1
|
Morlock GE, Zoller L. Fast unmasking toxicity of safe personal care products. J Chromatogr A 2025; 1752:465886. [PMID: 40252263 DOI: 10.1016/j.chroma.2025.465886] [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: 01/26/2025] [Revised: 03/09/2025] [Accepted: 03/19/2025] [Indexed: 04/21/2025]
Abstract
Personal care products are used several times a day and come into contact with the skin for many hours. As hazard-related data on products are rare, an imaging safety screening was developed. For the first time, it detected known and unknown hazardous compounds in 140 personal care products from 20 different product segments, including beauty and lifestyle products. The evidence-based chromatographic-hazard-related profiling detected genotoxic, cytotoxic, and estrogenic compounds, which were assigned to structural groups, and revealed that personal care products are not as safe as stated. Wound-healing and nipple creams in contact with babies as well as lipsticks used by adolescents were shown to contain highly toxic compounds able to enter the bloodstream via wounds, micro-injuries or bleeding gums and contaminate nature when washed off. Dose-response curves pointed to half-maximal genotoxic effect doses (EC50) of around 60 µg of such products which can easily be exceeded by a factor of 100,000 for only 6 g product applied or for an aggregated use of various products increasing genotoxic exposure, as observed in consumer profiles. Simulated metabolization via S9 liver enzymes did not lead to detoxification. The new safety screening not only unmasked quickly the toxicity of personal care products but also provided an understanding of how to produce hazard-free products. It empowers industries to produce future products in compliance with their standards and claims in terms of consumer health, ethics, environmental compatibility, and sustainability, and authorities to control the safety of products on the market and tackle the current underregulation.
Collapse
Affiliation(s)
- Gertrud E Morlock
- Institute of Nutritional Science, Chair of Food Science, and Interdisciplinary Research Center, Justus Liebig University Giessen, Heinrich-Buff-Ring 26-32, 35392 Giessen, Germany; Center for Sustainable Food Systems, Justus Liebig University Giessen, Senckenbergstr. 3, 35390 Giessen, Germany.
| | - Luisa Zoller
- Institute of Nutritional Science, Chair of Food Science, and Interdisciplinary Research Center, Justus Liebig University Giessen, Heinrich-Buff-Ring 26-32, 35392 Giessen, Germany
| |
Collapse
|
2
|
Deng B, Su P, Cheng L, Zhang J, Zhang X, Yu T, Bao G, Yan T, Yin Y, Shen L, Wang D, Hong L, Miao X, Yang W, Wang C, Xie J, Wang R. Iterative Optimization Yields Stapled Peptides with Superior Pharmacokinetics and Potency for Renal Fibrosis Treatment. J Med Chem 2025; 68:8516-8529. [PMID: 40199779 DOI: 10.1021/acs.jmedchem.5c00133] [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: 04/10/2025]
Abstract
Renal fibrosis, resulting from myofibroblast-mediated excessive extracellular matrix (ECM) deposition, lacks effective treatments. Novel peptide DR3penA developed by our group showed therapeutic potential for fibrotic diseases; however, its application was hindered by poor stability and bioavailability. To address this unmet need, we implemented stepwise optimization of DR3penA. The conformationally restricted analogs designed via structural predictions enhanced both activity and stability. Through structure-activity relationship analysis and cleavage site mapping, introducing unnatural amino acids improved stability. Fatty acid modifications conferred fibroblast-selective cytotoxicity and improved pharmacokinetics. After several rounds of progressive modification, peptide 27 exhibited remarkable stability, with a 5.68-fold extended half-life compared to DR3penA. Following profibrotic stimuli, peptide 27 effectively inhibited myofibroblast activation, epithelial-mesenchymal transition, and ECM synthesis. It also attenuated renal fibrosis in a unilateral ureteral obstruction model. Our study leverages multiple modifications that integrate cell and animal models to identify peptide 27 as a promising candidate for renal fibrosis therapy.
Collapse
Affiliation(s)
- Bochuan Deng
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou 730000, China
| | - Ping Su
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou 730000, China
| | - Lu Cheng
- School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen University, Shenzhen 518060, China
| | - Jiao Zhang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou 730000, China
| | - Xiang Zhang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou 730000, China
| | - Tingli Yu
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou 730000, China
| | - Guangjun Bao
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou 730000, China
| | - Tiantian Yan
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou 730000, China
| | - Yue Yin
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou 730000, China
| | - Lei Shen
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou 730000, China
| | - Dan Wang
- Institute of Basic Medicine, North Sichuan Medical College, Nanchong 637000, China
| | - Liang Hong
- Guangdong Provincial Key Laboratory of Chiral Molecular and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Xiaokang Miao
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou 730000, China
| | - Wenle Yang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou 730000, China
| | - Chenyu Wang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou 730000, China
| | - Junqiu Xie
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou 730000, China
| | - Rui Wang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou 730000, China
- Institute of Materia Medica and Research Unit of Peptide Science, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
3
|
Driedger D, Wilson DM, Britton R. A sequential esterification-ring closing metathesis-Nozaki-Hiyama-Kishi strategy for constructing a natural product-like library of tetrahydrofuran-containing macrolides. Chem Sci 2025; 16:5918-5930. [PMID: 40060091 PMCID: PMC11884414 DOI: 10.1039/d5sc00591d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 02/26/2025] [Indexed: 04/04/2025] Open
Abstract
Polyketide-like macrolides (pMLs) represent a privileged class of compounds with a high incidence of bioactivity, however their structural complexity challenges their synthesis and more general study. Here we report the synthesis of a library of tetrahydrofuran-containing pMLs underpinned by a robust and convergent build/couple/pair/couple synthetic approach. The library comprises 170 pMLs originating from 17 building blocks, 10 of which were synthesized using a proline-catalyzed α-chlorination aldol reaction. Northern and southern hemisphere building blocks were coupled using either an oxidation/Horner-Wadsworth-Emmons sequence or a saponification/Steglich esterification strategy. Coupled fragments were cyclized via ring closing metathesis to yield macrocycles 14-16 atoms in size, which we diversified using 3 eastern side chain vinyl iodides through a Nozaki-Hiyama-Kishi reaction.
Collapse
Affiliation(s)
- Daniel Driedger
- Department of Chemistry, Simon Fraser University Burnaby British Columbia V5A 1S6 Canada
| | - Darryl M Wilson
- Department of Chemistry, Simon Fraser University Burnaby British Columbia V5A 1S6 Canada
| | - Robert Britton
- Department of Chemistry, Simon Fraser University Burnaby British Columbia V5A 1S6 Canada
| |
Collapse
|
4
|
Du T, Wu J, Wang W, Liang W, Li Y, Guo C, Li X, Huang L, Yu H. Design, synthesis and structure-activity relationship of novel 1,2,4-triazolopyrimidin-5-one derivatives targeting GABA A1 and Na v1.2 with antiepileptic activity. Eur J Med Chem 2025; 286:117316. [PMID: 39874632 DOI: 10.1016/j.ejmech.2025.117316] [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: 10/14/2024] [Revised: 01/15/2025] [Accepted: 01/21/2025] [Indexed: 01/30/2025]
Abstract
A novel class of 7-phenyl-[1,2,4]triazol-5(4H)-one derivatives was designed and synthesized, and their in vivo anticonvulsant activities were evaluated using subcutaneous pentylenetetrazole (Sc-PTZ) and maximal electroshock (MES) tests. Compounds 3u, 4f and 4k exhibited significant anticonvulsant activities in the Sc-PTZ model with ED50 values of 23.7, 17.1 and 18.3 mg/kg, respectively. Neurotoxicity was accessed using the rotarod assay and none of the compounds demonstrated neurotoxicity at maximum solubility, with all TD50 values exceeding 267 mg/kg. This resulted in protective indexes (PI = TD50/ED50) values of greater than 11.3, 15.6 and 14.6, respectively. Compared to control drugs such as sodium phenytoin, sodium valproate, and carbamazepine, compounds 3u, 4f and 4k displayed superior anticonvulsant activities and reduced neurotoxicity. Mechanism results indicated that compounds 4k and 4f were sensitive to the subunit configuration of synaptic α1β2γ2 GABAA receptors, while compounds 3u and 4f dose-dependently reduced the peak amplitude of Nav1.2 currents. These structural compounds may provide a foundation for the further design of novel antiepileptic molecules with low neurotoxicity.
Collapse
Affiliation(s)
- Tongtong Du
- State Key Laboratory Base for Eco-Chemical Engineering, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao, 266042, China
| | - Jun Wu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xiannongtan Street, Xicheng district, Beijing, 100050, China
| | - Weina Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xiannongtan Street, Xicheng district, Beijing, 100050, China
| | - Wenhui Liang
- State Key Laboratory Base for Eco-Chemical Engineering, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao, 266042, China
| | - Yunjie Li
- State Key Laboratory Base for Eco-Chemical Engineering, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao, 266042, China
| | - Chuanlong Guo
- State Key Laboratory Base for Eco-Chemical Engineering, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao, 266042, China
| | - Xiufen Li
- State Key Laboratory Base for Eco-Chemical Engineering, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao, 266042, China
| | - Longjiang Huang
- State Key Laboratory Base for Eco-Chemical Engineering, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao, 266042, China; State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xiannongtan Street, Xicheng district, Beijing, 100050, China.
| | - Haibo Yu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xiannongtan Street, Xicheng district, Beijing, 100050, China.
| |
Collapse
|
5
|
Lv X, Shen J, Du X, Yue B, Zhang Q, Chang W, Miao Y, Ji Z, Chen L, Gong Y, Yang Y, Chen Q. The Optimized Lipid-Modified Prodrug for CNV Treatment. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2419263. [PMID: 39895155 DOI: 10.1002/adma.202419263] [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: 12/09/2024] [Revised: 01/25/2025] [Indexed: 02/04/2025]
Abstract
Choroidal neovascularization (CNV) is a prevalent cause of vision impairment. The primary treatment for CNV involves intravitreal injections of anti-vascular endothelial growth factor antibodies. Nevertheless, this approach still faces numerous limitations like poor patient compliance, high therapy expenditure and lack of response in some individuals. Herein, a series of anti-neovascularization prodrugs, SU5402 (SU), modified with lipids of varying chain lengths (C12, C16, C20, C24, C28) is synthesized (SU-C12, SU-C16, SU-C20, SU-C24, SU-C28). 1% polyvinyl alcohol (PVA) is used as a stabilizer to create nanoformulations of five prodrugs named SU-C12 NPs, SU-C16 NPs, SU-C20 NPs, SU-C24 NPs, SU-C28 NPs. Among these, SU-C20 NPs significantly prolong the retention of bioactive drug in the eye for up to 70 d. Moreover, SU-C20 NPs demonstrate superior tissue permeability via enhanced cellular endocytosis and exocytosis. With its prolonged retention and improved penetration, SU-C20 NPs reduce the fluorescence intensity of fundus leakage by 42.5% and the fluorescence area by 51.5% in CNV mice after four weeks, effectively inhibiting the progression of CNV. Altogether, small molecule drug SU is innovatively modified to improve its effectiveness for treating fundus neovascular diseases, proposing an alternative therapy for wet age-related macular degeneration (wAMD).
Collapse
Affiliation(s)
- Xinying Lv
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China
| | - Jingjing Shen
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China
| | - Xinwei Du
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Boyu Yue
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Qiang Zhang
- School of Materials Science and Engineering, Tongji University, Shanghai, 201804, China
| | - Wanwan Chang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China
| | - Yu Miao
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China
| | - Zhaoxin Ji
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China
| | - Linfu Chen
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China
| | - Yimou Gong
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Yang Yang
- School of Materials Science and Engineering, Tongji University, Shanghai, 201804, China
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
- Central Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
| | - Qian Chen
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China
| |
Collapse
|
6
|
Zheng M, Cao Y, Zhou Q, Si J, Huang G, Ji Y, Wu Y, Ge Z. Multifunctional Zwitterionic N-Oxide Polymers to Overcome Cascade Physiological Barriers for Efficient Anticancer Drug Delivery. Adv Healthc Mater 2025; 14:e2403852. [PMID: 39910882 DOI: 10.1002/adhm.202403852] [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: 10/06/2024] [Revised: 01/01/2025] [Indexed: 02/07/2025]
Abstract
For efficient anticancer drug delivery, cascade physiological barriers must be overcome, which requires the drug delivery vehicles to possess different or even opposite properties at different stages. Poly(tertiary amine-oxide) (PTAO) polymers with the zwitterionic feature have distinct antifouling properties in blood circulation, which can be reduced and protonated in hypoxic tumors to promote cellular internalization. Nevertheless, the effects of various PTAO structures have not been studied systemically and optimized. In this report, the side groups of PTAO are proposed to be optimized by modulating the structures. Poly(2-(N-oxide-hexamethyleneimino)ethyl methacrylate) (POC7A) with a cyclic seven-membered ring is screened as the optimized PTAO structure for in vivo applications. Moreover, the block copolymer POC7A-block-poly(ε-caprolactone) (POC7A-PCL) is prepared for the formation of micelles in aqueous solution for delivery of doxorubicin (DOX). The zwitterionic nature of POC7A shells with efficient bioreductive activity and protonation in the tumor microenvironment endows the micelles with excellent antifouling properties for long blood circulation, efficient tumor accumulation, deep penetration, and effective cellular internalization. Thus, the DOX-loaded micelles exhibit potent antitumor efficacy after intravenous administration. Zwitterionic POC7A can be used as antifouling shells of the anticancer drug delivery nanocarriers to overcome the cascade physiological barriers efficiently.
Collapse
Affiliation(s)
- Moujiang Zheng
- School of Chemistry, Xi'an Key Laboratory of Sustainable Polymer Materials, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, China
| | - Yufei Cao
- School of Chemistry, Xi'an Key Laboratory of Sustainable Polymer Materials, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, China
| | - Qinghao Zhou
- School of Chemistry, Xi'an Key Laboratory of Sustainable Polymer Materials, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, China
| | - Jiale Si
- School of Chemistry, Xi'an Key Laboratory of Sustainable Polymer Materials, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, China
| | - Guopu Huang
- School of Chemistry, Xi'an Key Laboratory of Sustainable Polymer Materials, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, China
| | - Yuanyuan Ji
- Department of Geriatric General Surgery, Scientific Research Center and Precision Medical Institute, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China
| | - Youshen Wu
- MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter School of Physics, Xian Jiaotong University, Xi'an, Shaanxi, 710049, China
| | - Zhishen Ge
- School of Chemistry, Xi'an Key Laboratory of Sustainable Polymer Materials, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, China
| |
Collapse
|
7
|
Wang DD, He L, Qi MH, Zhao HY, Yu AX, Huang SW. Mitochondria-targeting artesunate-rhein conjugates: Linker-modulated cell-permeability, heme-affinity and anticancer activity. Eur J Med Chem 2025; 282:117100. [PMID: 39615162 DOI: 10.1016/j.ejmech.2024.117100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Revised: 11/21/2024] [Accepted: 11/22/2024] [Indexed: 12/10/2024]
Abstract
Heme, abundant in the mitochondria of cancer cells, is a key target for the anticancer activity of artemisinin (ART). Current strategies to enhance the anticancer activity of ART focus solely on its delivery to heme-enriched subcellular localizations while overlooking the decisive effects of ART-heme interactions. Here, we propose an ingenious strategy that synergizes mitochondria-targeted drug delivery and linker-mediated drug conformation modulation, thereby significantly enhancing the anticancer activity of ART. By strategically conjugating artemisinin (ART) with the mitochondria-targeting rhein (R) using different linkers, we aimed to precisely adjust the conformation of the conjugates. Comprehensive computational analysis revealed that the conjugate with the optimal linker length (C4) displayed a favorable conformation that facilitated cell permeability and exhibited the highest binding affinity to heme and Fe ions. Moreover, it exhibited superior tumor suppression capabilities both in vitro and in vivo, overcoming the uncertainty of in vivo application caused by the rapid clearance of the conventional mitochondria-targeted cation TPP+, and even inducing immunogenic cell death associated with immunotherapy. This novel strategy opens up a new avenue for the development of drug conjugate systems.
Collapse
Affiliation(s)
- Dan-Dan Wang
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan, 430072, China
| | - Li He
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Ming-Hui Qi
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan, 430072, China
| | - Hong-Yang Zhao
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan, 430072, China
| | - Ai-Xi Yu
- Department of Orthopedic Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| | - Shi-Wen Huang
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan, 430072, China; Department of Orthopedic Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
8
|
Orsi M, Reymond J. Navigating a 1E+60 Chemical Space of Peptide/Peptoid Oligomers. Mol Inform 2025; 44:e202400186. [PMID: 39390672 PMCID: PMC11733718 DOI: 10.1002/minf.202400186] [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: 05/31/2024] [Revised: 08/21/2024] [Accepted: 08/27/2024] [Indexed: 10/12/2024]
Abstract
Herein we report a virtual library of 1E+60 members, a common estimate for the total size of the drug-like chemical space. The library is obtained from 100 commercially available peptide and peptoid building blocks assembled into linear or cyclic oligomers of up to 30 units, forming molecules within the size range of peptide drugs and potentially accessible by solid-phase synthesis. We demonstrate ligand-based virtual screening (LBVS) using the peptide design genetic algorithm (PDGA), which evolves a population of 50 members to resemble a given target molecule using molecular fingerprint similarity as fitness function. Target molecules are reached in less than 10,000 generations. Like in many journeys, the value of the chemical space journey using PDGA lies not in reaching the target but in the journey itself, here by encountering non-obvious analogs. We also show that PDGA can be used to generate median molecules and analogs of non-peptide target molecules.
Collapse
Affiliation(s)
- Markus Orsi
- Department of ChemistryBiochemistry and Pharmaceutical SciencesUniversity of BernFreiestrasse 33012BernSwitzerland
| | - Jean‐Louis Reymond
- Department of ChemistryBiochemistry and Pharmaceutical SciencesUniversity of BernFreiestrasse 33012BernSwitzerland
| |
Collapse
|
9
|
Tahara K, Nakamura A, Wang X, Mitamura K, Ichihashi Y, Kano K, Mishiro-Sato E, Aoki K, Urano Y, Komatsu T, Tsukiji S. γ-Secretase Cleaves Bifunctional Fatty Acid-Conjugated Small Molecules with Amide Bonds in Mammalian Cells. ACS Chem Biol 2024; 19:2438-2450. [PMID: 39567846 DOI: 10.1021/acschembio.4c00432] [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: 11/22/2024]
Abstract
Connecting two small molecules, such as ligands, fluorophores, or lipids, together via a linker with amide bonds is a widely used strategy to generate synthetic bifunctional molecules for various biological and biomedical applications. Such bifunctional molecules have been used in live-cell experiments under the assumption that they should be stable in cells. However, we recently found that a membrane-targeting bifunctional molecule, composed of a lipopeptide and the small-molecule ligand trimethoprim, referred to as mgcTMP, underwent amide-bond cleavage in mammalian cells. In this work, we first identified γ-secretase as the major protease degrading mgcTMP in cells. We next investigated the intracellular degradation of several different types of amide-linked bifunctional compounds and found that N-terminally fatty acid-conjugated small molecules are susceptible to γ-secretase-mediated amide-bond cleavage. In contrast, amide-linked bifunctional molecules composed of two small molecules, such as ligands and hydrophobic groups, which lack lipid modification, did not undergo intracellular degradation. These findings highlight a previously overlooked consideration for the development and application of lipid-based bifunctional molecules in chemical biology research.
Collapse
Affiliation(s)
- Kai Tahara
- Department of Nanopharmaceutical Sciences, Nagoya Institute of Technology, Gokiso-cho, Showa-ku, Nagoya 466-8555, Japan
| | - Akinobu Nakamura
- Quantitative Biology Research Group, Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji-cho, Okazaki, Aichi 444-8787, Japan
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba 305-8566, Japan
| | - Xiaotong Wang
- Department of Life Science and Applied Chemistry, Nagoya Institute of Technology, Gokiso-cho, Showa-ku, Nagoya 466-8555, Japan
| | - Keishi Mitamura
- Department of Life Science and Applied Chemistry, Nagoya Institute of Technology, Gokiso-cho, Showa-ku, Nagoya 466-8555, Japan
| | - Yuki Ichihashi
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Keiko Kano
- Institute of Transformative Bio-Molecules (WPI-ITbM), Nagoya University, Nagoya 464-8602, Japan
| | - Emi Mishiro-Sato
- Institute of Transformative Bio-Molecules (WPI-ITbM), Nagoya University, Nagoya 464-8602, Japan
| | - Kazuhiro Aoki
- Quantitative Biology Research Group, Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji-cho, Okazaki, Aichi 444-8787, Japan
- Division of Quantitative Biology, National Institute for Basic Biology, National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji-cho, Okazaki, Aichi 444-8787, Japan
- Laboratory of Cell Cycle Regulation, Graduate School of Biostudies, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
- Center for Living Systems Information Science (CeLiSIS), Graduate School of Biostudies, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Yasuteru Urano
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
- Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Toru Komatsu
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Shinya Tsukiji
- Department of Nanopharmaceutical Sciences, Nagoya Institute of Technology, Gokiso-cho, Showa-ku, Nagoya 466-8555, Japan
- Department of Life Science and Applied Chemistry, Nagoya Institute of Technology, Gokiso-cho, Showa-ku, Nagoya 466-8555, Japan
| |
Collapse
|
10
|
Yao C, Peng A, Wu P, Zuo J, Pan J, Kong C, Qian Z, Jin Z, Feng H. Side-chain-engineered fluorescent dyes for 3D and long-term dynamic tracking of the plasma membrane in living cells. Talanta 2024; 279:126583. [PMID: 39053364 DOI: 10.1016/j.talanta.2024.126583] [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: 05/22/2024] [Revised: 07/12/2024] [Accepted: 07/16/2024] [Indexed: 07/27/2024]
Abstract
The plasma membrane involves in many important biological events such as cell fusion and programmed cell death, but most of current plasma membrane probes cannot meet the requirement of long-term specific anchoring to the plasma membrane. Herein, we propose a molecular side-chain engineering strategy to modulate the long-term imaging performance of fluorescent dyes to the plasma membrane by regulating the cell permeability and anchoring ability. A series of FMR dyes with different lengths of side chains were designed and synthesized, and their transmembrane behaviours and staining performance were evaluated in living HeLa cells. We found that short-chain and medium-chain FMR dyes have excellent cell permeability without the labeling ability to the plasma membrane while the long-chain FMR dyes specifically stain the plasma membrane and can be firmly anchored to the plasma membrane for a long period of time. These long-chain FMR dyes have high stain specificality to the plasma membrane, and C10-FMR can be anchored to the plasma membrane of living cells for 2 h, which enables it to continuously monitor dynamic changes of the plasma membrane. The three-dimensional precision imaging of various cells was achieved using C10-FMR, which provides an opportunity to obtain complete information on the three-dimensional spatial morphology of the plasma membrane. The PEG-induced cell fusion of chicken red blood cells and H2O2-induced apoptosis of HeLa cells were monitored by real-time tracking of dynamic changes of the plasma membrane during these processes, which provide solid examples to prove the usefulness of these fluorescent dyes as long-term imaging tools. This work validates the hypothesis that cell permeability of membrane dyes can be readily regulated by tuning the side chains, and provides the effective design strategy of fluorescent dyes for 3D and long-term dynamic tracking of the plasma membrane of diverse animal cells.
Collapse
Affiliation(s)
- Chuangye Yao
- Key Laboratory of the Ministry for Advanced Catalysis Materials, College of Chemistry and Materials Science, Zhejiang Normal University, Jinhua, 321004, PR China
| | - Aohui Peng
- College of Life Sciences, Zhejiang Normal University, Jinhua, 321004, PR China
| | - Penglei Wu
- Key Laboratory of the Ministry for Advanced Catalysis Materials, College of Chemistry and Materials Science, Zhejiang Normal University, Jinhua, 321004, PR China
| | - Jiaqi Zuo
- Key Laboratory of the Ministry for Advanced Catalysis Materials, College of Chemistry and Materials Science, Zhejiang Normal University, Jinhua, 321004, PR China
| | - Junjun Pan
- Key Laboratory of the Ministry for Advanced Catalysis Materials, College of Chemistry and Materials Science, Zhejiang Normal University, Jinhua, 321004, PR China
| | - Chuixi Kong
- College of Life Sciences, Zhejiang Normal University, Jinhua, 321004, PR China
| | - Zhaosheng Qian
- Key Laboratory of the Ministry for Advanced Catalysis Materials, College of Chemistry and Materials Science, Zhejiang Normal University, Jinhua, 321004, PR China
| | - Zhigang Jin
- College of Life Sciences, Zhejiang Normal University, Jinhua, 321004, PR China.
| | - Hui Feng
- Key Laboratory of the Ministry for Advanced Catalysis Materials, College of Chemistry and Materials Science, Zhejiang Normal University, Jinhua, 321004, PR China.
| |
Collapse
|
11
|
Blake AD, Chao J, SantaMaria AM, Ekaputri S, Green KJ, Brown ST, Rakowski CK, Choi EK, Aring L, Chen PJ, Snead NM, Matje DM, Geng T, Octaviani A, Bailey K, Hollenbach SJ, Fan TM, Seo YA, Burke MD. Minimizing higher-order aggregation maximizes iron mobilization by small molecules. Nat Chem Biol 2024; 20:1282-1293. [PMID: 38664586 PMCID: PMC11831690 DOI: 10.1038/s41589-024-01596-3] [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] [Received: 06/16/2022] [Accepted: 03/01/2024] [Indexed: 09/28/2024]
Abstract
The natural product hinokitiol mobilizes iron across lipid bilayers at low concentrations and restores hemoglobinization in iron transporter protein-deficient systems. But hinokitiol fails to similarly mobilize iron at higher concentrations, limiting its uses in chemical biology and medicine. Here we show that at higher concentrations, hinokitiol3:Fe(III) complexes form large, higher-order aggregates, leading to loss of transmembrane iron mobilization. Guided by this understanding and systematic structure-function studies enabled by modular synthesis, we identified FeM-1269, which minimally aggregates and dose-dependently mobilizes iron across lipid bilayers even at very high concentrations. In contrast to hinokitiol, FeM-1269 is also well-tolerated in animals at high doses for extended periods of time. In a mouse model of anemia of inflammation, FeM-1269 increases serum iron, transferrin saturation, hemoglobin and hematocrit. This rationally developed iron-mobilizing small molecule has enhanced potential as a molecular prosthetic for understanding and potentially treating iron transporter deficiencies.
Collapse
Affiliation(s)
- Andrew D Blake
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | | | - Anna M SantaMaria
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Stella Ekaputri
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Kelsie J Green
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Samantha T Brown
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | | | - Eun-Kyung Choi
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Luisa Aring
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Peng-Jui Chen
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | | | | | - Tao Geng
- Ambys Medicines, South San Francisco, CA, USA
| | | | - Keith Bailey
- Alnylam Pharmaceuticals, Inc., Cambridge, MA, USA
| | | | - Timothy M Fan
- Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Young-Ah Seo
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA.
| | - Martin D Burke
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
- Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
- Molecule Maker Lab Institute, Arnold and Mabel Beckman Institute, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
- Carle Illinois College of Medicine, Champaign, IL, USA.
| |
Collapse
|
12
|
Ji Y, Li J, Chen C, Piao C, Zhou X, Yoon J. Wash-Free Bacterial Gram-Typing and Photodynamic Inactivation with Long-Chain-Tailed BODIPY Derivatives. Biomater Res 2024; 28:0069. [PMID: 39228999 PMCID: PMC11370751 DOI: 10.34133/bmr.0069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 07/29/2024] [Indexed: 09/05/2024] Open
Abstract
The rapid identification of bacterial Gram types and their viability, as well as efficient bacterial elimination are crucial for managing bacterial infections yet present important challenges. In this research, we utilized long-chain-tailed BODIPY derivatives to address these hurdles. Our data indicated that these derivatives can distinguish bacteria types and their viability in aqueous solutions through a concise turn-on fluorescent response. Among them, B-8 stained both live and dead bacteria, and B-14 offered a wash-free staining. B-18 demonstrated the highest affinity to selectively fluorescent label viable gram-positive bacteria with a 53.2-fold fluorescent enhancement. Confocal imaging confirmed that B-18 can serve as an effective membrane-specific probe for facilitating the typing between gram-negative and gram-positive bacteria in a wash-free manner. Additionally, B-18 displayed selective photodynamic inactivation at 1 μM toward gram-positive bacteria. In vivo studies variformed the ideal photodynamic therapeutic efficacy of B-18 against methicillin-resistant Staphylococcus aureus in mice wound infections.
Collapse
Affiliation(s)
- Yuefeng Ji
- College of Agriculture,
Yanbian University, Yanji, 133002, China
| | - Jigai Li
- College of Chemistry and Chemical Engineering,
Qingdao University, Qingdao, 266071, China
| | - Chunping Chen
- College of Agriculture,
Yanbian University, Yanji, 133002, China
| | - Chunxiang Piao
- College of Agriculture,
Yanbian University, Yanji, 133002, China
| | - Xin Zhou
- College of Chemistry and Chemical Engineering,
Qingdao University, Qingdao, 266071, China
| | - Juyoung Yoon
- Department of Chemistry and Nano Science,
Ewha Womans University, Seoul, 120-750, Korea
| |
Collapse
|
13
|
Jogadi W, Kshetri MB, Alqarni S, Sharma A, Cheline M, Amin MA, Sheets C, Nsoure-Engohang A, Zheng YR. Engineering Novel Amphiphilic Platinum(IV) Complexes to Co-Deliver Cisplatin and Doxorubicin. Molecules 2024; 29:4095. [PMID: 39274943 PMCID: PMC11397443 DOI: 10.3390/molecules29174095] [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: 07/17/2024] [Revised: 08/24/2024] [Accepted: 08/25/2024] [Indexed: 09/16/2024] Open
Abstract
In this study, we report a novel platinum-doxorubicin conjugate that demonstrates superior therapeutic indices to cisplatin, doxorubicin, or their combination, which are commonly used in cancer treatment. This new molecular structure (1) was formed by conjugating an amphiphilic Pt(IV) prodrug of cisplatin with doxorubicin. Due to its amphiphilic nature, the Pt(IV)-doxorubicin conjugate effectively penetrates cell membranes, delivering both cisplatin and doxorubicin payloads intracellularly. The intracellular accumulation of these payloads was assessed using graphite furnace atomic absorption spectrometry and fluorescence imaging. Since the therapeutic effects of cisplatin and doxorubicin stem from their ability to target nuclear DNA, we hypothesized that the amphiphilic Pt(IV)-doxorubicin conjugate (1) would effectively induce nuclear DNA damage toward killing cancer cells. To test this hypothesis, we used flow the cytometric analysis of phosphorylated H2AX (γH2AX), a biomarker of nuclear DNA damage. The Pt(IV)-doxorubicin conjugate (1) markedly induced γH2AX in treated MDA-MB-231 breast cancer cells, showing higher levels than cells treated with either cisplatin or doxorubicin alone. Furthermore, MTT cell viability assays revealed that the enhanced DNA-damaging capability of complex 1 resulted in superior cytotoxicity and selectivity against human cancer cells compared to cisplatin, doxorubicin, or their combination. Overall, the development of this amphiphilic Pt(IV)-doxorubicin conjugate represents a new form of combination therapy with improved therapeutic efficacy.
Collapse
Affiliation(s)
- Wjdan Jogadi
- Department of Chemistry and Biochemistry, Kent State University, 236 Integrated Sciences Building, Kent, OH 44242, USA
| | - Man B Kshetri
- Department of Chemistry and Biochemistry, Kent State University, 236 Integrated Sciences Building, Kent, OH 44242, USA
| | - Suha Alqarni
- Department of Chemistry and Biochemistry, Kent State University, 236 Integrated Sciences Building, Kent, OH 44242, USA
- Department of Chemistry, University of Bisha, Bisha 67714, Saudi Arabia
| | - Arpit Sharma
- Department of Chemistry and Biochemistry, Kent State University, 236 Integrated Sciences Building, Kent, OH 44242, USA
| | - May Cheline
- Department of Chemistry and Biochemistry, Kent State University, 236 Integrated Sciences Building, Kent, OH 44242, USA
| | - Md Al Amin
- Department of Chemistry and Biochemistry, Kent State University, 236 Integrated Sciences Building, Kent, OH 44242, USA
| | - Cynthia Sheets
- Department of Chemistry and Biochemistry, Kent State University, 236 Integrated Sciences Building, Kent, OH 44242, USA
| | - Angele Nsoure-Engohang
- Department of Chemistry and Biochemistry, Kent State University, 236 Integrated Sciences Building, Kent, OH 44242, USA
| | - Yao-Rong Zheng
- Department of Chemistry and Biochemistry, Kent State University, 236 Integrated Sciences Building, Kent, OH 44242, USA
| |
Collapse
|
14
|
Rahn HP, Liu X, Chosy MB, Sun J, Cegelski L, Wender PA. Biguanide-Vancomycin Conjugates are Effective Broad-Spectrum Antibiotics against Actively Growing and Biofilm-Associated Gram-Positive and Gram-Negative ESKAPE Pathogens and Mycobacteria. J Am Chem Soc 2024; 146:22541-22552. [PMID: 39088791 PMCID: PMC11624893 DOI: 10.1021/jacs.4c06520] [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: 08/03/2024]
Abstract
Strategies to increase the efficacy and/or expand the spectrum of activity of existing antibiotics provide a potentially fast path to clinically address the growing crisis of antibiotic-resistant infections. Here, we report the synthesis, antibacterial efficacy, and mechanistic activity of an unprecedented class of biguanide-antibiotic conjugates. Our lead biguanide-vancomycin conjugate, V-C6-Bg-PhCl (5e), induces highly effective cell killing with up to a 2 orders-of-magnitude improvement over its parent compound, vancomycin (V), against vancomycin-resistant enterococcus. V-C6-Bg-PhCl (5e) also exhibits improved activity against mycobacteria and each of the ESKAPE pathogens, including the Gram-negative organisms. Furthermore, we uncover broad-spectrum killing activity against biofilm-associated Gram-positive and Gram-negative bacteria as well as mycobacteria not observed for clinically used antibiotics such as oritavancin. Mode-of-action studies reveal that vancomycin-like cell wall synthesis inhibition with improved efficacy attributed to enhanced engagement at vancomycin binding sites through biguanide association with relevant cell-surface anions for Gram-positive and Gram-negative bacteria. Due to its potency, remarkably broad activity, and lack of acute mammalian cell toxicity, V-C6-Bg-PhCl (5e) is a promising candidate for treating antibiotic-resistant infections and notoriously difficult-to-treat slowly growing and antibiotic-tolerant bacteria associated with chronic and often incurable infections. More generally, this study offers a new strategy (biguanidinylation) to enhance antibiotic activity and facilitate clinical entry.
Collapse
Affiliation(s)
- Harrison P. Rahn
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | - Xinyu Liu
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | - Madeline B. Chosy
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | - Jiuzhi Sun
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | - Lynette Cegelski
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | - Paul A. Wender
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
15
|
Yao C, Zuo J, Wu P, Liu J, Pan J, Zhu E, Feng H, Zhang K, Qian Z. Molecular engineering of fluorescent dyes for long-term specific visualization of the plasma membrane based on alkyl-chain-regulated cell permeability. Talanta 2024; 275:126105. [PMID: 38640520 DOI: 10.1016/j.talanta.2024.126105] [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: 02/02/2024] [Revised: 04/09/2024] [Accepted: 04/11/2024] [Indexed: 04/21/2024]
Abstract
Long-term visualization of changes in plasma membrane dynamics during important physiological processes can provide intuitive and reliable information in a 4D mode. However, molecular tools that can visualize plasma membranes over extended periods are lacking due to the absence of effective design rules that can specifically track plasma membrane fluorescent dye molecules over time. Using plant plasma membranes as a model, we systematically investigated the effects of different alkyl chain lengths of FMR dye molecules on their performance in imaging plasma membranes. Our findings indicate that alkyl chain length can effectively regulate the permeability of dye molecules across plasma membranes. The study confirms that introducing medium-length alkyl chains improves the ability of dye molecules to target and anchor to plasma membranes, allowing for long-term imaging of plasma membranes. This provides useful design rules for creating dye molecules that enable long-term visualization of plasma membranes. Using the amphiphilic amino-styryl-pyridine fluorescent skeleton, we discovered that the inclusion of short alkyl chains facilitated rapid crossing of the plasma membrane by the dye molecules, resulting in staining of the cell nucleus and indicating improved cell permeability. Conversely, the inclusion of long alkyl chains hindered the crossing of the cell wall by the dye molecules, preventing staining of the cell membrane and demonstrating membrane impermeability to plant cells. The FMR dyes with medium-length alkyl chains rapidly crossed the cell wall, uniformly stained the cell membrane, and anchored to it for a long period without being transmembrane. This allowed for visualization and tracking of the morphological dynamics of the cell plasma membrane during water loss in a 4D mode. This suggests that the introduction of medium-length alkyl chains into amphiphilic fluorescent dyes can transform them from membrane-permeable fluorescent dyes to membrane-staining fluorescent dyes suitable for long-term imaging of the plasma membrane. In addition, we have successfully converted a membrane-impermeable fluorescent dye molecule into a membrane-staining fluorescent dye by introducing medium-length alkyl chains into the molecule. This molecular engineering of dye molecules with alkyl chains to regulate cell permeability provides a simple and effective design rule for long-term visualization of the plasma membrane, and a convenient and feasible means of chemical modification for efficient transmembrane transport of small molecule drugs.
Collapse
Affiliation(s)
- Chuangye Yao
- Laboratory of the Ministry of Education for Advanced Catalysis Materials, College of Chemistry and Materials Science, Zhejiang Normal University, Jinhua, 321004, People's Republic of China
| | - Jiaqi Zuo
- Laboratory of the Ministry of Education for Advanced Catalysis Materials, College of Chemistry and Materials Science, Zhejiang Normal University, Jinhua, 321004, People's Republic of China
| | - Penglei Wu
- Laboratory of the Ministry of Education for Advanced Catalysis Materials, College of Chemistry and Materials Science, Zhejiang Normal University, Jinhua, 321004, People's Republic of China
| | - Jie Liu
- College of Life Sciences, Zhejiang Normal University, Jinhua, 321004, People's Republic of China
| | - Junjun Pan
- Laboratory of the Ministry of Education for Advanced Catalysis Materials, College of Chemistry and Materials Science, Zhejiang Normal University, Jinhua, 321004, People's Republic of China
| | - Engao Zhu
- College of Life Sciences, Zhejiang Normal University, Jinhua, 321004, People's Republic of China
| | - Hui Feng
- Laboratory of the Ministry of Education for Advanced Catalysis Materials, College of Chemistry and Materials Science, Zhejiang Normal University, Jinhua, 321004, People's Republic of China
| | - Kewei Zhang
- College of Life Sciences, Zhejiang Normal University, Jinhua, 321004, People's Republic of China.
| | - Zhaosheng Qian
- Laboratory of the Ministry of Education for Advanced Catalysis Materials, College of Chemistry and Materials Science, Zhejiang Normal University, Jinhua, 321004, People's Republic of China.
| |
Collapse
|
16
|
Rahn HP, Sun J, Li Z, Waymouth RM, Levy R, Wender PA. Isoprenoid CARTs: In Vitro and In Vivo mRNA Delivery by Charge-Altering Releasable Transporters Functionalized with Archaea-inspired Branched Lipids. Biomacromolecules 2024; 25:4305-4316. [PMID: 38814265 DOI: 10.1021/acs.biomac.4c00373] [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: 05/31/2024]
Abstract
The delivery of oligonucleotides across biological barriers is a challenge of unsurpassed significance at the interface of materials science and medicine, with emerging clinical utility in prophylactic and therapeutic vaccinations, immunotherapies, genome editing, and cell rejuvenation. Here, we address the role of readily available branched lipids in the design, synthesis, and evaluation of isoprenoid charge-altering releasable transporters (CARTs), a pH-responsive oligomeric nanoparticle delivery system for RNA. Systematic variation of the lipid block reveals an emergent relationship between the lipid block and the neutralization kinetics of the polycationic block. Unexpectedly, iA21A11, a CART with the smallest lipid side chain, isoamyl-, was identified as the lead isoprenoid CART for the in vitro transfection of immortalized lymphoblastic cell lines. When administered intramuscularly in a murine model, iA21A11-mRNA complexes induce higher protein expression levels than our previous lead CART, ONA. Isoprenoid CARTs represent a new delivery platform for RNA vaccines and other polyanion-based therapeutics.
Collapse
Affiliation(s)
- Harrison P Rahn
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
| | - Jiuzhi Sun
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
| | - Zhijian Li
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
| | - Robert M Waymouth
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
| | - Ronald Levy
- Stanford Cancer Institute, Division of Oncology, Department of Medicine, Stanford University, Stanford, California 94305, United States
| | - Paul A Wender
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
- Department of Chemical and Systems Biology, Stanford University, Stanford, California 94305, United States
| |
Collapse
|
17
|
Su Y, Sun D, Cao C, Wang Y. Lanosterol regulates abnormal amyloid accumulation in LECs through the mediation of cholesterol pathway metabolism. Biochem Biophys Rep 2024; 38:101679. [PMID: 38501050 PMCID: PMC10945048 DOI: 10.1016/j.bbrep.2024.101679] [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: 11/26/2023] [Revised: 02/27/2024] [Accepted: 02/28/2024] [Indexed: 03/20/2024] Open
Abstract
Age-related cataract (ARC) is the predominant cause of global blindness, linked to the progressive aging of the lens, oxidative stress, perturbed calcium homeostasis, hydration irregularities, and modifications in crystallin proteins. Currently, surgical intervention remains the sole efficacious remedy, albeit carrying inherent risks of complications that may culminate in irreversible blindness. It is urgent to explore alternative, cost-effective, and uncomplicated treatment modalities for cataracts. Lanosterol has been widely reported to reverse cataracts, but the mechanism of action is not yet clear. In this study, we elucidated the mechanism through which lanosterol operates in the context of cataract reversal. Through the targeted suppression of sterol regulatory element-binding protein 2 (SREBP2) followed by lanosterol treatment, we observed the restoration of lipid metabolism disorders induced by SREBP2 knockdown in lens epithelial cells (LECs). Notably, lanosterol exhibited the ability to effectively counteract amyloid accumulation and cellular apoptosis triggered by lipid metabolism disorders. In summary, our findings suggest that lanosterol, a pivotal intermediate in lipid metabolism, may exert its therapeutic effects on cataracts by influencing lipid metabolism. This study shed light on the treatment and pharmaceutical development targeting Age-related Cataracts (ARC).
Collapse
Affiliation(s)
- Yingxue Su
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Engineering Research Center for Ophthalmic Drug Creation and Evaluation, Guangzhou, 510060, China
| | - Danyuan Sun
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Chen Cao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Engineering Research Center for Ophthalmic Drug Creation and Evaluation, Guangzhou, 510060, China
| | - Yandong Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Engineering Research Center for Ophthalmic Drug Creation and Evaluation, Guangzhou, 510060, China
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| |
Collapse
|
18
|
Guo Y, Shen Z, Zhao W, Lu J, Song Y, Shen L, Lu Y, Wu M, Shi Q, Zhuang W, Qiu Y, Sheng J, Zhou Z, Fang L, Che J, Dong X. Rational Identification of Novel Antibody-Drug Conjugate with High Bystander Killing Effect against Heterogeneous Tumors. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306309. [PMID: 38269648 PMCID: PMC10987111 DOI: 10.1002/advs.202306309] [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: 09/02/2023] [Revised: 12/15/2023] [Indexed: 01/26/2024]
Abstract
Bystander-killing payloads can significantly overcome the tumor heterogeneity issue and enhance the clinical potential of antibody-drug conjugates (ADC), but the rational design and identification of effective bystander warheads constrain the broader implementation of this strategy. Here, graph attention networks (GAT) are constructed for a rational bystander killing scoring model and ADC construction workflow for the first time. To generate efficient bystander-killing payloads, this model is utilized for score-directed exatecan derivatives design. Among them, Ed9, the most potent payload with satisfactory permeability and bioactivity, is further used to construct ADC. Through linker optimization and conjugation, novel ADCs are constructed that perform excellent anti-tumor efficacy and bystander-killing effect in vivo and in vitro. The optimal conjugate T-VEd9 exhibited therapeutic efficacy superior to DS-8201 against heterogeneous tumors. These results demonstrate that the effective scoring approach can pave the way for the discovery of novel ADC with promising bystander payloads to combat tumor heterogeneity.
Collapse
Affiliation(s)
- Yu Guo
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical SciencesZhejiang UniversityHangzhou310058P. R. China
| | - Zheyuan Shen
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical SciencesZhejiang UniversityHangzhou310058P. R. China
| | - Wenbin Zhao
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang UniversityHangzhou310018P. R. China
| | - Jialiang Lu
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical SciencesZhejiang UniversityHangzhou310058P. R. China
| | - Yi Song
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical SciencesZhejiang UniversityHangzhou310058P. R. China
| | - Liteng Shen
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical SciencesZhejiang UniversityHangzhou310058P. R. China
| | - Yang Lu
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical SciencesZhejiang UniversityHangzhou310058P. R. China
| | - Mingfei Wu
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical SciencesZhejiang UniversityHangzhou310058P. R. China
| | - Qiuqiu Shi
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical SciencesZhejiang UniversityHangzhou310058P. R. China
| | - Weihao Zhuang
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical SciencesZhejiang UniversityHangzhou310058P. R. China
| | - Yueping Qiu
- The Department of PharmacyZhejiang Cancer HospitalHangzhou310022P. R. China
| | - Jianpeng Sheng
- Department of Hepatobiliary and Pancreatic Surgerythe First Affiliated Hospital, Zhejiang University School of MedicineHangzhou310002P. R. China
| | - Zhan Zhou
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang UniversityHangzhou310018P. R. China
| | - Luo Fang
- The Department of PharmacyZhejiang Cancer HospitalHangzhou310022P. R. China
| | - Jinxin Che
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical SciencesZhejiang UniversityHangzhou310058P. R. China
| | - Xiaowu Dong
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical SciencesZhejiang UniversityHangzhou310058P. R. China
- Cancer CenterZhejiang UniversityHangzhou310058P. R. China
- Department of PharmacySecond Affiliated HospitalZhejiang University School of MedicineHangzhou310009P. R. China
| |
Collapse
|
19
|
Wilson DM, Driedger DJ, Liu DY, Keerthisinghe S, Hermann A, Bieniossek C, Linington RG, Britton RA. Targeted sampling of natural product space to identify bioactive natural product-like polyketide macrolides. Nat Commun 2024; 15:2534. [PMID: 38514617 PMCID: PMC10958047 DOI: 10.1038/s41467-024-46721-x] [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: 07/07/2023] [Accepted: 03/01/2024] [Indexed: 03/23/2024] Open
Abstract
Polyketide or polyketide-like macrolides (pMLs) continue to serve as a source of inspiration for drug discovery. However, their inherent structural and stereochemical complexity challenges efforts to explore related regions of chemical space more broadly. Here, we report a strategy termed the Targeted Sampling of Natural Product space (TSNaP) that is designed to identify and assess regions of chemical space bounded by this important class of molecules. Using TSNaP, a family of tetrahydrofuran-containing pMLs are computationally assembled from pML inspired building blocks to provide a large collection of natural product-like virtual pMLs. By scoring functional group and volumetric overlap against their natural counterparts, a collection of compounds are prioritized for targeted synthesis. Using a modular and stereoselective synthetic approach, a library of polyketide-like macrolides are prepared to sample these unpopulated regions of pML chemical space. Validation of this TSNaP approach by screening this library against a panel of whole-cell biological assays, reveals hit rates exceeding those typically encountered in small molecule libraries. This study suggests that the TSNaP approach may be more broadly useful for the design of improved chemical libraries for drug discovery.
Collapse
Affiliation(s)
- Darryl M Wilson
- Department of Chemistry, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada
| | - Daniel J Driedger
- Department of Chemistry, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada
| | - Dennis Y Liu
- Department of Chemistry, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada
| | - Sandra Keerthisinghe
- Center for High-Throughput Chemical Biology, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada
| | - Adrian Hermann
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070, Basel, Switzerland
| | - Christoph Bieniossek
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070, Basel, Switzerland
| | - Roger G Linington
- Department of Chemistry, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada.
- Center for High-Throughput Chemical Biology, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada.
| | - Robert A Britton
- Department of Chemistry, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada.
| |
Collapse
|
20
|
Kirsanov R, Khailova LS, Rokitskaya TI, Lyamzaev KG, Panteleeva AA, Nazarov PA, Firsov AM, Iaubasarova IR, Korshunova GA, Kotova EA, Antonenko YN. Synthesis of Triphenylphosphonium-Linked Derivative of 3,5-Di tert-butyl-4-hydroxybenzylidene-malononitrile (SF6847) via Knoevenagel Reaction Yields an Effective Mitochondria-Targeted Protonophoric Uncoupler. ACS OMEGA 2024; 9:11551-11561. [PMID: 38496966 PMCID: PMC10938414 DOI: 10.1021/acsomega.3c08621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 02/07/2024] [Accepted: 02/14/2024] [Indexed: 03/19/2024]
Abstract
Mitochondrial uncouplers are actively sought as potential therapeutics. Here, we report the first successful synthesis of mitochondria-targeted derivatives of the highly potent uncoupler 3,5-ditert-butyl-4-hydroxybenzylidene-malononitrile (SF6847), bearing a cationic alkyl(triphenyl)phosphonium (TPP) group. As a key step of the synthesis, we used condensation of a ketophenol with malononitrile via the Knoevenagel reaction. SF-C5-TPP with a pentamethylene linker between SF6847 and TPP, stimulating respiration and collapsing membrane potential of rat liver mitochondria at submicromolar concentrations, proved to be the most effective uncoupler of the series. SF-C5-TPP showed pronounced protonophoric activity on a model planar bilayer lipid membrane. Importantly, SF-C5-TPP exhibited rather low toxicity in fibroblast cell culture, causing mitochondrial depolarization in cells at concentrations that only slightly affected cell viability. SF-C5-TPP was more effective in decreasing the mitochondrial membrane potential in the cell culture than SF6847, in contrast to the case of isolated mitochondria. Like other zwitterionic uncouplers, SF-C5-TPP inhibited the growth of Bacillus subtilis in the micromolar concentration range.
Collapse
Affiliation(s)
- Roman
S. Kirsanov
- Belozersky
Institute of Physico-Chemical Biology, Lomonosov
Moscow State University, 119991 Moscow, Russia
| | - Ljudmila S. Khailova
- Belozersky
Institute of Physico-Chemical Biology, Lomonosov
Moscow State University, 119991 Moscow, Russia
| | - Tatyana I. Rokitskaya
- Belozersky
Institute of Physico-Chemical Biology, Lomonosov
Moscow State University, 119991 Moscow, Russia
| | - Konstantin G. Lyamzaev
- Belozersky
Institute of Physico-Chemical Biology, Lomonosov
Moscow State University, 119991 Moscow, Russia
- The
“Russian Clinical Research Center for Gerontology” of
the Ministry of Healthcare of the Russian Federation, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Alisa A. Panteleeva
- Belozersky
Institute of Physico-Chemical Biology, Lomonosov
Moscow State University, 119991 Moscow, Russia
| | - Pavel A. Nazarov
- Belozersky
Institute of Physico-Chemical Biology, Lomonosov
Moscow State University, 119991 Moscow, Russia
| | - Alexander M. Firsov
- Belozersky
Institute of Physico-Chemical Biology, Lomonosov
Moscow State University, 119991 Moscow, Russia
| | - Iliuza R. Iaubasarova
- Belozersky
Institute of Physico-Chemical Biology, Lomonosov
Moscow State University, 119991 Moscow, Russia
| | - Galina A. Korshunova
- Belozersky
Institute of Physico-Chemical Biology, Lomonosov
Moscow State University, 119991 Moscow, Russia
| | - Elena A. Kotova
- Belozersky
Institute of Physico-Chemical Biology, Lomonosov
Moscow State University, 119991 Moscow, Russia
| | - Yuri N. Antonenko
- Belozersky
Institute of Physico-Chemical Biology, Lomonosov
Moscow State University, 119991 Moscow, Russia
| |
Collapse
|
21
|
Chen K, Liu S, Wei Y. Sub-nanosized vanadate hybrid clusters maintain glucose homeostasis and restore treatment response in inflammatory disease in obese mice. NANO RESEARCH 2024; 17:1818-1826. [DOI: 10.1007/s12274-023-6366-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 11/21/2023] [Accepted: 11/23/2023] [Indexed: 09/11/2024]
|
22
|
Ji W, Zhang P, Zhou Y, Zhou X, Ma X, Tan T, Cao H. Hydrogel-encapsulated medium chain lipid-modified zeolite imidazole framework-90 as a promising platform for oral delivery of proteins. J Control Release 2024; 367:93-106. [PMID: 38237690 DOI: 10.1016/j.jconrel.2024.01.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 01/06/2024] [Accepted: 01/09/2024] [Indexed: 01/28/2024]
Abstract
The administration of protein therapeutics through oral means is seen as a convenient and painless experience for patients, making it a significant consideration in the field of drug delivery. Nevertheless, the challenging conditions within the gastrointestinal tract, along with the obstacles to absorption, impede the efficient transportation of proteins. Here, we successfully implemented post-synthetic modifications to attach medium-chain lipids (C10) onto the surface of zeolitic imidazole framework-90 (ZIF-90), then encapsulated the nanoparticles with sodium alginate, resulting in a potential platform for the oral administration of proteins. By means of biomimetic mineralization, ZIF-90 achieves a simple and efficient encapsulation of proteins of varying sizes, while shielding them against degradation by digestive enzymes. Sodium alginate hydrogel protects proteins against gastric acid and helps the cargo to rapidly penetrate the mucus layer. Through a mixed mechanism dominated by micropinocytosis, the C10-conjugated ZIF-90 (ZIF-90-C10) can be uptake by Caco-2 cells with a 200-400% increase and transported through the Golgi apparatus after escaping from lysosomes, exhibiting enhanced uptake in the overall gastrointestinal tract. Furthermore, ZIF-90-C10 retains its adenosine triphosphate-responsive release, which drastically lowers the likelihood of accumulation in vivo and allows targeted delivery for disease cells. Our work highlights mid-chain lipid conjugation as a potent approach to enhancing nanoparticle delivery efficiency and a potential strategy for oral delivery of biomacromolecules when combined with pH-responsive gels.
Collapse
Affiliation(s)
- Wei Ji
- National Energy R&D Center for Biorefinery, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, PR China; Beijing Key Lab of Bioprocess, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, PR China
| | - Peng Zhang
- National Energy R&D Center for Biorefinery, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, PR China
| | - Yegui Zhou
- Beijing Key Lab of Bioprocess, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, PR China
| | - Xiqin Zhou
- National Energy R&D Center for Biorefinery, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, PR China; Beijing Key Lab of Bioprocess, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, PR China
| | - Xiufan Ma
- National Energy R&D Center for Biorefinery, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, PR China; Beijing Key Lab of Bioprocess, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, PR China
| | - Tianwei Tan
- National Energy R&D Center for Biorefinery, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, PR China; Beijing Key Lab of Bioprocess, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, PR China.
| | - Hui Cao
- National Energy R&D Center for Biorefinery, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, PR China; Beijing Key Lab of Bioprocess, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, PR China.
| |
Collapse
|
23
|
Ng MY, Song ZJ, Venkatesan G, Rodriguez-Cuenca S, West JA, Yang S, Tan CH, Ho PCL, Griffin JL, Vidal-Puig A, Bassetto M, Hagen T. Conjugating uncoupler compounds with hydrophobic hydrocarbon chains to achieve adipose tissue selective drug accumulation. Sci Rep 2024; 14:4932. [PMID: 38418847 PMCID: PMC10901892 DOI: 10.1038/s41598-024-54466-2] [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: 12/05/2023] [Accepted: 02/13/2024] [Indexed: 03/02/2024] Open
Abstract
One potential approach for treating obesity is to increase energy expenditure in brown and white adipose tissue. Here we aimed to achieve this outcome by targeting mitochondrial uncoupler compounds selectively to adipose tissue, thus avoiding side effects from uncoupling in other tissues. Selective drug accumulation in adipose tissue has been observed with many lipophilic compounds and dyes. Hence, we explored the feasibility of conjugating uncoupler compounds with a lipophilic C8-hydrocarbon chain via an ether bond. We found that substituting the trifluoromethoxy group in the uncoupler FCCP with a C8-hydrocarbon chain resulted in potent uncoupling activity. Nonetheless, the compound did not elicit therapeutic effects in mice, likely as a consequence of metabolic instability resulting from rapid ether bond cleavage. A lipophilic analog of the uncoupler compound 2,6-dinitrophenol, in which a C8-hydrocarbon chain was conjugated via an ether bond in the para-position (2,6-dinitro-4-(octyloxy)phenol), exhibited increased uncoupling activity compared to the parent compound. However, in vivo pharmacokinetics studies suggested that 2,6-dinitro-4-(octyloxy)phenol was also metabolically unstable. In conclusion, conjugation of a hydrophobic hydrocarbon chain to uncoupler compounds resulted in sustained or improved uncoupling activity. However, an ether bond linkage led to metabolic instability, indicating the need to conjugate lipophilic groups via other chemical bonds.
Collapse
Affiliation(s)
- Mei Ying Ng
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Zhi Jian Song
- School of Physical and Mathematical Sciences, Division of Chemistry and Biological Chemistry, Nanyang Technological University, Singapore, Singapore
| | | | - Sergio Rodriguez-Cuenca
- Wellcome-MRC Institute of Metabolic Science and Medical Research Council Metabolic Diseases Unit, The University of Cambridge, Cambridge, UK
| | - James A West
- Department of Biochemistry, The University of Cambridge, Cambridge, UK
| | - Shili Yang
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore, Singapore
| | - Choon Hong Tan
- School of Physical and Mathematical Sciences, Division of Chemistry and Biological Chemistry, Nanyang Technological University, Singapore, Singapore
| | - Paul Chi-Lui Ho
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore, Singapore
- School of Pharmacy, Monash University Malaysia, 47500, Subang Jaya, Malaysia
| | - Julian L Griffin
- The Rowett Institute of Nutrition and Health, The University of Aberdeen, Aberdeen, UK
| | - Antonio Vidal-Puig
- Wellcome-MRC Institute of Metabolic Science and Medical Research Council Metabolic Diseases Unit, The University of Cambridge, Cambridge, UK
| | - Marcella Bassetto
- School of Pharmacy and Pharmaceutical Sciences, College of Biomedical and Life Sciences, Cardiff University, Cardiff, UK.
| | - Thilo Hagen
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
24
|
Morstein J, Amatuni A, Shuster A, Kuttenlochner W, Ko T, Abegg D, Groll M, Adibekian A, Renata H, Trauner DH. Optical Control of Proteasomal Protein Degradation with a Photoswitchable Lipopeptide. Angew Chem Int Ed Engl 2024; 63:e202314791. [PMID: 38109686 PMCID: PMC12079555 DOI: 10.1002/anie.202314791] [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: 10/02/2023] [Revised: 12/02/2023] [Accepted: 12/05/2023] [Indexed: 12/20/2023]
Abstract
Photolipids have emerged as attractive tools for the optical control of lipid functions. They often contain an azobenzene photoswitch that imparts a cis double-bond upon irradiation. Herein, we present the application of photoswitching to a lipidated natural product, the potent proteasome inhibitor cepafungin I. Several azobenzene-containing lipids were attached to the cyclopeptide core, yielding photoswitchable derivatives. Most notably, PhotoCep4 exhibited a 10-fold higher cellular potency in its light-induced cis-form, matching the potency of natural cepafungin I. The length of the photolipid tail and distal positioning of the azobenzene photoswitch with respect to the macrocycle is critical for this activity. In a proteome-wide experiment, light-triggered PhotoCep4 modulation showed high overlap with constitutively active cepafungin I. The mode of action was studied using crystallography and revealed an identical binding of the cyclopeptide in comparison to cepafungin I, suggesting that differences in their cellular activity originate from switching the tail structure. The photopharmacological approach described herein could be applicable to many other natural products as lipid conjugation is common and often necessary for potent activity. Such lipids are often introduced late in synthetic routes, enabling facile chemical modifications.
Collapse
Affiliation(s)
- Johannes Morstein
- Department of Cellular and Molecular Pharmacology and Howard Hughes Medical Institute, University of California, San Francisco, California 94158, USA
- Department of Chemistry, New York University, New York, New York 10003, USA
| | - Alexander Amatuni
- Skaggs Doctoral Program in the Chemical and Biological Sciences, Scripps Research, La Jolla, California 92037, USA
| | - Anton Shuster
- Skaggs Doctoral Program in the Chemical and Biological Sciences, Scripps Research, La Jolla, California 92037, USA
| | - Wolfgang Kuttenlochner
- Technical University of Munich, TUM School of Natural Sciences, Department of Bioscience, Center for Protein Assemblies, Chair of Biochemistry, Ernst-Otto-Fischer-Str. 8, 85748 Garching, Germany
| | - Tongil Ko
- Department of Chemistry, New York University, New York, New York 10003, USA
- Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Daniel Abegg
- Department of Chemistry, University of Illinois Chicago, Chicago, Illinois 60607, USA
| | - Michael Groll
- Technical University of Munich, TUM School of Natural Sciences, Department of Bioscience, Center for Protein Assemblies, Chair of Biochemistry, Ernst-Otto-Fischer-Str. 8, 85748 Garching, Germany
| | - Alexander Adibekian
- Department of Chemistry, University of Illinois Chicago, Chicago, Illinois 60607, USA
| | - Hans Renata
- Department of Chemistry, BioScience Research Collaborative, Rice University, Houston, Texas 77005, USA
| | - Dirk H. Trauner
- Department of Chemistry, New York University, New York, New York 10003, USA
- Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
25
|
Su Y, Mangus AM, Cordell WT, Pfleger BF. Overcoming barriers to medium-chain fatty alcohol production. Curr Opin Biotechnol 2024; 85:103063. [PMID: 38219523 PMCID: PMC10922944 DOI: 10.1016/j.copbio.2023.103063] [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: 11/28/2023] [Revised: 12/19/2023] [Accepted: 12/20/2023] [Indexed: 01/16/2024]
Abstract
Medium-chain fatty alcohols (mcFaOHs) are aliphatic primary alcohols containing six to twelve carbons that are widely used in materials, pharmaceuticals, and cosmetics. Microbial biosynthesis has been touted as a route to less-abundant chain-length molecules and as a sustainable alternative to current petrochemical processes. Several metabolic engineering strategies for producing mcFaOHs have been demonstrated in the literature, yet processes continue to suffer from poor selectivity and mcFaOH toxicity, leading to reduced titers, rates, and yields of the desired compounds. This opinion examines the current state of microbial mcFaOH biosynthesis, summarizing engineering efforts to tailor selectivity and improve product tolerance by implementing engineering strategies that circumvent or overcome mcFaOH toxicity.
Collapse
Affiliation(s)
- Yun Su
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Anna M Mangus
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - William T Cordell
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Brian F Pfleger
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA.
| |
Collapse
|
26
|
McFetridge ML, Kulkarni K, Lee TH, Del Borgo MP, Aguilar MI, Ricardo SD. Elucidating the cell penetrating properties of self-assembling β-peptides. NANOSCALE 2023; 15:14971-14980. [PMID: 37661822 DOI: 10.1039/d3nr03673a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Self-assembling lipopeptide hydrogels have been widely developed for the delivery of therapeutics due to their rapid gelation, injectability, and highly controlled physicochemical properties. Lipopeptides are also known for their membrane-associating and cell penetrating properties, which may impact on their application in cell-encapsulation. Self-assembling lipidated-β3-peptide materials developed in our laboratory have previously been used in cell culture as 2D substrates, thus as a continuation of this work we aimed to encapsulate cells in 3D by forming a hydrogel. We therefore assessed the self-assembling lipidated-β3-peptides for cell-penetrating properties in mesenchymal stems cells (MSC) using fluorescence microscopy and membrane association with surface plasmon resonance spectroscopy (SPR). The results demonstrated that lipidated β3-peptides penetrate the MSC plasma membrane and localise to the mitochondrial network. While self-assembling lipopeptide hydrogels have shown tremendous potential for delivery of therapeutics, further optimisation may be required to minimise the membrane uptake of the lipidated-β3-peptides for cell encapsulation applications.
Collapse
Affiliation(s)
- Meg L McFetridge
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, 3800, Australia.
| | - Ketav Kulkarni
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, 3800, Australia.
| | - Tzong-Hsien Lee
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, 3800, Australia.
| | - Mark P Del Borgo
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, 3800, Australia.
| | - Marie-Isabel Aguilar
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, 3800, Australia.
| | - Sharon D Ricardo
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, 3800, Australia.
| |
Collapse
|
27
|
Morstein J, Shrestha R, Van QN, López CA, Arora N, Tonelli M, Liang H, Chen D, Zhou Y, Hancock JF, Stephen AG, Turbyville TJ, Shokat KM. Direct Modulators of K-Ras-Membrane Interactions. ACS Chem Biol 2023; 18:2082-2093. [PMID: 37579045 PMCID: PMC10510109 DOI: 10.1021/acschembio.3c00413] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 07/31/2023] [Indexed: 08/16/2023]
Abstract
Protein-membrane interactions (PMIs) are ubiquitous in cellular signaling. Initial steps of signal transduction cascades often rely on transient and dynamic interactions with the inner plasma membrane leaflet to populate and regulate signaling hotspots. Methods to target and modulate these interactions could yield attractive tool compounds and drug candidates. Here, we demonstrate that the conjugation of a medium-chain lipid tail to the covalent K-Ras(G12C) binder MRTX849 at a solvent-exposed site enables such direct modulation of PMIs. The conjugated lipid tail interacts with the tethered membrane and changes the relative membrane orientation and conformation of K-Ras(G12C), as shown by molecular dynamics (MD) simulation-supported NMR studies. In cells, this PMI modulation restricts the lateral mobility of K-Ras(G12C) and disrupts nanoclusters. The described strategy could be broadly applicable to selectively modulate transient PMIs.
Collapse
Affiliation(s)
- Johannes Morstein
- Department
of Cellular and Molecular Pharmacology and Howard Hughes Medical Institute, University of California, San Francisco, California 94158, United States
| | - Rebika Shrestha
- NCI
RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland 21701, United States
| | - Que N. Van
- NCI
RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland 21701, United States
| | - César A. López
- Theoretical
Biology and Biophysics Group, Los Alamos
National Laboratory, Los Alamos, New Mexico 87545, United States
| | - Neha Arora
- Department
of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center, Houston, Texas 77030, United States
| | - Marco Tonelli
- National
Magnetic Resonance Facility at Madison, Biochemistry Department, University of Wisconsin-Madison, Madison, Wisconsin 53706, United States
| | - Hong Liang
- Department
of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center, Houston, Texas 77030, United States
| | - De Chen
- NCI
RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland 21701, United States
| | - Yong Zhou
- Department
of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center, Houston, Texas 77030, United States
| | - John F. Hancock
- Department
of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center, Houston, Texas 77030, United States
| | - Andrew G. Stephen
- NCI
RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland 21701, United States
| | - Thomas J. Turbyville
- NCI
RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland 21701, United States
| | - Kevan M. Shokat
- Department
of Cellular and Molecular Pharmacology and Howard Hughes Medical Institute, University of California, San Francisco, California 94158, United States
| |
Collapse
|
28
|
Kshetri M, Jogadi W, Alqarni S, Datta P, Cheline M, Sharma A, Betters T, Broyles D, Zheng YR. Exploring the Impact of Head Group Modifications on the Anticancer Activities of Fatty-Acid-like Platinum(IV) Prodrugs: A Structure-Activity Relationship Study. Int J Mol Sci 2023; 24:13301. [PMID: 37686109 PMCID: PMC10487970 DOI: 10.3390/ijms241713301] [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: 07/30/2023] [Revised: 08/18/2023] [Accepted: 08/23/2023] [Indexed: 09/10/2023] Open
Abstract
We conducted the first comprehensive investigation on the impact of head group modifications on the anticancer activities of fatty-acid-like Pt(IV) prodrugs (FALPs), which are a class of platinum-based metallodrugs that target mitochondria. We created a small library of FALPs (1-9) with diverse head group modifications. The outcomes of our study demonstrate that hydrophilic modifications exclusively enhance the potency of these metallodrugs, whereas hydrophobic modifications significantly decrease their cytotoxicity. To further understand this interesting structure-activity relationship, we chose two representative FALPs (compounds 2 and 7) as model compounds: one (2) with a hydrophilic polyethylene glycol (PEG) head group, and the other (7) with a hydrophobic hydrocarbon modification of the same molecular weight. Using these FALPs, we conducted a targeted investigation on the mechanism of action. Our study revealed that compound 2, with hydrophilic modifications, exhibited remarkable penetration into cancer cells and mitochondria, leading to subsequent mitochondrial and DNA damage, and effectively eradicating cancer cells. In contrast, compound 7, with hydrophobic modifications, displayed a significantly lower uptake and weaker cellular responses. The collective results present a different perspective, indicating that increased hydrophobicity may not necessarily enhance cellular uptake as is conventionally believed. These findings provide valuable new insights into the fundamental principles of developing metallodrugs.
Collapse
Affiliation(s)
- Man Kshetri
- Department of Chemistry and Biochemistry, Kent State University, 236 Integrated Sciences Building, Kent, OH 44242, USA (S.A.); (P.D.); (M.C.)
| | - Wjdan Jogadi
- Department of Chemistry and Biochemistry, Kent State University, 236 Integrated Sciences Building, Kent, OH 44242, USA (S.A.); (P.D.); (M.C.)
| | - Suha Alqarni
- Department of Chemistry and Biochemistry, Kent State University, 236 Integrated Sciences Building, Kent, OH 44242, USA (S.A.); (P.D.); (M.C.)
- Department of Chemistry, University of Bisha, Bisha 67714, Saudi Arabia
| | - Payel Datta
- Department of Chemistry and Biochemistry, Kent State University, 236 Integrated Sciences Building, Kent, OH 44242, USA (S.A.); (P.D.); (M.C.)
- Department of Chemistry, Case Western Reserve University, Cleveland, OH 44106, USA
| | - May Cheline
- Department of Chemistry and Biochemistry, Kent State University, 236 Integrated Sciences Building, Kent, OH 44242, USA (S.A.); (P.D.); (M.C.)
| | - Arpit Sharma
- Department of Chemistry and Biochemistry, Kent State University, 236 Integrated Sciences Building, Kent, OH 44242, USA (S.A.); (P.D.); (M.C.)
| | - Tyler Betters
- Department of Chemistry and Biochemistry, Kent State University, 236 Integrated Sciences Building, Kent, OH 44242, USA (S.A.); (P.D.); (M.C.)
| | - Deonya Broyles
- Department of Chemistry and Biochemistry, Kent State University, 236 Integrated Sciences Building, Kent, OH 44242, USA (S.A.); (P.D.); (M.C.)
| | - Yao-Rong Zheng
- Department of Chemistry and Biochemistry, Kent State University, 236 Integrated Sciences Building, Kent, OH 44242, USA (S.A.); (P.D.); (M.C.)
| |
Collapse
|
29
|
Millette G, Lacasse E, Binette R, Belley V, Chaumont LP, Ster C, Beaudry F, Boyapelly K, Boudreault PL, Malouin F. Rationally Designed Pyrimidine Compounds: Promising Novel Antibiotics for the Treatment of Staphylococcus aureus-Associated Bovine Mastitis. Antibiotics (Basel) 2023; 12:1344. [PMID: 37627764 PMCID: PMC10451377 DOI: 10.3390/antibiotics12081344] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/18/2023] [Accepted: 08/19/2023] [Indexed: 08/27/2023] Open
Abstract
Staphylococcus aureus is one of the major pathogens causing bovine mastitis, and antibiotic treatment is most often inefficient due to its virulence and antibiotic-resistance attributes. The development of new antibiotics for veterinary use should account for the One Health concept, in which humans, animals, and environmental wellbeing are all interconnected. S. aureus can infect cattle and humans alike and antibiotic resistance can impact both if the same classes of antibiotics are used. New effective antibiotic classes against S. aureus are thus needed in dairy farms. We previously described PC1 as a novel antibiotic, which binds the S. aureus guanine riboswitch and interrupts transcription of essential GMP synthesis genes. However, chemical instability of PC1 hindered its development, evaluation, and commercialization. Novel PC1 analogs with improved stability have now been rationally designed and synthesized, and their in vitro and in vivo activities have been evaluated. One of these novel compounds, PC206, remains stable in solution and demonstrates specific narrow-spectrum activity against S. aureus. It is active against biofilm-embedded S. aureus, its cytotoxicity profile is adequate, and in vivo tests in mice and cows show that it is effective and well tolerated. PC206 and structural analogs represent a promising new antibiotic class to treat S. aureus-induced bovine mastitis.
Collapse
Affiliation(s)
- Guillaume Millette
- Department of Biology, Institute of Sciences, University of Sherbrooke, Sherbrooke, QC J1K 2R1, Canada; (G.M.); (E.L.); (C.S.)
| | - Evelyne Lacasse
- Department of Biology, Institute of Sciences, University of Sherbrooke, Sherbrooke, QC J1K 2R1, Canada; (G.M.); (E.L.); (C.S.)
| | - Renaud Binette
- Department of Chemistry, Institute of Sciences, University of Sherbrooke, Sherbrooke, QC J1K 2R1, Canada;
| | - Véronique Belley
- Department of Biology, Institute of Sciences, University of Sherbrooke, Sherbrooke, QC J1K 2R1, Canada; (G.M.); (E.L.); (C.S.)
| | - Louis-Philippe Chaumont
- Department of Biology, Institute of Sciences, University of Sherbrooke, Sherbrooke, QC J1K 2R1, Canada; (G.M.); (E.L.); (C.S.)
| | - Céline Ster
- Department of Biology, Institute of Sciences, University of Sherbrooke, Sherbrooke, QC J1K 2R1, Canada; (G.M.); (E.L.); (C.S.)
| | - Francis Beaudry
- Department of Veterinary Biomedicine, Institute of Veterinary Medicine, University of Montreal, St-Hyacinthe, QC J2S 2M2, Canada;
| | - Kumaraswamy Boyapelly
- Department of Pharmacology and Physiology, Institute of Medicine and Health Sciences, Sherbrooke University, Sherbrooke, QC J1H 5N4, Canada
| | - Pierre-Luc Boudreault
- Department of Pharmacology and Physiology, Institute of Medicine and Health Sciences, Sherbrooke University, Sherbrooke, QC J1H 5N4, Canada
| | - François Malouin
- Department of Biology, Institute of Sciences, University of Sherbrooke, Sherbrooke, QC J1K 2R1, Canada; (G.M.); (E.L.); (C.S.)
| |
Collapse
|
30
|
Trottmann F, Fiedler J, Ishida K, Ishida-Ito M, Little RF, Hertweck C. Bacterial Pathogen Channels Medium-Sized Fatty Acids into Malleicyprol Biosynthesis. ACS Chem Biol 2023; 18:1557-1563. [PMID: 37319349 DOI: 10.1021/acschembio.3c00188] [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: 06/17/2023]
Abstract
Bacterial pathogens of the Burkholderia pseudomallei (BP) group cause life-threatening infections in both humans and animals. Critical for the virulence of these often antibiotic-resistant pathogens is the polyketide hybrid metabolite malleicyprol, which features two chains, a short cyclopropanol-substituted chain and a long hydrophobic alkyl chain. The biosynthetic origin of the latter has remained unknown. Here, we report the discovery of novel overlooked malleicyprol congeners with varied chain lengths and identify medium-sized fatty acids as polyketide synthase (PKS) starter units that constitute the hydrophobic carbon tails. Mutational and biochemical analyses show that a designated coenzyme A-independent fatty acyl-adenylate ligase (FAAL, BurM) is essential for recruiting and activating fatty acids in malleicyprol biosynthesis. In vitro reconstitution of the BurM-catalyzed PKS priming reaction and analysis of ACP-bound building blocks reveal a key role of BurM in the toxin assembly. Insights into the function and role of BurM hold promise for the development of enzyme inhibitors as novel antivirulence therapeutics to combat infections with BP pathogens.
Collapse
Affiliation(s)
- Felix Trottmann
- Department of Biomolecular Chemistry, Leibniz Institute for Natural Product Research and Infection Biology (HKI), Beutenbergstraße 11a, 07745 Jena, Germany
| | - Jonas Fiedler
- Department of Biomolecular Chemistry, Leibniz Institute for Natural Product Research and Infection Biology (HKI), Beutenbergstraße 11a, 07745 Jena, Germany
| | - Keishi Ishida
- Department of Biomolecular Chemistry, Leibniz Institute for Natural Product Research and Infection Biology (HKI), Beutenbergstraße 11a, 07745 Jena, Germany
| | - Mie Ishida-Ito
- Department of Biomolecular Chemistry, Leibniz Institute for Natural Product Research and Infection Biology (HKI), Beutenbergstraße 11a, 07745 Jena, Germany
| | - Rory F Little
- Department of Biomolecular Chemistry, Leibniz Institute for Natural Product Research and Infection Biology (HKI), Beutenbergstraße 11a, 07745 Jena, Germany
| | - Christian Hertweck
- Department of Biomolecular Chemistry, Leibniz Institute for Natural Product Research and Infection Biology (HKI), Beutenbergstraße 11a, 07745 Jena, Germany
- Natural Product Chemistry, Faculty of Biological Sciences, Friedrich Schiller University Jena, 07743 Jena, Germany
| |
Collapse
|
31
|
Kirsanov RS, Khailova LS, Rokitskaya TI, Iaubasarova IR, Nazarov PA, Panteleeva AA, Lyamzaev KG, Popova LB, Korshunova GA, Kotova EA, Antonenko YN. Ester-stabilized phosphorus ylides as protonophores on bilayer lipid membranes, mitochondria and chloroplasts. Bioelectrochemistry 2023; 150:108369. [PMID: 36638678 DOI: 10.1016/j.bioelechem.2023.108369] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 12/19/2022] [Accepted: 01/03/2023] [Indexed: 01/06/2023]
Abstract
Triphenylphosphonium ylides are commonly used as key intermediates in the Wittig reaction. Based on the known acidities of stabilized ylide precursors, we proposed that a methylene group adjacent to phosphorus in these compounds can ensure proton shuttling across lipid membranes. Here, we synthesized (decyloxycarbonylmethyl)triphenylphosphonium bromide (CMTPP-C10) by reaction of triphenylphosphine with decyl bromoacetate. This phosphonium salt precursor of the ester-stabilized phosphorus ylide along with its octyl (CMTPP-C8) and dodecyl (CMTPP-C12) analogues was found to be a carrier of protons in mitochondrial, chloroplast and artificial lipid membranes, suggesting that it can reversibly release hydrogen ions and diffuse through the membranes in both zwitterionic (ylide) and cationic forms. The CMTPP-C10-mediated electrical current across planar bilayer lipid membranes exhibited pronounced proton selectivity. Similar to conventional protonophores, known to uncouple electron transport and ATP synthesis, CMTPP-Cn (n = 8, 10, 12) stimulated mitochondrial respiration, while decreasing membrane potential, at micromolar concentrations, thereby showing the classical uncoupling activity in mitochondria. CMTPP-C12 also caused dissipation of transmembrane pH gradient on chloroplast membranes. Importantly, CMTPP-C10 exhibited substantially lower toxicity in cell culture, than C12TPP. Thus, we report the finding of a new class of ylide-type protonophores, which is of substantial interest due to promising therapeutic properties of uncouplers.
Collapse
Affiliation(s)
- Roman S Kirsanov
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Ljudmila S Khailova
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Tatyana I Rokitskaya
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Iliuza R Iaubasarova
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia; Faculty of Chemistry, Lomonosov Moscow State University, Leninskie Gory 1/3, Moscow 119991, Russia
| | - Pavel A Nazarov
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Alisa A Panteleeva
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Konstantin G Lyamzaev
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia; The "Russian Clinical Research Center for Gerontology" of the Ministry of Healthcare of the Russian Federation, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Lyudmila B Popova
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Galina A Korshunova
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Elena A Kotova
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Yuri N Antonenko
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia.
| |
Collapse
|
32
|
Xie J, Long ZQ, Chen AQ, Ding YG, Liu ST, Zhou X, Liu LW, Yang S. Novel Sulfonamide Derivatives Containing a Piperidine Moiety as New Bactericide Leads for Managing Plant Bacterial Diseases. Int J Mol Sci 2023; 24:ijms24065861. [PMID: 36982936 PMCID: PMC10054644 DOI: 10.3390/ijms24065861] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 03/15/2023] [Accepted: 03/16/2023] [Indexed: 03/30/2023] Open
Abstract
Plant bacterial diseases are an intractable problem due to the fact that phytopathogens have acquired strong resistances for traditional pesticides, resulting in restricting the quality and yield of agricultural products around the world. To develop new agrochemical alternatives, we prepared a novel series of sulfanilamide derivatives containing piperidine fragments and assessed their antibacterial potency. The bioassay results revealed that most molecules displayed excellent in vitro antibacterial potency towards Xanthomonas oryzae pv. oryzae (Xoo) and Xanthomonas axonopodis pv. citri (Xac). In particular, molecule C4 exhibited outstanding inhibitory activity toward Xoo with EC50 value of 2.02 µg mL-1, which was significantly better than those of the commercial agents bismerthiazol (EC50 = 42.38 µg mL-1) and thiodiazole copper (EC50 = 64.50 µg mL-1). A series of biochemical assays confirmed that compound C4 interacted with dihydropteroate synthase, and irreversibly damaged the cell membrane. In vivo assays showed that the molecule C4 presented acceptable curative and protection activities of 34.78% and 39.83%, respectively, at 200 µg mL-1, which were greater than those of thiodiazole and bismerthiazol. This study highlights the valuable insights for the excavation and development of new bactericides that can concurrently target dihydropteroate synthase and bacterial cell membranes.
Collapse
Affiliation(s)
- Jiao Xie
- National Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang 550025, China
| | - Zhou-Qing Long
- National Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang 550025, China
| | - Ai-Qun Chen
- National Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang 550025, China
| | - Ying-Guo Ding
- National Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang 550025, China
| | - Shi-Tao Liu
- National Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang 550025, China
| | - Xiang Zhou
- National Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang 550025, China
| | - Li-Wei Liu
- National Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang 550025, China
| | - Song Yang
- National Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang 550025, China
| |
Collapse
|
33
|
Linker S, Schellhaas C, Kamenik AS, Veldhuizen MM, Waibl F, Roth HJ, Fouché M, Rodde S, Riniker S. Lessons for Oral Bioavailability: How Conformationally Flexible Cyclic Peptides Enter and Cross Lipid Membranes. J Med Chem 2023; 66:2773-2788. [PMID: 36762908 PMCID: PMC9969412 DOI: 10.1021/acs.jmedchem.2c01837] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Indexed: 02/11/2023]
Abstract
Cyclic peptides extend the druggable target space due to their size, flexibility, and hydrogen-bonding capacity. However, these properties impact also their passive membrane permeability. As the "journey" through membranes cannot be monitored experimentally, little is known about the underlying process, which hinders rational design. Here, we use molecular simulations to uncover how cyclic peptides permeate a membrane. We show that side chains can act as "molecular anchors", establishing the first contact with the membrane and enabling insertion. Once inside, the peptides are positioned between headgroups and lipid tails─a unique polar/apolar interface. Only one of two distinct orientations at this interface allows for the formation of the permeable "closed" conformation. In the closed conformation, the peptide crosses to the lower leaflet via another "anchoring" and flipping mechanism. Our findings provide atomistic insights into the permeation process of flexible cyclic peptides and reveal design considerations for each step of the process.
Collapse
Affiliation(s)
- Stephanie
M. Linker
- Department
of Chemistry and Applied Biosciences, ETH
Zürich, Vladimir-Prelog-Weg 2, 8093 Zürich, Switzerland
| | - Christian Schellhaas
- Department
of Chemistry and Applied Biosciences, ETH
Zürich, Vladimir-Prelog-Weg 2, 8093 Zürich, Switzerland
| | - Anna S. Kamenik
- Department
of Chemistry and Applied Biosciences, ETH
Zürich, Vladimir-Prelog-Weg 2, 8093 Zürich, Switzerland
| | - Mac M. Veldhuizen
- Department
of Chemistry and Applied Biosciences, ETH
Zürich, Vladimir-Prelog-Weg 2, 8093 Zürich, Switzerland
| | - Franz Waibl
- Department
of Chemistry and Applied Biosciences, ETH
Zürich, Vladimir-Prelog-Weg 2, 8093 Zürich, Switzerland
| | - Hans-Jörg Roth
- Novartis
Institutes for BioMedical Research, Novartis
Pharma AG, Novartis Campus, 4056 Basel, Switzerland
| | - Marianne Fouché
- Novartis
Institutes for BioMedical Research, Novartis
Pharma AG, Novartis Campus, 4056 Basel, Switzerland
| | - Stephane Rodde
- Novartis
Institutes for BioMedical Research, Novartis
Pharma AG, Novartis Campus, 4056 Basel, Switzerland
| | - Sereina Riniker
- Department
of Chemistry and Applied Biosciences, ETH
Zürich, Vladimir-Prelog-Weg 2, 8093 Zürich, Switzerland
| |
Collapse
|
34
|
Suzuki S, Yoshikawa M, Sawada S, Devaraj NK, Tsukiji S. Miniaturized Synthetic Palmitoylation Motifs for Small-Molecule Localization in Living Cells. Bioconjug Chem 2023; 34:169-173. [PMID: 36534355 DOI: 10.1021/acs.bioconjchem.2c00517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Conjugating small-molecule ligands to synthetic motifs that can localize to specific organelles or membranes in living cells is a practical approach to develop compounds as chimeric tools or drugs that can manipulate biological processes in a subcellular site-specific manner. However, the number of available organelle-targeted synthetic motifs for small-molecule localization is limited. We have recently developed a synthetic myristoyl-DCys motif for small-molecule localization that undergoes S-palmitoylation via the cellular palmitoylation machinery and localizes to the Golgi surface. Herein, we show that the lipid acyl chain of the myristoyl (C14)-DCys motif can be as short as 10-carbons and still retain the palmitoylation-dependent Golgi localization property in cells. This discovery led to the identification of four new derivatives for small-molecule localization: tridecanoyl (C13)-, dodecanoyl (C12)-, undecanoyl (C11)-, and decanoyl (C10)-DCys motifs. We demonstrated that even the short decanoyl-DCys palmitoylation motif could be used to generate small-molecule ligand conjugates that functioned as chemical tools for controlling protein localization and cell signaling. The miniaturized synthetic palmitoylation motifs identified in this work may find applications in creating various Golgi-localizable chimeric molecules for use in chemical biology and drug development.
Collapse
Affiliation(s)
- Sachio Suzuki
- Department of Nanopharmaceutical Sciences, Nagoya Institute of Technology, Gokiso-cho, Showa-ku, Nagoya 466-8555, Japan
| | - Masaru Yoshikawa
- Department of Nanopharmaceutical Sciences, Nagoya Institute of Technology, Gokiso-cho, Showa-ku, Nagoya 466-8555, Japan
| | - Shunsuke Sawada
- Department of Nanopharmaceutical Sciences, Nagoya Institute of Technology, Gokiso-cho, Showa-ku, Nagoya 466-8555, Japan
| | - Neal K Devaraj
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, California 92093, United States
| | - Shinya Tsukiji
- Department of Nanopharmaceutical Sciences, Nagoya Institute of Technology, Gokiso-cho, Showa-ku, Nagoya 466-8555, Japan.,Department of Life Science and Applied Chemistry, Nagoya Institute of Technology, Gokiso-cho, Showa-ku, Nagoya 466-8555, Japan
| |
Collapse
|
35
|
How to smuggle a drug into cells: add a lipid 'tail'. Nature 2022. [PMID: 36266585 DOI: 10.1038/d41586-022-03282-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|