1
|
Chen Y, Xu J, Li P, Shi L, Zhang S, Guo Q, Yang Y. Advances in the use of local anesthetic extended-release systems in pain management. Drug Deliv 2024; 31:2296349. [PMID: 38130151 PMCID: PMC10763865 DOI: 10.1080/10717544.2023.2296349] [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: 07/13/2023] [Accepted: 11/15/2023] [Indexed: 12/23/2023] Open
Abstract
Pain management remains among the most common and largely unmet clinical problems today. Local anesthetics play an indispensable role in pain management. The main limitation of traditional local anesthetics is the limited duration of a single injection. To address this problem, catheters are often placed or combined with other drugs in clinical practice to increase the time that local anesthetics act. However, this method does not meet the needs of clinical analgesics. Therefore, many researchers have worked to develop local anesthetic extended-release types that can be administered in a single dose. In recent years, drug extended-release systems have emerged dramatically due to their long duration and efficacy, providing more possibilities for the application of local anesthetics. This paper summarizes the types of local anesthetic drug delivery systems and their clinical applications, discusses them in the context of relevant studies on local anesthetics, and provides a summary and outlook on the development of local anesthetic extended-release agents.
Collapse
Affiliation(s)
- Yulu Chen
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China
| | - Jingmei Xu
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China
| | - Ping Li
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Obstetrics, Xiangya Hospital, Central South University, Changsha, China
| | - Liyang Shi
- College of Biology, Hunan University, Changsha, China
| | - Sha Zhang
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China
| | - Qulian Guo
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yong Yang
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
2
|
Martinez J, Ingram N, Kapur N, Jayne DG, Beales PA. Vesicle-based formulations for pain treatment: a narrative review. Pain Rep 2024; 9:e1196. [PMID: 39399306 PMCID: PMC11469894 DOI: 10.1097/pr9.0000000000001196] [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: 05/10/2024] [Revised: 07/29/2024] [Accepted: 08/14/2024] [Indexed: 10/15/2024] Open
Abstract
Pain, a complex and debilitating condition, necessitates innovative therapeutic strategies to alleviate suffering and enhance patients' quality of life. Vesicular systems hold the potential to enhance precision of drug localisation and release, prolong the duration of therapeutic action and mitigate adverse events associated with long-term pharmacotherapy. This review critically assesses the current state-of-the-art in vesicle-based formulations (liposomes, polymersomes, ethosomes, and niosomes) for pain management applications. We highlight formulation engineering strategies used to optimise drug pharmacokinetics, present preclinical findings of experimental delivery systems, and discuss the clinical evidence for the benefits of clinically approved formulations. We present the challenges and outlook for future improvements in long-acting anaesthetic and analgesic formulation development.
Collapse
Affiliation(s)
- Juan Martinez
- School of Chemistry, University of Leeds, Leeds, West Yorkshire, United Kingdom
- Leeds Institute for Medical Research, University of Leeds, Leeds, West Yorkshire, United Kingdom
| | - Nicola Ingram
- Leeds Institute for Medical Research, University of Leeds, Leeds, West Yorkshire, United Kingdom
| | - Nikil Kapur
- School of Mechanical Engineering, University of Leeds, Leeds, West Yorkshire, United Kingdom
| | - David G. Jayne
- Leeds Institute for Medical Research, University of Leeds, Leeds, West Yorkshire, United Kingdom
- The John Goligher Colorectal Surgery Unit, St. James's University Hospital, Leeds Teaching Hospital Trust, Leeds, West Yorkshire, United Kingdom
| | - Paul A. Beales
- School of Chemistry, University of Leeds, Leeds, West Yorkshire, United Kingdom
| |
Collapse
|
3
|
Cetin FN, Mignon A, Van Vlierberghe S, Kolouchova K. Polymer- and Lipid-Based Nanostructures Serving Wound Healing Applications: A Review. Adv Healthc Mater 2024:e2402699. [PMID: 39543796 DOI: 10.1002/adhm.202402699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 10/18/2024] [Indexed: 11/17/2024]
Abstract
Management of hard-to-heal wounds often requires specialized care that surpasses the capabilities of conventional treatments. Even the most advanced commercial products lack the functionality to meet the needs of hard-to-heal wounds, especially those complicated by active infection, extreme bleeding, and chronic inflammation. The review explores how supramolecular nanovesicles and nanoparticles-such as dendrimers, micelles, polymersomes, and lipid-based nanocarriers-can be key to introducing advanced wound healing and monitoring properties to address the complex needs of hard-to-heal wounds. Their potential to enable advanced functions essential for next-generation wound healing products-such as hemostatic functions, transdermal penetration, macrophage polarization, targeted delivery, and controlled release of active pharmaceutical ingredients (antibiotics, gaseous products, anti-inflammatory drugs, growth factors)-is discussed via an extensive overview of the recent reports. These studies highlight that the integration of supramolecular systems in wound care is crucial for advancing toward a new generation of wound healing products and addressing significant gaps in current wound management practices. Current strategies and potential improvements regarding personalized therapies, transdermal delivery, and the promising critically evaluated but underexplored polymer-based nanovesicles, including polymersomes and proteinosomes, for wound healing.
Collapse
Affiliation(s)
- Fatma N Cetin
- Department of Organic and Macromolecular Chemistry, Ghent University, Krijgslaan 281, Gent, 9000, Belgium
| | - Arn Mignon
- Department of Engineering Technology, KU Leuven, Andreas Vesaliusstraat 13, Leuven, 3000, Belgium
| | - Sandra Van Vlierberghe
- Department of Organic and Macromolecular Chemistry, Ghent University, Krijgslaan 281, Gent, 9000, Belgium
| | - Kristyna Kolouchova
- Department of Organic and Macromolecular Chemistry, Ghent University, Krijgslaan 281, Gent, 9000, Belgium
| |
Collapse
|
4
|
Liu J, Liao ZH, Zhu T, Wu J, Wang F. In Situ Self-Inflating-Modeled Giant-Vesicle-Like Quantum Dot Assembly for Biomimetic Artificial Photosynthesis. ACS NANO 2024; 18:31537-31546. [PMID: 39475626 DOI: 10.1021/acsnano.4c12728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2024]
Abstract
The study of biomimetic self-assembly is crucial for scientists aiming to understand the origin of life and construct biomimetic functional structures. In our endeavor to create a biomimetic photosynthetic assembly, we discover a self-inflation behavior that drives the components, MPA-CdSe quantum dots (QDs) and a solid cationic polyelectrolyte, CPPA, to form a giant-vesicle-like (GVL) architecture, termed GVL-QDs@CPPA. The in situ generation of osmotic pressure during the self-assembly of QDs onto swollen CPPA in water was found to cause this self-inflation process. The resulting vesicle-like structure exhibits spatial characteristics similar to those of natural photosynthetic cells, with QDs acting as pigments uniformly distributed on the CPPA membranes, which have embedded cobalt catalytic centers. This architecture ensures optimal absorption of visible light and facilitates efficient electron transfer between the QDs and catalytic centers. As a result, GVL-QDs@CPPA assemblies efficiently harness photogenerated electrons and holes to convert protons and isopropanol into hydrogen (H2) and acetone, respectively, achieving a nearly 1:1 ratio of the reduction product (H2) to the oxidation product (acetone).
Collapse
Affiliation(s)
- Jing Liu
- Key Laboratory of Materials Chemistry for Energy Conversion and Storage (Huazhong University of Science and Technology) of Ministry of Education, Hubei Key Laboratory of Material Chemistry and Service Failure, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, P. R. China
| | - Zi-Hao Liao
- Key Laboratory of Materials Chemistry for Energy Conversion and Storage (Huazhong University of Science and Technology) of Ministry of Education, Hubei Key Laboratory of Material Chemistry and Service Failure, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, P. R. China
| | - Ting Zhu
- Key Laboratory of Materials Chemistry for Energy Conversion and Storage (Huazhong University of Science and Technology) of Ministry of Education, Hubei Key Laboratory of Material Chemistry and Service Failure, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, P. R. China
| | - Jin Wu
- Key Laboratory of Materials Chemistry for Energy Conversion and Storage (Huazhong University of Science and Technology) of Ministry of Education, Hubei Key Laboratory of Material Chemistry and Service Failure, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, P. R. China
| | - Feng Wang
- Key Laboratory of Materials Chemistry for Energy Conversion and Storage (Huazhong University of Science and Technology) of Ministry of Education, Hubei Key Laboratory of Material Chemistry and Service Failure, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, P. R. China
- Guangdong Provincial Key Laboratory of Manufacturing Equipment Digitization (2023B1212060012), Guangdong HUST (Huazhong University of Science and Technology) Industrial Technology Research Institute, Dongguan 523808, P. R. China
| |
Collapse
|
5
|
Feng S, Zhang Y, Wang Y, Gao Y, Song Y. Harnessing Gene Editing Technology for Tumor Microenvironment Modulation: An Emerging Anticancer Strategy. Chemistry 2024; 30:e202402485. [PMID: 39225329 DOI: 10.1002/chem.202402485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 08/30/2024] [Accepted: 09/02/2024] [Indexed: 09/04/2024]
Abstract
Cancer is a multifaceted disease influenced by both intrinsic cellular traits and extrinsic factors, with the tumor microenvironment (TME) being crucial for cancer progression. To satisfy their high proliferation and aggressiveness, cancer cells always plunder large amounts of nutrients and release various signals to their surroundings, forming a dynamic TME with special metabolic, immune, microbial and physical characteristics. Due to the neglect of interactions between tumor cells and the TME, traditional cancer therapies often struggle with challenges such as drug resistance, low efficacy, and recurrence. Importantly, the development of gene editing technologies, particularly the CRISPR-Cas system, offers promising new strategies for cancer treatment. Combined with nanomaterial strategies, CRISPR-Cas technology exhibits precision, affordability, and user-friendliness with reduced side effects, which holds great promise for profoundly altering the TME at the genetic level, potentially leading to lasting anticancer outcomes. This review will delve into how CRISPR-Cas can be leveraged to manipulate the TME, examining its potential as a transformative anticancer therapy.
Collapse
Affiliation(s)
- Shujun Feng
- College of Engineering and Applied Sciences, Nanjing University, 210023, Nanjing, China
| | - Yu Zhang
- School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, 210094, Nanjing, China
| | - Yanyi Wang
- College of Engineering and Applied Sciences, Nanjing University, 210023, Nanjing, China
| | - Yanfeng Gao
- School of Medical Imaging, Wannan Medical College, 241002, Wuhu, China
| | - Yujun Song
- College of Engineering and Applied Sciences, Nanjing University, 210023, Nanjing, China
| |
Collapse
|
6
|
Ventura-Cobos J, Climent E, Martínez-Máñez R, Llopis-Lorente A. Chemical Communication between Giant Vesicles and Gated Nanoparticles for Strip-Based Sensing. NANO LETTERS 2024; 24:14050-14057. [PMID: 39442006 PMCID: PMC11544697 DOI: 10.1021/acs.nanolett.4c04022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/18/2024] [Accepted: 10/18/2024] [Indexed: 10/25/2024]
Abstract
Inspired by nature, the development of artificial micro/nanosystems capable of communicating has become an emergent topic in nanotechnology, synthetic biology, and related areas. However, the demonstration of actual applications still has to come. Here, we demonstrate how chemical communication between micro- and nanoparticles can be used for the design of sensing systems. To realize this, we synergistically combine two different types of particles: i.e., giant unilamellar vesicles (GUVs) as senders and gated mesoporous nanoparticles as receivers. The use of engineered GUVs allows the detection of analytes based on responsive membranes, while the use of gated nanoparticles allows a straightforward application on test strips with smartphone-based detection. In addition, we demonstrate that the combined communication system exhibits signal amplification and its application in real samples employing the bacterial toxin α-hemolysin as target analyte. Altogether, our report presents a new route for engineering sensing systems based on the combination of communicative micro/nanoparticles.
Collapse
Affiliation(s)
- Jordi Ventura-Cobos
- Instituto
Interuniversitario de Investigación de Reconocimiento Molecular
y Desarrollo Tecnológico (IDM), Universitat
Politècnica de València, Universitat de València, Camino de Vera s/n, 46022 València, Spain
| | - Estela Climent
- Instituto
Interuniversitario de Investigación de Reconocimiento Molecular
y Desarrollo Tecnológico (IDM), Universitat
Politècnica de València, Universitat de València, Camino de Vera s/n, 46022 València, Spain
- CIBER
de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN),
Instituto de Salud Carlos III, 28029 Madrid, Spain
- Unidad
Mixta de Investigación en Nanomedicina y Sensores, Universitat Politècnica de València,
Instituto de Investigación Sanitaria La Fe (IISLAFE), Avenida Fernando Abril Martorell
106, 46026 Valencia, Spain
| | - Ramón Martínez-Máñez
- Instituto
Interuniversitario de Investigación de Reconocimiento Molecular
y Desarrollo Tecnológico (IDM), Universitat
Politècnica de València, Universitat de València, Camino de Vera s/n, 46022 València, Spain
- CIBER
de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN),
Instituto de Salud Carlos III, 28029 Madrid, Spain
- Unidad
Mixta de Investigación en Nanomedicina y Sensores, Universitat Politècnica de València,
Instituto de Investigación Sanitaria La Fe (IISLAFE), Avenida Fernando Abril Martorell
106, 46026 Valencia, Spain
- Unidad
Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades
y Nanomedicina, Universitat Politècnica
de València, Centro de Investigación Príncipe
Felipe, C/Eduardo Primo
Yúfera 3, 46100 Valencia, Spain
- Departamento
de Química, Universitat Politècnica
de València, Camino
de Vera s/n, 46022 Valencia, Spain
| | - Antoni Llopis-Lorente
- Instituto
Interuniversitario de Investigación de Reconocimiento Molecular
y Desarrollo Tecnológico (IDM), Universitat
Politècnica de València, Universitat de València, Camino de Vera s/n, 46022 València, Spain
- CIBER
de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN),
Instituto de Salud Carlos III, 28029 Madrid, Spain
- Departamento
de Química, Universitat Politècnica
de València, Camino
de Vera s/n, 46022 Valencia, Spain
| |
Collapse
|
7
|
Marinho A, Reis S, Nunes C. On the design of cell membrane-coated nanoparticles to treat inflammatory conditions. NANOSCALE HORIZONS 2024. [PMID: 39499543 DOI: 10.1039/d4nh00457d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2024]
Abstract
Biomimetic-based drug delivery systems (DDS) attempt to recreate the complex interactions that occur naturally between cells. Cell membrane-coated nanoparticles (CMCNPs) have been one of the main strategies in this area to prevent opsonization and clearance. Moreover, coating nanoparticles with cell membranes allows them to acquire functions and properties inherent to the mother cells. In particular, cells from bloodstream show to have specific advantages depending on the cell type to be used for that application, specifically in cases of chronic inflammation. Thus, this review focuses on the biomimetic strategies that use membranes from blood cells to target and treat inflammatory conditions.
Collapse
Affiliation(s)
- Andreia Marinho
- LAQV, REQUIMTE, Faculdade de Farmácia, Universidade do Porto, R. Jorge de Viterbo Ferreira 228, 4500-313 Porto, Portugal.
- LAQV, REQUIMTE, Faculdade de Ciências, Universidade do Porto, R. Campo Alegre s/n, 4169-007 Porto, Portugal
| | - Salette Reis
- LAQV, REQUIMTE, Faculdade de Farmácia, Universidade do Porto, R. Jorge de Viterbo Ferreira 228, 4500-313 Porto, Portugal.
| | - Cláudia Nunes
- LAQV, REQUIMTE, Faculdade de Farmácia, Universidade do Porto, R. Jorge de Viterbo Ferreira 228, 4500-313 Porto, Portugal.
- LAQV, REQUIMTE, Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, R. Jorge de Viterbo Ferreira 228, 4500-313 Porto, Portugal
| |
Collapse
|
8
|
Heuberger L, Korpidou M, Guinart A, Doellerer D, López DM, Schoenenberger CA, Milinkovic D, Lörtscher E, Feringa BL, Palivan CG. Photoreceptor-Like Signal Transduction Between Polymer-Based Protocells. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024:e2413981. [PMID: 39491508 DOI: 10.1002/adma.202413981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 10/14/2024] [Indexed: 11/05/2024]
Abstract
Deciphering inter- and intracellular signaling pathways is pivotal for understanding the intricate communication networks that orchestrate life's dynamics. Communication models involving bottom-up construction of protocells are emerging but often lack specialized compartments sufficiently robust and hierarchically organized to perform spatiotemporally defined signaling. Here, the modular construction of communicating polymer-based protocells designed to mimic the transduction of information in retinal photoreceptors is presented. Microfluidics is used to generate polymeric protocells subcompartmentalized by specialized artificial organelles. In one protocell population, light triggers artificial organelles with membrane-embedded photoresponsive rotary molecular motors to set off a sequence of reactions starting with the release of encapsulated signaling molecules into the lumen. Intercellular communication is mediated by signal transfer across membranes to protocells containing catalytic artificial organelles as subcompartments, whose signal conversion can be modulated by environmental calcium. Signal propagation also requires selective permeability of the diverse compartments. By segregating artificial organelles in distinct protocells, a sequential chain of reactions mediating intercellular communication is created that is further modulated by adding extracellular messengers. This connective behavior offers the potential for a deeper understanding of signaling pathways and faster integration of proto- and living cells, with the unique advantage of controlling each step by bio-relevant signals.
Collapse
Affiliation(s)
- Lukas Heuberger
- Department of Chemistry, University of Basel, Basel, 4002, Switzerland
| | - Maria Korpidou
- Department of Chemistry, University of Basel, Basel, 4002, Switzerland
| | - Ainoa Guinart
- Faculty of Science and Engineering, Stratingh Institute for Chemistry, University of Groningen, AG Groningen, 9747, The Netherlands
| | - Daniel Doellerer
- Faculty of Science and Engineering, Stratingh Institute for Chemistry, University of Groningen, AG Groningen, 9747, The Netherlands
| | | | | | - Daela Milinkovic
- Department of Chemistry, University of Basel, Basel, 4002, Switzerland
| | - Emanuel Lörtscher
- IBM Research Europe-Zürich, Säumerstrasse 4, Rüschlikon, 8803, Switzerland
- NCCR - Molecular Systems Engineering, Mattenstrasse 22, Basel, 4002, Switzerland
| | - Ben L Feringa
- Faculty of Science and Engineering, Stratingh Institute for Chemistry, University of Groningen, AG Groningen, 9747, The Netherlands
| | - Cornelia G Palivan
- Department of Chemistry, University of Basel, Basel, 4002, Switzerland
- NCCR - Molecular Systems Engineering, Mattenstrasse 22, Basel, 4002, Switzerland
- Swiss Nanoscience Institute (SNI), University of Basel, Klingelbergstrasse 80, Basel, 4056, Switzerland
| |
Collapse
|
9
|
Westensee IN, de Dios Andres P, Brodszkij E, Descours PL, Perez-Rodriguez D, Spinazzola A, Mookerjee RP, Städler B. Engineered Lipids for Intracellular Reactive Oxygen Species Scavenging in Steatotic Hepatocytes. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2400816. [PMID: 38949047 DOI: 10.1002/smll.202400816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 05/20/2024] [Indexed: 07/02/2024]
Abstract
Intracellular reactive oxygen species (ROS) in steatotic cells pose a problem due to their potential to cause oxidative stress and cellular damage. Delivering engineered phospholipids to intracellular lipid droplets in steatotic hepatic cells, using the cell's inherent intracellular lipid transport mechanisms are investigated. Initially, it is shown that tail-labeled fluorescent lipids assembled into liposomes are able to be transported to intracellular lipid droplets in steatotic HepG2 cells and HHL-5 cells. Further, an antioxidant, an EUK salen-manganese derivative, which has superoxide dismutase-like and catalase-like activity, is covalently conjugated to the tail of a phospholipid and formulated as liposomes for administration. Steatotic HepG2 cells and HHL-5 cells incubated with these antioxidant liposomes have lower intracellular ROS levels compared to untreated controls and non-covalently formulated antioxidants. This first proof-of-concept study illustrates an alternative strategy to equip native organelles in mammalian cells with engineered enzyme activity.
Collapse
Affiliation(s)
- Isabella N Westensee
- Interdisciplinarly Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, Aarhus, 8000, Denmark
| | - Paula de Dios Andres
- Interdisciplinarly Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, Aarhus, 8000, Denmark
| | - Edit Brodszkij
- Interdisciplinarly Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, Aarhus, 8000, Denmark
| | - Pierre-Louis Descours
- Interdisciplinarly Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, Aarhus, 8000, Denmark
| | - Diego Perez-Rodriguez
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, Royal Free Campus, London, NW3 2PF, UK
| | - Antonella Spinazzola
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, Royal Free Campus, London, NW3 2PF, UK
| | - Rajeshwar Prosad Mookerjee
- Institute for Liver and Digestive Health, University College London, Royal Free Campus, Rowland Hill Street, Hampstead, London, NW3 2PF, UK
| | - Brigitte Städler
- Interdisciplinarly Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, Aarhus, 8000, Denmark
| |
Collapse
|
10
|
Lin M, Lv X, Wang H, Shu L, Wang H, Zhang G, Sun J, Chen X. Coacervation-Driven Semipermeable Nanoreactors for Enzymatic Cascade-Mediated Cancer Combination Therapy with Enhanced Efficacy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2407378. [PMID: 39235373 DOI: 10.1002/adma.202407378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 08/13/2024] [Indexed: 09/06/2024]
Abstract
Utilizing enzyme cascades as a promising approach for targeted cancer therapies holds significant potential, yet its clinical effectiveness is substantially hindered by functional losses during delivery. Complex coacervation emerges as an intriguing strategy for designing functional nanoreactors. In this study, a noteworthy achievement is presented in the development of lactobionic acid-modified tumor microenvironment (TME)-responsive polyelectrolyte complex vesicles (HGS-PCVs) based on bioinspired homopolypeptoids, which serve as a facile, intelligent, and highly efficient nanoreactor tunable for glucose oxidase, hemoglobin, and sorafenib (SRF) to hepatic cancer cells. The TME-responsive permeability of HGS-PCVs enables the selective entry of glucose into their interior, triggering an enzymatic cascade reaction within the tumor. This intricate process generates toxic hydroxyl radicals while concurrently lowering the pH. Consequently, this pH shift enhances the SRF release, effectively promoting ferroptosis and apoptosis in the target cancer cells. Further, the administration of the HGS-PCVs not only initiates immunogenic cell death but also plays a crucial role in inducing the maturation of dendritic cells within lymph nodes. It stimulates an adaptive T-cell response, a crucial mechanism that contributes to impeding the growth of distant tumors in vivo, demonstrating the promising potential of PCVs for cancer immunotherapy.
Collapse
Affiliation(s)
- Min Lin
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, Jilin, 130012, P. R. China
| | - Xueli Lv
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, Jilin, 130012, P. R. China
| | - Hepeng Wang
- China-Japan Union Hospital of Jilin University, Changchun, Jilin, 130033, P. R. China
| | - Lilei Shu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, Jilin, 130012, P. R. China
| | - Helin Wang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, Jilin, 130012, P. R. China
| | - Guojing Zhang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, Jilin, 130012, P. R. China
| | - Jing Sun
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, Jilin, 130012, P. R. China
| | - Xuesi Chen
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, Jilin, 130012, P. R. China
| |
Collapse
|
11
|
Han H, Santos HA. Nano- and Micro-Platforms in Therapeutic Proteins Delivery for Cancer Therapy: Materials and Strategies. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2409522. [PMID: 39263818 DOI: 10.1002/adma.202409522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 08/26/2024] [Indexed: 09/13/2024]
Abstract
Proteins have emerged as promising therapeutics in oncology due to their great specificity. Many treatment strategies are developed based on protein biologics, such as immunotherapy, starvation therapy, and pro-apoptosis therapy, while some protein biologics have entered the clinics. However, clinical translation is severely impeded by instability, short circulation time, poor transmembrane transportation, and immunogenicity. Micro- and nano-particles-based drug delivery platforms are designed to solve those problems and enhance protein therapeutic efficacy. This review first summarizes the different types of therapeutic proteins in clinical and research stages, highlighting their administration limitations. Next, various types of micro- and nano-particles are described to demonstrate how they can overcome those limitations. The potential of micro- and nano-particles are then explored to enhance the therapeutic efficacy of proteins by combinational therapies. Finally, the challenges and future directions of protein biologics carriers are discussed for optimized protein delivery.
Collapse
Affiliation(s)
- Huijie Han
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen (UMCG), University of Groningen, Ant. Deusinglaan 1, Groningen, 9713 AV, The Netherlands
| | - Hélder A Santos
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen (UMCG), University of Groningen, Ant. Deusinglaan 1, Groningen, 9713 AV, The Netherlands
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland
| |
Collapse
|
12
|
Shi Y, Landfester K, Morris SM. Fine-Tuning the Microstructure and Photophysical Characteristics of Fluorescent Conjugated Copolymers Using Photoalignment and Liquid-Crystal Ordering. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2407117. [PMID: 39206683 PMCID: PMC11538637 DOI: 10.1002/advs.202407117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 07/29/2024] [Indexed: 09/04/2024]
Abstract
Replicating the microstructural basis and the near 100% excitation energy transfer efficiency in naturally occurring light-harvesting complexes (LHCs) remains challenging in synthetic energy-harvesting devices. Biological photosynthesis regulates active ensembles of light-absorbing and funneling chlorophylls in proteins in response to fluctuating sunlight. Here, use of long-range liquid crystal (LC) ordering to tailor chain orientation and packing structure in liquid crystalline conjugated polymer (LCCP) layers for bio-mimicry of certain structural basis and light-harvesting properties of LHCs is reported. It is found that long-range orientational ordering in an LC phase of poly(9,9-dioctylfluorene-co-benzothiadiazole) (F8BT) copolymer stabilizes a small fraction of randomly-oriented F8BT nanocrystals dispersed in an amorphous matrix of F8BT chains, resembling a self-doped host-guest system whereby excitation energy funneling and photoluminescence quantum efficiencies are enhanced significantly by triggering 3D donor-to-acceptor Förster resonance energy transfer (FRET) and dominant intrachain emission in the nano-crystal acceptor. Further, photoalignment of nematic F8BT layers is combined with LC orientational ordering to fabricate large-area-extended monodomains exhibiting >60% crystallinity and ≈20 nm-long interchain packing order. Remarkably, these monodomains demonstrate strong linearly polarized emission, whilst also promoting a new band-edge absorption species and an extra emissive interchain excited state as compared to the non-aligned films.
Collapse
Affiliation(s)
- Yuping Shi
- Max Planck Institute for Polymer ResearchAckermannweg 1055128MainzGermany
- Department of Engineering ScienceUniversity of OxfordParks RoadOxfordOX1 3PJUK
| | | | - Stephen M. Morris
- Department of Engineering ScienceUniversity of OxfordParks RoadOxfordOX1 3PJUK
| |
Collapse
|
13
|
Banerjee T, Dan K, Ghosh S. pH-Responsive self-assembled polymer-photosensitizer conjugate for activable photodynamic therapy. NANOSCALE 2024; 16:19756-19762. [PMID: 39373067 DOI: 10.1039/d4nr03249g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
This paper reports synthesis, aqueous self-assembly and relevance of the pH-triggered activable photodynamic therapy of amphiphilic polyurethane (P1S) functionalized with a heavy-atom free organic photosensitizer. Condensation polymerization between 1,4-diisocyanatobutane and two different dihydroxy monomers (one having a pendant hydrophilic wedge and the other having 1,3-dihydroxypropan-2-one with a reactive carbonyl group) in the presence of a measured amount of (S)-2-methylbutan-1-ol (chain-stopper) and DABCO catalyst produces a reactive pre-polymer P1. Hydrazide-functionalized thionated-naphthalenemonoimide (NMIS), which acts as a photosensitizer, reacted with the carbonyl-functionality of P1 to obtain the desired polymer-photosensitizer conjugate P1S in which the dye was attached to the polymer backbone via an acid-labile hydrazone linker. In water, P1S adopted an intra-chain H-bonding stabilized folded structure, which further assembled to produce a polymersome structure (Dh ≈ 200 nm), in which the hydrophobic membrane consists of aggregated NMIS and trialkoxy-benzene chromophores, as evident from UV/vis, CD and small-angle X-ray diffraction studies. In the aggregated state, NMIS loses its reactive oxygen species (ROS) generation ability and remains in a dormant state. However, under acidic conditions (pH 5.5), the photosensitizer is detached (presumably because of the cleavage of the hydrazone linker) and regains its full ROS-generation activity under photoirradiation, as evidenced from the standard DCFH assay. To test the possibility of such pH-activable intra-cellular ROS generation, P1S was treated with HeLa cells, as it is known that cancer cells are more acidic than normal cells. Indeed, photoirradiation-induced intra-cellular ROS generation was evident by the DCFH assay, resulting in efficient cell killing.
Collapse
Affiliation(s)
- Tanushri Banerjee
- School of Applied and Interdisciplinary Sciences, Indian Association for the Cultivation of Science, 2A and 2B Raja S. C. Mullick Road, Kolkata, 700032, India.
| | - Krishna Dan
- School of Applied and Interdisciplinary Sciences, Indian Association for the Cultivation of Science, 2A and 2B Raja S. C. Mullick Road, Kolkata, 700032, India.
| | - Suhrit Ghosh
- School of Applied and Interdisciplinary Sciences, Indian Association for the Cultivation of Science, 2A and 2B Raja S. C. Mullick Road, Kolkata, 700032, India.
| |
Collapse
|
14
|
Faizi HA, Granek R, Vlahovska PM. Curvature fluctuations of fluid vesicles reveal hydrodynamic dissipation within the bilayer. Proc Natl Acad Sci U S A 2024; 121:e2413557121. [PMID: 39441635 PMCID: PMC11536141 DOI: 10.1073/pnas.2413557121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 09/24/2024] [Indexed: 10/25/2024] Open
Abstract
The biological function of membranes is closely related to their softness, which is often studied through the membranes' thermally driven fluctuations. Typically, the analysis assumes that the relaxation rate of a pure bending deformation is determined by the competition between membrane bending rigidity and viscous dissipation in the surrounding medium. Here, we reexamine this assumption and demonstrate that viscous flows within the membrane dominate the dynamics of bending fluctuations of nonplanar membranes with a radius of curvature smaller than the Saffman-Delbrück length. Using flickering spectroscopy of giant vesicles made of dipalmitoylphosphatidylcholine, DPPC:cholesterol mixtures and pure diblock-copolymer membranes, we experimentally detect the signature of membrane dissipation in curvature fluctuations. We show that membrane viscosity can be reliably obtained from the short time behavior of the shape time correlations. The results indicate that the DPPC:cholesterol membranes behave as a Newtonian fluid, while the polymer membranes exhibit more complex rheology. Our study provides physical insights into the time scales of curvature remodeling of biological and synthetic membranes.
Collapse
Affiliation(s)
- Hammad A. Faizi
- Department of Mechanical Engineering, Northwestern University, Evanston, IL60208
| | - Rony Granek
- Avram and Stella Goldstein-Goren Department of Biotechnology Engineering, and Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of The Negev, Beer Sheva84105, Israel
| | - Petia M. Vlahovska
- Department of Engineering Sciences and Applied Mathematics, Northwestern University, Evanston, IL60208
- National Institute for Theory and Mathematics in Biology, Northwestern University and The University of Chicago, Chicago, IL60611
| |
Collapse
|
15
|
Zhao Z, Song H, Qi M, Liu Y, Zhang Y, Li S, Zhang H, Sun Y, Sun Y, Gao Z. Brain targeted polymeric micelles as drug carriers for ischaemic stroke treatment. J Drug Target 2024:1-17. [PMID: 39403962 DOI: 10.1080/1061186x.2024.2417190] [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: 08/24/2024] [Revised: 09/30/2024] [Accepted: 10/09/2024] [Indexed: 10/19/2024]
Abstract
Ischaemic stroke is a central nervous system disease with high morbidity, recurrence and mortality rates. Thrombolytic and neuroprotective therapies are the main therapeutic strategies for ischaemic stroke, however, the poor delivery efficiency of thrombolytic and neuroprotective drugs to the brain limits their clinical application. So far, the development of nanomedicine has brought opportunities for the above challenges, which can not only realise the effective accumulation of drugs in the target site, but also improve the pharmacokinetic behaviour of the drugs. Among the most rapidly developing nanoparticles, micelles gradually emerging as an effective strategy for ischaemic stroke treatment due to their own unique advantages. This review provided an overview of targeted and response-release micelles based on the physicochemical properties of the ischaemic stroke microenvironment, summarised the targeting strategies for delivering micellar formulations to the thrombus, blood-brain barrier, and brain parenchyma, and finally described the potentials and challenges of polymeric micelles in the treatment of ischaemic stroke.
Collapse
Affiliation(s)
- Zirui Zhao
- Department of Pharmacy, School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
| | - Huijia Song
- Department of Pharmacy, School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
- State Key Laboratory Breeding Base - Hebei Province Key Laboratory of Molecular Chemistry for Drugs, Hebei University of Science and Technology, Shijiazhuang, China
- Hebei Research Center of Pharmaceutical and Chemical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
| | - Mengge Qi
- Department of Pharmacy, School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
| | - Yurong Liu
- Department of Pharmacy, School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
| | - Yanchao Zhang
- Department of Pharmacy, School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
| | - Shuo Li
- Department of Pharmacy, School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
- State Key Laboratory Breeding Base - Hebei Province Key Laboratory of Molecular Chemistry for Drugs, Hebei University of Science and Technology, Shijiazhuang, China
- Hebei Research Center of Pharmaceutical and Chemical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
| | - Huimin Zhang
- Department of Pharmacy, School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
- State Key Laboratory Breeding Base - Hebei Province Key Laboratory of Molecular Chemistry for Drugs, Hebei University of Science and Technology, Shijiazhuang, China
- Hebei Research Center of Pharmaceutical and Chemical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
| | - Yongjun Sun
- Department of Pharmacy, School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
- State Key Laboratory Breeding Base - Hebei Province Key Laboratory of Molecular Chemistry for Drugs, Hebei University of Science and Technology, Shijiazhuang, China
- Hebei Research Center of Pharmaceutical and Chemical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
| | - Yanping Sun
- Department of Pharmacy, School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
- State Key Laboratory Breeding Base - Hebei Province Key Laboratory of Molecular Chemistry for Drugs, Hebei University of Science and Technology, Shijiazhuang, China
- Hebei Research Center of Pharmaceutical and Chemical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
| | - Zibin Gao
- Department of Pharmacy, School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
- State Key Laboratory Breeding Base - Hebei Province Key Laboratory of Molecular Chemistry for Drugs, Hebei University of Science and Technology, Shijiazhuang, China
- Hebei Research Center of Pharmaceutical and Chemical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
| |
Collapse
|
16
|
Thanapongpibul C, Rifaie-Graham O, Ojansivu M, Najer A, Kim H, Bakker SE, Chami M, Peeler DJ, Liu C, Yeow J, Stevens MM. Unlocking Intracellular Protein Delivery by Harnessing Polymersomes Synthesized at Microliter Volumes using Photo-PISA. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024:e2408000. [PMID: 39417762 DOI: 10.1002/adma.202408000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 09/06/2024] [Indexed: 10/19/2024]
Abstract
Efficient delivery of therapeutic proteins and vaccine antigens to intracellular targets is challenging due to generally poor cell membrane permeation and endolysosomal entrapment causing degradation. Herein, these challenges are addressed by developing an oxygen-tolerant photoinitiated polymerization-induced self-assembly (Photo-PISA) process, allowing for the microliter-scale (10 µL) synthesis of protein-loaded polymersomes directly in 1536-well plates. High-resolution techniques capable of analysis at a single particle level are employed to analyze protein encapsulation and release mechanisms. Using confocal microscopy and super-resolution stochastic optical reconstruction microscopy (STORM) imaging, their ability to deliver proteins into the cytosol following endosomal escape is subsequently visualized. Lastly, the adaptability of these polymersomes is exploited to encapsulate and deliver a prototype vaccine antigen, demonstrating its ability to activate antigen-presenting cells and support antigen cross-presentation for applications in subunit vaccines and cancer immunotherapy. This combination of ultralow volume synthesis and efficient intracellular delivery holds significant promise for unlocking the high throughput screening of a broad range of otherwise cost-prohibitive or early-stage therapeutic protein and vaccine antigen candidates that can be difficult to obtain in large quantities. The versatility of this platform for rapid screening of intracellular protein delivery can result in significant advancements across the fields of nanomedicine and biomedical engineering.
Collapse
Affiliation(s)
- Chalaisorn Thanapongpibul
- Department of Materials, Department of Bioengineering, and Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ, UK
| | - Omar Rifaie-Graham
- Department of Materials, Department of Bioengineering, and Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ, UK
| | - Miina Ojansivu
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, 17177, Sweden
| | - Adrian Najer
- Department of Materials, Department of Bioengineering, and Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ, UK
| | - Hyemin Kim
- Department of Materials, Department of Bioengineering, and Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ, UK
| | - Saskia E Bakker
- Advanced Bioimaging Research Technology Platform, University of Warwick, Gibbet Hill Road, Coventry, CV4 7AL, UK
| | - Mohamed Chami
- BioEM Lab, Biozentrum, University of Basel, Basel, 4058, Switzerland
| | - David J Peeler
- Department of Materials, Department of Bioengineering, and Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ, UK
| | - Chenchen Liu
- Department of Materials, Department of Bioengineering, and Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ, UK
- Kavli Institute for Nanoscience Discovery, Department of Physiology, Anatomy and Genetics, Department of Engineering Science, University of Oxford, Oxford, OX1 3QU, UK
| | - Jonathan Yeow
- Department of Materials, Department of Bioengineering, and Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ, UK
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Molly M Stevens
- Department of Materials, Department of Bioengineering, and Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ, UK
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, 17177, Sweden
- Kavli Institute for Nanoscience Discovery, Department of Physiology, Anatomy and Genetics, Department of Engineering Science, University of Oxford, Oxford, OX1 3QU, UK
| |
Collapse
|
17
|
Mardešić I, Boban Z, Raguz M. Electroformation of Giant Unilamellar Vesicles from Damp Films in Conditions Involving High Cholesterol Contents, Charged Lipids, and Saline Solutions. MEMBRANES 2024; 14:215. [PMID: 39452827 PMCID: PMC11510074 DOI: 10.3390/membranes14100215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 10/01/2024] [Accepted: 10/11/2024] [Indexed: 10/26/2024]
Abstract
Giant unilamellar vesicles (GUVs) are frequently used as membrane models in studies of membrane properties. They are most often produced using the electroformation method. However, there are a number of parameters that can influence the success of the procedure. Some of the most common conditions that have been shown to have a negative effect on GUV electroformation are the presence of high cholesterol (Chol) concentrations, the use of mixtures containing charged lipids, and the solutions with an elevated ionic strength. High Chol concentrations are problematic for the traditional electroformation protocol as it involves the formation of a dry lipid film by complete evaporation of the organic solvent from the lipid mixture. During drying, anhydrous Chol crystals form. They are not involved in the formation of the lipid bilayer, resulting in a lower Chol concentration in the vesicle bilayer compared to the original lipid mixture. Motivated primarily by the issue of artifactual Chol demixing, we have modified the electroformation protocol by incorporating the techniques of rapid solvent exchange (RSE), ultrasonication, plasma cleaning, and spin-coating for reproducible production of GUVs from damp lipid films. Aside from decreasing Chol demixing, we have shown that the method can also be used to produce GUVs from lipid mixtures with charged lipids and in ionic solutions used as internal solutions. A high yield of GUVs was obtained for Chol/1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPC) samples with mixing ratios ranging from 0 to 2.5. We also succeeded in preparing GUVs from mixtures containing up to 60 mol% of the charged lipid 1-palmitoyl-2-oleoyl-sn-glycero-3-phospho-L-serine (POPS) and in NaCl solutions with low ionic strength (<25 mM).
Collapse
Affiliation(s)
- Ivan Mardešić
- Department of Medical Physics and Biophysics, University of Split School of Medicine, 21000 Split, Croatia; (I.M.); (Z.B.)
- Doctoral Study of Biophysics, Faculty of Science, University of Split, 21000 Split, Croatia
| | - Zvonimir Boban
- Department of Medical Physics and Biophysics, University of Split School of Medicine, 21000 Split, Croatia; (I.M.); (Z.B.)
| | - Marija Raguz
- Department of Medical Physics and Biophysics, University of Split School of Medicine, 21000 Split, Croatia; (I.M.); (Z.B.)
| |
Collapse
|
18
|
Havelikar U, Ghorpade KB, Kumar A, Patel A, Singh M, Banjare N, Gupta PN. Comprehensive insights into mechanism of nanotoxicity, assessment methods and regulatory challenges of nanomedicines. DISCOVER NANO 2024; 19:165. [PMID: 39365367 PMCID: PMC11452581 DOI: 10.1186/s11671-024-04118-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 09/26/2024] [Indexed: 10/05/2024]
Abstract
Nanomedicine has the potential to transform healthcare by offering targeted therapies, precise diagnostics, and enhanced drug delivery systems. The National Institutes of Health has coined the term "nanomedicine" to describe the use of nanotechnology in biological system monitoring, control, diagnosis, and treatment. Nanomedicine continues to receive increasing interest for the rationalized delivery of therapeutics and pharmaceutical agents to achieve the required response while reducing its side effects. However, as nanotechnology continues to advance, concerns about its potential toxicological effects have also grown. This review explores the current state of nanomedicine, focusing on the types of nanoparticles used and their associated properties that contribute to nanotoxicity. It examines the mechanisms through which nanoparticles exert toxicity, encompassing various cellular and molecular interactions. Furthermore, it discusses the assessment methods employed to evaluate nanotoxicity, encompassing in-vitro and in-vivo models, as well as emerging techniques. The review also addresses the regulatory issues surrounding nanotoxicology, highlighting the challenges in developing standardized guidelines and ensuring the secure translation of nanomedicine into clinical settings. It also explores into the challenges and ethical issues associated with nanotoxicology, as understanding the safety profile of nanoparticles is essential for their effective translation into therapeutic applications.
Collapse
Affiliation(s)
- Ujwal Havelikar
- Department of Pharmaceutics, NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, 303121, India
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu, 180001, India
| | - Kabirdas B Ghorpade
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) - Raebareli, Lucknow, Uttar Pradesh, 226002, India
| | - Amit Kumar
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) - Raebareli, Lucknow, Uttar Pradesh, 226002, India
| | - Akhilesh Patel
- Department of Pharmaceutics, NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, 303121, India
| | - Manisha Singh
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu, 180001, India
| | - Nagma Banjare
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu, 180001, India
| | - Prem N Gupta
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu, 180001, India.
| |
Collapse
|
19
|
Lin Y, Chen H, Wang L, Su J, Li J, Huang X. Lipase activated endocytosis-like behavior of oil-in-water emulsion. Nat Commun 2024; 15:8517. [PMID: 39353937 PMCID: PMC11445447 DOI: 10.1038/s41467-024-52802-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 09/23/2024] [Indexed: 10/03/2024] Open
Abstract
Oil-in-water emulsion is a system with extensive applications in foods, cosmetics and coating industries, and it could also be designed into an artificial lipid droplet in recent works. However, the insights into the biophysical dynamic behaviors of such artificial lipid droplets are lacking. Here, we reveal an enzymatic reaction triggered endocytosis-like behavior in the oil-in-water emulsion lipid droplets. A thermodynamically favored recruitment of lipases onto the membrane of the droplets is demonstrated. We confirm that the hydrolysis of tributyrin by lipases can decrease the interfacial tension and increase the compressive force on the membrane, which are the two main driving forces for triggering the endocytosis-like behavior. The endocytosis-like behavior induced various emerging functionalities of the lipid droplets, including proteins, DNA or inorganic particles being efficiently sequestered into the oil droplet with reversible release as well as enhanced cascade enzymatic reaction. Overall, our studies are expected to open up a way to functionalize oil-in-water emulsions capable of life-inspired behaviors and tackle emerging challenges in bottom-up synthetic biology, revealing the unknown dynamic behaviors of lipid droplets in living organisms.
Collapse
Affiliation(s)
- Youping Lin
- MIIT Key Laboratory of Critical Materials Technology for New Energy Conversion and Storage, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150001, China
| | - Haixu Chen
- MIIT Key Laboratory of Critical Materials Technology for New Energy Conversion and Storage, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150001, China
| | - Lei Wang
- MIIT Key Laboratory of Critical Materials Technology for New Energy Conversion and Storage, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150001, China
| | - Jiaojiao Su
- MIIT Key Laboratory of Critical Materials Technology for New Energy Conversion and Storage, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150001, China
| | - Junbo Li
- MIIT Key Laboratory of Critical Materials Technology for New Energy Conversion and Storage, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150001, China
| | - Xin Huang
- MIIT Key Laboratory of Critical Materials Technology for New Energy Conversion and Storage, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150001, China.
| |
Collapse
|
20
|
Lin P, Zhang S, Komatsubara F, Konishi H, Nakata E, Morii T. Artificial Compartments Encapsulating Enzymatic Reactions: Towards the Construction of Artificial Organelles. Chempluschem 2024:e202400483. [PMID: 39351818 DOI: 10.1002/cplu.202400483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 09/30/2024] [Indexed: 11/08/2024]
Abstract
Cells have used compartmentalization to implement complex biological processes involving thousands of enzyme cascade reactions. Enzymes are spatially organized into the cellular compartments to carry out specific and efficient reactions in a spatiotemporally controlled manner. These compartments are divided into membrane-bound and membraneless organelles. Mimicking such cellular compartment systems has been a challenge for years. A variety of artificial scaffolds, including liposomes, polymersomes, proteins, nucleic acids, or hybrid materials have been used to construct artificial membrane-bound or membraneless compartments. These artificial compartments may have great potential for applications in biosynthesis, drug delivery, diagnosis and therapeutics, among others. This review first summarizes the typical examples of cellular compartments. In particular, the recent studies on cellular membraneless organelles (biomolecular condensates) are reviewed. We then summarize the recent advances in the construction of artificial compartments using engineered platforms. Finally, we provide our insights into the construction of biomimetic systems and the applications of these systems. This review article provides a timely summary of the relevant perspectives for the future development of artificial compartments, the building blocks for the construction of artificial organelles or cells.
Collapse
Affiliation(s)
- Peng Lin
- Institute of Advanced Energy, Kyoto University, Uji-shi, Kyoto, 611-0011, Japan
| | - Shiwei Zhang
- Institute of Advanced Energy, Kyoto University, Uji-shi, Kyoto, 611-0011, Japan
| | - Futa Komatsubara
- Graduate School of Energy Science, Kyoto University, Yoshida-hommachi, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Hiroaki Konishi
- Institute of Advanced Energy, Kyoto University, Uji-shi, Kyoto, 611-0011, Japan
| | - Eiji Nakata
- Institute of Advanced Energy, Kyoto University, Uji-shi, Kyoto, 611-0011, Japan
| | - Takashi Morii
- Institute of Advanced Energy, Kyoto University, Uji-shi, Kyoto, 611-0011, Japan
- Department of Health and Nutrition, Kyoto Koka Women's University, Ukyo-ku, Kyoto, 615-0882, Japan
| |
Collapse
|
21
|
Huang J, Jaekel A, van den Boom J, Podlesainski D, Elnaggar M, Heuer-Jungemann A, Kaiser M, Meyer H, Saccà B. A modular DNA origami nanocompartment for engineering a cell-free, protein unfolding and degradation pathway. NATURE NANOTECHNOLOGY 2024; 19:1521-1531. [PMID: 39075293 PMCID: PMC11486656 DOI: 10.1038/s41565-024-01738-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 05/28/2024] [Indexed: 07/31/2024]
Abstract
Within the cell, chemical reactions are often confined and organized through a modular architecture. This facilitates the targeted localization of molecular species and their efficient translocation to subsequent sites. Here we present a cell-free nanoscale model that exploits compartmentalization strategies to carry out regulated protein unfolding and degradation. Our synthetic model comprises two connected DNA origami nanocompartments (each measuring 25 nm × 41 nm × 53 nm): one containing the protein unfolding machine, p97, and the other housing the protease chymotrypsin. We achieve the unidirectional immobilization of p97 within the first compartment, establishing a gateway mechanism that controls substrate recruitment, translocation and processing within the second compartment. Our data show that, whereas spatial confinement increases the rate of the individual reactions by up to tenfold, the physical connection of the compartmentalized enzymes into a chimera efficiently couples the two reactions and reduces off-target proteolysis by almost sixfold. Hence, our modular approach may serve as a blueprint for engineering artificial nanofactories with reshaped catalytic performance and functionalities beyond those observed in natural systems.
Collapse
Affiliation(s)
- J Huang
- Bionanotechnology, CENIDE and ZMB, University of Duisburg-Essen, Essen, Germany
| | - A Jaekel
- Bionanotechnology, CENIDE and ZMB, University of Duisburg-Essen, Essen, Germany
| | - J van den Boom
- Molecular Biology, ZMB, University of Duisburg-Essen, Essen, Germany
| | - D Podlesainski
- Chemical Biology, ZMB, University of Duisburg-Essen, Essen, Germany
| | - M Elnaggar
- Max Planck Institute of Biochemistry, Martinsried, Germany
| | | | - M Kaiser
- Chemical Biology, ZMB, University of Duisburg-Essen, Essen, Germany
| | - H Meyer
- Molecular Biology, ZMB, University of Duisburg-Essen, Essen, Germany.
| | - B Saccà
- Bionanotechnology, CENIDE and ZMB, University of Duisburg-Essen, Essen, Germany.
| |
Collapse
|
22
|
Rosso AP, de Oliveira FA, Guégan P, Jager E, Giacomelli FC. Evaluation of polymersome permeability as a fundamental aspect towards the development of artificial cells and nanofactories. J Colloid Interface Sci 2024; 671:88-99. [PMID: 38795537 DOI: 10.1016/j.jcis.2024.05.133] [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: 03/06/2024] [Revised: 05/06/2024] [Accepted: 05/18/2024] [Indexed: 05/28/2024]
Abstract
Polymersomes are synthetic vesicles with potential use in healthcare, chemical transformations in confined environment (nanofactories), and in the construction of artificial cells and organelles. In this framework, one of the most important features of such supramolecular structures is the permeability behavior allowing for selective control of mass exchange between the inner and outer compartments. The use of biological and synthetic nanopores in this regard is the most common strategy to impart permeability nevertheless, this typically requires fairly complex strategies to enable porosity. Yet, investigations concerning the permeability of polymer vesicles to different analytes still requires further exploration and, taking these considerations into account, we have detailed investigated the permeability behavior of a variety of polymersomes with regard to different analytes (water, protons, and rhodamine B) which were selected as models for solvents, ions, and small molecules. Polymersomes based on hydrophilic blocks of poly[N-(2-hydroxypropyl)methacrylamide] (PHPMA) or PEO (poly(ethylene oxide)) linked to the non-responsive blocks poly[N-(4-isopropylphenylacetamide)ethyl methacrylate] (PPPhA) or poly(methyl methacrylate) (PMMA), or to the stimuli pH-responsive block poly[2-(diisopropylamino)ethyl methacrylate] (PDPA) have been investigated. Interestingly, the produced PEO-based vesicles are notably larger than the ones produced using PHPMA-containing block copolymers. The experimental results reveal that all the vesicles are inherently permeable to some extent with permeability behavior following exponential profiles. Nevertheless, polymersomes based on PMMA as the hydrophobic component were demonstrated to be the least permeable to the small molecule rhodamine B as well as to water. The synthetic vesicles based on the pH-responsive PDPA block exhibited restrictive and notably slow proton permeability as attributed to partial chain protonation upon acidification of the medium. The dye permeability was evidenced to be much slower than ion or solvent diffusion, and in the case of pH-responsive assemblies, it was demonstrated to also depend on the ionic strength of the environment. These findings are understood to be highly relevant towards polymer selection for the production of synthetic vesicles with selective and time-dependent permeability, and it may thus contribute in advancing biomimicry and nanomedicine.
Collapse
Affiliation(s)
- Anabella P Rosso
- Centro de Ciências Naturais e Humanas, Universidade Federal do ABC, Santo André, Brazil
| | | | - Philippe Guégan
- Equipe Chimie des Polymères, Institut Parisien de Chimie Moléculaire (UMR-CNRS 8232), Sorbonne Université, Paris, France
| | - Eliezer Jager
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Prague, Czech Republic
| | - Fernando C Giacomelli
- Centro de Ciências Naturais e Humanas, Universidade Federal do ABC, Santo André, Brazil.
| |
Collapse
|
23
|
Wong CK, Lai RY, Stenzel MH. Polymersomes with micellar patches. J Colloid Interface Sci 2024; 671:449-456. [PMID: 38815380 DOI: 10.1016/j.jcis.2024.05.177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 04/29/2024] [Accepted: 05/22/2024] [Indexed: 06/01/2024]
Abstract
Hollow block copolymer particles called polymer vesicles (polymersomes) serve as versatile containers for compartmentalization in synthetic biology and drug delivery. Recently, there has been growing interest in using polymersomes as colloidal building blocks for creating higher-order clustered structures. Most reports thus far rely on the use of DNA base-pairing interactions to "glue" polymersomes with other colloidal components. In this study, we present two alternative electrostatically driven approaches to assemble polymersomes and model colloids (micelles) into hybrid clusters. The first approach uses pH to manipulate electrostatic interactions and effectively control the clustering extent of micellar subunits on polymersomes, while the second approach relies on the hydrolysis of an acid trigger, glucono delta-lactone (GDL), to introduce temporal control over clustering. We envisage our approaches and structures reported herein will help inspire the creation of new prospects for materials science and biomedical applications.
Collapse
Affiliation(s)
- Chin Ken Wong
- School of Chemistry, University of New South Wales (UNSW), Sydney 2052, NSW, Australia.
| | - Rebecca Y Lai
- School of Chemistry, University of New South Wales (UNSW), Sydney 2052, NSW, Australia
| | - Martina H Stenzel
- School of Chemistry, University of New South Wales (UNSW), Sydney 2052, NSW, Australia.
| |
Collapse
|
24
|
Hazegh Nikroo A, Altenburg WJ, van Veldhuisen TW, Brunsveld L, van Hest JCM. Spatiotemporal Control Over Protein Release from Artificial Cells via a Light-Activatable Protease. Adv Biol (Weinh) 2024:e2400353. [PMID: 39334525 DOI: 10.1002/adbi.202400353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 09/11/2024] [Indexed: 09/30/2024]
Abstract
The regulation of protein uptake and secretion by cells is paramount for intercellular signaling and complex multicellular behavior. Mimicking protein-mediated communication in artificial cells holds great promise to elucidate the underlying working principles, but remains challenging without the stimulus-responsive regulatory machinery of living cells. Therefore, systems to precisely control when and where protein release occurs should be incorporated in artificial cells. Here, a light-activatable TEV protease (LaTEV) is presented that enables spatiotemporal control over protein release from a coacervate-based artificial cell platform. Due to the presence of Ni2+-nitrilotriacetic acid moieties within the coacervates, His-tagged proteins are effectively sequestered into the coacervates. LaTEV is first photocaged, effectively blocking its activity. Upon activation by irradiation with 365 nm light, LaTEV cleaves the His-tags from sequestered cargo proteins, resulting in their release. The successful blocking and activation of LaTEV provides control over protein release rate and triggerable protein release from specific coacervates via selective irradiation. Furthermore, light-activated directional transfer of proteins between two artificial cell populations is demonstrated. Overall, this system opens up avenues to engineer light-responsive protein-mediated communication in artificial cell context, which can advance the probing of intercellular signaling and the development of protein delivery platforms.
Collapse
Affiliation(s)
- Arjan Hazegh Nikroo
- Laboratory of Bio-Organic Chemistry, Department of Biomedical Engineering, and Institute for Complex Molecular Systems, Eindhoven University of Technology, P.O. Box 513, Eindhoven, 5600 MB, The Netherlands
| | - Wiggert J Altenburg
- Laboratory of Bio-Organic Chemistry, Department of Biomedical Engineering, and Institute for Complex Molecular Systems, Eindhoven University of Technology, P.O. Box 513, Eindhoven, 5600 MB, The Netherlands
| | - Thijs W van Veldhuisen
- Laboratory of Bio-Organic Chemistry, Department of Biomedical Engineering, and Institute for Complex Molecular Systems, Eindhoven University of Technology, P.O. Box 513, Eindhoven, 5600 MB, The Netherlands
- Laboratory of Chemical Biology, Department of Biomedical Engineering, and Institute for Complex Molecular Systems, Eindhoven University of Technology, P.O. Box 513, Eindhoven, 5600 MB, The Netherlands
| | - Luc Brunsveld
- Laboratory of Chemical Biology, Department of Biomedical Engineering, and Institute for Complex Molecular Systems, Eindhoven University of Technology, P.O. Box 513, Eindhoven, 5600 MB, The Netherlands
| | - Jan C M van Hest
- Laboratory of Bio-Organic Chemistry, Department of Biomedical Engineering, and Institute for Complex Molecular Systems, Eindhoven University of Technology, P.O. Box 513, Eindhoven, 5600 MB, The Netherlands
| |
Collapse
|
25
|
Wang H, Zhang G, Lin M, Hartinger CG, Sun J. Zwitterionic Polyelectrolyte Complex Vesicles Assembled from Homopoly(2-Oxazoline)s as Enzyme Catalytic Nanoreactors for Potent Anti-Tumor Efficiency. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:19423-19429. [PMID: 39083025 DOI: 10.1021/acs.langmuir.4c01729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/18/2024]
Abstract
Enzymes are known for their remarkable catalytic efficiency across a wide range of applications. Here, we present a novel and convenient nanoreactor platform based on zwitterionic polyelectrolyte complex vesicles (PCVs), assembled from oppositely charged homopoly(2-oxazoline)s, facilitating enzyme immobilization. We show remarkable enhancements in catalytic activity and stability by encapsulation of lipase as a model enzyme. Even as the temperature rises, the performance of the lipase remains robust. Further, the structural characteristics of PCVs, including hollow architecture and semipermeable membranes, endow them with unique advantages for enzyme cascade reactions involving glucose oxidase (GOx) and horseradish peroxidase (HRP). A decline in catalytic efficiency is shown when the enzymes are individually loaded and subsequently mixed, in contrast to the coloaded GOx-HRP-PCV group. We demonstrate that the vesicle structures establish confined environments where precise enzyme-substrate interactions facilitate enhanced catalytic efficiency. In addition, the nanoreactors exhibit excellent biocompatibility and efficient anti-tumor activity, which hold significant promise for biomedical applications within enzyme-based technologies.
Collapse
Affiliation(s)
- Hepeng Wang
- China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P. R. China
| | - Guojing Zhang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, Jilin 130012, China
| | - Min Lin
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, Jilin 130012, China
| | - Christian G Hartinger
- School of Chemical Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Jing Sun
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, Jilin 130012, China
| |
Collapse
|
26
|
Song S, Ivanov T, Yuan D, Wang J, da Silva LC, Xie J, Cao S. Peptide-Based Biomimetic Condensates via Liquid-Liquid Phase Separation as Biomedical Delivery Vehicles. Biomacromolecules 2024; 25:5468-5488. [PMID: 39178343 DOI: 10.1021/acs.biomac.4c00814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/25/2024]
Abstract
Biomolecular condensates are dynamic liquid droplets through intracellular liquid-liquid phase separation that function as membraneless organelles, which are highly involved in various complex cellular processes and functions. Artificial analogs formed via similar pathways that can be integrated with biological complexity and advanced functions have received tremendous research interest in the field of synthetic biology. The coacervate droplet-based compartments can partition and concentrate a wide range of solutes, which are regarded as attractive candidates for mimicking phase-separation behaviors and biophysical features of biomolecular condensates. The use of peptide-based materials as phase-separating components has advantages such as the diversity of amino acid residues and customized sequence design, which allows for programming their phase-separation behaviors and the physicochemical properties of the resulting compartments. In this Perspective, we highlight the recent advancements in the design and construction of biomimicry condensates from synthetic peptides relevant to intracellular phase-separating protein, with specific reference to their molecular design, self-assembly via phase separation, and biorelated applications, to envisage the use of peptide-based droplets as emerging biomedical delivery vehicles.
Collapse
Affiliation(s)
- Siyu Song
- Life-Like Materials and Systems, Department of Chemistry, University of Mainz, Mainz 55128, Germany
| | | | - Dandan Yuan
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Jianqiang Wang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | | | - Jing Xie
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Shoupeng Cao
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| |
Collapse
|
27
|
Palivan CG, Heuberger L, Gaitzsch J, Voit B, Appelhans D, Borges Fernandes B, Battaglia G, Du J, Abdelmohsen L, van Hest JCM, Hu J, Liu S, Zhong Z, Sun H, Mutschler A, Lecommandoux S. Advancing Artificial Cells with Functional Compartmentalized Polymeric Systems - In Honor of Wolfgang Meier. Biomacromolecules 2024; 25:5454-5467. [PMID: 39196319 DOI: 10.1021/acs.biomac.4c00769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Abstract
The fundamental building block of living organisms is the cell, which is the universal biological base of all living entities. This micrometric mass of cytoplasm and the membrane border have fascinated scientists due to the highly complex and multicompartmentalized structure. This specific organization enables numerous metabolic reactions to occur simultaneously and in segregated spaces, without disturbing each other, but with a promotion of inter- and intracellular communication of biomolecules. At present, artificial nano- and microcompartments, whether as single components or self-organized in multicompartment architectures, hold significant value in the study of life development and advanced functional materials and in the fabrication of molecular devices for medical applications. These artificial compartments also possess the properties to encapsulate, protect, and control the release of bio(macro)molecules through selective transport processes, and they are capable of embedding or being connected with other types of compartments. The self-assembly mechanism of specific synthetic compartments and thus the fabrication of a simulated organelle membrane are some of the major aspects to gain insight. Considerable efforts have now been devoted to design various nano- and microcompartments and understand their functionality for precise control over properties. Of particular interest is the use of polymeric vesicles for communication in synthetic cells and colloidal systems to reinitiate chemical and biological communication and thus close the gap toward biological functions. Multicompartment systems can now be effectively created with a high level of hierarchical control. In this way, these structures can not only be explored to deepen our understanding of the functional organization of living cells, but also pave the way for many more exciting developments in the biomedical field.
Collapse
Affiliation(s)
- Cornelia G Palivan
- Department of Chemistry, University of Basel, Mattenstrasse 22, 4002 Basel, Switzerland
| | - Lukas Heuberger
- Department of Chemistry, University of Basel, Mattenstrasse 22, 4002 Basel, Switzerland
| | - Jens Gaitzsch
- Leibniz-Institut für Polymerforschung Dresden e.V., Hohe Straße 6, Dresden 01069, Germany
| | - Brigitte Voit
- Leibniz-Institut für Polymerforschung Dresden e.V., Hohe Straße 6, Dresden 01069, Germany
| | - Dietmar Appelhans
- Leibniz-Institut für Polymerforschung Dresden e.V., Hohe Straße 6, Dresden 01069, Germany
| | - Barbara Borges Fernandes
- Institute for Bioengineering of Catalonia, Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
- Department of Condensed Matter Physics, University of Barcelona, 08028 Barcelona, Spain
| | - Giuseppe Battaglia
- Institute for Bioengineering of Catalonia, Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
- Catalan Institution for Research and Advanced Studies, 08010 Barcelona, Spain
| | - Jianzhong Du
- Department of Polymeric Materials, School of Materials Science and Engineering, Tongji University, 4800 Caoan Road, Shanghai 201804, China
- School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Loai Abdelmohsen
- Department of Chemistry and Chemical Engineering, Institute for Complex Molecular Systems, Bio-Organic Chemistry, Eindhoven University of Technology, Helix, P.O. Box 513, 5600MB Eindhoven, The Netherlands
| | - Jan C M van Hest
- Department of Chemistry and Chemical Engineering, Institute for Complex Molecular Systems, Bio-Organic Chemistry, Eindhoven University of Technology, Helix, P.O. Box 513, 5600MB Eindhoven, The Netherlands
| | - Jinming Hu
- Department of Pharmacy, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine and Laboratory of Precision and Intelligent Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui Province 230026, China
| | - Shiyong Liu
- Department of Pharmacy, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine and Laboratory of Precision and Intelligent Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui Province 230026, China
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, P. R. China
- College of Pharmaceutical Sciences, and International College of Pharmaceutical Innovation, Soochow University, Suzhou 215123, P. R. China
| | - Huanli Sun
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, P. R. China
| | - Angela Mutschler
- Univ. Bordeaux, CNRS, Bordeaux INP, LCPO, UMR 5629, F-33600 Pessac, France
| | | |
Collapse
|
28
|
Yang ZS, Gao S, Zhang JL. Magnetic manipulation of the reactivity of singlet oxygen: from test tubes to living cells. Natl Sci Rev 2024; 11:nwae069. [PMID: 39144743 PMCID: PMC11321247 DOI: 10.1093/nsr/nwae069] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 01/14/2024] [Accepted: 02/17/2024] [Indexed: 08/16/2024] Open
Abstract
Although magnetism undoubtedly influences life on Earth, the science behind biological magnetic sensing is largely a mystery, and it has proved challenging, especially in the life sciences, to harness the interactions of magnetic fields (MFs) with matter to achieve specific ends. Using the well-established radical pair (RP) mechanism, we here demonstrate a bottom-up strategy for the exploitation of MF effects in living cells by translating knowledge from studies of RP reactions performed in vitro. We found an unprecedented MF dependence of the reactivity of singlet oxygen (1O2) towards electron-rich substrates (S) such as anthracene, lipids and iodide, in which [S ˙+ O2 ˙-] RPs are formed as a basis for MFs influencing molecular redox events in biological systems. The close similarity of the observed MF effects on the biologically relevant process of lipid peroxidation in solution, in membrane mimics and in living cells, shows that MFs can reliably be used to manipulate 1O2-induced cytotoxicity and cell-apoptosis-related protein expression. These findings led to a 'proof-of-concept' study on MF-assisted photodynamic therapy in vivo, highlighting the potential of MFs as a non-invasive tool for controlling cellular events.
Collapse
Affiliation(s)
- Zi-Shu Yang
- Institute of Inorganic Chemistry, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Song Gao
- Institute of Inorganic Chemistry, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
- Spin-X Institute and Guangdong-Hong Kong-Macao Joint Laboratory of Optoelectronic and Magnetic Functional Materials, South China University of Technology, Guangzhou 510641, China
- Institute of Inorganic and Material Chemistry, School of Chemistry and Chemical Engineering, Sun Yat-sen University, Guangzhou 510275, China
- Chemistry and Chemical Engineering Guangdong Laboratory, Shantou 515031, China
| | - Jun-Long Zhang
- Institute of Inorganic Chemistry, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
- Chemistry and Chemical Engineering Guangdong Laboratory, Shantou 515031, China
| |
Collapse
|
29
|
Mustafa YL, Balestri A, Huang X, Palivan C. Redefining drug therapy: innovative approaches using catalytic compartments. Expert Opin Drug Deliv 2024; 21:1395-1413. [PMID: 39259136 DOI: 10.1080/17425247.2024.2403476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 08/22/2024] [Accepted: 09/09/2024] [Indexed: 09/12/2024]
Abstract
INTRODUCTION Rapid excretion of drug derivatives often results in short drug half-lives, necessitating frequent administrations. Catalytic compartments, also known as nano- and microreactors, offer a solution by providing confined environments for in situ production of therapeutic agents. Inspired by natural compartments, polymer-based catalytic compartments have been developed to improve reaction efficiency and enable site-specific therapeutic applications. AREAS COVERED Polymer-based compartments provide stability, permeability control, and responsiveness to stimuli, making them ideal for generating localized compounds/signals. These sophisticated systems, engineered to carry active compounds and enable selective molecular release, represent a significant advancement in pharmaceutical research. They mimic cellular functions, creating controlled catalytic environments for bio-relevant processes. This review explores the latest advancements in synthetic catalytic compartments, focusing on design approaches, building blocks, active molecules, and key bio-applications. EXPERT OPINION Catalytic compartments hold transformative potential in precision medicine by improving therapeutic outcomes through precise, on-site production of therapeutic agents. While promising, challenges like scalable manufacturing, biodegradability, and regulatory hurdles must be addressed to realize their full potential. Addressing these will be crucial for their successful application in healthcare.
Collapse
Affiliation(s)
| | - Arianna Balestri
- Department of Chemistry, University of Basel, Basel, Switzerland
| | - Xinan Huang
- Department of Chemistry, University of Basel, Basel, Switzerland
| | - Cornelia Palivan
- Department of Chemistry, University of Basel, Basel, Switzerland
- National Centre of Competence in Research-Molecular Systems Engineering, Basel, Switzerland
| |
Collapse
|
30
|
Dhara TK, Khawas S, Sharma N. Lipid nanoparticles for pulmonary fibrosis: A comprehensive review. Pulm Pharmacol Ther 2024; 87:102319. [PMID: 39216596 DOI: 10.1016/j.pupt.2024.102319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 08/07/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a fatal progressive and irreversible ailment associated with the proliferation of fibroblast and accumulation of extracellular matrix (ECM) with gradual scarring of lung tissue. Despite several research studies, the treatments available are not efficient enough for the reversal of the disease and are constantly in progress. No drugs other than Pirfenidone and Nintedanib have been approved for the treatment of IPF, necessitating the exploration of novel therapeutic strategies. Recently, lipid-based nanoparticles (LNPs) have drawn more attention because of their potential to enhance the solubility of drugs, cross biological barriers of the lungs and specifically target lung fibrotic tissues, overcoming various challenges in treating IPF. LNPs offer a versatile platform to encapsulate a wide range of drugs, both hydrophilic and lipophilic, improving their bioavailability, allowing sustained release and reducing toxicity, which radiates their significant role in addressing the complexities of IPF. This review summarizes the pathogenesis and conventional treatment of idiopathic pulmonary fibrosis, along with their drawbacks. The review focuses on different types of lipid-based nanoparticles that have been tested in the treatment of idiopathic pulmonary fibrosis, including nanoemulsions, liposomes, solid lipid nanoparticles, nanostructured lipid carriers, niosomes and lipid-polymer hybrid nanoparticles. The review also highlights the future prospects that can offer a potential approach for developing novel strategies to treat idiopathic pulmonary fibrosis.
Collapse
Affiliation(s)
- Tushar Kanti Dhara
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, India
| | - Sayak Khawas
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, India
| | - Neelima Sharma
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, India.
| |
Collapse
|
31
|
Yang X, Yang J, Wei L, Zhang Y, Yang J, Ni M, Dong Y. Formation of a planar biomimetic membrane with a novel zwitterionic polymer for nanopore sequencing. J Mater Chem B 2024; 12:8189-8199. [PMID: 39082061 DOI: 10.1039/d4tb01007h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2024]
Abstract
Biological membranes containing transmembrane channels play a crucial role in numerous cellular processes, and mimicking of cell membranes has garnered significant interest in various biomedical applications, particularly nanopore sequencing technology, where remarkable progress has been made with nanopore membranes. Considering the fragility of biomimetic membranes formed by artificial lipids and the limited mimicry of those formed by common block copolymers, this study developed a novel amphiphilic polymer by covalently linking hydrophilic heads of phospholipids to the ends of hydrophobic poly(dimethyl siloxane) (PDMS) chains. The absence of hydrophilic blocks allowed for good control over the polydispersity of this polymer within a narrow range. The high flexibility of PDMS chains, combined with relatively uniform molecular weights, would confer enhanced stability and robustness to polymeric membranes. Dynamic light scattering measurements and microdroplet formation tests demonstrated good amphipathic properties of these novel polymers when maintaining an appropriate hydrophilic-hydrophobic ratio. Moreover, the high similarity between the hydrophilic heads and natural phospholipids makes this polymer more compatible with biomolecules. A successful protein insertion experiment confirmed both the stability of this polymeric membrane and its compatibility with membrane proteins. As a result, this novel amphiphilic polymer exhibits great potential for biomembrane mimicking and paves a new path for material design in biomedical applications.
Collapse
Affiliation(s)
| | | | - Lai Wei
- BGI Research, Shenzhen 518083, China.
| | | | | | - Ming Ni
- BGI Research, Shenzhen 518083, China.
| | | |
Collapse
|
32
|
Zhou Y, Zhang K, Moreno S, Temme A, Voit B, Appelhans D. Continuous Transformation from Membrane-Less Coacervates to Membranized Coacervates and Giant Vesicles: Toward Multicompartmental Protocells with Complex (Membrane) Architectures. Angew Chem Int Ed Engl 2024; 63:e202407472. [PMID: 38847278 DOI: 10.1002/anie.202407472] [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: 04/19/2024] [Indexed: 07/25/2024]
Abstract
The membranization of membrane-less coacervates paves the way for the exploitation of complex protocells with regard to structural and cell-like functional behaviors. However, the controlled transformation from membranized coacervates to vesicles remains a challenge. This can provide stable (multi)phase and (multi)compartmental architectures through the reconfiguration of coacervate droplets in the presence of (bioactive) polymers, bio(macro)molecules and/or nanoobjects. Herein, we present a continuous protocell transformation from membrane-less coacervates to membranized coacervates and, ultimately, to giant hybrid vesicles. This transformation process is orchestrated by altering the balance of non-covalent interactions through varying concentrations of an anionic terpolymer, leading to dynamic processes such as spontaneous membranization of terpolymer nanoparticles at the coacervate surface, disassembly of the coacervate phase mediated by the excess anionic charge, and the redistribution of coacervate components in membrane. The diverse protocells during the transformation course provide distinct structural features and molecular permeability. Notably, the introduction of multiphase coacervates in this continuous transformation process signifies advancements toward the creation of synthetic cells with different diffusible compartments. Our findings emphasize the highly controlled continuous structural reorganization of coacervate protocells and represents a novel step toward the development of advanced and sophisticated synthetic protocells with more precise compositions and complex (membrane) structures.
Collapse
Affiliation(s)
- Yang Zhou
- Leibniz-Institut für Polymerforschung Dresden e.V., Hohe Straße 6, Dresden, 01069, Germany
- Organic Chemistry of Polymers, TUD Dresden University of Technology, Dresden, 01062, Germany
| | - Kehu Zhang
- Leibniz-Institut für Polymerforschung Dresden e.V., Hohe Straße 6, Dresden, 01069, Germany
- Organic Chemistry of Polymers, TUD Dresden University of Technology, Dresden, 01062, Germany
| | - Silvia Moreno
- Leibniz-Institut für Polymerforschung Dresden e.V., Hohe Straße 6, Dresden, 01069, Germany
| | - Achim Temme
- Department of Neurosurgery, Section Experimental Neurosurgery/Tumor Immunology, University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, 01307, Germany
- German Cancer Consortium (DKTK), partner site Dresden, Germany, German Cancer Research Center (DKFZ), Heidelberg, Germany, National Center for Tumor Diseases (NCT), Fetscherstraße 74, Dresden, 01307, Germany
| | - Brigitte Voit
- Leibniz-Institut für Polymerforschung Dresden e.V., Hohe Straße 6, Dresden, 01069, Germany
- Organic Chemistry of Polymers, TUD Dresden University of Technology, Dresden, 01062, Germany
| | - Dietmar Appelhans
- Leibniz-Institut für Polymerforschung Dresden e.V., Hohe Straße 6, Dresden, 01069, Germany
| |
Collapse
|
33
|
Thaden O, Schneider N, Walther T, Spiller E, Taoum A, Göpfrich K, Duarte Campos D. Bioprinting of Synthetic Cell-like Lipid Vesicles to Augment the Functionality of Tissues after Manufacturing. ACS Synth Biol 2024; 13:2436-2446. [PMID: 39025476 PMCID: PMC11334175 DOI: 10.1021/acssynbio.4c00137] [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: 02/27/2024] [Revised: 06/27/2024] [Accepted: 06/27/2024] [Indexed: 07/20/2024]
Abstract
Bioprinting is an automated bioassembly method that enables the formation of human tissue-like constructs to restore or replace damaged tissues. Regardless of the employed bioprinting method, cells undergo mechanical stress that can impact their survival and function postprinting. In this study, we investigate the use of a synthetic cell-like unit, giant unilamellar vesicles (GUVs), as adjuvants of the cellular function of human cells postprinting, or in future as the complete replacement of human cells. We analyzed the impact of two nozzle-based bioprinting methods (drop-on-demand and extrusion bioprinting) on the structure, stability, and function of GUVs. We showed that over 65% of the GUVs remain intact when printing at 0.5 bar, demonstrating the potential of using GUVs as a synthetic cell source. We further increased the stability of GUVs in a cell culture medium by introducing polyethylene glycol (PEG) into the GUV lipid membrane. The presence of PEG, however, diminished the structural properties of GUVs postprinting, and reduced the interaction of GUVs with human cells. Although the design of PEG-GUVs can still be modified in future studies for better cell-GUV interactions, we demonstrated that GUVs are functional postprinting. Chlorin e6-PEG-GUVs loaded with a fluorescent dye were bioprinted, and they released the dye postprinting only upon illumination. This is a new strategy to deliver carriers, such as growth factors, drugs, nutrients, or gases, inside large bioprinted specimens on a millimeter to centimeter scale. Overall, we showed that printed GUVs can augment the functionality of manufactured human tissues.
Collapse
Affiliation(s)
- Ole Thaden
- Bioprinting
& Tissue Engineering Group, Center for
Molecular Biology of Heidelberg University (ZMBH), Heidelberg 69120, Germany
| | - Nicole Schneider
- Bioprinting
& Tissue Engineering Group, Center for
Molecular Biology of Heidelberg University (ZMBH), Heidelberg 69120, Germany
| | - Tobias Walther
- Biophysical
Engineering of Life Group, Center for Molecular
Biology of Heidelberg University (ZMBH), Heidelberg 69120, Germany
- Max
Planck Institute for Medical Research, Heidelberg 69120, Germany
| | - Erin Spiller
- Bioprinting
& Tissue Engineering Group, Center for
Molecular Biology of Heidelberg University (ZMBH), Heidelberg 69120, Germany
| | - Alexandre Taoum
- Bioprinting
& Tissue Engineering Group, Center for
Molecular Biology of Heidelberg University (ZMBH), Heidelberg 69120, Germany
| | - Kerstin Göpfrich
- Biophysical
Engineering of Life Group, Center for Molecular
Biology of Heidelberg University (ZMBH), Heidelberg 69120, Germany
- Max
Planck Institute for Medical Research, Heidelberg 69120, Germany
| | - Daniela Duarte Campos
- Bioprinting
& Tissue Engineering Group, Center for
Molecular Biology of Heidelberg University (ZMBH), Heidelberg 69120, Germany
| |
Collapse
|
34
|
Wei Y, Weng X, Wang Y, Yang W. Stimuli-Responsive Polymersomes: Reshaping the Immunosuppressive Tumor Microenvironment. Biomacromolecules 2024; 25:4663-4676. [PMID: 39054960 DOI: 10.1021/acs.biomac.4c00706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2024]
Abstract
The progression of cancer involves mutations in normal cells, leading to uncontrolled division and tissue destruction, highlighting the complexity of tumor microenvironments (TMEs). Immunotherapy has emerged as a transformative approach, yet the balance between efficacy and safety remains a challenge. Nanoparticles such as polymersomes offer the possibility to precisely target tumors, deliver drugs in a controlled way, effectively modulate the antitumor immunity, and notably reduce side effects. Herein, stimuli-responsive polymersomes, with capabilities for carrying multiple therapeutics, are highlighted for their potential in enhancing antitumor immunity through mechanisms like inducing immunogenic cell death and activating STING (stimulator of interferon genes), etc. The recent progress of utilizing stimuli-responsive polymersomes to reshape the TME is reviewed here. The advantages and limitations to applied stimuli-responsive polymersomes are outlined. Additionally, challenges and future prospects in leveraging polymersomes for cancer therapy are discussed, emphasizing the need for future research and clinical translation.
Collapse
Affiliation(s)
- Yaohua Wei
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Xiao Weng
- School of Pharmaceutical Sciences, Henan Key Laboratory of Nanomedicine for Targeting Diagnosis and Treatment, Zhengzhou University, Zhengzhou, Henan 450001 China
| | - Yayun Wang
- School of Pharmaceutical Sciences, Henan Key Laboratory of Nanomedicine for Targeting Diagnosis and Treatment, Zhengzhou University, Zhengzhou, Henan 450001 China
| | - Weijing Yang
- School of Pharmaceutical Sciences, Henan Key Laboratory of Nanomedicine for Targeting Diagnosis and Treatment, Zhengzhou University, Zhengzhou, Henan 450001 China
| |
Collapse
|
35
|
Skowicki M, Tarvirdipour S, Kraus M, Schoenenberger CA, Palivan CG. Nanoassemblies designed for efficient nuclear targeting. Adv Drug Deliv Rev 2024; 211:115354. [PMID: 38857762 DOI: 10.1016/j.addr.2024.115354] [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: 04/12/2024] [Revised: 05/23/2024] [Accepted: 06/04/2024] [Indexed: 06/12/2024]
Abstract
One of the key aspects of coping efficiently with complex pathological conditions is delivering the desired therapeutic compounds with precision in both space and time. Therefore, the focus on nuclear-targeted delivery systems has emerged as a promising strategy with high potential, particularly in gene therapy and cancer treatment. Here, we explore the design of supramolecular nanoassemblies as vehicles to deliver specific compounds to the nucleus, with the special focus on polymer and peptide-based carriers that expose nuclear localization signals. Such nanoassemblies aim at maximizing the concentration of genetic and therapeutic agents within the nucleus, thereby optimizing treatment outcomes while minimizing off-target effects. A complex scenario of conditions, including cellular uptake, endosomal escape, and nuclear translocation, requires fine tuning of the nanocarriers' properties. First, we introduce the principles of nuclear import and the role of nuclear pore complexes that reveal strategies for targeting nanosystems to the nucleus. Then, we provide an overview of cargoes that rely on nuclear localization for optimal activity as their integrity and accumulation are crucial parameters to consider when designing a suitable delivery system. Considering that they are in their early stages of research, we present various cargo-loaded peptide- and polymer nanoassemblies that promote nuclear targeting, emphasizing their potential to enhance therapeutic response. Finally, we briefly discuss further advancements for more precise and effective nuclear delivery.
Collapse
Affiliation(s)
- Michal Skowicki
- Department of Chemistry, University of Basel, BPR 1096, Mattenstrasse 22, 4058 Basel, Switzerland; NCCR-Molecular Systems Engineering, BPR 1095, Mattenstrasse 24a, 4058 Basel, Switzerland
| | - Shabnam Tarvirdipour
- Department of Chemistry, University of Basel, BPR 1096, Mattenstrasse 22, 4058 Basel, Switzerland
| | - Manuel Kraus
- Department of Chemistry, University of Basel, BPR 1096, Mattenstrasse 22, 4058 Basel, Switzerland
| | - Cora-Ann Schoenenberger
- Department of Chemistry, University of Basel, BPR 1096, Mattenstrasse 22, 4058 Basel, Switzerland; NCCR-Molecular Systems Engineering, BPR 1095, Mattenstrasse 24a, 4058 Basel, Switzerland.
| | - Cornelia G Palivan
- Department of Chemistry, University of Basel, BPR 1096, Mattenstrasse 22, 4058 Basel, Switzerland; NCCR-Molecular Systems Engineering, BPR 1095, Mattenstrasse 24a, 4058 Basel, Switzerland.
| |
Collapse
|
36
|
Ngocho K, Yang X, Wang Z, Hu C, Yang X, Shi H, Wang K, Liu J. Synthetic Cells from Droplet-Based Microfluidics for Biosensing and Biomedical Applications. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2400086. [PMID: 38563581 DOI: 10.1002/smll.202400086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 03/13/2024] [Indexed: 04/04/2024]
Abstract
Synthetic cells function as biological mimics of natural cells by mimicking salient features of cells such as metabolism, response to stimuli, gene expression, direct metabolism, and high stability. Droplet-based microfluidic technology presents the opportunity for encapsulating biological functional components in uni-lamellar liposome or polymer droplets. Verified by its success in the fabrication of synthetic cells, microfluidic technology is widely replacing conventional labor-intensive, expensive, and sophisticated techniques justified by its ability to miniaturize and perform batch production operations. In this review, an overview of recent research on the preparation of synthetic cells through droplet-based microfluidics is provided. Different synthetic cells including lipid vesicles (liposome), polymer vesicles (polymersome), coacervate microdroplets, and colloidosomes, are systematically discussed. Efforts are then made to discuss the design of a variety of microfluidic chips for synthetic cell preparation since the combination of microfluidics with bottom-up synthetic biology allows for reproductive and tunable construction of batches of synthetic cell models from simple structures to higher hierarchical structures. The recent advances aimed at exploiting them in biosensors and other biomedical applications are then discussed. Finally, some perspectives on the challenges and future developments of synthetic cell research with microfluidics for biomimetic science and biomedical applications are provided.
Collapse
Affiliation(s)
- Kleins Ngocho
- State key laboratory of Chemo/Biosensing and Chemometrics College of Chemistry and Chemical Engineering Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University, Changsha, 410082, P. R. China
| | - Xilei Yang
- State key laboratory of Chemo/Biosensing and Chemometrics College of Chemistry and Chemical Engineering Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University, Changsha, 410082, P. R. China
| | - Zefeng Wang
- State key laboratory of Chemo/Biosensing and Chemometrics College of Chemistry and Chemical Engineering Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University, Changsha, 410082, P. R. China
| | - Cunjie Hu
- State key laboratory of Chemo/Biosensing and Chemometrics College of Chemistry and Chemical Engineering Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University, Changsha, 410082, P. R. China
| | - Xiaohai Yang
- State key laboratory of Chemo/Biosensing and Chemometrics College of Chemistry and Chemical Engineering Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University, Changsha, 410082, P. R. China
| | - Hui Shi
- State key laboratory of Chemo/Biosensing and Chemometrics College of Chemistry and Chemical Engineering Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University, Changsha, 410082, P. R. China
| | - Kemin Wang
- State key laboratory of Chemo/Biosensing and Chemometrics College of Chemistry and Chemical Engineering Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University, Changsha, 410082, P. R. China
| | - Jianbo Liu
- State key laboratory of Chemo/Biosensing and Chemometrics College of Chemistry and Chemical Engineering Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University, Changsha, 410082, P. R. China
| |
Collapse
|
37
|
Bouazzaoui A, Abdellatif AA. Vaccine delivery systems and administration routes: Advanced biotechnological techniques to improve the immunization efficacy. Vaccine X 2024; 19:100500. [PMID: 38873639 PMCID: PMC11170481 DOI: 10.1016/j.jvacx.2024.100500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 03/21/2024] [Accepted: 05/14/2024] [Indexed: 06/15/2024] Open
Abstract
Since the first use of vaccine tell the last COVID-19 pandemic caused by spread of SARS-CoV-2 worldwide, the use of advanced biotechnological techniques has accelerated the development of different types and methods for immunization. The last pandemic showed that the nucleic acid-based vaccine, especially mRNA, has an advantage in terms of development time; however, it showed a very critical drawback namely, the higher costs when compared to other strategies, and its inability to protect against new variants. This showed the need of more improvement to reach a better delivery and efficacy. In this review we will describe different vaccine delivery systems including, the most used viral vector, and also variable strategies for delivering of nucleic acid-based vaccines especially lipid-based nanoparticles formulation, polymersomes, electroporation and also the new powerful tools for the delivery of mRNA, which is based on the use of cell-penetrating peptides (CPPs). Additionally, we will also discuss the main challenges associated with each system. Finlay, the efficacy and safety of the vaccines depends not only on the formulations and delivery systems, but also the dosage and route of administration are also important players, therefore we will see the different routes for the vaccine administration including traditionally routes (intramuscular, Transdermal, subcutaneous), oral inhalation or via nasal mucosa, and will describe the advantages and disadvantage of each administration route.
Collapse
Affiliation(s)
- Abdellatif Bouazzaoui
- Department of Medical Genetics, Faculty of Medicine, Umm Al-Qura University, P.O. Box 715, Makkah 21955, Saudi Arabia
- Science and Technology Unit, Umm Al Qura University, P.O. Box 715, Makkah 21955, Saudi Arabia
| | - Ahmed A.H. Abdellatif
- Department of Pharmaceutics, College of Pharmacy, Qassim University, 51452 Qassim, Saudi Arabia
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Al-Azhar University, 71524 Assiut, Egypt
| |
Collapse
|
38
|
Ghosh R, Singh B, Basu S, Mondal A, Maiti PK, De M. Reversing the Trend: Deciphering Self-Assembly of Unconventional Amphiphiles Having Both Alkyl-Chain and PEG. Chempluschem 2024; 89:e202400147. [PMID: 38623044 DOI: 10.1002/cplu.202400147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 04/15/2024] [Accepted: 04/15/2024] [Indexed: 04/17/2024]
Abstract
In the field of molecular self-assembly, the core of an assembly is always made up of hydrophobic moiety like a long alkyl chain, whereas the outer part has always been a hydrophilic moiety such as poly(ethylene glycol) (PEG), or charged species. Hence, reversing the trend to manifest self-assembled structures with a PEG core and a surface consisting of alkyl chains in aqueous system is incredibly challenging. Herein, we architected a unique class of cationic bolaamphiphiles containing low molecular weight PEG and alkyl chains of different lengths. The bolaamphiphiles spontaneously form vesicles without external stimuli. These vesicles are unprecedented because PEG makes up the vesicle core, while the alkyl chains appear on the vesicles' exterior. Hence, this particular design reverses the usual trend of self-assembly formation. The vesicle size increases with the increase in alkyl chain-length. To our great surprise, we obtained large micelles for longest alkyl-chain amphiphile, which in turn act as a gemini amphiphile. The shift from a particular bolaamphiphile to gemini amphiphile with the variation of alkyl chain is also unexplored. Therefore, this specific class of self-assembled structure would compound a new paradigm in molecular self-assembly and supramolecular chemistry.
Collapse
Affiliation(s)
- Rita Ghosh
- Department of Organic Chemistry, Indian Institute of Science, Bengaluru, 560012, India
| | - Bharat Singh
- Department of Physics, Indian Institute of Science, Bengaluru, 560012, India
| | - Subhadip Basu
- Department of Physics, Indian Institute of Science, Bengaluru, 560012, India
| | - Avijit Mondal
- Department of Organic Chemistry, Indian Institute of Science, Bengaluru, 560012, India
| | - Prabal Kumar Maiti
- Department of Physics, Indian Institute of Science, Bengaluru, 560012, India
| | - Mrinmoy De
- Department of Organic Chemistry, Indian Institute of Science, Bengaluru, 560012, India
| |
Collapse
|
39
|
Wang Y, Zhang C, Cheng J, Yan T, He Q, Huang D, Liu J, Wang Z. Cutting-Edge Biomaterials in Intervertebral Disc Degeneration Tissue Engineering. Pharmaceutics 2024; 16:979. [PMID: 39204324 PMCID: PMC11359550 DOI: 10.3390/pharmaceutics16080979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/12/2024] [Accepted: 07/18/2024] [Indexed: 09/04/2024] Open
Abstract
Intervertebral disc degeneration (IVDD) stands as the foremost contributor to low back pain (LBP), imposing a substantial weight on the world economy. Traditional treatment modalities encompass both conservative approaches and surgical interventions; however, the former falls short in halting IVDD progression, while the latter carries inherent risks. Hence, the quest for an efficacious method to reverse IVDD onset is paramount. Biomaterial delivery systems, exemplified by hydrogels, microspheres, and microneedles, renowned for their exceptional biocompatibility, biodegradability, biological efficacy, and mechanical attributes, have found widespread application in bone, cartilage, and various tissue engineering endeavors. Consequently, IVD tissue engineering has emerged as a burgeoning field of interest. This paper succinctly introduces the intervertebral disc (IVD) structure and the pathophysiology of IVDD, meticulously classifies biomaterials for IVD repair, and reviews recent advances in the field. Particularly, the strengths and weaknesses of biomaterials in IVD tissue engineering are emphasized, and potential avenues for future research are suggested.
Collapse
Affiliation(s)
- Yifan Wang
- Department of Orthopedics, Chinese PLA General Hospital, Beijing 100853, China; (Y.W.); (C.Z.); (J.C.); (T.Y.)
| | - Chuyue Zhang
- Department of Orthopedics, Chinese PLA General Hospital, Beijing 100853, China; (Y.W.); (C.Z.); (J.C.); (T.Y.)
| | - Junyao Cheng
- Department of Orthopedics, Chinese PLA General Hospital, Beijing 100853, China; (Y.W.); (C.Z.); (J.C.); (T.Y.)
| | - Taoxu Yan
- Department of Orthopedics, Chinese PLA General Hospital, Beijing 100853, China; (Y.W.); (C.Z.); (J.C.); (T.Y.)
| | - Qing He
- College of Biological Science and Engineering, Fuzhou University, Fuzhou 350108, China; (Q.H.); (D.H.)
| | - Da Huang
- College of Biological Science and Engineering, Fuzhou University, Fuzhou 350108, China; (Q.H.); (D.H.)
| | - Jianheng Liu
- Department of Orthopedics, Chinese PLA General Hospital, Beijing 100853, China; (Y.W.); (C.Z.); (J.C.); (T.Y.)
| | - Zheng Wang
- Department of Orthopedics, Chinese PLA General Hospital, Beijing 100853, China; (Y.W.); (C.Z.); (J.C.); (T.Y.)
| |
Collapse
|
40
|
Liu F, Qu P, Weiss J, Guo K, Weck M. Substrate Channeling in Compartmentalized Nanoreactors. Macromolecules 2024; 57:6805-6815. [PMID: 39071043 PMCID: PMC11270995 DOI: 10.1021/acs.macromol.4c00697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 05/28/2024] [Accepted: 06/18/2024] [Indexed: 07/30/2024]
Abstract
Thermo- and photoresponsive nanoreactors based on shell cross-linked micelles (SCMs) for the rhodium-catalyzed asymmetric transfer hydrogenation (ATH) of ketones have been developed from poly(2-oxazoline) triblock terpolymers. The nanoreactors incorporate thermoresponsive poly(2-isopropyl-2-oxazoline) as the hydrophilic corona and are covalently cross-linked with a photoswitchable spiropyran molecule. UV irradiation or changes in temperature trigger morphology switching of the polymer-based nanoreactors that alters the hydrophobicity in separate layers of the SCMs, resulting in dynamic substrate selectivity of the ATH in water. Control experiments and kinetic studies show that the thermoresponsive outer layer induces the gated behavior for more hydrophobic substrates, whereas the photoresponsive cross-linking layer induces the gated behavior for less hydrophobic substrates. The nanoreactors mimic the multichannels in Nature, transporting substrates and reagents into the catalytic core which can be controlled through external triggers such as temperature and light wavelengths. Additionally, the nanoreactors can be easily recovered and reused with continued high activity and selectivities.
Collapse
Affiliation(s)
- Fangbei Liu
- Molecular Design Institute and Department
of Chemistry, New York University, New York, New York 10003-6688, United
States
| | | | - Jeremy Weiss
- Molecular Design Institute and Department
of Chemistry, New York University, New York, New York 10003-6688, United
States
| | - Kunhao Guo
- Molecular Design Institute and Department
of Chemistry, New York University, New York, New York 10003-6688, United
States
| | - Marcus Weck
- Molecular Design Institute and Department
of Chemistry, New York University, New York, New York 10003-6688, United
States
| |
Collapse
|
41
|
Fielden SDP. Kinetically Controlled and Nonequilibrium Assembly of Block Copolymers in Solution. J Am Chem Soc 2024; 146:18781-18796. [PMID: 38967256 PMCID: PMC11258791 DOI: 10.1021/jacs.4c03314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 06/21/2024] [Accepted: 06/24/2024] [Indexed: 07/06/2024]
Abstract
Covalent polymers are versatile macromolecules that have found widespread use in society. Contemporary methods of polymerization have made it possible to construct sequence polymers, including block copolymers, with high precision. Such copolymers assemble in solution when the blocks have differing solubilities. This produces nano- and microparticles of various shapes and sizes. While it is straightforward to draw an analogy between such amphiphilic block copolymers and phospholipids, these two classes of molecules show quite different assembly characteristics. In particular, block copolymers often assemble under kinetic control, thus producing nonequilibrium structures. This leads to a rich variety of behaviors being observed in block copolymer assembly, such as pathway dependence (e.g., thermal history), nonergodicity and responsiveness. The dynamics of polymer assemblies can be readily controlled using changes in environmental conditions and/or integrating functional groups situated on polymers with external chemical reactions. This perspective highlights that kinetic control is both pervasive and a useful attribute in the mechanics of block copolymer assembly. Recent examples are highlighted in order to show that toggling between static and dynamic behavior can be used to generate, manipulate and dismantle nonequilibrium states. New methods to control the kinetics of block copolymer assembly will provide endless unanticipated applications in materials science, biomimicry and medicine.
Collapse
Affiliation(s)
- Stephen D. P. Fielden
- School of Chemistry, University of Birmingham, Edgbaston, Birmingham B15 2TT, United
Kingdom
| |
Collapse
|
42
|
Brodszkij E, Städler B. Advances in block copolymer-phospholipid hybrid vesicles: from physical-chemical properties to applications. Chem Sci 2024; 15:10724-10744. [PMID: 39027291 PMCID: PMC11253165 DOI: 10.1039/d4sc01444h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 06/23/2024] [Indexed: 07/20/2024] Open
Abstract
Hybrid vesicles, made of lipids and amphiphilic block copolymers, have become increasingly popular thanks to their versatile properties that enable the construction of intricate membranes mimicking cellular structures. This tutorial review gives an overview over the different hybrid vesicle designs, and provides a detailed analysis of their properties, including their composition, membrane fluidity, membrane homogeneity, permeability, stability. The review puts emphasis on the application of these hybrid vesicles in bottom-up synthetic biology and aims to offer an overview of design guidelines, particularly focusing on composition, to eventually realize the intended applications of these hybrid vesicles.
Collapse
Affiliation(s)
- Edit Brodszkij
- Interdisciplinary Nanoscience Center (iNANO) Aarhus University Gustave Wieds Vej 14 8000 Aarhus C Denmark
| | - Brigitte Städler
- Interdisciplinary Nanoscience Center (iNANO) Aarhus University Gustave Wieds Vej 14 8000 Aarhus C Denmark
| |
Collapse
|
43
|
You J, Qian Y, Xiong S, Zhang P, Mukwaya V, Levi-Kalisman Y, Raviv U, Dou H. Poly(ferrocenylsilane)-Based Redox-Active Artificial Organelles for Biomimetic Cascade Reactions. Chemistry 2024; 30:e202401435. [PMID: 38739532 DOI: 10.1002/chem.202401435] [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: 04/13/2024] [Revised: 05/09/2024] [Accepted: 05/11/2024] [Indexed: 05/16/2024]
Abstract
Artificial organelles serve as functional counterparts to natural organelles, which are primarily employed to artificially replicate, restore, or enhance cellular functions. While most artificial organelles exhibit basic functions, we diverge from this norm by utilizing poly(ferrocenylmethylethylthiocarboxypropylsilane) microcapsules (PFC MCs) to construct multifunctional artificial organelles through water/oil interfacial self-assembly. Within these PFC MCs, enzymatic cascades are induced through active molecular exchange across the membrane to mimic the functions of enzymes in mitochondria. We harness the inherent redox properties of the PFC polymer, which forms the membrane, to facilitate in-situ redox reactions similar to those supported by the inner membrane of natural mitochondria. Subsequent studies have demonstrated the interaction between PFC MCs and living cell including extended lifespans within various cell types. We anticipate that functional PFC MCs have the potential to serve as innovative platforms for organelle mimics capable of executing specific cellular functions.
Collapse
Affiliation(s)
- Jiayi You
- The State Key Laboratory of Metal Matrix Composites, School of Materials Science and Engineering., Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Yonghui Qian
- The State Key Laboratory of Metal Matrix Composites, School of Materials Science and Engineering., Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Shuhan Xiong
- The State Key Laboratory of Metal Matrix Composites, School of Materials Science and Engineering., Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Peipei Zhang
- The State Key Laboratory of Metal Matrix Composites, School of Materials Science and Engineering., Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Vincent Mukwaya
- The State Key Laboratory of Metal Matrix Composites, School of Materials Science and Engineering., Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Yael Levi-Kalisman
- Institute of Life Sciences and the Center for Nanoscience and Nanotechnology, The Hebrew University of Jerusalem, Edmond J. Safra Campus, Givat Ram, 9190401, Jerusalem, Israel
| | - Uri Raviv
- Institute of Chemistry and the Center for Nanoscience and Nanotechnology, The Hebrew University of Jerusalem, Edmond J. Safra Campus, Givat Ram, 9190401, Jerusalem, Israel
| | - Hongjing Dou
- The State Key Laboratory of Metal Matrix Composites, School of Materials Science and Engineering., Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| |
Collapse
|
44
|
Koball A, Obst F, Gaitzsch J, Voit B, Appelhans D. Boosting Microfluidic Enzymatic Cascade Reactions with pH-Responsive Polymersomes by Spatio-Chemical Activity Control. SMALL METHODS 2024:e2400282. [PMID: 38989686 DOI: 10.1002/smtd.202400282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 06/26/2024] [Indexed: 07/12/2024]
Abstract
Microfluidic flow reactors permit the implementation of sensitive biocatalysts in polymeric environments (e.g., hydrogel dots), mimicking nature through the use of diverse microstructures within defined confinements. However, establishing complex hybrid structures to mimic biological processes and functions under continuous flow with optimal utilization of all components involved in the reaction process represents a significant scientific challenge. To achieve spatial, chemical, and temporal control for any microfluidic application, compartmentalization is required, as well as the unification of different sensitive compartments in the reaction chamber for the microfluidic flow design. This study presents a self-regulating microfluidic system fabricated by a sequential photostructuring process with an intermediate chemical process step to realize pH-sensitive hybrid structures for the fabrication of a microfluidic double chamber reactor for controlled enzymatic cascade reaction (ECR). The key point is the adaptation and retention of the function of pH-responsive horseradish peroxidase-loaded polymersomes in a microfluidic chip under continuous flow. ECR is successfully triggered and controlled by an interplay between glucose oxidase-converted glucose, the membrane state of pH-responsive polymersomes, and other parameters (e.g., flow rate and fluid composition). This study establishes a promising noninvasive regulatory platform for extended spatio-chemical control of current and future ECR and other cascade reaction systems.
Collapse
Affiliation(s)
- Andrea Koball
- Leibniz-Institut für Polymerforschung Dresden e.V., Hohe Straße 6, D-01069, Dresden, Germany
- Technische Universität Dresden, Fakultät Chemie und Lebensmittelchemie, Organische Chemie der Polymere, D-01062, Dresden, Germany
| | - Franziska Obst
- Leibniz-Institut für Polymerforschung Dresden e.V., Hohe Straße 6, D-01069, Dresden, Germany
- Technische Universität Dresden, Institut für Halbleiter- und Mikrosystemtechnik, Nöthnitzer Straße 64, D-01187, Dresden, Germany
| | - Jens Gaitzsch
- Leibniz-Institut für Polymerforschung Dresden e.V., Hohe Straße 6, D-01069, Dresden, Germany
| | - Brigitte Voit
- Leibniz-Institut für Polymerforschung Dresden e.V., Hohe Straße 6, D-01069, Dresden, Germany
- Technische Universität Dresden, Fakultät Chemie und Lebensmittelchemie, Organische Chemie der Polymere, D-01062, Dresden, Germany
| | - Dietmar Appelhans
- Leibniz-Institut für Polymerforschung Dresden e.V., Hohe Straße 6, D-01069, Dresden, Germany
| |
Collapse
|
45
|
Jäger E, Černoch P, Vragovic M, Calumby Albuquerque LJ, Sincari V, Heizer T, Jäger A, Kučka J, Janoušková OŠ, Pavlova E, Šefc L, Giacomelli FC. Membrane Permeability and Responsiveness Drive Performance: Linking Structural Features with the Antitumor Effectiveness of Doxorubicin-Loaded Stimuli-Triggered Polymersomes. Biomacromolecules 2024; 25:4192-4202. [PMID: 38917475 PMCID: PMC11238342 DOI: 10.1021/acs.biomac.4c00282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 06/07/2024] [Accepted: 06/10/2024] [Indexed: 06/27/2024]
Abstract
The permeability and responsiveness of polymer membranes are absolutely relevant in the design of polymersomes for cargo delivery. Accordingly, we herein correlate the structural features, permeability, and responsiveness of doxorubicin-loaded (DOX-loaded) nonresponsive and stimuli-responsive polymersomes with their in vitro and in vivo antitumor performance. Polymer vesicles were produced using amphiphilic block copolymers containing a hydrophilic poly[N-(2-hydroxypropyl)methacrylamide] (PHPMA) segment linked to poly[N-(4-isopropylphenylacetamide)ethyl methacrylate] (PPPhA, nonresponsive block), poly[4-(4,4,5,5-tetra-methyl-1,3,2-dioxaborolan-2-yl)benzyl methacrylate] [PbAPE, reactive oxygen species (ROS)-responsive block], or poly[2-(diisopropylamino)ethyl methacrylate] (PDPA, pH-responsive block). The PDPA-based polymersomes demonstrated outstanding biological performance with antitumor activity notably enhanced compared to their counterparts. We attribute this behavior to a fast-triggered DOX release in acidic tumor environments as induced by pH-responsive polymersome disassembly at pH < 6.8. Possibly, an insufficient ROS concentration in the selected tumor model attenuates the rate of ROS-responsive vesicle degradation, whereas the nonresponsive nature of the PPPhA block remarkably impacts the performance of such potential nanomedicines.
Collapse
Affiliation(s)
- Eliézer Jäger
- Institute
of Macromolecular Chemistry, Czech Academy
of Sciences, Prague 162 00, Czech Republic
| | - Peter Černoch
- Institute
of Macromolecular Chemistry, Czech Academy
of Sciences, Prague 162 00, Czech Republic
| | - Martina Vragovic
- Institute
of Macromolecular Chemistry, Czech Academy
of Sciences, Prague 162 00, Czech Republic
| | - Lindomar Jose Calumby Albuquerque
- Institute
of Macromolecular Chemistry, Czech Academy
of Sciences, Prague 162 00, Czech Republic
- Centro
de Ciências Naturais e Humanas, Universidade
Federal do ABC, Santo Andre 09280-560, Brazil
| | - Vladimir Sincari
- Institute
of Macromolecular Chemistry, Czech Academy
of Sciences, Prague 162 00, Czech Republic
| | - Tomáš Heizer
- Center
for Advanced Preclinical Imaging (CAPI), First Faculty of Medicine, Charles University, Prague 120 00, Czech Republic
| | - Alessandro Jäger
- Institute
of Macromolecular Chemistry, Czech Academy
of Sciences, Prague 162 00, Czech Republic
| | - Jan Kučka
- Institute
of Macromolecular Chemistry, Czech Academy
of Sciences, Prague 162 00, Czech Republic
| | | | - Ewa Pavlova
- Institute
of Macromolecular Chemistry, Czech Academy
of Sciences, Prague 162 00, Czech Republic
| | - Luděk Šefc
- Center
for Advanced Preclinical Imaging (CAPI), First Faculty of Medicine, Charles University, Prague 120 00, Czech Republic
| | | |
Collapse
|
46
|
Holkar A, Gao S, Villaseñor K, Lake M, Srivastava S. Quantitative turbidimetric characterization of stabilized complex coacervate dispersions. SOFT MATTER 2024; 20:5060-5070. [PMID: 38743276 DOI: 10.1039/d3sm01761c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Stabilizing complex coacervate microdroplets is desirable due to their various applications, such as bioreactors, drug delivery vehicles, and encapsulants. Here, we present quantitative characterization of complex coacervate dispersion stability inferred by turbidimetry measurements. The stability of the dispersions is shown to be modulated by the concentrations of comb polyelectrolyte (cPE) stabilizers and salt. We demonstrate cPEs as effective stabilizers for complex coacervate dispersions independent of the chemistry or length of the constituent polyelectrolytes, salts, or preparation routes. By monitoring the temporal evolution of dispersion turbidity, we show that cPEs suppress microdroplet coalescence with minimal change in microdroplet sizes over 48 hours, even at salt concentrations up to 300 mM. The number density and average microdroplet size are shown to be controlled by varying the cPE and salt concentrations. Lastly, turbidity maps, akin to binodal phase maps, depict an expansion of the turbid two-phase region and an increase in the salt resistance of the coacervates upon the introduction of cPEs. The coacervate salt resistance is shown to increase by >3×, and this increase is maintained for up to 15 days, demonstrating that cPEs impart higher salt resistance over extended durations.
Collapse
Affiliation(s)
- Advait Holkar
- Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - Shang Gao
- Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - Kathleen Villaseñor
- Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - Michael Lake
- NSF BioPACIFIC MIP, University of California, Los Angeles, Los Angeles, CA 90095, USA
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Samanvaya Srivastava
- Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, CA 90095, USA.
- NSF BioPACIFIC MIP, University of California, Los Angeles, Los Angeles, CA 90095, USA
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Institute for Carbon Management, University of California, Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
47
|
Chang L, Cui H, Li F, Job Zhang YHP, Zhang L. ATP regeneration by ATPases for in vitro biotransformation. Biotechnol Adv 2024; 73:108377. [PMID: 38763231 DOI: 10.1016/j.biotechadv.2024.108377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 04/10/2024] [Accepted: 05/12/2024] [Indexed: 05/21/2024]
Abstract
Adenosine triphosphate (ATP) regeneration is a significant step in both living cells and in vitro biotransformation (ivBT). Rotary motor ATP synthases (ATPases), which regenerate ATP in living cells, have been widely assembled in biomimetic structures for in vitro ATP synthesis. In this review, we present a comprehensive overview of ATPases, including the working principle, orientation and distribution density properties of ATPases, as well as the assembly strategies and applications of ATPase-based ATP regeneration modules. The original sources of ATPases for in vitro ATP regeneration include chromatophores, chloroplasts, mitochondria, and inverted Escherichia coli (E. coli) vesicles, which are readily accessible but unstable. Although significant advances have been made in the assembly methods for ATPase-artificial membranes in recent decades, it remains challenging to replicate the high density and orientation of ATPases observed in vivo using in vitro assembly methods. The use of bioproton pumps or chemicals for constructing proton motive forces (PMF) enables the versatility and potential of ATPase-based ATP regeneration modules. Additionally, overall robustness can be achieved via membrane component selection, such as polymers offering great mechanical stability, or by constructing a solid supporting matrix through layer-by-layer assembly techniques. Finally, the prospects of ATPase-based ATP regeneration modules can be expected with the technological development of ATPases and artificial membranes.
Collapse
Affiliation(s)
- Lijing Chang
- Key Laboratory of Engineering Biology for Low-Carbon Manufacturing, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, PR China; In vitro Synthetic Biology Center, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, PR China
| | - Huijuan Cui
- Key Laboratory of Engineering Biology for Low-Carbon Manufacturing, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, PR China; In vitro Synthetic Biology Center, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, PR China
| | - Fei Li
- Key Laboratory of Engineering Biology for Low-Carbon Manufacturing, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, PR China; In vitro Synthetic Biology Center, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, PR China
| | - Yi-Heng P Job Zhang
- Key Laboratory of Engineering Biology for Low-Carbon Manufacturing, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, PR China; In vitro Synthetic Biology Center, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Lingling Zhang
- Key Laboratory of Engineering Biology for Low-Carbon Manufacturing, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, PR China; In vitro Synthetic Biology Center, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China.
| |
Collapse
|
48
|
Yildiz SN, Entezari M, Paskeh MDA, Mirzaei S, Kalbasi A, Zabolian A, Hashemi F, Hushmandi K, Hashemi M, Raei M, Goharrizi MASB, Aref AR, Zarrabi A, Ren J, Orive G, Rabiee N, Ertas YN. Nanoliposomes as nonviral vectors in cancer gene therapy. MedComm (Beijing) 2024; 5:e583. [PMID: 38919334 PMCID: PMC11199024 DOI: 10.1002/mco2.583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 04/19/2024] [Accepted: 04/26/2024] [Indexed: 06/27/2024] Open
Abstract
Nonviral vectors, such as liposomes, offer potential for targeted gene delivery in cancer therapy. Liposomes, composed of phospholipid vesicles, have demonstrated efficacy as nanocarriers for genetic tools, addressing the limitations of off-targeting and degradation commonly associated with traditional gene therapy approaches. Due to their biocompatibility, stability, and tunable physicochemical properties, they offer potential in overcoming the challenges associated with gene therapy, such as low transfection efficiency and poor stability in biological fluids. Despite these advancements, there remains a gap in understanding the optimal utilization of nanoliposomes for enhanced gene delivery in cancer treatment. This review delves into the present state of nanoliposomes as carriers for genetic tools in cancer therapy, sheds light on their potential to safeguard genetic payloads and facilitate cell internalization alongside the evolution of smart nanocarriers for targeted delivery. The challenges linked to their biocompatibility and the factors that restrict their effectiveness in gene delivery are also discussed along with exploring the potential of nanoliposomes in cancer gene therapy strategies by analyzing recent advancements and offering future directions.
Collapse
Affiliation(s)
| | - Maliheh Entezari
- Department of GeneticsFaculty of Advanced Science and TechnologyTehran Medical SciencesIslamic Azad UniversityTehranIran
- Department of Medical Convergence SciencesFarhikhtegan Hospital Tehran Medical SciencesIslamic Azad UniversityTehranIran
| | - Mahshid Deldar Abad Paskeh
- Department of GeneticsFaculty of Advanced Science and TechnologyTehran Medical SciencesIslamic Azad UniversityTehranIran
- Department of Medical Convergence SciencesFarhikhtegan Hospital Tehran Medical SciencesIslamic Azad UniversityTehranIran
| | - Sepideh Mirzaei
- Department of BiologyFaculty of ScienceIslamic Azad UniversityScience and Research BranchTehranIran
| | - Alireza Kalbasi
- Department of PharmacyBrigham and Women's HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - Amirhossein Zabolian
- Department of OrthopedicsShahid Beheshti University of Medical SciencesTehranIran
| | - Farid Hashemi
- Department of Comparative BiosciencesFaculty of Veterinary MedicineUniversity of TehranTehranIran
| | - Kiavash Hushmandi
- Department of Clinical Sciences InstituteNephrology and Urology Research CenterBaqiyatallah University of Medical SciencesTehranIran
| | - Mehrdad Hashemi
- Department of GeneticsFaculty of Advanced Science and TechnologyTehran Medical SciencesIslamic Azad UniversityTehranIran
- Department of Medical Convergence SciencesFarhikhtegan Hospital Tehran Medical SciencesIslamic Azad UniversityTehranIran
| | - Mehdi Raei
- Department of Epidemiology and BiostatisticsSchool of HealthBaqiyatallah University of Medical SciencesTehranIran
| | | | - Amir Reza Aref
- Belfer Center for Applied Cancer ScienceDana‐Farber Cancer InstituteHarvard Medical SchoolBostonMassachusettsUSA
- Department of Translational SciencesXsphera Biosciences Inc.BostonMassachusettsUSA
| | - Ali Zarrabi
- Department of Biomedical EngineeringFaculty of Engineering and Natural SciencesIstinye UniversityIstanbulTurkey
| | - Jun Ren
- Shanghai Institute of Cardiovascular DiseasesDepartment of CardiologyZhongshan HospitalFudan UniversityShanghaiChina
| | - Gorka Orive
- NanoBioCel Research GroupSchool of PharmacyUniversity of the Basque Country (UPV/EHU)Vitoria‐GasteizSpain
- University Institute for Regenerative Medicine and Oral Implantology ‐ UIRMI (UPV/EHU‐Fundación Eduardo Anitua)Vitoria‐GasteizSpain
- Bioaraba, NanoBioCel Research GroupVitoria‐GasteizSpain
- The AcademiaSingapore Eye Research InstituteSingaporeSingapore
| | - Navid Rabiee
- Centre for Molecular Medicine and Innovative TherapeuticsMurdoch UniversityPerthWestern AustraliaAustralia
| | - Yavuz Nuri Ertas
- Department of Biomedical EngineeringErciyes UniversityKayseriTurkey
- ERNAM—Nanotechnology Research and Application CenterErciyes UniversityKayseriTurkey
- UNAM−National Nanotechnology Research CenterBilkent UniversityAnkaraTurkey
| |
Collapse
|
49
|
Ramezani P, De Smedt SC, Sauvage F. Supramolecular dye nanoassemblies for advanced diagnostics and therapies. Bioeng Transl Med 2024; 9:e10652. [PMID: 39036081 PMCID: PMC11256156 DOI: 10.1002/btm2.10652] [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: 09/12/2023] [Revised: 01/09/2024] [Accepted: 01/19/2024] [Indexed: 07/23/2024] Open
Abstract
Dyes have conventionally been used in medicine for staining cells, tissues, and organelles. Since these compounds are also known as photosensitizers (PSs) which exhibit photoresponsivity upon photon illumination, there is a high desire towards formulating these molecules into nanoparticles (NPs) to achieve improved delivery efficiency and enhanced stability for novel imaging and therapeutic applications. Furthermore, it has been shown that some of the photophysical properties of these molecules can be altered upon NP formation thereby playing a major role in the outcome of their application. In this review, we primarily focus on introducing dye categories, their formulation strategies and how these strategies affect their photophysical properties in the context of photothermal and non-photothermal applications. More specifically, the most recent progress showing the potential of dye supramolecular assemblies in modalities such as photoacoustic and fluorescence imaging, photothermal and photodynamic therapies as well as their employment in photoablation as a novel modality will be outlined. Aside from their photophysical activity, we delve shortly into the emerging application of dyes as drug stabilizing agents where these molecules are used together with aggregator molecules to form stable nanoparticles.
Collapse
Affiliation(s)
- Pouria Ramezani
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences Ghent University Ghent Belgium
| | - Stefaan C De Smedt
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences Ghent University Ghent Belgium
| | - Félix Sauvage
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences Ghent University Ghent Belgium
| |
Collapse
|
50
|
Rudzińska M, Grygier A, Knight G, Kmiecik D. Liposomes as Carriers of Bioactive Compounds in Human Nutrition. Foods 2024; 13:1814. [PMID: 38928757 PMCID: PMC11202941 DOI: 10.3390/foods13121814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 06/05/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
This article provides an overview of the literature data on the role of liposomal structures and encapsulated substances in food technology and human nutrition. The paper briefly describes how liposomes are created and how they encapsulate food ingredients, which can either be individual compounds or plant extracts. Another very interesting application of liposomes is their use as antimicrobial carriers to protect food products from spoilage during storage. The encapsulation of food ingredients in liposomes can increase their bioavailability, which is particularly important for compounds with health-promoting properties but low bioavailability. Particular attention was paid to compounds such as phytosterols, which lower blood cholesterol levels but have very low absorption in the human body. In addition, consumer expectations and regulations for liposomes in food are discussed. To date, no in vivo human studies have been conducted to indicate which encapsulation methods give the best results for gastrointestinal effects and which food-added substances are most stable during food storage and processing. The paper identifies further lines of research that are needed before liposomes can be introduced into food.
Collapse
Affiliation(s)
- Magdalena Rudzińska
- Faculty of Food Science and Nutrition, University of Life Sciences, 60-637 Poznań, Poland; (A.G.); (G.K.); (D.K.)
| | | | | | | |
Collapse
|