1
|
Cimino C, Zingale E, Bonaccorso A, Musumeci T, Carbone C, Pignatello R. From Preformulative Design to In Vivo Tests: A Complex Path of Requisites and Studies for Nanoparticle Ocular Application. Part 1: Design, Characterization, and Preliminary In Vitro Studies. Mol Pharm 2024. [PMID: 39441703 DOI: 10.1021/acs.molpharmaceut.4c00554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Ocular pathologies are widely diffused worldwide, and their effective treatment, combined with a high patient compliance, is sometimes challenging to achieve due to the barriers of the eye; in this context, the use of nanoparticles for topical ophthalmic application could represent a successful strategy. Aiming to develop nanoplatforms with potential clinical applications, great attention has to be paid to their features, in relation to the route of administration and to the pharmacopoeial requirements. This review (part 1) thus embraces the preliminary steps of nanoparticle development and characterization. At the beginning, the main barriers of the eye and the different administration routes are resumed, followed by a general description of the advantages of the employment of nanoparticles for ocular topical administration. Subsequently, the preformulative steps are discussed, deepening the choice of raw materials and determining the quantitative composition. Then, a detailed report of the physicochemical and technological characterization of nanoparticles is presented, analyzing the most relevant tests that should be performed on nanoparticles to verify their properties and the requisites (both mandatory and suggested) demanded by regulatory agencies. In conclusion, some preliminary noncellular in vitro evaluation methods are described. Studies from in vitro cellular assays to in vivo tests will be discussed in a separate (part 2) review paper. Hence, this overview aims to offer a comprehensive tool to guide researchers in the choice of the most relevant studies to develop a nanoplatform for ophthalmic drug administration.
Collapse
Affiliation(s)
- Cinzia Cimino
- Laboratory of Drug Delivery Technology, Department of Drug and Health Sciences, University of Catania, Viale A. Doria 6, 95124 Catania, Italy
- NANOMED, Research Centre for Nanomedicine and Pharmaceutical Nanotechnology, University of Catania, 95124 Catania, Italy
| | - Elide Zingale
- Laboratory of Drug Delivery Technology, Department of Drug and Health Sciences, University of Catania, Viale A. Doria 6, 95124 Catania, Italy
- NANOMED, Research Centre for Nanomedicine and Pharmaceutical Nanotechnology, University of Catania, 95124 Catania, Italy
| | - Angela Bonaccorso
- Laboratory of Drug Delivery Technology, Department of Drug and Health Sciences, University of Catania, Viale A. Doria 6, 95124 Catania, Italy
- NANOMED, Research Centre for Nanomedicine and Pharmaceutical Nanotechnology, University of Catania, 95124 Catania, Italy
| | - Teresa Musumeci
- Laboratory of Drug Delivery Technology, Department of Drug and Health Sciences, University of Catania, Viale A. Doria 6, 95124 Catania, Italy
- NANOMED, Research Centre for Nanomedicine and Pharmaceutical Nanotechnology, University of Catania, 95124 Catania, Italy
| | - Claudia Carbone
- Laboratory of Drug Delivery Technology, Department of Drug and Health Sciences, University of Catania, Viale A. Doria 6, 95124 Catania, Italy
- NANOMED, Research Centre for Nanomedicine and Pharmaceutical Nanotechnology, University of Catania, 95124 Catania, Italy
| | - Rosario Pignatello
- Laboratory of Drug Delivery Technology, Department of Drug and Health Sciences, University of Catania, Viale A. Doria 6, 95124 Catania, Italy
- NANOMED, Research Centre for Nanomedicine and Pharmaceutical Nanotechnology, University of Catania, 95124 Catania, Italy
| |
Collapse
|
2
|
Wang KN, Li ZZ, Zhou K, Liu B, Rao L, Bu LL. Cell Membrane-Coated Nanoparticles for Dental, Oral, and Craniofacial Diseases. RESEARCH (WASHINGTON, D.C.) 2024; 7:0478. [PMID: 39296987 PMCID: PMC11409001 DOI: 10.34133/research.0478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 08/26/2024] [Accepted: 08/29/2024] [Indexed: 09/21/2024]
Abstract
Dental, oral, and craniofacial diseases can substantially impact the quality of human life, thereby posing a serious public health concern. Although conventional therapies such as surgery have solved these problems largely, the prognosis of patients is not always satisfactory. Cell membrane-coated nanoparticles (CMCNPs) carry nanodrugs with the help of natural cell membranes, therefore utilizing their remarkable ability to interface and interact with their surrounding environment. These nanoparticles have demonstrated substantial advantages in drug targeting, prolonging blood circulation time, penetrating biofilms, and immune escape. With the assistance of CMCNPs, the therapeutic effects of dental, oral, and craniofacial diseases can reach a higher level. CMCNPs have been applied for dental, oral, and craniofacial diseases for various conditions such as head and neck cancer, periodontal disease, and oral biosignal detection. For the therapies of head and neck cancer, CMCNPs have been widely utilized as a tool of chemotherapy, phototherapy, and immunotherapy, while yet to be exploited in imaging technique. In the end, we summarized the challenges and prospectives of CMCNPs for dental, oral, and craniofacial diseases: large-scale production with uniform standards and high quantity, extensive application directions in dental, oral, and craniofacial regions (implant, endodontics), and the promotion of its clinical application.
Collapse
Affiliation(s)
- Kang-Ning Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Zi-Zhan Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Kan Zhou
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Bing Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
- Department of Oral & Maxillofacial-Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Lang Rao
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen 518132, China
| | - Lin-Lin Bu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
- Department of Oral & Maxillofacial-Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| |
Collapse
|
3
|
Aljabali AAA, Obeid MA, Gammoh O, El-Tanani M, Mishra V, Mishra Y, Kapre S, Srivatsa Palakurthi S, Hassan SS, Nawn D, Lundstrom K, Hromić-Jahjefendić A, Serrano-Aroca Á, Redwan EM, Uversky VN, Tambuwala MM. Nanomaterial-Driven Precision Immunomodulation: A New Paradigm in Therapeutic Interventions. Cancers (Basel) 2024; 16:2030. [PMID: 38893150 PMCID: PMC11171400 DOI: 10.3390/cancers16112030] [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: 04/26/2024] [Revised: 05/15/2024] [Accepted: 05/21/2024] [Indexed: 06/21/2024] Open
Abstract
Immunotherapy is a rapidly advancing field of research in the treatment of conditions such as cancer and autoimmunity. Nanomaterials can be designed for immune system manipulation, with precise targeted delivery and improved immunomodulatory efficacy. Here, we elaborate on various strategies using nanomaterials, including liposomes, polymers, and inorganic NPs, and discuss their detailed design intricacies, mechanisms, and applications, including the current regulatory issues. This type of nanomaterial design for targeting specific immune cells or tissues and controlling release kinetics could push current technological frontiers and provide new and innovative solutions for immune-related disorders and diseases without off-target effects. These materials enable targeted interactions with immune cells, thereby enhancing the effectiveness of checkpoint inhibitors, cancer vaccines, and adoptive cell therapies. Moreover, they allow for fine-tuning of immune responses while minimizing side effects. At the intersection of nanotechnology and immunology, nanomaterial-based platforms have immense potential to revolutionize patient-centered immunotherapy and reshape disease management. By prioritizing safety, customization, and compliance with regulatory standards, these systems can make significant contributions to precision medicine, thereby significantly impacting the healthcare landscape.
Collapse
Affiliation(s)
- Alaa A. A. Aljabali
- Faculty of Pharmacy, Department of Pharmaceutics & Pharmaceutical Technology, Yarmouk University, Irbid 21163, Jordan; (A.A.A.A.); (M.A.O.)
| | - Mohammad A. Obeid
- Faculty of Pharmacy, Department of Pharmaceutics & Pharmaceutical Technology, Yarmouk University, Irbid 21163, Jordan; (A.A.A.A.); (M.A.O.)
| | - Omar Gammoh
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmacy, Yarmouk University, Irbid 21163, Jordan;
| | - Mohamed El-Tanani
- College of Pharmacy, Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah P.O. Box 11172, United Arab Emirates;
| | - Vijay Mishra
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, Punjab, India;
| | - Yachana Mishra
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara 144411, Punjab, India;
| | - Sumedha Kapre
- Department of Pharmaceutical Sciences, Irma Lerma Rangel School of Pharmacy, Texas A&M University, Kingsville, TX 78363, USA; (S.K.); (S.S.P.)
| | - Sushesh Srivatsa Palakurthi
- Department of Pharmaceutical Sciences, Irma Lerma Rangel School of Pharmacy, Texas A&M University, Kingsville, TX 78363, USA; (S.K.); (S.S.P.)
| | - Sk. Sarif Hassan
- Department of Mathematics, Pingla Thana Mahavidyalaya, Maligram, Paschim Medinipur 721140, West Bengal, India;
| | - Debaleena Nawn
- Indian Research Institute for Integrated Medicine (IRIIM), Unsani, Howrah 711302, West Bengal, India;
| | | | - Altijana Hromić-Jahjefendić
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Hrasnicka Cesta 15, 71000 Sarajevo, Bosnia and Herzegovina;
| | - Ángel Serrano-Aroca
- Biomaterials and Bioengineering Lab, Centro de Investigación Traslacional San Alberto Magno, Universidad Católica de Valencia San Vicente Mártir, c/Guillem de Castro 94, 46001 Valencia, Spain;
| | - Elrashdy M. Redwan
- Department of Biological Science, Faculty of Science, King Abdulaziz University, P.O. Box 80203, Jeddah 21589, Saudi Arabia;
- Centre of Excellence in Bionanoscience Research, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Therapeutic and Protective Proteins Laboratory, Protein Research Department, Genetic Engineering and Biotechnology Research Institute, City for Scientific Research and Technology Applications, New Borg EL-Arab, Alexandria 21934, Egypt
| | - Vladimir N. Uversky
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Murtaza M. Tambuwala
- College of Pharmacy, Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah P.O. Box 11172, United Arab Emirates;
| |
Collapse
|
4
|
Wang N, Floriano Marcelino T, Ade C, Pendlmayr S, Ramos Docampo MA, Städler B. Collagenase motors in gelatine-based hydrogels. NANOSCALE 2024; 16:9935-9943. [PMID: 38690802 DOI: 10.1039/d3nr05712g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2024]
Abstract
Nano/micromotors outperform Brownian motion due to their self-propulsive capabilities and hold promise as carriers for drug delivery across biological barriers such as the extracellular matrix. This study employs poly(2-(diethylamino)ethyl methacrylate) polymer brushes to enhance the collagenase-loading capacity of silica particle-based motors with the aim to systematically investigate the impact of gelatine viscosity, motors' size, and morphology on their propulsion velocity. Notably, 500 nm and 1 μm motors achieve similar speeds as high as ∼15 μm s-1 in stiff gelatine-based hydrogels when triggered with calcium. Taken together, our findings highlight the potential of collagenase-based motors for navigating the extracellular matrix, positioning them as promising candidates for efficient drug delivery.
Collapse
Affiliation(s)
- Nanying Wang
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, 8000, Aarhus, Denmark.
| | - Thaís Floriano Marcelino
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, 8000, Aarhus, Denmark.
- Sino-Danish Center for Education and Research, University of Chinese Academy of Sciences, Beijing, China
| | - Carina Ade
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, 8000, Aarhus, Denmark.
| | - Stefan Pendlmayr
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, 8000, Aarhus, Denmark.
- Sino-Danish Center for Education and Research, University of Chinese Academy of Sciences, Beijing, China
| | - Miguel A Ramos Docampo
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, 8000, Aarhus, Denmark.
| | - Brigitte Städler
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, 8000, Aarhus, Denmark.
| |
Collapse
|
5
|
Tan J, Zhu C, Li L, Wang J, Xia XH, Wang C. Engineering Cell Membranes: From Extraction Strategies to Emerging Biosensing Applications. Anal Chem 2024; 96:7880-7894. [PMID: 38272835 DOI: 10.1021/acs.analchem.3c01746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2024]
Affiliation(s)
- Jing Tan
- College of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, P.R. China
| | - Chengcheng Zhu
- College of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, P.R. China
| | - Lulu Li
- College of Environmental and Chemical Engineering, Jiangsu University of Science and Technology, Zhenjiang 212000, P.R. China
| | - Jin Wang
- College of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, P.R. China
| | - Xing-Hua Xia
- State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing 210023, P.R. China
| | - Chen Wang
- College of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, P.R. China
- State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing 210023, P.R. China
| |
Collapse
|
6
|
Franco Machado J, Sá M, Pires I, da Silva MT, Marques F, Coelho JAS, Mendes F, Piedade MFM, Machuqueiro M, Jiménez MA, Garcia MH, Correia JDG, Morais TS. Dual FGFR-targeting and pH-activatable ruthenium-peptide conjugates for targeted therapy of breast cancer. Dalton Trans 2024; 53:7682-7693. [PMID: 38573236 DOI: 10.1039/d4dt00497c] [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: 04/05/2024]
Abstract
Dysregulation of Fibroblast Growth Factor Receptors (FGFRs) signaling has been associated with breast cancer, yet employing FGFR-targeted delivery systems to improve the efficacy of cytotoxic agents is still sparsely exploited. Herein, we report four new bi-functional ruthenium-peptide conjugates (RuPCs) with FGFR-targeting and pH-dependent releasing abilities, envisioning the selective delivery of cytotoxic Ru complexes to FGFR(+)-breast cancer cells, and controlled activation at the acidic tumoral microenvironment. The antiproliferative potential of the RuPCs and free Ru complexes was evaluated in four breast cancer cell lines with different FGFR expression levels (SKBR-3, MDA-MB-134-VI, MCF-7, and MDA-MB-231) and in human dermal fibroblasts (HDF), at pH 6.8 and pH 7.4 aimed at mimicking the tumor microenvironment and normal tissues/bloodstream pHs, respectively. The RuPCs showed higher cytotoxicity in cells with higher level of FGFR expression at acidic pH. Additionally, RuPCs showed up to 6-fold higher activity in the FGFR(+) breast cancer lines compared to the normal cell line. The release profile of Ru complexes from RuPCs corroborates the antiproliferative effects observed. Remarkably, the cytotoxicity and releasing ability of RuPCs were shown to be strongly dependent on the conjugation of the peptide position in the Ru complex. Complementary molecular dynamic simulations and computational calculations were performed to help interpret these findings at the molecular level. In summary, we identified a lead bi-functional RuPC that holds strong potential as a FGFR-targeted chemotherapeutic agent.
Collapse
Affiliation(s)
- João Franco Machado
- Centro de Química Estrutural, Institute of Molecular Sciences, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal.
- Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10 (km 139, 7), 2695-066 Bobadela LRS, Portugal.
| | - Marco Sá
- Centro de Química Estrutural, Institute of Molecular Sciences, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal.
| | - Inês Pires
- BioISI - Biosystems & Integrative Sciences Institute, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
| | - Miguel Tarita da Silva
- Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10 (km 139, 7), 2695-066 Bobadela LRS, Portugal.
| | - Fernanda Marques
- Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10 (km 139, 7), 2695-066 Bobadela LRS, Portugal.
- Departamento de Engenharia e Ciências Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10 (km 139, 7), 2695-066 Bobadela LRS, Portugal
| | - Jaime A S Coelho
- Centro de Química Estrutural, Institute of Molecular Sciences, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal.
| | - Filipa Mendes
- Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10 (km 139, 7), 2695-066 Bobadela LRS, Portugal.
- Departamento de Engenharia e Ciências Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10 (km 139, 7), 2695-066 Bobadela LRS, Portugal
| | - M Fátima M Piedade
- Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
- Centro de Química Estrutural, Institute of Molecular Sciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Miguel Machuqueiro
- BioISI - Biosystems & Integrative Sciences Institute, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
- Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
| | - María Angeles Jiménez
- Institute of Physical Chemistry Blas Cabreras (IQF-CSIC), Serrano 119, E-28006 Madrid, Spain
| | - Maria Helena Garcia
- Centro de Química Estrutural, Institute of Molecular Sciences, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal.
- Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
| | - João D G Correia
- Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10 (km 139, 7), 2695-066 Bobadela LRS, Portugal.
- Departamento de Engenharia e Ciências Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10 (km 139, 7), 2695-066 Bobadela LRS, Portugal
| | - Tânia S Morais
- Centro de Química Estrutural, Institute of Molecular Sciences, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal.
- Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
| |
Collapse
|
7
|
Mazlee MTF, Heidelberg T, Ariffin A, Zain SM. Cation-stimulated drug delivery via lipid assembly comprising macrocyclized disaccharides - A DFT study. Carbohydr Res 2023; 532:108923. [PMID: 37598565 DOI: 10.1016/j.carres.2023.108923] [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: 06/11/2023] [Revised: 08/07/2023] [Accepted: 08/09/2023] [Indexed: 08/22/2023]
Abstract
In the attempt to create a delivery system for an alkali-cation stimulated drug release, a computational study was conducted, aiming for the evaluation of synthetic access towards glycolipid crown ethers analogs and their potential for coordination-induced changes of packing constraints for molecular assemblies. The results disfavor amide-linkages for the creation of macrocycles around the inter-glycosidic bond of a disaccharide. Conformational changes upon cation coordination of the macrocycle decrease the intersection area for easily accessible macrocycles based on lactose. This leads to shrinking intersection areas upon alkali complexation. Maltose-based analogs, on the other hand, exhibited the targeted increase of the glycolipid intersection area and, hence, may be considered as a promising resource.
Collapse
Affiliation(s)
| | - Thorsten Heidelberg
- Chemistry Department, Faculty of Science, University of Malaya, 50603, Kuala Lumpur, Malaysia.
| | - Azhar Ariffin
- Chemistry Department, Faculty of Science, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Sharifuddin Md Zain
- Chemistry Department, Faculty of Science, University of Malaya, 50603, Kuala Lumpur, Malaysia
| |
Collapse
|
8
|
Wang Y, Mou X, Ji Y, Pan F, Li S. Interaction of Macromolecular Chain with Phospholipid Membranes in Solutions: A Dissipative Particle Dynamics Simulation Study. Molecules 2023; 28:5790. [PMID: 37570760 PMCID: PMC10420874 DOI: 10.3390/molecules28155790] [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: 07/03/2023] [Revised: 07/24/2023] [Accepted: 07/27/2023] [Indexed: 08/13/2023] Open
Abstract
The interaction between macromolecular chains and phospholipid membranes in aqueous solution was investigated using dissipative particle dynamics simulations. Two cases were considered, one in which the macromolecular chains were pulled along parallel to the membrane surfaces and another in which they were pulled vertical to the membrane surfaces. Several parameters, including the radius of gyration, shape factor, particle number, and order parameter, were used to investigate the interaction mechanisms during the dynamics processes by adjusting the pulling force strength of the chains. In both cases, the results showed that the macromolecular chains undergo conformational transitions from a coiled to a rod-like structure. Furthermore, the simulations revealed that the membranes can be damaged and repaired during the dynamic processes. The role of the pulling forces and the adsorption interactions between the chains and membranes differed in the parallel and perpendicular pulling cases. These findings contribute to our understanding of the interaction mechanisms between macromolecules and membranes, and they may have potential applications in biology and medicine.
Collapse
Affiliation(s)
- Yuane Wang
- Department of Physics, Wenzhou University, Wenzhou 325035, China; (Y.W.); (X.M.); (Y.J.)
| | - Xuankang Mou
- Department of Physics, Wenzhou University, Wenzhou 325035, China; (Y.W.); (X.M.); (Y.J.)
| | - Yongyun Ji
- Department of Physics, Wenzhou University, Wenzhou 325035, China; (Y.W.); (X.M.); (Y.J.)
| | - Fan Pan
- School of Data Science and Artificial Intelligence, Wenzhou University of Technology, Wenzhou 325035, China
| | - Shiben Li
- Department of Physics, Wenzhou University, Wenzhou 325035, China; (Y.W.); (X.M.); (Y.J.)
| |
Collapse
|
9
|
Gavutis M, Schulze-Niemand E, Lee HH, Liedberg B, Stein M, Valiokas R. Bilayer lipid membrane formation on surface assemblies with sparsely distributed tethers. NANOSCALE 2023. [PMID: 37128711 DOI: 10.1039/d2nr07069c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
A combined computational and experimental study of small unilamellar vesicle (SUV) fusion on mixed self-assembled monolayers (SAMs) terminated with different deuterated tether moieties (-(CD2)7CD3 or -(CD2)15CD3) is reported. Tethered bilayer lipid membrane (tBLM) formation of synthetic 1-stearoyl-2-oleoyl-sn-glycero-3-phosphocholine was initially probed on SAMs with controlled tether (d-alkyl tail) surface densities and lateral molecular packing using quartz crystal microbalance with dissipation monitoring (QCM-D). Long time-scale coarse-grained molecular dynamics (MD) simulations were then employed to elucidate the mechanisms behind the interaction between the SUVs and the different phases formed by the -(CD2)7CD3 and -(CD2)15CD3 tethers. Furthermore, a series of real time kinetics was recorded under different osmotic conditions using QCM-D to determine the accumulated lipid mass and for probing the fusion process. It is shown that the key factors driving the SUV fusion and tBLM formation on this type of surfaces involve tether insertion into the SUVs along with vesicle deformation. It is also evident that surface densities of the tethers as small as a few mol% are sufficient to obtain stable tBLMs with a high reproducibility. The described "sparsely tethered" tBLM system can be advantageous in studying different biophysical phenomena, such as membrane protein insertion, effects of receptor clustering, and raft formation.
Collapse
Affiliation(s)
- Martynas Gavutis
- Department of Nanoengineering, Center for Physical Sciences and Technology, Savanorių 231, 02300 Vilnius, Lithuania.
| | - Eric Schulze-Niemand
- Molecular Simulations and Design Group, Max Planck Institute for Dynamics of Complex Technical System, Magdeburg, Germany
- Institute of Experimental Internal Medicine, Medical Faculty, Otto von Guericke University, 39120 Magdeburg, Germany
| | - Hung-Hsun Lee
- Division of Molecular Physics, Department of Physics, Chemistry and Biology, Linköping University, 58183 Linköping, Sweden
| | - Bo Liedberg
- Division of Molecular Physics, Department of Physics, Chemistry and Biology, Linköping University, 58183 Linköping, Sweden
| | - Matthias Stein
- Molecular Simulations and Design Group, Max Planck Institute for Dynamics of Complex Technical System, Magdeburg, Germany
| | - Ramūnas Valiokas
- Department of Nanoengineering, Center for Physical Sciences and Technology, Savanorių 231, 02300 Vilnius, Lithuania.
| |
Collapse
|
10
|
Szczukowski Ł, Maniewska J, Wiatrak B, Jawień P, Krzyżak E, Kotynia A, Marciniak A, Janeczek M, Redzicka A. Interactions of N-Mannich Bases of Pyrrolo[3,4- c]pyrrole with Artificial Models of Cell Membranes and Plasma Proteins, Evaluation of Anti-Inflammatory and Antioxidant Activity. MEMBRANES 2023; 13:349. [PMID: 36984737 PMCID: PMC10057445 DOI: 10.3390/membranes13030349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/10/2023] [Accepted: 03/14/2023] [Indexed: 06/18/2023]
Abstract
Despite the widespread and easy access to NSAIDs, effective and safe treatment of various inflammatory disorders is still a serious challenge because of the severe adverse effects distinctive to these drugs. The Mannich base derivatives of pyrrolo[3,4-c]pyrrole are potent, preferential COX-2 inhibitors with a COX-2/COX-1 inhibitory ratio better than meloxicam. Therefore, we chose the six most promising molecules and subjected them to further in-depth research. The current study presents the extensive biological, spectroscopic and in silico evaluation of the activity and physicochemical properties of pyrrolo[3,4-c]pyrrole derivatives. Aware of the advantages of dual COX-LOX inhibition, we investigated the 15-LOX inhibitory activity of these molecules. We also examined their antioxidant effect in several in vitro experiments in a protection and regeneration model. Furthermore, we defined how studied compounds interact with artificial models of cell membranes, which is extremely important for drugs administered orally with an intracellular target. The interactions and binding mode of the derivatives with the most abundant plasma proteins-human serum albumin and alpha-1-acid glycoprotein-are also described. Finally, we used computational techniques to evaluate their pharmacokinetic properties. According to the obtained results, we can state that pyrrolo[3,4-c]pyrrole derivatives are promising anti-inflammatory and antioxidant agents with potentially good membrane permeability.
Collapse
Affiliation(s)
- Łukasz Szczukowski
- Department of Medicinal Chemistry, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211, 50-556 Wroclaw, Poland
| | - Jadwiga Maniewska
- Department of Medicinal Chemistry, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211, 50-556 Wroclaw, Poland
| | - Benita Wiatrak
- Department of Pharmacology, Faculty of Medicine, Wroclaw Medical University, Mikulicza-Radeckiego 2, 50-345 Wroclaw, Poland
| | - Paulina Jawień
- Department of Biostructure and Animal Physiology, Division of Animal Anatomy, Faculty of Veterinary Medicine, Wroclaw University of Environmental and Life Sciences, Kożuchowska 1, 51-631 Wroclaw, Poland
| | - Edward Krzyżak
- Department of Basic Chemical Sciences, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211a, 50-556 Wroclaw, Poland
| | - Aleksandra Kotynia
- Department of Basic Chemical Sciences, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211a, 50-556 Wroclaw, Poland
| | - Aleksandra Marciniak
- Department of Basic Chemical Sciences, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211a, 50-556 Wroclaw, Poland
| | - Maciej Janeczek
- Department of Biostructure and Animal Physiology, Division of Animal Anatomy, Faculty of Veterinary Medicine, Wroclaw University of Environmental and Life Sciences, Kożuchowska 1, 51-631 Wroclaw, Poland
| | - Aleksandra Redzicka
- Department of Medicinal Chemistry, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211, 50-556 Wroclaw, Poland
| |
Collapse
|
11
|
Fopase R, Panda C, Rajendran AP, Uludag H, Pandey LM. Potential of siRNA in COVID-19 therapy: Emphasis on in silico design and nanoparticles based delivery. Front Bioeng Biotechnol 2023; 11:1112755. [PMID: 36814718 PMCID: PMC9939533 DOI: 10.3389/fbioe.2023.1112755] [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: 11/30/2022] [Accepted: 01/13/2023] [Indexed: 02/09/2023] Open
Abstract
Small interfering RNA (siRNA)-mediated mRNA degradation approach have imparted its eminence against several difficult-to-treat genetic disorders and other allied diseases. Viral outbreaks and resulting pandemics have repeatedly threatened public health and questioned human preparedness at the forefront of drug design and biomedical readiness. During the recent pandemic caused by the SARS-CoV-2, mRNA-based vaccination strategies have paved the way for a new era of RNA therapeutics. RNA Interference (RNAi) based approach using small interfering RNA may complement clinical management of the COVID-19. RNA Interference approach will primarily work by restricting the synthesis of the proteins required for viral replication, thereby hampering viral cellular entry and trafficking by targeting host as well as protein factors. Despite promising benefits, the stability of small interfering RNA in the physiological environment is of grave concern as well as site-directed targeted delivery and evasion of the immune system require immediate attention. In this regard, nanotechnology offers viable solutions for these challenges. The review highlights the potential of small interfering RNAs targeted toward specific regions of the viral genome and the features of nanoformulations necessary for the entrapment and delivery of small interfering RNAs. In silico design of small interfering RNA for different variants of SARS-CoV-2 has been discussed. Various nanoparticles as promising carriers of small interfering RNAs along with their salient properties, including surface functionalization, are summarized. This review will help tackle the real-world challenges encountered by the in vivo delivery of small interfering RNAs, ensuring a safe, stable, and readily available drug candidate for efficient management of SARS-CoV-2 in the future.
Collapse
Affiliation(s)
- Rushikesh Fopase
- Bio-Interface & Environmental Engineering Lab, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam, India
| | - Chinmaya Panda
- Bio-Interface & Environmental Engineering Lab, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam, India
| | - Amarnath P. Rajendran
- Department of Chemical & Materials Engineering, Faculty of Engineering, University of Alberta, Edmonton, AB, Canada
| | - Hasan Uludag
- Department of Chemical & Materials Engineering, Faculty of Engineering, University of Alberta, Edmonton, AB, Canada
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
- Department of Biomedical Engineering, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Lalit M. Pandey
- Bio-Interface & Environmental Engineering Lab, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam, India
| |
Collapse
|
12
|
Starosta R, Santos TC, Dinis de Sousa AF, Santos MS, Corvo ML, Tomaz AI, de Almeida RFM. Assessing the role of membrane lipids in the action of ruthenium(III) anticancer compounds. Front Mol Biosci 2023; 9:1059116. [PMID: 36660430 PMCID: PMC9845782 DOI: 10.3389/fmolb.2022.1059116] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 12/12/2022] [Indexed: 01/05/2023] Open
Abstract
This work addresses the possible role of the cell membrane in the molecular mechanism of action of two salan-type ruthenium complexes that were previously shown to be active against human tumor cells, namely [Ru(III)(L1)(PPh3)Cl] and [Ru(III)(L2)(PPh3)Cl] (where L1 is 6,6'-(1R,2R)-cyclohexane-1,2-diylbis(azanediyl)bis(methylene)bis(3-methoxyphenol); and L2 is 2,2'-(1R,2R)-cyclohexane-1,2-diylbis(azanediyl)bis(methylene)bis(4-methoxyphenol)). One-component membrane models were first used, a disordered fluid bilayer of dioleoylphosphatodylcholine (DOPC), and an ordered rigid gel bilayer of dipalmitoylphosphatidylcholine. In addition, two quaternary mixtures of phosphatidylcholine, phosphatidylethanolamine, sphingomyelin and cholesterol were used to mimic the lipid composition either of mammalian plasma membrane (1:1:1:1 mol ratio) or of a cancer cell line membrane (36.2:23.6:6.8:33.4 mol ratio). The results show that both salan ligands L1 and L2 bind relatively strongly to DOPC bilayers, but without significantly affecting their structure. The ruthenium complexes have moderate affinity for DOPC. However, their impact on the membranes was notable, leading to a significant increase in the permeability of the lipid vesicles. None of the compounds compromised liposome integrity, as revealed by dynamic light scattering. Fluorescence spectroscopy studies revealed changes in the biophysical properties of all membrane models analyzed in the presence of the two complexes, which promoted an increased fluidity and water penetration into the lipid bilayer in the one-component systems. In the quaternary mixtures, one of the complexes had an analogous effect (increasing water penetration), whereas the other complex reorganized the liquid ordered and liquid disordered domains. Thus, small structural differences in the metal ligands may lead to different outcomes. To better understand the effect of these complexes in cancer cells, the membrane dipole potential was also measured. For both Ru complexes, an increase in the dipole potential was observed for the cancer cell membrane model, while no alteration was detected on the non-cancer plasma membrane model. Our results show that the action of the Ru(III) complexes tested involves changes in the biophysical properties of the plasma membrane, and that it also depends on membrane lipid composition, which is frequently altered in cancer cells when compared to their normal counterparts.
Collapse
Affiliation(s)
- Radoslaw Starosta
- Faculty of Chemistry, University of Wroclaw, Wroclaw, Poland,Centro de Química Estrutural, Institute of Molecular Sciences, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Lisbon, Portugal
| | - Telma C. Santos
- Centro de Química Estrutural, Institute of Molecular Sciences, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Lisbon, Portugal
| | - Andreia F. Dinis de Sousa
- Centro de Química Estrutural, Institute of Molecular Sciences, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Lisbon, Portugal
| | - Maria Soledade Santos
- Centro de Química Estrutural, Institute of Molecular Sciences, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Lisbon, Portugal
| | - M. Luisa Corvo
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal
| | - Ana Isabel Tomaz
- Centro de Química Estrutural, Institute of Molecular Sciences, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Lisbon, Portugal,*Correspondence: Rodrigo F. M. de Almeida, ; Ana Isabel Tomaz,
| | - Rodrigo F. M. de Almeida
- Centro de Química Estrutural, Institute of Molecular Sciences, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Lisbon, Portugal,*Correspondence: Rodrigo F. M. de Almeida, ; Ana Isabel Tomaz,
| |
Collapse
|
13
|
Guo J, Du X, Huang J, Liu C, Zhou Y, Li Y, Du B. Robust Dual Enzyme Cascade-Catalytic Cholesterol Depletion for Reverse Tumor Multidrug Resistance. Adv Healthc Mater 2022; 11:e2200859. [PMID: 35906730 DOI: 10.1002/adhm.202200859] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 06/30/2022] [Indexed: 01/27/2023]
Abstract
Although combination drugs and P-glycoprotein inhibitors are the main methods to solve multidrug resistance, these methods ignore the pathological structure of drug-resistant cells and extremely limit curative effect. Herein, a new paradigm of reversing multidrug resistance with abnormal expression of cholesterol as the target is proposed, which uses the cascade catalysis of "natural enzyme" cholesterol oxidase (COD) and "nanoenzyme" Cu2+ -modified zirconium-based metal-organic framework (ZrMOF(Cu)) to convert cholesterol into the highly cytotoxic hydroxyl radicals. The doxorubicin (DOX)-loaded nanoparticles (DOX@COD-MOF) can significantly reduce the cholesterol content of cancer cells via COD, which decrease the rigidity of drug resistant cancer cell membranes and restore the sensitivity of multidrug-resistant cells to DOX. Afterward, DOX@COD-MOF is encapsulated by cancer cell membranes (CCM) to construct a bionic "dual enzyme catalytic cascade nanoreactor" (DOX@COD-MOF@CCM). Such a rational design presents a preferential accumulation tendency to tumor sites due to the homologous targeting mechanism of CCM, and affords 94.4% in tumor growth suppression without systemic toxicity in vivo. This work aims to achieve the therapeutic purpose of high efficiency and low toxicity. It has the characteristics of "converting enemy into friend, " and opens up a promising way for effectively reversing multidrug resistance of tumors.
Collapse
Affiliation(s)
- Jialing Guo
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Xiaoming Du
- Key Laboratory of Photochemistry, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
| | - Jingshu Huang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Chenxin Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Yingying Zhou
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Ying Li
- Department of Pharmacy, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, 450007, China
| | - Bin Du
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China.,Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou, Henan Province, 450001, China
| |
Collapse
|
14
|
Pedrosa M, Maldonado-Valderrama J, Gálvez-Ruiz MJ. Interactions between curcumin and cell membrane models by Langmuir monolayers. Colloids Surf B Biointerfaces 2022; 217:112636. [PMID: 35738079 DOI: 10.1016/j.colsurfb.2022.112636] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 06/09/2022] [Accepted: 06/11/2022] [Indexed: 10/18/2022]
Abstract
Studying interactions between potential anticancer drugs and cell membrane models is of great interest to explore the capability of novel drugs in the development of anticancer treatments. Lipid membrane models are useful to understand cellular interactions and to discern drug mechanism action. Here, the interactions of curcumin, as a bioactive natural compound with anti-cancer properties, with both healthy and cancerous or tumor cell membrane models, based on Langmuir monolayers, have been studied. The healthy-cell membrane model is composed of cholesterol 67%, and saturated lipid dipalmitoylphosphatidylcholine 33%. The cancerous-cell-membrane-model is composed of a lower proportion of cholesterol, 25%, and unsaturated lipid sphingomyelin 75%. To compare their interaction with curcumin we report the compression isotherms registered for both lipid membrane models and curcumin in different proportions, their compression moduli and the thermodynamic interaction parameters. From this analysis, we evidence a destabilizing interaction between curcumin and the cancerous cell membrane model in comparison with the healthy one. This interaction is further visualized by micro-Brewster Angle and Atomic Force Microscopies. Our experiments show that the drug enhances cohesion in the healthy membrane model whereas it fluidifies the cancerous cell membrane model causing thermodynamic destabilization. These are useful results to improve the selectivity of the drug avoiding adverse side effects of most current anticancer therapies.
Collapse
Affiliation(s)
- María Pedrosa
- Biocolloids and Fluid Physics Group, Department of Applied Physics, University of Granada, Campus Fuente Nueva, s/n, C.P. 18071, Granada, Spain; Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Cuesta del Hospicio, s/n, C.P. 18010, Granada, Spain
| | - Julia Maldonado-Valderrama
- Biocolloids and Fluid Physics Group, Department of Applied Physics, University of Granada, Campus Fuente Nueva, s/n, C.P. 18071, Granada, Spain; Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Cuesta del Hospicio, s/n, C.P. 18010, Granada, Spain
| | - María José Gálvez-Ruiz
- Biocolloids and Fluid Physics Group, Department of Applied Physics, University of Granada, Campus Fuente Nueva, s/n, C.P. 18071, Granada, Spain; Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Cuesta del Hospicio, s/n, C.P. 18010, Granada, Spain.
| |
Collapse
|
15
|
El-Beyrouthy J, Makhoul-Mansour MM, Freeman EC. Studying the Mechanics of Membrane Permeabilization through Mechanoelectricity. ACS APPLIED MATERIALS & INTERFACES 2022; 14:6120-6130. [PMID: 35073482 DOI: 10.1021/acsami.1c19880] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
In this research, real-time monitoring of lipid membrane disruption is made possible by exploiting the dynamic properties of model lipid bilayers formed at oil-water interfaces. This involves tracking an electrical signal generated through rhythmic membrane perturbation translated into the adsorption and penetration of charged species within the membrane. Importantly, this allows for the detection of membrane surface interactions that occur prior to pore formation that may be otherwise undetected. The requisite dynamic membranes for this approach are made possible through the droplet interface bilayer (DIB) technique. Membranes are formed at the interface of lipid monolayer-coated aqueous droplets submerged in oil. We present how cyclically alternating the membrane area leads to the generation of mechanoelectric current. This current is negligible without a transmembrane voltage until a composition mismatch between the membrane monolayers is produced, such as a one-sided accumulation of disruptive agents. The generated mechanoelectric current is then eliminated when an applied electric field compensates for this asymmetry, enabling measurement of the transmembrane potential offset. Tracking the compensating voltage with respect to time then reveals the gradual accumulation of disruptive agents prior to membrane permeabilization. The innovation of this work is emphasized in its ability to continuously track membrane surface activity, highlighting the initial interaction steps of membrane disruption. In this paper, we begin by validating our proposed approach against measurements taken for fixed composition membranes using standard electrophysiological techniques. Next, we investigate surfactant adsorption, including hexadecyltrimethylammonium bromide (CTAB, cationic) and sodium decyl sulfate (SDS, anionic), demonstrating the ability to track adsorption prior to disruption. Finally, we investigate the penetration of lipid membranes by melittin, confirming that the peptide insertion and disruption mechanics are, in part, modulated by membrane composition.
Collapse
Affiliation(s)
- Joyce El-Beyrouthy
- Biomembranes Engineering Laboratory, School of Environment, Civil, Agriculture and Mechanical Engineering, The University of Georgia, Athens, Georgia 30602, United States
| | - Michelle M Makhoul-Mansour
- Biomembranes Engineering Laboratory, School of Environment, Civil, Agriculture and Mechanical Engineering, The University of Georgia, Athens, Georgia 30602, United States
| | - Eric C Freeman
- Biomembranes Engineering Laboratory, School of Environment, Civil, Agriculture and Mechanical Engineering, The University of Georgia, Athens, Georgia 30602, United States
| |
Collapse
|
16
|
Camacho SA, Kobal MB, Moreira LG, Bistaffa MJ, Roque TC, Pazin WM, Toledo KA, Oliveira ON, Aoki PHB. The efficiency of photothermal action of gold shell-isolated nanoparticles against tumor cells depends on membrane interactions. Colloids Surf B Biointerfaces 2021; 211:112301. [PMID: 34968778 DOI: 10.1016/j.colsurfb.2021.112301] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 11/17/2021] [Accepted: 12/16/2021] [Indexed: 12/26/2022]
Abstract
Photoinduced hyperthermia with nanomaterials has been proven effective in photothermal therapy (PTT) of tumor tissues, but a precise control in PTT requires determination of the molecular-level mechanisms. In this paper, we determined the mechanisms responsible for the action of photoexcited gold shell-isolated nanoparticles (AuSHINs) in reducing the viability of MCF7 (glandular breast cancer) and especially A549 (lung adenocarcinoma) cells in vitro experiments, while the photoinduced damage to healthy cells was much smaller. The photoinduced effects were more significant than using other nanomaterials, and could be explained by the different effects from incorporating AuSHINs on Langmuir monolayers from lipid extracts of tumoral (MCF7 and A549) and healthy cells. The incorporation of AuSHINs caused similar expansion of the Langmuir monolayers, but Fourier-transform infrared spectroscopy (FTIR) data of Langmuir-Schaefer films (LS) indicated distinct levels of penetration into the monolayers. AuSHINs penetrated deeper into the A549 extract monolayers, affecting the vibrational modes of polar groups and carbon chains, while in MCF7 monolayers penetration was limited to the surroundings of the polar groups. Even smaller insertion was observed for monolayers of the healthy cell extract. The photochemical reactions were modulated by AuSHINs penetration, since upon irradiation the surface area of A549 monolayer decreased owing to lipid chain cleavage by oxidative reactions. For MCF7 monolayers, hydroperoxidation under illumination led to a ca. 5% increase in surface area. The monolayers of healthy cell lipid extract were barely affected by irradiation, consistent with the lowest degree of AuSHINs insertion. In summary, efficient photothermal therapy may be devised by producing AuSHINs capable of penetrating the chain region of tumor cell membranes.
Collapse
Affiliation(s)
- Sabrina A Camacho
- São Paulo State University (UNESP), School of Sciences, Humanities and Languages, Assis, SP 19806-900, Brazil; IFSC, São Carlos Institute of Physics, University of São Paulo (USP), São Carlos, SP 13566-590, Brazil
| | - Mirella B Kobal
- São Paulo State University (UNESP), School of Sciences, Humanities and Languages, Assis, SP 19806-900, Brazil
| | - Lucas G Moreira
- São Paulo State University (UNESP), School of Sciences, Humanities and Languages, Assis, SP 19806-900, Brazil
| | - Maria J Bistaffa
- São Paulo State University (UNESP), School of Sciences, Humanities and Languages, Assis, SP 19806-900, Brazil
| | - Thamires C Roque
- São Paulo State University (UNESP), School of Sciences, Humanities and Languages, Assis, SP 19806-900, Brazil
| | - Wallance M Pazin
- IFSC, São Carlos Institute of Physics, University of São Paulo (USP), São Carlos, SP 13566-590, Brazil; São Paulo State University (UNESP), School of Technology and Applied Sciences, Presidente Prudente, SP 19060-900, Brazil
| | - Karina A Toledo
- São Paulo State University (UNESP), School of Sciences, Humanities and Languages, Assis, SP 19806-900, Brazil; São Paulo State University (UNESP), Institute of Biosciences, Letters and Exact Sciences, São José do Rio Preto 15054-000, Brazil
| | - Osvaldo N Oliveira
- São Paulo State University (UNESP), School of Sciences, Humanities and Languages, Assis, SP 19806-900, Brazil; IFSC, São Carlos Institute of Physics, University of São Paulo (USP), São Carlos, SP 13566-590, Brazil
| | - Pedro H B Aoki
- São Paulo State University (UNESP), School of Sciences, Humanities and Languages, Assis, SP 19806-900, Brazil.
| |
Collapse
|
17
|
Materón EM, Shimizu FM, Figueiredo Dos Santos K, Nascimento GF, Geraldo VPN, Oliveira ON, Faria RC. Membrane model as key tool in the study of glutathione-s-transferase mediated anticancer drug resistance. Biomed Pharmacother 2021; 145:112426. [PMID: 34861633 DOI: 10.1016/j.biopha.2021.112426] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 11/05/2021] [Accepted: 11/12/2021] [Indexed: 11/02/2022] Open
Abstract
Glutathione-s-transferase is believed to be involved in the resistance to chemotherapeutic drugs, which depends on the interaction with the cell membranes. In this study, we employed Langmuir monolayers of a mixture of phospholipids and cholesterol (MIX) as models for tumor cell membranes and investigated their interaction with the anticancer drugs cisplatin (CDDP) and doxorubicin (DOX). We found that both DOX and CDDP expand and affect the elasticity of MIX monolayers, but these effects are hindered when glutathione-s-transferase (GST) and its cofactor glutathione (GSH) are incorporated. Changes are induced by DOX or CDDP on the polarization-modulated infrared reflection absorption spectroscopy (PM-IRRAS) data for MIX/GST/GSH monolayers, thus denoting some degree of interaction that is not sufficient to alter the monolayer mechanical properties. Overall, the results presented here give support to the hypothesis of the inactivation of DOX and CDDP by GST and point to possible directions to detect and fight drug resistance.
Collapse
Affiliation(s)
- Elsa M Materón
- Chemistry Department, Federal University of São Carlos, CP 676, São Carlos 13565-905, São Paulo, Brazil; São Carlos Institute of Physics, University of São Paulo, P.O Box 369, 13560-970 São Carlos, SP, Brazil.
| | - Flavio M Shimizu
- São Carlos Institute of Physics, University of São Paulo, P.O Box 369, 13560-970 São Carlos, SP, Brazil; Department of Applied Physics, "Gleb Wataghin" Institute of Physics (IFGW), University of Campinas (UNICAMP), Campinas, SP 13083-859, Brazil.
| | | | - Gustavo F Nascimento
- São Carlos Institute of Physics, University of São Paulo, P.O Box 369, 13560-970 São Carlos, SP, Brazil
| | - Vananélia P N Geraldo
- São Carlos Institute of Physics, University of São Paulo, P.O Box 369, 13560-970 São Carlos, SP, Brazil
| | - Osvaldo N Oliveira
- São Carlos Institute of Physics, University of São Paulo, P.O Box 369, 13560-970 São Carlos, SP, Brazil.
| | - Ronaldo C Faria
- Chemistry Department, Federal University of São Carlos, CP 676, São Carlos 13565-905, São Paulo, Brazil.
| |
Collapse
|
18
|
Wang XY, Lv J, Hong Q, Zhou ZR, Li DW, Qian RC. Nanopipette-Based Nanosensor for Label-Free Electrochemical Monitoring of Cell Membrane Rupture under H 2O 2 Treatment. Anal Chem 2021; 93:13967-13973. [PMID: 34623143 DOI: 10.1021/acs.analchem.1c03313] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
H2O2 is an essential signaling molecule in living cells that can cause direct damage to lipids, proteins, and DNA, resulting in cell membrane rupture. However, current studies mostly focus on probe-based sensing of intracellular H2O2, and these methods usually require sophisticated probe synthesis and instruments. In particular, local H2O2 treatment induces cell membrane rupture, but the level of cell membrane destruction is unknown because the mechanical properties of the cell membrane are difficult to accurately determine. Therefore, highly sensitive and label-free methods are required to measure and reflect mechanical changes in the cell membrane. Here, using an ultrasmall quartz nanopipette with a tip diameter less than 90 nm as a nanosensor, label-free and noninvasive electrochemical single-cell measurement is achieved for real-time monitoring of cell membrane rupture under H2O2 treatment. By spatially controlling the nanopipette tip to precisely approach a specific location on the membrane of a single living cell, stable cyclic membrane oscillations are observed under a constant direct current voltage. Specifically, upon nanopipette advancement, the mechanical status of the cell membrane can be sensibly displayed by continuous current versus time traces. The electrical signals are collected and processed, ultimately revealing the mechanical properties of the cell membrane and the degree of cell apoptosis. This nanopipette-based nanosensor paves the way for developing a facile, label-free, and noninvasive strategy to assay the mechanical properties of the cell membrane during external stimulation at the single-cell level.
Collapse
Affiliation(s)
- Xiao-Yuan Wang
- Key Laboratory for Advanced Materials & School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai 200237, P. R. China
| | - Jian Lv
- Key Laboratory for Advanced Materials & School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai 200237, P. R. China
| | - Qin Hong
- Key Laboratory for Advanced Materials & School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai 200237, P. R. China
| | - Ze-Rui Zhou
- Key Laboratory for Advanced Materials & School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai 200237, P. R. China
| | - Da-Wei Li
- Key Laboratory for Advanced Materials & School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai 200237, P. R. China
| | - Ruo-Can Qian
- Key Laboratory for Advanced Materials & School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai 200237, P. R. China
| |
Collapse
|
19
|
Phyo P, Zhao X, Templeton AC, Xu W, Cheung JK, Su Y. Understanding molecular mechanisms of biologics drug delivery and stability from NMR spectroscopy. Adv Drug Deliv Rev 2021; 174:1-29. [PMID: 33609600 DOI: 10.1016/j.addr.2021.02.007] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 01/20/2021] [Accepted: 02/07/2021] [Indexed: 02/06/2023]
Abstract
Protein therapeutics carry inherent limitations of membrane impermeability and structural instability, despite their predominant role in the modern pharmaceutical market. Effective formulations are needed to overcome physiological and physicochemical barriers, respectively, for improving bioavailability and stability. Knowledge of membrane affinity, cellular internalization, encapsulation, and release of drug-loaded carrier vehicles uncover the structural basis for designing and optimizing biopharmaceuticals with enhanced delivery efficiency and therapeutic efficacy. Understanding stabilizing and destabilizing interactions between protein drugs and formulation excipients provide fundamental mechanisms for ensuring the stability and quality of biological products. This article reviews the molecular studies of biologics using solution and solid-state NMR spectroscopy on structural attributes pivotal to drug delivery and stability. In-depth investigation of the structure-function relationship of drug delivery systems based on cell-penetrating peptides, lipid nanoparticles and polymeric colloidal, and biophysical and biochemical stability of peptide, protein, monoclonal antibody, and vaccine, as the integrative efforts on drug product design, will be elaborated.
Collapse
Affiliation(s)
- Pyae Phyo
- Pharmaceutical Sciences, Merck & Co., Inc., Kenilworth, NJ 07033, United States
| | - Xi Zhao
- Pharmaceutical Sciences, Merck & Co., Inc., Kenilworth, NJ 07033, United States
| | - Allen C Templeton
- Pharmaceutical Sciences, Merck & Co., Inc., Kenilworth, NJ 07033, United States
| | - Wei Xu
- Pharmaceutical Sciences, Merck & Co., Inc., Kenilworth, NJ 07033, United States
| | - Jason K Cheung
- Pharmaceutical Sciences, Merck & Co., Inc., Kenilworth, NJ 07033, United States
| | - Yongchao Su
- Pharmaceutical Sciences, Merck & Co., Inc., Kenilworth, NJ 07033, United States.
| |
Collapse
|
20
|
Yablonskii S, Bodnarchuk V, Geivandov A, Romero-Hasler P, Soto-Bustamante E, Morales J. Dember photovoltaic effect as method for structural characterization of phospholipidic membranes. J Mol Liq 2021. [DOI: 10.1016/j.molliq.2021.115409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
21
|
Matamoros-Recio A, Franco-Gonzalez JF, Forgione RE, Torres-Mozas A, Silipo A, Martín-Santamaría S. Understanding the Antibacterial Resistance: Computational Explorations in Bacterial Membranes. ACS OMEGA 2021; 6:6041-6054. [PMID: 33718695 PMCID: PMC7948216 DOI: 10.1021/acsomega.0c05590] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 02/09/2021] [Indexed: 05/05/2023]
Affiliation(s)
- Alejandra Matamoros-Recio
- Department of Structural and Chemical Biology, Centro de Investigaciones Biológicas Research Margarita Salas, CIB-CSIC, C/Ramiro de Maeztu, 9, 28040 Madrid, Spain
| | - Juan Felipe Franco-Gonzalez
- Department of Structural and Chemical Biology, Centro de Investigaciones Biológicas Research Margarita Salas, CIB-CSIC, C/Ramiro de Maeztu, 9, 28040 Madrid, Spain
| | - Rosa Ester Forgione
- Department of Structural and Chemical Biology, Centro de Investigaciones Biológicas Research Margarita Salas, CIB-CSIC, C/Ramiro de Maeztu, 9, 28040 Madrid, Spain
- Dipartimento di Scienze Chimiche, Complesso Universitario Monte Sant’Angelo, Università di Napoli Federico II, Via Cintia 4, 80126 Napoli, Italy
| | - Angel Torres-Mozas
- Department of Structural and Chemical Biology, Centro de Investigaciones Biológicas Research Margarita Salas, CIB-CSIC, C/Ramiro de Maeztu, 9, 28040 Madrid, Spain
| | - Alba Silipo
- Dipartimento di Scienze Chimiche, Complesso Universitario Monte Sant’Angelo, Università di Napoli Federico II, Via Cintia 4, 80126 Napoli, Italy
| | - Sonsoles Martín-Santamaría
- Department of Structural and Chemical Biology, Centro de Investigaciones Biológicas Research Margarita Salas, CIB-CSIC, C/Ramiro de Maeztu, 9, 28040 Madrid, Spain
| |
Collapse
|
22
|
Fernandes E, Benfeito S, Cagide F, Gonçalves H, Bernstorff S, Nieder JB, Cd Real Oliveira ME, Borges F, Lúcio M. Lipid Nanosystems and Serum Protein as Biomimetic Interfaces: Predicting the Biodistribution of a Caffeic Acid-Based Antioxidant. Nanotechnol Sci Appl 2021; 14:7-27. [PMID: 33603350 PMCID: PMC7882595 DOI: 10.2147/nsa.s289355] [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: 10/29/2020] [Accepted: 01/16/2021] [Indexed: 11/30/2022] Open
Abstract
Purpose AntiOxCIN3 is a novel mitochondriotropic antioxidant developed to minimize the effects of oxidative stress on neurodegenerative diseases. Prior to an investment in pre-clinical in vivo studies, it is important to apply in silico and biophysical cell-free in vitro studies to predict AntiOxCIN3 biodistribution profile, respecting the need to preserve animal health in accordance with the EU principles (Directive 2010/63/EU). Accordingly, we propose an innovative toolbox of biophysical studies and mimetic models of biological interfaces, such as nanosystems with different compositions mimicking distinct membrane barriers and human serum albumin (HSA). Methods Intestinal and cell membrane permeation of AntiOxCIN3 was predicted using derivative spectrophotometry. AntiOxCIN3 –HSA binding was evaluated by intrinsic fluorescence quenching, synchronous fluorescence, and dynamic/electrophoretic light scattering. Steady-state and time-resolved fluorescence quenching was used to predict AntiOxCIN3-membrane orientation. Fluorescence anisotropy, synchrotron small- and wide-angle X-ray scattering were used to predict lipid membrane biophysical impairment caused by AntiOxCIN3 distribution. Results and Discussion We found that AntiOxCIN3 has the potential to permeate the gastrointestinal tract. However, its biodistribution and elimination from the body might be affected by its affinity to HSA (>90%) and by its steady-state volume of distribution (VDSS=1.89± 0.48 L∙Kg−1). AntiOxCIN3 is expected to locate parallel to the membrane phospholipids, causing a bilayer stiffness effect. AntiOxCIN3 is also predicted to permeate through blood-brain barrier and reach its therapeutic target – the brain. Conclusion Drug interactions with biological interfaces may be evaluated using membrane model systems and serum proteins. This knowledge is important for the characterization of drug partitioning, positioning and orientation of drugs in membranes, their effect on membrane biophysical properties and the study of serum protein binding. The analysis of these interactions makes it possible to collect valuable knowledge on the transport, distribution, accumulation and, eventually, therapeutic impact of drugs which may aid the drug development process.
Collapse
Affiliation(s)
- Eduarda Fernandes
- Departamento de Física da Universidade do Minho, CF-UM-UP, Centro de Física das Universidades do Minho e Porto, Campus de Gualtar, Braga, 4710-057, Portugal.,Ultrafast Bio- and Nanophotonics Group, INL - International Iberian Nanotechnology Laboratory, Braga, Portugal
| | - Sofia Benfeito
- CIQUP/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto, Portugal
| | - Fernando Cagide
- CIQUP/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto, Portugal
| | | | - Sigrid Bernstorff
- Elettra-Sincrotrone Trieste S. C.p.A.,, Basovizza, Trieste, I-34149, Italy
| | - Jana B Nieder
- Ultrafast Bio- and Nanophotonics Group, INL - International Iberian Nanotechnology Laboratory, Braga, Portugal
| | - M Elisabete Cd Real Oliveira
- Departamento de Física da Universidade do Minho, CF-UM-UP, Centro de Física das Universidades do Minho e Porto, Campus de Gualtar, Braga, 4710-057, Portugal
| | - Fernanda Borges
- CIQUP/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto, Portugal
| | - Marlene Lúcio
- Departamento de Física da Universidade do Minho, CF-UM-UP, Centro de Física das Universidades do Minho e Porto, Campus de Gualtar, Braga, 4710-057, Portugal.,CBMA, Centro de Biologia Molecular e Ambiental, Departamento de Biologia, Universidade do Minho, Campus de Gualtar, Braga 4710-057, Portugal
| |
Collapse
|
23
|
Dai X, Ji Y, Wang Z, He L, Wang X, Li S. Interaction between Bottlebrush Polymers and Phospholipid Membranes in Solutions. Polymers (Basel) 2020; 12:E3033. [PMID: 33348889 PMCID: PMC7766109 DOI: 10.3390/polym12123033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 12/04/2020] [Accepted: 12/15/2020] [Indexed: 11/23/2022] Open
Abstract
In this work, the interactions between bottlebrush polymers and phospholipid membranes were investigated using dissipative particle dynamics simulations. The weak and strong adsorption phenomena between the polymers and membranes were examined by calculating the system parameters. A spring model was introduced to explain the variances in the shape factors and the radius of gyration of the bottlebrush polymers, as well as the order parameters of the phospholipid membrane in the pulling processes. This work provides further understanding for the application of bottlebrush polymers in biological processes.
Collapse
Affiliation(s)
| | | | - Zhenguo Wang
- Department of Physics, Wenzhou University, Wenzhou 325035, China; (X.D.); (Y.J.); (L.H.); (X.W.)
| | | | | | - Shiben Li
- Department of Physics, Wenzhou University, Wenzhou 325035, China; (X.D.); (Y.J.); (L.H.); (X.W.)
| |
Collapse
|