1
|
Simms CH, Kovacs D, Hacker L, Sarson ET, Sokolova D, Christensen KE, Khrapichev A, Martin LAW, Vincent K, Conway SJ, Hammond EM, Langton MJ, Faulkner S. Binuclear Lanthanide Complexes as Magnetic Resonance and Optical Imaging Probes for Redox Sensing. Chemistry 2025:e202404748. [PMID: 40226941 DOI: 10.1002/chem.202404748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 04/11/2025] [Accepted: 04/14/2025] [Indexed: 04/15/2025]
Abstract
We report a family of lanthanide(III) complexes that act as redox probes via both magnetic resonance (MR) and luminescence outputs. The ligands are functionalized with nitro, azobenzene and azide groups which are reduced to a common aniline product, and each responds to both chemical and biocatalytic reductive conditions at different cathodic onset potentials. By judicious choice of complexed Ln(III), the probes can be optimized either for use in MR imaging (Ln = Gd), or as highly efficient turn-on luminescence probes (Ln = Tb). The Tb(III) analogues are essentially nonemissive, until reductive generation of the aniline affords a complex which when excited by visible light (488 nm) emits green light with a quantum yield of 45% and millisecond long luminescent lifetimes (ms). The tunable redox response and imaging modalities of these versatile complexes have the potential to open up new approaches to redox sensing, such as the imaging of hypoxic environments in biology.
Collapse
Affiliation(s)
- Charlie H Simms
- Department of Chemistry, Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford, OX1 3TA, UK
| | - Daniel Kovacs
- Department of Chemistry, Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford, OX1 3TA, UK
| | - Lina Hacker
- Department of Oncology, University of Oxford, Oxford, OX3 7DQ, UK
| | - Euan T Sarson
- Department of Chemistry, Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford, OX1 3TA, UK
| | - Daria Sokolova
- Department of Chemistry, Inorganic Chemistry Laboratory, University of Oxford, South Parks Road, Oxford, OX1 3QR, UK
| | - Kirsten E Christensen
- Department of Chemistry, Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford, OX1 3TA, UK
| | | | | | - Kylie Vincent
- Department of Chemistry, Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford, OX1 3TA, UK
| | - Stuart J Conway
- Department of Chemistry and Biochemistry, University of California, Los Angeles, 607 Charles E. Young Drive East, Box 951569, Los Angeles, CA, 90095-1569, USA
| | - Ester M Hammond
- Department of Oncology, University of Oxford, Oxford, OX3 7DQ, UK
| | - Matthew J Langton
- Department of Chemistry, Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford, OX1 3TA, UK
| | - Stephen Faulkner
- Department of Chemistry, Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford, OX1 3TA, UK
| |
Collapse
|
2
|
Zhang Y, Cao GY, Zhou Z, Hu TY, Xu BX, Chen DA, Du JB, Wang J, Wang G, Zhen L. Colon-Targeting Supersulfide Donor-Drug Conjugates Align Forces against Inflammation. JACS AU 2025; 5:642-652. [PMID: 40017786 PMCID: PMC11862929 DOI: 10.1021/jacsau.4c00868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 01/17/2025] [Accepted: 01/20/2025] [Indexed: 03/01/2025]
Abstract
Supersulfides are rising stars in regulating redox. Their distinctive redox biological functions are becoming increasingly evident with the advancement of supersulfide donors. However, most existing donors are limited to releasing hydropersulfide (RSSH) only, and in addition, there is still a knowledge gap in translating supersulfides into therapeutic molecules. To this end, in this work, we devised and synthesized a supersulfide donor-drug conjugate, RSSS-ASA, containing an azo moiety. This bifunctional prodrug enables the production of hydrotrisulfide (RSSSH) catalyzed by intestinal azoreductase, accompanied by the release of the anti-inflammatory molecule 5-aminosalicylic acid (ASA). Notably, the corelease of the supersulfides with ASA exhibited colonic-targeting attributes, thereby synergistically contributing to the potent anti-inflammatory and antioxidant activities observed in cellular and animal models. This prodrug design is worthy of further development and translation in donor development and disease treatment.
Collapse
Affiliation(s)
| | | | - Zhihao Zhou
- Key Laboratory of Drug Metabolism
and Pharmacokinetics, China Pharmaceutical
University, 24 Tongjia Xiang, Nanjing, Jiangsu 210009, China
| | - Tian-Yu Hu
- Key Laboratory of Drug Metabolism
and Pharmacokinetics, China Pharmaceutical
University, 24 Tongjia Xiang, Nanjing, Jiangsu 210009, China
| | - Bi-Xin Xu
- Key Laboratory of Drug Metabolism
and Pharmacokinetics, China Pharmaceutical
University, 24 Tongjia Xiang, Nanjing, Jiangsu 210009, China
| | - De-Ao Chen
- Key Laboratory of Drug Metabolism
and Pharmacokinetics, China Pharmaceutical
University, 24 Tongjia Xiang, Nanjing, Jiangsu 210009, China
| | - Jin-Biao Du
- Key Laboratory of Drug Metabolism
and Pharmacokinetics, China Pharmaceutical
University, 24 Tongjia Xiang, Nanjing, Jiangsu 210009, China
| | - Jiankun Wang
- Key Laboratory of Drug Metabolism
and Pharmacokinetics, China Pharmaceutical
University, 24 Tongjia Xiang, Nanjing, Jiangsu 210009, China
| | - Guangji Wang
- Key Laboratory of Drug Metabolism
and Pharmacokinetics, China Pharmaceutical
University, 24 Tongjia Xiang, Nanjing, Jiangsu 210009, China
| | - Le Zhen
- Key Laboratory of Drug Metabolism
and Pharmacokinetics, China Pharmaceutical
University, 24 Tongjia Xiang, Nanjing, Jiangsu 210009, China
| |
Collapse
|
3
|
Tian JH, Huang S, Wang ZH, Li JJ, Song X, Jiang ZT, Shi BS, Zhao YY, Zhang HY, Wang KR, Hu XY, Zhang X, Guo DS. Supramolecular discrimination and diagnosis-guided treatment of intracellular bacteria. Nat Commun 2025; 16:1016. [PMID: 39863571 PMCID: PMC11762306 DOI: 10.1038/s41467-025-56308-9] [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: 06/28/2024] [Accepted: 01/15/2025] [Indexed: 01/27/2025] Open
Abstract
Pathogenic intracellular bacteria pose a significant threat to global public health due to the barriers presented by host cells hindering the timely detection of hidden bacteria and the effective delivery of therapeutic agents. To address these challenges, we propose a tandem diagnosis-guided treatment paradigm. A supramolecular sensor array is developed for simple, rapid, accurate, and high-throughput identification of intracellular bacteria. This diagnostic approach executes the significant guiding missions of screening a customized host-guest drug delivery system by disclosing the rationale behind the discrimination. We design eight azocalix[4]arenes with differential active targeting, cellular internalization, and hypoxia responsiveness to penetrate cells and interact with bacteria. Loaded with fluorescent indicators, these azocalix[4]arenes form a sensor array capable of discriminating eight intracellular bacterial species without cell lysis or separation. By fingerprinting specimens collected from bacteria-infected mice, the facilitated accurate diagnosis offers valuable guidance for selecting appropriate antibiotics. Moreover, mannose-modified azocalix[4]arene (ManAC4A) is screened as a drug carrier efficiently taken up by macrophages. Doxycycline loaded with ManAC4A exhibits improved efficacy against methicillin-resistant Staphylococcus aureus-infected peritonitis. This study introduces an emerging paradigm to intracellular bacterial diagnosis and treatment, offering broad potential in combating bacterial infectious diseases.
Collapse
Affiliation(s)
- Jia-Hong Tian
- College of Chemistry, Nankai University, Tianjin, China
- State Key Laboratory of Elemento-Organic Chemistry, Nankai University, Tianjin, China
- Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin, China
- Frontiers Science Center for New Organic Matter, Nankai University, Tianjin, China
- Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin, China
| | - Siyuan Huang
- College of Chemistry, Nankai University, Tianjin, China
- Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin, China
- Tianjin Key Laboratory of Functional Polymer Materials, Nankai University, Tianjin, China
| | - Ze-Han Wang
- College of Chemistry, Nankai University, Tianjin, China
- State Key Laboratory of Elemento-Organic Chemistry, Nankai University, Tianjin, China
- Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin, China
- Frontiers Science Center for New Organic Matter, Nankai University, Tianjin, China
- Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin, China
| | - Juan-Juan Li
- College of Chemistry, Nankai University, Tianjin, China
- State Key Laboratory of Elemento-Organic Chemistry, Nankai University, Tianjin, China
- Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin, China
- Frontiers Science Center for New Organic Matter, Nankai University, Tianjin, China
- Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin, China
| | - Xianhui Song
- College of Chemistry, Nankai University, Tianjin, China
- Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin, China
- Tianjin Key Laboratory of Functional Polymer Materials, Nankai University, Tianjin, China
| | - Ze-Tao Jiang
- College of Chemistry, Nankai University, Tianjin, China
- State Key Laboratory of Elemento-Organic Chemistry, Nankai University, Tianjin, China
- Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin, China
- Frontiers Science Center for New Organic Matter, Nankai University, Tianjin, China
- Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin, China
| | - Bing-Sen Shi
- College of Chemistry and Materials Science, Hebei University, Baoding, China
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, Hebei University, Baoding, China
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis (Ministry of Education), Hebei University, Baoding, China
- Key Laboratory of Chemical Biology of Hebei Province, Hebei University, Baoding, China
- Hebei Research Center of the Basic Discipline of Synthetic Chemistry, Hebei University, Baoding, China
| | - Ying-Ying Zhao
- College of Chemistry and Materials Science, Hebei University, Baoding, China
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, Hebei University, Baoding, China
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis (Ministry of Education), Hebei University, Baoding, China
- Key Laboratory of Chemical Biology of Hebei Province, Hebei University, Baoding, China
- Hebei Research Center of the Basic Discipline of Synthetic Chemistry, Hebei University, Baoding, China
| | - Hui-Yan Zhang
- College of Chemistry and Materials Science, Hebei University, Baoding, China
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, Hebei University, Baoding, China
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis (Ministry of Education), Hebei University, Baoding, China
- Key Laboratory of Chemical Biology of Hebei Province, Hebei University, Baoding, China
- Hebei Research Center of the Basic Discipline of Synthetic Chemistry, Hebei University, Baoding, China
| | - Ke-Rang Wang
- College of Chemistry and Materials Science, Hebei University, Baoding, China
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, Hebei University, Baoding, China
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis (Ministry of Education), Hebei University, Baoding, China
- Key Laboratory of Chemical Biology of Hebei Province, Hebei University, Baoding, China
- Hebei Research Center of the Basic Discipline of Synthetic Chemistry, Hebei University, Baoding, China
| | - Xin-Yue Hu
- College of Chemistry, Nankai University, Tianjin, China.
- State Key Laboratory of Elemento-Organic Chemistry, Nankai University, Tianjin, China.
- Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin, China.
- Frontiers Science Center for New Organic Matter, Nankai University, Tianjin, China.
- Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin, China.
| | - Xinge Zhang
- College of Chemistry, Nankai University, Tianjin, China.
- Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin, China.
- Tianjin Key Laboratory of Functional Polymer Materials, Nankai University, Tianjin, China.
| | - Dong-Sheng Guo
- College of Chemistry, Nankai University, Tianjin, China.
- State Key Laboratory of Elemento-Organic Chemistry, Nankai University, Tianjin, China.
- Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin, China.
- Frontiers Science Center for New Organic Matter, Nankai University, Tianjin, China.
- Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin, China.
- Xinjiang Key Laboratory of Novel Functional Materials Chemistry, Kashi University, Kashi, China.
- College of Chemistry and Environmental Sciences, Kashi University, Kashi, China.
| |
Collapse
|
4
|
Peng YJ, Xu B, Rokita SE. Breaking the Myth of Enzymatic Azoreduction. ACS Chem Biol 2025; 20:229-237. [PMID: 39707960 DOI: 10.1021/acschembio.4c00779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2024]
Abstract
Flavin-dependent azoreductases have been applied to a wide range of tasks from decolorizing numerous azo dyes to releasing azo-conjugated prodrugs. A general narrative reiterated in much of the literature suggests that this enzyme promotes sequential reduction of both the azo-containing substrate and its corresponding hydrazo product to release the aryl amine components while consuming two equivalents of NAD(P)H. Indeed, such aryl amines can be formed by incubation of certain azo compounds with azoreductases, but the nature of the substrates capable of this apparent azo bond lysis remained unknown. We have now prepared a set of azobenzene derivatives and characterized their turnover and products after treatment with azoreductase from Escherichia coli to discover the structural basis regulating aryl amine formation. Without resonance donation by aryl substituents, reduction ceases at the hydrazo product. This indicates that azoreductases do not act on the hydrazo bond. Instead, aryl amine formation depends on a spontaneous hydrazo bond lysis that is promoted by resonance stabilization and subsequent reduction of the quinone-like intermediate by azoreductase. Experimental and computational approaches confirm the substituent dependence of this process. With knowledge of this requirement, full release of aryl amines from azo-conjugates can now be designed and applied with confidence.
Collapse
Affiliation(s)
- Yu-Ju Peng
- Department of Chemistry, Johns Hopkins University, 3400 N. Charles St., Baltimore, Maryland 21218, United States
| | - Bing Xu
- Department of Chemistry, Johns Hopkins University, 3400 N. Charles St., Baltimore, Maryland 21218, United States
| | - Steven E Rokita
- Department of Chemistry, Johns Hopkins University, 3400 N. Charles St., Baltimore, Maryland 21218, United States
| |
Collapse
|
5
|
Ma B, Shi J, Zhang Y, Li Z, Yong H, Zhou YN, Liu S, A S, Zhou D. Enzymatically Activatable Polymers for Disease Diagnosis and Treatment. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2306358. [PMID: 37992728 DOI: 10.1002/adma.202306358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 11/03/2023] [Indexed: 11/24/2023]
Abstract
The irregular expression or activity of enzymes in the human body leads to various pathological disorders and can therefore be used as an intrinsic trigger for more precise identification of disease foci and controlled release of diagnostics and therapeutics, leading to improved diagnostic accuracy, sensitivity, and therapeutic efficacy while reducing systemic toxicity. Advanced synthesis strategies enable the preparation of polymers with enzymatically activatable skeletons or side chains, while understanding enzymatically responsive mechanisms promotes rational incorporation of activatable units and predictions of the release profile of diagnostics and therapeutics, ultimately leading to promising applications in disease diagnosis and treatment with superior biocompatibility and efficiency. By overcoming the challenges, new opportunities will emerge to inspire researchers to develop more efficient, safer, and clinically reliable enzymatically activatable polymeric carriers as well as prodrugs.
Collapse
Affiliation(s)
- Bin Ma
- School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Jiahao Shi
- School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Yuhe Zhang
- School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Zhili Li
- School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Haiyang Yong
- School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Ya-Nan Zhou
- School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Shuai Liu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Sigen A
- School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
- School of Medicine, Anhui University of Science and Technology, Huainan, 232001, China
| | - Dezhong Zhou
- School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| |
Collapse
|
6
|
Han J, Sheng T, Zhang Y, Cheng H, Gao J, Yu J, Gu Z. Bioresponsive Immunotherapeutic Materials. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2209778. [PMID: 36639983 DOI: 10.1002/adma.202209778] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 12/31/2022] [Indexed: 06/17/2023]
Abstract
The human immune system is an interaction network of biological processes, and its dysfunction is closely associated with a wide array of diseases, such as cancer, infectious diseases, tissue damage, and autoimmune diseases. Manipulation of the immune response network in a desired and controlled fashion has been regarded as a promising strategy for maximizing immunotherapeutic efficacy and minimizing side effects. Integration of "smart" bioresponsive materials with immunoactive agents including small molecules, biomacromolecules, and cells can achieve on-demand release of agents at targeted sites to reduce overdose-related toxicity and alleviate off-target effects. This review highlights the design principles of bioresponsive immunotherapeutic materials and discusses the critical roles of controlled release of immunoactive agents from bioresponsive materials in recruiting, housing, and manipulating immune cells for evoking desired immune responses. Challenges and future directions from the perspective of clinical translation are also discussed.
Collapse
Affiliation(s)
- Jinpeng Han
- Zhejiang Provincial Key Laboratory for Advanced Drug Delivery Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Tao Sheng
- Zhejiang Provincial Key Laboratory for Advanced Drug Delivery Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yuqi Zhang
- Zhejiang Provincial Key Laboratory for Advanced Drug Delivery Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Department of Burns and Wound Center, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Hao Cheng
- Department of Materials Science and Engineering, Drexel University, Philadelphia, PA, 19104, USA
| | - Jianqing Gao
- Zhejiang Provincial Key Laboratory for Advanced Drug Delivery Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Cancer Center, Zhejiang University, Hangzhou, 310058, China
- Jinhua Institute of Zhejiang University, Jinhua, 321299, China
| | - Jicheng Yu
- Zhejiang Provincial Key Laboratory for Advanced Drug Delivery Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Jinhua Institute of Zhejiang University, Jinhua, 321299, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, 311121, China
- Department of General Surgery, Sir Run Run Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
| | - Zhen Gu
- Zhejiang Provincial Key Laboratory for Advanced Drug Delivery Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Jinhua Institute of Zhejiang University, Jinhua, 321299, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, 311121, China
- Department of General Surgery, Sir Run Run Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| |
Collapse
|
7
|
González A, Fullaondo A, Odriozola I, Odriozola A. Microbiota and other detrimental metabolites in colorectal cancer. ADVANCES IN GENETICS 2024; 112:309-365. [PMID: 39396839 DOI: 10.1016/bs.adgen.2024.08.006] [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: 10/15/2024]
Abstract
Increasing scientific evidence demonstrates that gut microbiota plays an essential role in the onset and development of Colorectal cancer (CRC). However, the mechanisms by which these microorganisms contribute to cancer development are complex and far from completely clarified. Specifically, the impact of gut microbiota-derived metabolites on CRC is undeniable, exerting both protective and detrimental effects. This paper examines the effects and mechanisms by which important bacterial metabolites exert detrimental effects associated with increased risk of CRC. Metabolites considered include heterocyclic amines and polycyclic aromatic hydrocarbons, heme iron, secondary bile acids, ethanol, and aromatic amines. It is necessary to delve deeper into the mechanisms of action of these metabolites in CRC and identify the microbiota members involved in their production. Furthermore, since diet is the main factor capable of modifying the intestinal microbiota, conducting studies that include detailed descriptions of dietary interventions is crucial. All this knowledge is essential for developing precision nutrition strategies to optimise a protective intestinal microbiota against CRC.
Collapse
Affiliation(s)
- Adriana González
- Hologenomics Research Group, Department of Genetics, Physical Anthropology, and Animal Physiology, University of the Basque Country, Spain
| | - Asier Fullaondo
- Hologenomics Research Group, Department of Genetics, Physical Anthropology, and Animal Physiology, University of the Basque Country, Spain
| | - Iñaki Odriozola
- Health Department of Basque Government, Donostia-San Sebastián, Spain
| | - Adrian Odriozola
- Hologenomics Research Group, Department of Genetics, Physical Anthropology, and Animal Physiology, University of the Basque Country, Spain.
| |
Collapse
|
8
|
Gu Y, Yang R, Chen J, Fan Y, Xie W, Wu H, Ding J. Design and Synthesis of an Azo Reductase Responsive Flavonol-Indomethacin Hybrid Used for the Diagnosis and Treatment of Colitis. Molecules 2024; 29:4244. [PMID: 39275092 PMCID: PMC11397019 DOI: 10.3390/molecules29174244] [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: 06/09/2024] [Revised: 08/17/2024] [Accepted: 08/29/2024] [Indexed: 09/16/2024] Open
Abstract
Human intestinal bacteria are the primary producers of azo reductase, and the content of azo reductase is closely associated with various intestinal diseases, including ulcerative colitis (UC). The rapid detection of changes in azo reductase levels is crucial for diagnosing and promptly intervening in UC. In this study, a therapeutic agent, FAI, specifically targeting UC, was designed and synthesized. This agent was developed by linking the anti-inflammatory drug indomethacin to flavonols with antioxidant activity via an azo bond (off-on). Breakage of the azo bond breaks results in the release of both fluorophores and drugs, achieving targeted tracing and integrated treatment effects. In vivo and in vitro fluorescence imaging experiments were used to demonstrate the potential of FAI in the diagnosis of UC, together with synergistic therapeutic effects through the release of both fluorophores and anti-inflammatory agents. Therefore, this diagnostic agent shows promise as a potential tool for diagnosing and treating UC.
Collapse
Affiliation(s)
- Yaqin Gu
- School of Pharmacy, Jiangsu Vocational College of Medicine, Yancheng 224005, China
| | - Rui Yang
- School of Pharmacy, Jiangsu Vocational College of Medicine, Yancheng 224005, China
| | - Jine Chen
- School of Pharmacy, Jiangsu Vocational College of Medicine, Yancheng 224005, China
| | - Yu Fan
- School of Pharmacy, Nantong University, Nantong 226019, China
| | - Wenna Xie
- School of Pharmacy, Jiangsu Vocational College of Medicine, Yancheng 224005, China
| | - Hongyan Wu
- School of Pharmacy, Jiangsu Vocational College of Medicine, Yancheng 224005, China
| | - Jinfeng Ding
- School of Pharmacy, Jiangsu Vocational College of Medicine, Yancheng 224005, China
| |
Collapse
|
9
|
Kopeček J. Hydrophilic biomaterials: From crosslinked and self-assembled hydrogels to polymer-drug conjugates and drug-free macromolecular therapeutics. J Control Release 2024; 373:1-22. [PMID: 38734315 PMCID: PMC11384549 DOI: 10.1016/j.jconrel.2024.05.012] [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: 03/15/2024] [Revised: 05/05/2024] [Accepted: 05/07/2024] [Indexed: 05/13/2024]
Abstract
This "Magnum Opus" accentuates my lifelong belief that the future of science is in the interdisciplinary approach to hypotheses formulation and problem solving. Inspired by the invention of hydrogels and soft contact lenses by my mentors, my six decades of research have continuously proceeded from the synthesis of biocompatible hydrogels to the development of polymer-drug conjugates, then generation of drug-free macromolecular therapeutics (DFMT) and finally to multi-antigen T cell hybridizers (MATCH). This interdisciplinary journey was inspiring; the lifetime feeling that one is a beginner in some aspects of the research is a driving force that keeps the enthusiasm high. Also, I wanted to illustrate that systematic research in one wide area can be a life-time effort without the need to jump to areas that are temporarily en-vogue. In addition to generating general scientific knowledge, hydrogels from my laboratory have been transferred to the clinic, polymer-drug conjugates to clinical trials, and drug-free macromolecular systems have an excellent potential for personalizing patient therapies. There is a limit to life but no limit to imagination. I anticipate that systematic basic research will contribute to the expansion of our knowledge and create a foundation for the design of new paradigms based on the comprehension of mechanisms of physiological processes. The emerging novel platform technologies in biomaterial-based devices and implants as well as in personalized nanomedicines will ultimately impact clinical practice.
Collapse
Affiliation(s)
- Jindřich Kopeček
- Center for Controlled Chemical Delivery, Department of Molecular Pharmaceutics, Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA.
| |
Collapse
|
10
|
Kolay S, Das M, Mondal A, Sengupta A, Bag S, De P, Molla MR. Enzyme-Triggered Degradation of Supramolecularly Cross-Linked Polymersomes of Azobenzene-Based Polyurethane: Cell-Selective Anticancer Drug Release. Biomacromolecules 2024; 25:5068-5080. [PMID: 39041235 DOI: 10.1021/acs.biomac.4c00485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/24/2024]
Abstract
Enzyme-responsive self-assembled nanostructures for drug delivery applications have gained a lot of attention, as enzymes exhibit dysregulation in many disease-associated microenvironments. Azoreductase enzyme levels are strongly elevated in many tumor tissues; hence, here, we exploited the altered enzyme activity of the azoreductase enzyme and designed a main-chain azobenzene-based amphiphilic polyurethane, which self-assembles into a vesicular nanostructure and is programmed to disassemble in response to a specific enzyme, azoreductase, with the help of the nicotinamide adenine dinucleotide phosphate (NADPH) coenzyme in the hypoxic environment of solid tumors. The vesicular nanostructure sequesters, stabilizes the hydrophobic anticancer drug, and releases the drug in a controlled fashion in response to enzyme-triggered degradation of azo-bonds and disruption of vesicular assembly. The biological evaluation revealed tumor extracellular matrix pH-induced surface charge modulation, selective activated cellular uptake to azoreductase overexpressed lung cancer cells (A549), and the release of the anticancer drug followed by cell death. In contrast, the benign nature of the drug-loaded vesicular nanostructure toward normal cells (H9c2) suggested excellent cell specificity. We envision that the main-chain azobenzene-based polyurethane discussed in this manuscript could be considered as a possible selective chemotherapeutic cargo against the azoreductase overexpressed cancer cells while shielding the normal cells from off-target toxicity.
Collapse
Affiliation(s)
- Soumya Kolay
- Department of Chemistry, University of Calcutta, 92 A. P. C. Road, Kolkata 700009, India
| | - Madhuchhanda Das
- Department of Life Science & Biotechnology, Jadavpur University, 188, Raja S. C. M Road, Kolkata 700032, India
| | - Arun Mondal
- Department of Chemistry, University of Calcutta, 92 A. P. C. Road, Kolkata 700009, India
| | - Arunima Sengupta
- Department of Life Science & Biotechnology, Jadavpur University, 188, Raja S. C. M Road, Kolkata 700032, India
| | - Sagar Bag
- Department of Chemical Science, Indian Institute of Science Education and Research, Mohanpur, Nadia, Kolkata, West Bengal 741246, India
| | - Priyadarsi De
- Department of Chemical Science, Indian Institute of Science Education and Research, Mohanpur, Nadia, Kolkata, West Bengal 741246, India
| | - Mijanur Rahaman Molla
- Department of Chemistry, University of Calcutta, 92 A. P. C. Road, Kolkata 700009, India
| |
Collapse
|
11
|
Simpson JB, Walker ME, Sekela JJ, Ivey SM, Jariwala PB, Storch CM, Kowalewski ME, Graboski AL, Lietzan AD, Walton WG, Davis KA, Cloer EW, Borlandelli V, Hsiao YC, Roberts LR, Perlman DH, Liang X, Overkleeft HS, Bhatt AP, Lu K, Redinbo MR. Gut microbial β-glucuronidases influence endobiotic homeostasis and are modulated by diverse therapeutics. Cell Host Microbe 2024; 32:925-944.e10. [PMID: 38754417 PMCID: PMC11176022 DOI: 10.1016/j.chom.2024.04.018] [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: 11/10/2023] [Revised: 03/18/2024] [Accepted: 04/24/2024] [Indexed: 05/18/2024]
Abstract
Hormones and neurotransmitters are essential to homeostasis, and their disruptions are connected to diseases ranging from cancer to anxiety. The differential reactivation of endobiotic glucuronides by gut microbial β-glucuronidase (GUS) enzymes may influence interindividual differences in the onset and treatment of disease. Using multi-omic, in vitro, and in vivo approaches, we show that germ-free mice have reduced levels of active endobiotics and that distinct gut microbial Loop 1 and FMN GUS enzymes drive hormone and neurotransmitter reactivation. We demonstrate that a range of FDA-approved drugs prevent this reactivation by intercepting the catalytic cycle of the enzymes in a conserved fashion. Finally, we find that inhibiting GUS in conventional mice reduces free serotonin and increases its inactive glucuronide in the serum and intestines. Our results illuminate the indispensability of gut microbial enzymes in sustaining endobiotic homeostasis and indicate that therapeutic disruptions of this metabolism promote interindividual response variabilities.
Collapse
Affiliation(s)
- Joshua B Simpson
- Department of Chemistry, University of North Carolina, Chapel Hill, NC, USA
| | - Morgan E Walker
- Department of Chemistry, University of North Carolina, Chapel Hill, NC, USA
| | - Joshua J Sekela
- Department of Chemistry, University of North Carolina, Chapel Hill, NC, USA
| | - Samantha M Ivey
- Department of Chemistry, University of North Carolina, Chapel Hill, NC, USA
| | - Parth B Jariwala
- Department of Chemistry, University of North Carolina, Chapel Hill, NC, USA
| | - Cameron M Storch
- Department of Chemistry, University of North Carolina, Chapel Hill, NC, USA
| | - Mark E Kowalewski
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC, USA
| | - Amanda L Graboski
- Department of Pharmacology, University of North Carolina, Chapel Hill, NC, USA
| | - Adam D Lietzan
- Division of Oral and Craniofacial Health Sciences, Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - William G Walton
- Department of Chemistry, University of North Carolina, Chapel Hill, NC, USA
| | - Kacey A Davis
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC, USA
| | - Erica W Cloer
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Valentina Borlandelli
- Department of Bioorganic Synthesis, Leiden Institute of Chemistry, Leiden University, Leiden, the Netherlands
| | - Yun-Chung Hsiao
- Department of Environmental Sciences and Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Lee R Roberts
- Exploratory Science Center, Merck & Co., Inc., Cambridge, MA 02141, USA
| | - David H Perlman
- Exploratory Science Center, Merck & Co., Inc., Cambridge, MA 02141, USA
| | - Xue Liang
- Exploratory Science Center, Merck & Co., Inc., Cambridge, MA 02141, USA
| | - Hermen S Overkleeft
- Department of Bioorganic Synthesis, Leiden Institute of Chemistry, Leiden University, Leiden, the Netherlands
| | - Aadra P Bhatt
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Division of Gastroenterology and Hepatology, Department of Medicine, Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Kun Lu
- Department of Environmental Sciences and Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Matthew R Redinbo
- Department of Chemistry, University of North Carolina, Chapel Hill, NC, USA; Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
12
|
Zhao Z, Li J, Yuan W, Cheng D, Ma S, Li YF, Shi ZJ, Hu K. Nature-Inspired Photocatalytic Azo Bond Cleavage with Red Light. J Am Chem Soc 2024; 146:1364-1373. [PMID: 38082478 DOI: 10.1021/jacs.3c09837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2024]
Abstract
The emerging field of photoredox catalysis in mammalian cells enables spatiotemporal regulation of a wealth of biological processes. However, the selective cleavage of stable covalent bonds driven by low-energy visible light remains a great challenge. Herein, we report that red light excitation of a commercially available dye, abbreviated NMB+, leads to catalytic cleavage of stable azo bonds in both aqueous solutions and hypoxic cells and hence a means to photodeliver drugs or functional molecules. Detailed mechanistic studies reveal that azo bond cleavage is triggered by a previously unknown consecutive two-photon process. The first photon generates a triplet excited state, 3NMB+*, that is reductively quenched by an electron donor to generate a protonated NMBH•+. The NMBH•+ undergoes a disproportionation reaction that yields the initial NMB+ and two-electron-reduced NMBH (i.e., leuco-NMB, abbreviated as LNMB). Interestingly, LNMB forms a charge transfer complex with all four azo substrates that possess an intense absorption band in the red region. A second red photon induces electron transfer from LNMB to the azo substrate, resulting in azo bond cleavage. The charge transfer complex mediated two-photon catalytic mechanism reported herein is reminiscent of the flavin-dependent natural photoenzyme that catalyzes bond cleavage reactions with high-energy photons. The red-light-driven photocatalytic strategy offers a new approach to bioorthogonal azo bond cleavage for photodelivery of drugs or functional molecules.
Collapse
Affiliation(s)
- Zijian Zhao
- Department of Chemistry, Fudan University, 220 Handan Road, Shanghai 200433, People's Republic of China
| | - Jili Li
- Department of Chemistry, Fudan University, 220 Handan Road, Shanghai 200433, People's Republic of China
| | - Wei Yuan
- Shanghai Frontiers Science Research Base of Intelligent Optoelectronics and Perception, Institute of Optoelectronics, Fudan University, 2005 Songhu Road, Shanghai 200438, People's Republic of China
- Institute of Optoelectronics, Fudan University, 2005 Songhu Road, Shanghai 200438, People's Republic of China
| | - Dajiao Cheng
- Department of Chemistry, Fudan University, 220 Handan Road, Shanghai 200433, People's Republic of China
| | - Suze Ma
- Department of Chemistry, Fudan University, 220 Handan Road, Shanghai 200433, People's Republic of China
| | - Ye-Fei Li
- Department of Chemistry, Fudan University, 220 Handan Road, Shanghai 200433, People's Republic of China
| | - Zhang-Jie Shi
- Department of Chemistry, Fudan University, 220 Handan Road, Shanghai 200433, People's Republic of China
| | - Ke Hu
- Department of Chemistry, Fudan University, 220 Handan Road, Shanghai 200433, People's Republic of China
| |
Collapse
|
13
|
Janse van Rensburg HD, N'Da DD, Suganuma K. In vitro trypanocidal potency and in vivo treatment efficacy of oligomeric ethylene glycol-tethered nitrofurantoin derivatives. Eur J Pharm Sci 2024; 192:106668. [PMID: 38065268 DOI: 10.1016/j.ejps.2023.106668] [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: 10/24/2023] [Revised: 12/05/2023] [Accepted: 12/06/2023] [Indexed: 12/22/2023]
Abstract
African trypanosomiasis is a significant vector-borne disease of humans and animals in the tsetse fly belt of Africa, particularly affecting production animals such as cattle, and thus, hindering food security. Trypanosoma congolense (T. congolense), the causative agent of nagana, is livestock's most virulent trypanosome species. There is currently no vaccine against trypanosomiasis; its treatment relies solely on chemotherapy. However, pathogenic resistance has been established against trypanocidal agents in clinical use. This underscores the need to develop new therapeutics to curb trypanosomiasis. Many nitroheterocyclic drugs or compounds, including nitrofurantoin, possess antiparasitic activities in addition to their clinical use as antibiotics. The current study evaluated the in vitro trypanocidal potency and in vivo treatment efficacy of previously synthesized antileishmanial active oligomeric ethylene glycol derivatives of nitrofurantoin. The trypanocidal potency of analogues 2a-o varied among the trypanosome species; however, T. congolense strain IL3000 was more susceptible to these drug candidates than the other human and animal trypanosomes. The arylated analogues 2k (IC50 0.04 µM; SI >6365) and 2l (IC50 0.06 µM; SI 4133) featuring 4-chlorophenoxy and 4-nitrophenoxy moieties, respectively, were revealed as the most promising antitrypanosomal agents of all analogues against T. congolense strain IL3000 trypomastigotes with nanomolar activities. In a preliminary in vivo study involving T. congolense strain IL3000 infected BALB/c mice, the oral administration of 100 mg/kg/day of 2k caused prolonged survival up to 18 days post-infection relative to the infected but untreated control mice which survived 9 days post-infection. However, no cure was achieved due to its poor solubility in the in vivo testing medium, assumably leading to low oral bioavailability. These results confirm the importance of the physicochemical properties lipophilicity and water solubility in attaining not only in vitro trypanocidal potency but also in vivo treatment efficacy. Future work will focus on the chemical optimization of 2k through the investigation of analogues containing solubilizing groups at certain positions on the core structure to improve solubility in the in vivo testing medium which, in the current investigation, is the biggest stumbling block in successfully treating either animal or human Trypanosoma infections.
Collapse
Affiliation(s)
| | - David D N'Da
- Centre of Excellence for Pharmaceutical Sciences, North-West University, Potchefstroom 2520, South Africa
| | - Keisuke Suganuma
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada, Obihiro, Hokkaido 080-8555, Japan
| |
Collapse
|
14
|
Sparatore F, Sparatore A. 3,3-Disubstituted 3,4-Dihydro-1,2,4-benzotriazines: Chemistry, Biological Activity, and Affinity to Sigma Receptors. Molecules 2023; 29:132. [PMID: 38202715 PMCID: PMC10780181 DOI: 10.3390/molecules29010132] [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: 11/14/2023] [Revised: 12/17/2023] [Accepted: 12/18/2023] [Indexed: 01/12/2024] Open
Abstract
By reducing the 2-nitrophenylhydrazone of cyclohexanone with sodium dithionite, an unexpected yellow compound was obtained instead of the corresponding colorless amino derivative. Many years later, the structure of this compound, namely, cyclohexane-3-spiro-3,4-dihydro-1,2,4-benzotriazine, was demonstrated. From that time, the reduction of 2-nitrophenylhydrazones of different kinds of ketones, followed by air oxidation of the initially formed amino compounds, has represented a general way to synthesize a variety of 3,3-disubstituted 3,4-dihydro-1,2,4-benzotriazines. Many derivatives have been obtained so far by a single research group, and most of them have demonstrated interesting pharmacological activities, mainly antihypertensive, anti-inflammatory, and diuretic effects and other activities with lower diffusion. Moreover, 3,3-disubstituted 3,4-dihydro-1,2,4-benzotriazines represent a novel class of ligands for sigma receptors, with nanomolar affinity to the σ1 subtype. This property might promote the development of agents for cardiovascular, neurodegenerative, and proliferative pathologies. The present commentary, by collecting compounds and biological results obtained so far, intends to celebrate the centennial of the discovery of the first member of this class of compounds and to promote further investigation in the field.
Collapse
Affiliation(s)
- Fabio Sparatore
- Department of Pharmacy, University of Genova, 16132 Genova, Italy
| | - Anna Sparatore
- Department of Pharmaceutical Sciences (DISFARM), University of Milano, 20133 Milano, Italy;
| |
Collapse
|
15
|
Khojasteh SC, Argikar UA, Cheruzel L, Cho S, Crouch RD, Dhaware D, Heck CJS, Johnson KM, Kalgutkar AS, King L, Liu J, Ma B, Maw H, Miller GP, Seneviratne HK, Takahashi RH, Wang S, Wei C, Jackson KD. Biotransformation research advances - 2022 year in review. Drug Metab Rev 2023; 55:301-342. [PMID: 37737116 DOI: 10.1080/03602532.2023.2262161] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 06/05/2023] [Indexed: 09/23/2023]
Abstract
This annual review is the eighth of its kind since 2016 (Baillie et al. 2016, Khojasteh et al. 2017, Khojasteh et al. 2018, Khojasteh et al. 2019, Khojasteh et al. 2020, Khojasteh et al. 2021, Khojasteh et al. 2022). Our objective is to explore and share articles which we deem influential and significant in the field of biotransformation.
Collapse
Affiliation(s)
- S Cyrus Khojasteh
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc, South San Francisco, CA, USA
| | - Upendra A Argikar
- Non-clinical Development, Bill and Melinda Gates Medical Research Institute, Cambridge, MA, USA
| | - Lionel Cheruzel
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc, South San Francisco, CA, USA
| | - Sungjoon Cho
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc, South San Francisco, CA, USA
| | - Rachel D Crouch
- Department of Pharmacy and Pharmaceutical Sciences, Lipscomb University College of Pharmacy, Nashville, TN, USA
| | | | - Carley J S Heck
- Medicine Design, Pfizer Worldwide Research, Development and Medical, Groton, CT, USA
| | - Kevin M Johnson
- Drug Metabolism and Pharmacokinetics, Inotiv, MD Heights, MO, USA
| | - Amit S Kalgutkar
- Medicine Design, Pfizer Worldwide Research, Development and Medical, Cambridge, MA, USA
| | - Lloyd King
- Quantitative Drug Discovery, UCB Biopharma UK, Slough UK
| | - Joyce Liu
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc, South San Francisco, CA, USA
| | - Bin Ma
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc, South San Francisco, CA, USA
| | - Hlaing Maw
- Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals, Inc, Ridgefield, CT, USA
| | - Grover P Miller
- Department of Biochemistry and Molecular Biology, University of AR for Medical Sciences, Little Rock, AR, USA
| | | | - Ryan H Takahashi
- Drug Metabolism and Pharmacokinetics, Denali Therapeutics, South San Francisco, CA, USA
| | - Shuai Wang
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc, South San Francisco, CA, USA
| | - Cong Wei
- Drug Metabolism and Pharmacokinetics, Biogen Inc, Cambridge, MA, USA
| | - Klarissa D Jackson
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, Chapel Hill, NC, USA
| |
Collapse
|
16
|
Pieper LM, Spanogiannopoulos P, Volk RF, Miller CJ, Wright AT, Turnbaugh PJ. The global anaerobic metabolism regulator fnr is necessary for the degradation of food dyes and drugs by Escherichia coli. mBio 2023; 14:e0157323. [PMID: 37642463 PMCID: PMC10653809 DOI: 10.1128/mbio.01573-23] [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: 06/21/2023] [Accepted: 07/06/2023] [Indexed: 08/31/2023] Open
Abstract
IMPORTANCE This work has broad relevance due to the ubiquity of dyes containing azo bonds in food and drugs. We report that azo dyes can be degraded by human gut bacteria through both enzymatic and nonenzymatic mechanisms, even from a single gut bacterial species. Furthermore, we revealed that environmental factors, oxygen, and L-Cysteine control the ability of E. coli to degrade azo dyes due to their impacts on bacterial transcription and metabolism. These results open up new opportunities to manipulate the azoreductase activity of the gut microbiome through the manipulation of host diet, suggest that azoreductase potential may be altered in patients suffering from gastrointestinal disease, and highlight the importance of studying bacterial enzymes for drug metabolism in their natural cellular and ecological context.
Collapse
Affiliation(s)
- Lindsey M. Pieper
- Department of Microbiology & Immunology, University of California, San Francisco, California, USA
| | - Peter Spanogiannopoulos
- Department of Microbiology & Immunology, University of California, San Francisco, California, USA
| | - Regan F. Volk
- Department of Microbiology & Immunology, University of California, San Francisco, California, USA
| | - Carson J. Miller
- Biological Sciences Group, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - Aaron T. Wright
- Biological Sciences Group, Pacific Northwest National Laboratory, Richland, Washington, USA
- Department of Biology, Baylor University, Waco, Texas, USA
- Department of Chemistry and Biochemistry, Baylor University, Waco, Texas, USA
| | - Peter J. Turnbaugh
- Department of Microbiology & Immunology, University of California, San Francisco, California, USA
- Chan Zuckerberg Biohub-San Francisco, San Francisco, California, USA
| |
Collapse
|
17
|
Bai W, Hu Y, Zhao J, Shi L, Ge C, Zhu Z, Rao J. Precision Dosing of Antibiotics and Potentiators by Hypoxia-Responsive Nanoparticles for Overcoming Antibiotic Resistance in Gram-Negative Bacteria. ACS Macro Lett 2023; 12:1193-1200. [PMID: 37590266 DOI: 10.1021/acsmacrolett.3c00365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/19/2023]
Abstract
The stalling development of antibiotics, especially against intrinsically resistant Gram-negative pathogens associated with outer membranes, leads to an emerging antibiotic crisis across the globe. To breathe life into existing drugs, we herein report a hypoxia-responsive nanoparticle (NP) that encapsulates a hydrophobic antibiotic, rifampicin, and a cationic potentiator, polysulfonium. The simultaneous release of antibiotics and potentiators can be promoted and inhibited in response to the severity of bacterial-induced hypoxia, leading to antimicrobial dosing in a precision manner. Under the synergism of polysulfoniums with membrane-disruption capability, the NPs can intensively decrease the antibiotic dose by up to 66-95% in eliminating planktonic Gram-negative P. aeruginosa bacteria and achieve an 8-log reduction of bacteria in mature biofilms at rifampicin MIC. The NP formulation demonstrates that precision dosing of antibiotics and potentiators regulated by hypoxia provides a promising strategy to maximize efficacy and minimize toxicity in treating Gram-negative bacterial infection.
Collapse
Affiliation(s)
- Weiguang Bai
- Hubei Key Laboratory of Material Chemistry and Service Failure, Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan, Hubei 430074, PR China
- College of Chemistry, Liaoning University, Shenyang 110036, PR China
| | - Yongjin Hu
- Hubei Key Laboratory of Material Chemistry and Service Failure, Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan, Hubei 430074, PR China
| | - Jinghua Zhao
- Hubei Key Laboratory of Material Chemistry and Service Failure, Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan, Hubei 430074, PR China
| | - Liuqi Shi
- Hubei Key Laboratory of Material Chemistry and Service Failure, Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan, Hubei 430074, PR China
| | - Chunhua Ge
- College of Chemistry, Liaoning University, Shenyang 110036, PR China
| | - Zhiyuan Zhu
- Taizhou Research Institute, Southern University of Science and Technology, Taizhou, Zhejiang 318001, PR China
| | - Jingyi Rao
- Hubei Key Laboratory of Material Chemistry and Service Failure, Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan, Hubei 430074, PR China
| |
Collapse
|
18
|
Josephy PD, Allen-Vercoe E. Reductive metabolism of azo dyes and drugs: Toxicological implications. Food Chem Toxicol 2023; 178:113932. [PMID: 37451600 DOI: 10.1016/j.fct.2023.113932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 06/24/2023] [Accepted: 07/04/2023] [Indexed: 07/18/2023]
Abstract
Azo compounds are widely distributed synthetic chemicals in the modern world. Their most important applications are as dyes, but, in addition, several azo compounds are used as pharmaceuticals. Ingested azo compounds can be reduced by the action of bacteria in the gut, where the oxygen tension is low, and the development of microbiome science has allowed more precise delineation of the roles of specific bacteria in these processes. Reduction of the azo bond of an azo compound generates two distinct classes of aromatic amine metabolites: the starting material that was used in the synthesis of the azo compound and a product which is formed de novo by metabolism. Reductive metabolism of azo compounds can have toxic consequences, because many aromatic amines are toxic/genotoxic. In this review, we discuss aspects of the development and application of azo compounds in industry and medicine. Current understanding of the toxicology of azo compounds and their metabolites is illustrated with four specific examples - Disperse Dyes used for dyeing textiles; the drugs phenazopyridine and eltrombopag; and the ubiquitous food dye, tartrazine - and knowledge gaps are identified. SUBMISSION TO: FCT VSI: Toxicology of Dyes.
Collapse
Affiliation(s)
- P David Josephy
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada.
| | - Emma Allen-Vercoe
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| |
Collapse
|
19
|
Wood GE, Kim CM, Aguila LKT, Cichewicz RH. In Vitro Susceptibility and Resistance of Mycoplasma genitalium to Nitroimidazoles. Antimicrob Agents Chemother 2023; 67:e0000623. [PMID: 37070857 PMCID: PMC10112249 DOI: 10.1128/aac.00006-23] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2023] Open
Abstract
Mycoplasma genitalium is a sexually transmitted reproductive tract pathogen of men and women. M. genitalium infections are increasingly difficult to treat due to poor efficacy of doxycycline and acquired resistance to azithromycin and moxifloxacin. A recent clinical trial suggested that metronidazole may improve cure rates for women with pelvic inflammatory disease and reduced the detection of M. genitalium when included with standard doxycycline plus ceftriaxone treatment. As data regarding susceptibility of mycoplasmas to nitroimidazoles are lacking in the scientific literature, we determined the in vitro susceptibility of 10 M. genitalium strains to metronidazole, secnidazole, and tinidazole. MICs ranged from 1.6 to 12.5 μg/mL for metronidazole, 3.1 to 12.5 μg/mL for secnidazole, and 0.8 to 6.3 μg/mL for tinidazole. None of these agents was synergistic with doxycycline in checkerboard broth microdilution assays. Tinidazole was superior to metronidazole and secnidazole in terms of MIC and time-kill kinetics and was bactericidal (>99.9% killing) at concentrations below reported serum concentrations. Mutations associated with nitroimidazole resistance were identified by whole-genome sequencing of spontaneous resistant mutants, suggesting a mechanism for reductive activation of the nitroimidazole prodrug by a predicted NAD(P)H-dependent flavin mononucleotide (FMN) oxidoreductase. The presence of oxygen did not affect MICs of wild-type M. genitalium, but a nitroimidazole-resistant mutant was defective for growth under anaerobic conditions, suggesting that resistant mutants may have a fitness disadvantage in anaerobic genital sites. Clinical studies are needed to determine if nitroimidazoles, especially tinidazole, are effective for eradicating M. genitalium infections in men and women.
Collapse
Affiliation(s)
- Gwendolyn E Wood
- Department of Medicine, Division of Allergy and Infectious Diseases, University of Washington, Seattle, Washington, USA
| | - Caroline M Kim
- Department of Medicine, Division of Allergy and Infectious Diseases, University of Washington, Seattle, Washington, USA
| | - Laarni Kendra T Aguila
- Department of Medicine, Division of Allergy and Infectious Diseases, University of Washington, Seattle, Washington, USA
| | - Robert H Cichewicz
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma, USA
- Natural Products Discovery Group, University of Oklahoma, Norman, Oklahoma, USA
- Institute for Natural Products Applications and Research Technologies, University of Oklahoma, Norman, Oklahoma, USA
| |
Collapse
|
20
|
Becker HEF, Demers K, Derijks LJJ, Jonkers DMAE, Penders J. Current evidence and clinical relevance of drug-microbiota interactions in inflammatory bowel disease. Front Microbiol 2023; 14:1107976. [PMID: 36910207 PMCID: PMC9996055 DOI: 10.3389/fmicb.2023.1107976] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 02/06/2023] [Indexed: 02/25/2023] Open
Abstract
Background Inflammatory bowel disease (IBD) is a chronic relapsing-remitting disease. An adverse immune reaction toward the intestinal microbiota is involved in the pathophysiology and microbial perturbations are associated with IBD in general and with flares specifically. Although medical drugs are the cornerstone of current treatment, responses vary widely between patients and drugs. The intestinal microbiota can metabolize medical drugs, which may influence IBD drug (non-)response and side effects. Conversely, several drugs can impact the intestinal microbiota and thereby host effects. This review provides a comprehensive overview of current evidence on bidirectional interactions between the microbiota and relevant IBD drugs (pharmacomicrobiomics). Methods Electronic literature searches were conducted in PubMed, Web of Science and Cochrane databases to identify relevant publications. Studies reporting on microbiota composition and/or drug metabolism were included. Results The intestinal microbiota can both enzymatically activate IBD pro-drugs (e.g., in case of thiopurines), but also inactivate certain drugs (e.g., mesalazine by acetylation via N-acetyltransferase 1 and infliximab via IgG-degrading enzymes). Aminosalicylates, corticosteroids, thiopurines, calcineurin inhibitors, anti-tumor necrosis factor biologicals and tofacitinib were all reported to alter the intestinal microbiota composition, including changes in microbial diversity and/or relative abundances of various microbial taxa. Conclusion Various lines of evidence have shown the ability of the intestinal microbiota to interfere with IBD drugs and vice versa. These interactions can influence treatment response, but well-designed clinical studies and combined in vivo and ex vivo models are needed to achieve consistent findings and evaluate clinical relevance.
Collapse
Affiliation(s)
- Heike E. F. Becker
- Division Gastroenterology-Hepatology, Department of Internal Medicine, NUTRIM School of Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, Netherlands
- Department of Medical Microbiology, Infectious Diseases and Infection Prevention, NUTRIM School of Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, Netherlands
| | - Karlijn Demers
- Division Gastroenterology-Hepatology, Department of Internal Medicine, NUTRIM School of Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, Netherlands
| | - Luc J. J. Derijks
- Department of Clinical Pharmacy and Pharmacology, Máxima Medical Center, Veldhoven, Netherlands
- Department of Clinical Pharmacy and Toxicology, Maastricht University Medical Centre+, Maastricht, Netherlands
| | - Daisy M. A. E. Jonkers
- Division Gastroenterology-Hepatology, Department of Internal Medicine, NUTRIM School of Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, Netherlands
| | - John Penders
- Department of Medical Microbiology, Infectious Diseases and Infection Prevention, NUTRIM School of Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, Netherlands
- Department of Medical Microbiology, Infectious Diseases and Infection Prevention, CAPHRI School of Public Health and Primary Care, Maastricht University Medical Centre+, Maastricht, Netherlands
| |
Collapse
|
21
|
Ndlovu K, Kannigadu C, Aucamp J, van Rensburg HDJ, N'Da DD. Exploration of ethylene glycol linked nitrofurantoin derivatives against Leishmania: Synthesis and in vitro activity. Arch Pharm (Weinheim) 2023; 356:e2200529. [PMID: 36759973 DOI: 10.1002/ardp.202200529] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/27/2022] [Accepted: 12/30/2022] [Indexed: 02/11/2023]
Abstract
Leishmaniasis is a neglected tropical disease that is caused by the Leishmania parasite. It is estimated that there are more than 350 million people at risk of infection annually. Current treatments that are in clinical use are expensive, have toxic side effects, and are facing parasitic resistance. Therefore, new drugs are urgently required. In the quest for new, safe, and cost-effective drugs, a series of novel ethylene glycol derivatives of nitrofurantoin was synthesised and the in vitro antileishmanial efficacy of the compounds tested against Leishmania donovani and Leishmania major strains. Arylated ethylene glycol derivatives were found to be the most potent, with submicromolar activity up to 294-fold greater than the parent compound nitrofurantoin. Analogues 2j and 2k had the best antipromastigote activities with submicromolar IC50 values against L. major IR-173 and antimonial-resistant L. donovani 9515 strains.
Collapse
Affiliation(s)
- Keitumetsi Ndlovu
- Centre of Excellence for Pharmaceutical Sciences, North-West University, Potchefstroom, South Africa
| | - Christina Kannigadu
- Centre of Excellence for Pharmaceutical Sciences, North-West University, Potchefstroom, South Africa
| | - Janine Aucamp
- Centre of Excellence for Pharmaceutical Sciences, North-West University, Potchefstroom, South Africa
| | | | - David D N'Da
- Centre of Excellence for Pharmaceutical Sciences, North-West University, Potchefstroom, South Africa
| |
Collapse
|
22
|
Zuma NH, Aucamp J, Janse van Rensburg HD, N'Da DD. Synthesis and in vitro antileishmanial activity of alkylene-linked nitrofurantoin-triazole hybrids. Eur J Med Chem 2023; 246:115012. [PMID: 36516584 DOI: 10.1016/j.ejmech.2022.115012] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 12/01/2022] [Accepted: 12/06/2022] [Indexed: 12/12/2022]
Abstract
Leishmaniasis is a vector-borne parasitic disease that mostly affects populations in tropical and sub-tropical countries. There is currently no protective anti-leishmanial vaccine and only a paucity of clinical drugs is available to treat this disease albeit their toxicity. Leishmaniasis is curable but its eradication and elimination have been hampered by the emergence of multidrug resistant strains of the causative pathogens. This heightens the necessity for new and effective antileishmanial drugs. In search for such agents, nitrofurantoin, a clinical antibiotic, was appended to triazole scaffold through alkylene linkers of various length, and the resulting hybrids were evaluated for in vitro antileishmanial efficacy against Leishmania (L.) parasite of two strains. The hybrid 13, harboring a n-pentylene linker was uncovered as a leishmanicidal hit with micromolar activity against antimonial-resistant L. donovani, the causative of deadly visceral Leishmaniasis.
Collapse
Affiliation(s)
- Nonkululeko H Zuma
- Centre of Excellence for Pharmaceutical Sciences, North-West University, Potchefstroom, 2520, South Africa
| | - Janine Aucamp
- Centre of Excellence for Pharmaceutical Sciences, North-West University, Potchefstroom, 2520, South Africa
| | | | - David D N'Da
- Centre of Excellence for Pharmaceutical Sciences, North-West University, Potchefstroom, 2520, South Africa.
| |
Collapse
|
23
|
Chen W, Sheng P, Chen Y, Liang Y, Wu S, Jia L, He X, Zhang CF, Wang CZ, Yuan CS. Hypoxia-responsive Immunostimulatory Nanomedicines Synergize with Checkpoint Blockade Immunotherapy for Potentiating Cancer Immunotherapy. CHEMICAL ENGINEERING JOURNAL (LAUSANNE, SWITZERLAND : 1996) 2023. [PMID: 37033201 DOI: 10.1016/j.cej.2022.134869] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Inducing cell death while simultaneously enhancing antitumor immune responses is a promising therapeutic approach for multiple cancers. Celastrol (Cel) and 7-ethyl-10-hydroxycamptothecin (SN38) have contrasting physicochemical properties, but strong synergy in immunogenic cell death induction and anticancer activity. Herein, a hypoxia-sensitive nanosystem (CS@TAP) was designed to demonstrate effective immunotherapy for colorectal cancer by systemic delivery of an immunostimulatory chemotherapy combination. Furthermore, the combination of CS@TAP with anti-PD-L1 mAb (αPD-L1) exhibited a significant therapeutic benefit of delaying tumor growth and increased local doses of immunogenic signaling and T-cell infiltration, ultimately extending survival. We conclude that CS@TAP is an effective inducer of immunogenic cell death (ICD) in cancer immunotherapy. Therefore, this study provides an encouraging strategy to synergistically induce immunogenic cell death to enhance tumor cytotoxic T lymphocytes (CTLs) infiltration for anticancer immunotherapy.
Collapse
Affiliation(s)
- Weiguo Chen
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Ping Sheng
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Yujiang Chen
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Yi Liang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Sixin Wu
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Liying Jia
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Xin He
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Chun-Feng Zhang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Chong-Zhi Wang
- Tang Center of Herbal Medicine Research and Department of Anesthesia & Critical Care, University of Chicago, Chicago, IL, 60637, USA
| | - Chun-Su Yuan
- Tang Center of Herbal Medicine Research and Department of Anesthesia & Critical Care, University of Chicago, Chicago, IL, 60637, USA
| |
Collapse
|
24
|
Simpson JB, Sekela JJ, Carry BS, Beaty V, Patel S, Redinbo MR. Diverse but desolate landscape of gut microbial azoreductases: A rationale for idiopathic IBD drug response. Gut Microbes 2023; 15:2203963. [PMID: 37122075 PMCID: PMC10132220 DOI: 10.1080/19490976.2023.2203963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 02/01/2023] [Accepted: 04/04/2023] [Indexed: 05/02/2023] Open
Abstract
Prodrugs reliant on microbial activation are widely used but exhibit a range of efficacies that remain poorly understood. The anti-inflammatory compound 5-aminosalicylic acid (5-ASA), which is packaged in a variety of azo-linked prodrugs provided to most Ulcerative Colitis (UC) patients, shows confounding inter-individual variabilities in response. Such prodrugs must be activated by azo-bond reduction to form 5-ASA, a process that has been attributed to both enzymatic and non-enzymatic catalysis. Gut microbial azoreductases (AzoRs) are the first catalysts shown to activate azo-linked drugs and to metabolize toxic azo-chemicals. Here, we chart the scope of the structural and functional diversity of AzoRs in health and in patients with the inflammatory bowel diseases (IBDs) UC and Crohn's Disease (CD). Using structural metagenomics, we define the landscape of gut microbial AzoRs in 413 healthy donor and 1059 IBD patient fecal samples. Firmicutes encode a significantly higher number of unique AzoRs compared to other phyla. However, structural and biochemical analyses of distinct AzoRs from the human microbiome reveal significant differences between prevalent orthologs in the processing of toxic azo-dyes, and their generally poor activation of IBD prodrugs. Furthermore, while individuals with IBD show higher abundances of AzoR-encoding gut microbial taxa than healthy controls, the overall abundance of AzoR-encoding microbes is markedly low in both disease and health. Together, these results establish that gut microbial AzoRs are functionally diverse but sparse in both health and disease, factors that may contribute to non-optimal processing of azo-linked prodrugs and idiopathic IBD drug responses.
Collapse
Affiliation(s)
- Joshua B. Simpson
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Josh J. Sekela
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Benjamin S. Carry
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Violet Beaty
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Shakshi Patel
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Matthew. R. Redinbo
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Biochemistry and Biophysics, Department of Microbiology and Immunology, and the Integrated Program for Biological and Genome Sciences, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
25
|
Chen W, Sheng P, Chen Y, Liang Y, Wu S, Jia L, He X, Zhang CF, Wang CZ, Yuan CS. Hypoxia-responsive Immunostimulatory Nanomedicines Synergize with Checkpoint Blockade Immunotherapy for Potentiating Cancer Immunotherapy. CHEMICAL ENGINEERING JOURNAL (LAUSANNE, SWITZERLAND : 1996) 2023; 451:138781. [PMID: 37033201 PMCID: PMC10079280 DOI: 10.1016/j.cej.2022.138781] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Inducing cell death while simultaneously enhancing antitumor immune responses is a promising therapeutic approach for multiple cancers. Celastrol (Cel) and 7-ethyl-10-hydroxycamptothecin (SN38) have contrasting physicochemical properties, but strong synergy in immunogenic cell death induction and anticancer activity. Herein, a hypoxia-sensitive nanosystem (CS@TAP) was designed to demonstrate effective immunotherapy for colorectal cancer by systemic delivery of an immunostimulatory chemotherapy combination. Furthermore, the combination of CS@TAP with anti-PD-L1 mAb (αPD-L1) exhibited a significant therapeutic benefit of delaying tumor growth and increased local doses of immunogenic signaling and T-cell infiltration, ultimately extending survival. We conclude that CS@TAP is an effective inducer of immunogenic cell death (ICD) in cancer immunotherapy. Therefore, this study provides an encouraging strategy to synergistically induce immunogenic cell death to enhance tumor cytotoxic T lymphocytes (CTLs) infiltration for anticancer immunotherapy.
Collapse
Affiliation(s)
- Weiguo Chen
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Ping Sheng
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Yujiang Chen
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Yi Liang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Sixin Wu
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Liying Jia
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Xin He
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Chun-Feng Zhang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Chong-Zhi Wang
- Tang Center of Herbal Medicine Research and Department of Anesthesia & Critical Care, University of Chicago, Chicago, IL, 60637, USA
| | - Chun-Su Yuan
- Tang Center of Herbal Medicine Research and Department of Anesthesia & Critical Care, University of Chicago, Chicago, IL, 60637, USA
| |
Collapse
|
26
|
Zhang C, Zheng R, Li S, Yang K, Tai S, Tao Y, Zhang S, Zhang K. Using a dual-emission Sm( iii)-macrocycle as the perceptive lab-on-a-molecule chemosensor toward selective and discriminative detection of nitroaromatic explosives. NEW J CHEM 2023. [DOI: 10.1039/d3nj00627a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/07/2023]
Abstract
A dual-emission Sm(iii)-macrocycle Sm-2l is designed as the perceptive lab-on-a-molecule toward selective and discriminative detection of nitroaromatic explosives by statistical analysis.
Collapse
Affiliation(s)
- Chengjian Zhang
- School of Chemistry and Chemical Engineering, Key Laboratory of Surface & Interface Science of Polymer Materials of Zhejiang Province, Zhejiang Sci-Tech University, Hangzhou 310018, P. R. China
| | - Ruijie Zheng
- School of Chemistry and Chemical Engineering, Key Laboratory of Surface & Interface Science of Polymer Materials of Zhejiang Province, Zhejiang Sci-Tech University, Hangzhou 310018, P. R. China
| | - Sichen Li
- School of Chemistry and Chemical Engineering, Key Laboratory of Surface & Interface Science of Polymer Materials of Zhejiang Province, Zhejiang Sci-Tech University, Hangzhou 310018, P. R. China
| | - Kang Yang
- School of Chemistry and Chemical Engineering, Key Laboratory of Surface & Interface Science of Polymer Materials of Zhejiang Province, Zhejiang Sci-Tech University, Hangzhou 310018, P. R. China
| | - Shengdi Tai
- School of Chemistry and Chemical Engineering, Key Laboratory of Surface & Interface Science of Polymer Materials of Zhejiang Province, Zhejiang Sci-Tech University, Hangzhou 310018, P. R. China
| | - Yinsong Tao
- School of Chemistry and Chemical Engineering, Key Laboratory of Surface & Interface Science of Polymer Materials of Zhejiang Province, Zhejiang Sci-Tech University, Hangzhou 310018, P. R. China
| | - Shishen Zhang
- School of Chemistry and Chemical Engineering, Key Laboratory of Surface & Interface Science of Polymer Materials of Zhejiang Province, Zhejiang Sci-Tech University, Hangzhou 310018, P. R. China
| | - Kun Zhang
- School of Chemistry and Chemical Engineering, Key Laboratory of Surface & Interface Science of Polymer Materials of Zhejiang Province, Zhejiang Sci-Tech University, Hangzhou 310018, P. R. China
| |
Collapse
|
27
|
Simpson JB, Redinbo MR. Multi-omic analysis of host-microbial interactions central to the gut-brain axis. Mol Omics 2022; 18:896-907. [PMID: 36169030 DOI: 10.1039/d2mo00205a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The gut microbiota impact numerous aspects of human physiology, including the central nervous system (CNS). Emerging work is now focusing on the microbial factors underlying the bi-directional communication network linking host and microbial systems within the gastrointestinal tract to the CNS, the "gut-brain axis". Neurotransmitters are key coordinators of this network, and their dysregulation has been linked to numerous neurological disease states. As the bioavailability of neurotransmitters is modified by gut microbes, it is critical to unravel the influence of the microbiota on neurotransmitters in the context of the gut-brain axis. Here we review foundational studies that defined molecular relationships between the microbiota, neurotransmitters, and the gut-brain axis. We examine links between the gut microbiome, behavior, and neurological diseases, as well as microbial influences on neurotransmitter bioavailability and physiology. Finally, we review multi-omics technologies uniquely applicable to this area, including high-throughput genetics, modern metabolomics, structure-guided metagenomics, targeted proteomics, and chemogenetics. Interdisciplinary studies will continue to drive the discovery of molecular mechanisms linking the gut microbiota to clinical manifestations of neurobiology.
Collapse
Affiliation(s)
- Joshua B Simpson
- Department of Chemistry, University of North Carolina at Chapel Hill, USA
| | - Matthew R Redinbo
- Department of Chemistry, University of North Carolina at Chapel Hill, USA
- Department of Biochemistry & Biophysics, Department of Microbiology & Immunology, and the Integrated Program in Biological & Genome Sciences, University of North Carolina at Chapel Hill, USA.
| |
Collapse
|
28
|
Lactose azocalixarene drug delivery system for the treatment of multidrug-resistant pseudomonas aeruginosa infected diabetic ulcer. Nat Commun 2022; 13:6279. [PMID: 36270992 PMCID: PMC9586954 DOI: 10.1038/s41467-022-33920-7] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 10/06/2022] [Indexed: 12/25/2022] Open
Abstract
Diabetic wound is one of the most intractable chronic wounds that is prone to bacterial infection. Hypoxia is an important feature in its microenvironment. However, it is challenging for antimicrobial therapy to directly apply the existing hypoxia-responsive drug delivery systems due to the active targeting deficiency and the biofilm obstacle. Herein, we customizes a hypoxia-responsive carrier, lactose-modified azocalix[4]arene (LacAC4A) with the ability to actively target and inhibit biofilm. By loading ciprofloxacin (Cip), the resultant supramolecular nanoformulation Cip@LacAC4A demonstrates enhanced antibacterial efficacy resulting from both the increased drug accumulation and the controlled release at the site of infection. When applied on diabetic wounds together with multidrug-resistant Pseudomonas aeruginosa infection in vivo, Cip@LacAC4A induces definitely less inflammatory infiltration than free Cip, which translates into high wound healing performance. Importantly, such design principle provides a direction for developing antimicrobial drug delivery systems.
Collapse
|
29
|
Cipriano A, Milite C, Feoli A, Viviano M, Pepe G, Campiglia P, Sarno G, Picaud S, Imaide S, Makukhin N, Filippakopoulos P, Ciulli A, Castellano S, Sbardella G. Discovery of Benzo[d]imidazole-6-sulfonamides as Bromodomain and Extra-Terminal Domain (BET) Inhibitors with Selectivity for the First Bromodomain. ChemMedChem 2022; 17:e202200343. [PMID: 36040095 PMCID: PMC9826262 DOI: 10.1002/cmdc.202200343] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/29/2022] [Indexed: 01/11/2023]
Abstract
The bromodomain and extra-terminal (BET) family of proteins includes BRD2, BRD3, BRD4, and the testis-specific protein, BRDT, each containing two N-terminal tandem bromodomain (BRD) modules. Potent and selective inhibitors targeting the two bromodomains are required to elucidate their biological role(s), with potential clinical applications. In this study, we designed and synthesized a series of benzimidazole-6-sulfonamides starting from the azobenzene compounds MS436 (7 a) and MS611 (7 b) that exhibited preference for the first (BD1) over the second (BD2) BRD of BET family members. The most-promising compound (9 a) showed good binding potency and improved metabolic stability and selectivity towards BD1 with respect to the parent compounds.
Collapse
Affiliation(s)
- Alessandra Cipriano
- Department of PharmacyUniversity of Salernovia Giovanni Paolo II 13284084Fisciano (SA)Italy
| | - Ciro Milite
- Department of PharmacyUniversity of Salernovia Giovanni Paolo II 13284084Fisciano (SA)Italy
| | - Alessandra Feoli
- Department of PharmacyUniversity of Salernovia Giovanni Paolo II 13284084Fisciano (SA)Italy
| | - Monica Viviano
- Department of PharmacyUniversity of Salernovia Giovanni Paolo II 13284084Fisciano (SA)Italy
| | - Giacomo Pepe
- Department of PharmacyUniversity of Salernovia Giovanni Paolo II 13284084Fisciano (SA)Italy
| | - Pietro Campiglia
- Department of PharmacyUniversity of Salernovia Giovanni Paolo II 13284084Fisciano (SA)Italy
| | - Giuliana Sarno
- Department of PharmacyUniversity of Salernovia Giovanni Paolo II 13284084Fisciano (SA)Italy
| | - Sarah Picaud
- Nuffield Department of MedicineOxford UniversityOX3 7DQOxfordUK
| | - Satomi Imaide
- Division of Biological Chemistry and Drug DiscoverySchool of Life SciencesUniversity of DundeeDow StreetDundeeDD1 5EH, ScotlandUK,Discovery Technology Research LaboratoriesOno Pharmaceutical Co., Ltd.618-8585OsakaJapan
| | - Nikolai Makukhin
- Division of Biological Chemistry and Drug DiscoverySchool of Life SciencesUniversity of DundeeDow StreetDundeeDD1 5EH, ScotlandUK,Oncology R&DTumour Targeted DeliveryAstraZenecaQMB Innovation Centre42 New RoadLondonE1 2AXUK
| | | | - Alessio Ciulli
- Division of Biological Chemistry and Drug DiscoverySchool of Life SciencesUniversity of DundeeDow StreetDundeeDD1 5EH, ScotlandUK
| | - Sabrina Castellano
- Department of PharmacyUniversity of Salernovia Giovanni Paolo II 13284084Fisciano (SA)Italy
| | - Gianluca Sbardella
- Department of PharmacyUniversity of Salernovia Giovanni Paolo II 13284084Fisciano (SA)Italy
| |
Collapse
|
30
|
Wolfson SJ, Hitchings R, Peregrina K, Cohen Z, Khan S, Yilmaz T, Malena M, Goluch ED, Augenlicht L, Kelly L. Bacterial hydrogen sulfide drives cryptic redox chemistry in gut microbial communities. Nat Metab 2022; 4:1260-1270. [PMID: 36266544 PMCID: PMC11328334 DOI: 10.1038/s42255-022-00656-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 09/07/2022] [Indexed: 01/20/2023]
Abstract
Microbial biochemistry contributes to a dynamic environment in the gut. Yet, how bacterial metabolites such as hydrogen sulfide (H2S) mechanistically alter the gut chemical landscape is poorly understood. Here we show that microbially generated H2S drives the abiotic reduction of azo (R-N = N-R') xenobiotics, which are commonly found in Western food dyes and drugs. This nonenzymatic reduction of azo compounds is demonstrated in Escherichia coli cultures, in human faecal microbial communities and in vivo in male mice. Changing dietary levels of the H2S xenobiotic redox partner Red 40 transiently decreases mouse faecal sulfide levels, demonstrating that a xenobiotic can attenuate sulfide concentration and alleviate H2S accumulation in vivo. Cryptic H2S redox chemistry thus can modulate sulfur homeostasis, alter the chemical landscape in the gut and contribute to azo food dye and drug metabolism. Interactions between chemicals derived from microbial communities may be a key feature shaping metabolism in the gut.
Collapse
Affiliation(s)
- Sarah J Wolfson
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Reese Hitchings
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Karina Peregrina
- Departments of Medicine and Cell Biology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Ziv Cohen
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Saad Khan
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Tugba Yilmaz
- Department of Chemical Engineering, Northeastern University, Boston, MA, USA
| | - Marcel Malena
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Edgar D Goluch
- Department of Chemical Engineering, Northeastern University, Boston, MA, USA
| | - Leonard Augenlicht
- Departments of Medicine and Cell Biology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Libusha Kelly
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, Bronx, NY, USA.
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
31
|
Bu A, Zhao Y, Xiao H, Tung C, Wu L, Cong H. A Conjugated Covalent Template Strategy for All‐Benzene Catenane Synthesis. Angew Chem Int Ed Engl 2022; 61:e202209449. [DOI: 10.1002/anie.202209449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Indexed: 11/11/2022]
Affiliation(s)
- An Bu
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials Technical Institute of Physics and Chemistry Chinese Academy of Sciences Beijing 100190 China
- School of Future Technology University of Chinese Academy of Sciences Chinese Academy of Sciences Beijing 100190 China
| | - Yongye Zhao
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials Technical Institute of Physics and Chemistry Chinese Academy of Sciences Beijing 100190 China
| | - Hongyan Xiao
- Key Laboratory of Bio-inspired Materials and Interfacial Science Technical Institute of Physics and Chemistry Chinese Academy of Sciences Beijing 100190 China
| | - Chen‐Ho Tung
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials Technical Institute of Physics and Chemistry Chinese Academy of Sciences Beijing 100190 China
- School of Future Technology University of Chinese Academy of Sciences Chinese Academy of Sciences Beijing 100190 China
| | - Li‐Zhu Wu
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials Technical Institute of Physics and Chemistry Chinese Academy of Sciences Beijing 100190 China
- School of Future Technology University of Chinese Academy of Sciences Chinese Academy of Sciences Beijing 100190 China
| | - Huan Cong
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials Technical Institute of Physics and Chemistry Chinese Academy of Sciences Beijing 100190 China
- School of Future Technology University of Chinese Academy of Sciences Chinese Academy of Sciences Beijing 100190 China
| |
Collapse
|
32
|
Wang X, Sun B, Ye Z, Zhang W, Xu W, Gao S, Zhou N, Wu F, Shen J. Enzyme-Responsive COF-Based Thiol-Targeting Nanoinhibitor for Curing Bacterial Infections. ACS APPLIED MATERIALS & INTERFACES 2022; 14:38483-38496. [PMID: 35989491 DOI: 10.1021/acsami.2c08845] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Pathogen infections impose severe challenges in clinical practice, especially for patients infected with antibiotic-resistant microbes. The thioredoxin (Trx) system in Gram-positive bacteria serves as an ideal antimicrobial target for novel medicine design due to the structural differences from corresponding system in mammals. However, a backup thiol-dependent antioxidant glutathione (GSH) system limits the effectiveness of drugs in many Gram-negative bacteria. Herein, we synthesize a thiol-targeting nanoinhibitor based on an enzyme-responsive covalent organic framework (COF) coloaded with silver nanoparticles (AgNPs) and ebselen (EBS) (Ag-TA-CON@EBS@PEG) to exert synergistic antibacterial effects. Since azoreductase can dissociate the enzyme-responsive COF, we adopt this strategy to achieve the accurate release of EBS and Ag+ at infection sites. Our research identifies that the functionalized nanoinhibitor shows excellent bactericidal performance for Gram-positive and Gram-negative bacteria in vitro and exhibits low toxicity to normal cells. Besides, the nanoinhibitor presents favorable biocompatibility, anti-inflammatory property, and effective wound healing ability in mice. This paper provides a promising clinical strategy for synergistic antibacterial therapy and enhanced wound healing properties via an optimized combination of the targeted nanomedicines with an intelligent drug conveying platform.
Collapse
Affiliation(s)
- Xinye Wang
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Baohong Sun
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Ziqiu Ye
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Wenjia Zhang
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Wang Xu
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Shurui Gao
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Ninglin Zhou
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Fan Wu
- Key Laboratory of Cardiovascular & Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Jian Shen
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| |
Collapse
|
33
|
Walker ME, Simpson JB, Redinbo MR. A structural metagenomics pipeline for examining the gut microbiome. Curr Opin Struct Biol 2022; 75:102416. [PMID: 35841748 PMCID: PMC10039758 DOI: 10.1016/j.sbi.2022.102416] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 04/25/2022] [Accepted: 05/18/2022] [Indexed: 12/13/2022]
Abstract
Metagenomic sequencing data provide a rich resource from which to expand our understanding of differential protein functions involved in human health. Here, we outline a pipeline that combines microbial whole genome sequencing with protein structure data to yield a structural metagenomics-informed atlas of microbial enzyme families of interest. Visualizing metagenomics data through a structural lens facilitates downstream studies including targeted inhibition and probe-based proteomics to define at the molecular level how different enzyme orthologs impact in vivo function. Application of this pipeline to gut microbial enzymes like glucuronidases, TMA lyases, and bile salt hydrolases is expected to pinpoint their involvement in health and disease and may aid in the development of therapeutics that target specific enzymes within the microbiome.
Collapse
Affiliation(s)
- Morgan E Walker
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Joshua B Simpson
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Matthew R Redinbo
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Integrated Program for Biological and Genome Sciences, And Departments of Biochemistry and Microbiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
34
|
Bu A, Zhao Y, Xiao H, Tung CH, Wu LZ, Cong H. Conjugated Covalent Template Strategy for All‐Benzene Catenane Synthesis. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202209449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- An Bu
- Technical Institute of Physics and Chemistry Key Laboratory of Photochemical Conversion and Optoelectronic Materials CHINA
| | - Yongye Zhao
- Technical Institute of Physics and Chemistry Key Laboratory of Photochemical Conversion and Optoelectronic Materials CHINA
| | - Hongyan Xiao
- Technical Institute of Physics and Chemistry Key Laboratory of Bio-inspired Materials and Interfacial Science CHINA
| | - Chen-Ho Tung
- Technical Institute of Physics and Chemistry Key Laboratory of Photochemical Conversion and Optoelectronic Materials CHINA
| | - Li-Zhu Wu
- Technical Institute of Physics and Chemistry Key Laboratory of Photochemical Conversion and Optoelectronic Materials CHINA
| | - Huan Cong
- Technical Institute of Physics and Chemistry CAS: Technical Institute of Physics and Chemistry Key Laboratory of Photochemical Conversion and Optoelectronic Materials No.29 Zhongguancun East Road 100190 Beijing CHINA
| |
Collapse
|
35
|
New Bioprecursor Prodrugs of Sulfadiazine: Synthesis, X-ray Structure and Hirshfeld Analysis. CRYSTALS 2022. [DOI: 10.3390/cryst12081016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Sulphonamide motif is found extensively in numerous chemotherapeutic drug candidates, it acts by stopping the production of folate inside the bacterial cell. Current research has established the synthesis and characterization of new bioprecursor prodrugs of sulfadiazine. The first prodrug, 3, was synthesized via the coupling of diazonium salt of sulfadiazine with ethyl acetoacetate in AcONa at 0 °C. The second prodrug, sulfadiazine-pyrazole, 5, was furnished via cyclocondensation of the hydrazono derivative, 3, and 2-pyridyl hydrazine, 4. The generated data from the X-ray analysis is interpreted and refined to obtain the crystal structure of the target compound, 5. Density functional theory (DFT) method was used to calculate the optimized geometrical parameters, electronic state (HOMO–LUMO), and the electronic properties. Moreover, Hirshfeld analysis revealed that the most important contributions to the crystal packing of the prodrug 5 are H···H, O···H and H···C contacts.
Collapse
|
36
|
Kannigadu C, Aucamp J, N'Da DD. Exploring novel nitrofuranyl sulfonohydrazides as anti-Leishmania and anti-cancer agents: Synthesis, in vitro efficacy and hit identification. Chem Biol Drug Des 2022; 100:267-279. [PMID: 35648075 PMCID: PMC9546217 DOI: 10.1111/cbdd.14097] [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: 03/11/2022] [Revised: 05/06/2022] [Accepted: 05/29/2022] [Indexed: 11/28/2022]
Abstract
Leishmaniasis and cancer are two deadly diseases that plague the human population. There are a limited number of drugs available for the treatment of these diseases; however, their overuse has resulted in pathogenic resistance. Recent studies have indicated the repurposing of nitro‐containing compounds to be a new avenue into finding new treatments. In this study, new nitrofuranyl sulfonohydrazide derivatives were synthesized and evaluated for their in vitro antileishmanial and anticancer activities. The analogue 2h, featuring biphenyl moiety exhibited selective (SI > 10) submicromolar activity (IC50 0.97 μM) against acute promyelocytic leukemia cells hence was identified anticancer hit. This study revealed no antileishmanial hit. However, several promising analogues were uncovered and are worthy of further structural modifications to improve their toxicity and bioactivity profiles.
Collapse
Affiliation(s)
- Christina Kannigadu
- Centre of Excellence for Pharmaceutical Sciences, North-West University, Potchefstroom, South Africa
| | - Janine Aucamp
- Centre of Excellence for Pharmaceutical Sciences, North-West University, Potchefstroom, South Africa
| | - David D N'Da
- Centre of Excellence for Pharmaceutical Sciences, North-West University, Potchefstroom, South Africa
| |
Collapse
|
37
|
Zuma NH, Aucamp J, Viljoen M, N'Da DD. Synthesis, in vitro Antileishmanial Efficacy and Hit/Lead Identification of Nitrofurantoin-Triazole Hybrids. ChemMedChem 2022; 17:e202200023. [PMID: 35388649 PMCID: PMC9322565 DOI: 10.1002/cmdc.202200023] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 04/04/2022] [Indexed: 11/09/2022]
Abstract
Leishmaniasis is a vector-borne neglected parasitic infection affecting thousands of individuals, mostly among populations in low- to moderate-income developing countries. In the absence of protective vaccines, the management of the disease banks solely on chemotherapy. However, the clinical usefulness of current antileishmanial drugs is threatened by their toxicity and the emergence of multidrug-resistant strains of the causative pathogens. This emphasizes the imperative for the development of new and effective antileishmanial agents. In this regard, we synthesized and evaluated in vitro the antileishmanial activity and cytotoxicity profile of a series of nitrofurantoin-triazole hybrids. The nitrofurantoin derivative 1 featuring propargyl moiety was distinctively the most active of all, was nontoxic to human cells and possessed submicromolar cellular activity selectively directed towards the pathogens of the life threatening visceral leishmaniasis. Hence it was identified as potential antileishmanial lead for further investigation into its prospective to act as alternative to therapies.
Collapse
Affiliation(s)
- Nonkululeko H. Zuma
- Centre of Excellence for Pharmaceutical Sciences (Pharmacen)Faculty of Health SciencesNorth-West University11 Hoffmann StreetPotchefstroom2520South Africa
| | - Janine Aucamp
- Centre of Excellence for Pharmaceutical Sciences (Pharmacen)Faculty of Health SciencesNorth-West University11 Hoffmann StreetPotchefstroom2520South Africa
| | - Maryna Viljoen
- School of Pharmacy, Faculty of Health SciencesNorth-West University11 Hoffmann StreetPotchefstroom2520South Africa
| | - David D. N'Da
- Centre of Excellence for Pharmaceutical Sciences (Pharmacen)Faculty of Health SciencesNorth-West University11 Hoffmann StreetPotchefstroom2520South Africa
| |
Collapse
|
38
|
Microbial Degradation of Azo Dyes: Approaches and Prospects for a Hazard-Free Conversion by Microorganisms. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19084740. [PMID: 35457607 PMCID: PMC9026373 DOI: 10.3390/ijerph19084740] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/08/2022] [Accepted: 04/11/2022] [Indexed: 11/16/2022]
Abstract
Azo dyes have become a staple in various industries, as colors play an important role in consumer choices. However, these dyes pose various health and environmental risks. Although different wastewater treatments are available, the search for more eco-friendly options persists. Bioremediation utilizing microorganisms has been of great interest to researchers and industries, as the transition toward greener solutions has become more in demand through the years. This review tackles the health and environmental repercussions of azo dyes and its metabolites, available biological approaches to eliminate such dyes from the environment with a focus on the use of different microorganisms, enzymes that are involved in the degradation of azo dyes, and recent trends that could be applied for the treatment of azo dyes.
Collapse
|
39
|
Basharat Z, Yasmin A. Sulphonated azo dye decolorization by Alcaligenes faecalis subsp. phenolicus MB207: Insights from laboratory and computational analysis. Biophys Chem 2022; 286:106806. [DOI: 10.1016/j.bpc.2022.106806] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 03/14/2022] [Accepted: 03/20/2022] [Indexed: 01/02/2023]
|
40
|
Liu H, Yang J, Yan X, Li C, Elsabahy M, Chen L, Yang YW, Gao H. A dendritic polyamidoamine supramolecular system composed of pillar[5]arene and azobenzene for targeting drug-resistant colon cancer. J Mater Chem B 2021; 9:9594-9605. [PMID: 34783814 DOI: 10.1039/d1tb02134f] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Fusobacterium nucleatum caused drug-resistant around tumor sites often leads to the failure of chemotherapy during colorectal cancer (CRC) treatment. Multifunctional cationic quaternary ammonium materials have been widely used as broad-spectrum antibacterial agents in antibacterial and anticancer fields. Herein, we design a smart supramolecular quaternary ammonium nanoparticle, namely quaternary ammonium PAMAM-AZO@CP[5]A (Q-P-A@CP[5]A), consisting of azobenzene (AZO)-conjugated dendritic cationic quaternary ammonium polyamidoamine (PAMAM) as the core and carboxylatopillar[5]arene (CP[5]A)-based switch, for antibacterial and anti-CRC therapies. The quaternary ammonium-PAMAM-AZO (Q-P-A) core endows the supramolecular system with enhanced antibacterial and anticancer properties. -N+CH3 groups on the surface of Q-P-A are accommodated in the CP[5]A cavity under normal conditions, which significantly improves the biocompatibility of Q-P-A@CP[5]A. Meanwhile, the CP[5]A host can be detached from -N+CH3 groups under pathological conditions, achieving efficient antibacterial and antitumor therapies. Furthermore, azoreductase in the tumor site can break the -NN- bonds of AZO in Q-P-A@CP[5]A, leading to the morphology recovery of supramolecular nanoparticles and CRC therapy through inducing cell membrane rupture. Both in vitro and in vivo experiments demonstrate that Q-P-A@CP[5]A possesses good biocompatibility, excellent antibacterial effect, and CRC treatment capability with negligible side effects. This supramolecular quaternary ammonium system provides an effective treatment method to overcome chemotherapy-resistant cancer caused by bacteria.
Collapse
Affiliation(s)
- Hongyu Liu
- State Key Laboratory of Separation Membranes and Membrane Processes, School of Materials Science and Engineering, Tiangong University, Tianjin 300387, P. R. China. .,Tianjin Key Laboratory of Drug Targeting and Bioimaging, Tianjin Enterprise Key Laboratory for Application Research of Hyaluronic Acid, School of Chemistry and Chemical Engineering, Tianjin University of Technology, Tianjin 300384, P. R. China
| | - Jie Yang
- International Joint Research Laboratory of Nano-Micro Architecture Chemistry, College of Chemistry, Jilin University, 2699 Qianjin Street, Changchun 130012, P. R. China.
| | - Xiangjie Yan
- Tianjin Key Laboratory of Drug Targeting and Bioimaging, Tianjin Enterprise Key Laboratory for Application Research of Hyaluronic Acid, School of Chemistry and Chemical Engineering, Tianjin University of Technology, Tianjin 300384, P. R. China
| | - Chaoqi Li
- Tianjin Key Laboratory of Drug Targeting and Bioimaging, Tianjin Enterprise Key Laboratory for Application Research of Hyaluronic Acid, School of Chemistry and Chemical Engineering, Tianjin University of Technology, Tianjin 300384, P. R. China
| | - Mahmoud Elsabahy
- Science Academy, School of Biotechnology, Badr University in Cairo, Badr City, Cairo 11829, Egypt
| | - Li Chen
- State Key Laboratory of Separation Membranes and Membrane Processes, School of Materials Science and Engineering, Tiangong University, Tianjin 300387, P. R. China.
| | - Ying-Wei Yang
- International Joint Research Laboratory of Nano-Micro Architecture Chemistry, College of Chemistry, Jilin University, 2699 Qianjin Street, Changchun 130012, P. R. China.
| | - Hui Gao
- State Key Laboratory of Separation Membranes and Membrane Processes, School of Materials Science and Engineering, Tiangong University, Tianjin 300387, P. R. China. .,Tianjin Key Laboratory of Drug Targeting and Bioimaging, Tianjin Enterprise Key Laboratory for Application Research of Hyaluronic Acid, School of Chemistry and Chemical Engineering, Tianjin University of Technology, Tianjin 300384, P. R. China
| |
Collapse
|
41
|
Kapalatiya H, Madav Y, Tambe VS, Wairkar S. Enzyme-responsive smart nanocarriers for targeted chemotherapy: an overview. Drug Deliv Transl Res 2021; 12:1293-1305. [PMID: 34251612 DOI: 10.1007/s13346-021-01020-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/22/2021] [Indexed: 02/02/2023]
Abstract
Nanocarriers play pivotal roles in the field of biomedical applications, particularly in anticancer therapy. One of the prominent strategies for the transport of anticancer drugs with site-specific release and improved therapeutic efficacy is the use of an enzyme-responsive drug delivery system. There is an emerging class of cancer therapeutics engineered to control the release of a drug via enzymatic degradation. Enzymes, being an essential component of bio-nanotechnology toolbox, hold exceptional biorecognition abilities as well as outstanding catalytic properties. Often, abnormal enzyme expression observed in cancer offers many opportunities in designing nanocarriers modified with enzyme-labile linkage. Through altered physical or chemical characteristics of these nanocarriers or cleavage of the drug in response to the bio-action of enzyme, an on-demand drug release can be obtained. In this review, several classes of enzymes performing critical roles in cancer such as hydrolases, lipases, and oxidoreductases are summarized. Insights on various approaches that interfere with the mechanism of these enzymes have also been included. Finally, various smart nanocarriers such as mesoporous silica nanoparticles, gold nanoparticles, carbon-nanotubes, micelles, liposomes, and dendrimers serving as excellent platforms for enzyme-responsive formulations have been discussed.
Collapse
Affiliation(s)
- Hiral Kapalatiya
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKMs NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai, Maharashtra, 400056, India
| | - Yamini Madav
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKMs NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai, Maharashtra, 400056, India
| | - Varunesh Sanjay Tambe
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKMs NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai, Maharashtra, 400056, India
| | - Sarika Wairkar
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKMs NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai, Maharashtra, 400056, India.
| |
Collapse
|
42
|
Targeted Cancer Therapy Using Compounds Activated by Light. Cancers (Basel) 2021; 13:cancers13133237. [PMID: 34209493 PMCID: PMC8269035 DOI: 10.3390/cancers13133237] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 06/18/2021] [Accepted: 06/24/2021] [Indexed: 12/21/2022] Open
Abstract
Cancer chemotherapy is affected by a modest selectivity and toxic side effects of pharmacological interventions. Among novel approaches to overcome this limitation and to bring to therapy more potent and selective agents is the use of light for selective activation of anticancer compounds. In this review, we focus on the anticancer applications of two light-activated approaches still in the experimental phase: photoremovable protecting groups ("photocages") and photoswitches. We describe the structural considerations behind the development of novel compounds and the plethora of assays used to confirm whether the photochemical and pharmacological properties are meeting the stringent criteria for an efficient in vivo light-dependent activation. Despite its immense potential, light activation brings many challenges, and the complexity of the task is very demanding. Currently, we are still deeply in the phase of pharmacological tools, but the vivid research and rapid development bring the light of hope for potential clinical use.
Collapse
|
43
|
Sidhu JS, Kaur N, Singh N. Trends in small organic fluorescent scaffolds for detection of oxidoreductase. Biosens Bioelectron 2021; 191:113441. [PMID: 34167075 DOI: 10.1016/j.bios.2021.113441] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 05/23/2021] [Accepted: 06/11/2021] [Indexed: 12/18/2022]
Abstract
Oxidoreductases are diverse class of enzymes engaged in modulating the redox homeostasis and cellular signaling cascades. Abnormal expression of oxidoreductases including thioredoxin reductase, azoreductase, cytochrome oxidoreductase, tyrosinase and monoamine oxidase leads to the initiation of numerous disorders. Thus, enzymes are the promising biomarkers of the diseased cells and their accurate detection has utmost significance for clinical diagnosis. The detection method must be extremely selective, sensitive easy to use, long self-life, mass manufacturable and disposable. Fluorescence assay approach has been developed potential substitute to conventional techniques used in enzyme's quantification. The fluorescent probes possess excellent stability, high spatiotemporal ratio and reproducibility represent applications in real sample analysis. Therefore, the enzymatic transformations have been monitored by small activatable organic fluorescent probes. These probes are generally integrated with enzyme's substrate/inhibitors to improve their binding affinity toward the enzyme's catalytic site. As the recognition unit bio catalyzed, the signaling unit produces the readout signals and provides novel insights to understand the biochemical reactions for diagnosis and development of point of care devices. Several structural modifications are required in fluorogenic scaffolds to tune the selectivity for a particular enzyme. Hence, the fluorescent probes with their structural features and enzymatic reaction mechanism of oxidoreductase are the key points discussed in this review. The basic strategies to detect each enzyme are discussed. The selectivity, sensitivity and real-time applications are critically compared. The kinetic parameters and futuristic opportunities are present, which would be enormous benefits for chemists and biologists to understand the facts to design and develop unique fluorophore molecules for clinical applications.
Collapse
Affiliation(s)
- Jagpreet Singh Sidhu
- Department of Chemistry, Indian Institute of Technology Ropar, Rupnagar, Punjab, 140001, India; Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Bathinda, India
| | - Navneet Kaur
- Department of Chemistry, Panjab University, Chandigarh, 160014, India
| | - Narinder Singh
- Department of Chemistry, Indian Institute of Technology Ropar, Rupnagar, Punjab, 140001, India.
| |
Collapse
|
44
|
Thomas C, Gwenin CD. The Role of Nitroreductases in Resistance to Nitroimidazoles. BIOLOGY 2021; 10:388. [PMID: 34062712 PMCID: PMC8147198 DOI: 10.3390/biology10050388] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 02/04/2021] [Accepted: 02/10/2021] [Indexed: 01/14/2023]
Abstract
Antimicrobial resistance is a major challenge facing modern medicine, with an estimated 700,000 people dying annually and a global cost in excess of $100 trillion. This has led to an increased need to develop new, effective treatments. This review focuses on nitroimidazoles, which have seen a resurgence in interest due to their broad spectrum of activity against anaerobic Gram-negative and Gram-positive bacteria. The role of nitroreductases is to activate the antimicrobial by reducing the nitro group. A decrease in the activity of nitroreductases is associated with resistance. This review will discuss the resistance mechanisms of different disease organisms, including Mycobacterium tuberculosis, Helicobacter pylori and Staphylococcus aureus, and how these impact the effectiveness of specific nitroimidazoles. Perspectives in the field of nitroimidazole drug development are also summarised.
Collapse
Affiliation(s)
- Carol Thomas
- School of Natural Sciences, Bangor University, Bangor LL57 2UW, UK;
| | - Christopher D. Gwenin
- Department of Chemistry, Xi’an Jiaotong-Liverpool University, 111 Ren’ai Road, Suzhou Industrial Park, Suzhou 215123, China
| |
Collapse
|
45
|
Cerón-Carrasco JP, Jacquemin D. Using Theory To Extend the Scope of Azobenzene Drugs in Chemotherapy: Novel Combinations for a Specific Delivery. ChemMedChem 2021; 16:1764-1774. [PMID: 33619857 DOI: 10.1002/cmdc.202100046] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 02/18/2021] [Indexed: 12/12/2022]
Abstract
Gut microorganisms metabolize azobenzene compounds (Ph1 -N=N-Ph2 ) into free aniline products (Ph1 -NH2 +H2 N-Ph2 ), a process that has been largely investigated to reduce dyes residues in the textile industry. However, the action of bacterial core enzymes such as azoreductases (AzoR) might also help to deliver prodrugs that become active when they reach the colonic region, a mechanism with potential applications for the treatment of inflammatory bowel disease (IBD) and colorectal cancer. So far, three azo-bonded prodrugs of 5-aminosalicylic acid (5-ASA), for example, sulfasalazine, olsalazine and balsalazide, have been used for colon-targeted delivery. The present contribution describes the first rational design of a novel azobenzene prodrug thanks to a computational approach, with a focus on linking 5-ASA to another approved anti-inflammatory drug. The resulting prodrugs were assessed for their degradation upon AzoR action. Replacing the original carriers by irsogladine is found to improve action.
Collapse
Affiliation(s)
- José P Cerón-Carrasco
- Reconocimiento y Encapsulación Molecular, Universidad Católica San Antonio de Murcia (UCAM) Campus los Jerónimos, 30107, Murcia, Spain
| | - Denis Jacquemin
- CEISAM UMR CNRS 6230, Université de Nantes, 44000, Nantes, France
| |
Collapse
|
46
|
Quantification and Metabolite Identification of Sulfasalazine in Mouse Brain and Plasma Using Quadrupole-Time-of-Flight Mass Spectrometry. Molecules 2021; 26:molecules26041179. [PMID: 33671835 PMCID: PMC7926890 DOI: 10.3390/molecules26041179] [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: 01/21/2021] [Revised: 02/10/2021] [Accepted: 02/18/2021] [Indexed: 11/16/2022] Open
Abstract
Sulfasalazine (SAS), an anti-inflammatory drug with potent cysteine/glutamate antiporter system xc-(SXC) inhibition has recently shown beneficial effects in brain-related diseases. Despite many reports related to central nervous system (CNS) effect of SAS, pharmacokinetics (PK) and metabolite identification studies in the brain for SAS were quite limited. The aim of this study was to investigate the pharmacokinetics and metabolite identification of SAS and their distributions in mouse brain. Using in vivo brain exposure studies (neuro PK), the PK parameters of SAS was calculated for plasma as well as brain following intravenous and oral administration at 10 mg/kg and 50 mg/kg in mouse, respectively. In addition, in vivo metabolite identification (MetID) studies of SAS in plasma and brain were also conducted. The concentration of SAS in brain was much lower than that in plasma and only 1.26% of SAS was detected in mouse brain when compared to the SAS concentration in plasma (brain to plasma ratio (%): 1.26). In the MetID study, sulfapyridine (SP), hydroxy-sulfapyridine (SP-OH), and N-acetyl sulfapyridine (Ac-SP) were identified in plasma, whereas only SP and Ac-SP were identified as significant metabolites in brain. As a conclusion, our results suggest that the metabolites of SAS such as SP and Ac-SP might be responsible for the pharmacological effect in brain, not the SAS itself.
Collapse
|
47
|
Bafana A, Khan F, Suguna K. Purification, characterization, and crystal structure of YhdA-type azoreductase from Bacillus velezensis. Proteins 2020; 89:483-492. [PMID: 33289153 DOI: 10.1002/prot.26032] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 11/06/2020] [Accepted: 11/28/2020] [Indexed: 12/13/2022]
Abstract
Azoreductases are being extensively investigated for their ability to initiate degradation of recalcitrant azo dyes through reduction of azo bonds. There is great interest in studying their diversity, structure, and function to facilitate better understanding and effective application. Current study reports azoreductase enzyme from Bacillus velezensis, which showed 69.5% identity to the Bacillus subtilis azoreductase YhdA. The enzyme was homotetrameric and molecular weight of each subunit was 20 kDa. It decolorized azo dyes with different structures. The Vmax for decolorization of congo red, methyl orange and methyl red was 14.7, 28.6, and 77.9 nmol/min/mg, respectively. The enzyme contained FMN as cofactor and used NADPH as the favored co-substrate. It was oxygen-insensitive, but the presence of reducing agents enhanced its activity, which is a new finding. The azoreductase expression in B. velezensis was found to be unaffected by addition of azo dyes, although azo dyes are known to induce azoreductase expression in few organisms. The enzyme was thermostable with melting temperature of 89.5°C and functioned in wide temperature range. Further, the enzyme was crystallized and its structure was solved. The structural basis of its functional attributes is discussed. In our knowledge, this is the first report on characterization of azoreductase enzyme from B. velezensis.
Collapse
Affiliation(s)
- Amit Bafana
- Director's Research Cell, CSIR-NEERI (National Environmental Engineering Research Institute), Nagpur, India
| | - Farha Khan
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, India.,David Geffen School of Medicine, Department of Physiology, University of California, Los Angeles, Los Angeles, USA
| | - Kaza Suguna
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, India
| |
Collapse
|
48
|
Kannigadu C, Aucamp J, N'Da DD. Synthesis and in vitro antileishmanial efficacy of benzyl analogues of nifuroxazide. Drug Dev Res 2020; 82:287-295. [PMID: 33141473 DOI: 10.1002/ddr.21755] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 10/12/2020] [Accepted: 10/16/2020] [Indexed: 12/22/2022]
Abstract
Leishmaniasis is a vector-borne parasitic disease that mostly affects populations in tropical and subtropical countries. There is currently no vaccine to protect against and only a handful of drugs are available to treat this disease. Leishmaniasis is curable, but its eradication and elimination are hindered by the emergence of multidrug resistant strains of the causative pathogens, accentuating the need for new and effective antileishmanial drugs. In search for such agents, nifuroxazide, a clinical antibiotic, was evaluated through investigation of its benzyl analogues for in vitro antileishmanial efficacy against promastigotes of various Leishmania (L.) strains. The monobenzylated analogues 1 and 2 were the most potent of all, possessing nanomolar activities up to 10-fold higher than the parent drug nifuroxazide against all three tested Leishmania strains. Both analogues stand as antipromastigote hits for further lead investigation into their potential to act as new antileishmanial agents.
Collapse
Affiliation(s)
- Christina Kannigadu
- Centre of Excellence for Pharmaceutical Sciences, North-West University, Potchefstroom, South Africa
| | - Janine Aucamp
- Centre of Excellence for Pharmaceutical Sciences, North-West University, Potchefstroom, South Africa
| | - David D N'Da
- Centre of Excellence for Pharmaceutical Sciences, North-West University, Potchefstroom, South Africa
| |
Collapse
|
49
|
Yan VC, Butterfield HE, Poral AH, Yan MJ, Yang KL, Pham CD, Muller FL. Why Great Mitotic Inhibitors Make Poor Cancer Drugs. Trends Cancer 2020; 6:924-941. [PMID: 32536592 PMCID: PMC7606322 DOI: 10.1016/j.trecan.2020.05.010] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 05/12/2020] [Accepted: 05/19/2020] [Indexed: 12/13/2022]
Abstract
Chemotherapy is central to oncology, perceived to operate only on prolific cancerous tissue. Yet, many non-neoplastic tissues are more prolific compared with typical tumors. Chemotherapies achieve sufficient therapeutic windows to exert antineoplastic activity because they are prodrugs that are bioactivated in cancer-specific environments. The advent of precision medicine has obscured this concept, favoring the development of high-potency kinase inhibitors. Inhibitors of essential mitotic kinases exemplify this paradigm shift, but intolerable on-target toxicities in more prolific normal tissues have led to repeated failures in the clinic. Proliferation rates alone cannot be used to achieve cancer specificity. Here, we discuss integrating the cancer specificity of prodrugs from classical chemotherapeutics and the potency of mitotic kinase inhibitors to generate a class of high-precision cancer therapeutics.
Collapse
Affiliation(s)
- Victoria C Yan
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA.
| | | | - Anton H Poral
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Matthew J Yan
- Department of Chemistry, Boston College, Chestnut Hill, MA 02467, USA
| | - Kristine L Yang
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Cong-Dat Pham
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Florian L Muller
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA.
| |
Collapse
|
50
|
Mechanistic Understanding Enables the Rational Design of Salicylanilide Combination Therapies for Gram-Negative Infections. mBio 2020; 11:mBio.02068-20. [PMID: 32934086 PMCID: PMC7492738 DOI: 10.1128/mbio.02068-20] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
There is a critical need for more-effective treatments to combat multidrug-resistant Gram-negative infections. Combination therapies are a promising strategy, especially when these enable existing clinical drugs to be repurposed as antibiotics. We examined the mechanisms of action and basis of innate Gram-negative resistance for the anthelmintic drug niclosamide and subsequently exploited this information to demonstrate that niclosamide and analogs kill Gram-negative bacteria when combined with antibiotics that inhibit drug efflux or permeabilize membranes. We confirm the synergistic potential of niclosamide in vitro against a diverse range of recalcitrant Gram-negative clinical isolates and in vivo in a mouse abscess model. We also demonstrate that nitroreductases can confer resistance to niclosamide but show that evolution of these enzymes for enhanced niclosamide resistance confers a collateral sensitivity to other clinical antibiotics. Our results highlight how detailed mechanistic understanding can accelerate the evaluation and implementation of new combination therapies. One avenue to combat multidrug-resistant Gram-negative bacteria is the coadministration of multiple drugs (combination therapy), which can be particularly promising if drugs synergize. The identification of synergistic drug combinations, however, is challenging. Detailed understanding of antibiotic mechanisms can address this issue by facilitating the rational design of improved combination therapies. Here, using diverse biochemical and genetic assays, we examine the molecular mechanisms of niclosamide, a clinically approved salicylanilide compound, and demonstrate its potential for Gram-negative combination therapies. We discovered that Gram-negative bacteria possess two innate resistance mechanisms that reduce their niclosamide susceptibility: a primary mechanism mediated by multidrug efflux pumps and a secondary mechanism of nitroreduction. When efflux was compromised, niclosamide became a potent antibiotic, dissipating the proton motive force (PMF), increasing oxidative stress, and reducing ATP production to cause cell death. These insights guided the identification of diverse compounds that synergized with salicylanilides when coadministered (efflux inhibitors, membrane permeabilizers, and antibiotics that are expelled by PMF-dependent efflux), thus suggesting that salicylanilide compounds may have broad utility in combination therapies. We validate these findings in vivo using a murine abscess model, where we show that niclosamide synergizes with the membrane permeabilizing antibiotic colistin against high-density infections of multidrug-resistant Gram-negative clinical isolates. We further demonstrate that enhanced nitroreductase activity is a potential route to adaptive niclosamide resistance but show that this causes collateral susceptibility to clinical nitro-prodrug antibiotics. Thus, we highlight how mechanistic understanding of mode of action, innate/adaptive resistance, and synergy can rationally guide the discovery, development, and stewardship of novel combination therapies.
Collapse
|