived natural products have remained an attractive choice for drug development owing to their specific biological functions relevant to human health and also the high degree of potency and specificity they offer. In this context, one such esteemed phytoconstituent with inexplicable biological potential is psoralen, a furanocoumarin. Psoralen was the first constituent isolated from the plant Psoralea corylifolia, commonly known as Bauchi. Despite being a life-saver for psoriasis, vitiligo, and leukoderma, it also showed immense anticancer, anti-inflammatory, and anti-osteoporotic potential. This review brings attention to the possible application of psoralen as an attractive target for rational drug design and medicinal chemistry. It discusses the various methods for the total synthesis of psoralen, its extraction, the pharmacological spectrum of psoralen, and the derivatization done on psoralen.
Collapse
Affiliation(s)
- Nancy Tripathi
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology BHU, Varanasi, 221005, India
| | - Nivedita Bhardwaj
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology BHU, Varanasi, 221005, India
| | - Sanjay Kumar
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology BHU, Varanasi, 221005, India
| | - Shreyans K Jain
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology BHU, Varanasi, 221005, India
| |
Collapse
|
9
|
Jessen-Howard D, Pan Q, Ascher DB. Identifying the Molecular Drivers of Pathogenic Aldehyde Dehydrogenase Missense Mutations in Cancer and Non-Cancer Diseases. Int J Mol Sci 2023; 24:10157. [PMID: 37373306 DOI: 10.3390/ijms241210157] [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: 04/30/2023] [Revised: 06/07/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
Human aldehyde dehydrogenases (ALDHs) comprising 19 isoenzymes play a vital role on both endogenous and exogenous aldehyde metabolism. This NAD(P)-dependent catalytic process relies on the intact structural and functional activity of the cofactor binding, substrate interaction, and the oligomerization of ALDHs. Disruptions on the activity of ALDHs, however, could result in the accumulation of cytotoxic aldehydes, which have been linked with a wide range of diseases, including both cancers as well as neurological and developmental disorders. In our previous works, we have successfully characterised the structure-function relationships of the missense variants of other proteins. We, therefore, applied a similar analysis pipeline to identify potential molecular drivers of pathogenic ALDH missense mutations. Variants data were first carefully curated and labelled as cancer-risk, non-cancer diseases, and benign. We then leveraged various computational biophysical methods to describe the changes caused by missense mutations, informing a bias of detrimental mutations with destabilising effects. Cooperating with these insights, several machine learning approaches were further utilised to investigate the combination of features, revealing the necessity of the conservation of ALDHs. Our work aims to provide important biological perspectives on pathogenic consequences of missense mutations of ALDHs, which could be invaluable resources in the development of cancer treatment.
Collapse
Affiliation(s)
- Dana Jessen-Howard
- School of Chemistry and Molecular Bioscience, University of Queensland, Brisbane, QLD 4072, Australia
| | - Qisheng Pan
- School of Chemistry and Molecular Bioscience, University of Queensland, Brisbane, QLD 4072, Australia
- Computational Biology and Clinical Informatics, Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
| | - David B Ascher
- School of Chemistry and Molecular Bioscience, University of Queensland, Brisbane, QLD 4072, Australia
- Computational Biology and Clinical Informatics, Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
| |
Collapse
|
10
|
Virtual Screening of FDA-Approved Drugs for Enhanced Binding with Mitochondrial Aldehyde Dehydrogenase. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27248773. [PMID: 36557906 PMCID: PMC9781114 DOI: 10.3390/molecules27248773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/07/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022]
Abstract
Mitochondrial aldehyde dehydrogenase (ALDH2) is a potential target for the treatment of substance use disorders such as alcohol addiction. Here, we adopted computational methods of molecular dynamics (MD) simulation, docking, and molecular mechanics Poisson-Boltzmann surface area (MM-PBSA) analysis to perform a virtual screening of FDA-approved drugs, hitting potent inhibitors against ALDH2. Using MD-derived conformations as receptors, butenafine (net charge q = +1 e) and olaparib (q = 0) were selected as promising compounds with a low toxicity and a binding strength equal to or stronger than previously reported potent inhibitors of daidzin and CVT-10216. A few negatively charged compounds were also hit from the docking with the Autodock Vina software, while the MM-PBSA analysis yielded positive binding energies (unfavorable binding) for these compounds, mainly owing to electrostatic repulsion in association with a negatively charged receptor (q = -6 e for ALDH2 plus the cofactor NAD+). This revealed a deficiency of the Vina scoring in dealing with strong charge-charge interactions between binding partners, due to its built-in protocol of not using atomic charges for electrostatic interactions. These observations indicated a requirement of further verification using MD and/or MM-PBSA after docking prediction. The identification of key residues for the binding implied that the receptor residues at the bottom and entrance of the substrate-binding hydrophobic tunnel were able to offer additional interactions with different inhibitors such as π-π, π-alkyl, van der Waals contacts, and polar interactions, and that the rational use of these interactions is beneficial to the design of potent inhibitors against ALDH2.
Collapse
|
11
|
Alkyl esters of 7-hydroxycoumarin-3-carboxylic acid as potent tissue-specific uncouplers of oxidative phosphorylation: Involvement of ATP/ADP translocase in mitochondrial uncoupling. Arch Biochem Biophys 2022; 728:109366. [PMID: 35878680 DOI: 10.1016/j.abb.2022.109366] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 07/15/2022] [Accepted: 07/19/2022] [Indexed: 11/22/2022]
Abstract
An impressive body of evidence has been accumulated now on sound beneficial effects of mitochondrial uncouplers in struggling with the most dangerous pathologies such as cancer, infective diseases, neurodegeneration and obesity. To increase their efficacy while gaining further insight in the mechanism of the uncoupling action has been remaining a challenge. Encouraged by our previous promising results on lipophilic derivatives of 7-hydroxycoumarin-4-acetic acid (UB-4 esters), here, we use a 7-hydroxycoumarin-3-carboxylic acid scaffold to synthesize a new series of 7-hydroxycoumarin (umbelliferone, UB)-derived uncouplers of oxidative phosphorylation - alkyl esters of umbelliferone-3-carboxylic acid (UB-3 esters) with varying carbon chain length. Compared to the UB-4 derivatives, UB-3 esters proved to be stronger uncouplers: the most effective of them caused a pronounced increase in the respiration rate of isolated rat heart mitochondria (RHM) at submicromolar concentrations. Both of these series of UB derivatives exhibited a striking difference between their uncoupling patterns in mitochondria isolated from liver and heart or kidney, namely: a pronounced but transient decrease in membrane potential, followed by its recovery, was observed after the addition of these compounds to isolated rat liver mitochondria (RLM), while the depolarization of RHM and rat kidney mitochondria (RKM) was rather stable under the same conditions. Interestingly, partial reversal of this depolarization in RHM and RKM was caused by carboxyatractyloside, an inhibitor of ATP/ADP translocase, thereby pointing to the involvement of this mitochondrial membrane protein in the uncoupling activity of both UB-3 and UB-4 esters. The fast membrane potential recovery in RLM uncoupled by the addition of the UB esters was apparently associated with hydrolysis of these compounds, catalyzed by mitochondrial aldehyde dehydrogenase (ALDH2), being in high abundance in liver compared to other tissues. Protonophoric properties of the UB derivatives in isolated mitochondria were confirmed by measurements of RHM swelling in the presence of potassium acetate. In model bilayer lipid membranes (BLM), proton-carrying activity of UB-3 esters was demonstrated by measuring fluorescence response of the pH-dependent dye pyranine in liposomes. Electrophysiological experiments on identified neurons from Lymnaea stagnalis demonstrated low neurotoxicity of UB-3 esters. Resazurin-based cell viability assay showed low toxicity of UB-3 esters to HEK293 cells and primary human fibroblasts. Thus, the present results enable us to consider UB-3 esters as effective tissue-specific protonophoric mitochondrial uncouplers.
Collapse
|
12
|
Muralikrishnan V, Fang F, Given TC, Podicheti R, Chtcherbinine M, Metcalfe TX, Sriramkumar S, O’Hagan HM, Hurley TD, Nephew KP. A Novel ALDH1A1 Inhibitor Blocks Platinum-Induced Senescence and Stemness in Ovarian Cancer. Cancers (Basel) 2022; 14:3437. [PMID: 35884498 PMCID: PMC9318275 DOI: 10.3390/cancers14143437] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 07/08/2022] [Accepted: 07/10/2022] [Indexed: 02/04/2023] Open
Abstract
Ovarian cancer is a deadly disease attributed to late-stage detection as well as recurrence and the development of chemoresistance. Ovarian cancer stem cells (OCSCs) are hypothesized to be largely responsible for the emergence of chemoresistant tumors. Although chemotherapy may initially succeed at decreasing the size and number of tumors, it leaves behind residual malignant OCSCs. In this study, we demonstrate that aldehyde dehydrogenase 1A1 (ALDH1A1) is essential for the survival of OCSCs. We identified a first-in-class ALDH1A1 inhibitor, compound 974, and used 974 as a tool to decipher the mechanism of stemness regulation by ALDH1A1. The treatment of OCSCs with 974 significantly inhibited ALDH activity, the expression of stemness genes, and spheroid and colony formation. An in vivo limiting dilution assay demonstrated that 974 significantly inhibited CSC frequency. A transcriptomic sequencing of cells treated with 974 revealed a significant downregulation of genes related to stemness and chemoresistance as well as senescence and the senescence-associated secretory phenotype (SASP). We confirmed that 974 inhibited the senescence and stemness induced by platinum-based chemotherapy in functional assays. Overall, these data establish that ALDH1A1 is essential for OCSC survival and that ALDH1A1 inhibition suppresses chemotherapy-induced senescence and stemness. Targeting ALDH1A1 using small-molecule inhibitors in combination with chemotherapy therefore presents a promising strategy to prevent ovarian cancer recurrence and has the potential for clinical translation.
Collapse
Affiliation(s)
- Vaishnavi Muralikrishnan
- Cell, Molecular and Cancer Biology Graduate Program, Medical Sciences Department, Indiana University School of Medicine, Bloomington, IN 47405, USA; (V.M.); (T.C.G.); (T.X.M.); (S.S.); (H.M.O.)
| | - Fang Fang
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
| | - Tyler C. Given
- Cell, Molecular and Cancer Biology Graduate Program, Medical Sciences Department, Indiana University School of Medicine, Bloomington, IN 47405, USA; (V.M.); (T.C.G.); (T.X.M.); (S.S.); (H.M.O.)
| | - Ram Podicheti
- Center for Genomics and Bioinformatics, Indiana University, Bloomington, IN 46202, USA;
| | - Mikhail Chtcherbinine
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
| | - Tara X. Metcalfe
- Cell, Molecular and Cancer Biology Graduate Program, Medical Sciences Department, Indiana University School of Medicine, Bloomington, IN 47405, USA; (V.M.); (T.C.G.); (T.X.M.); (S.S.); (H.M.O.)
| | - Shruthi Sriramkumar
- Cell, Molecular and Cancer Biology Graduate Program, Medical Sciences Department, Indiana University School of Medicine, Bloomington, IN 47405, USA; (V.M.); (T.C.G.); (T.X.M.); (S.S.); (H.M.O.)
| | - Heather M. O’Hagan
- Cell, Molecular and Cancer Biology Graduate Program, Medical Sciences Department, Indiana University School of Medicine, Bloomington, IN 47405, USA; (V.M.); (T.C.G.); (T.X.M.); (S.S.); (H.M.O.)
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN 46202, USA
| | - Thomas D. Hurley
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
| | - Kenneth P. Nephew
- Cell, Molecular and Cancer Biology Graduate Program, Medical Sciences Department, Indiana University School of Medicine, Bloomington, IN 47405, USA; (V.M.); (T.C.G.); (T.X.M.); (S.S.); (H.M.O.)
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
- Department of Anatomy, Cell Biology and Physiology, Department of Obstetrics and Gynecology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
13
|
Krasnov VS, Kirsanov RS, Khailova LS, Firsov AM, Nazarov PA, Tashlitsky VN, Korshunova GA, Kotova EA, Antonenko YN. Alkyl esters of umbelliferone-4-acetic acid as protonophores in bilayer lipid membranes and ALDH2-dependent soft uncouplers in rat liver mitochondria. Bioelectrochemistry 2022; 145:108081. [DOI: 10.1016/j.bioelechem.2022.108081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 01/25/2022] [Accepted: 01/28/2022] [Indexed: 02/08/2023]
|
14
|
Zhang Y, Qiu Y, Zhang H. Computational Investigation of Structural Basis for Enhanced Binding of Isoflavone Analogues with Mitochondrial Aldehyde Dehydrogenase. ACS OMEGA 2022; 7:8115-8127. [PMID: 35284766 PMCID: PMC8908493 DOI: 10.1021/acsomega.2c00032] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 02/11/2022] [Indexed: 06/14/2023]
Abstract
Isoflavone compounds are potent inhibitors against mitochondrial aldehyde dehydrogenase (ALDH2) for the treatment of alcoholism and drug addiction, and an in-depth understanding of the underlying structural basis helps design new inhibitors for enhanced binding. Here, we investigated the binding poses and strengths of eight isoflavone analogues (including CVT-10216 and daidzin) with ALDH2 via computational methods of molecular docking, molecular dynamics (MD) simulation, molecular mechanics Poisson-Boltzmann surface area (MM-PBSA), steered MD, and umbrella sampling. Neither the Vina scoring of docked and MD-sampled complexes nor the nonbonded protein-inhibitor interaction energy from MD simulations is able to reproduce the relative binding strength of the inhibitors compared to experimental IC50 values. Considering the solvation contribution, MM-PBSA and relatively expensive umbrella sampling yield good performance for the relative binding (free) energies. The isoflavone skeleton prefers to form π-π stacking, π-sulfur, and π-alkyl interactions with planar (Phe and Trp) or sulfur-containing (Cys and Met) residues. The enhanced inhibition of CVT-10216 originates from both end groups of the isoflavone skeleton offering strong van der Waals contacts and from the methylsulfonamide group at the 4' position by hydrogen bonding (HB) with neighboring receptor residues. These results indicate that the hydrophobic binding tunnel of ALDH2 is larger than the isoflavone skeleton in length and thus an extended hydrophobic core is likely a premise for potent inhibitors.
Collapse
|
15
|
Omran Z. Novel Disulfiram Derivatives as ALDH1a1-Selective Inhibitors. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27020480. [PMID: 35056791 PMCID: PMC8778300 DOI: 10.3390/molecules27020480] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/04/2022] [Accepted: 01/11/2022] [Indexed: 01/16/2023]
Abstract
Aldehyde dehydrogenase-1a1 (ALDH1a1), the enzyme responsible for the oxidation of retinal into retinoic acid, represents a key therapeutic target for the treatment of debilitating disorders such as cancer, obesity, and inflammation. Drugs that can inhibit ALDH1a1 include disulfiram, an FDA-approved drug to treat chronic alcoholism. Disulfiram, by carbamylation of the catalytic cysteines, irreversibly inhibits ALDH1a1 and ALDH2. The latter is the isozyme responsible for important physiological processes such as the second stage of alcohol metabolism. Given the fact that ALDH1a1 has a larger substrate tunnel than that in ALDH2, replacing disulfiram ethyl groups with larger motifs will yield selective ALDH1a1 inhibitors. We report herein the synthesis of new inhibitors of ALDH1a1 where (hetero)aromatic rings were introduced into the structure of disulfiram. Most of the developed compounds retained the anti-ALDH1a1 activity of disulfiram; however, they were completely devoid of inhibitory activity against ALDH2.
Collapse
Affiliation(s)
- Ziad Omran
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, Jeddah 21442, Saudi Arabia;
- College of Pharmacy, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| |
Collapse
|
16
|
Xia D, Liu H, Cheng X, Maraswami M, Chen Y, Lv X. Recent Developments of Coumarin-based Hybrids in Drug Discovery. Curr Top Med Chem 2022; 22:269-283. [PMID: 34986774 DOI: 10.2174/1568026622666220105105450] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 11/23/2021] [Accepted: 12/05/2021] [Indexed: 11/22/2022]
Abstract
Coumarin scaffold is a highly significant O-heterocycle, namely benzopyran-2-ones, form an elite class of naturally occurring compounds that possess promising therapeutic perspectives. Based on its broad spectrum of biological activities, the privileged coumarin scaffold is applied to medicinal and pharmacological treatments by several rational design strategies and approaches. Structure-activity relationships of the coumarin-based hybrids with various bioactivity fragments revealed significant information toward the further development of highly potent and selective disorder therapeutic agents. The molecular docking studies between coumarins and critical therapeutic enzymes demonstrated mode of action by forming noncovalent interactions with more than one receptor, further rationally confirm information about structure-activity relationships. This review summarizes recent developments relating to coumarin-based hybrids with other pharmacophores aiming to numerous feasible therapeutic enzymatic targets to combat various therapeutic fields, including anticancer, antimicrobic, anti-Alzheimer, anti-inflammatory activities.
Collapse
Affiliation(s)
- Dongguo Xia
- School of Science, Anhui Agricultural University, 230036, Hefei, China
| | - Hao Liu
- School of Science, Anhui Agricultural University, 230036, Hefei, China
| | - Xiang Cheng
- School of Science, Anhui Agricultural University, 230036, Hefei, China
| | - Manikantha Maraswami
- Division of Chemistry and Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University, 637371, Singapore
| | - Yiting Chen
- Fujian Provincial University Engineering Research Center of Green Materials and Chemical Engineering, Minjiang University, 350108, Fuzhou, China
| | - Xianhai Lv
- School of Science, Anhui Agricultural University, 230036, Hefei, China
| |
Collapse
|
17
|
Narendra G, Raju B, Verma H, Sapra B, Silakari O. Multiple machine learning models combined with virtual screening and molecular docking to identify selective human ALDH1A1 inhibitors. J Mol Graph Model 2021; 107:107950. [PMID: 34089986 DOI: 10.1016/j.jmgm.2021.107950] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 05/18/2021] [Accepted: 05/20/2021] [Indexed: 12/19/2022]
Abstract
Aldehyde dehydrogenases (ALDHs) are the enzymes of oxidoreductase family that are responsible for the aldehyde metabolism. The unbalanced expression of these enzymes may be associated with a variety of disease conditions including cancers. ALDH1A1 is one of the isoform of ALDHs majorly overexpressed in a variety of tumors and responsible for the anti-cancer drug resistance. This makes ALDH1A1 as a specific target to develop small molecule ALDH1A1 inhibitors for resistant cancer condition. Number of ALDH1A1 inhibitors have been developed and reported in the literature, but because of non-selectivity and inappropriate pharmacokinetic properties till now none of these have reached in the market for clinical use. Therefore, multiple machine learning models of different isoforms of ALDHs are integrated with in-silico techniques including virtual screening, docking, ADMET profiling, and MD simulation to identify selective ALDH1A1 inhibitors. Total ten selective ALDH1A1 inhibitors with diverse scaffolds and appropriate ADMET were identified that can be further developed as adjuvant therapy in cyclophosphamide and cisplatin resistance cancer.
Collapse
Affiliation(s)
- Gera Narendra
- Molecular Modeling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, 147002, India
| | - Baddipadige Raju
- Molecular Modeling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, 147002, India
| | - Himanshu Verma
- Molecular Modeling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, 147002, India
| | - Bharti Sapra
- Molecular Modeling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, 147002, India
| | - Om Silakari
- Molecular Modeling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, 147002, India.
| |
Collapse
|
18
|
Evaluation of spice and herb as phyto-derived selective modulators of human retinaldehyde dehydrogenases using a simple in vitro method. Biosci Rep 2021; 41:228584. [PMID: 33950219 PMCID: PMC8493444 DOI: 10.1042/bsr20210491] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 04/19/2021] [Accepted: 05/05/2021] [Indexed: 11/17/2022] Open
Abstract
Selective modulation of retinaldehyde dehydrogenases (RALDHs)-the main aldehyde dehydrogenase (ALDH) enzymes converting retinal into retinoic acid (RA), is very important not only in the RA signaling pathway but also for the potential regulatory effects on RALDH isozyme-specific processes and RALDH-related cancers. However, very few selective modulators for RALDHs have been identified, partly due to variable overexpression protocols of RALDHs and insensitive activity assay that needs to be addressed. In the present study, deletion of the N-terminal disordered regions is found to enable simple preparation of all RALDHs and their closest paralog ALDH2 using a single protocol. Fluorescence-based activity assay was employed for enzymatic activity investigation and screening for RALDH-specific modulators from extracts of various spices and herbs that are well-known for containing many phyto-derived anti-cancer constituents. Under the established conditions, spice and herb extracts exhibited differential regulatory effects on RALDHs/ALDH2 with several extracts showing potential selective inhibition of the activity of RALDHs. In addition, the presence of magnesium ions was shown to significantly increase the activity for the natural substrate retinal of RALDH3 but not the others, while His-tag cleavage considerably increased the activity of ALDH2 for the non-specific substrate retinal. Altogether we propose a readily reproducible workflow to find selective modulators for RALDHs and suggest potential sources of selective modulators from spices and herbs.
Collapse
|
19
|
Litwin K, Crowley VM, Suciu RM, Boger DL, Cravatt BF. Chemical proteomic identification of functional cysteines with atypical electrophile reactivities. Tetrahedron Lett 2021; 67. [PMID: 33776155 DOI: 10.1016/j.tetlet.2021.152861] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Cysteine-directed covalent ligands have emerged as a versatile category of chemical probes and drugs that leverage thiol nucleophilicity to form permanent adducts with proteins of interest. Understanding the scope of cysteines that can be targeted by covalent ligands, as well as the types of electrophiles that engage these residues, represent important challenges for fully realizing the potential of cysteine-directed chemical probe discovery. Although chemical proteomic strategies have begun to address these important questions, only a limited number of electrophilic chemotypes have been explored to date. Here, we describe a diverse set of candidate electrophiles appended to a common core 6-methoxy-1,2,3,4-tetrahydroquinoline fragment and evaluate their global cysteine reactivity profiles in human cancer cell proteomes. This work uncovered atypical reactivity patterns for a discrete set of cysteines, including residues involved in enzymatic catalysis and located in proximity to protein-protein interactions. These findings thus point to potentially preferred electrophilic groups for site-selectively targeting functional cysteines in the human proteome.
Collapse
Affiliation(s)
- Kevin Litwin
- The Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA 92307, United States
| | - Vincent M Crowley
- The Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA 92307, United States
| | - Radu M Suciu
- The Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA 92307, United States
| | - Dale L Boger
- The Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA 92307, United States
| | - Benjamin F Cravatt
- The Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA 92307, United States
| |
Collapse
|
20
|
Huddle BC, Grimley E, Chtcherbinine M, Buchman CD, Takahashi C, Debnath B, McGonigal SC, Mao S, Li S, Felton J, Pan S, Wen B, Sun D, Neamati N, Buckanovich RJ, Hurley TD, Larsen SD. Development of 2,5-dihydro-4H-pyrazolo[3,4-d]pyrimidin-4-one inhibitors of aldehyde dehydrogenase 1A (ALDH1A) as potential adjuncts to ovarian cancer chemotherapy. Eur J Med Chem 2020; 211:113060. [PMID: 33341649 DOI: 10.1016/j.ejmech.2020.113060] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 11/21/2020] [Accepted: 11/25/2020] [Indexed: 02/06/2023]
Abstract
There is strong evidence that inhibition of one or more Aldehyde Dehydrogenase 1A (ALDH1A) isoforms may be beneficial in chemotherapy-resistant ovarian cancer and other tumor types. While many previous efforts have focused on development of ALDH1A1 selective inhibitors, the most deadly ovarian cancer subtype, high-grade serous (HGSOC), exhibits elevated expression of ALDH1A3. Herein, we report continued development of pan-ALDH1A inhibitors to assess whether broad spectrum ALDH1A inhibition is an effective adjunct to chemotherapy in this critical tumor subtype. Optimization of the CM39 scaffold, aided by metabolite ID and several new ALDH1A1 crystal structures, led to improved biochemical potencies, improved cellular ALDH inhibition in HGSOC cell lines, and substantial improvements in microsomal stability culminating in orally bioavailable compounds. We demonstrate that two compounds 68 and 69 are able to synergize with chemotherapy in a resistant cell line and patient-derived HGSOC tumor spheroids, indicating their suitability for future in vivo proof of concept experiments.
Collapse
Affiliation(s)
- Brandt C Huddle
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Edward Grimley
- Division of Hematology-Oncology, Departments of Internal Medicine and Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh Medical Center and the Magee-Womens Research Institute, Pittsburgh, PA, 15213, USA
| | - Mikhail Chtcherbinine
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Cameron D Buchman
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Cyrus Takahashi
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Bikash Debnath
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Stacy C McGonigal
- Division of Hematology-Oncology, Departments of Internal Medicine and Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh Medical Center and the Magee-Womens Research Institute, Pittsburgh, PA, 15213, USA
| | - Shuai Mao
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Siwei Li
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Jeremy Felton
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Shu Pan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Bo Wen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Duxin Sun
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Nouri Neamati
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Ronald J Buckanovich
- Division of Hematology-Oncology, Departments of Internal Medicine and Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh Medical Center and the Magee-Womens Research Institute, Pittsburgh, PA, 15213, USA
| | - Thomas D Hurley
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Scott D Larsen
- Vahlteich Medicinal Chemistry Core, College of Pharmacy, University of Michigan, Ann Arbor, MI, 48109, USA; Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
21
|
Yuan B, El Dana F, Ly S, Yan Y, Ruvolo V, Shpall EJ, Konopleva M, Andreeff M, Battula VL. Bone marrow stromal cells induce an ALDH+ stem cell-like phenotype and enhance therapy resistance in AML through a TGF-β-p38-ALDH2 pathway. PLoS One 2020; 15:e0242809. [PMID: 33253299 PMCID: PMC7703975 DOI: 10.1371/journal.pone.0242809] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 11/10/2020] [Indexed: 12/11/2022] Open
Abstract
The bone marrow microenvironment (BME) in acute myeloid leukemia (AML) consists of various cell types that support the growth of AML cells and protect them from chemotherapy. Mesenchymal stromal cells (MSCs) in the BME have been shown to contribute immensely to leukemogenesis and chemotherapy resistance in AML cells. However, the mechanism of stroma-induced chemotherapy resistance is not known. Here, we hypothesized that stromal cells promote a stem-like phenotype in AML cells, thereby inducing tumorigenecity and therapy resistance. To test our hypothesis, we co-cultured AML cell lines and patient samples with BM-derived MSCs and determined aldehyde dehydrogenase (ALDH) activity and performed gene expression profiling by RNA sequencing. We found that the percentage of ALDH+ cells increased dramatically when AML cells were co-cultured with MSCs. However, among the 19 ALDH isoforms, ALDH2 and ALDH1L2 were the only two that were significantly upregulated in AML cells co-cultured with stromal cells compared to cells cultured alone. Mechanistic studies revealed that the transforming growth factor-β1 (TGF-β1)-regulated gene signature is activated in AML cells co-cultured with MSCs. Knockdown of TGF-β1 in BM-MSCs inhibited stroma-induced ALDH activity and ALDH2 expression in AML cells, whereas treatment with recombinant TGF-β1 induced the ALDH+ phenotype in AML cells. We also found that TGF-β1-induced ALDH2 expression in AML cells is mediated by the non-canonical pathway through the activation of p38. Interestingly, inhibition of ALDH2 with diadzin and CVT-10216 significantly inhibited MSC-induced ALDH activity in AML cells and sensitized them to chemotherapy, even in the presence of MSCs. Collectively, BM stroma induces ALDH2 activity in AML cells through the non-canonical TGF-β pathway. Inhibition of ALDH2 sensitizes AML cells to chemotherapy.
Collapse
Affiliation(s)
- Bin Yuan
- Department of Leukemia, Section of Molecular Hematology and Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Fouad El Dana
- Department of Leukemia, Section of Molecular Hematology and Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Stanley Ly
- Department of Leukemia, Section of Molecular Hematology and Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Yuanqing Yan
- Department of Neurosurgery, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Vivian Ruvolo
- Department of Leukemia, Section of Molecular Hematology and Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Elizabeth J. Shpall
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Marina Konopleva
- Department of Leukemia, Section of Molecular Hematology and Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Michael Andreeff
- Department of Leukemia, Section of Molecular Hematology and Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Venkata Lokesh Battula
- Department of Leukemia, Section of Molecular Hematology and Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| |
Collapse
|
22
|
Li B, Yang K, Liang D, Jiang C, Ma Z. Discovery and development of selective aldehyde dehydrogenase 1A1 (ALDH1A1) inhibitors. Eur J Med Chem 2020; 209:112940. [PMID: 33328099 DOI: 10.1016/j.ejmech.2020.112940] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 09/29/2020] [Accepted: 10/11/2020] [Indexed: 12/24/2022]
Abstract
ALDH1A1, one important member of 19 ALDHs, can metabolize reactive aldehydes to their corresponding carboxylic acid derivatives and play important physiological and toxicological roles in many areas, including CNS, metabolic disorders, and cancers. Overexpression of ALDH1A1 correlates with poor prognosis and tumor aggressiveness, is associated with drug resistance in traditional chemotherapy for cancer treatment and contributes to obesity, diabetes, and inflammation. So, inhibition of ALDH1A1 may offer new therapeutic options for patients with cancer, obesity, diabetes, and inflammation. Up to now, many ALDH1A1 inhibitors with different scaffolds have been identified and developed as useful chemical tools for better understanding of the functions of ALDH1A1 in physiologic and pathophysiologic conditions. In this review, the advances in the discovery and development of selective ALDH1A1 inhibitors are summarized, and opportunities and challenges associated with this field are also discussed.
Collapse
Affiliation(s)
- Bingyan Li
- Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Tongjiaxiang 24, Nanjing, 210009, China; Department of Medicinal Chemistry, China Pharmaceutical University, Tongjiaxiang 24, Nanjing, 210009, China
| | - Kang Yang
- Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Tongjiaxiang 24, Nanjing, 210009, China
| | - Dailin Liang
- Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Tongjiaxiang 24, Nanjing, 210009, China; Department of Medicinal Chemistry, China Pharmaceutical University, Tongjiaxiang 24, Nanjing, 210009, China
| | - Cheng Jiang
- Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Tongjiaxiang 24, Nanjing, 210009, China; Department of Medicinal Chemistry, China Pharmaceutical University, Tongjiaxiang 24, Nanjing, 210009, China.
| | - Zonghui Ma
- Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Tongjiaxiang 24, Nanjing, 210009, China; Department of Medicinal Chemistry, China Pharmaceutical University, Tongjiaxiang 24, Nanjing, 210009, China.
| |
Collapse
|
23
|
Buhimschi AD, Gooden DM, Jing H, Fels DR, Hansen KS, Beyer WF, Dewhirst MW, Walder H, Gasparro FP. Psoralen Derivatives with Enhanced Potency. Photochem Photobiol 2020; 96:1014-1031. [PMID: 32221980 DOI: 10.1111/php.13263] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Accepted: 02/28/2020] [Indexed: 12/22/2022]
Abstract
Psoralen is a furocoumarin natural product that intercalates within DNA and forms covalent adducts when activated by ultraviolet radiation. It is well known that this property contributes to psoralen's clinical efficacy in several disease contexts, which include vitiligo, psoriasis, graft-versus-host disease and cutaneous T-cell lymphoma. Given the therapeutic relevance of psoralen and its derivatives, we attempted to synthesize psoralens with even greater potency. In this study, we report a library of 73 novel psoralens, the largest collection of its kind. When screened for the ability to reduce cell proliferation, we identified two derivatives even more cytotoxic than 4'-aminomethyl-4,5',8-trimethylpsoralen (AMT), one of the most potent psoralens identified to date. Using MALDI-TOF MS, we studied the DNA adduct formation for a subset of novel psoralens and found that in most cases enhanced DNA binding correlated well with cytotoxicity. Generally, our most potent derivatives contain positively charged substituents, which we believe increase DNA affinity and enhance psoralen intercalation. Thus, we provide a rational approach to guide efforts toward further optimizing psoralens to fully capitalize on this drug class' therapeutic potential. Finally, the structure-activity insights we have gained shed light on several opportunities to study currently underappreciated aspects of psoralen's mechanism.
Collapse
Affiliation(s)
- Alexandru D Buhimschi
- Medical Scientist Training Program, Northwestern University Feinberg School of Medicine, Chicago, IL.,Photobiology Laboratory, Hamden Hall Country Day School, Hamden, CT
| | | | - Hongwu Jing
- Department of Obstetrics and Gynecology, University of Illinois College of Medicine, Chicago, IL
| | - Diane R Fels
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC.,Duke Cancer Institute, Duke University Medical Center, Durham, NC
| | - Katherine S Hansen
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC.,Duke Cancer Institute, Duke University Medical Center, Durham, NC
| | | | - Mark W Dewhirst
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC.,Duke Cancer Institute, Duke University Medical Center, Durham, NC
| | | | | |
Collapse
|
24
|
Vaz WF, Custodio JMF, D'Oliveira GDC, Neves BJ, Junior PSC, Filho JTM, Andrade CH, Perez CN, Silveira-Lacerda EP, Napolitano HB. Dihydroquinoline derivative as a potential anticancer agent: synthesis, crystal structure, and molecular modeling studies. Mol Divers 2020; 25:55-66. [PMID: 31900682 DOI: 10.1007/s11030-019-10024-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Accepted: 12/05/2019] [Indexed: 01/16/2023]
Abstract
Cancer is one of the leading causes of death worldwide and requires intense and growing research investments from the public and private sectors. This is expected to lead to the development of new medicines. A determining factor in this process is the structural understanding of molecules with potential anticancer properties. Since the major compounds used in cancer therapies fail to encompass every spectrum of this disease, there is a clear need to research new molecules for this purpose. As it follows, we have studied the class of quinolinones that seem effective for such therapy. This paper describes the structural elucidation of a novel dihydroquinoline by single-crystal X-ray diffraction and spectroscopy characterization. Topology studies were carried through Hirshfeld surfaces analysis and molecular electrostatic potential map; electronic stability was evaluated from the calculated energy of frontier molecular orbitals. Additionally, in silico studies by molecular docking indicated that this dihydroquinoline could act as an anticancer agent due to their higher binding affinity with human aldehyde dehydrogenase 1A1 (ALDH 1A1). Tests in vitro were performed for VERO (normal human skin keratinocytes), B16F10 (mouse melanoma), and MDA-MB-231 (metastatic breast adenocarcinoma), and the results certified that compound as a potential anticancer agent. A Dihydroquinoline derivative was tested against three cancer cell lines and the results attest that compound as potential anticancer agent.
Collapse
Affiliation(s)
- W F Vaz
- Universidade Estadual de Goiás, Anápolis, GO, 75132-400, Brazil.
- Instituto Federal de Educação, Ciência e Tecnologia de Mato Grosso, Lucas do Rio Verde, MT, 78455-000, Brazil.
| | - J M F Custodio
- Universidade Federal de Goiás, Goiânia, GO, 74690-900, Brazil
| | | | - B J Neves
- LabMol, Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, GO, 74605-170, Brazil
| | - P S C Junior
- Universidade Federal de Mato Grosso do Sul, Nova Andradina, MS, 79750-000, Brazil
| | - J T M Filho
- Universidade Federal de Goiás, Goiânia, GO, 74690-900, Brazil
| | - C H Andrade
- LabMol, Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, GO, 74605-170, Brazil
| | - C N Perez
- Universidade Federal de Goiás, Goiânia, GO, 74690-900, Brazil
| | - E P Silveira-Lacerda
- Laboratório de Genética Molecular e Citogenética, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia, GO, 74605-170, Brazil
| | - H B Napolitano
- Universidade Estadual de Goiás, Anápolis, GO, 75132-400, Brazil
| |
Collapse
|
25
|
Michorowska S, Giebułtowicz J, Wolinowska R, Konopka A, Wilkaniec A, Krajewski P, Bulska E, Wroczyński P. Detection of ALDH3B2 in Human Placenta. Int J Mol Sci 2019; 20:E6292. [PMID: 31847104 PMCID: PMC6941052 DOI: 10.3390/ijms20246292] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 12/11/2019] [Accepted: 12/11/2019] [Indexed: 01/28/2023] Open
Abstract
Aldehyde dehydrogenase 3B2 (ALDH3B2) gene contains a premature termination codon, which can be skipped or suppressed resulting in full-length protein expression. Alternatively, the longest putative open reading frame starting with the second in-frame start codon would encode short isoform. No unequivocal evidence of ALDH3B2 expression in healthy human tissues is available. The aim of this study was to confirm its expression in human placenta characterized by the highest ALDH3B2 mRNA abundance. ALDH3B2 DNA and mRNA were sequenced. The expression was investigated using western blot. The identity of the protein was confirmed using mass spectrometry (MS). The predicted tertiary and quaternary structures, subcellular localization, and phosphorylation sites were assessed using bioinformatic analyses. All DNA and mRNA isolates contained the premature stop codon. In western blot analyses, bands corresponding to the mass of full-length protein were detected. MS analysis led to the identification of two unique peptides, one of which is encoded by the nucleotide sequence located upstream the second start codon. Bioinformatic analyses suggest cytoplasmic localization and several phosphorylation sites. Despite premature stop codon in DNA and mRNA sequences, full-length ALDH3B2 was found. It can be formed as a result of premature stop codon readthrough, complex phenomenon enabling stop codon circumvention.
Collapse
Affiliation(s)
- Sylwia Michorowska
- Department of Bioanalysis and Drug Analysis, Faculty of Pharmacy, Medical University of Warsaw, 02-097 Warsaw, Poland; (J.G.); (P.W.)
| | - Joanna Giebułtowicz
- Department of Bioanalysis and Drug Analysis, Faculty of Pharmacy, Medical University of Warsaw, 02-097 Warsaw, Poland; (J.G.); (P.W.)
| | - Renata Wolinowska
- Department of Pharmaceutical Microbiology, Centre for Preclinical Research and Technology (CePT), Faculty of Pharmacy, Medical University of Warsaw, 02-097 Warsaw, Poland;
| | - Anna Konopka
- Biological and Chemical Research Centre, Faculty of Chemistry, University of Warsaw, 02-097 Warsaw, Poland; (A.K.); (E.B.)
| | - Anna Wilkaniec
- Department of Cellular Signaling, Mossakowski Research Centre, Polish Academy of Sciences, Pawińskiego 5, 02-106 Warsaw, Poland;
| | - Paweł Krajewski
- Forensic Medicine Department, First Faculty of Medicine, Medical University of Warsaw, 02-097 Warsaw, Poland;
| | - Ewa Bulska
- Biological and Chemical Research Centre, Faculty of Chemistry, University of Warsaw, 02-097 Warsaw, Poland; (A.K.); (E.B.)
| | - Piotr Wroczyński
- Department of Bioanalysis and Drug Analysis, Faculty of Pharmacy, Medical University of Warsaw, 02-097 Warsaw, Poland; (J.G.); (P.W.)
| |
Collapse
|
26
|
Liang D, Fan Y, Yang Z, Zhang Z, Liu M, Liu L, Jiang C. Discovery of coumarin-based selective aldehyde dehydrogenase 1A1 inhibitors with glucose metabolism improving activity. Eur J Med Chem 2019; 187:111923. [PMID: 31816557 DOI: 10.1016/j.ejmech.2019.111923] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Revised: 11/28/2019] [Accepted: 11/28/2019] [Indexed: 02/06/2023]
Abstract
Overexpression of aldehyde dehydrogenase 1A1 (ALDH1A1) is associated with the occurrence and development of obesity and insulin resistance. Herein, a series of coumarin-based ALDH1A1 inhibitors were designed, synthesized and evaluated. Among them, compounds 10, 14 and 26 exhibited potent inhibitory activity against ALDH1A1 and high selectivity over ALDH1A2, ALDH1A3, ALDH2 and ALDH3A1. Optimized compound 10 showed markedly improved pharmacokinetic characters and ADME profiles comparing to the lead compound 1. In vitro study demonstrated that 10 alleviated palmitic acid-induced impairment of glucose consumption in HepG2 cells.
Collapse
Affiliation(s)
- Dailin Liang
- Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Tongjiaxiang 24, Nanjing, 210009, China; Department of Medicinal Chemistry, China Pharmaceutical University, Tongjiaxiang 24, Nanjing, 210009, China
| | - Yazhou Fan
- Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Tongjiaxiang 24, Nanjing, 210009, China
| | - Zhou Yang
- Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Tongjiaxiang 24, Nanjing, 210009, China; Department of Medicinal Chemistry, China Pharmaceutical University, Tongjiaxiang 24, Nanjing, 210009, China
| | - Zhenguo Zhang
- Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Tongjiaxiang 24, Nanjing, 210009, China; Department of Medicinal Chemistry, China Pharmaceutical University, Tongjiaxiang 24, Nanjing, 210009, China
| | - Meiyang Liu
- Department of Medicinal Chemistry, China Pharmaceutical University, Tongjiaxiang 24, Nanjing, 210009, China
| | - Li Liu
- Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Tongjiaxiang 24, Nanjing, 210009, China.
| | - Cheng Jiang
- Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Tongjiaxiang 24, Nanjing, 210009, China; Department of Medicinal Chemistry, China Pharmaceutical University, Tongjiaxiang 24, Nanjing, 210009, China.
| |
Collapse
|
27
|
Carbone A, Montalbano A, Spanò V, Musante I, Galietta LJV, Barraja P. Furocoumarins as multi-target agents in the treatment of cystic fibrosis. Eur J Med Chem 2019; 180:283-290. [PMID: 31319264 DOI: 10.1016/j.ejmech.2019.07.025] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 07/07/2019] [Accepted: 07/07/2019] [Indexed: 02/06/2023]
Abstract
Multi-target molecular entities, offer a path to progress both in understanding causes of disease and in defining effective small molecule treatments. Coumarin and its derivatives belong to an important group of natural compounds with diverse biological properties. They are found in vegetables and plants for which literature reports thousands of publications for the great variety of biological applications among which the photoprotective effects, thus being considered multi-targeting agents. Their furan condensed analogues constitute the family of furocoumarins, less represented in the literature, endowed with photosensitizing properties and often used for the treatment of skin diseases such as vitiligo and psoriasis. Despite the study of biological properties of linear and angular furocumarins dates back to ancient times, mainly as photosensitizers, these small molecules still represent an attractive scaffold for further development and applications in several therapeutic fields. The aim of the present review is to summarize the most promising chemical entities belonging to the class of furocumarins and coumarins, emerged in the last decades, and the methods used for their synthesis with a particular focus on main targets involved in the cystic fibrosis treatment.
Collapse
Affiliation(s)
- Anna Carbone
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Via Archirafi 32, 90123, Palermo, Italy
| | - Alessandra Montalbano
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Via Archirafi 32, 90123, Palermo, Italy
| | - Virginia Spanò
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Via Archirafi 32, 90123, Palermo, Italy
| | - Ilaria Musante
- Telethon Institute of Genetics and Medicine (TIGEM), Campi Flegrei 34, 80078, Pozzuoli, NA, Italy
| | - Luis J V Galietta
- Telethon Institute of Genetics and Medicine (TIGEM), Campi Flegrei 34, 80078, Pozzuoli, NA, Italy; Department of Translational Medical Sciences (DISMET), University of Naples, "Federico II", 80131, Naples, Italy
| | - Paola Barraja
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Via Archirafi 32, 90123, Palermo, Italy.
| |
Collapse
|
28
|
Verma H, Singh Bahia M, Choudhary S, Kumar Singh P, Silakari O. Drug metabolizing enzymes-associated chemo resistance and strategies to overcome it. Drug Metab Rev 2019; 51:196-223. [DOI: 10.1080/03602532.2019.1632886] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Affiliation(s)
- Himanshu Verma
- MolecularModelling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| | | | - Shalki Choudhary
- MolecularModelling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| | - Pankaj Kumar Singh
- MolecularModelling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| | - Om Silakari
- MolecularModelling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| |
Collapse
|
29
|
Natural and semisynthetic oxyprenylated aromatic compounds as stimulators or inhibitors of melanogenesis. Bioorg Chem 2019; 87:181-190. [DOI: 10.1016/j.bioorg.2019.03.026] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 03/08/2019] [Accepted: 03/13/2019] [Indexed: 12/11/2022]
|
30
|
Pan L, Lei D, Jin L, He Y, Yang Q. Promising Fungicides from Allelochemicals: Synthesis of Umbelliferone Derivatives and Their Structure⁻Activity Relationships. Molecules 2018; 23:molecules23113002. [PMID: 30453559 PMCID: PMC6278345 DOI: 10.3390/molecules23113002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Revised: 11/07/2018] [Accepted: 11/12/2018] [Indexed: 01/02/2023] Open
Abstract
Umbelliferone was discovered to be an important allelochemical in our previous study, but the contribution of its activity and structure has not yet been revealed. In this study, a series of analogues were synthesized to determine the skeleton of umbelliferone and examine its fungicidal activity. Furthermore, targeted modifications were conducted with three plant parasitic fungi to examine the lead compounds. Among those tested, compounds 2f and 10 were found to show excellent antifungal activity with an inhibitory rate over 80% at 100 ug/mL. The study proves that umbelliferone can be a promising skeleton for fungicides discovery. In addition, the primary structure–activity relationship provides a good guidance for the discovery of novel fungicides based on natural products in the future.
Collapse
Affiliation(s)
- Le Pan
- Chemical Engineering College, Xinjiang Agricultural University, Urumqi 830052, China.
| | - Dongyu Lei
- Department of Physiology, Preclinical School, Xinjiang Medical University, Urumqi 830011, China.
| | - Lu Jin
- Chemical Engineering College, Xinjiang Agricultural University, Urumqi 830052, China.
| | - Yuan He
- Chemical Engineering College, Xinjiang Agricultural University, Urumqi 830052, China.
| | - Qingqing Yang
- Chemical Engineering College, Xinjiang Agricultural University, Urumqi 830052, China.
| |
Collapse
|
31
|
Huddle BC, Grimley E, Buchman CD, Chtcherbinine M, Debnath B, Mehta P, Yang K, Morgan CA, Li S, Felton J, Sun D, Mehta G, Neamati N, Buckanovich RJ, Hurley TD, Larsen SD. Structure-Based Optimization of a Novel Class of Aldehyde Dehydrogenase 1A (ALDH1A) Subfamily-Selective Inhibitors as Potential Adjuncts to Ovarian Cancer Chemotherapy. J Med Chem 2018; 61:8754-8773. [PMID: 30221940 DOI: 10.1021/acs.jmedchem.8b00930] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Aldehyde dehydrogenase (ALDH) activity is commonly used as a marker to identify cancer stem-like cells. The three ALDH1A isoforms have all been individually implicated in cancer stem-like cells and in chemoresistance; however, which isoform is preferentially expressed varies between cell lines. We sought to explore the structural determinants of ALDH1A isoform selectivity in a series of small-molecule inhibitors in support of research into the role of ALDH1A in cancer stem cells. An SAR campaign guided by a cocrystal structure of the HTS hit CM39 (7) with ALDH1A1 afforded first-in-class inhibitors of the ALDH1A subfamily with excellent selectivity over the homologous ALDH2 isoform. We also discovered the first reported modestly selective single isoform 1A2 and 1A3 inhibitors. Two compounds, 13g and 13h, depleted the CD133+ putative cancer stem cell pool, synergized with cisplatin, and achieved efficacious concentrations in vivo following IP administration. Compound 13h additionally synergized with cisplatin in a patient-derived ovarian cancer spheroid model.
Collapse
Affiliation(s)
| | | | - Cameron D Buchman
- Department of Biochemistry and Molecular Biology , Indiana University School of Medicine , Indianapolis , Indiana 46202 , United States
| | - Mikhail Chtcherbinine
- Department of Biochemistry and Molecular Biology , Indiana University School of Medicine , Indianapolis , Indiana 46202 , United States
| | | | - Pooja Mehta
- Department of Materials Science Engineering , University of Michigan , Ann Arbor , Michigan 48109 , United States
| | - Kun Yang
- Division of Hematology Oncology, Department of Internal Medicine , University of Michigan , Ann Arbor , Michigan 48109 , United States
| | - Cynthia A Morgan
- Department of Biochemistry and Molecular Biology , Indiana University School of Medicine , Indianapolis , Indiana 46202 , United States
| | - Siwei Li
- Department of Pharmaceutical Sciences, College of Pharmacy ; University of Michigan , Ann Arbor , Michigan 48109 , United States
| | - Jeremy Felton
- Department of Pharmaceutical Sciences, College of Pharmacy ; University of Michigan , Ann Arbor , Michigan 48109 , United States
| | - Duxin Sun
- Department of Pharmaceutical Sciences, College of Pharmacy ; University of Michigan , Ann Arbor , Michigan 48109 , United States
| | - Geeta Mehta
- Department of Materials Science Engineering , University of Michigan , Ann Arbor , Michigan 48109 , United States.,Department of Biomedical Engineering , University of Michigan , Ann Arbor , Michigan 48109 , United States.,Macromolecular Science and Engineering , University of Michigan , Ann Arbor , Michigan 48109 , United States
| | | | - Ronald J Buckanovich
- Division of Hematology Oncology, Department of Internal Medicine , University of Michigan , Ann Arbor , Michigan 48109 , United States
| | - Thomas D Hurley
- Department of Biochemistry and Molecular Biology , Indiana University School of Medicine , Indianapolis , Indiana 46202 , United States
| | | |
Collapse
|
32
|
Chen Y, Zhu JY, Ho Hong K, Mikles DC, Georg GI, Goldstein AS, Amory JK, Schönbrunn E. Structural Basis of ALDH1A2 Inhibition by Irreversible and Reversible Small Molecule Inhibitors. ACS Chem Biol 2018; 13:582-590. [PMID: 29240402 PMCID: PMC6089219 DOI: 10.1021/acschembio.7b00685] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Enzymes of the ALDH1A subfamily of aldehyde dehydrogenases are crucial in regulating retinoic acid (RA) signaling and have received attention as potential drug targets. ALDH1A2 is the primary RA-synthesizing enzyme in mammalian spermatogenesis and is therefore considered a viable drug target for male contraceptive development. However, only a small number of ALDH1A2 inhibitors have been reported, and information on the structure of ALDH1A2 was limited to the NAD-liganded enzyme void of substrate or inhibitors. Herein, we describe the mechanism of action of structurally unrelated reversible and irreversible inhibitors of human ALDH1A2 using direct binding studies and X-ray crystallography. All inhibitors bind to the active sites of tetrameric ALDH1A2. Compound WIN18,446 covalently reacts with the side chain of the catalytic residue Cys320, resulting in a chiral adduct in ( R) configuration. The covalent adduct directly affects the neighboring NAD molecule, which assumes a contracted conformation suboptimal for the dehydrogenase reaction. The reversible inhibitors interact predominantly through direct hydrogen bonding interactions with residues in the vicinity of Cys320 without affecting NAD. Upon interaction with inhibitors, a large flexible loop assumes regular structure, thereby shielding the active site from solvent. The precise knowledge of the binding modes provides a new framework for the rational design of novel inhibitors of ALDH1A2 with improved potency and selectivity profiles.
Collapse
Affiliation(s)
- Yan Chen
- Drug Discovery Department, Moffitt Cancer Center, Tampa, Florida 33612, United States
| | - Jin-Yi Zhu
- Drug Discovery Department, Moffitt Cancer Center, Tampa, Florida 33612, United States
- Present Address Dart Neuroscience, San Diego, CA 92131, USA
| | - Kwon Ho Hong
- Department of Medicinal Chemistry and Institute for Therapeutics Discovery and Development, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota 55414, United States
| | - David C. Mikles
- Drug Discovery Department, Moffitt Cancer Center, Tampa, Florida 33612, United States
| | - Gunda I. Georg
- Department of Medicinal Chemistry and Institute for Therapeutics Discovery and Development, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota 55414, United States
| | | | - John K Amory
- Department of Medicine, University of Washington, Seattle, Washington 98195, United States
| | - Ernst Schönbrunn
- Drug Discovery Department, Moffitt Cancer Center, Tampa, Florida 33612, United States
| |
Collapse
|
33
|
Abdul W, Aliyu SR, Lin L, Sekete M, Chen X, Otieno FJ, Yang T, Lin Y, Norvienyeku J, Wang Z. Family-Four Aldehyde Dehydrogenases Play an Indispensable Role in the Pathogenesis of Magnaporthe oryzae. FRONTIERS IN PLANT SCIENCE 2018; 9:980. [PMID: 30135691 PMCID: PMC6092734 DOI: 10.3389/fpls.2018.00980] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Accepted: 06/15/2018] [Indexed: 05/22/2023]
Abstract
The oxidative degradation of lipids through lipid peroxidation processes results in the generation of free fatty acid radicals. These free radicals including reactive oxygen species (ROS) serve as a substrate for generating reactive aldehydes. The accumulation of free fatty acid radicals, ROS, and reactive aldehydes in cell compartments beyond physiological threshold levels tends to exert a damaging effect on proximal membranes and distal tissues. Living organisms deploy a wide array of efficient enzymes including superoxide dismutase (SOD), catalase (CAT), peroxidase (POD), and aldehyde dehydrogenases (ALDHs) for scavenging reactive molecules and intermediates produced from membrane lipid peroxidation events. Although the contributions of SOD, CAT, and POD to the pathogenesis of microbial plant pathogens are well known, the influence of ALDH genes on the morphological and infectious development of plant pathogenic microbes is not well understood. In this study, we deployed RNA interference (RNAi) techniques and successfully silenced two putative family-four aldehyde dehydrogenase genes potassium-activated aldehyde dehydrogenase (MoKDCDH) and delta-1-pyrrorine-5-carboxylate dehydrogenase (MoP5CDH) in the rice blast pathogen Magnaporthe oryzae. The results obtained from the phenotypic analysis of individual knock-down strains showed that the RNAi-mediated inactivation of MoKDCDH and MoP5CDH triggered a significant reduction in conidiogenesis and vegetative growth of ΔMokdcdh and ΔMop5cdh strains. We further observed that downregulating the expression of MoKDCDH and MoP5CDH severely compromised the pathogenesis of the rice blast fungus. Also, the disruption of MoKDCDH and MoP5CDH M. oryzae undermined membrane integrity and rendered the mutant strains highly sensitive to membrane stress inducing osmolytes. However, the MoKDCDH and MoP5CDH knock-down strains generated in this study displayed unaltered cell wall integrity and thus suggested that family-four ALDHs play a dispensable role in enforcing cell wall-directed stress tolerance in M. oryzae. From these results, we deduced that family-four ALDHs play a conserved role in fostering membrane integrity in M. oryzae possibly by scavenging reactive aldehydes, fatty acid radicals, and other alcohol derivatives. The observation that downregulating the expression activities of MoKDCDH had a lethal effect on potential mutants further emphasized the need for comprehensive and holistic evaluation of the numerous ALDHs amassed by the rice blast fungus for their possible engagement as suitable targets as antiblast agents.
Collapse
Affiliation(s)
- Waheed Abdul
- Fujian University Key Laboratory for Functional Genomics of Plant Fungal Pathogens, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
- Fujian and Taiwan Joint Center for Ecological Control of Crop Pests, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Sami R. Aliyu
- Fujian and Taiwan Joint Center for Ecological Control of Crop Pests, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Lili Lin
- Fujian and Taiwan Joint Center for Ecological Control of Crop Pests, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Malota Sekete
- Department of Crop Science, Faculty of Agriculture, National University of Lesotho, Lesotho, Southern Africa
| | - Xiaomin Chen
- Fujian and Taiwan Joint Center for Ecological Control of Crop Pests, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Frankline J. Otieno
- Fujian University Key Laboratory for Functional Genomics of Plant Fungal Pathogens, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Tao Yang
- Fujian and Taiwan Joint Center for Ecological Control of Crop Pests, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Yahong Lin
- Fujian University Key Laboratory for Functional Genomics of Plant Fungal Pathogens, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Justice Norvienyeku
- Fujian University Key Laboratory for Functional Genomics of Plant Fungal Pathogens, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
- Fujian and Taiwan Joint Center for Ecological Control of Crop Pests, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, China
- *Correspondence: Justice Norvienyeku, Zonghua Wang,
| | - Zonghua Wang
- Fujian University Key Laboratory for Functional Genomics of Plant Fungal Pathogens, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
- Fujian and Taiwan Joint Center for Ecological Control of Crop Pests, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, China
- Institute of Oceanography, Minjiang University, Fuzhou, China
- *Correspondence: Justice Norvienyeku, Zonghua Wang,
| |
Collapse
|