Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (576)

Search Parameters:
Keywords = exosomal delivery

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
20 pages, 1647 KB  
Review
Advances in Plant-Derived Extracellular Vesicles: Implications for Apple-Derived EVs
by Hao Fu, Shunyuan Yong, Yanping Song, Jiangbo Dang, Danlong Jing, Di Wu and Qigao Guo
Plants 2025, 14(22), 3425; https://doi.org/10.3390/plants14223425 - 9 Nov 2025
Viewed by 436
Abstract
Plant-derived extracellular vesicles (PDEVs) are nanoscale membrane vesicles released by edible plants that deliver proteins, lipids, nucleic acids, and small metabolites to recipient cells, thereby modulating inflammation, barrier function, metabolism, and intercellular signaling. In recent years, PDEV research has advanced from concept and [...] Read more.
Plant-derived extracellular vesicles (PDEVs) are nanoscale membrane vesicles released by edible plants that deliver proteins, lipids, nucleic acids, and small metabolites to recipient cells, thereby modulating inflammation, barrier function, metabolism, and intercellular signaling. In recent years, PDEV research has advanced from concept and in vitro observations to engineering-ready systems with validation in animal models, encompassing oral, transdermal, and intranasal delivery paradigms. Among edible plants, the apple has broad consumption and a favorable safety profile; however, studies on apple-derived extracellular vesicles (ADEVs) lag behind those on other plant EVs. Accordingly, this review systematically summarizes ADEV progress across extraction methods, characterization, molecular cargo, and roles in disease settings. We highlight evidence gaps in animal efficacy and translation, and propose priorities including process standardization, harmonized critical quality attributes, in vivo biodistribution, and long-term safety. Our aim is to provide a reference for ADEV research and to accelerate the development of safe, low-cost, scalable bionanocarriers for disease therapy. Full article
Show Figures

Figure 1

20 pages, 3312 KB  
Article
Application of VHH-Immobilized Cryogel-Based Immunoaffinity Chromatography for Isolation of Extracellular Vesicles
by Jovana Terzić, Lidija Filipović, Ninoslav Mitić, Sanja Stevanović, Jugoslav Krstić, Ario de Marco, Julie Courraud and Milica Popović
Molecules 2025, 30(22), 4337; https://doi.org/10.3390/molecules30224337 - 8 Nov 2025
Viewed by 267
Abstract
Extracellular vesicles (EVs) are nanosized structures involved in intercellular communication that have high potential as disease biomarkers and for the delivery of therapeutic cargos. However, translation to the clinic is hampered by time-consuming, low-yield, and poorly reproducible EV isolation methods. We describe a [...] Read more.
Extracellular vesicles (EVs) are nanosized structures involved in intercellular communication that have high potential as disease biomarkers and for the delivery of therapeutic cargos. However, translation to the clinic is hampered by time-consuming, low-yield, and poorly reproducible EV isolation methods. We describe a cryogel-based immunoaffinity chromatography system that exploits single-domain VHH antibodies as capture elements for the selective isolation of EVs from human plasma. Supermacroporous cryogels functionalized with five unique anti-EV VHHs (total immobilization capacity ~500 µg/g) were prepared, yielding a highly permeable and hydrophilic support. They were captured and eluted under mild conditions, and their morphology and identity were confirmed by SEM, AFM, NTA, and flow cytometry. Proteomic profiling of the isolated samples identified 234 proteins, of which 63% were ExoCarta-listed exosomal proteins; contaminants such as albumin and apolipoproteins were also identified. The purification method provided samples with ~2 × 109 EVs/mL, with EV median size of 135 nm and consistent protein-to-lipid ratio across three independent isolations (CV < 10%). This study demonstrates that VHH-functionalized cryogels (VHH-SMC) are a rapid and reproducible EV purification method that represents a promising alternative to conventional ultracentrifugation- or precipitation-based protocols. While optimization of nanobody density and reduction in plasma protein carryover are still necessary, the platform holds potential for scalable EV enrichment, a condition that can significantly speed up biomarker research and clinical diagnostics. Full article
(This article belongs to the Section Chemical Biology)
Show Figures

Figure 1

34 pages, 1371 KB  
Review
Advances in Cell-Mediated Drug Delivery for Dermatologic Diseases: Mechanisms and Current Applications
by Lara Shqair, Iyla Draw, Tala Maya, Christopher G. Bunick, Hossein Akbarialiabad, Todd Schlesinger, Giovanni Damiani, Mahmoud Ghannoum and Ayman Grada
Pharmaceutics 2025, 17(11), 1438; https://doi.org/10.3390/pharmaceutics17111438 - 7 Nov 2025
Viewed by 569
Abstract
Cell-mediated drug delivery systems represent a promising frontier in dermatologic therapy by offering enhanced targeting precision, prolonged drug release, and reduced systemic toxicity. These systems leverage the intrinsic properties of immune cells, stem cells, and skin-resident cells to migrate toward inflamed or diseased [...] Read more.
Cell-mediated drug delivery systems represent a promising frontier in dermatologic therapy by offering enhanced targeting precision, prolonged drug release, and reduced systemic toxicity. These systems leverage the intrinsic properties of immune cells, stem cells, and skin-resident cells to migrate toward inflamed or diseased skin and deliver therapeutic agents in a controlled and biocompatible manner. This review explores the mechanistic foundations of cell-mediated delivery, including chemotaxis, phagocytosis, and immune modulation, and examines current applications in inflammatory skin diseases such as atopic dermatitis and psoriasis, cutaneous malignancies such as melanoma and cutaneous T-cell lymphoma, and chronic wound healing. Engineering approaches such as cell surface modification, exosome loading, and integration with gene editing technologies are also discussed. Finally, we highlight translational challenges related to immunogenicity, manufacturing scalability, and regulatory considerations, and propose future directions for clinical adoption in dermatology. This review provides a comprehensive overview of the current landscape and outlines the potential for cell-based delivery systems to transform the treatment of chronic and refractory skin diseases. Full article
(This article belongs to the Special Issue Cell-Mediated Delivery Systems)
Show Figures

Figure 1

36 pages, 1826 KB  
Review
Platelet-Rich Plasma (PRP): Molecular Mechanisms, Actions and Clinical Applications in Human Body
by Wen-Shan Wu, Li-Ru Chen and Kuo-Hu Chen
Int. J. Mol. Sci. 2025, 26(21), 10804; https://doi.org/10.3390/ijms262110804 - 6 Nov 2025
Viewed by 751
Abstract
Platelet-rich plasma (PRP) is an autologous blood-derived concentrate increasingly utilized in regenerative medicine for its ability to accelerate healing and tissue repair. PRP is broadly classified by leukocyte content, fibrin architecture, and platelet concentration, with classification systems developed to standardize characterization. Preparation methods, [...] Read more.
Platelet-rich plasma (PRP) is an autologous blood-derived concentrate increasingly utilized in regenerative medicine for its ability to accelerate healing and tissue repair. PRP is broadly classified by leukocyte content, fibrin architecture, and platelet concentration, with classification systems developed to standardize characterization. Preparation methods, including single- or double-spin centrifugation and buffy coat techniques, influence the final composition of PRP, determining the relative proportions of platelets, leukocytes, plasma proteins, and extracellular vesicles. These components act synergistically, with platelets releasing growth factors (e.g., VEGF, PDGF, TGF-β) that stimulate angiogenesis and matrix synthesis, leukocytes providing immunomodulation, plasma proteins facilitating scaffolding, and exosomes regulating intercellular signaling. Mechanistically, PRP enhances tissue repair through four key pathways: platelet adhesion molecules promote hemostasis and cell recruitment; immunomodulation reduces pro-inflammatory cytokines and favors M2 macrophage polarization; angiogenesis supports vascular remodeling and nutrient delivery; and serotonin-mediated pathways contribute to analgesia. These processes establish a regenerative microenvironment that supports both structural repair and functional recovery. Clinically, PRP has been applied across multiple specialties. In orthopedics, it promotes tendon, cartilage, and bone healing in conditions such as tendinopathy and osteoarthritis. In dermatology, PRP enhances skin rejuvenation, scar remodeling, and hair restoration. Gynecology has adopted PRP for ovarian rejuvenation, endometrial repair, and vulvovaginal atrophy. In dentistry and oral surgery, PRP accelerates wound closure and osseointegration, while chronic wound care benefits from its angiogenic and anti-inflammatory effects. PRP has also favored gingival recession coverage, regeneration of intrabony periodontal defects, and sinus grafting. Although preparation heterogeneity remains a challenge, PRP offers a versatile, biologically active therapy with expanding clinical utility. Full article
(This article belongs to the Section Biochemistry)
Show Figures

Figure 1

19 pages, 392 KB  
Review
MicroRNAs as Emerging Therapeutic Targets Modulating the Tumor Microenvironment in Head and Neck Squamous Cell Carcinoma
by Roxana Daniela Brata, Lavinia Marcut, Alina Cristina Barb, Alexia Manole, Alexandru Ciolofan, Cristina Stefania Dumitru, Flavia Zara and Raul Patrascu
Int. J. Mol. Sci. 2025, 26(21), 10794; https://doi.org/10.3390/ijms262110794 - 6 Nov 2025
Viewed by 400
Abstract
Head and neck squamous cell carcinoma (HNSCC) remains one of the most aggressive solid tumors, characterized by marked molecular heterogeneity and a complex tumor microenvironment (TME). Recent evidence highlights the pivotal role of microRNAs (miRNAs) in regulating tumor progression, immune evasion, angiogenesis, and [...] Read more.
Head and neck squamous cell carcinoma (HNSCC) remains one of the most aggressive solid tumors, characterized by marked molecular heterogeneity and a complex tumor microenvironment (TME). Recent evidence highlights the pivotal role of microRNAs (miRNAs) in regulating tumor progression, immune evasion, angiogenesis, and stromal remodeling. This review synthesizes current insights into miRNA-mediated molecular pathways that modulate the TME in HNSCC and discusses emerging therapeutic strategies, including nanocarrier- and exosome-based miRNA delivery systems, targeting these molecules. Key miRNAs, including miR-21, miR-146a, and miR-221, orchestrate bidirectional signaling between cancer cells, fibroblasts, and immune infiltrates, thereby shaping tumor aggressiveness and therapy resistance. Advances in nanotechnology have facilitated the development of miRNA-based therapeutics—such as mimics, antagomiRs, and exosome-mediated systems—capable of restoring physiological expression patterns and reprogramming the TME toward an anti-tumor state. However, clinical translation remains hindered by challenges in targeted delivery, molecular stability, and tumor heterogeneity. By integrating molecular and translational perspectives, this review underscores how miRNA-targeting strategies may evolve into a new generation of precision therapies, bridging the gap between molecular oncology and personalized treatment of head and neck cancer. Full article
Show Figures

Figure 1

23 pages, 7504 KB  
Review
Mesenchymal Stem Cell-Mediated Targeted Drug Delivery Systems for Hepatocellular Carcinoma: Current Advances and Future Directions
by Yang Gao, Jian-Ping Wang, De-Fei Hong, Chang Yang and Hua Naranmandura
Bioengineering 2025, 12(11), 1206; https://doi.org/10.3390/bioengineering12111206 - 4 Nov 2025
Viewed by 759
Abstract
Hepatocellular carcinoma (HCC) ranks as the second most lethal malignancy worldwide, presenting formidable therapeutic challenges including tumor heterogeneity, complex microenvironment, and inefficient drug delivery. Conventional therapies such as surgery, chemotherapy, and immunotherapy are limited by systemic toxicity, drug resistance, and poor targeting specificity. [...] Read more.
Hepatocellular carcinoma (HCC) ranks as the second most lethal malignancy worldwide, presenting formidable therapeutic challenges including tumor heterogeneity, complex microenvironment, and inefficient drug delivery. Conventional therapies such as surgery, chemotherapy, and immunotherapy are limited by systemic toxicity, drug resistance, and poor targeting specificity. Mesenchymal stem cells (MSCs) have emerged as promising drug delivery vehicles, leveraging their innate tumor-homing capacity, immunomodulatory properties, and exosome-mediated cargo transport. Preclinical studies demonstrate that MSC-based systems triple drug accumulation in tumors and synergize with immunotherapy, extending survival in HCC models. This review systematically examines recent advances in MSC-based delivery systems for HCC, focusing on engineering strategies to enhance targeting precision and controlled drug release, including genetic modification, exosome engineering, and stimuli-response systems. Despite progress, challenges such as MSC heterogeneity and scalable production persist. Emerging solutions like single-cell RNA sequencing for subpopulation selection and 3D bioprinting for standardized culture are highlighted. This work provides a roadmap for developing MSC-based precision therapies, bridging translational gaps in HCC treatment. Full article
(This article belongs to the Special Issue Advances in Drug Delivery in Cancer Treatment)
Show Figures

Figure 1

22 pages, 2074 KB  
Review
Non-Coding RNAs as Emerging Regulators in Kidney Pathophysiology: From Molecular Mechanisms to Therapeutic Potential
by Petar Todorović, Nikola Pavlović, Mirko Maglica, Patricija Bajt, Nela Kelam, Fila Raguž and Katarina Vukojević
Genes 2025, 16(11), 1328; https://doi.org/10.3390/genes16111328 - 3 Nov 2025
Viewed by 434
Abstract
The kidney’s intricate physiology relies on finely tuned gene regulatory networks that coordinate cellular responses to metabolic, inflammatory, and fibrotic stress. Beyond protein-coding transcripts, non-coding RNAs (ncRNAs), including microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs), have emerged as pivotal regulators [...] Read more.
The kidney’s intricate physiology relies on finely tuned gene regulatory networks that coordinate cellular responses to metabolic, inflammatory, and fibrotic stress. Beyond protein-coding transcripts, non-coding RNAs (ncRNAs), including microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs), have emerged as pivotal regulators of renal biology. By modulating transcriptional, post-transcriptional, and epigenetic pathways, ncRNAs govern podocyte integrity, tubular adaptation, intercellular signaling, and immune activation. Dysregulation of these networks is now recognized as a hallmark of major kidney diseases, ranging from diabetic nephropathy and acute kidney injury to chronic kidney disease, glomerulopathies, and polycystic kidney disease. Mechanistic studies have revealed how pathogenic ncRNAs drive apoptosis, inflammation, fibrosis, and cystic remodeling, while protective ncRNAs mitigate these processes, highlighting their dual roles as both disease mediators and therapeutic targets. The exceptional stability of ncRNAs in urine, plasma, and exosomes further positions them as minimally invasive biomarkers with diagnostic and prognostic value. Translational advances include anti-miR and mimic-based therapies (e.g., lademirsen targeting miR-21, miR-29 mimics, anti-miR-17 oligonucleotides), alongside lncRNA silencing strategies, although challenges in delivery, safety, and redundancy remain significant. This review integrates molecular mechanisms with translational perspectives, providing a comprehensive synthesis of how ncRNAs shape renal pathophysiology. By bridging mechanistic insights with emerging diagnostic and therapeutic applications, we highlight the potential of ncRNAs to transform nephrology, paving the way for biomarker-driven precision medicine and novel interventions aimed at intercepting kidney injury at its regulatory roots. In clinical terms, ncRNA-based biomarkers and therapeutics promise earlier detection, more precise risk stratification, and individualized treatment selection within precision nephrology. Full article
(This article belongs to the Special Issue From Genetic to Molecular Basis of Kidney Diseases)
Show Figures

Figure 1

31 pages, 1993 KB  
Review
Sepsis-Induced Cardiomyopathy and Cardiac Arrhythmias: Pathophysiology and Implications for Novel Therapeutic Approaches
by Konstantinos Pamporis, Paschalis Karakasis, Antonia Pantelidaki, Panagiotis Antonios Goutis, Konstantinos Grigoriou, Panagiotis Theofilis, Athanasia Katsaouni, Michail Botis, Aikaterini-Eleftheria Karanikola, Nikias Milaras, Konstantinos Vlachos, Dimitrios Tsiachris, Constantinos Pantos and Iordanis Mourouzis
Biomedicines 2025, 13(11), 2643; https://doi.org/10.3390/biomedicines13112643 - 28 Oct 2025
Viewed by 1206
Abstract
In the context of multi-organ involvement in sepsis, cardiac toxicity is manifested as sepsis-induced cardiomyopathy (SICM). To date, no unified SICM definition exists, though a left ventricular ejection fraction ≤ 50% and/or an absolute drop ≥ 10% from baseline are the most widely [...] Read more.
In the context of multi-organ involvement in sepsis, cardiac toxicity is manifested as sepsis-induced cardiomyopathy (SICM). To date, no unified SICM definition exists, though a left ventricular ejection fraction ≤ 50% and/or an absolute drop ≥ 10% from baseline are the most widely accepted components. Several molecular pathways have been associated with SICM, including (i) pro-inflammatory mediator-induced cardiac depression; (ii) sarcolemmal membrane dysfunction; (iii) autonomic nervous system (ANS) imbalance; (iv) blunted cardiovascular response to catecholamines; (v) dysfunctional intracellular calcium handling; (vi) mitochondrial dysfunction; (vii) metabolic reprogramming; and (viii) disturbed endothelial and microcirculatory function. Atrial and ventricular arrhythmias—particularly atrial fibrillation—commonly complicate disease management and are associated with adverse outcomes. Key mechanisms outlining sepsis-induced arrhythmogenesis are (i) inflammation; (ii) electrolyte imbalances; (iii) myocardial ischemia; (iv) QT prolongation/dispersion; (v) adrenergic overactivation; (vi) calcium mishandling; and (vii) fever-induced arrhythmogenesis in Brugada. Established therapeutic approaches include prompt treatment with antibiotics, hemodynamic optimization, and/or selective use of beta-blockers. Furthermore, several molecules are currently being investigated targeting numerous pathways activated in sepsis. Vitamin C, ginsenoside Rc, Schistosoma Japonicum cystatin, and gasmerdin-D inhibitor Y2 exert anti-inflammatory actions, while melatonin and α-ketoglutarate regulate mitochondrial homeostasis. Triiodothyronine targets microcirculatory optimization and regulates protective pathways against stress-related cell death. Engineered exosomes may facilitate targeted drug delivery, inflammatory response modulation, and activation of pathways related to cell survival, while sodium octanoate exhibits anti-inflammatory actions coupled with improved energy metabolism. Finally, gene-regulating therapies aiming at inflammatory response optimization have also been proposed and are currently under development. Future research should aim to standardize the SICM definition, translate emerging therapeutics into clinical practice, identify novel molecular targets, and implement personalized treatment strategies for SICM. Full article
Show Figures

Figure 1

15 pages, 1249 KB  
Review
Regenerative Applications and Performance of Periodontal Ligament Stem Cells: A Comprehensive Review of In Vivo Studies
by Romain Podembski, Isabel Barahona, Catarina Izidoro, Alexis Romero and Paulo Mascarenhas
Appl. Sci. 2025, 15(21), 11444; https://doi.org/10.3390/app152111444 - 26 Oct 2025
Viewed by 539
Abstract
Periodontal ligament stem cells (PDLSCs) represent a promising cell source for true periodontal regeneration due to their ability to form bone, cementum, and functional ligament. This review critically synthesised twelve in vivo studies (rats = 5, pigs = 2, dogs = 2, sheep [...] Read more.
Periodontal ligament stem cells (PDLSCs) represent a promising cell source for true periodontal regeneration due to their ability to form bone, cementum, and functional ligament. This review critically synthesised twelve in vivo studies (rats = 5, pigs = 2, dogs = 2, sheep = 2, one human trial) evaluating PDLSC transplantation for periodontal defects. A comprehensive search of PubMed, Web of Science, Embase, and the Cochrane Library (to May 2025) identified 358 records, of which 12 met predefined inclusion criteria. Data extraction encompassed cell source, scaffold, dose, follow-up, and quantitative regenerative outcomes. Nine studies reported cell doses (5 × 105–2 × 107 cells) and six PDLSC regeneration rates (33–100%). After normalisation for host mass, effective delivery ranged from 105 to 106 cells·kg−1, with optimal outcomes typically above 105 cells·kg−1. PDLSC transplantation consistently enhanced alveolar bone, cementum, and periodontal-ligament regeneration compared with scaffold-alone or untreated controls, with the highest outcomes obtained using biocompatible scaffolds such as Hydroxyapatite/Tricalcium Phosphate (HA/TCP), Gelfoam, or amniotic membrane. Both autologous and allogeneic PDLSC achieved equivalent performance and excellent safety, while xenogeneic models confirmed immune tolerance. Despite encouraging results, the evidence remains preliminary—most studies were short-term and small-scale, and only one randomised human trial has been published. Standardisation of cell preparation, scaffold selection, dosing (absolute and mass-normalised), and follow-up is urgently needed. Future research should include Good Manufacturing Practice (GMP)-compliant clinical trials and mechanistic studies on PDLSC differentiation, paracrine signalling, and exosome-mediated effects to consolidate their translational potential for predictable periodontal regeneration. Full article
Show Figures

Figure 1

44 pages, 1002 KB  
Review
The Heart’s Small Molecules: The Importance of MicroRNAs in Cardiovascular Health
by Mustafa Yildiz, Ugur Ozkan and Metin Budak
J. Clin. Med. 2025, 14(21), 7454; https://doi.org/10.3390/jcm14217454 - 22 Oct 2025
Viewed by 486
Abstract
This comprehensive review explores the critical roles of microRNAs (miRNAS) in cardiovascular diseases, emphasizing their regulatory functions in gene expression and their involvement in disease progression. miRNAS are small, evolutionarily conserved non-coding RNAs that regulate gene expression post-transcriptionally and play essential roles in [...] Read more.
This comprehensive review explores the critical roles of microRNAs (miRNAS) in cardiovascular diseases, emphasizing their regulatory functions in gene expression and their involvement in disease progression. miRNAS are small, evolutionarily conserved non-coding RNAs that regulate gene expression post-transcriptionally and play essential roles in various cardiac conditions, including fibrosis, cardiac remodeling, apoptosis, ischemia/reperfusion injury, hypertrophy, heart failure, arrhythmias, coronary artery disease (CAD), congenital heart diseases (CHDs), cardiomyopathies, and valvular heart disease (VHD). miRNAS are increasingly recognized as sensitive and specific biomarkers for early diagnosis, disease monitoring, and evaluation of therapeutic responses across the cardiovascular disease spectrum. Ischemia/reperfusion injury leads to significant cardiac damage through elevated oxidative stress, mitochondrial dysfunction, and apoptosis. CAD, a major contributor to global morbidity and mortality, is primarily driven by atherosclerosis and chronic inflammation. Cardiac hypertrophy is initially an adaptive response to stress but may progress to heart failure if sustained. Arrhythmias arise from electrical disturbances such as reentry, abnormal automaticity, and triggered activity. Heart failure is a complex and progressive syndrome marked by poor prognosis and increasing global prevalence. VHD involves intricate molecular alterations, including myocardial fibrosis and calcification, which contribute to disease progression and adverse outcomes. Cardiomyopathies—including hypertrophic, dilated, restrictive, and arrhythmogenic forms—are influenced by genetic mutations, systemic diseases, and disrupted molecular signaling. CHDs, the most common congenital malformations, stem from structural abnormalities in cardiac development and remain a major cause of infant morbidity and mortality. Novel therapeutic methods, such as antisense oligonucleotides, miR mimics, and exosome-based delivery mechanisms, demonstrate the translational promise of miRNAs in the realm of personalized cardiovascular medicine. However, issues such as small sample sizes, inconsistent results, interspecies differences, and delivery challenges restrict the clinical application of miRNA-based therapies. This review integrates mechanistic insights, critiques the quality of available evidence, and identifies translational shortcomings. It highlights the diagnostic, prognostic, and therapeutic potential of miRNAs in reshaping cardiovascular disease treatment. Full article
Show Figures

Figure 1

18 pages, 9549 KB  
Article
Fused Membrane-Targeted Nanoscale Gene Delivery System Based on an Asymmetric Membrane Structure for Ischemic Stroke
by Jing Shi, Xinyi Zhao, Yue Zhang, Zitong Zhao, Jing Wang, Jia Mi, Zhaowei Xu, Chunhua Yang, Jing Qin and Hong Zhang
Pharmaceutics 2025, 17(10), 1357; https://doi.org/10.3390/pharmaceutics17101357 - 21 Oct 2025
Viewed by 402
Abstract
Background: Bone marrow-derived mesenchymal stem cell exosomes (EXOs) are attractive in biotechnology and biomedical research, as they possess natural cell-targeting properties and can cross biological barriers by influencing the SDF-1/CXCR4 axis. Lipid calcium phosphate (LCP) consists of a calcium phosphate core and [...] Read more.
Background: Bone marrow-derived mesenchymal stem cell exosomes (EXOs) are attractive in biotechnology and biomedical research, as they possess natural cell-targeting properties and can cross biological barriers by influencing the SDF-1/CXCR4 axis. Lipid calcium phosphate (LCP) consists of a calcium phosphate core and an asymmetric phospholipid bilayer containing abundant Ca2+ ions. AMD3100 modification of targeted LCP (T-LCP) can achieve targeted delivery to ischemic lesions via specific binding to CXCR4 receptors on various neuronal cell surfaces. Methods: Herein, a fused membrane formulation that simultaneously possesses EXO characteristics and enables targeted modification with AMD3100 was produced. The characteristics of biologically derived EXOs, artificially designed T-LCP, and the fused membrane formulation, including targeted delivery and gene loading efficiency, were then compared. Results: The fusion of artificially designed T-LCP with EXOs of natural origin is feasible and combines the advantages of both to achieve more prominent targeted delivery effects. Conclusions: MiRNA210-based gene therapy was effective in this study and provides a strategy for therapeutic efficacy in delivery systems with different targeting efficiencies. Full article
(This article belongs to the Section Gene and Cell Therapy)
Show Figures

Graphical abstract

23 pages, 1851 KB  
Review
Biological Nanoparticles for Enhancing Chronic Wound Regeneration
by Daniil Zotikov, Natalia Ponomareva, Sergey Brezgin, Anastasiia Kostyusheva, Anastasiya Frolova, Vladimir Chulanov, Alexander Lukashev, Peter Timashev and Dmitry Kostyushev
Cells 2025, 14(20), 1637; https://doi.org/10.3390/cells14201637 - 21 Oct 2025
Viewed by 766
Abstract
Chronic wounds (CWs) represent a growing global health concern with profound clinical and socioeconomic implications. Studies indicate that approximately 15% of CWs remain unhealed one year after the initial treatment. At the same time, it is assumed that from 1% to 2% of [...] Read more.
Chronic wounds (CWs) represent a growing global health concern with profound clinical and socioeconomic implications. Studies indicate that approximately 15% of CWs remain unhealed one year after the initial treatment. At the same time, it is assumed that from 1% to 2% of the population of developed countries will suffer from chronic wounds during their lifetime. CWs severely impair patients’ quality of life. Current therapies (compression bandages, antibiotics, hyperbaric oxygen, and skin grafts) face limitations, including toxicity, contraindications, inefficacy in patients with comorbidities like diabetes, and high cost. Biological nanoparticles (BNPs), particularly extracellular vesicles (EVs), emerge as transformative solutions due to their innate biocompatibility, targeted biodistribution, and multifunctional regenerative properties. This review examines the mechanisms by which BNPs promote CW healing and drug delivery. Innovative BNP delivery platforms (chitosan hydrogels, alginate films) are evaluated, enabling sustained release and responsiveness to the wound microenvironment. Clinical advances, including exosome-laden hydrogels that accelerate healing in diabetic ulcers, underscore BNPs’ potential to overcome conventional therapy limitations. By addressing the challenges of both pathophysiological complexity and healthcare system burden, BNPs demonstrate the potential to improve patient outcomes in the management of chronic wounds. Full article
Show Figures

Figure 1

26 pages, 2438 KB  
Review
Exosomes in HPV-Associated Cancers: From Biomarkers to Engineered Therapeutics
by Muharrem Okan Cakir, Melis Selek, Betul Yilmaz, Mustafa Ozdogan and G. Hossein Ashrafi
Cancers 2025, 17(20), 3386; https://doi.org/10.3390/cancers17203386 - 21 Oct 2025
Viewed by 881
Abstract
Background/Objectives: Human papillomavirus (HPV) is the main causative agent of cervical cancer and contributes to a significant proportion of other anogenital and oropharyngeal malignancies. The need for better biomarkers and therapeutic approaches in HPV-associated cancers has drawn attention to exosomes, small extracellular vesicles [...] Read more.
Background/Objectives: Human papillomavirus (HPV) is the main causative agent of cervical cancer and contributes to a significant proportion of other anogenital and oropharyngeal malignancies. The need for better biomarkers and therapeutic approaches in HPV-associated cancers has drawn attention to exosomes, small extracellular vesicles known for their stability, biomolecule transport capabilities, and role in cell-to-cell communication. Methods: This review comprehensively evaluates recent literature on the diagnostic, prognostic, and therapeutic applications of small extracellular vesicles, particularly exosomes, in HPV-related cancers. It analyzes findings on exosomal nucleic acids, proteins, and long non-coding RNAs, as well as engineered exosome-based therapies. Results: Exosomal miRNAs (e.g., miR-204-5p, miR-99a-5p, miR-21), proteins (e.g., glycolytic enzymes, HSP90), and lncRNAs (e.g., HOTAIR, DLEU1) have emerged as promising biomarkers for disease detection and monitoring. Exosomal cargo actively participates in HPV-related tumor progression. For example, miRNAs such as miR-21 and miR-146a modulate immune cell polarization and inflammatory signaling, while lncRNAs like HOTAIR promote oncogenic transcriptional programs. Exosomal proteins including HSP90 and ANXA1 facilitate extracellular matrix remodeling and immune evasion, thereby influencing tumor growth and metastasis. In HPV-positive head and neck and cervical cancers, exosomal cargo reflects HPV status, tumor progression, and treatment response. Therapeutic studies demonstrate the utility of exosomes in vaccine delivery, immune modulation, and drug delivery systems, including the use of PROTACs. However, clinical translation faces barriers including isolation protocol standardization, biomarker validation, and scalable production. Conclusions: Exosomes hold great promise for integration into diagnostic and therapeutic workflows for HPV-related cancers. Future research should focus on resolving standardization issues, validating biomarkers in diverse cohorts, and optimizing engineered exosome platforms for targeted therapy. Full article
(This article belongs to the Collection The Development of Anti-cancer Agents)
Show Figures

Figure 1

42 pages, 1602 KB  
Review
Exosome-Based Drug Delivery: A Next-Generation Platform for Cancer, Infection, Neurological and Immunological Diseases, Gene Therapy and Regenerative Medicine
by Dolores R. Serrano, Francisco Juste, Brayan J. Anaya, Bianca I. Ramirez, Sergio A. Sánchez-Guirales, John M. Quispillo, Ester M. Hernandez, Jesus A. Simon, Jose M. Trallero, Celia Serrano, Satyavati Rawat and Aikaterini Lalatsa
Pharmaceutics 2025, 17(10), 1336; https://doi.org/10.3390/pharmaceutics17101336 - 15 Oct 2025
Viewed by 2348
Abstract
Exosomes, naturally derived extracellular vesicles, have emerged as powerful bio-nanocarriers in precision medicine. Their endogenous origin, biocompatibility, and ability to encapsulate and deliver diverse therapeutic payloads position them as transformative tools in drug delivery, gene therapy, and regenerative medicine. This review presents a [...] Read more.
Exosomes, naturally derived extracellular vesicles, have emerged as powerful bio-nanocarriers in precision medicine. Their endogenous origin, biocompatibility, and ability to encapsulate and deliver diverse therapeutic payloads position them as transformative tools in drug delivery, gene therapy, and regenerative medicine. This review presents a comprehensive analysis of exosome-based therapeutics across multiple biomedical domains, including cancer, neurological and infectious diseases, immune modulation, and tissue repair. Exosomes derived from stem cells, immune cells, or engineered lines can be loaded with small molecules, RNA, or CRISPR-Cas systems, offering highly specific and low-immunogenic alternatives to viral vectors or synthetic nanoparticles. We explore endogenous and exogenous loading strategies, surface functionalization techniques for targeted delivery, and innovations that allow exosomes to traverse physiological barriers such as the blood–brain barrier. Furthermore, exosomes demonstrate immunomodulatory and regenerative properties in autoimmune and degenerative conditions, with promising roles in skin rejuvenation and cosmeceuticals. Despite their potential, challenges remain in large-scale production, cargo loading efficiency, and regulatory translation. Recent clinical trials and industry efforts underscore the accelerating momentum in this field. Exosomes represent a promising platform in precision medicine, though further standardization and validation are required before widespread clinical use. This review offers critical insights into current technologies, therapeutic mechanisms, and future directions to unlock the full translational potential of exosomes in clinical practice. Full article
(This article belongs to the Special Issue Vesicle-Based Drug Delivery Systems)
Show Figures

Graphical abstract

18 pages, 1164 KB  
Review
Advances in β-Thalassemia Gene Therapy: CRISPR/Cas Systems and Delivery Innovations
by Hongmei Liu and Peng Zhang
Cells 2025, 14(20), 1595; https://doi.org/10.3390/cells14201595 - 14 Oct 2025
Viewed by 1550
Abstract
β-thalassemia is an inherited blood disorder caused by mutations in the β-globin (HBB) gene, leading to reduced or absent β-globin production, resulting in chronic anemia. While current therapies, including blood transfusions and hematopoietic stem cell transplantation, offer symptomatic relief, they are limited by [...] Read more.
β-thalassemia is an inherited blood disorder caused by mutations in the β-globin (HBB) gene, leading to reduced or absent β-globin production, resulting in chronic anemia. While current therapies, including blood transfusions and hematopoietic stem cell transplantation, offer symptomatic relief, they are limited by complications and their limited accessibility. CRISPR-based gene editing technologies provide new therapeutic avenues by enabling the precise correction of HBB mutations or the reactivation of fetal hemoglobin (HbF) through the targeting of regulatory elements such as BCL11A. These approaches have shown promising preclinical and clinical outcomes. However, efficient and safe delivery remains a major challenge. Viral vectors offer high efficiency but raise concerns about immunogenicity and insertional mutagenesis, whereas non-viral systems such as lipid nanoparticles and engineered exosomes offer lower toxicity and modularity but face targeting limitations. This review highlights recent progress in CRISPR-based therapies for β-thalassemia and emerging delivery strategies to enhance clinical translation. Full article
(This article belongs to the Special Issue CRISPR-Based Genome Editing in Translational Research—Third Edition)
Show Figures

Figure 1

Back to TopTop