1
|
Martinez J, Ingram N, Kapur N, Jayne DG, Beales PA. Composition-dependent tunability of the cell interactions of hybrid lipid - block copolymer vesicles. J Colloid Interface Sci 2025; 694:137664. [PMID: 40294502 DOI: 10.1016/j.jcis.2025.137664] [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: 02/22/2025] [Revised: 04/08/2025] [Accepted: 04/20/2025] [Indexed: 04/30/2025]
Abstract
Hybrid vesicles composed of phospholipids and block copolymers are of interest for a wide range of applications due to the broad tunability of their material properties that can synergistically combine desirable properties of liposomes and polymersomes. A major application of vesicles in biotechnology has been in the field of drug delivery, where understanding and controlling vesicle interactions with cells is of vital importance. Here, we investigate the tunability of hybrid vesicle interaction with three distinct cell lines through modulating non-specific interactions. We formulate vesicles composed of three different constituents, the zwitterionic lipid 1-palmitoyl-2-oleoyl-glycero-3-phosphocholine (POPC), the cationic lipid 1,2-dioleoyl-3-trimethylammonium-propane (DOTAP) and the amphiphilic diblock copolymer Poly(1,2-butadiene)-b-poly(ethylene oxide) (PBD22-PEO14). This enables the tunability of cell interactions through electrostatic attraction to anionic cellular membranes and steric repulsion from the polymeric PEO brush layer. We establish a microfluidic flow protocol to enhance the reproducibility of vesicle-cell interactions by controlling the hydrodynamic stresses during incubation and washing steps. We demonstrate a high degree of tunability of cell interactions and low cytotoxicity across the three cell lines investigated (HFFF2, HEK293, HepG2). These initial findings offer critical insights into the engineering of hybrid vesicles and their potential applications in drug delivery.
Collapse
Affiliation(s)
- Juan Martinez
- School of Chemistry and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, West Yorkshire LS2 9JT, UK; Leeds Institute for Medical Research, University of Leeds, Leeds, West Yorkshire LS9 7TF, UK
| | - Nicola Ingram
- Leeds Institute for Medical Research, University of Leeds, Leeds, West Yorkshire LS9 7TF, UK
| | - Nikil Kapur
- School of Mechanical Engineering, University of Leeds, Leeds, West Yorkshire LS2 9JT, UK
| | - David G Jayne
- Leeds Institute for Medical Research, University of Leeds, Leeds, West Yorkshire LS9 7TF, UK; The John Goligher Colorectal Surgery Unit, St. James's University Hospital, Leeds Teaching Hospital Trust, Beckett Street, Leeds, West Yorkshire LS9 7TF, UK
| | - Paul A Beales
- School of Chemistry and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, West Yorkshire LS2 9JT, UK.
| |
Collapse
|
2
|
Zhang L, Gan L, Lin Y, Mei Z, Liao S. FTO Promotes Hepatocellular Carcinoma Progression by Mediating m6A Modification of BUB1 and Targeting TGF-βR1 to Activate the TGF-β Signaling Pathway. J Clin Transl Hepatol 2025; 13:385-394. [PMID: 40385944 PMCID: PMC12078176 DOI: 10.14218/jcth.2025.00007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 03/01/2025] [Accepted: 03/17/2025] [Indexed: 05/20/2025] Open
Abstract
Background and Aims Fat mass and obesity-associated protein (FTO) has been linked to various cancers, though its role in hepatocellular carcinoma (HCC) remains unclear. This study aimed to investigate FTO expression, its clinical relevance, functional role in HCC progression, and the underlying molecular mechanisms. Methods Quantitative reverse-transcription polymerase chain reaction and immunohistochemical analysis were used to assess FTO expression in HCC. Functional assays, including proliferation, invasion, and epithelial-mesenchymal transition studies, were conducted using HCC cell lines with FTO knockdown. N6-methyladenosine (m6A) RNA immunoprecipitation and RNA stability assays further elucidated the role of FTO in BUB1 mRNA methylation and stability. Co-immunoprecipitation studies were employed to confirm the interaction between BUB1 and TGF-βR1. In vivo studies in nude mice were conducted to evaluate tumor growth following FTO knockdown. Results FTO was significantly upregulated in HCC tissues compared to normal liver tissues, with higher expression observed in advanced tumor-node-metastasis stages and metastatic HCC. Elevated FTO correlated with poor overall survival in patients. Silencing FTO decreased HCC cell proliferation, colony formation, invasion, epithelial-mesenchymal transition, and tumor growth in nude mice. Mechanistically, FTO downregulation led to increased m6A modification of BUB1 mRNA, thereby promoting its degradation via the YTH domain family 2-dependent pathway and reducing BUB1 protein levels. Additionally, BUB1 physically interacted with TGF-βR1, activating downstream TGF-β signaling. Conclusions FTO is overexpressed in HCC and is associated with poor clinical outcomes. Mechanistically, FTO promotes HCC progression by stabilizing BUB1 mRNA through an m6A-YTH domain family 2-dependent pathway, which activates TGF-β signaling. Targeting the FTO-BUB1-TGF-βR1 regulatory network may offer a promising therapeutic strategy for HCC.
Collapse
Affiliation(s)
- Lin Zhang
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Gastroenterology, Chongqing Jiangjin Central Hospital, Chongqing, China
| | - Li Gan
- Department of Anatomy, and Laboratory of Neuroscience and Tissue Engineering, Basic Medical College, Chongqing Medical University, Chongqing, China
| | - Yuru Lin
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhechuan Mei
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shengtao Liao
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
3
|
Redi G, Del Piano F, Cappellini S, Paladino M, den Breejen A, Fens MHAM, Caiazzo M. Delivery Systems in Neuronal Direct Cell Reprogramming. Cell Reprogram 2025. [PMID: 40372965 DOI: 10.1089/cell.2025.0008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/17/2025] Open
Abstract
Neuronal direct cell reprogramming approach allows direct conversion of somatic cells into neurons via forced expression of neuronal cell-lineage transcription factors (TFs). These so-called induced neuronal cells have significant potential as research tools and for therapeutic applications, such as in cell replacement therapy. However, the optimization of TF delivery strategies is crucial to reach clinical practice. In this review, we outlined the currently explored delivery technologies in neuronal direct cell reprogramming and their limitations and advantages. The first employed delivery strategies were mainly integrating viral systems, such as lentiviruses that exert consistently high transgene expression in most cell types. On the other hand, viral systems cause major safety concerns, including the risk for insertional mutagenesis and inflammation. More recently, several safer nonviral delivery systems have been investigated as well; however, these systems generally exert inferior reprogramming efficiency compared with viral systems. Emerging delivery technologies could provide new opportunities in the achievement of safe and effective delivery for neuronal direct cell reprogramming.
Collapse
Affiliation(s)
- Giulia Redi
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II," Naples, Italy
| | - Filomena Del Piano
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II," Naples, Italy
| | - Sara Cappellini
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands
| | - Martina Paladino
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II," Naples, Italy
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands
| | - Anne den Breejen
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands
| | - Marcel H A M Fens
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands
| | - Massimiliano Caiazzo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II," Naples, Italy
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands
| |
Collapse
|
4
|
Karmaker S, Rosales PD, Tirumuruhan B, Viravalli A, Boehnke N. More than a delivery system: the evolving role of lipid-based nanoparticles. NANOSCALE 2025; 17:11864-11893. [PMID: 40293317 DOI: 10.1039/d4nr04508d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Lipid-based nanoparticles, including liposomes and lipid nanoparticles (LNPs), make up an important class of drug delivery systems. Their modularity enables encapsulation of a wide range of therapeutic cargoes, their ease of functionalization allows for incorporation of targeting motifs and anti-fouling coatings, and their scalability facilitates rapid translation to the clinic. While the discovery and early understanding of lipid-based nanoparticles is heavily rooted in biology, formulation development has largely focused on materials properties, such as how liposome and lipid nanoparticle composition can be altered to maximize drug loading, stability and circulation. To achieve targeted delivery and enable improved accumulation of therapeutics at target tissues or disease sites, emphasis is typically placed on the use of external modifications, such as peptide, protein, and polymer motifs. However, these approaches can increase the complexity of the nanocarrier and complicate scale up. In this review, we focus on how our understanding of lipid structure and function in biological contexts can be used to design intrinsically functional and targeted nanocarriers. We highlight formulation-based strategies, such as the incorporation of bioactive lipids, that have been used to modulate liposome and lipid nanoparticle properties and improve their functionality while retaining simple nanocarrier designs. We also highlight classes of naturally occurring lipids, their functions, and how they have been incorporated into lipid-based nanoparticles. We will additionally position these approaches into the historical context of both liposome and LNP development.
Collapse
Affiliation(s)
- Senjuti Karmaker
- Department of Chemical Engineering and Materials Science, University of Minnesota, Twin Cities Minneapolis, MN 55455, USA.
| | - Plinio D Rosales
- Department of Chemical Engineering and Materials Science, University of Minnesota, Twin Cities Minneapolis, MN 55455, USA.
| | - Barath Tirumuruhan
- Department of Chemical Engineering and Materials Science, University of Minnesota, Twin Cities Minneapolis, MN 55455, USA.
| | - Amartya Viravalli
- Department of Chemical Engineering and Materials Science, University of Minnesota, Twin Cities Minneapolis, MN 55455, USA.
| | - Natalie Boehnke
- Department of Chemical Engineering and Materials Science, University of Minnesota, Twin Cities Minneapolis, MN 55455, USA.
| |
Collapse
|
5
|
Tregoning JS, Sanders NN. The past, present, and future of RNA vaccines. Mol Ther 2025; 33:1876-1878. [PMID: 39978334 DOI: 10.1016/j.ymthe.2025.02.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 02/06/2025] [Accepted: 02/06/2025] [Indexed: 02/22/2025] Open
Affiliation(s)
- John S Tregoning
- Department of Infectious Disease, Imperial College London, London SW7 2AZ, UK.
| | - Niek N Sanders
- Laboratory of Gene Therapy, Department of Veterinary and Biosciences, Ghent University, 9820 Merelbeke, Belgium
| |
Collapse
|
6
|
Suri K, Pfeifer L, Cvet D, Li A, McCoy M, Singh A, Amiji MM. Oral delivery of stabilized lipid nanoparticles for nucleic acid therapeutics. Drug Deliv Transl Res 2025; 15:1755-1769. [PMID: 39320435 PMCID: PMC11968485 DOI: 10.1007/s13346-024-01709-4] [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] [Accepted: 09/09/2024] [Indexed: 09/26/2024]
Abstract
Gastrointestinal disorders originate in the gastrointestinal tract (GIT), and the therapies can benefit from direct access to the GIT achievable through the oral route. RNA molecules show great promise therapeutically but are highly susceptible to degradation and often require a carrier for cytoplasmic access. Lipid nanoparticles (LNPs) are clinically proven drug-delivery agents, primarily administered parenterally. An ideal Orally Delivered (OrD) LNP formulation should overcome the diverse GI environment, successfully delivering the drug to the site of action. A versatile OrD LNP formulation has been developed to encapsulate and deliver siRNA and mRNA in this paper. The formulations were prepared by the systematic addition of cationic lipid to the base LNP formulation, keeping the total of cationic lipid and ionizable lipid to 50 mol%. Biorelevant media stability depicted increased resistance to bile salt mediated destabilization upon the addition of the cationic lipid, however the in vitro efficacy data underscored the importance of the ionizable lipid. Based on this, OrD LNP was selected comprising of 20% cationic lipid and 30% ionizable lipid. Further investigation revealed the enhanced efficacy of OrD LNP in vitro after incubation in different dilutions of fasted gastric, fasted intestinal media, and mucin. Confocal imaging and flow cytometry confirmed uptake while in vivo studies demonstrated efficacy with siRNA and mRNA as payloads. Taken together, this research introduces OrD LNP to deliver nucleic acid locally to the GIT.
Collapse
Affiliation(s)
- Kanika Suri
- Takeda Development Center Americas, Cambridge, MA, USA
- Department of Bioengineering, College of Engineering, Northeastern University, Boston, MA, USA
| | - Liam Pfeifer
- Takeda Development Center Americas, Cambridge, MA, USA
| | - Donna Cvet
- Takeda Development Center Americas, Cambridge, MA, USA
| | - Angela Li
- Takeda Development Center Americas, Cambridge, MA, USA
| | - Michael McCoy
- Takeda Development Center Americas, Cambridge, MA, USA
| | - Amit Singh
- Takeda Development Center Americas, Cambridge, MA, USA
| | - Mansoor M Amiji
- Department of Pharmaceutical Sciences, Bouve College of Health Sciences, Northeastern University, Boston, MA, USA.
- Department of Chemical Engineering, College of Engineering, Northeastern University, Boston, MA, USA.
| |
Collapse
|
7
|
Fang R, Vallius T, Zhang A, Van Cura D, Alicandri F, Fischer G, Draper E, Xu S, Pelletier R, Wang J, Mandinova A, Katsyv I, Sorger PK, Murphy GF, Lian CG. PRAME Expression in Melanoma is Negatively Regulated by TET2-Mediated DNA Hydroxymethylation. J Transl Med 2025; 105:104123. [PMID: 40024557 DOI: 10.1016/j.labinv.2025.104123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 02/12/2025] [Accepted: 02/21/2025] [Indexed: 03/04/2025] Open
Abstract
Preferentially expressed Antigen in Melanoma (PRAME) and Ten-Eleven Translocation (TET) dioxygenase-mediated 5-hydroxymethylcytosine (5hmC) are emerging melanoma biomarkers. We observed an inverse correlation between PRAME expression and 5hmC levels in benign nevi, melanoma in situ, primary invasive melanoma, and metastatic melanomas via immunohistochemistry and multiplex immunofluorescence: nevi exhibited high 5hmC and low PRAME, whereas melanomas showed the opposite pattern. Single-cell multiplex imaging of melanoma precursors revealed that diminished 5hmC coincides with PRAME upregulation in premalignant cells. Analysis of The Cancer Genome Atlas and Genotype-Tissue Expression databases confirmed a negative relationship between TET2 and PRAME messenger RNA expression in melanoma. Additionally, 5hmC levels were reduced at the PRAME 5' promoter in melanoma compared with nevi, suggesting a role for 5hmC in PRAME transcription. Restoring 5hmC levels via TET2 overexpression notably reduced PRAME expression in melanoma cell lines. These findings establish a function of TET2-mediated DNA hydroxymethylation in regulating PRAME expression and demonstrate epigenetic reprogramming as pivotal in melanoma tumorigenesis.
Collapse
Affiliation(s)
- Rui Fang
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Tuulia Vallius
- Laboratory of Systems Pharmacology, Harvard Medical School, Harvard Program in Therapeutic Science, Boston, Massachusetts; Ludwig Center at Harvard Medical School, Boston, Massachusetts
| | - Arianna Zhang
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Devon Van Cura
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts
| | - Francisco Alicandri
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Grant Fischer
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Elizabeth Draper
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Shuyun Xu
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Roxanne Pelletier
- Laboratory of Systems Pharmacology, Harvard Medical School, Harvard Program in Therapeutic Science, Boston, Massachusetts; Ludwig Center at Harvard Medical School, Boston, Massachusetts
| | - Justina Wang
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Anna Mandinova
- Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts; Broad Institute of Harvard and MIT, Cambridge, Massachusetts; Harvard Stem Cell Institute, Cambridge, Massachusetts
| | - Igor Katsyv
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Peter K Sorger
- Laboratory of Systems Pharmacology, Harvard Medical School, Harvard Program in Therapeutic Science, Boston, Massachusetts; Ludwig Center at Harvard Medical School, Boston, Massachusetts; Department of Systems Biology, Harvard Medical School, Boston, Massachusetts.
| | - George F Murphy
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.
| | - Christine G Lian
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
8
|
Al-Shihabi AM, Al-Mohaya M, Haider M, Demiralp B. Exploring the promise of lipoplexes: From concept to clinical applications. Int J Pharm 2025; 674:125424. [PMID: 40043964 DOI: 10.1016/j.ijpharm.2025.125424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 02/02/2025] [Accepted: 02/28/2025] [Indexed: 03/09/2025]
Abstract
Lipoplexes are non-viral lipid vectors that effectively form complexes with genetic material, positioning them as promising alternatives to viral vectors in gene therapy. Their advantages include lower toxicity, reduced immunogenicity, improved targetability, and ease of large-scale production. A typical lipoplex is composed of cationic lipids, neutral lipids, and anionic nucleic acids (e.g., DNA, mRNA, miRNA, siRNA, shRNA). Neutral lipids play an auxiliary role and are often used as transfection enhancers. Enhancing lipoplex efficiency often involves modifying the cationic lipid structure through functional groups like PEG polymers and targeting ligands. The assembly of lipoplexes occurs spontaneously. This process involves the binding of the positively charged polar head group of the cationic lipid to the negatively charged DNA spontaneously as a result of electrostatic interaction, then irreversible rearrangement and condensation of the lipoplex occurs to form either lamellar or hexagonal structures. The transfection process encompasses several steps: cellular entry, endosomal escape and cargo release, cytoplasmic trafficking, and nuclear entry. The physicochemical and biological properties of lipoplexes are influenced by factors such as lipid structure, charge ratio, and environmental conditions. Despite certain limitations like low gene transfer efficiency and rapid clearance by serum proteins, lipoplexes show promise for clinical applications. They can be administered through various routes, offering potential treatments for diseases such as cancer, bone damage, infection, and cystic fibrosis. The study aims to examine the potential of lipoplexes as a promising vehicle for delivering therapeutic agents and their progression from theoretical concepts to practical clinical applications.
Collapse
Affiliation(s)
- Alaa M Al-Shihabi
- Institute of Health Sciences, Istanbul University, 34216, Beyazıt, Istanbul, Turkey; Istanbul University, Faculty of Pharmacy, Pharmaceutical Technology Dept., 34126, Beyazıt, Istanbul, Turkey
| | - Mazen Al-Mohaya
- Institute of Health Sciences, Istanbul University, 34216, Beyazıt, Istanbul, Turkey; Istanbul University, Faculty of Pharmacy, Pharmaceutical Technology Dept., 34126, Beyazıt, Istanbul, Turkey
| | - Mohamed Haider
- Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, University of Sharjah 27272 Sharjah, United Arab Emirates; Research Institute of Medical & Health Sciences, University of Sharjah 27272 Sharjah, United Arab Emirates.
| | - Burcu Demiralp
- Istanbul University, Faculty of Pharmacy, Pharmaceutical Technology Dept., 34126, Beyazıt, Istanbul, Turkey.
| |
Collapse
|
9
|
Honrath S, Burger M, Leroux JC. Hurdles to healing: Overcoming cellular barriers for viral and nonviral gene therapy. Int J Pharm 2025; 674:125470. [PMID: 40112901 DOI: 10.1016/j.ijpharm.2025.125470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 02/26/2025] [Accepted: 03/11/2025] [Indexed: 03/22/2025]
Abstract
Gene delivery offers great potential for treating various diseases, yet its success requires overcoming several biological barriers. These hurdles span from extracellular degradation, reaching the target cells, and inefficient cellular uptake to endosomal entrapment, cytoplasmic transport, nuclear entry, and transcription limitations. Viruses and non-viral vectors deal with these barriers via different mechanisms. Viral vectors, such as adenoviruses, adeno-associated viruses, and lentiviruses use natural mechanisms to efficiently deliver genetic material but face limitations including immunogenicity, cargo capacity, and production complexity. Nonviral vectors, including lipid nanoparticles, polymers, and protein-based systems, offer scalable and safer alternatives but often fall short in overcoming intracellular barriers and achieving high transfection efficiencies. Recent advancements in vector engineering have partially overcome several of these challenges. Ionizable lipids improve endosomal escape while minimizing toxicity. Biodegradable polymers balance efficacy with safety, and engineered protein systems, inspired by viral or bacterial entry mechanisms, integrate multifunctionality for enhanced delivery. Despite these advances, challenges, particularly in achieving robust in vivo translatability, scalability, and reduced immunogenicity, remain. This review synthesizes current knowledge of cellular barriers and the approaches to overcome them, providing a roadmap for designing more efficient gene delivery systems. By addressing these barriers, the field can advance toward safer, and more effective therapies.
Collapse
Affiliation(s)
- Steffen Honrath
- ETH Zurich, Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, Vladimir-Prelog-Weg 3, 8093 Zurich, Switzerland
| | - Michael Burger
- ETH Zurich, Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, Vladimir-Prelog-Weg 3, 8093 Zurich, Switzerland.
| | - Jean-Christophe Leroux
- ETH Zurich, Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, Vladimir-Prelog-Weg 3, 8093 Zurich, Switzerland.
| |
Collapse
|
10
|
Thongbamrer C, Kunkeaw N, Nguitragool W, Roobsoong W, Sattabongkot J, Pengnam S, Opanasopit P, Yingyongnarongkul BE. Enhancing Transfection Efficiency of Spermine-Based Cationic Lipids with a Lysine-Based Spacer. Chem Asian J 2025:e202401751. [PMID: 40229172 DOI: 10.1002/asia.202401751] [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: 12/02/2024] [Revised: 03/27/2025] [Accepted: 03/31/2025] [Indexed: 04/16/2025]
Abstract
L-shape spermine-based cationic lipids with a lysine spacer and both identical and non-identical hydrophobic tails were successfully synthesized. Liposomes prepared from these lipids, either alone or in combination with DOPE, demonstrated DNA-binding capability, as confirmed by gel electrophoresis assays. The physicochemical properties, such as size, zeta-potential, and stability of the lipoplexes formed from the cationic lipids were investigated. The liposomes efficiently condensed DNA into compact structures at an N/P ratio of approximately 5-10. Interestingly, liposomes without DOPE exhibited higher transfection efficiency than those containing DOPE, with the cationic lipid featuring a spermine polar head bonded to a lysine spacer and C12-C16 hydrocarbon tails (Sper-Lys-C12,16) achieved the greatest transfection efficiency, as revealed by fluorescence microscopy and flow cytometry. Notably, this formulation maintained high transfection efficiencies even in serum concentrations up to 40%, outperforming the commercial standard Lipofectamine 3000. Additionally, these liposomes exhibited low cytotoxicity, highlighting their potential as safe and effective gene delivery agents.
Collapse
Affiliation(s)
- Chopaka Thongbamrer
- Department of Chemistry and Center of Excellence for Innovation in Chemistry (PERCH-CIC) Faculty of Science, Ramkhamhaeng University, Bangkok, 10240, Thailand
| | - Nawapol Kunkeaw
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Ratchathewi, Bangkok, 10400, Thailand
| | - Wang Nguitragool
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Ratchathewi, Bangkok, 10400, Thailand
| | - Wanlapa Roobsoong
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Ratchathewi, Bangkok, 10400, Thailand
| | - Jetsumon Sattabongkot
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Ratchathewi, Bangkok, 10400, Thailand
| | - Supusson Pengnam
- Pharmaceutical Development of Green Innovations Group (PDGIG), Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, 73000, Thailand
| | - Praneet Opanasopit
- Pharmaceutical Development of Green Innovations Group (PDGIG), Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, 73000, Thailand
| | - Boon-Ek Yingyongnarongkul
- Department of Chemistry and Center of Excellence for Innovation in Chemistry (PERCH-CIC) Faculty of Science, Ramkhamhaeng University, Bangkok, 10240, Thailand
| |
Collapse
|
11
|
Pakian S, Nabid MR, Satarian L, Abandansari HS, Mirkani A. Topical Administration of Mucoadhesive Liposomes-Epoetin-β for Targeting the Ocular Posterior Segment. Mol Pharm 2025; 22:2287-2305. [PMID: 40100128 DOI: 10.1021/acs.molpharmaceut.5c00079] [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] [Indexed: 03/20/2025]
Abstract
Delivering drugs to the posterior eye segment is a complex task, particularly for treating retinal diseases. Neuroprotective approaches to maintain neuronal integrity have garnered significant attention in recent research. Here, we developed a mucoadhesive nanoparticulate system based on thiolated hyaluronic acid-modified cationic liposomes (HA-SH@liposomes) for topical administration. To fabricate these liposomes, we utilized microfluidic technology with a toroidal mixer to ensure consistent size and stability. Cationic liposomes were prepared by using the microfluidic method, and Epoetin-β (EPOβ), a neuroprotective agent, was loaded into the liposomes. Following this, HA-SH was conjugated to the EPOβ/HA-SH@liposomes using a postmicrofluidics conjugation method, wherein HA-SH was added dropwise to facilitate electrostatic interactions between the cationic liposomes and the anionic polymer. The resulting liposomes exhibited a mean size of 144 ± 1.3 nm and a polydispersity index (PDI) of 0.09 ± 0.01, indicating their uniformity. We evaluated the biocompatibility of the EPOβ/HA-SH@liposomes in vitro using live/dead and MTS assays on the RGC-5 cell line, demonstrating no notable cytotoxicity compared to the controls. To assess the in vivo performance, we conducted extensive ophthalmological examinations in C57/BL6 mice, including immunofluorescence staining to track the distribution of EPOβ and EPOβ/HA-SH@liposomes within the eyeball. Additionally, we quantified EPOβ levels in the retina using an enzyme-linked immunosorbent assay (ELISA) kit after the topical application of free EPOβ and the EPOβ/HA-SH@liposome formulation. The immunofluorescence staining revealed efficient delivery of EPOβ into the retina and choroid via the transcorneal route when administered as EPOβ/HA-SH@liposomes. ELISA results showed that the liposomal formulation achieved approximately 1.9× greater penetration efficiency than free EPOβ. Furthermore, optokinetic response (OKR) assays indicated that animals treated with EPOβ/HA-SH@liposomes exhibited slightly improved visual acuity compared with those treated with free EPOβ, though the difference was not statistically significant. In conclusion, the topical ocular administration of EPOβ/HA-SH@liposomes facilitated the efficient delivery of EPOβ to the retina, promoting retinal recovery and confirming its neuroprotective properties. This preclinical study provides a foundation for innovative strategies in the topical delivery of neuroprotective agents in ocular therapy.
Collapse
Affiliation(s)
- Sarvenaz Pakian
- Department of Polymer and Materials Chemistry, Faculty of Chemistry & Petroleum Sciences, Shahid Beheshti University, Tehran 1983969411, Iran
| | - Mohammad Reza Nabid
- Department of Polymer and Materials Chemistry, Faculty of Chemistry & Petroleum Sciences, Shahid Beheshti University, Tehran 1983969411, Iran
| | - Leila Satarian
- Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 16635-148, Iran
| | - Hamid Sadeghi Abandansari
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 16635-148, Iran
- Department of Stem Cells and Developmental Biology, Faculty of Basic Sciences and Advanced Technologies in Biology, University of Science and Culture, Tehran 1461968151, Iran
| | - Ahmad Mirkani
- Department of Polymer and Materials Chemistry, Faculty of Chemistry & Petroleum Sciences, Shahid Beheshti University, Tehran 1983969411, Iran
| |
Collapse
|
12
|
Tajer BJ, Kalu G, Jay S, Wynn E, Decaux A, Gilbert P, Singer HD, Kidd MD, Nelson JA, Harake N, Lopez NJ, Souchet NR, Luong AG, Savage AM, Min S, Karabacak A, Böhm S, Kim RT, Froitzheim T, Sousounis K, Courtemanche K, Han J, Payzin-Dogru D, Blair SJ, Roy S, Fei JF, Tanaka EM, Whited JL. Optimized toolkit for the manipulation of immortalized axolotl fibroblasts. Methods 2025; 240:21-34. [PMID: 40187387 DOI: 10.1016/j.ymeth.2025.03.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 02/27/2025] [Accepted: 03/27/2025] [Indexed: 04/07/2025] Open
Abstract
The axolotl salamander model has broad utility for regeneration studies, but this model is limited by a lack of efficient cell-culture-based tools. The Axolotl Limb-1 (AL-1) fibroblast line, the only available immortalized axolotl cell line, was first published over 20 years ago, but many established molecular biology techniques, such as lipofectamine transfection, CRISPR-Cas9 mutagenesis, and antibiotic selection, work poorly or remain untested in AL-1 cells. Innovating technologies to manipulate AL-1 cells in culture and study their behavior following transplantation into the axolotl will complement in-vivo studies, decrease the number of animals used, and enable the faster, more streamlined investigation of regenerative biology questions. Here, we establish transfection, mutagenesis, antibiotic selection, and in-vivo transplantation techniques in axolotl AL-1 cells. These techniques will enable efficient culture with AL-1 cells and guide future tool development for the culture and manipulation of other salamander cell lines.
Collapse
Affiliation(s)
- Benjamin J Tajer
- Departmet of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Ave., Cambridge, MA 02318, USA
| | - Glory Kalu
- Departmet of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Ave., Cambridge, MA 02318, USA
| | - Sarah Jay
- Departmet of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Ave., Cambridge, MA 02318, USA; Master de Biologie, École Normale Supérieure de Lyon, Université Claude Bernard Lyon 1, Université de Lyon, 69342 Lyon Cedex 07, France
| | - Eric Wynn
- Departmet of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Ave., Cambridge, MA 02318, USA
| | - Antoine Decaux
- Departmet of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Ave., Cambridge, MA 02318, USA; Master de Biologie, École Normale Supérieure de Lyon, Université Claude Bernard Lyon 1, Université de Lyon, 69342 Lyon Cedex 07, France
| | - Paul Gilbert
- Departmet of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Ave., Cambridge, MA 02318, USA
| | - Hani D Singer
- Departmet of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Ave., Cambridge, MA 02318, USA
| | - Maddeline D Kidd
- Departmet of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Ave., Cambridge, MA 02318, USA
| | - Jeffery A Nelson
- Bauer Core Facility, Harvard University, Northwest Building, Room B239, 52 Oxford St., Cambridge, MA 02138, USA
| | - Noora Harake
- Departmet of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Ave., Cambridge, MA 02318, USA
| | - Noah J Lopez
- Departmet of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Ave., Cambridge, MA 02318, USA
| | - Nathan R Souchet
- Departmet of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Ave., Cambridge, MA 02318, USA
| | - Anna G Luong
- Departmet of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Ave., Cambridge, MA 02318, USA
| | - Aaron M Savage
- Departmet of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Ave., Cambridge, MA 02318, USA
| | - Sangwon Min
- Departmet of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Ave., Cambridge, MA 02318, USA
| | - Alparslan Karabacak
- Departmet of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Ave., Cambridge, MA 02318, USA
| | - Sebastian Böhm
- Departmet of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Ave., Cambridge, MA 02318, USA
| | - Ryan T Kim
- Departmet of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Ave., Cambridge, MA 02318, USA
| | - Tim Froitzheim
- Departmet of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Ave., Cambridge, MA 02318, USA
| | - Konstantinos Sousounis
- Departmet of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Ave., Cambridge, MA 02318, USA
| | - Katherine Courtemanche
- Departmet of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Ave., Cambridge, MA 02318, USA
| | - Jihee Han
- Departmet of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Ave., Cambridge, MA 02318, USA
| | - Duygu Payzin-Dogru
- Departmet of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Ave., Cambridge, MA 02318, USA
| | - Steven J Blair
- Departmet of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Ave., Cambridge, MA 02318, USA
| | - Stéphane Roy
- Department of Stomatology, Faculty of Dentistry, Université de Montréal, Montréal, Québec H3T 1J4, Canada
| | - Ji-Feng Fei
- Department of Pathology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, China
| | - Elly M Tanaka
- Institute of Molecular Biotechnology, Austrian Academy of Sciences (IMBA), Vienna Biocenter (VBC), Dr.-Bohr-Gasse 3, 1030 Vienna, Austria
| | - Jessica L Whited
- Departmet of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Ave., Cambridge, MA 02318, USA; Broad Institute, 415 Main St., Cambridge, MA 02142, USA; Department of Orthopedic Surgery, Brigham & Women's Hospital, Mass General Brigham, 75 Francis St., Boston, MA 02115, USA.
| |
Collapse
|
13
|
Gruenberger R, Baek C, Spitzenberg C, Min J, Baeumner AJ. Studying Cationic Liposomes for Quick, Simple, and Effective Nucleic Acid Preconcentration and Isolation. Anal Chem 2025; 97:6018-6026. [PMID: 39878247 PMCID: PMC11948181 DOI: 10.1021/acs.analchem.4c05936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 01/14/2025] [Accepted: 01/16/2025] [Indexed: 01/31/2025]
Abstract
To ensure high quality of food and water, the identification of traces of pathogens is mandatory. Rapid nucleic acid-based tests shorten traditional detection times while maintaining low detection limits. Challenging is the loss of nucleic acids during necessary purification processes, since elution off solid surfaces is not efficient. We therefore propose the development of a vanishing surface strategy in which cationic liposomes efficiently capture nucleic acids. A lipoplex is formed that can be easily centrifuged down and washed if needed. Adding the lipoplex to detergent solutions or nucleic acid amplification reactions dissolves the liposomes, releasing 100% of the nucleic acid into the reaction. After initial protocol optimization, it was applied to isolate DNA from Escherichia coli, Staphylococcus aureus, and adenovirus in buffer followed by qPCR detection. This enabled the detection of these pathogens down to concentrations of 1 CFU or 1 PFU/mL, respectively. Comparing it to a standard commercial DNA extraction kit, it was superior, as evidenced by lower Ct-values in the qPCR for all pathogen concentrations. Scaling up to larger volumes, samples containing bacteria were first concentrated through nitrocellulose filters (pore size = 0.45 μm). Tap water, lake water, and rinse water of fresh produce were investigated, leading to relevant limits of detection of 100 CFU in 100 mL of tap water, 1000 CFU in 100 mL of lake water, and 100 CFU in 10 g of iceberg lettuce, respectively. Since the liposome protocol is a homogeneous, simple incubation step, it is a valuable alternative to standard commercial nucleic acid extraction kits.
Collapse
Affiliation(s)
- Rahel Gruenberger
- Institute
of Analytical Chemistry, Chemo- and Biosensors, University of Regensburg, Universitaetsstr. 31, Regensburg 93053, Germany
| | - Changyoon Baek
- School
of Integrative Engineering, Chung-Ang University, Heukseok-Dong, Dongjak-Gu, Seoul 06974, Republic of Korea
| | - Clemens Spitzenberg
- Institute
of Analytical Chemistry, Chemo- and Biosensors, University of Regensburg, Universitaetsstr. 31, Regensburg 93053, Germany
| | - Junhong Min
- School
of Integrative Engineering, Chung-Ang University, Heukseok-Dong, Dongjak-Gu, Seoul 06974, Republic of Korea
| | - Antje J. Baeumner
- Institute
of Analytical Chemistry, Chemo- and Biosensors, University of Regensburg, Universitaetsstr. 31, Regensburg 93053, Germany
| |
Collapse
|
14
|
Bruckhoff R, Becker O, Steinhilber D, Suess B. Potential and Optimization of Mammalian Splice Riboswitches for the Regulation of Exon Skipping-Dependent Gene Expression and Isoform Switching within the ALOX5 Gene. ACS Synth Biol 2025; 14:804-818. [PMID: 40011207 PMCID: PMC11934966 DOI: 10.1021/acssynbio.4c00731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 02/18/2025] [Accepted: 02/18/2025] [Indexed: 02/28/2025]
Abstract
Synthetic riboswitches are attracting increasing interest for a diverse range of applications, including synthetic biology, functional genomics, and prospective therapeutic strategies. This study demonstrates that controlling alternative splicing with synthetic riboswitches represents a promising approach to effectively regulating transgene expression in mammalian cells. However, the function of synthetic riboswitches in the eukaryotic system in controlling gene expression is often limited to certain genes or cell types. So far, strategies to increase the dynamic range of regulation have been focused on adapting and modifying the riboswitch sequence itself without taking into account the context in which the riboswitch was inserted. In the present study, the tetracycline riboswitch was chosen to investigate the effects of the context and insertion site of a cassette exon within the gene to control the expression of an artificial arachidonate 5-lipoxygenase gene (ALOX5) in HEK293 cells. We demonstrate here that the use of riboswitch-controlled cassette exons for the control of gene expression via alternative splicing can be easily transferred to another gene through the process of contextual sequence adaptation. This was achieved through the introduction of gene-specific intronic and exonic sequences with different intron lengths and positions being tested. In contrast, the introduction of nonadapted constructs resulted in an unanticipated functionality outcome of the gene switch. Furthermore, we demonstrate that the combination of two cassette exons into a single gene resulted in a notable enhancement in the dynamic range. Finally, we generated a novel riboswitch-controlled splicing concept that enabled us to switch 5-LO wild-type to expression of an ALOX5 isoform that lacks exon 13 (5-LOΔ13). Taken together, this study demonstrates that synthetic riboswitches that control alternative splicing are a powerful tool to regulate gene expression when applied in combination with gene-specific intronic and exonic sequences.
Collapse
Affiliation(s)
- Robin
W. Bruckhoff
- Department
of Biology, Technical University of Darmstadt, Schnittspahnstraße 10, Darmstadt D-64287, Germany
| | - Olga Becker
- Department
of Biology, Technical University of Darmstadt, Schnittspahnstraße 10, Darmstadt D-64287, Germany
| | - Dieter Steinhilber
- Institute
of Pharmaceutical Chemistry, Goethe University, Max-von-Laue-Straße 9, Frankfurt am Main D-60438, Germany
| | - Beatrix Suess
- Department
of Biology, Technical University of Darmstadt, Schnittspahnstraße 10, Darmstadt D-64287, Germany
- Centre
for Synthetic Biology, Technical University
of Darmstadt, Darmstadt 64287, Germany
| |
Collapse
|
15
|
Park J, Rhoo KY, Kim Y, Kim YS, Paik SR. Cell-Division-Independent Rapid Expression of DNA Delivered with α-Synuclein-Gold Nanoparticle Conjugates. ACS APPLIED MATERIALS & INTERFACES 2025; 17:14846-14858. [PMID: 40014054 DOI: 10.1021/acsami.4c17967] [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: 02/28/2025]
Abstract
Gene delivery is a primary technology employed in diverse areas of biomedical science, from gene therapy to gene editing, cancer treatment, and stem cell research. Here, we introduce a gene delivery system utilizing an intrinsically disordered protein of α-synuclein (αS) demonstrated to interact with lipid membranes by transforming its original random structure to an α-helix. Since the helix bundle formation is a signature of cell-penetrating peptides for membrane translocation, a multitude of αS(Y136C)s replacing tyrosine at the C-terminus with cysteine were covalently attached onto gold nanoparticles (AuNPs) in a specific orientation with the helix-forming basic N-termini exposed outward. The resulting αS(Y136C)-AuNP conjugates were found to exhibit a rapid gene expression without causing cytotoxicity when the gene of the enhanced green fluorescent protein (EGFP) was delivered with the conjugates into the cells. Based on inhibition studies toward endocytosis and mitosis, the αS(Y136C)-AuNP/DNA complex was demonstrated to take both endosomal and non-endosomal intracellular transport pathways. The DNA translocation into the nucleus was independent of cell division. This nondisruptive and rapid DNA transfection by αS(Y136C)-AuNPs allowed a successful delivery of granzyme A gene leading to cellular pyroptosis. Modifications of αS(Y136C)-AuNP/DNA complex, such as antibody immobilization and replacement of DNA with biological suprastructures including RNA, protein, and nonbiological fusion materials, would allow the intracellular delivery system to be applied in diverse areas of future biotechnology.
Collapse
Affiliation(s)
- Jeongha Park
- School of Chemical and Biological Engineering, Institute of Engineering Research, College of Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Kun Yil Rhoo
- Interdisciplinary Program of Bioengineering, College of Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Yunsoo Kim
- School of Chemical and Biological Engineering, Institute of Engineering Research, College of Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Young-Sik Kim
- Department of Pathology, Ansan Hospital, Korea University College of Medicine, Ansan 15355, Republic of Korea
| | - Seung R Paik
- School of Chemical and Biological Engineering, Institute of Engineering Research, College of Engineering, Seoul National University, Seoul 08826, Republic of Korea
- Interdisciplinary Program of Bioengineering, College of Engineering, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
16
|
Cavazza A, Molina-Estévez FJ, Reyes ÁP, Ronco V, Naseem A, Malenšek Š, Pečan P, Santini A, Heredia P, Aguilar-González A, Boulaiz H, Ni Q, Cortijo-Gutierrez M, Pavlovic K, Herrera I, de la Cerda B, Garcia-Tenorio EM, Richard E, Granados-Principal S, López-Márquez A, Köber M, Stojanovic M, Vidaković M, Santos-Garcia I, Blázquez L, Haughton E, Yan D, Sánchez-Martín RM, Mazini L, Aseguinolaza GG, Miccio A, Rio P, Desviat LR, Gonçalves MA, Peng L, Jiménez-Mallebrera C, Molina FM, Gupta D, Lainšček D, Luo Y, Benabdellah K. Advanced delivery systems for gene editing: A comprehensive review from the GenE-HumDi COST Action Working Group. MOLECULAR THERAPY. NUCLEIC ACIDS 2025; 36:102457. [PMID: 39991472 PMCID: PMC11847086 DOI: 10.1016/j.omtn.2025.102457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
In the past decade, precise targeting through genome editing has emerged as a promising alternative to traditional therapeutic approaches. Genome editing can be performed using various platforms, where programmable DNA nucleases create permanent genetic changes at specific genomic locations due to their ability to recognize precise DNA sequences. Clinical application of this technology requires the delivery of the editing reagents to transplantable cells ex vivo or to tissues and organs for in vivo approaches, often representing a barrier to achieving the desired editing efficiency and safety. In this review, authored by members of the GenE-HumDi European Cooperation in Science and Technology (COST) Action, we described the plethora of delivery systems available for genome-editing components, including viral and non-viral systems, highlighting their advantages, limitations, and potential application in a clinical setting.
Collapse
Affiliation(s)
- Alessia Cavazza
- Molecular and Cellular Immunology Section, Department of Infection, Immunity & Inflammation, UCL Great Ormond Street Institute of Child Health, University College London, 20 Guilford Street, London WC1N 1DZ, UK
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Via del Pozzo 71, 41125 Modena, Italy
| | - Francisco J. Molina-Estévez
- Department of Genomic Medicine, Pfizer-University of Granada-Andalusian Regional Government Centre for Genomics and Oncological Research (GENYO), Av. de la Ilustración, 114, 18016 Granada, Spain
- Fundación para la Investigación Biosanitaria de Andalucía Oriental, Alejandro Otero (FIBAO), Avda. de Madrid 15, 18012 Granada, Spain
- Biosanitary Research Institute of Granada (ibs. GRANADA), University of Granada, Av. de Madrid, 15, Beiro, 18012 Granada, Spain
| | - Álvaro Plaza Reyes
- Department of Regeneration and Cell Therapy, Andalusian Molecular Biology and Regenerative Medicine Centre (CABIMER), Avda. Americo Vespucio, 24, 41092 Seville, Spain
| | - Victor Ronco
- Department of Genomic Medicine, Pfizer-University of Granada-Andalusian Regional Government Centre for Genomics and Oncological Research (GENYO), Av. de la Ilustración, 114, 18016 Granada, Spain
| | - Asma Naseem
- Molecular and Cellular Immunology Section, Department of Infection, Immunity & Inflammation, UCL Great Ormond Street Institute of Child Health, University College London, 20 Guilford Street, London WC1N 1DZ, UK
| | - Špela Malenšek
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia
- Graduate School of Biomedicine, University of Ljubljana, Kongresni trg, 1000 Ljubljana, Slovenia
| | - Peter Pečan
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia
- Graduate School of Biomedicine, University of Ljubljana, Kongresni trg, 1000 Ljubljana, Slovenia
| | - Annalisa Santini
- Imagine Institute, UMR 163 INSERM, 24 Bd du Montparnasse, 75015 Paris, France
- Paris City University, 45 Rue des Saints-Pères, 75006 Paris, France
| | - Paula Heredia
- Department of Genomic Medicine, Pfizer-University of Granada-Andalusian Regional Government Centre for Genomics and Oncological Research (GENYO), Av. de la Ilustración, 114, 18016 Granada, Spain
- Department of Anatomy and Human Embryology, Faculty of Medicine, University of Granada, Avenida de la Investigación 11, 18016 Granada, Spain
| | - Araceli Aguilar-González
- Department of Genomic Medicine, Pfizer-University of Granada-Andalusian Regional Government Centre for Genomics and Oncological Research (GENYO), Av. de la Ilustración, 114, 18016 Granada, Spain
- Biosanitary Research Institute of Granada (ibs. GRANADA), University of Granada, Av. de Madrid, 15, Beiro, 18012 Granada, Spain
- Department of Medicinal & Organic Chemistry and Excellence Research Unit of “Chemistry applied to Bio-medicine and the Environment, ” Faculty of Pharmacy, University of Granada, Campus de Cartuja s/n, 18071 Granada, Spain
| | - Houria Boulaiz
- Biosanitary Research Institute of Granada (ibs. GRANADA), University of Granada, Av. de Madrid, 15, Beiro, 18012 Granada, Spain
- Department of Anatomy and Human Embryology, Faculty of Medicine, University of Granada, Avenida de la Investigación 11, 18016 Granada, Spain
| | - Qianqian Ni
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Marina Cortijo-Gutierrez
- Department of Genomic Medicine, Pfizer-University of Granada-Andalusian Regional Government Centre for Genomics and Oncological Research (GENYO), Av. de la Ilustración, 114, 18016 Granada, Spain
| | - Kristina Pavlovic
- Department of Genomic Medicine, Pfizer-University of Granada-Andalusian Regional Government Centre for Genomics and Oncological Research (GENYO), Av. de la Ilustración, 114, 18016 Granada, Spain
| | - Inmaculada Herrera
- Department of Hematology, Reina Sofía University Hospital, Av. Menéndez Pidal, Poniente Sur, 14004 Córdoba, Spain
- Maimonides Institute of Biomedical Research in Cordoba (IMIBIC), Cell Therapy, Av. Menéndez Pidal, Poniente Sur, 14004 Córdoba, Spain
| | - Berta de la Cerda
- Department of Regeneration and Cell Therapy, Andalusian Molecular Biology and Regenerative Medicine Centre (CABIMER), Avda. Americo Vespucio, 24, 41092 Seville, Spain
| | - Emilio M. Garcia-Tenorio
- Centro de Biología Molecular Severo Ochoa UAM-CSIC, IUBM, CIBERER, IDIPAZ, Universidad Autónoma de Madrid, C. de Pedro Rico, 6, Fuencarral-El Pardo, 28029 Madrid, Spain
| | - Eva Richard
- Centro de Biología Molecular Severo Ochoa UAM-CSIC, IUBM, CIBERER, IDIPAZ, Universidad Autónoma de Madrid, C. de Pedro Rico, 6, Fuencarral-El Pardo, 28029 Madrid, Spain
| | - Sergio Granados-Principal
- Department of Genomic Medicine, Pfizer-University of Granada-Andalusian Regional Government Centre for Genomics and Oncological Research (GENYO), Av. de la Ilustración, 114, 18016 Granada, Spain
- Biosanitary Research Institute of Granada (ibs. GRANADA), University of Granada, Av. de Madrid, 15, Beiro, 18012 Granada, Spain
- Department of Biochemistry and Molecular Biology 2, Faculty of Pharmacy, University of Granada, Campus de Cartuja s/n, 18071 Granada, Spain
| | - Arístides López-Márquez
- Neuromuscular Unit, Institut de Recerca Sant Joan de Déu, Hospital Sant Joan de Déu, C. de Sta. Rosa, 39, 08950 Barcelona, Spain
- Biomedical Research Network on Rare Diseases (CIBERER), C. de Melchor Fernández Almagro, 3, Fuencarral-El Pardo, 28029 Madrid, Spain
- Department of Genetics, Microbiology and Statistics, University of Barcelona, Gran Via de les Corts Catalanes, 585, L'Eixample, 08007 Barcelona, Spain
| | - Mariana Köber
- Biomedical Research Network on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), C/ Monforte de Lemos 3-5, Pabellón 11, Planta 0, 28029 Madrid, Spain
- Institut de Ciència de Materials de Barcelona (ICMAB-CSIC), Campus UAB, Universitat Autònoma de Barcelona, 08193 Bellaterra, Barcelona, Spain
| | - Marijana Stojanovic
- Institute for Biological Research “Siniša Stanković”, University of Belgrade, Bulevar despota Stefana 142, 10060 Belgrade, Serbia
| | - Melita Vidaković
- Institute for Biological Research “Siniša Stanković”, University of Belgrade, Bulevar despota Stefana 142, 10060 Belgrade, Serbia
| | - Irene Santos-Garcia
- Department of Neurosciences, Biogipuzkoa Health Research Institute, Paseo Dr. Begiristain, s/n, 20014 San Sebastián, Gipuzkoa, Spain
| | - Lorea Blázquez
- Department of Neurosciences, Biogipuzkoa Health Research Institute, Paseo Dr. Begiristain, s/n, 20014 San Sebastián, Gipuzkoa, Spain
- CIBERNED, ISCIII CIBER, Carlos III Institute, Spanish Ministry of Sciences and Innovation), Av. de Monforte de Lemos, 5, Fuencarral-El Pardo, 28029 Madrid, Spain
- Ikerbasque, Basque Foundation for Science, Euskadi Pl., 5, Abando, 48009 Bilbao, Biscay, Spain
| | - Emily Haughton
- Institute of Developmental & Regenerative Medicine, University of Oxford, Campus, Old Rd, Roosevelt Dr, Headington, Oxford OX3 7TY, UK
| | - Dongnan Yan
- Institute of Developmental & Regenerative Medicine, University of Oxford, Campus, Old Rd, Roosevelt Dr, Headington, Oxford OX3 7TY, UK
- Nuffield Department of Women’s and Reproductive Health, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Rosario María Sánchez-Martín
- Department of Genomic Medicine, Pfizer-University of Granada-Andalusian Regional Government Centre for Genomics and Oncological Research (GENYO), Av. de la Ilustración, 114, 18016 Granada, Spain
- Biosanitary Research Institute of Granada (ibs. GRANADA), University of Granada, Av. de Madrid, 15, Beiro, 18012 Granada, Spain
- Department of Medicinal & Organic Chemistry and Excellence Research Unit of “Chemistry applied to Bio-medicine and the Environment, ” Faculty of Pharmacy, University of Granada, Campus de Cartuja s/n, 18071 Granada, Spain
| | - Loubna Mazini
- Technological, Medical and Academic Park (TMAP), N°109, Abdelkrim Elkhatabi, Bd Abdelkrim Al Khattabi, Marrakech 40000, Morocco
| | - Gloria Gonzalez Aseguinolaza
- DNA & RNA Medicine Division, Gene Therapy for Rare Diseases Department, Center for Applied Medical Research (CIMA), University of Navarra, IdisNA, Av. de Pío XII, 55, 31008 Pamplona, Navarra, Spain
- Vivet Therapeutics, Av. de Pío XII 31, 31008 Pamplona, Navarra, Spain
| | - Annarita Miccio
- Imagine Institute, UMR 163 INSERM, 24 Bd du Montparnasse, 75015 Paris, France
- Paris City University, 45 Rue des Saints-Pères, 75006 Paris, France
| | - Paula Rio
- Biomedical Research Network on Rare Diseases (CIBERER), C. de Melchor Fernández Almagro, 3, Fuencarral-El Pardo, 28029 Madrid, Spain
- Division of Hematopoietic Innovative Therapies, CIEMAT, Av. Complutense, 40, Moncloa - Aravaca, 28040 Madrid, Spain
- Advanced Therapies Unit, IIS-Fundación Jimenez Diaz (IIS-FJD, UAM), Av. de los Reyes Católicos, 2, Moncloa - Aravaca, 28040 Madrid, Spain
| | - Lourdes R. Desviat
- Centro de Biología Molecular Severo Ochoa UAM-CSIC, IUBM, CIBERER, IDIPAZ, Universidad Autónoma de Madrid, C. de Pedro Rico, 6, Fuencarral-El Pardo, 28029 Madrid, Spain
| | - Manuel A.F.V. Gonçalves
- Leiden University Medical Center, Department of Cell and Chemical Biology, Einthovenweg 20, 2333 ZC Leiden, the Netherlands
| | - Ling Peng
- Aix-Marseille Universite, CNRS, Centre Interdisciplinaire de Nanoscience de Marseille, UMR 7325, “Equipe Labellisee Ligue Ćontre le Cancer”, Campus de Luminy, case 913, 13009 Marseille, France
| | - Cecilia Jiménez-Mallebrera
- Neuromuscular Unit, Institut de Recerca Sant Joan de Déu, Hospital Sant Joan de Déu, C. de Sta. Rosa, 39, 08950 Barcelona, Spain
- Biomedical Research Network on Rare Diseases (CIBERER), C. de Melchor Fernández Almagro, 3, Fuencarral-El Pardo, 28029 Madrid, Spain
| | - Francisco Martin Molina
- Department of Genomic Medicine, Pfizer-University of Granada-Andalusian Regional Government Centre for Genomics and Oncological Research (GENYO), Av. de la Ilustración, 114, 18016 Granada, Spain
- Biosanitary Research Institute of Granada (ibs. GRANADA), University of Granada, Av. de Madrid, 15, Beiro, 18012 Granada, Spain
- Department of Biochemistry and Molecular Biology III and Immunology, Faculty of Medicine, University of Granada, Avenida de la Investigación 11, 18016 Granada, Spain
| | - Dhanu Gupta
- Institute of Developmental & Regenerative Medicine, University of Oxford, Campus, Old Rd, Roosevelt Dr, Headington, Oxford OX3 7TY, UK
- Department of Laboratory Medicine, Karolinska Institutet, Alfred Nobels allé 8, 141 52 Huddinge, Sweden
| | - Duško Lainšček
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia
- Centre for Technologies of Gene and Cell Therapy, National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia
- EN-FIST Centre of Excellence, Trg Osvobodilne fronte 13, 1000 Ljubljana, Slovenia
| | - Yonglun Luo
- Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, 8200 Aarhus N, Denmark
| | - Karim Benabdellah
- Department of Genomic Medicine, Pfizer-University of Granada-Andalusian Regional Government Centre for Genomics and Oncological Research (GENYO), Av. de la Ilustración, 114, 18016 Granada, Spain
| |
Collapse
|
17
|
Paloncýová M, Valério M, Dos Santos RN, Kührová P, Šrejber M, Čechová P, Dobchev DA, Balsubramani A, Banáš P, Agarwal V, Souza PCT, Otyepka M. Computational Methods for Modeling Lipid-Mediated Active Pharmaceutical Ingredient Delivery. Mol Pharm 2025; 22:1110-1141. [PMID: 39879096 PMCID: PMC11881150 DOI: 10.1021/acs.molpharmaceut.4c00744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 01/06/2025] [Accepted: 01/06/2025] [Indexed: 01/31/2025]
Abstract
Lipid-mediated delivery of active pharmaceutical ingredients (API) opened new possibilities in advanced therapies. By encapsulating an API into a lipid nanocarrier (LNC), one can safely deliver APIs not soluble in water, those with otherwise strong adverse effects, or very fragile ones such as nucleic acids. However, for the rational design of LNCs, a detailed understanding of the composition-structure-function relationships is missing. This review presents currently available computational methods for LNC investigation, screening, and design. The state-of-the-art physics-based approaches are described, with the focus on molecular dynamics simulations in all-atom and coarse-grained resolution. Their strengths and weaknesses are discussed, highlighting the aspects necessary for obtaining reliable results in the simulations. Furthermore, a machine learning, i.e., data-based learning, approach to the design of lipid-mediated API delivery is introduced. The data produced by the experimental and theoretical approaches provide valuable insights. Processing these data can help optimize the design of LNCs for better performance. In the final section of this Review, state-of-the-art of computer simulations of LNCs are reviewed, specifically addressing the compatibility of experimental and computational insights.
Collapse
Affiliation(s)
- Markéta Paloncýová
- Regional
Center of Advanced Technologies and Materials, Czech Advanced Technology and Research Institute (CATRIN), Palacký
University Olomouc, Šlechtitelů 27, 779 00 Olomouc, Czech Republic
| | - Mariana Valério
- Laboratoire
de Biologie et Modélisation de la Cellule, CNRS, UMR 5239,
Inserm, U1293, Université Claude Bernard Lyon 1, Ecole Normale
Supérieure de Lyon, 46 Allée d’Italie, 69364 Lyon, France
- Centre Blaise
Pascal de Simulation et de Modélisation Numérique, Ecole Normale Supérieure de Lyon, 46 Allée d’Italie, 69364 Lyon, France
| | | | - Petra Kührová
- Regional
Center of Advanced Technologies and Materials, Czech Advanced Technology and Research Institute (CATRIN), Palacký
University Olomouc, Šlechtitelů 27, 779 00 Olomouc, Czech Republic
| | - Martin Šrejber
- Regional
Center of Advanced Technologies and Materials, Czech Advanced Technology and Research Institute (CATRIN), Palacký
University Olomouc, Šlechtitelů 27, 779 00 Olomouc, Czech Republic
| | - Petra Čechová
- Regional
Center of Advanced Technologies and Materials, Czech Advanced Technology and Research Institute (CATRIN), Palacký
University Olomouc, Šlechtitelů 27, 779 00 Olomouc, Czech Republic
| | | | - Akshay Balsubramani
- mRNA Center
of Excellence, Sanofi, Waltham, Massachusetts 02451, United States
| | - Pavel Banáš
- Regional
Center of Advanced Technologies and Materials, Czech Advanced Technology and Research Institute (CATRIN), Palacký
University Olomouc, Šlechtitelů 27, 779 00 Olomouc, Czech Republic
| | - Vikram Agarwal
- mRNA Center
of Excellence, Sanofi, Waltham, Massachusetts 02451, United States
| | - Paulo C. T. Souza
- Laboratoire
de Biologie et Modélisation de la Cellule, CNRS, UMR 5239,
Inserm, U1293, Université Claude Bernard Lyon 1, Ecole Normale
Supérieure de Lyon, 46 Allée d’Italie, 69364 Lyon, France
- Centre Blaise
Pascal de Simulation et de Modélisation Numérique, Ecole Normale Supérieure de Lyon, 46 Allée d’Italie, 69364 Lyon, France
| | - Michal Otyepka
- Regional
Center of Advanced Technologies and Materials, Czech Advanced Technology and Research Institute (CATRIN), Palacký
University Olomouc, Šlechtitelů 27, 779 00 Olomouc, Czech Republic
- IT4Innovations,
VŠB − Technical University of Ostrava, 17. listopadu 2172/15, 708 00 Ostrava-Poruba, Czech Republic
| |
Collapse
|
18
|
Yang H, Li J, Song C, Li H, Luo Q, Chen M. Emerging Gene Therapy Based on Nanocarriers: A Promising Therapeutic Alternative for Cardiovascular Diseases and a Novel Strategy in Valvular Heart Disease. Int J Mol Sci 2025; 26:1743. [PMID: 40004206 PMCID: PMC11855571 DOI: 10.3390/ijms26041743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 02/10/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025] Open
Abstract
Cardiovascular disease remains a leading cause of global mortality, with many unresolved issues in current clinical treatment strategies despite years of extensive research. Due to the great progress in nanotechnology and gene therapy in recent years, the emerging gene therapy based on nanocarriers has provided a promising therapeutic alternative for cardiovascular diseases. This review outlines the status of nanocarriers as vectors in gene therapy for cardiovascular diseases, including coronary heart disease, pulmonary hypertension, hypertension, and valvular heart disease. It discusses challenges and future prospects, aiming to support emerging clinical treatments. This review is the first to summarize gene therapy using nanocarriers for valvular heart disease, highlighting their potential in targeting challenging tissues.
Collapse
Affiliation(s)
- Haoran Yang
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, Chengdu 610041, China; (H.Y.); (J.L.); (C.S.)
- Department of Cardiology, West China Hospital, Sichuan University, No.37 Guoxue Street, Chengdu 610041, China
| | - Junli Li
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, Chengdu 610041, China; (H.Y.); (J.L.); (C.S.)
- Cardiac Structure and Function Research Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Chengxiang Song
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, Chengdu 610041, China; (H.Y.); (J.L.); (C.S.)
- Department of Cardiology, West China Hospital, Sichuan University, No.37 Guoxue Street, Chengdu 610041, China
| | - Hongde Li
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, Chengdu 610041, China; (H.Y.); (J.L.); (C.S.)
- Department of Cardiology, West China Hospital, Sichuan University, No.37 Guoxue Street, Chengdu 610041, China
| | - Qiang Luo
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, Chengdu 610041, China; (H.Y.); (J.L.); (C.S.)
- Department of Cardiology, West China Hospital, Sichuan University, No.37 Guoxue Street, Chengdu 610041, China
- Cardiac Structure and Function Research Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Mao Chen
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, Chengdu 610041, China; (H.Y.); (J.L.); (C.S.)
- Department of Cardiology, West China Hospital, Sichuan University, No.37 Guoxue Street, Chengdu 610041, China
- Cardiac Structure and Function Research Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
19
|
Tenchov R, Hughes KJ, Ganesan M, Iyer KA, Ralhan K, Lotti Diaz LM, Bird RE, Ivanov JM, Zhou QA. Transforming Medicine: Cutting-Edge Applications of Nanoscale Materials in Drug Delivery. ACS NANO 2025; 19:4011-4038. [PMID: 39823199 PMCID: PMC11803921 DOI: 10.1021/acsnano.4c09566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 12/23/2024] [Accepted: 12/27/2024] [Indexed: 01/19/2025]
Abstract
Since their inception in the early 1960s, the development and use of nanoscale materials have progressed tremendously, and their roles in diverse fields ranging from human health to energy and electronics are undeniable. The application of nanotechnology inventions has revolutionized many aspects of everyday life including various medical applications and specifically drug delivery systems, maximizing the therapeutic efficacy of the contained drugs by means of bioavailability enhancement or minimization of adverse effects. In this review, we utilize the CAS Content Collection, a vast repository of scientific information extracted from journal and patent publications, to analyze trends in nanoscience research relevant to drug delivery in an effort to provide a comprehensive and detailed picture of the use of nanotechnology in this field. We examine the publication landscape in the area to provide insights into current knowledge advances and developments. We review the major classes of nanosized drug delivery systems, their delivery routes, and targeted diseases. We outline the most discussed concepts and assess the advantages of various nanocarriers. The objective of this review is to provide a broad overview of the evolving landscape of current knowledge regarding nanosized drug delivery systems, to outline challenges, and to evaluate growth opportunities. The merit of the review stems from the extensive, wide-ranging coverage of the most up-to-date scientific information, allowing unmatched breadth of landscape analysis and in-depth insights.
Collapse
Affiliation(s)
- Rumiana Tenchov
- CAS,
a division of the American Chemical Society, Columbus, Ohio 43210, United States
| | - Kevin J. Hughes
- CAS,
a division of the American Chemical Society, Columbus, Ohio 43210, United States
| | | | - Kavita A. Iyer
- CAS,
a division of the American Chemical Society, Columbus, Ohio 43210, United States
| | | | - Leilani M. Lotti Diaz
- CAS,
a division of the American Chemical Society, Columbus, Ohio 43210, United States
| | - Robert E. Bird
- CAS,
a division of the American Chemical Society, Columbus, Ohio 43210, United States
| | - Julian M. Ivanov
- CAS,
a division of the American Chemical Society, Columbus, Ohio 43210, United States
| | | |
Collapse
|
20
|
Clarissa EM, Karmacharya M, Choi H, Kumar S, Cho YK. Nature Inspired Delivery Vehicles for CRISPR-Based Genome Editing. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025:e2409353. [PMID: 39901476 DOI: 10.1002/smll.202409353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 01/16/2025] [Indexed: 02/05/2025]
Abstract
The advent of Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)-based genome editing technologies has opened up groundbreaking possibilities for treating a wide spectrum of genetic disorders and diseases. However, the success of these technologies relies heavily on the development of efficient and safe delivery systems. Among the most promising approaches are natural and synthetic nanocarrier-mediated delivery systems, including viral vectors, extracellular vesicles (EVs), engineered cellular membrane particles, liposomes, and various nanoparticles. These carriers enhance the efficacy of the CRISPR system by providing a unique combination of efficiency, specificity, and reduced immunogenicity. Synthetic carriers such as liposomes and nanoparticles facilitate CRISPR delivery with high reproducibility and customizable functions. Viral vectors, renowned for their high transduction efficiency and broad tropism, serve as powerful vehicles for delivering CRISPR components to various cell types. EVs, as natural carriers of RNA and proteins, offer a stealth mechanism to evade immune detection, allowing for the targeted delivery of genome editors with minimal off-target effects. Engineered cellular membrane particles further improve delivery by simulating the cellular environment, enhancing uptake, and minimizing immune response. This review explores the innovative integration of CRISPR genome editors with various nanocarrier systems, focusing on recent advancements, applications, and future directions in therapeutic genome editing.
Collapse
Affiliation(s)
- Elizabeth Maria Clarissa
- Center for Algorithmic and Robotized Synthesis, Institute for Basic Science (IBS), UNIST-gil 50, Ulsan, 44919, Republic of Korea
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), UNIST-gil 50, Ulsan, 44919, Republic of Korea
| | - Mamata Karmacharya
- Center for Algorithmic and Robotized Synthesis, Institute for Basic Science (IBS), UNIST-gil 50, Ulsan, 44919, Republic of Korea
| | - Hyunmin Choi
- Center for Algorithmic and Robotized Synthesis, Institute for Basic Science (IBS), UNIST-gil 50, Ulsan, 44919, Republic of Korea
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), UNIST-gil 50, Ulsan, 44919, Republic of Korea
| | - Sumit Kumar
- Center for Algorithmic and Robotized Synthesis, Institute for Basic Science (IBS), UNIST-gil 50, Ulsan, 44919, Republic of Korea
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), UNIST-gil 50, Ulsan, 44919, Republic of Korea
| | - Yoon-Kyoung Cho
- Center for Algorithmic and Robotized Synthesis, Institute for Basic Science (IBS), UNIST-gil 50, Ulsan, 44919, Republic of Korea
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), UNIST-gil 50, Ulsan, 44919, Republic of Korea
| |
Collapse
|
21
|
Stokes EG, Vasquez JJ, Azouz G, Nguyen M, Tierno A, Zhuang Y, Galinato VM, Hui M, Toledano M, Tyler I, Shi X, Hunt RF, Aoto J, Beier KT. Cationic peptides cause memory loss through endophilin-mediated endocytosis. Nature 2025; 638:479-489. [PMID: 39814881 DOI: 10.1038/s41586-024-08413-w] [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: 11/04/2023] [Accepted: 11/14/2024] [Indexed: 01/18/2025]
Abstract
The zeta inhibitory peptide (ZIP) interferes with memory maintenance and long-term potentiation (LTP)1 when administered to mice. However, mice lacking its putative target, protein kinase PKMζ, exhibit normal learning and memory as well as LTP2,3, making the mechanism of ZIP unclear. Here we show that ZIP disrupts LTP by removing surface AMPA receptors through its cationic charge alone. This effect requires endophilin-A2-mediated endocytosis and is fully blocked by drugs suppressing macropinocytosis. ZIP and other cationic peptides remove newly inserted AMPA receptor nanoclusters at potentiated synapses, providing a mechanism by which these peptides erase memories without altering basal synaptic function. When delivered in vivo, cationic peptides can modulate memories on local and brain-wide scales, and these mechanisms can be leveraged to prevent memory loss in a model of traumatic brain injury. Our findings uncover a previously unknown synaptic mechanism by which memories are maintained or lost.
Collapse
Affiliation(s)
- Eric G Stokes
- Pharmacology Graduate Program, University of Colorado Anschutz, Aurora, CO, USA
| | - Jose J Vasquez
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Ghalia Azouz
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Megan Nguyen
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Alexa Tierno
- Department of Anatomy and Neurobiology, University of California, Irvine, Irvine, CA, USA
| | - Yinyin Zhuang
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
| | - Vivienne Mae Galinato
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - May Hui
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Michael Toledano
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Isabella Tyler
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Xiaoyu Shi
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
- Department of Chemistry, University of California, Irvine, Irvine, CA, USA
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, USA
| | - Robert F Hunt
- Department of Anatomy and Neurobiology, University of California, Irvine, Irvine, CA, USA
- Epilepsy Research Center, University of California, Irvine, Irvine, CA, USA
| | - Jason Aoto
- Pharmacology Graduate Program, University of Colorado Anschutz, Aurora, CO, USA
- Department of Pharmacology, University of Colorado Anschutz, Aurora, CO, USA
| | - Kevin T Beier
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA.
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, USA.
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA, USA.
- Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, USA.
| |
Collapse
|
22
|
Sheehan K, Jeon H, Corr SC, Hayes JM, Mok KH. Antibody Aggregation: A Problem Within the Biopharmaceutical Industry and Its Role in AL Amyloidosis Disease. Protein J 2025; 44:1-20. [PMID: 39527351 DOI: 10.1007/s10930-024-10237-6] [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] [Accepted: 10/24/2024] [Indexed: 11/16/2024]
Abstract
Due to the large size and rapid growth of the global therapeutic antibody market, there is major interest in understanding the aggregation of protein products as it can compromise efficacy, concentration, and safety. Various production and storage conditions have been identified as capable of inducing aggregation of polyclonal and monoclonal antibody (mAb) therapies such as low pH, freezing, light exposure, lyophilisation and increased ionic strength. The addition of stabilising excipients to these therapeutics helps to combat the formation of aggregates with future aggregation inhibition mechanisms involving the introduction of point mutations and glycoengineering within aggregation prone regions (APRs). Antibody aggregation also plays an integral role in the pathogenesis of a condition known as amyloid light chain (AL) amyloidosis which is characterised by the production of improperly folded and amyloidogenic immunoglobulin light chains (LCs). Current diagnostic tools rely heavily on histological staining with their future moving towards amyloid component identification and proteomic analysis. For many years, treatment options designed for multiple myeloma (MM) have been applied to AL amyloidosis patients by depleting plasma cell numbers. More recently, treatment strategies more specific to this condition have been developed with many designed to recognize amyloid fibrils and trigger their degradation without causing systemic plasma cell cytotoxicity. Amyloid fibrils in AL disease and aggregates in antibody therapeutics are both formed through the oligomerisation of misfolded / modified proteins attempting to reach a thermodynamically stable, free energy minimum that is lower than the respective monomers themselves. Although the final morphologies are different, by understanding the principles underlying such aggregation, we expect to find common insights that may contribute to the development of new and effective methods of antibody aggregation and/or amyloidosis management. We envision that this area of research will continue to be very relevant in both industry and clinical settings.
Collapse
Affiliation(s)
- Kate Sheehan
- Trinity Biomedical Sciences Institute (TBSI), School of Biochemistry & Immunology, Trinity College Dublin, The University of Dublin, Dublin 2, Ireland
- School of Genetics & Microbiology, Trinity College Dublin, The University of Dublin, Dublin 2, Ireland
| | - Hyesoo Jeon
- Trinity Biomedical Sciences Institute (TBSI), School of Biochemistry & Immunology, Trinity College Dublin, The University of Dublin, Dublin 2, Ireland
- Lonza Biologics Tuas Pte. Ltd., 35 Tuas South Ave 6, Singapore, 637377, Republic of Singapore
| | - Sinéad C Corr
- School of Genetics & Microbiology, Trinity College Dublin, The University of Dublin, Dublin 2, Ireland
- School of Microbiology and APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Jerrard M Hayes
- Trinity Biomedical Sciences Institute (TBSI), School of Biochemistry & Immunology, Trinity College Dublin, The University of Dublin, Dublin 2, Ireland
| | - K H Mok
- Trinity Biomedical Sciences Institute (TBSI), School of Biochemistry & Immunology, Trinity College Dublin, The University of Dublin, Dublin 2, Ireland.
- Centre for Research on Adaptive Nanostructures and Nanodevices (CRANN), Trinity College Dublin, The University of Dublin, Dublin 2, Ireland.
| |
Collapse
|
23
|
Abedi A, Moosazadeh Moghaddam M, Kachuei R, Imani Fooladi AA. Exosomes as a Therapeutic Strategy in Cancer: Potential Roles as Drug Carriers and Immune Modulators. Biochim Biophys Acta Rev Cancer 2025; 1880:189238. [PMID: 39674417 DOI: 10.1016/j.bbcan.2024.189238] [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: 07/22/2024] [Revised: 12/04/2024] [Accepted: 12/05/2024] [Indexed: 12/16/2024]
Abstract
Exosome-based cancer immunotherapy is advancing quickly on the concept of artificially activating the immune system to combat cancer. They can mechanistically change the tumor microenvironment, increase immune responses, and function as efficient drug delivery vehicles because of their inherent bioactivity, low toxicity, and immunogenicity. Accurate identification of the mechanisms of action of exosomes in tumor environments, along with optimization of their isolation, purification, and characterization methods, is necessary to increase clinical applications. Exosomes can be modified through cargo loading and surface modification to enhance their therapeutic applications, either before or after the donor cells' isolation. These engineered exosomes can directly target tumor cells at the tumor site or indirectly activate innate and adaptive immune responses in the tumor microenvironment. This approach is particularly effective when combined with traditional cancer immunotherapy techniques such as vaccines, immune checkpoints, and CAR-T cells. It can improve anti-tumor responses, induce long-term immunity, and address the limitations of traditional therapies, such as poor penetration in solid tumors and immunosuppressive environments. This review aims to provide a comprehensive and detailed overview of the direct role of engineered exosomes as drug delivery systems and their immunomodulatory effects on tumors as an indirect approach to fighting cancer. Additionally, it will discuss novel immunotherapy options.
Collapse
Affiliation(s)
- Azam Abedi
- Tissue Engineering and Regenerative Medicine Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mehrdad Moosazadeh Moghaddam
- Tissue Engineering and Regenerative Medicine Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Reza Kachuei
- Molecular Biology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Abbas Ali Imani Fooladi
- Applied Microbiology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
24
|
Xu L, Shao Z, Fang X, Xin Z, Zhao S, Zhang H, Zhang Y, Zheng W, Yu X, Zhang Z, Sun L. Exploring precision treatments in immune-mediated inflammatory diseases: Harnessing the infinite potential of nucleic acid delivery. EXPLORATION (BEIJING, CHINA) 2025; 5:20230165. [PMID: 40040830 PMCID: PMC11875455 DOI: 10.1002/exp.20230165] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 04/22/2024] [Indexed: 03/06/2025]
Abstract
Immune-mediated inflammatory diseases (IMIDs) impose an immeasurable burden on individuals and society. While the conventional use of immunosuppressants and disease-modifying drugs has provided partial relief and control, their inevitable side effects and limited efficacy cast a shadow over finding a cure. Promising nucleic acid drugs have shown the potential to exert precise effects at the molecular level, with different classes of nucleic acids having regulatory functions through varying mechanisms. For the better delivery of nucleic acids, safe and effective viral vectors and non-viral delivery systems (including liposomes, polymers, etc.) have been intensively explored. Herein, after describing a range of nucleic acid categories and vectors, we focus on the application of therapeutic nucleic acid delivery in various IMIDs, including rheumatoid arthritis, inflammatory bowel disease, psoriasis, multiple sclerosis, asthma, ankylosing spondylitis, systemic lupus erythematosus, and uveitis. Molecules implicated in inflammation and immune dysregulation are abnormally expressed in a series of IMIDs, and their meticulous modulation through nucleic acid therapy results in varying degrees of remission and improvement of these diseases. By synthesizing findings centered on specific molecular targets, this review delivers a systematic elucidation and perspective towards advancing and utilization of nucleic acid therapeutics for managing IMIDs.
Collapse
Affiliation(s)
- Lingxiao Xu
- Department of Orthopedic SurgeryThe Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Orthopedics Research Institute of Zhejiang UniversityZhejiang University School of MedicineHangzhouChina
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang ProvinceZhejiang University School of MedicineHangzhouChina
- Clinical Research Center of Motor System Disease of Zhejiang ProvinceZhejiang University School of MedicineHangzhouChina
| | - Zhenxuan Shao
- Department of Orthopedic SurgeryThe Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Orthopedics Research Institute of Zhejiang UniversityZhejiang University School of MedicineHangzhouChina
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang ProvinceZhejiang University School of MedicineHangzhouChina
- Clinical Research Center of Motor System Disease of Zhejiang ProvinceZhejiang University School of MedicineHangzhouChina
| | - Xia Fang
- Department of Plastic SurgeryThe Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Zengfeng Xin
- Department of Orthopedic SurgeryThe Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Orthopedics Research Institute of Zhejiang UniversityZhejiang University School of MedicineHangzhouChina
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang ProvinceZhejiang University School of MedicineHangzhouChina
- Clinical Research Center of Motor System Disease of Zhejiang ProvinceZhejiang University School of MedicineHangzhouChina
| | - Shenzhi Zhao
- Department of Orthopedic SurgeryThe Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Orthopedics Research Institute of Zhejiang UniversityZhejiang University School of MedicineHangzhouChina
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang ProvinceZhejiang University School of MedicineHangzhouChina
- Clinical Research Center of Motor System Disease of Zhejiang ProvinceZhejiang University School of MedicineHangzhouChina
| | - Hongbo Zhang
- Pharmaceutical Sciences LaboratoryAbo Akademi UniversityTurkuFinland
| | - Yu Zhang
- Pharmaceutical Sciences LaboratoryAbo Akademi UniversityTurkuFinland
| | - Wenbiao Zheng
- Department of OrthopedicsTaizhou Municipal HospitalTaizhouChina
| | - Xiaohua Yu
- Department of Orthopedic SurgeryThe Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Orthopedics Research Institute of Zhejiang UniversityZhejiang University School of MedicineHangzhouChina
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang ProvinceZhejiang University School of MedicineHangzhouChina
- Clinical Research Center of Motor System Disease of Zhejiang ProvinceZhejiang University School of MedicineHangzhouChina
| | - Zengjie Zhang
- Department of Orthopedic SurgeryThe Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Orthopedics Research Institute of Zhejiang UniversityZhejiang University School of MedicineHangzhouChina
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang ProvinceZhejiang University School of MedicineHangzhouChina
- Clinical Research Center of Motor System Disease of Zhejiang ProvinceZhejiang University School of MedicineHangzhouChina
| | - Lingling Sun
- Department of Orthopedic SurgeryThe Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Orthopedics Research Institute of Zhejiang UniversityZhejiang University School of MedicineHangzhouChina
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang ProvinceZhejiang University School of MedicineHangzhouChina
- Clinical Research Center of Motor System Disease of Zhejiang ProvinceZhejiang University School of MedicineHangzhouChina
| |
Collapse
|
25
|
Yang ZF, Jiang XC, Gao JQ. Present insights into the progress in gene therapy delivery systems for central nervous system diseases. Int J Pharm 2025; 669:125069. [PMID: 39662855 DOI: 10.1016/j.ijpharm.2024.125069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 12/06/2024] [Accepted: 12/08/2024] [Indexed: 12/13/2024]
Abstract
Central nervous system (CNS) diseases, including Alzheimer's disease (AD), Parkinson's disease (PD), spinal cord injury (SCI), and ischemic strokes and certain rare diseases, such as amyotrophic lateral sclerosis (ALS) and ataxia, present significant obstacles to treatment using conventional molecular pharmaceuticals. Gene therapy, with its ability to target previously "undruggable" proteins with high specificity and safety, is increasingly utilized in both preclinical and clinical research for CNS ailments. As our comprehension of the pathophysiology of these conditions deepens, gene therapy stands out as a versatile and promising strategy with the potential to both prevent and treat these diseases. Despite the remarkable progress in refining and enhancing the structural design of gene therapy agents, substantial obstacles persist in their effective and safe delivery within living systems. To surmount these obstacles, a diverse array of gene delivery systems has been devised and continuously improved. Notably, Adeno-Associated Virus (AAVs)-based viral gene vectors and lipid-based nanocarriers have each advanced the in vivo delivery of gene therapies to various extents. This review aims to concisely summarize the pathophysiological foundations of CNS diseases and to shed light on the latest advancements in gene delivery vector technologies. It discusses the primary categories of these vectors, their respective advantages and limitations, and their specialized uses in the context of gene therapy delivery.
Collapse
Affiliation(s)
- Ze-Feng Yang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Xin-Chi Jiang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China..
| | - Jian-Qing Gao
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China..
| |
Collapse
|
26
|
Chen S, Dou Y, Zhang Y, Sun X, Liu X, Yang Q. Innovating intervertebral disc degeneration therapy: Harnessing the power of extracellular vesicles. J Orthop Translat 2025; 50:44-55. [PMID: 39868351 PMCID: PMC11761297 DOI: 10.1016/j.jot.2024.09.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 09/11/2024] [Accepted: 09/26/2024] [Indexed: 01/28/2025] Open
Abstract
Intervertebral disc degeneration is the leading cause of low back pain, imposing significant burdens on patients, societies, and economies. Advancements in regenerative medicine have spotlighted extracellular vesicles as promising nanoparticles for intervertebral disc degeneration treatment. Extracellular vesicles retain the potential of cell therapy and serve as carriers to deliver their cargo to target cells, thereby regulating cell activity. This review summarizes the biogenesis and molecular composition of extracellular vesicles and explores their therapeutic roles in intervertebral disc degeneration treatment through various mechanisms. These mechanisms include mitigating cell loss and senescence, delaying extracellular matrix degeneration, and modulating the inflammatory microenvironment. Additionally, it highlights recent efforts in engineering extracellular vesicles to enhance their targeting and therapeutic efficacy. The integration of extracellular vesicle-based acellular therapy is anticipated to drive significant advancements in disc regenerative medicine. The translational potential of this article Existing clinical treatment strategies often fail to effectively address the challenges associated with regenerating degenerated intervertebral discs. As a new regenerative medicine strategy, the extracellular vesicle strategy avoids the risks associated with cell transplantation and shows great promise in treating intervertebral disc degeneration by carrying therapeutic cargo. This review comprehensively examines the latest research, underlying mechanisms, and therapeutic potential of extracellular vesicles, offering a promising new strategy for intervertebral disc degeneration treatment.
Collapse
Affiliation(s)
- Shanfeng Chen
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, China
- Clinical School of Orthopedics, Tianjin Medical University, Tianjin, China
| | - Yiming Dou
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, China
| | - Yiming Zhang
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, China
- Clinical School of Orthopedics, Tianjin Medical University, Tianjin, China
| | - Xun Sun
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, China
| | - Xinyu Liu
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Qiang Yang
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, China
| |
Collapse
|
27
|
Kendre PN, Kayande DR, Pote AK, Kanthale SB, Prajapati BG, Kendre Y, Jain S. Emerging Lipid-based Carriers for Systematic Utilization in the Pharmaceutical and Biomedical Sciences: A Review. Pharm Nanotechnol 2025; 13:2-21. [PMID: 38284709 DOI: 10.2174/0122117385268268231204061938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 10/07/2023] [Accepted: 10/16/2023] [Indexed: 01/30/2024]
Abstract
Emerging lipid-based carriers are revolutionizing drug delivery in the pharmaceutical and biomedical sciences. These innovative carriers harness the unique properties of lipids to improve the solubility, stability, and targeted delivery of therapeutic agents, ushering in a new era of precision medicine. Lipid- based carriers, such as liposomes, lipid nanoparticles, and solid lipid nanoparticles, offer several advantages. They can encapsulate both hydrophilic and hydrophobic drugs, enabling the delivery of a wide range of compounds. Additionally, lipids are biocompatible and biodegradable, minimizing the risk of toxicity. Their ability to mimic cell membranes allows for enhanced cellular uptake and controlled release, optimizing drug efficacy while minimizing side effects. Furthermore, lipid-based carriers are ideal for delivering drugs to specific sites within the body. By modifying the lipid composition, surface charge, and size, researchers can tailor these carriers to target tumours, inflamed tissues, or specific cells, improving therapeutic outcomes and reducing systemic toxicity. In summary, emerging lipid-based carriers are poised to transform pharmaceutical and biomedical sciences by addressing critical challenges in drug delivery. These carriers enhance drug stability, bioavailability, and targeted delivery, offering the potential to revolutionize the treatment of various diseases and improve patient outcomes. As research in this field continues to advance, we can expect even more sophisticated lipid-based carrier systems to emerge, further expanding the possibilities for precision medicine. This review focuses on the contribution of lipid carriers in the pharmaceutical and biomedical sciences.
Collapse
Affiliation(s)
- Prakash N Kendre
- Rajarshi Shahu College of Pharmacy, Buldhana, 443001, Maharashtra, India
| | - Dhiraj R Kayande
- Rajarshi Shahu College of Pharmacy, Buldhana, 443001, Maharashtra, India
| | - Ajinkya K Pote
- Department of Pharmaceutics, Rajarshi Shahu College of Pharmacy, Khamgaon-Botha Road, Malvihir, Buldhana, 443001, India
| | | | - Bhupendra G Prajapati
- S. K. Patel College of Pharmaceutical Education and Research, Ganpat University, Ganpat Vidya Nagar, Mehsana, 384012, Gujrat, India
| | - Yuvraj Kendre
- Podar International School, Buldhana, 443001, Maharashtra, India
| | - Shirish Jain
- Rajarshi Shahu College of Pharmacy, Buldhana, 443001, Maharashtra, India
| |
Collapse
|
28
|
Cao Y, Yan W, Yi W, Yin Q, Li Y. Bioengineered therapeutic systems for improving antitumor immunity. Natl Sci Rev 2025; 12:nwae404. [PMID: 40114728 PMCID: PMC11925021 DOI: 10.1093/nsr/nwae404] [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: 08/22/2024] [Revised: 09/29/2024] [Accepted: 11/08/2024] [Indexed: 03/22/2025] Open
Abstract
Immunotherapy, a monumental advancement in antitumor therapy, still yields limited clinical benefits owing to its unguaranteed efficacy and safety. Therapeutic systems derived from cellular, bacterial and viral sources possess inherent properties that are conducive to antitumor immunotherapy. However, crude biomimetic systems have restricted functionality and may produce undesired toxicity. With advances in biotechnology, various toolkits are available to add or subtract certain properties of living organisms to create flexible therapeutic platforms. This review elaborates on the creation of bioengineered systems, via gene editing, synthetic biology and surface engineering, to enhance immunotherapy. The modifying strategies of the systems are discussed, including equipment for navigation and recognition systems to improve therapeutic precision, the introduction of controllable components to control the duration and intensity of treatment, the addition of immunomodulatory components to amplify immune activation, and the removal of toxicity factors to ensure biosafety. Finally, we summarize the advantages of bioengineered immunotherapeutic systems and possible directions for their clinical translation.
Collapse
Affiliation(s)
- Ying Cao
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Life Sciences, Jilin University, Changchun 130012, China
| | - Wenlu Yan
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wenzhe Yi
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qi Yin
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Yantai 264000, China
| | - Yaping Li
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Yantai 264000, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264000, China
| |
Collapse
|
29
|
Yi Z, Jia Y, Lu R, Li C, Wen L, Yin X, Yi J, Li L. E2F1-driven CENPM expression promotes glycolytic reprogramming and tumorigenicity in glioblastoma. Cell Biol Toxicol 2024; 41:4. [PMID: 39707034 PMCID: PMC11662047 DOI: 10.1007/s10565-024-09945-7] [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: 03/18/2024] [Accepted: 11/04/2024] [Indexed: 12/23/2024]
Abstract
Centromere protein M (CENPM), traditionally associated with chromosome segregation, is now recognized for its significant role in cancer biology. Particularly in glioblastoma (GBM), where less is known about CENPM compared to other centromere proteins (CENPs), it appears crucially involved in regulating tumor cell proliferation, invasion, and metabolic reprogramming-key factors in GBM's aggressiveness. Initial analyses using the GEPIA database (TCGA/GTEx datasets) reveal distinct patterns of CENPM expression in GBM, suggesting its potential as a therapeutic target. Our study manipulated CENPM expression through shRNA-mediated knockdown and vector-based overexpression in GBM cell lines LN229 and U251. Knockdown resulted in a 50% reduction in cell proliferation and a 70% decrease in invasion, accompanied by diminished glycolytic markers such as glucose consumption, lactate production, and ATP levels. Conversely, overexpression of CENPM enhanced both metabolic activity and invasive capacities. The introduction of the glycolytic inhibitor 2-DG effectively reversed the effects of CENPM modulation, highlighting a dependency on glycolytic pathways. Moreover, we identified E2F1 as a key regulator of CENPM, linking it to GBM's metabolic alterations. In vivo studies using a BALB/c nude mouse xenograft model demonstrated that CENPM knockdown significantly inhibits tumor growth, with treated groups showing a 60% reduction in tumor volume over four weeks. These findings underscore the E2F1-CENPM axis as a promising target for therapeutic strategies, aiming to disrupt the metabolic and invasive pathways facilitated by CENPM in GBM. These insights establish a foundation for targeting the metabolic dependencies of tumor cells, potentially leading to innovative treatments for GBM.
Collapse
Affiliation(s)
- Zhiqiang Yi
- Department of Neurosurgery, Peking University First Hospital, No. 8 Xishiku Street, Xicheng District, Beijing, 100034, China
| | - Yanfei Jia
- Department of Neurosurgery, Peking University First Hospital, No. 8 Xishiku Street, Xicheng District, Beijing, 100034, China
| | - Runchun Lu
- Department of Neurosurgery, Peking University First Hospital, No. 8 Xishiku Street, Xicheng District, Beijing, 100034, China
| | - Chunwei Li
- Department of Neurosurgery, Peking University First Hospital, No. 8 Xishiku Street, Xicheng District, Beijing, 100034, China
| | - Long Wen
- Department of Neurosurgery, Peking University First Hospital, No. 8 Xishiku Street, Xicheng District, Beijing, 100034, China
| | - Xiangdong Yin
- Department of Neurosurgery, Peking University First Hospital, No. 8 Xishiku Street, Xicheng District, Beijing, 100034, China
| | - Junfei Yi
- Department of Neurosurgery, Peking University First Hospital, No. 8 Xishiku Street, Xicheng District, Beijing, 100034, China
| | - Liang Li
- Department of Neurosurgery, Peking University First Hospital, No. 8 Xishiku Street, Xicheng District, Beijing, 100034, China.
| |
Collapse
|
30
|
Pavlov RV, Akimov SA, Dashinimaev EB, Bashkirov PV. Boosting Lipofection Efficiency Through Enhanced Membrane Fusion Mechanisms. Int J Mol Sci 2024; 25:13540. [PMID: 39769303 PMCID: PMC11677079 DOI: 10.3390/ijms252413540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 12/11/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025] Open
Abstract
Gene transfection is a fundamental technique in the fields of biological research and therapeutic innovation. Due to their biocompatibility and membrane-mimetic properties, lipid vectors serve as essential tools in transfection. The successful delivery of genetic material into the cytoplasm is contingent upon the fusion of the vector and cellular membranes, which enables hydrophilic polynucleic acids to traverse the hydrophobic barriers of two intervening membranes. This review examines the critical role of membrane fusion in lipofection efficiency, with a particular focus on the molecular mechanisms that govern lipoplex-membrane interactions. This analysis will examine the key challenges inherent to the fusion process, from achieving initial membrane proximity to facilitating final content release through membrane remodeling. In contrast to viral vectors, which utilize specialized fusion proteins, lipid vectors necessitate a strategic formulation and environmental optimization to enhance their fusogenicity. This review discusses recent advances in vector design and fusion-promoting strategies, emphasizing their potential to improve gene delivery yield. It highlights the importance of understanding lipoplex-membrane fusion mechanisms for developing next-generation delivery systems and emphasizes the need for continued fundamental research to advance lipid-mediated transfection technology.
Collapse
Affiliation(s)
- Rais V. Pavlov
- Research Institute for Systems Biology and Medicine, 18 Nauchniy Proezd, Moscow 117246, Russia
| | - Sergey A. Akimov
- Frumkin Institute of Physical Chemistry and Electrochemistry, Russian Academy of Sciences, 31/4 Leninskiy Prospekt, Moscow 119071, Russia;
| | - Erdem B. Dashinimaev
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow 117997, Russia;
| | - Pavel V. Bashkirov
- Research Institute for Systems Biology and Medicine, 18 Nauchniy Proezd, Moscow 117246, Russia
| |
Collapse
|
31
|
Wang X, Dong J, Gao M, Wang Y, Hou F, Wang J, Li Y, Wang Z. Radial mechanical properties of deoxyribonucleic acid molecules. NANOTECHNOLOGY 2024; 36:085101. [PMID: 39660784 DOI: 10.1088/1361-6528/ad9930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 12/02/2024] [Indexed: 12/12/2024]
Abstract
Given the small diameter of deoxyribonucleic acid (DNA), the difficulty in studying its radial mechanical properties laid in the challenge of applying a precise and controlled small force. In this work, the radial mechanical properties of DNA were measured in the AFM. DNA adhesion properties were analyzed through force-distance curves and adhesion images. The adhesion force values applied on DNA obtained from the force-distance curves were consistent with those obtained from the adhesion images. The Young's modulus of DNA was determined by collecting the data of indentation depth and the force applied on DNA and using the Hertz model for calculation. At the same compression speed, the Young's moduli increased with increasing forces, but exhibited a nonlinear growth. This reflected the complex stress-strain behavior of DNA. The impact of speeds on mechanical properties of DNA was explored. Higher speed resulted in greater Young's moduli and adhesion. This study not only deepens the understanding the mechanical properties of DNA, but also provides a strategy for investigating the mechanical properties of other thin and soft materials.
Collapse
Affiliation(s)
- Xia Wang
- International Research Centre for Nano Handling and Manufacturing of China, Changchun University of Science and Technology, Changchun 130022, People's Republic of China
- Centre for Opto/Bio-Nano Measurement and Manufacturing, Zhongshan Institute of Changchun University of Science and Technology, Zhongshan 528437, People's Republic of China
- Ministry of Education Key Laboratory for Cross-Scale Micro and Nano Manufacturing, Changchun University of Science and Technology, Changchun 130022, People's Republic of China
| | - Jianjun Dong
- International Research Centre for Nano Handling and Manufacturing of China, Changchun University of Science and Technology, Changchun 130022, People's Republic of China
- Centre for Opto/Bio-Nano Measurement and Manufacturing, Zhongshan Institute of Changchun University of Science and Technology, Zhongshan 528437, People's Republic of China
- Ministry of Education Key Laboratory for Cross-Scale Micro and Nano Manufacturing, Changchun University of Science and Technology, Changchun 130022, People's Republic of China
| | - Mingyan Gao
- International Research Centre for Nano Handling and Manufacturing of China, Changchun University of Science and Technology, Changchun 130022, People's Republic of China
- Centre for Opto/Bio-Nano Measurement and Manufacturing, Zhongshan Institute of Changchun University of Science and Technology, Zhongshan 528437, People's Republic of China
- Ministry of Education Key Laboratory for Cross-Scale Micro and Nano Manufacturing, Changchun University of Science and Technology, Changchun 130022, People's Republic of China
| | - Ying Wang
- International Research Centre for Nano Handling and Manufacturing of China, Changchun University of Science and Technology, Changchun 130022, People's Republic of China
- Centre for Opto/Bio-Nano Measurement and Manufacturing, Zhongshan Institute of Changchun University of Science and Technology, Zhongshan 528437, People's Republic of China
| | - Fengyan Hou
- International Research Centre for Nano Handling and Manufacturing of China, Changchun University of Science and Technology, Changchun 130022, People's Republic of China
- Centre for Opto/Bio-Nano Measurement and Manufacturing, Zhongshan Institute of Changchun University of Science and Technology, Zhongshan 528437, People's Republic of China
- Ministry of Education Key Laboratory for Cross-Scale Micro and Nano Manufacturing, Changchun University of Science and Technology, Changchun 130022, People's Republic of China
| | - Jingyu Wang
- International Research Centre for Nano Handling and Manufacturing of China, Changchun University of Science and Technology, Changchun 130022, People's Republic of China
- Centre for Opto/Bio-Nano Measurement and Manufacturing, Zhongshan Institute of Changchun University of Science and Technology, Zhongshan 528437, People's Republic of China
- Ministry of Education Key Laboratory for Cross-Scale Micro and Nano Manufacturing, Changchun University of Science and Technology, Changchun 130022, People's Republic of China
| | - Ye Li
- International Research Centre for Nano Handling and Manufacturing of China, Changchun University of Science and Technology, Changchun 130022, People's Republic of China
- Centre for Opto/Bio-Nano Measurement and Manufacturing, Zhongshan Institute of Changchun University of Science and Technology, Zhongshan 528437, People's Republic of China
- Ministry of Education Key Laboratory for Cross-Scale Micro and Nano Manufacturing, Changchun University of Science and Technology, Changchun 130022, People's Republic of China
| | - Zuobin Wang
- International Research Centre for Nano Handling and Manufacturing of China, Changchun University of Science and Technology, Changchun 130022, People's Republic of China
- Centre for Opto/Bio-Nano Measurement and Manufacturing, Zhongshan Institute of Changchun University of Science and Technology, Zhongshan 528437, People's Republic of China
- Ministry of Education Key Laboratory for Cross-Scale Micro and Nano Manufacturing, Changchun University of Science and Technology, Changchun 130022, People's Republic of China
- JR3CN & IRAC, University of Bedfordshire, Luton LU1 3JU, United Kingdom
| |
Collapse
|
32
|
Han Y, Wang M, Chen Y, Ouyang D, Zheng Y, Hu Y. Profiling patent compounds in lipid nanoparticle formulations of siRNA. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102362. [PMID: 39554995 PMCID: PMC11565460 DOI: 10.1016/j.omtn.2024.102362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 10/16/2024] [Indexed: 11/19/2024]
Abstract
Lipid nanoparticles (LNPs) have emerged as a prominent delivery system for nucleic acid drugs, attracting significant attention, especially through the successful development of several commercial products. As a key component in LNPs, cationic lipids have long served as a key technical barrier to block competitors by building up a complex patent thicket. However, there have been few studies as yet that have comprehensively analyzed the patented compounds in LNP formulations, despite a large number of technical reviews and original articles. In this context, this study focuses on analyzing the macroscopic landscapes and microscopic molecular characteristics of LNP patents, aiming to provide a valuable reference for researchers and developers in making informed technological and commercial decisions. By mining 2,994 patents, 265 formulas, 7,674 compounds, and 28,789 fragments, this work sketches the empirical golden ratio of lipid materials in LNP formulation, discloses the advanced technology in the formulation, characterizes high-frequency fragments of heads, linkers and tails in both novel cationic lipids as well as targeting lipids, and establishes a virtual focus library of LNP materials.
Collapse
Affiliation(s)
- Yunfeng Han
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China
| | - Mengyang Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China
| | - Yu Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China
| | - Defang Ouyang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China
- Centre for Pharmaceutical Regulatory Sciences, University of Macau, Macao SAR, China
- DPM, Faculty of Health Sciences, University of Macau, Macao SAR, China
| | - Ying Zheng
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China
- DPS, Faculty of Health Sciences, University of Macau, Macao SAR, China
| | - Yuanjia Hu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China
- Centre for Pharmaceutical Regulatory Sciences, University of Macau, Macao SAR, China
- DPM, Faculty of Health Sciences, University of Macau, Macao SAR, China
| |
Collapse
|
33
|
Haque MA, Shrestha A, Mikelis CM, Mattheolabakis G. Comprehensive analysis of lipid nanoparticle formulation and preparation for RNA delivery. Int J Pharm X 2024; 8:100283. [PMID: 39309631 PMCID: PMC11415597 DOI: 10.1016/j.ijpx.2024.100283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 08/21/2024] [Accepted: 09/07/2024] [Indexed: 09/25/2024] Open
Abstract
Nucleic acid-based therapeutics are a common approach that is increasingly popular for a wide spectrum of diseases. Lipid nanoparticles (LNPs) are promising delivery carriers that provide RNA stability, with strong transfection efficiency, favorable and tailorable pharmacokinetics, limited toxicity, and established translatability. In this review article, we describe the lipid-based delivery systems, focusing on lipid nanoparticles, the need of their use, provide a comprehensive analysis of each component, and highlight the advantages and disadvantages of the existing manufacturing processes. We further summarize the ongoing and completed clinical trials utilizing LNPs, indicating important aspects/questions worth of investigation, and analyze the future perspectives of this significant and promising therapeutic approach.
Collapse
Affiliation(s)
- Md. Anamul Haque
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA
| | - Archana Shrestha
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA
| | - Constantinos M. Mikelis
- Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras 26504, Greece
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - George Mattheolabakis
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA
| |
Collapse
|
34
|
Mirhadi E, Kesharwani P, Jha SK, Karav S, Sahebkar A. Utilizing ionic liquids as eco-friendly and sustainable carriers for delivering nucleic acids: A review on the revolutionary advancement in nano delivery systems. Int J Biol Macromol 2024; 283:137582. [PMID: 39542300 DOI: 10.1016/j.ijbiomac.2024.137582] [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: 09/07/2024] [Revised: 11/04/2024] [Accepted: 11/11/2024] [Indexed: 11/17/2024]
Abstract
Ionic liquids (ILs) are an extremely versatile class of chemicals. It has been shown that they can effectively pass through many biological barriers in the human body to deliver medications. ILs are solvents noted for their ecological friendliness; they contain equal amounts of cations and anions and remain liquid at temperatures below 100 °C. Hence, these are ideal for biomedical applications owing to their advantageous properties such as biocompatibility, solubility, and adaptability. ILs are widely reported to improve the solubility and stability of nucleic acids (DNA and RNA) in aqueous conditions, allowing for more effective delivery. Certain ILs have shown the ability to enhance the absorption of nucleic acids into cells. In addition, ILs can also be used to create vectors for gene delivery, such as liposomes and nanoparticles, thereby improving the transfection efficiency of plasmid DNA and siRNA. Subsequently, the application of ILs for nucleic acid delivery has increased significantly in recent years. In this context, we believe that using ILs to enhance the transport of nucleic acids will have a considerable effect as a novel and crucial therapeutic method in the upcoming decades. The use of ILs as solvents to preserve the natural structure of DNA and RNA shows promise for a variety of biotechnological and medical applications. Notably, ILs may be utilized for a variety of functions, including extracting, concentrating, stabilizing, and spreading nucleic acids inside cells. Our review emphasizes the key findings of research works published in this domain, wherein outstanding effectiveness of delivering RNA to the desired areas was achieved, and was made possible through the utilization of ILs.
Collapse
Affiliation(s)
- Elaheh Mirhadi
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India.
| | - Saurav Kumar Jha
- Department of Biological Sciences and Bioengineering (BSBE), Indian Institute of Technology, Kanpur 208016, Uttar Pradesh, India
| | - Sercan Karav
- Department of Molecular Biology and Genetics, Canakkale Onsekiz Mart University, Canakkale 17100, Turkey
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
35
|
Eker Fidan EB, Bal K, Şentürk S, Kaplan Ö, Demir K, Gök MK. Enhancing gene delivery efficiency with amphiphilic chitosan modified by myristic acid and tertiary amino groups. Int J Biol Macromol 2024; 282:136775. [PMID: 39442853 DOI: 10.1016/j.ijbiomac.2024.136775] [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: 03/25/2024] [Revised: 10/13/2024] [Accepted: 10/19/2024] [Indexed: 10/25/2024]
Abstract
The aim of this study is to synthesize new amphiphilic chitosan containing myristic acid as the hydrophobic tail and tertiary amine groups as the hydrophilic head and to evaluate the gene delivery efficiency. In this context, the primary amine groups of chitosan were first modified with myristic acid (Chi-M), followed by the modification of the methylol groups with 3-dimethylamino-1-propyl chloride hydrochloride. The chemical characterization of this chitosan formulation (Chi-MA) was determined using nuclear magnetic resonance (NMR), Fourier-transform infrared spectroscopy (FTIR) analysis and gel permeation chromatography-size exclusion chromatography. Chi-MA nanoparticles were prepared via ionic gelation, and particle size, polydispersity and zeta potential were determined. The nanoparticles were evaluated for their proton buffering capacity and gene complexing capacity. Additionally, the cytotoxicity of Chi-MA on HEK293T cells was determined via MTT assay, and the transfection efficiency of Chi-MA was analyzed by a flow cytometer. The results indicate a significant increase in gene complexing capacity (8-fold) and nanoparticle formation ability of Chi-MA compared to other chitosan formulations. Chi-MA nanoparticles showed no toxicity against HEK293T cells and exhibited the highest transfection efficiency with significantly lower nanoparticle: gene ratios compared to previous studies. These findings demonstrate the effective use of amphiphilic Chi-MA as a gene carrier.
Collapse
Affiliation(s)
- Emine Büşra Eker Fidan
- Istanbul University-Cerrahpasa, Faculty of Engineering, Department of Chemical Engineering, Istanbul, Turkey
| | - Kevser Bal
- Istanbul University-Cerrahpasa, Faculty of Engineering, Department of Chemical Engineering, Istanbul, Turkey
| | - Sema Şentürk
- Istanbul University-Cerrahpasa, Faculty of Engineering, Department of Chemical Engineering, Istanbul, Turkey
| | - Özlem Kaplan
- Alanya Alaaddin Keykubat University, Rafet Kayış Faculty of Engineering, Department of Genetics and Bioengineering, Antalya, Turkey
| | - Kamber Demir
- Department of Reproduction and Artificial Insemination, Faculty of Veterinary Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Mehmet Koray Gök
- Istanbul University-Cerrahpasa, Faculty of Engineering, Department of Chemical Engineering, Istanbul, Turkey.
| |
Collapse
|
36
|
Piñeiro-Silva C, Gadea J. Optimization of lipofection protocols for CRISPR/Cas9 delivery in porcine zona pellucida intact oocytes: A study of coincubation duration and reagent efficacy. Theriogenology 2024; 230:121-129. [PMID: 39293174 DOI: 10.1016/j.theriogenology.2024.09.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 09/03/2024] [Accepted: 09/13/2024] [Indexed: 09/20/2024]
Abstract
A priority to facilitate the application of lipofection to generate genetically modified porcine embryos and animals will be the use of zona pellucida (ZP)-intact oocytes and zygotes. Recently, our group produced genetically modified embryos by lipofection of ZP-intact oocytes during in vitro fertilization (IVF). This study investigates the effect of two commercial lipofection reagents, Lipofectamine 3000 and Lipofectamine CRISPRMAX, on embryo development and mutation efficiency in ZP-intact porcine oocytes. We compared these reagents with the electroporation method and a control group using two sgRNAs targeting the CAPN3 and CD163 genes. The detrimental effects on cleavage rates were observed in both lipofection treatments compared to the control and electroporated groups. However, blastocyst rates were higher in the Lipofectamine 3000 group than in the electroporated group for both genes. Mutation parameters varied by target gene, with Lipofectamine 3000 achieving higher mutation rates for CD163, while all groups were similar for the CAPN3 gene. Overall efficiency was similar for both lipofectamines, confirming their feasibility for use. In addition, we evaluated the effect of coincubation time (4, 8, and 24 h) on IVF outcomes, embryo development, and mutation parameters. Results indicated that an 8-h coincubation period optimized fertilization and mutation efficiency without significant toxic effects. This study demonstrates that lipofection with either Lipofectamine 3000 or CRISPRMAX during IVF is an effective method for generating genetically modified porcine embryos without the need for specialized equipment or trained personnel, with efficiencies similar to or greater than electroporation. This study also highlights the importance of optimizing reagent selection and coincubation times. There is no difference between Lipofectamine 3000 and CRISPRMAXTM in terms of embryo development and mutation efficiency, and under our experimental conditions, the optimal coincubation time with lipofectamine is 8 h.
Collapse
Affiliation(s)
- Celia Piñeiro-Silva
- University of Murcia Dept. Physiology, Murcia, Spain; International Excellence Campus for Higher Education and Research "Campus Mare Nostrum" and Institute for Biomedical Research of Murcia (IMIB-Arrixaca), Murcia, Spain
| | - Joaquín Gadea
- University of Murcia Dept. Physiology, Murcia, Spain; International Excellence Campus for Higher Education and Research "Campus Mare Nostrum" and Institute for Biomedical Research of Murcia (IMIB-Arrixaca), Murcia, Spain.
| |
Collapse
|
37
|
Hameed H, Faheem S, Younas K, Jamshaid M, Ereej N, Hameed A, Munir R, Khokhar R. A comprehensive review on lipid-based nanoparticles via nose to brain targeting as a novel approach. J Microencapsul 2024; 41:681-714. [PMID: 39286884 DOI: 10.1080/02652048.2024.2404414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Accepted: 09/10/2024] [Indexed: 09/19/2024]
Abstract
The central nervous system (CNS) has been a chief concern for millions of people worldwide, and many therapeutic medications are unable to penetrate the blood-brain barrier. Advancements in nanotechnology have enabled safe, effective, and precise delivery of medications towards specific brain regions by utilising a nose-to-brain targeting route. This method reduces adverse effects, increases medication bioavailability, and facilitates mucociliary clearance while promoting accumulation of drug in the targeted brain region. Recent developments in lipid-based nanoparticles, for instance solid lipid nanoparticles (SLNs), liposomes, nanoemulsions, and nano-structured lipid carriers have been explored. SLNs are currently the most promising drug carrier system because of their capability of transporting drugs across the blood-brain barrier at the intended brain site. This approach offers higher efficacy, controlled drug delivery, target specificity, longer circulation time, and a reduction in toxicity through a biomimetic mechanism.
Collapse
Affiliation(s)
- Huma Hameed
- Faculty of Pharmaceutical Sciences, University of Central Punjab, Lahore, Pakistan
| | - Saleha Faheem
- Faculty of Pharmaceutical Sciences, University of Central Punjab, Lahore, Pakistan
| | - Komel Younas
- Faculty of Pharmacy, University Paris Saclay, Orsay, France
| | - Muhammad Jamshaid
- Faculty of Pharmaceutical Sciences, University of Central Punjab, Lahore, Pakistan
| | - Nelofer Ereej
- Faculty of Pharmaceutical Sciences, University of Central Punjab, Lahore, Pakistan
| | - Anam Hameed
- Department of Human Nutrition and Dietetics, Faculty of Rehabilitation and Allied Health Sciences, Riphah International University, Lahore, Pakistan
| | - Rabia Munir
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Government College University Faisalabad, Faisalabad, Pakistan
| | - Rabia Khokhar
- Institute of Pharmaceutical Sciences, University of Veterinary and Animal Sciences, Lahore, Pakistan
| |
Collapse
|
38
|
de Souza AP, Bastos AP, da Fonseca FN, Pandolfi JR, Costamilan CADVLR, Marques MG. Polyethyleneimine-mediated gene transfection in porcine fetal fibroblasts. Anim Reprod 2024; 21:e20240026. [PMID: 39629009 PMCID: PMC11614137 DOI: 10.1590/1984-3143-ar2024-0026] [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: 05/06/2024] [Accepted: 09/04/2024] [Indexed: 12/06/2024] Open
Abstract
Polyethylenimine (PEI) has been explored as an efficient non-viral system for delivering genes to cells; however, there were no protocols for its use in porcine fetal fibroblasts (PFF). Therefore, we compared different concentrations of FITC-PEI (0.625, 1.25, 2.5, 5, 10, 20, 40, or 80 µg/mL) and incubation times (30 min, 1 h, or 2 h). It was observed that the incubation time did not affect the internalization of the PEI-FITC and that 30 min was sufficient to capture the complex. The concentrations higher than 10 µg/mL could reach many marked PFF (>90%). Then, two PEI concentrations were tested, 10 or 40 µg/mL, combined with an N/P of 2 with the pmhyGENIE-5 for 30 min. The percentage of PFF-GFP positive was similar between the PEI concentrations in the evaluation time points (24 h, 48 h, and 72 h). However, 40 µg/mL caused higher membrane damage rates. Thus, it can be concluded that concentrations between 10 - 80 µg/ml of PEI promote high incorporation rates, even in periods as short as 30 minutes. Furthermore, it can be stated that the transfection condition used in Polyplexes 1 (10 µg/mL of PEI and 37.5 µg/mL of pmhyGENIE-5 for 30 min) efficiently produces genetically edited porcine fetal fibroblasts with low cell damage.
Collapse
Affiliation(s)
| | - Ana Paula Bastos
- Empresa Brasileira de Pesquisa Agropecuária, Embrapa Suínos e Aves, Concórdia, SC, Brasil
| | | | - José Rodrigo Pandolfi
- Empresa Brasileira de Pesquisa Agropecuária, Embrapa Suínos e Aves, Concórdia, SC, Brasil
| | | | - Mariana Groke Marques
- Empresa Brasileira de Pesquisa Agropecuária, Embrapa Suínos e Aves, Concórdia, SC, Brasil
- Pós-Graduação em Produção e Sanidade Animal, Instituto Federal Catarinense, Concórdia, SC, Brasil
| |
Collapse
|
39
|
Mrksich K, Padilla MS, Mitchell MJ. Breaking the final barrier: Evolution of cationic and ionizable lipid structure in lipid nanoparticles to escape the endosome. Adv Drug Deliv Rev 2024; 214:115446. [PMID: 39293650 PMCID: PMC11900896 DOI: 10.1016/j.addr.2024.115446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 08/18/2024] [Accepted: 09/07/2024] [Indexed: 09/20/2024]
Abstract
In the past decade, nucleic acid therapies have seen a boon in development and clinical translation largely due to advances in nanotechnology that have enabled their safe and targeted delivery. Nanoparticles can protect nucleic acids from degradation by serum enzymes and can facilitate entry into cells. Still, achieving endosomal escape to allow nucleic acids to enter the cytoplasm has remained a significant barrier, where less than 5% of nanoparticles within the endo-lysosomal pathway are able to transfer their cargo to the cytosol. Lipid-based drug delivery vehicles, particularly lipid nanoparticles (LNPs), have been optimized to achieve potent endosomal escape, and thus have been the vector of choice in the clinic as demonstrated by their utilization in the COVID-19 mRNA vaccines. The success of LNPs is in large part due to the rational design of lipids that can specifically overcome endosomal barriers. In this review, we chart the evolution of lipid structure from cationic lipids to ionizable lipids, focusing on structure-function relationships, with a focus on how they relate to endosomal escape. Additionally, we examine recent advancements in ionizable lipid structure as well as discuss the future of lipid design.
Collapse
Affiliation(s)
- Kaitlin Mrksich
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Marshall S Padilla
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael J Mitchell
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn Institute for RNA Innovation, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
40
|
Bai S, Luo H, Tong H, Wu Y, Yuan Y. Advances on transfer and maintenance of large DNA in bacteria, fungi, and mammalian cells. Biotechnol Adv 2024; 76:108421. [PMID: 39127411 DOI: 10.1016/j.biotechadv.2024.108421] [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: 03/24/2023] [Revised: 05/07/2024] [Accepted: 08/04/2024] [Indexed: 08/12/2024]
Abstract
Advances in synthetic biology allow the design and manipulation of DNA from the scale of genes to genomes, enabling the engineering of complex genetic information for application in biomanufacturing, biomedicine and other areas. The transfer and subsequent maintenance of large DNA are two core steps in large scale genome rewriting. Compared to small DNA, the high molecular weight and fragility of large DNA make its transfer and maintenance a challenging process. This review outlines the methods currently available for transferring and maintaining large DNA in bacteria, fungi, and mammalian cells. It highlights their mechanisms, capabilities and applications. The transfer methods are categorized into general methods (e.g., electroporation, conjugative transfer, induced cell fusion-mediated transfer, and chemical transformation) and specialized methods (e.g., natural transformation, mating-based transfer, virus-mediated transfection) based on their applicability to recipient cells. The maintenance methods are classified into genomic integration (e.g., CRISPR/Cas-assisted insertion) and episomal maintenance (e.g., artificial chromosomes). Additionally, this review identifies the major technological advantages and disadvantages of each method and discusses the development for large DNA transfer and maintenance technologies.
Collapse
Affiliation(s)
- Song Bai
- Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, 300072 Tianjin, China; Frontiers Research Institute for Synthetic Biology, Tianjin University, Tianjin 300072, China
| | - Han Luo
- Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, 300072 Tianjin, China; Frontiers Research Institute for Synthetic Biology, Tianjin University, Tianjin 300072, China
| | - Hanze Tong
- Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, 300072 Tianjin, China; Frontiers Research Institute for Synthetic Biology, Tianjin University, Tianjin 300072, China
| | - Yi Wu
- Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, 300072 Tianjin, China; Frontiers Research Institute for Synthetic Biology, Tianjin University, Tianjin 300072, China. @tju.edu.cn
| | - Yingjin Yuan
- Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, 300072 Tianjin, China; Frontiers Research Institute for Synthetic Biology, Tianjin University, Tianjin 300072, China
| |
Collapse
|
41
|
Wei PS, Thota N, John G, Chang E, Lee S, Wang Y, Ma Z, Tsai YH, Mei KC. Enhancing RNA-lipid nanoparticle delivery: Organ- and cell-specificity and barcoding strategies. J Control Release 2024; 375:366-388. [PMID: 39179112 PMCID: PMC11972657 DOI: 10.1016/j.jconrel.2024.08.030] [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: 04/24/2024] [Revised: 07/25/2024] [Accepted: 08/19/2024] [Indexed: 08/26/2024]
Abstract
Recent advancements in RNA therapeutics highlight the critical need for precision gene delivery systems that target specific organs and cells. Lipid nanoparticles (LNPs) have emerged as key vectors in delivering mRNA and siRNA, offering protection against enzymatic degradation, enabling targeted delivery and cellular uptake, and facilitating RNA cargo release into the cytosol. This review discusses the development and optimization of organ- and cell-specific LNPs, focusing on their design, mechanisms of action, and therapeutic applications. We explore innovations such as DNA/RNA barcoding, which facilitates high-throughput screening and precise adjustments in formulations. We address major challenges, including improving endosomal escape, minimizing off-target effects, and enhancing delivery efficiencies. Notable clinical trials and recent FDA approvals illustrate the practical applications and future potential of LNP-based RNA therapies. Our findings suggest that while considerable progress has been made, continued research is essential to resolve existing limitations and bridge the gap between preclinical and clinical evaluation of the safety and efficacy of RNA therapeutics. This review highlights the dynamic progress in LNP research. It outlines a roadmap for future advancements in RNA-based precision medicine.
Collapse
Affiliation(s)
- Pu-Sheng Wei
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Binghamton, Johnson City, New York, NY 13790, USA
| | - Nagasri Thota
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Binghamton, Johnson City, New York, NY 13790, USA
| | - Greshma John
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Binghamton, Johnson City, New York, NY 13790, USA
| | - Evelyn Chang
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Binghamton, Johnson City, New York, NY 13790, USA
| | - Sunjae Lee
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Binghamton, Johnson City, New York, NY 13790, USA
| | - Yuanjun Wang
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Binghamton, Johnson City, New York, NY 13790, USA
| | - Zitao Ma
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Binghamton, Johnson City, New York, NY 13790, USA
| | - Yu-Hsuan Tsai
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Binghamton, Johnson City, New York, NY 13790, USA
| | - Kuo-Ching Mei
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Binghamton, Johnson City, New York, NY 13790, USA.
| |
Collapse
|
42
|
Fajardo C, De Donato M, Macedo M, Charoonnart P, Saksmerprome V, Yang L, Purton S, Mancera JM, Costas B. RNA Interference Applied to Crustacean Aquaculture. Biomolecules 2024; 14:1358. [PMID: 39595535 PMCID: PMC11592254 DOI: 10.3390/biom14111358] [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: 09/20/2024] [Revised: 10/21/2024] [Accepted: 10/23/2024] [Indexed: 11/28/2024] Open
Abstract
RNA interference (RNAi) is a powerful tool that can be used to specifically knock-down gene expression using double-stranded RNA (dsRNA) effector molecules. This approach can be used in aquaculture as an investigation instrument and to improve the immune responses against viral pathogens, among other applications. Although this method was first described in shrimp in the mid-2000s, at present, no practical approach has been developed for the use of dsRNA in shrimp farms, as the limiting factor for farm-scale usage in the aquaculture sector is the lack of cost-effective and simple dsRNA synthesis and administration procedures. Despite these limitations, different RNAi-based approaches have been successfully tested at the laboratory level, with a particular focus on shrimp. The use of RNAi technology is particularly attractive for the shrimp industry because crustaceans do not have an adaptive immune system, making traditional vaccination methods unfeasible. This review summarizes recent studies and the state-of-the-art on the mechanism of action, design, use, and administration methods of dsRNA, as applied to shrimp. In addition, potential constraints that may hinder the deployment of RNAi-based methods in the crustacean aquaculture sector are considered.
Collapse
Affiliation(s)
- Carlos Fajardo
- Department of Biology, Faculty of Marine and Environmental Sciences, Instituto Universitario de Investigación Marina (INMAR), Campus de Excelencia Internacional del Mar (CEI-MAR), University of Cadiz (UCA), 11510 Puerto Real, Spain;
- Interdisciplinary Centre of Marine and Environmental Research, The University of Porto (CIIMAR), 4450-208 Matosinhos, Portugal; (M.M.); (B.C.)
| | - Marcos De Donato
- Center for Aquaculture Technologies (CAT), San Diego, CA 92121, USA;
- Escuela de Medicina y Ciencias de la Salud, Tecnológico de Monterrey, Querétaro 76130, Mexico
| | - Marta Macedo
- Interdisciplinary Centre of Marine and Environmental Research, The University of Porto (CIIMAR), 4450-208 Matosinhos, Portugal; (M.M.); (B.C.)
- Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto (UP), 4050-313 Porto, Portugal
| | - Patai Charoonnart
- Center of Excellence for Shrimp Molecular Biology and Biotechnology (Centex Shrimp), Faculty of Science, Mahidol University, Bangkok 10400, Thailand; (P.C.); (V.S.)
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Bangkok 12120, Thailand
| | - Vanvimon Saksmerprome
- Center of Excellence for Shrimp Molecular Biology and Biotechnology (Centex Shrimp), Faculty of Science, Mahidol University, Bangkok 10400, Thailand; (P.C.); (V.S.)
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Bangkok 12120, Thailand
| | - Luyao Yang
- Department of Structural and Molecular Biology, University College London (UCL), London WC1E 6BT, UK; (L.Y.); (S.P.)
| | - Saul Purton
- Department of Structural and Molecular Biology, University College London (UCL), London WC1E 6BT, UK; (L.Y.); (S.P.)
| | - Juan Miguel Mancera
- Department of Biology, Faculty of Marine and Environmental Sciences, Instituto Universitario de Investigación Marina (INMAR), Campus de Excelencia Internacional del Mar (CEI-MAR), University of Cadiz (UCA), 11510 Puerto Real, Spain;
| | - Benjamin Costas
- Interdisciplinary Centre of Marine and Environmental Research, The University of Porto (CIIMAR), 4450-208 Matosinhos, Portugal; (M.M.); (B.C.)
- Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto (UP), 4050-313 Porto, Portugal
| |
Collapse
|
43
|
Wang Y, Wang C, Lu Y. Spleen Targeting Nucleic Acid Delivery Vector Based on Metal-Organic Frameworks. ACS APPLIED MATERIALS & INTERFACES 2024; 16:56826-56836. [PMID: 39390629 DOI: 10.1021/acsami.4c13519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Nucleic acids have attracted increasing attention as drugs due to their fascinating advantages, such as long-term efficacy and ease of preparation compared to proteins. The nucleic acid therapy relies heavily on delivery vectors, which can prevent the degradation of nucleic acids while assisting them in cellular internalization. However, commonly used nonviral vector liposomes easily accumulate in the liver, which can limit their application in extrahepatic diseases. Herein, a potential spleen targeting vector for nucleic acids is developed based on the metal-organic frameworks. The plasmids are encapsulated inside the nanoscale zeolitic imidazolate framework (ZIF) via coprecipitation. The co-encapsulation of the cationic polymer poly(ether imide) (PEI) and the stabilizer polyvinylpyrrolidone (PVP) can significantly improve particle dispersion and stability. The prepared nanoparticles allow efficient transfection in vitro, mainly through clathrin-mediated and caveolae-mediated endocytosis. The biodistribution in mice shows that 46% of the nanoparticles accumulate in the spleen, which is much higher than that of the liposomes. The vector can successfully deliver plasmids to extrahepatic organs for protein synthesis and even induce an immune response. The elaborate ZIF-based nanoparticle may offer a new route for extrahepatic, especially spleen targeting delivery for the nucleic acids.
Collapse
Affiliation(s)
- Yiming Wang
- Department of Chemical Engineering, Tsinghua University, Beijing 100084, China
- Key Laboratory of Industrial Biocatalysis, Ministry of Education, Tsinghua University, Beijing 100084, China
| | - Chen Wang
- Department of Chemical Engineering, Tsinghua University, Beijing 100084, China
- Key Laboratory of Industrial Biocatalysis, Ministry of Education, Tsinghua University, Beijing 100084, China
| | - Yuan Lu
- Department of Chemical Engineering, Tsinghua University, Beijing 100084, China
- Key Laboratory of Industrial Biocatalysis, Ministry of Education, Tsinghua University, Beijing 100084, China
| |
Collapse
|
44
|
Tchounwou C, Jobanputra AJ, Lasher D, Fletcher BJ, Jacinto J, Bhaduri A, Best RL, Fisher WS, Ewert KK, Li Y, Feinstein SC, Safinya CR. Mixtures of Intrinsically Disordered Neuronal Protein Tau and Anionic Liposomes Reveal Distinct Anionic Liposome-Tau Complexes Coexisting with Tau Liquid-Liquid Phase-Separated Coacervates. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:21041-21051. [PMID: 39340452 DOI: 10.1021/acs.langmuir.4c02471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/30/2024]
Abstract
Tau, an intrinsically disordered neuronal protein and polyampholyte with an overall positive charge, is a microtubule (MT) associated protein that binds to anionic domains of MTs and suppresses their dynamic instability. Aberrant tau-MT interactions are implicated in Alzheimer's and other neurodegenerative diseases. Here, we studied the interactions between full-length human protein tau and other negatively charged binding substrates, as revealed by differential interference contrast (DIC) and fluorescence microscopy. As a binding substrate, we chose anionic liposomes (ALs) containing either 1,2-dioleoyl-sn-glycero-3-phosphatidylserine (DOPS, -1e) or 1,2-dioleoyl-sn-glycero-3-phosphatidylglycerol (DOPG, -1e) mixed with zwitterionic 1,2-dioleoyl-sn-glycero-3-phosphatidylcholine (DOPC) to mimic anionic plasma membranes of axons where tau resides. At low salt concentrations (0 to 10 mM KCl or NaCl) with minimal charge screening, reaction mixtures of tau and ALs resulted in the formation of distinct states of AL-tau complexes coexisting with liquid-liquid phase-separated tau self-coacervates arising from the polyampholytic nature of tau containing cationic and anionic domains. AL-tau complexes (i.e. tau-lipoplexes) exhibited distinct types of morphologies. This included large ∼20-30 μm tau-decorated giant vesicles with additional smaller liposomes with bound tau attached to the giant vesicles and tau-mediated finite-size assemblies of small liposomes. As the salt concentration was increased to near and above 150 mM for 1:1 electrolytes, AL-tau complexes remained stable, while tau self-coacervate droplets were found to dissolve, indicative of the breaking of (anionic/cationic) electrostatic bonds between tau chains due to increased charge screening. The findings are consistent with the hypothesis that distinct cationic domains of tau may interact with anionic lipid domains of the lumen-facing monolayer of the axon's plasma membrane, suggesting the possibility of transient yet robust interactions near relevant ionic strengths found in neurons.
Collapse
Affiliation(s)
- Christine Tchounwou
- Materials Department, University of California, Santa Barbara, California 93106, United States
- Molecular, Cellular, and Developmental Biology Department, University of California, Santa Barbara, California 93106, United States
| | - Anjali J Jobanputra
- Materials Department, University of California, Santa Barbara, California 93106, United States
- Molecular, Cellular, and Developmental Biology Department, University of California, Santa Barbara, California 93106, United States
- Biomolecular Science & Engineering Program, University of California, Santa Barbara, California 93106, United States
| | - Dylan Lasher
- Materials Department, University of California, Santa Barbara, California 93106, United States
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, California 93106, United States
| | - Bretton J Fletcher
- Materials Department, University of California, Santa Barbara, California 93106, United States
- Biomolecular Science & Engineering Program, University of California, Santa Barbara, California 93106, United States
| | - Jorge Jacinto
- Materials Department, University of California, Santa Barbara, California 93106, United States
- Molecular, Cellular, and Developmental Biology Department, University of California, Santa Barbara, California 93106, United States
| | - Arjun Bhaduri
- Materials Department, University of California, Santa Barbara, California 93106, United States
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, California 93106, United States
| | - Rebecca L Best
- Molecular, Cellular, and Developmental Biology Department, University of California, Santa Barbara, California 93106, United States
- Neuroscience Research Institute, University of California, Santa Barbara, California 93106, United States
| | - William S Fisher
- Materials Department, University of California, Santa Barbara, California 93106, United States
- Biomolecular Science & Engineering Program, University of California, Santa Barbara, California 93106, United States
| | - Kai K Ewert
- Materials Department, University of California, Santa Barbara, California 93106, United States
| | - Youli Li
- Materials Research Laboratory, University of California, Santa Barbara, California 93106, United States
| | - Stuart C Feinstein
- Molecular, Cellular, and Developmental Biology Department, University of California, Santa Barbara, California 93106, United States
- Neuroscience Research Institute, University of California, Santa Barbara, California 93106, United States
| | - Cyrus R Safinya
- Materials Department, University of California, Santa Barbara, California 93106, United States
- Molecular, Cellular, and Developmental Biology Department, University of California, Santa Barbara, California 93106, United States
- Biomolecular Science & Engineering Program, University of California, Santa Barbara, California 93106, United States
- Department of Physics, University of California, Santa Barbara, California 93106, United States
| |
Collapse
|
45
|
Inoh Y, Ito N, Yokawa S, Suzuki R, Furuno T. Surface charge, but not size, of liposomes is involved in the suppression of rat basophilic leukemia (RBL-2H3) cell degranulation mediated by Akt phosphorylation. Cell Biol Int 2024; 48:1463-1472. [PMID: 39418631 DOI: 10.1002/cbin.12205] [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: 04/23/2024] [Revised: 05/28/2024] [Accepted: 06/03/2024] [Indexed: 10/19/2024]
Abstract
Cationic liposomes composed of cholesteryl-3β-carboxyamidoethylene-N-hydroxyethylamine (OH-chol) and 1,2-dioleoyl-sn-glycero-3-phosphatidylethanolamine (DOPE) inhibit mast cell degranulation mediated via crosslinking of high-affinity IgE receptors (FcεRI). Although the inhibitory efficiency of mast cell degranulation is altered by modifying the ratio of OH-chol and DOPE in cationic liposomes, the manner in which physicochemical properties, such as surface charge and size, influence suppression is not clear. We observed that positive surface charge, but not the size, of liposomes plays a role in suppressing rat basophilic leukemia (RBL-2H3) cell activation. Pretreatment with middle-ratio OH-chol liposomes (zeta potential, 62.2 ± 0.5 mV; diameter, 325.4 ± 7.3 nm) exhibited a larger suppression of RBL-2H3 cell degranulation evoked by FcεRI crosslinking compared with that by low-ratio OH-chol liposomes (zeta potential, 48.6 ± 1.9 mV; diameter, 344.4 ± 25.0 nm), although both liposomes were similarly attached to RBL-2H3 cells. Preparation of middle-ratio OH-chol liposomes, classified roughly by size using an extrusion method, revealed that the liposomal size did not affect the inhibitory efficiency of RBL-2H3 cell activation. Mechanistically, we found that middle-ratio OH-chol liposomes increased the inhibition of antigen-induced Akt phosphorylation compared to low-ratio OH-chol liposomes. We measured the phosphorylation of linker for activation of T cells (LAT) and paxillin, which are important proteins in FcεRI- and focal adhesions (FAs)-mediated signaling, respectively. Middle ratio OH-chol liposomes significantly suppressed antigen-induced paxillin phosphorylation, but did not affect LAT phosphorylation, suggesting that middle-ratio OH-chol liposomes attached to RBL-2H3 cells suppress the degranulation by impairing FA-mediated Akt phosphorylation evoked by FcεRI crosslinking.
Collapse
Affiliation(s)
- Yoshikazu Inoh
- School of Pharmacy, Aichi Gakuin University, Nagoya, Japan
| | - Nanami Ito
- School of Pharmacy, Aichi Gakuin University, Nagoya, Japan
| | - Satoru Yokawa
- School of Pharmacy, Aichi Gakuin University, Nagoya, Japan
| | - Ruriko Suzuki
- School of Pharmacy, Aichi Gakuin University, Nagoya, Japan
| | | |
Collapse
|
46
|
Lokras AG, Bobak TR, Baghel SS, Sebastiani F, Foged C. Advances in the design and delivery of RNA vaccines for infectious diseases. Adv Drug Deliv Rev 2024; 213:115419. [PMID: 39111358 DOI: 10.1016/j.addr.2024.115419] [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: 03/01/2024] [Revised: 07/19/2024] [Accepted: 07/30/2024] [Indexed: 08/23/2024]
Abstract
RNA medicines represent a paradigm shift in treatment and prevention of critical diseases of global significance, e.g., infectious diseases. The highly successful messenger RNA (mRNA) vaccines against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) were developed at record speed during the coronavirus disease 2019 pandemic. A consequence of this is exceptionally shortened vaccine development times, which in combination with adaptability makes the RNA vaccine technology highly attractive against infectious diseases and for pandemic preparedness. Here, we review state of the art in the design and delivery of RNA vaccines for infectious diseases based on different RNA modalities, including linear mRNA, self-amplifying RNA, trans-amplifying RNA, and circular RNA. We provide an overview of the clinical pipeline of RNA vaccines for infectious diseases, and present analytical procedures, which are paramount for characterizing quality attributes and guaranteeing their quality, and we discuss future perspectives for using RNA vaccines to combat pathogens beyond SARS-CoV-2.
Collapse
Affiliation(s)
- Abhijeet Girish Lokras
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Denmark
| | - Thomas Rønnemoes Bobak
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Denmark
| | - Saahil Sandeep Baghel
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Denmark
| | - Federica Sebastiani
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Denmark; Division of Physical Chemistry, Department of Chemistry, Lund University, 22100, Lund, Sweden
| | - Camilla Foged
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Denmark.
| |
Collapse
|
47
|
Thongbamrer C, Kunkeaw N, Nguitragool W, Roobsoong W, Sattabongkot J, Opanasopit P, Yingyongnarongkul BE. Optimizing Transfection Efficiency of Spermine Polar Head Cholesterol-Based Cationic Lipids with Amino Acid Linker. Chembiochem 2024:e202400490. [PMID: 39353853 DOI: 10.1002/cbic.202400490] [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: 06/04/2024] [Revised: 09/26/2024] [Accepted: 10/01/2024] [Indexed: 10/04/2024]
Abstract
In this work, a series of spermine polar head cholesterol-based cationic lipids with various amino acid spacers were synthesized and evaluated as non-viral gene delivery systems. The physicochemical properties of the resulting lipoplexes, formed from these lipids and DOPE, were assessed, including zeta-potential, DNA binding and DNA protection from serum. Transfection efficiency and cytotoxicity were examined under serum-free and 10-40 % serum-containing conditions. The results showed that the physicochemical properties of cationic lipids, both with and without amino acid spacers, were not significantly different. Cationic liposomes composed of lipid Sper-Ahx-Chol, which has a 6-aminohexanoic acid spacer, and DOPE exhibited greater transfection efficiency in HeLa cells compared to Lipofectamine3000, both in the absence and presence of 10-40 % serum. Additionally, lipid Sper-His-Chol with a histidine spacer and Sper-Ahx-Chol showed higher efficiency than Lipofectamine3000 against HEK293T under 40 % serum conditions. These results suggest that the incorporation of amino acids into the cationic lipids can significantly enhance their DNA delivery efficiency. Specifically, certain amino acid modifications improved transfection efficiency while maintaining low cytotoxicity. Our findings highlight the potential of amino acid-tailored cationic lipids as promising vectors for enhanced DNA delivery.
Collapse
Affiliation(s)
- Chopaka Thongbamrer
- Department of Chemistry and Center of Excellence for Innovation in Chemistry (PERCH-CIC), Faculty of Science, Ramkhamhaeng University, Bangkapi, Bangkok, 10240, Thailand
| | - Nawapol Kunkeaw
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Rachathewi, Bangkok, 10400, Thailand
| | - Wang Nguitragool
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, 10400, Thailand
| | - Wanlapa Roobsoong
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Rachathewi, Bangkok, 10400, Thailand
| | - Jetsumon Sattabongkot
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Rachathewi, Bangkok, 10400, Thailand
| | - Praneet Opanasopit
- Pharmaceutical Development of Green Innovations Group (PDGIG), Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, 73000, Thailand
| | - Boon-Ek Yingyongnarongkul
- Department of Chemistry and Center of Excellence for Innovation in Chemistry (PERCH-CIC), Faculty of Science, Ramkhamhaeng University, Bangkapi, Bangkok, 10240, Thailand
| |
Collapse
|
48
|
Dastgerdi NK, Dastgerdi NK, Bayraktutan H, Costabile G, Atyabi F, Dinarvand R, Longobardi G, Alexander C, Conte C. Enhancing siRNA cancer therapy: Multifaceted strategies with lipid and polymer-based carrier systems. Int J Pharm 2024; 663:124545. [PMID: 39098747 DOI: 10.1016/j.ijpharm.2024.124545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 07/29/2024] [Accepted: 07/30/2024] [Indexed: 08/06/2024]
Abstract
Cancers are increasing in prevalence and many challenges remain for their treatment, such as chemoresistance and toxicity. In this context, siRNA-based therapeutics have many potential advantages for cancer therapies as a result of their ability to reduce or prevent expression of specific cancer-related genes. However, the direct delivery of naked siRNA is hindered by issues like enzymatic degradation, insufficient cellular uptake, and poor pharmacokinetics. Hence, the discovery of a safe and efficient delivery vehicle is essential. This review explores various lipid and polymer-based delivery systems for siRNA in cancer treatment. Both polymers and lipids have garnered considerable attention as carriers for siRNA delivery. While all of these systems protect siRNA and enhance transfection efficacy, each exhibits its unique strengths. Lipid-based delivery systems, for instance, demonstrate high entrapment efficacy and utilize cost-effective materials. Conversely, polymeric-based delivery systems offer advantages through chemical modifications. Nonetheless, certain drawbacks still limit their usage. To address these limitations, combining different materials in formulations (lipid, polymer, or targeting agent) could enhance pharmaceutical properties, boost transfection efficacy, and reduce side effects. Furthermore, co-delivery of siRNA with other therapeutic agents presents a promising strategy to overcome cancer resistance. Lipid-based delivery systems have been demonstrated to encapsulate many therapeutic agents and with high efficiency, but most are limited in terms of the functionalities they display. In contrast, polymeric-based delivery systems can be chemically modified by a wide variety of routes to include multiple components, such as release or targeting elements, from the same materials backbone. Accordingly, by incorporating multiple materials such as lipids, polymers, and/or targeting agents in RNA formulations it is possible to improve the pharmaceutical properties and therapeutic efficacy while reducing side effects. This review focuses on strategies to improve siRNA cancer treatments and discusses future prospects in this important field.
Collapse
Affiliation(s)
- Nazgol Karimi Dastgerdi
- Division of Molecular Therapeutics and Formulation, School of Pharmacy, University of Nottingham, NG7 2RD, UK; Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Nazanin Karimi Dastgerdi
- Pharmaceutical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hulya Bayraktutan
- Division of Molecular Therapeutics and Formulation, School of Pharmacy, University of Nottingham, NG7 2RD, UK
| | | | - Fatemeh Atyabi
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 1417614315, Iran
| | - Rassoul Dinarvand
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 1417614315, Iran.
| | | | - Cameron Alexander
- Division of Molecular Therapeutics and Formulation, School of Pharmacy, University of Nottingham, NG7 2RD, UK
| | - Claudia Conte
- Department of Pharmacy, University of Napoli Federico II, Napoli, Italy.
| |
Collapse
|
49
|
Hameed H, Sarwar HS, Younas K, Zaman M, Jamshaid M, Irfan A, Khalid M, Sohail MF. Exploring the potential of nanomedicine for gene therapy across the physicochemical and cellular barriers. Funct Integr Genomics 2024; 24:177. [PMID: 39340586 DOI: 10.1007/s10142-024-01459-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 09/13/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024]
Abstract
After COVID-19, a turning point in the way of pharmaceutical technology is gene therapy with beneficial potential to start a new medical era. However, commercialization of such pharmaceuticals would never be possible without the help of nanotechnology. Nanomedicine can fulfill the growing needs linked to safety, efficiency, and site-specific targeted delivery of Gene therapy-based pharmaceuticals. This review's goal is to investigate how nanomedicine may be used to transfer nucleic acids by getting beyond cellular and physicochemical barriers. Firstly, we provide a full description of types of gene therapy, their mechanism, translation, transcription, expression, type, and details of diseases with possible mechanisms that can only be treated with genes-based pharmaceuticals. Additionally, we also reviewed different types of physicochemical barriers, physiological and cellular barriers in nucleic acids (DNA/RNA) based drug delivery. Finally, we highlight the need and importance of cationic lipid-based nanomedicine/nanocarriers in gene-linked drug delivery and how nanotechnology can help to overcome the above-discussed barrier in gene therapy and their biomedical applications.
Collapse
Affiliation(s)
- Huma Hameed
- Faculty of Pharmaceutical Sciences, University of Central Punjab (UCP), Lahore, 54000, Pakistan.
| | - Hafiz Shoaib Sarwar
- Faculty of Pharmaceutical Sciences, University of Central Punjab (UCP), Lahore, 54000, Pakistan
| | - Komel Younas
- Faculty of Pharmacy, University Paris Saclay, 17 Avenue des sciences, 91190, Orsay, France
| | - Muhammad Zaman
- Faculty of Pharmaceutical Sciences, University of Central Punjab (UCP), Lahore, 54000, Pakistan
| | - Muhammad Jamshaid
- Faculty of Pharmaceutical Sciences, University of Central Punjab (UCP), Lahore, 54000, Pakistan
| | - Ali Irfan
- Department of Chemistry, Government College University Faisalabad, Faisalabad, 38000, Pakistan
| | - Maha Khalid
- Faculty of Pharmaceutical Sciences, University of Central Punjab (UCP), Lahore, 54000, Pakistan
| | - Muhammad Farhan Sohail
- Department of Chemistry, SBASSE, Lahore University of Management Sciences (LUMS), Lahore, 54000, Pakistan
- Alliant College of Pharmacy and Allied Health Sciences, Lahore, 54000, Pakistan
| |
Collapse
|
50
|
Aggarwal T, Bellomo A, Stevenson ER, Herbert J, Laskin DL, Gow AJ, Izgu EC. Protocol for detecting nitrative stress in biological lipid membranes in murine cells and tissues. STAR Protoc 2024; 5:103268. [PMID: 39215997 PMCID: PMC11403049 DOI: 10.1016/j.xpro.2024.103268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 07/14/2024] [Accepted: 08/01/2024] [Indexed: 09/04/2024] Open
Abstract
Detection of nitrative stress is crucial to understanding redox signaling and pathophysiology. Dysregulated nitrative stress, which generates high levels of peroxynitrite, can damage lipid membranes and cause activation of proinflammatory pathways associated with pulmonary complications. Here, we present a protocol for implementing a peroxynitrite-sensing phospholipid to investigate nitrative stress in murine cells and lung tissue. We detail procedures for sensing ONOO- in stimulated cells, both ex vivo and in vivo, using murine models of acute lung injury (ALI). For complete details on the use and execution of this protocol, please refer to Gutierrez and Aggarwal et al.1.
Collapse
Affiliation(s)
- Tushar Aggarwal
- Department of Chemistry and Chemical Biology, Rutgers University-New Brunswick, Piscataway, NJ 08854, USA
| | - Alyssa Bellomo
- Ernest Mario School of Pharmacy, Department of Pharmacology & Toxicology, Rutgers University, New Brunswick, NJ 08901, USA
| | - Emily R Stevenson
- Ernest Mario School of Pharmacy, Department of Pharmacology & Toxicology, Rutgers University, New Brunswick, NJ 08901, USA
| | - Julia Herbert
- Ernest Mario School of Pharmacy, Department of Pharmacology & Toxicology, Rutgers University, New Brunswick, NJ 08901, USA
| | - Debra L Laskin
- Ernest Mario School of Pharmacy, Department of Pharmacology & Toxicology, Rutgers University, New Brunswick, NJ 08901, USA
| | - Andrew J Gow
- Ernest Mario School of Pharmacy, Department of Pharmacology & Toxicology, Rutgers University, New Brunswick, NJ 08901, USA
| | - Enver Cagri Izgu
- Department of Chemistry and Chemical Biology, Rutgers University-New Brunswick, Piscataway, NJ 08854, USA; Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ 08901, USA; Rutgers Center for Lipid Research, New Jersey Institute for Food, Nutrition, and Health, Rutgers University-New Brunswick, New Brunswick, NJ 08901, USA.
| |
Collapse
|