1
|
Zixin Y, Lulu C, Xiangchang Z, Qing F, Binjie Z, Chunyang L, Tai R, Dongsheng O. TMAO as a potential biomarker and therapeutic target for chronic kidney disease: A review. Front Pharmacol 2022; 13:929262. [PMID: 36034781 PMCID: PMC9411716 DOI: 10.3389/fphar.2022.929262] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 06/30/2022] [Indexed: 11/13/2022] Open
Abstract
The gut microbiota and its metabolites have become a hotspot of recent research. Trimethylamine N-oxide (TMAO) metabolized by the gut microbiota is closely related to many diseases such as cardiovascular disease, chronic kidney disease, type 2 diabetes, etc. Chronic kidney disease (CKD) is an important contributor to morbidity and mortality from non-communicable diseases. Recently, increasing focus has been put on the role of TMAO in the development and progress of chronic kidney disease. The level of TMAO in patients with chronic kidney disease is significantly increased, and a high level of TMAO deteriorates chronic kidney disease. This article describes the relationship between TMAO and chronic kidney disease and the research progress of drugs targeted TMAO, providing a reference for the development of anti-chronic kidney disease drugs targeted TMAO.
Collapse
Affiliation(s)
- Ye Zixin
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Changsha, China
| | - Chen Lulu
- Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha, China
- Department of Clinical Pharmacy, Affiliated Hospital of Xiangnan University, Chenzhou, China
| | - Zeng Xiangchang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Changsha, China
| | - Fang Qing
- Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha, China
| | - Zheng Binjie
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Changsha, China
| | - Luo Chunyang
- Department of Clinical Pharmacy, Affiliated Hospital of Xiangnan University, Chenzhou, China
| | - Rao Tai
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Changsha, China
| | - Ouyang Dongsheng
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Changsha, China
- Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha, China
| |
Collapse
|
2
|
George N, Jawaid Akhtar M, Al Balushi KA, Alam Khan S. Rational drug design strategies for the development of promising multi-target directed indole hybrids as Anti-Alzheimer agents. Bioorg Chem 2022; 127:105941. [PMID: 35714473 DOI: 10.1016/j.bioorg.2022.105941] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 06/03/2022] [Accepted: 06/07/2022] [Indexed: 11/26/2022]
Abstract
Alzheimer's disease (AD) is a neurological disorder that leads to dementia i.e., progressive memory loss accompanied with worsening of thinking ability of an individual. The cause of AD is not fully understood but it progresses with age where brain cells gradually die over time. According to the World Health Organization (WHO), currently 50 million people worldwide are affected by dementia and 60-70% of the cases belong to AD. Cumulative research over the past few decades have shown that molecules that act at a single target possess limited efficacy since these investigational drugs are not able to act against complex pathologies and thus do not provide permanent cure. Designing of multi-target directed ligands (MTDLs) appears to be more beneficial and a rational approach to treat chronic complex diseases including neurodegenerative diseases. Recently, MTDLs are being extensively researched by the medicinal chemists for the development of drugs for the treatment of various multifactorial diseases. Indole is one of the privileged scaffolds which is considered as an essential mediator between the gut-brain axis because of its neuroprotective, anti-inflammatory, β-amyloid anti-aggregation and antioxidant activities. Herein, we have reviewed the potential of some indole-hybrids acting at multiple targets in the pathogenesis of AD. We have reviewed research articles from the year 2014-2021 from various scientific databases and highlighted the synthetic strategies, mechanisms of neuroprotection, toxicity, structure activity relationships and molecular docking studies of various indole-hybrid derivatives. This literature review of published data on indole derivatives indicated that developing indole hybrids have improved the pharmacokinetic profile with lower toxicity, provided synergistic effect, helped to develop more potent compounds and prevented drug-drug interactions. It is evident that this class of compounds have potential to inhibit multiple enzymes targets involved in the pathogenesis of AD and therefore indole hybrids as MTDLs may play an important role in the development of anti-AD molecules.
Collapse
Affiliation(s)
- Namy George
- College of Pharmacy, National University of Science and Technology, PO Box 620, PC 130, Muscat, Oman
| | - Md Jawaid Akhtar
- College of Pharmacy, National University of Science and Technology, PO Box 620, PC 130, Muscat, Oman
| | - Khalid A Al Balushi
- College of Pharmacy, National University of Science and Technology, PO Box 620, PC 130, Muscat, Oman
| | - Shah Alam Khan
- College of Pharmacy, National University of Science and Technology, PO Box 620, PC 130, Muscat, Oman.
| |
Collapse
|
3
|
Burhop A, Bag S, Grigalunas M, Woitalla S, Bodenbinder P, Brieger L, Strohmann C, Pahl A, Sievers S, Waldmann H. Synthesis of Indofulvin Pseudo-Natural Products Yields a New Autophagy Inhibitor Chemotype. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2102042. [PMID: 34346568 PMCID: PMC8498912 DOI: 10.1002/advs.202102042] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 07/14/2021] [Indexed: 06/01/2023]
Abstract
Chemical and biological limitations in bioactive compound design based on natural product (NP) structure can be overcome by the combination of NP-derived fragments in unprecedented arrangements to afford "pseudo-natural products" (pseudo-NPs). A new pseudo-NP design principle is described, i.e., the combination of NP-fragments by transformations that are not part of current biosynthesis pathways. A collection of indofulvin pseudo-NPs is obtained from 2-hydroxyethyl-indoles and ketones derived from the fragment-sized NP griseofulvin by means of an iso-oxa-Pictet-Spengler reaction. Cheminformatic analysis indicates that the indofulvins reside in an area of chemical space sparsely covered by NPs, drugs, and drug-like compounds and they may combine favorable properties of these compound classes. Biological evaluation of the compound collection in different cell-based assays and the unbiased high content cell painting assay reveal that the indofulvins define a new autophagy inhibitor chemotype that targets mitochondrial respiration.
Collapse
Affiliation(s)
- Annina Burhop
- Max Planck Institute of Molecular PhysiologyDepartment of Chemical BiologyDortmund44227Germany
- Technical University DortmundFaculty of ChemistryChemical BiologyDortmund44227Germany
| | - Sukdev Bag
- Max Planck Institute of Molecular PhysiologyDepartment of Chemical BiologyDortmund44227Germany
| | - Michael Grigalunas
- Max Planck Institute of Molecular PhysiologyDepartment of Chemical BiologyDortmund44227Germany
| | - Sophie Woitalla
- Max Planck Institute of Molecular PhysiologyDepartment of Chemical BiologyDortmund44227Germany
- Technical University DortmundFaculty of ChemistryChemical BiologyDortmund44227Germany
| | - Pia Bodenbinder
- Max Planck Institute of Molecular PhysiologyDepartment of Chemical BiologyDortmund44227Germany
- Technical University DortmundFaculty of ChemistryChemical BiologyDortmund44227Germany
| | - Lukas Brieger
- Technical University DortmundFaculty of ChemistryInorganic ChemistryDortmund44227Germany
| | - Carsten Strohmann
- Technical University DortmundFaculty of ChemistryInorganic ChemistryDortmund44227Germany
| | - Axel Pahl
- Max Planck Institute of Molecular PhysiologyDepartment of Chemical BiologyDortmund44227Germany
- Compound Management and Screening CenterDortmund44227Germany
| | - Sonja Sievers
- Max Planck Institute of Molecular PhysiologyDepartment of Chemical BiologyDortmund44227Germany
- Compound Management and Screening CenterDortmund44227Germany
| | - Herbert Waldmann
- Max Planck Institute of Molecular PhysiologyDepartment of Chemical BiologyDortmund44227Germany
- Technical University DortmundFaculty of ChemistryChemical BiologyDortmund44227Germany
| |
Collapse
|
4
|
Pecnard S, Hamze A, Pozzo JL, Alami M, Provot O. Synthesis of Oxazino[4,3-a]indoles and biological applications. Eur J Med Chem 2021; 224:113728. [PMID: 34340043 DOI: 10.1016/j.ejmech.2021.113728] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/20/2021] [Accepted: 07/27/2021] [Indexed: 12/23/2022]
Abstract
This review brings together the various pathways to the oxazino[4,3-a]indole motif over the last decades. Representative examples showing the scope of these processes will illustrate the synthetic pathways and the biological activity of the synthesized oxazinoindoles will be mentioned wherever possible.
Collapse
Affiliation(s)
- Shannon Pecnard
- Université Paris-Saclay, CNRS, BioCIS, 92290, Châtenay-Malabry, France
| | - Abdallah Hamze
- Université Paris-Saclay, CNRS, BioCIS, 92290, Châtenay-Malabry, France
| | - Jean-Luc Pozzo
- Univ. Bordeaux, CNRS, Bordeaux INP, ISM, UMR5255, 351 cours Libération, F-33405, Bordeaux, France
| | - Mouad Alami
- Université Paris-Saclay, CNRS, BioCIS, 92290, Châtenay-Malabry, France
| | - Olivier Provot
- Université Paris-Saclay, CNRS, BioCIS, 92290, Châtenay-Malabry, France.
| |
Collapse
|
5
|
Steinke I, Ghanei N, Govindarajulu M, Yoo S, Zhong J, Amin RH. Drug Discovery and Development of Novel Therapeutics for Inhibiting TMAO in Models of Atherosclerosis and Diabetes. Front Physiol 2020; 11:567899. [PMID: 33192565 PMCID: PMC7658318 DOI: 10.3389/fphys.2020.567899] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 09/23/2020] [Indexed: 12/19/2022] Open
Abstract
Diabetes mellitus exists as a comorbidity with congestive heart failure (CHF). However, the exact molecular signaling mechanism linking CHF as the major form of mortality from diabetes remains unknown. Type 2 diabetic patients display abnormally high levels of metabolic products associated with gut dysbiosis. One such metabolite, trimethylamine N-oxide (TMAO), has been observed to be directly related with increased incidence of cardiovascular diseases (CVD) in human patients. TMAO a gut-liver metabolite, comes from the metabolic degenerative product trimethylamine (TMA) that is produced from gut microbial metabolism. Elevated levels of TMAO in diabetics and obese patients are observed to have a direct correlation with increased risk for major adverse cardiovascular events. The pro-atherogenic effect of TMAO is attributed to enhancing inflammatory pathways with cholesterol and bile acid dysregulation, promoting foam cell formation. Recent studies have revealed several potential therapeutic strategies for reducing TMAO levels and will be the central focus for the current review. However, few have focused on developing rational drug therapeutics and may be due to the gaps in knowledge for understanding the mechanism by which microbial TMA producing enzymes and hepatic flavin-containing monoxygenase (FMO) can work together in preventing elevation of TMAO levels. Therefore, it is critical to understand the advantages of developing a novel rational drug design strategy that manipulates FMO production of TMAO and TMA production by microbial enzymes. This review will focus on the inspection of FMO manipulation, as well as gut microbiota dysbiosis and its influence on metabolic disorders including cardiovascular disease and describe novel potential pharmacological therapeutic development.
Collapse
Affiliation(s)
- Ian Steinke
- Drug Discovery and Development, Auburn University, Auburn, AL, United States
| | - Nila Ghanei
- Drug Discovery and Development, Auburn University, Auburn, AL, United States
| | - Manoj Govindarajulu
- Drug Discovery and Development, Auburn University, Auburn, AL, United States
| | - Sieun Yoo
- Department of Anatomy, Physiology and Pharmacology, Auburn University, Auburn, AL, United States
| | - Juming Zhong
- Department of Anatomy, Physiology and Pharmacology, Auburn University, Auburn, AL, United States
| | - Rajesh H Amin
- Drug Discovery and Development, Auburn University, Auburn, AL, United States
| |
Collapse
|
6
|
Zinad DS, Mahal A, Mohapatra RK, Sarangi AK, Pratama MRF. Medicinal chemistry of oxazines as promising agents in drug discovery. Chem Biol Drug Des 2019; 95:16-47. [PMID: 31583840 DOI: 10.1111/cbdd.13633] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 09/02/2019] [Accepted: 09/04/2019] [Indexed: 12/18/2022]
Affiliation(s)
- Dhafer S. Zinad
- Applied Science Department University of Technology Baghdad Iraq
| | - Ahmed Mahal
- Key Laboratory of Plant Resources Conservation and Sustainable Utilization and Guangdong Provincial Key Laboratory of Applied Botany South China Botanical Garden Chinese Academy of Sciences Guangzhou China
- Guangzhou HC Pharmaceutical Co., Ltd. Guangzhou China
| | - Ranjan K. Mohapatra
- Department of Chemistry Government College of Engineering Keonjhar Odisha India
| | - Ashish K. Sarangi
- Department of Chemistry Government College of Engineering Keonjhar Odisha India
| | - Mohammad Rizki Fadhil Pratama
- Department of Pharmacy Faculty of Health Sciences Muhammadiyah University of Palangkaraya Palangka Raya Indonesia
- Department of Pharmaceutical Chemistry Faculty of Pharmacy Airlangga University Surabaya Indonesia
| |
Collapse
|
7
|
Guo E, Hu Y, Du T, Zhu H, Chen L, Qu W, Zhang J, Xie N, Liu W, Feng F, Xu J. Effects of Picrasma quassioides and its active constituents on Alzheimer's disease in vitro and in vivo. Bioorg Chem 2019; 92:103258. [PMID: 31520892 DOI: 10.1016/j.bioorg.2019.103258] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 07/30/2019] [Accepted: 09/04/2019] [Indexed: 01/18/2023]
Abstract
Alzheimer disease (AD), a prevalent neurodegenerative disorder, is one of the leading causes of dementia. However, there is no effective drug for this disease to date. Picrasma quassioides (D.Don) Benn, a Chinese traditional medicine, was used mainly for the treatment of inflammation, fever, microbial infection and dysentery. In this paper, we reported that the EtOAc extract of Picrasma quassioides stems showed potential neuroprotective activities in l-glutamate-stimulated PC12 and Aβ25-35-stimulated SH-SY5Y cell models, as well as improved memory and cognitive abilities in AD mice induced by amyloid-β peptide. Moreover, it was revealed that the anti-AD mechanism was related to suppressing neuroinflammatory and reducing Aβ1-42 deposition using ELISA assay kits. To clarify the active components of the EtOAc extract of Picrasma quassioides stems, a systematic phytochemistry study led to isolate and identify six β-carboline alkaloids (1-6), seven canthin-6-one alkaloids (7-13), and five quassinoids (14-18). Among them, four β-carbolines (1-3, and 6) and six canthin-6-ones (7-11, and 13) exhibited potential neuroprotective activities in vitro. Based on these date, the structure-activity relationships of alkaloids were discussed. Furthermore, molecular docking experiments showed that compounds 2 and 3 have high affinity for both of dual-specificity tyrosine phosphorylation-regulated kinase 1A (DYPKIA) and butyrylcholinesterase (BuChE).
Collapse
Affiliation(s)
- Eryan Guo
- Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Yunwei Hu
- Key Laboratory of Drug Quality Control and Pharmacovigilance (China Pharmaceutical University), Ministry of Education, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Tao Du
- Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Huilin Zhu
- Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Lei Chen
- Key Laboratory of Drug Quality Control and Pharmacovigilance (China Pharmaceutical University), Ministry of Education, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Wei Qu
- Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, People's Republic of China; Key Laboratory of Biomedical Functional Materials, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Jie Zhang
- Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Ning Xie
- State Key Laboratory of Innovative Natural Medicines and TCM Injections, Jiangxi Qingfeng Pharmaceutical Co., Ltd., Ganzhou 341000, Jiangxi, China
| | - Wenyuan Liu
- Key Laboratory of Drug Quality Control and Pharmacovigilance (China Pharmaceutical University), Ministry of Education, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Feng Feng
- Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, People's Republic of China; Key Laboratory of Biomedical Functional Materials, China Pharmaceutical University, Nanjing 211198, People's Republic of China; Jiangsu Food & Pharmaceutical Science College, Huaian 223003, People's Republic of China.
| | - Jian Xu
- Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, People's Republic of China.
| |
Collapse
|
8
|
Rhodes S, Short S, Sharma S, Kaur R, Jha M. One-pot mild and efficient synthesis of [1,3]thiazino[3,2-a]indol-4-ones and their anti-proliferative activity. Org Biomol Chem 2019; 17:3914-3920. [DOI: 10.1039/c9ob00500e] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
One-pot synthesis of [1,3]thiazino[3,2-a]indol-4-one frameworks is developed in aqueous medium and the anti-proliferative activity of the synthesized compounds is evaluated against two triple negative breast cancer cell lines.
Collapse
Affiliation(s)
- Steven Rhodes
- Department of Biology and Chemistry
- Nipissing University
- North Bay
- Canada
| | - Spencer Short
- Department of Biology and Chemistry
- Nipissing University
- North Bay
- Canada
| | - Sidhika Sharma
- Department of Biology
- University of North Georgia
- Oakwood
- USA
| | - Ramneet Kaur
- Department of Biology
- University of North Georgia
- Oakwood
- USA
| | - Mukund Jha
- Department of Biology and Chemistry
- Nipissing University
- North Bay
- Canada
| |
Collapse
|
9
|
Wani IA, Das S, Mondal S, Ghorai MK. Stereoselective Construction of Pyrazinoindoles and Oxazinoindoles via Ring-Opening/Pictet-Spengler Reaction of Aziridines and Epoxides with 3-Methylindoles and Carbonyls. J Org Chem 2018; 83:14553-14567. [DOI: 10.1021/acs.joc.8b02339] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Imtiyaz Ahmad Wani
- Department of Chemistry, Indian Institute of Technology, 208016 Kanpur, Uttar Pradesh, India
| | - Subhomoy Das
- Department of Chemistry, Indian Institute of Technology, 208016 Kanpur, Uttar Pradesh, India
| | - Shobhan Mondal
- Department of Chemistry, Indian Institute of Technology, 208016 Kanpur, Uttar Pradesh, India
| | - Manas K. Ghorai
- Department of Chemistry, Indian Institute of Technology, 208016 Kanpur, Uttar Pradesh, India
| |
Collapse
|
10
|
Yan J, Zhou Y, Zhang N, Chen H, He X, Xiao S, Zheng K. Facile synthesis of novel [1,3]oxazino[2,3-c][1,2,4] thiadiazin-12-one derivatives. RESEARCH ON CHEMICAL INTERMEDIATES 2017. [DOI: 10.1007/s11164-017-2934-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|