1
|
Mete R, Das S, Saha A, Roy S, Mondal S, Bose A, Basu B, Elossaily GM, Prajapati B. Transgenesis in Drug Discovery: Enhancing Target Identification and Validation. Mol Biotechnol 2025:10.1007/s12033-025-01426-4. [PMID: 40148722 DOI: 10.1007/s12033-025-01426-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 03/05/2025] [Indexed: 03/29/2025]
Abstract
Transgenesis, the introduction of foreign genetic material into the genome of an organism, has become a crucial and transformative technique in the realm of drug discovery. This review article provides a comprehensive overview of the integral role that transgenesis plays in the drug discovery process, with a specific focus on target identification and target validation. By examining the recent advancements and innovative approaches, this article aims to shed light on the importance of transgenesis in accelerating drug development. In the context of target identification, transgenesis has allowed for the creation of relevant disease models, enabling researchers to study the genetic and molecular basis of various disorders. The use of transgenic animals, such as mice and zebrafish, has facilitated the identification of potential drug targets by mimicking specific human disease conditions. This review also discusses emerging technologies, such as CRISPR-Cas9 and other genome editing tools, which have revolutionized the field of transgenesis. These technologies have enhanced the precision and efficiency of genetic manipulations in transgenic animals, making the creation of disease-relevant models more accessible and cost-effective. Moreover, integration of omics technologies, such as genomics, transcriptomics, proteomics, and metabolomics, has provided a holistic view of the molecular changes in transgenic models, further aiding in target identification and validation. This review emphasizes the importance of transgenesis in target identification and validation and underscores its vital role in shaping the future of drug discovery.
Collapse
Affiliation(s)
- Rumela Mete
- TAAB Biostudy Services, Jadavpur, Kolkata, 700032, India
| | - Sourav Das
- Department of Pharmaceutics, School of Pharmacy, The Neotia University, Sarisa, West Bengal, 743368, India
| | - Arindam Saha
- Cognizant Technology Solutions Private Limited, Salt Lake, Kolkata, 700091, India
| | - Sukanta Roy
- Department of Pharmaceutics, School of Pharmacy, The Neotia University, Sarisa, West Bengal, 743368, India
| | - Smritilekha Mondal
- Department of Biopharmaceutics, Dr. Reddy's Laboratory, Bachupally Village, Hyderabad, Telangana, 500090, India
| | - Anirbandeep Bose
- Department of Pharmaceutical Technology, School of Health & Medical Sciences, Adamas University, Barasat, Kolkata, West Bengal, 700126, India
| | - Biswajit Basu
- Department of Pharmaceutical Technology, School of Health & Medical Sciences, Adamas University, Barasat, Kolkata, West Bengal, 700126, India
| | - Gehan M Elossaily
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, P.O. Box 71666, Riyadh, 13713, Saudi Arabia
| | - Bhupendra Prajapati
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India.
- Department of Industrial Pharmacy, Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, 73000, Thailand.
| |
Collapse
|
2
|
Huang W, Jia C, Ren C. Artificial Ion Transporters as Potent Therapeutics for Channelopathies. ChemMedChem 2025; 20:e202400811. [PMID: 39572385 DOI: 10.1002/cmdc.202400811] [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: 10/12/2024] [Revised: 11/21/2024] [Indexed: 12/10/2024]
Abstract
Ion channels are essential for the selective transport of ions, playing a fundamental role in critical physiological processes. Dysfunctions in these channels, often arising from genetic mutations or environmental factors, give rise to a class of disorders collectively known as channelopathies. In recent years, artificial ion transporters have been developed to mimic the essential function of natural channels, offering potential therapeutic approaches for these conditions. Although significant progress has been made in improving the activity and selectivity of these synthetic transporters, their application in treating diseases associated with ion transport dysregulation remains in its infancy. This concept provides an overview of recent advancements in artificial ion transporters for treating channelopathies, while highlighting the key challenges and prospects in translating these developments into practical therapies.
Collapse
Affiliation(s)
- Wei Huang
- Department of Respiratory Medicine, The People's Hospital of Gongan County, Gongan, Hubei, 434300, China
- State Key Laboratory of Cellular Stress Biology and Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| | - Chunyan Jia
- State Key Laboratory of Cellular Stress Biology and Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| | - Changliang Ren
- State Key Laboratory of Cellular Stress Biology and Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| |
Collapse
|
3
|
Konopka M, Halgreen L, Dascalu AE, Chvojka M, Valkenier H. Controlling the transmembrane transport of chloride by dynamic covalent chemistry with azines. Chem Sci 2025; 16:3509-3515. [PMID: 39877820 PMCID: PMC11770589 DOI: 10.1039/d4sc08580a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 01/19/2025] [Indexed: 01/31/2025] Open
Abstract
Stimuli-responsive transmembrane ion transport has become a prominent area of research due to its fundamental importance in cellular processes and potential therapeutic applications. Commonly used stimuli include pH, light, and reduction or oxidation agents. This paper presents the use of dynamic covalent chemistry to activate and modulate the transmembrane transport of chloride in liposomes. An active chloride transporter was obtained in situ within the lipid bilayer by dynamic azine metathesis. The transport activity was further tuned by changing the structure of the added azines, while the dynamic covalent chemistry could be activated by lowering the pH. This dynamic covalent chemistry opens a new approach towards controlling transmembrane transport.
Collapse
Affiliation(s)
- Marcin Konopka
- Engineering of Molecular NanoSystems, Université libre de Bruxelles (ULB) Avenue F.D. Roosevelt 50, CP165/64 B-1050 Brussels Belgium
| | - Lau Halgreen
- Engineering of Molecular NanoSystems, Université libre de Bruxelles (ULB) Avenue F.D. Roosevelt 50, CP165/64 B-1050 Brussels Belgium
| | - Anca-Elena Dascalu
- Engineering of Molecular NanoSystems, Université libre de Bruxelles (ULB) Avenue F.D. Roosevelt 50, CP165/64 B-1050 Brussels Belgium
| | - Matúš Chvojka
- Engineering of Molecular NanoSystems, Université libre de Bruxelles (ULB) Avenue F.D. Roosevelt 50, CP165/64 B-1050 Brussels Belgium
- Department of Chemistry and RECETOX Faculty of Science, Masaryk University Brno 62500 Czech Republic
| | - Hennie Valkenier
- Engineering of Molecular NanoSystems, Université libre de Bruxelles (ULB) Avenue F.D. Roosevelt 50, CP165/64 B-1050 Brussels Belgium
| |
Collapse
|
4
|
Ghazikhani H, Butler G. Ion channel classification through machine learning and protein language model embeddings. J Integr Bioinform 2024; 21:jib-2023-0047. [PMID: 39572876 PMCID: PMC11698620 DOI: 10.1515/jib-2023-0047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 09/04/2024] [Indexed: 01/06/2025] Open
Abstract
Ion channels are critical membrane proteins that regulate ion flux across cellular membranes, influencing numerous biological functions. The resource-intensive nature of traditional wet lab experiments for ion channel identification has led to an increasing emphasis on computational techniques. This study extends our previous work on protein language models for ion channel prediction, significantly advancing the methodology and performance. We employ a comprehensive array of machine learning algorithms, including k-Nearest Neighbors, Random Forest, Support Vector Machines, and Feed-Forward Neural Networks, alongside a novel Convolutional Neural Network (CNN) approach. These methods leverage fine-tuned embeddings from ProtBERT, ProtBERT-BFD, and MembraneBERT to differentiate ion channels from non-ion channels. Our empirical findings demonstrate that TooT-BERT-CNN-C, which combines features from ProtBERT-BFD and a CNN, substantially surpasses existing benchmarks. On our original dataset, it achieves a Matthews Correlation Coefficient (MCC) of 0.8584 and an accuracy of 98.35 %. More impressively, on a newly curated, larger dataset (DS-Cv2), it attains an MCC of 0.9492 and an ROC AUC of 0.9968 on the independent test set. These results not only highlight the power of integrating protein language models with deep learning for ion channel classification but also underscore the importance of using up-to-date, comprehensive datasets in bioinformatics tasks. Our approach represents a significant advancement in computational methods for ion channel identification, with potential implications for accelerating research in ion channel biology and aiding drug discovery efforts.
Collapse
Affiliation(s)
- Hamed Ghazikhani
- Department of Computer Science and Software Engineering, Concordia University, Montreal, Canada
| | - Gregory Butler
- Department of Computer Science and Software Engineering, Concordia University, Montreal, Canada
| |
Collapse
|
5
|
Wu M, Liao J, Meng F, Chen C. Calculating linear and nonlinear multi-ensemble slow collective variables for protein folding. J Chem Phys 2024; 161:184102. [PMID: 39513439 DOI: 10.1063/5.0232102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 10/21/2024] [Indexed: 11/15/2024] Open
Abstract
Traditional molecular dynamics simulation of biomolecules suffers from the conformational sampling problem. It is often difficult to produce enough valid data for post analysis such as free energy calculation and transition path construction. To improve the sampling, one practical solution is putting an adaptive bias potential on some predefined collective variables. The quality of collective variables strongly affects the sampling ability of a molecule in the simulation. In the past, collective variables were built with the sampling data at a constant temperature. This is insufficient because of the same sampling problem. In this work, we apply the standard weighted histogram analysis method to calculate the multi-ensemble averages of pairs of time-lagged features for the construction of both linear and nonlinear slow collective variables. Compared to previous single-ensemble methods, the presented method produces averages with much smaller statistical uncertainties. The generated collective variables help a peptide and a miniprotein fold to their near-native states in a short simulation time period. By using the method, enhanced sampling simulations could be more effective and productive.
Collapse
Affiliation(s)
- Mincong Wu
- Institute of Biophysics, School of Physics, Huazhong University of Science and Technology, Wuhan 430074, Hubei, China
| | - Jun Liao
- Institute of Biophysics, School of Physics, Huazhong University of Science and Technology, Wuhan 430074, Hubei, China
| | - Fanjun Meng
- Institute of Biophysics, School of Physics, Huazhong University of Science and Technology, Wuhan 430074, Hubei, China
| | - Changjun Chen
- Institute of Biophysics, School of Physics, Huazhong University of Science and Technology, Wuhan 430074, Hubei, China
| |
Collapse
|
6
|
Qureshi A, Shah A, Iftikhar FJ, Haleem A, Zia MA. Electrochemical analysis of anticancer and antibiotic drugs in water and biological specimens. RSC Adv 2024; 14:36633-36655. [PMID: 39559583 PMCID: PMC11570916 DOI: 10.1039/d4ra05685j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 11/08/2024] [Indexed: 11/20/2024] Open
Abstract
The increasing prevalence of pharmaceuticals in water and complex matrices necessitates accurate measurement and monitoring of their environmental contamination levels. This is crucial not only for environmental conservation but also for comprehending the intricate mechanisms involved and developing more effective treatment approaches. In this context, electrochemical techniques show significant potential for the detection of pharmaceuticals across various matrices. Specifically, voltammetry is advantageous due to its rapid, straightforward, and cost-effective nature, allowing for the simultaneous analysis of multiple anticancer and antibiotic drugs. By utilizing nanomaterial-modified electrochemical sensors, the sensitivity and selectivity of detection methods can be significantly improved. The small size and customizable properties of nanomaterials enable these sensors to identify trace amounts of drugs in diverse samples. However, challenges persist in achieving reliable and accurate electrochemical monitoring of drugs in water and biological samples. Biofluids such as saliva, urine, and blood/serum, along with environmental samples from lakes and rivers, often contain numerous interfering substances that can diminish analyte signals. This review examines electrochemical methods and their potential applications for detecting pharmaceuticals and their metabolites, while also addressing the mechanisms of action and harmful effects of these drugs on both ecosystems and human health. Recent developments in electrochemical sensors utilizing nanomaterials for the detection of health-threatening pharmaceutical contaminants are examined, providing important insights into their underlying mechanisms. The emphasis is placed on the detection of anticancer agents and antibiotics, which relies on the electrocatalytic properties of the sensor materials. Additionally, discussions on density functional theory studies are included, along with an exploration of the emerging challenges and future directions in this area, aimed at enhancing readers' comprehension of the field and underscoring the necessary actions for a sustainable future.
Collapse
Affiliation(s)
- Ayesha Qureshi
- Department of Chemistry, Quaid-i-Azam University Islamabad 45320 Pakistan
| | - Afzal Shah
- Department of Chemistry, Quaid-i-Azam University Islamabad 45320 Pakistan
| | | | - Abdul Haleem
- Department of Chemistry, Quaid-i-Azam University Islamabad 45320 Pakistan
| | - Muhammad Abid Zia
- Department of Chemistry, University of Education Attock Punjab 43600 Pakistan
| |
Collapse
|
7
|
Yang X, Li M, Jia ZC, Liu Y, Wu SF, Chen MX, Hao GF, Yang Q. Unraveling the secrets: Evolution of resistance mediated by membrane proteins. Drug Resist Updat 2024; 77:101140. [PMID: 39244906 DOI: 10.1016/j.drup.2024.101140] [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: 04/28/2024] [Revised: 08/08/2024] [Accepted: 08/14/2024] [Indexed: 09/10/2024]
Abstract
Membrane protein-mediated resistance is a multidisciplinary challenge that spans fields such as medicine, agriculture, and environmental science. Understanding its complexity and devising innovative strategies are crucial for treating diseases like cancer and managing resistant pests in agriculture. This paper explores the dual nature of resistance mechanisms across different organisms: On one hand, animals, bacteria, fungi, plants, and insects exhibit convergent evolution, leading to the development of similar resistance mechanisms. On the other hand, influenced by diverse environmental pressures and structural differences among organisms, they also demonstrate divergent resistance characteristics. Membrane protein-mediated resistance mechanisms are prevalent across animals, bacteria, fungi, plants, and insects, reflecting their shared survival strategies evolved through convergent evolution to address similar survival challenges. However, variations in ecological environments and biological characteristics result in differing responses to resistance. Therefore, examining these differences not only enhances our understanding of adaptive resistance mechanisms but also provides crucial theoretical support and insights for addressing drug resistance and advancing pharmaceutical development.
Collapse
Affiliation(s)
- Xue Yang
- State 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.
| | - Min Li
- State Key Laboratory of Crop Biology, College of Life Science, Shandong Agricultural University, Tai'an 271018, China.
| | - Zi-Chang Jia
- State 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.
| | - Yan Liu
- State Key Laboratory of Crop Biology, College of Life Science, Shandong Agricultural University, Tai'an 271018, China.
| | - Shun-Fan Wu
- College of Plant Protection, Nanjing Agricultural University, State & Local Joint Engineering Research Center of Green Pesticide Invention and Application, Weigang Road 1, Nanjing, Jiangsu 210095, China.
| | - Mo-Xian Chen
- State 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.
| | - Ge-Fei Hao
- State 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.
| | - Qing Yang
- Institute of Plant Protection, Chinese Academy of Agricultural Science, No. 2 West Yuanmingyuan Road, Haidian District, Beijing 100193, China.
| |
Collapse
|
8
|
Berscheid A, Straetener J, Schilling NA, Ruppelt D, Konnerth MC, Schittek B, Krismer B, Peschel A, Steinem C, Grond S, Brötz-Oesterhelt H. The microbiome-derived antibacterial lugdunin acts as a cation ionophore in synergy with host peptides. mBio 2024; 15:e0057824. [PMID: 39133006 PMCID: PMC11389392 DOI: 10.1128/mbio.00578-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 07/02/2024] [Indexed: 08/13/2024] Open
Abstract
Lugdunin is a microbiome-derived antibacterial agent with good activity against Gram-positive pathogens in vitro and in animal models of nose colonization and skin infection. We have previously shown that lugdunin depletes bacterial energy resources by dissipating the membrane potential of Staphylococcus aureus. Here, we explored the mechanism of action of lugdunin in more detail and show that lugdunin quickly depolarizes cytoplasmic membranes of different bacterial species and acidifies the cytoplasm of S. aureus within minutes due to protonophore activity. Varying the salt species and concentrations in buffers revealed that not only protons are transported, and we demonstrate the binding of the monovalent cations K+, Na+, and Li+ to lugdunin. By comparing known ionophores with various ion transport mechanisms, we conclude that the ion selectivity of lugdunin largely resembles that of 15-mer linear peptide gramicidin A. Direct interference with the main bacterial metabolic pathways including DNA, RNA, protein, and cell wall biosyntheses can be excluded. The previously observed synergism of lugdunin with dermcidin-derived peptides such as DCD-1 in killing S. aureus is mechanistically based on potentiated membrane depolarization. We also found that lugdunin was active against certain eukaryotic cells, however strongly depending on the cell line and growth conditions. While adherent lung epithelial cell lines were almost unaffected, more sensitive cells showed dissipation of the mitochondrial membrane potential. Lugdunin seems specifically adapted to its natural environment in the respiratory tract. The ionophore mechanism is refractory to resistance development and benefits from synergy with host-derived antimicrobial peptides. IMPORTANCE The vast majority of antimicrobial peptides produced by members of the microbiome target the bacterial cell envelope by many different mechanisms. These compounds and their producers have evolved side-by-side with their host and were constantly challenged by the host's immune system. These molecules are optimized to be well tolerated at their physiological site of production, and their modes of action have proven efficient in vivo. Imbalancing the cellular ion homeostasis is a prominent mechanism among antibacterial natural products. For instance, over 120 naturally occurring polyether ionophores are known to date, and antimicrobial peptides with ionophore activity have also been detected in microbiomes. In this study, we elucidated the mechanism underlying the membrane potential-dissipating activity of the thiazolidine-containing cycloheptapeptide lugdunin, the first member of the fibupeptides discovered in a commensal bacterium from the human nose, which is a promising future probiotic candidate that is not prone to resistance development.
Collapse
Affiliation(s)
- Anne Berscheid
- Interfaculty Institute of Microbiology and Infection Medicine, Microbial Bioactive Compounds, University of Tübingen, Tübingen, Germany
- Microbial Bioactive Compounds, University of Tübingen, German Centre for Infection Research (DZIF), Partner Site Tübingen, Tübingen, Germany
| | - Jan Straetener
- Interfaculty Institute of Microbiology and Infection Medicine, Microbial Bioactive Compounds, University of Tübingen, Tübingen, Germany
| | - Nadine A Schilling
- Institute of Organic Chemistry, University of Tübingen, Tübingen, Germany
| | - Dominik Ruppelt
- Georg-August-Universität Göttingen, Institute of Organic and Biomolecular Chemistry, Göttingen, Germany
| | - Martin C Konnerth
- Institute of Organic Chemistry, University of Tübingen, Tübingen, Germany
| | - Birgit Schittek
- Department of Dermatology, Division of Dermatooncology, University of Tübingen, Tübingen, Germany
| | - Bernhard Krismer
- Microbial Bioactive Compounds, University of Tübingen, German Centre for Infection Research (DZIF), Partner Site Tübingen, Tübingen, Germany
- Interfaculty Institute of Microbiology and Infection Medicine, Infection Biology, University of Tübingen, Tübingen, Germany
- Microbial Bioactive Compounds, University of Tübingen, Cluster of Excellence EXC 2124-Controlling Microbes to Fight Infections, Tubingen, Germany
| | - Andreas Peschel
- Microbial Bioactive Compounds, University of Tübingen, German Centre for Infection Research (DZIF), Partner Site Tübingen, Tübingen, Germany
- Interfaculty Institute of Microbiology and Infection Medicine, Infection Biology, University of Tübingen, Tübingen, Germany
- Microbial Bioactive Compounds, University of Tübingen, Cluster of Excellence EXC 2124-Controlling Microbes to Fight Infections, Tubingen, Germany
| | - Claudia Steinem
- Georg-August-Universität Göttingen, Institute of Organic and Biomolecular Chemistry, Göttingen, Germany
- Max-Planck-Institute for Dynamics and Self Organization, Göttingen, Germany
| | - Stephanie Grond
- Institute of Organic Chemistry, University of Tübingen, Tübingen, Germany
- Microbial Bioactive Compounds, University of Tübingen, Cluster of Excellence EXC 2124-Controlling Microbes to Fight Infections, Tubingen, Germany
| | - Heike Brötz-Oesterhelt
- Interfaculty Institute of Microbiology and Infection Medicine, Microbial Bioactive Compounds, University of Tübingen, Tübingen, Germany
- Microbial Bioactive Compounds, University of Tübingen, German Centre for Infection Research (DZIF), Partner Site Tübingen, Tübingen, Germany
- Microbial Bioactive Compounds, University of Tübingen, Cluster of Excellence EXC 2124-Controlling Microbes to Fight Infections, Tubingen, Germany
| |
Collapse
|
9
|
Johnson TG, Sadeghi-Kelishadi A, Langton MJ. Length dependent reversible off-on activation of photo-switchable relay anion transporters. Chem Commun (Camb) 2024; 60:7160-7163. [PMID: 38910566 DOI: 10.1039/d4cc02603a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/25/2024]
Abstract
A homologous series of azobenzene-derived photo-switchable ion relay transporters is reported. We reveal that both the length and geometry of the relay strongly affect transport rate, allowing the relative activity of the E and Z isomers to be reversed and hence the wavelengths of light used for on and off switching to be exchanged.
Collapse
Affiliation(s)
- Toby G Johnson
- Chemistry Research Laboratory, Mansfield Road, Oxford, OX1 3TA, UK.
| | | | | |
Collapse
|
10
|
Paul R, Dutta D, Mukhopadhyay TK, Müller D, Lala B, Datta A, Schwalbe H, Dash J. A non-B DNA binding peptidomimetic channel alters cellular functions. Nat Commun 2024; 15:5275. [PMID: 38902227 PMCID: PMC11190219 DOI: 10.1038/s41467-024-49534-0] [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: 08/08/2022] [Accepted: 06/07/2024] [Indexed: 06/22/2024] Open
Abstract
DNA binding transcription factors possess the ability to interact with lipid membranes to construct ion-permeable pathways. Herein, we present a thiazole-based DNA binding peptide mimic TBP2, which forms transmembrane ion channels, impacting cellular ion concentration and consequently stabilizing G-quadruplex DNA structures. TBP2 self-assembles into nanostructures, e.g., vesicles and nanofibers and facilitates the transportation of Na+ and K+ across lipid membranes with high conductance (~0.6 nS). Moreover, TBP2 exhibits increased fluorescence when incorporated into the membrane or in cellular nuclei. Monomeric TBP2 can enter the lipid membrane and localize to the nuclei of cancer cells. The coordinated process of time-dependent membrane or nuclear localization of TBP2, combined with elevated intracellular cation levels and direct G-quadruplex (G4) interaction, synergistically promotes formation and stability of G4 structures, triggering cancer cell death. This study introduces a platform to mimic and control intricate biological functions, leading to the discovery of innovative therapeutic approaches.
Collapse
Affiliation(s)
- Raj Paul
- School of Chemical Sciences, Indian Association for the Cultivation of Science, Kolkata, 700032, India
| | - Debasish Dutta
- School of Chemical Sciences, Indian Association for the Cultivation of Science, Kolkata, 700032, India
| | - Titas Kumar Mukhopadhyay
- School of Chemical Sciences, Indian Association for the Cultivation of Science, Kolkata, 700032, India
| | - Diana Müller
- Institute of Organic Chemistry and Chemical Biology, Center for Biomolecular Magnetic Resonance (BMRZ), Goethe, University Frankfurt, Max-von-Laue Strasse 7, 60438, Frankfurt am Main, Germany
| | - Binayak Lala
- School of Chemical Sciences, Indian Association for the Cultivation of Science, Kolkata, 700032, India
| | - Ayan Datta
- School of Chemical Sciences, Indian Association for the Cultivation of Science, Kolkata, 700032, India
| | - Harald Schwalbe
- Institute of Organic Chemistry and Chemical Biology, Center for Biomolecular Magnetic Resonance (BMRZ), Goethe, University Frankfurt, Max-von-Laue Strasse 7, 60438, Frankfurt am Main, Germany
| | - Jyotirmayee Dash
- School of Chemical Sciences, Indian Association for the Cultivation of Science, Kolkata, 700032, India.
| |
Collapse
|
11
|
Mukherjee S, Shinde SV, Talukdar P, Haldar J. Unveiling the potent activity of a synthetic ion transporter against multidrug-resistant Gram-positive bacteria and biofilms. RSC Med Chem 2024; 15:2127-2137. [PMID: 38911153 PMCID: PMC11187549 DOI: 10.1039/d4md00002a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 04/25/2024] [Indexed: 06/25/2024] Open
Abstract
The increasing prevalence of drug-resistant infections caused by Gram-positive bacteria poses a significant threat to public healthcare. These pathogens exhibit not only smart resistance mechanisms but also form impenetrable biofilms on various surfaces, rendering them resilient to conventional therapies. In this study, we present the potent antibacterial activity of a synthetic ion transporter T against multi-drug resistant (MDR) Gram-positive pathogens, with minimum inhibitory concentration (MIC) values ranging from 0.5 to 2 μg mL-1. The compound demonstrates high selectivity with negligible toxicity towards mammalian cells (HC50 = 810 μg mL-1). It exhibits fast killing kinetics, completely eliminating >5 log bacterial cells within 12 h. Moreover, the compound displays efficacy against both planktonic bacteria and preformed biofilms of methicillin-resistant S. aureus (MRSA), reducing the bacterial burden within the biofilm by 2 log. Mechanistic investigations reveal that the ion transporter depolarizes the bacterial membrane potential and enhances membrane permeability. Additionally, it generates reactive oxygen species, contributing to its bactericidal activity. Notably, MRSA did not exhibit detectable resistance to the ion transporter even after serial passaging for 10 days. Collectively, this novel class of ion transporter holds promise as a therapeutic candidate for combating infections caused by multi-drug resistant Gram-positive bacteria.
Collapse
Affiliation(s)
- Sudip Mukherjee
- Antimicrobial Research Laboratory, New Chemistry Unit, JNCASR Jakkur Bangalore-560064 India
| | - Sopan Valiba Shinde
- Department of Chemistry, Indian Institute of Science Education and Research Pune Dr. Homi Bhabha Road, Pashan Pune 411008 Maharashtra India
| | - Pinaki Talukdar
- Department of Chemistry, Indian Institute of Science Education and Research Pune Dr. Homi Bhabha Road, Pashan Pune 411008 Maharashtra India
| | - Jayanta Haldar
- Antimicrobial Research Laboratory, New Chemistry Unit, JNCASR Jakkur Bangalore-560064 India
- School of Advanced Materials, JNCASR Jakkur Bangalore-560064 India
| |
Collapse
|
12
|
Huang WL, Wang XD, Ao YF, Wang QQ, Wang DX. Reversing the ion transport selectivity through arm modification of an artificial molecular hourglass. Chem Commun (Camb) 2023. [PMID: 37997041 DOI: 10.1039/d3cc04573k] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2023]
Abstract
An arm modification strategy, by replacing relatively rigid, electron-deficient side arms with flexible ether chain arms and linking them onto a tetraoxacalix[2]arene[2]triazine skeleton, was utilized to design an artificial molecular hourglass. The planar bilayer experiments confirmed the unimolecular channel mechanism and suggested reversed ion selectivity from the previously reported anion selectivity to weak cation selectivity.
Collapse
Affiliation(s)
- Wen-Long Huang
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Molecular Recognition and Function, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China.
| | - Xu-Dong Wang
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Molecular Recognition and Function, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China.
| | - Yu-Fei Ao
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Molecular Recognition and Function, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China.
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qi-Qiang Wang
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Molecular Recognition and Function, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China.
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - De-Xian Wang
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Molecular Recognition and Function, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China.
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
13
|
Alonso-Carrillo D, Arias-Betancur A, Carreira-Barral I, Fontova P, Soto-Cerrato V, García-Valverde M, Pérez-Tomás R, Quesada R. Small molecule anion carriers facilitate lactate transport in model liposomes and cells. iScience 2023; 26:107898. [PMID: 37790273 PMCID: PMC10543179 DOI: 10.1016/j.isci.2023.107898] [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: 05/22/2023] [Revised: 08/29/2023] [Accepted: 09/08/2023] [Indexed: 10/05/2023] Open
Abstract
An excessive production of lactate by cancer cells fosters tumor growth and metastasis. Therefore, targeting lactate metabolism and transport offers a new therapeutic strategy against cancer, based on dependency of some cancer cells for lactate as energy fuel or as oncogenic signal. Herein we present a family of anionophores based on the structure of click-tambjamines that have proved to be extremely active lactate carriers across phospholipid membranes. Compound 1, the most potent lactate transmembrane carrier, was studied in HeLa cells. The use of a monocarboxylate transporters (MCTs) inhibitor proved that 1 is an active lactate transporter in living cells, confirming the results obtained in phospholipid vesicles. Moreover, an additive effect of compound 1 with cisplatin was observed in HeLa cells. Identification of active lactate anionophores working in living cells opens up ways to exploit this class of compounds as molecular tools and drugs addressing dysregulated lactate metabolism.
Collapse
Affiliation(s)
- Daniel Alonso-Carrillo
- Departamento de Química, Facultad de Ciencias, Universidad de Burgos, 09001 Burgos, Spain
| | - Alain Arias-Betancur
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Universitat de Barcelona, L’Hospitalet de Llobregat, 08907, Spain
- Molecular Signalling, Oncobell Program, Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), L’Hospitalet de Llobregat, 08908, Spain
- Department of Integral Adult Dentistry, Dental School, Research Centre for Dental Sciences, Universidad de La Frontera, Temuco 4811230, Chile
| | - Israel Carreira-Barral
- Departamento de Química, Facultad de Ciencias, Universidad de Burgos, 09001 Burgos, Spain
| | - Pere Fontova
- Departamento de Química, Facultad de Ciencias, Universidad de Burgos, 09001 Burgos, Spain
| | - Vanessa Soto-Cerrato
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Universitat de Barcelona, L’Hospitalet de Llobregat, 08907, Spain
- Molecular Signalling, Oncobell Program, Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), L’Hospitalet de Llobregat, 08908, Spain
| | - María García-Valverde
- Departamento de Química, Facultad de Ciencias, Universidad de Burgos, 09001 Burgos, Spain
| | - Ricardo Pérez-Tomás
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Universitat de Barcelona, L’Hospitalet de Llobregat, 08907, Spain
- Molecular Signalling, Oncobell Program, Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), L’Hospitalet de Llobregat, 08908, Spain
| | - Roberto Quesada
- Departamento de Química, Facultad de Ciencias, Universidad de Burgos, 09001 Burgos, Spain
| |
Collapse
|
14
|
Tabana Y, Babu D, Fahlman R, Siraki AG, Barakat K. Target identification of small molecules: an overview of the current applications in drug discovery. BMC Biotechnol 2023; 23:44. [PMID: 37817108 PMCID: PMC10566111 DOI: 10.1186/s12896-023-00815-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 09/22/2023] [Indexed: 10/12/2023] Open
Abstract
Target identification is an essential part of the drug discovery and development process, and its efficacy plays a crucial role in the success of any given therapy. Although protein target identification research can be challenging, two main approaches can help researchers make significant discoveries: affinity-based pull-down and label-free methods. Affinity-based pull-down methods use small molecules conjugated with tags to selectively isolate target proteins, while label-free methods utilize small molecules in their natural state to identify targets. Target identification strategy selection is essential to the success of any drug discovery process and must be carefully considered when determining how to best pursue a specific project. This paper provides an overview of the current target identification approaches in drug discovery related to experimental biological assays, focusing primarily on affinity-based pull-down and label-free approaches, and discusses their main limitations and advantages.
Collapse
Affiliation(s)
- Yasser Tabana
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | - Dinesh Babu
- Li Ka Shing Applied Virology Institute, University of Alberta, Edmonton, AB, T6G 2E1, Canada
| | - Richard Fahlman
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | - Arno G Siraki
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - Khaled Barakat
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
15
|
Alonso-Carrillo D, Carreira-Barral I, Mielczarek M, Sancho-Medina A, Herran E, Vairo C, Del Pozo A, Luzuriaga I, Lazcanoiturburu N, Ibarrola O, Ponce S, Villar-Vidal M, García-Valverde M, Quesada R. Formulation and evaluation of anion transporters in nanostructured lipid carriers. Org Biomol Chem 2023; 21:7753-7757. [PMID: 37691616 DOI: 10.1039/d3ob01182h] [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: 09/12/2023]
Abstract
Six novel click-tambjamines (1-6) bearing an alkyl chain of varying length linked to the imine moiety have been formulated in nanostructured lipid carriers (NLCs) to evaluate their transmembrane anion transport activity both when free (i.e., not encapsulated) and nanoformulated. Nanostructured lipid carriers (NLCs) are an example of drug delivery systems (DDSs) that stand out because of their versatility. In this work we show that NLCs can be used to efficiently formulate highly lipophilic anionophores and experiments conducted in model liposomes reveal that these formulations are adequate to deliver anionophores without compromising their transport activity. This result paves the way to facilitate the study of highly lipophilic anionophores and their potential use as future drugs.
Collapse
Affiliation(s)
| | | | - Marcin Mielczarek
- Departamento de Química, Universidad de Burgos, Burgos 09001, Spain.
| | | | - Enara Herran
- Biokeralty Research Institute AIE, Hermanos Lumière 5, 01510 Miñano, Spain
| | - Claudia Vairo
- Biokeralty Research Institute AIE, Hermanos Lumière 5, 01510 Miñano, Spain
| | - Angel Del Pozo
- Biokeralty Research Institute AIE, Hermanos Lumière 5, 01510 Miñano, Spain
| | - Iris Luzuriaga
- Biokeralty Research Institute AIE, Hermanos Lumière 5, 01510 Miñano, Spain
| | | | - Oihane Ibarrola
- Biokeralty Research Institute AIE, Hermanos Lumière 5, 01510 Miñano, Spain
| | - Sara Ponce
- Biokeralty Research Institute AIE, Hermanos Lumière 5, 01510 Miñano, Spain
| | - María Villar-Vidal
- Biokeralty Research Institute AIE, Hermanos Lumière 5, 01510 Miñano, Spain
| | | | - Roberto Quesada
- Departamento de Química, Universidad de Burgos, Burgos 09001, Spain.
| |
Collapse
|
16
|
Bellotto O, D'Andrea P, Marchesan S. Nanotubes and water-channels from self-assembling dipeptides. J Mater Chem B 2023. [PMID: 36790014 DOI: 10.1039/d2tb02643k] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Dipeptides are attractive building blocks for biomaterials in light of their inherent biocompatibility, biodegradability, and simplicity of preparation. Since the discovery of diphenylalanine (Phe-Phe) self-assembling ability into nanotubes, research efforts have been devoted towards the identification of other dipeptide sequences capable of forming these interesting nanomorphologies, although design rules towards nanotube formation are still elusive. In this review, we analyze the dipeptide sequences reported thus far for their ability to form nanotubes, which often feature water-filled supramolecular channels as revealed by single-crystal X-ray diffraction, as well as their properties, and their potential biological applications, which span from drug delivery and regenerative medicine, to bioelectronics and bioimaging.
Collapse
Affiliation(s)
- Ottavia Bellotto
- Chem. Pharm. Sc. Dept., University of Trieste, Via Giorgieri 1, 34127 Trieste, Italy.
| | - Paola D'Andrea
- Life Sc. Dept., University of Trieste, Via Weiss 2, 34128 Trieste, Italy
| | - Silvia Marchesan
- Chem. Pharm. Sc. Dept., University of Trieste, Via Giorgieri 1, 34127 Trieste, Italy. .,INSTM, Unit of Trieste, Via Giorgieri 1, 34127 Trieste, Italy
| |
Collapse
|
17
|
Hitchhiking into a cell: flavonoids may produce complexes with transition metals for transmembrane translocation. Biometals 2022; 35:1299-1306. [PMID: 36161545 DOI: 10.1007/s10534-022-00445-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 09/12/2022] [Indexed: 12/14/2022]
Abstract
Flavonoids are a group of food polyphenols that are delivered to the human body with plant foods. In recent years, these substances have attracted the attention of researchers due to their effectiveness in preventing a wide variety of diseases, including neurodegenerative, oncological, autoimmune, and cardiovascular. Similar pathologies may also occur with a lack of some first-row transition metals, including Cu(II), Zn(II), Mn(II), Fe(II/III). It is noteworthy that flavonoids are known as transition metal chelators. When a complex with these metals is formed, the therapeutic effect of flavonoids can be enhanced, assuming the possibility of synergy. Molecular models have shown that the lipophilicity of flavonoid-metal complexes can vary significantly depending on their binding stoichiometry. Therefore, a unique process of translocation of flavonoid-metal complexes of various lipophilicity through cell membranes is assumed, based on the possibility of their sequential association and dissociation, called "hitchhiking". It is expected that studies of the interaction of flavonoids with metals will improve the effectiveness of drugs based on flavonoids.
Collapse
|
18
|
Self-Assembly and Gelation Study of Dipeptide Isomers with Norvaline and Phenylalanine. CHEMISTRY 2022. [DOI: 10.3390/chemistry4040093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Dipeptides have emerged as attractive building blocks for supramolecular materials thanks to their low-cost, inherent biocompatibility, ease of preparation, and environmental friendliness as they do not persist in the environment. In particular, hydrophobic amino acids are ideal candidates for self-assembly in polar and green solvents, as a certain level of hydrophobicity is required to favor their aggregation and reduce the peptide solubility. In this work, we analyzed the ability to self-assemble and the gel of dipeptides based on the amino acids norvaline (Nva) and phenylalanine (Phe), studying all their combinations and not yielding to enantiomers, which display the same physicochemical properties, and hence the same self-assembly behavior in achiral environments as those studied herein. A single-crystal X-ray diffraction of all the compounds revealed fine details over their molecular packing and non-covalent interactions.
Collapse
|
19
|
Díaz-Cabrera S, Carreira-Barral I, García-Valverde M, Quesada R. Roseophilin-inspired derivatives as transmembrane anion carriers. Supramol Chem 2022. [DOI: 10.1080/10610278.2022.2099277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Affiliation(s)
| | | | | | - Roberto Quesada
- Departamento de Química, Universidad de Burgos, Burgos, Spain
| |
Collapse
|
20
|
Bellotto O, Pierri G, Rozhin P, Polentarutti M, Kralj S, D'Andrea P, Tedesco C, Marchesan S. Dipeptide self-assembly into water-channels and gel biomaterial. Org Biomol Chem 2022; 20:6211-6218. [PMID: 35575102 DOI: 10.1039/d2ob00622g] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Dipeptides are convenient building blocks for supramolecular gel biomaterials that can be produced on a large scale at low cost and do not persist in the environment. In the case of unprotected sequences, hydrophobicity is a key requirement to enable gelation, with Phe-Phe standing out for its self-assembling ability. Conversely, more hydrophilic sequences such as homochiral dipeptides Phe-Val and Val-Phe neither fibrillate nor gel aqueous buffers and their crystal structures reveal amphipathic layers. In this work, we test emerging rules for the design of self-assembling dipeptides using heterochiral Phe-Val and Val-Phe. Each dipeptide is characterized by 1H- and 13C-NMR, LC-MS, circular dichroism, infrared and Raman spectroscopies, rheology, electron microscopy, and single-crystal X-ray diffraction. In particular, D-Phe-L-Val is the first heterochiral dipeptide to self-assemble into supramolecular water-channels whose cavity is defined by four peptide molecules arranged head-to-tail. This minimalistic sequence is devoid of amyloid character as probed by thioflavin T fluorescence and it displays excellent biocompatibility in vitro. The dataset provided, through comparison with the literature, significantly advances the definition of molecular design rules for minimalistic unprotected dipeptides that self-assemble into water-channels and biocompatible gels, to assist with the future development of supramolecular biomaterials with fine control over nanomorphological features for a variety of applications.
Collapse
Affiliation(s)
- Ottavia Bellotto
- University of Trieste, Chem. Pharm. Sc. Dept., Via Giorgieri 1, 34127 Trieste, Italy.
| | - Giovanni Pierri
- University of Salerno, Dept. of Chemistry & Biologi "A. Zambelli", Via Giovanni Paolo II 132, 84084 Fisciano, SA, Italy.
| | - Petr Rozhin
- University of Trieste, Chem. Pharm. Sc. Dept., Via Giorgieri 1, 34127 Trieste, Italy.
| | | | - Slavko Kralj
- Jožef Stefan Institute, Materials Synthesis Dept., Jamova 39, 1000 Ljubljana, Slovenia.,University of Ljubljana, Pharmaceutical Technology Dept., Faculty of Pharmacy, Aškerčeva 7, 1000 Ljubljana, Slovenia
| | - Paola D'Andrea
- University of Trieste, Life Sciences Dept., Via L. Giorgieri 5, 34127 Trieste, Italy
| | - Consiglia Tedesco
- University of Salerno, Dept. of Chemistry & Biologi "A. Zambelli", Via Giovanni Paolo II 132, 84084 Fisciano, SA, Italy.
| | - Silvia Marchesan
- University of Trieste, Chem. Pharm. Sc. Dept., Via Giorgieri 1, 34127 Trieste, Italy.
| |
Collapse
|