1
|
Xue Z, Wang L, Pan S, Yan J, You M, Yao C. The nucleic acid reactions on the nanomaterials surface for biomedicine. J Nanobiotechnology 2025; 23:308. [PMID: 40269855 PMCID: PMC12016162 DOI: 10.1186/s12951-025-03374-2] [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: 01/26/2025] [Accepted: 04/06/2025] [Indexed: 04/25/2025] Open
Abstract
Integrating nucleic acids (NAs) with nanomaterials has substantially advanced biomedical research, enabling critical applications in biosensing, drug delivery, therapeutics, and the synthesis of nanomaterials. At the core of these advances are the reactions of NAs on nanomaterial surfaces, encompassing conjugation (covalent and non-covalent), detachment (physical and chemical), and signal amplification (enzyme-mediated signal amplification, enzyme-free signal amplification, and DNA Walker). Here, we review the fundamental mechanisms and recent progress in nucleic acid reactions on nanomaterial surfaces, discuss emerging applications for diagnostics, nanomedicine, and gene therapy, and explore persistent challenges in the field. We offer a forward-looking perspective on how future developments could better control, optimize, and harness these reactions for transformative advances in nanomedicine and biomedical engineering.
Collapse
Affiliation(s)
- Zhenrui Xue
- Department of Transfusion Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, People's Republic of China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China
| | - Lu Wang
- Department of Transfusion Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, People's Republic of China
| | - Shengnan Pan
- Department of Transfusion Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, People's Republic of China
| | - Jie Yan
- Department of Transfusion Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, People's Republic of China
| | - Minli You
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China.
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China.
| | - Chunyan Yao
- Department of Transfusion Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, People's Republic of China.
| |
Collapse
|
2
|
Tiwari P, Shukla RP, Yadav K, Sharma M, Bakshi AK, Panwar D, Singh N, Agarwal N, Mugale MN, Mishra PR. YIGSR Functionalized Hybrid Exosomes Spatially Target Dasatinib to Laminin Receptors for Precision Therapy in Breast Cancer. Adv Healthc Mater 2025; 14:e2402673. [PMID: 39962816 DOI: 10.1002/adhm.202402673] [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: 07/21/2024] [Revised: 12/17/2024] [Indexed: 04/08/2025]
Abstract
In this study, YIGSR-functionalized exosomes (Exo) are engineered and hybridized with lipid polymeric nanoparticles (LPNPs) followed by loading of chemotherapy Dasatinib (DST) to spatially target laminin receptors on tumors. Exo derived from differentiated macrophages are engineered with YIGSR targeting peptides.These YIGSR-Exo are subsequently fused with LPNPs membranes using the freeze-thaw method, resulting in fused hybrid YIGSR-Exo, which are then loaded with DST, creating DST-FuNP@YIGSR-Exo and targeted breast cancer (BC), leading to enhanced mitochondrial membrane potential (54.50 ±5.0%), increased reactive oxygen species (59.50 ± 6.0%), and apoptosis (63 ± 6.5%), ultimately inducing cell death. Further, cellular uptake and receptor blocking studies confirm the binding affinity and interaction of DST-FuNP@YIGSR-Exo with laminin receptors, Intravenous pharmacokinetic analysis of DST-FuNP@YIGSR-Exo reveals a significant improvement in AUC0-∞, with a 20.84-fold increase compared to free DST and a 1.61-fold enhancement over DST-FuNP@Exo. This is further supported by in vivo imaging and demonstrated improved tumor localization. A tumor regression study shows a 6.8-fold reduction in tumors. Tumor tissue-specific IHC for the Ki67 proliferative marker is significantly reduced in the targeted formulation. The potential of DST-FuNP@YIGSR-Exo as an effective carrier for delivering chemotherapeutic drugs, paving the path for the advancement of biologically obtained nanocarriers for targeted breast cancer.
Collapse
Affiliation(s)
- Pratiksha Tiwari
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Ravi Prakash Shukla
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Krishna Yadav
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Madhu Sharma
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Avijit Kumar Bakshi
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Dilip Panwar
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Neha Singh
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Neha Agarwal
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Madhav Nilakanth Mugale
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Prabhat Ranjan Mishra
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
- Academy of Scientific and Innovation Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| |
Collapse
|
3
|
Abdulmonem WA, Ahsan M, Mallick AK, Mohamed AH, Waggiallah HA, Shafie A, Alzahrani HS, Ashour AA, Rab SO, Mirdad MT, Ali HTO. The Role of Exosomal miRNAs in Female Infertility: Therapeutic Potential and Mechanisms of Action. Stem Cell Rev Rep 2025:10.1007/s12015-025-10869-w. [PMID: 40126819 DOI: 10.1007/s12015-025-10869-w] [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] [Accepted: 03/17/2025] [Indexed: 03/26/2025]
Abstract
Reproductive disorders, including preeclampsia (PE), endometriosis, premature ovarian failure (POF), and polycystic ovary syndrome (PCOS), present substantial challenges to women's reproductive health. Exosomes (EXOs) are cell-derived vesicles containing molecules that influence target cells' gene expression and cellular behavior. Among their cargo, microRNAs (miRNAs)-short, non-coding RNAs typically 19-25 nucleotides in length-play a crucial role in post-transcriptional gene regulation and have been extensively studied for their therapeutic potential. miRNAs are considered therapeutic targets because they regulate key cellular pathways such as proliferation, apoptosis, angiogenesis, and tissue repair. This review examines the role of exosomal miRNAs from sources such as mesenchymal stem cells (MSCs), plasma, and amniotic fluid in female reproductive disorders, including PE, POF, PCOS, and endometriosis. We discuss their biological origins, mechanisms of miRNA sorting and packaging, and their therapeutic applications in modulating disease progression. By categorizing miRNAs according to their beneficial or detrimental effects in specific conditions, we aim to simplify the understanding of their roles in female infertility.
Collapse
Affiliation(s)
- Waleed Al Abdulmonem
- Department of Pathology, College of Medicine, Qassim University, Buraydah, Saudi Arabia
| | - Marya Ahsan
- Department of Pharmacology, College of Medicine, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, 13317, Saudi Arabia
| | - Ayaz Khurram Mallick
- Department of Clinical Biochemistry, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Asma'a H Mohamed
- Department of Optometry Techniques, Technical College Al-Mussaib, Al-Furat Al-Awsat Technical University, Najaf, Iraq.
| | - Hisham Ali Waggiallah
- Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam Bin Abdulaziz University, Alkharj, 11942, Saudi Arabia
| | - Alaa Shafie
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, P.O.Box 11099, Taif, 21944, Saudi Arabia
| | - Hassan Swed Alzahrani
- Counseling Healthy Marriage, Jeddah Regional Laboratory, Jeddah First Cluster , Jeddah, Saudi Arabia
| | - Amal Adnan Ashour
- Department of Oral & Maxillofacial Surgery and Diagnostic Sciences, Faculty of Dentistry, Taif University, P.O. Box 11099, Taif, 21944, Saudi Arabia
| | - Safia Obaidur Rab
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia
| | - Mohammed Tarek Mirdad
- Medical Intern MBBS, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Hatim T O Ali
- Obstetrics and Gynecology, College of Medicine, King Khalid University, Abha, Saudi Arabia
| |
Collapse
|
4
|
Sangubotla R, Gubbiyappa KS, Devarapogu R, Kim J. Modern insights of nanotheranostics in the glioblastoma: An updated review. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167653. [PMID: 39756713 DOI: 10.1016/j.bbadis.2024.167653] [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: 08/29/2024] [Revised: 12/08/2024] [Accepted: 12/28/2024] [Indexed: 01/07/2025]
Abstract
Glioblastoma multiforme (GBM) is a highly malignant subtype of glioma, originating from the glial cells that provide support to other neurons in the brain. GBM predominantly impacts the cerebral hemisphere of the brain, with minimal effects on the cerebellum, brain stem, or spinal cord. Individuals diagnosed with GBM commonly encounter a range of symptoms, starting from auditory abnormalities to seizures. Recently, cell membrane-camouflaged nanoparticles (CMCNPs) are evolving as promising theranostic agents that can carry specific biological moieties from their biological origin and effectively target GBM cells. Moreover, exosomes have gained widespread scientific attention as an effective drug delivery approach due to their excellent stability in the bloodstream, high biocompatibility, low immune response, and inherent targeting capabilities. Exosomes derived from specific cell types can transport endogenous signaling molecules that have therapeutic promise for GBM therapy. In this context, researchers are utilizing various techniques to isolate exosomes from liquid biomarkers from patients, such as serum and cerebrospinal fluid (CSF). Proper isolation of exosomes may induce the clinical diagnosis in GBM due to their commercial accessibility and real-time monitoring options. Since exosomes are unable to penetrate the blood-brain barrier (BBB), strategic theranostic methods are ideal. For this, understanding interactions between glioma-specific exosomes in the TME and biomarkers is necessary. The versatile characteristics of NPs and their capacity to cross the BBB enable them to be indispensable against GBM. In this review article, we discussed the recent theranostic applications of nanotechnology by comparing the limitations of existing nanotechnology-based approaches.
Collapse
Affiliation(s)
- Roopkumar Sangubotla
- Department of Chemical and Biological Engineering, Gachon University, 1342 Seongnam Daero, Seongnam-Si, Gyeonggi-Do 13120, Republic of Korea
| | - Kumar Shiva Gubbiyappa
- GITAM School of Pharmacy, GITAM Deemed to be University, Rudraram, Patencheru, Sangareddy Dist, 502329, Telangana, India
| | - Rajakumari Devarapogu
- Department of Zoology, Sri Venkateswara University, Tirupati, Andhra Pradesh 517502, India
| | - Jongsung Kim
- Department of Chemical and Biological Engineering, Gachon University, 1342 Seongnam Daero, Seongnam-Si, Gyeonggi-Do 13120, Republic of Korea.
| |
Collapse
|
5
|
He F, Du W, Liu Y, Ling Y, Xu M, Liu J, Song P, Fang Z, Yue Z, Duan J, Wang L. Exosome-equipped TNF antisense oligodeoxynucleotide or 2-deoxy-D-glucose ameliorated nonalcoholic steatohepatitis by modulating superoxide dismutase 1 in mice. Redox Biol 2025; 80:103488. [PMID: 39778469 PMCID: PMC11763583 DOI: 10.1016/j.redox.2025.103488] [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: 10/05/2024] [Revised: 12/09/2024] [Accepted: 01/02/2025] [Indexed: 01/11/2025] Open
Abstract
Inflammatory mediators tumor necrosis factor (TNF) and interleukin 1 beta (IL1β), primarily derived from hepatic macrophages in the liver, play a crucial role in the progression of nonalcoholic steatohepatitis (NASH). Meanwhile, intravenously injected exosomes are mainly distributed in the liver and predominantly taken up by hepatic macrophage. Herein, we aimed to evaluate the feasibility of targeted inhibition of TNF and IL1β expression in hepatic macrophages via exosomes as a potential therapeutic strategy for NASH. In this study, we demonstrated that antisense oligodeoxynucleotide targeting TNF (ASO-TNF) or 2-deoxy-d-glucose (2DG) effectively suppressed the expression of TNF and/or IL1β in macrophages. Exosomes loaded with ASO-TNF or 2DG were able to suppress the expression of TNF and/or IL1β in macrophages in vitro or in vivo. Furthermore, infusion of Exo/ASO-TNF or Exo/2DG significantly attenuated experimental steatohepatitis in choline deficient amino acid-defined (CDAA) or methionine and choline deficient (MCD) diet-fed mice. RNA-seq results showed that treatment with Exo/ASO-TNF or Exo/2DG significantly inhibited pro-inflammatory signaling pathways. Mechanistically, we observed that administration of Exo/ASO-TNF or Exo/2DG could attenuate NASH progression by up-regulating the expression of superoxide dismutase 1 (Sod1). Combined, our findings demonstrated that infusion of exosomes loaded with ASO-TNF or 2DG alleviated experimental steatohepatitis in murine models. Thus, infusion of exosomes loaded with anti-inflammatory agents holds promise as a potential therapeutic strategy for NASH treatment.
Collapse
Affiliation(s)
- Fei He
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, 710032, Xi'an, China.
| | - Wei Du
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, 710032, Xi'an, China.
| | - Yingying Liu
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, 710032, Xi'an, China.
| | - Yuwei Ling
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, 710032, Xi'an, China.
| | - Ming Xu
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, 710032, Xi'an, China.
| | - Jingjing Liu
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, 710032, Xi'an, China.
| | - Ping Song
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, 710032, Xi'an, China.
| | - Zhiqiang Fang
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, 710032, Xi'an, China.
| | - Zhensheng Yue
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, 710032, Xi'an, China.
| | - Juanli Duan
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, 710032, Xi'an, China.
| | - Lin Wang
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, 710032, Xi'an, China.
| |
Collapse
|
6
|
Wang B, Zhao Y, Zhao M, Han Y, Zhang H, Kong W, Qu F. Al-Based Metal-Organic Nanoslice-Coupled Phospholipid Layer Enables Highly Enriched Biological External Vesicles. Anal Chem 2025; 97:3319-3327. [PMID: 39899402 DOI: 10.1021/acs.analchem.4c05089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2025]
Abstract
Systematic analysis of the information content of biological external vesicles (BEVs) is essential for understanding the complex relationships between metabolic processes and cells at a systemic level. Although considerable efforts have been made to enrich BEVs, their comprehensive analysis in a way that maintains and stabilizes information maintenance at high doses remains a challenge. To address this issue, we developed a metal-organic nanoslice-coupled exocytosis phospholipid layer method that utilized subtle interactions between the metal-organic nanoslice and the phospholipid molecular layer of the membrane to achieve the convenient and efficient enrichment of BEVs. The method bypassed the conventional ultracentrifugation and size exclusion separation and helped to highly preserve the internal information on BEVs. By integrating and analyzing the enriched BEVs for elements such as membrane proteins and endotoxins, the simplicity and robustness of the metal-organic nanoslice-coupled vesicle surface phospholipid layer technology were verified, which provided a reliable platform for studying the cellular events associated with outer vesicles and expanded the biological applications of the metal-organic nanoslice.
Collapse
Affiliation(s)
- Bangming Wang
- Key Laboratory of Life-Organic Analysis of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China
| | - Yan Zhao
- Key Laboratory of Life-Organic Analysis of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China
| | - Mingzhu Zhao
- Key Laboratory of Life-Organic Analysis of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China
| | - Yuke Han
- Key Laboratory of Life-Organic Analysis of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China
| | - Hui Zhang
- School of Molecular Medicine, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, P. R. China
| | - Weiheng Kong
- Key Laboratory of Life-Organic Analysis of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China
| | - Fengli Qu
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou 310022, P. R. China
- School of Molecular Medicine, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, P. R. China
| |
Collapse
|
7
|
Lee HY, Lee JW. Spheroid-Exosome-Based Bioprinting Technology in Regenerative Medicine. J Funct Biomater 2024; 15:345. [PMID: 39590549 PMCID: PMC11595066 DOI: 10.3390/jfb15110345] [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: 10/11/2024] [Revised: 11/13/2024] [Accepted: 11/13/2024] [Indexed: 11/28/2024] Open
Abstract
Since the discovery that exosomes can exchange genes, their potential use as tools for tissue regeneration, disease diagnosis, and therapeutic applications has drawn significant attention. Emerging three-dimensional (3D) printing technologies, such as bioprinting, which allows the printing of cells, proteins, DNA, and other biological materials, have demonstrated the potential to create complex body tissues or personalized 3D models. The use of 3D spheroids in bioprinting facilitates volumetric tissue reconstruction and accelerates tissue regeneration via exosome secretion. In this review, we discussed a convergence approach between two promising technologies for bioprinting and exosomes in regenerative medicine. Among the various 3D cell culture methods used for exosome production, we focused on spheroids, which are suitable for mass production by bioprinting. We then summarized the research results on cases of bioprinting applications using the spheroids and exosomes produced. If a large number of spheroids can be supplied through bioprinting, the spheroid-exosome-based bioprinting technology will provide new possibilities for application in tissue regeneration, disease diagnosis, and treatment.
Collapse
Affiliation(s)
- Hwa-Yong Lee
- Division of Science Education, Kangwon National University, Chuncheon 24341, Republic of Korea;
| | - Jin Woo Lee
- Department of Molecular Medicine, College of Medicine, Gachon University, Incheon 21999, Republic of Korea
| |
Collapse
|
8
|
Chen Y, Liu Z, Zhang B, Wu H, Lv X, Zhang Y, Lin Y. Biomedical Utility of Non-Enzymatic DNA Amplification Reaction: From Material Design to Diagnosis and Treatment. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2404641. [PMID: 39152925 DOI: 10.1002/smll.202404641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 08/04/2024] [Indexed: 08/19/2024]
Abstract
Nucleic acid nanotechnology has become a promising strategy for disease diagnosis and treatment, owing to remarkable programmability, precision, and biocompatibility. However, current biosensing and biotherapy approaches by nucleic acids exhibit limitations in sensitivity, specificity, versatility, and real-time monitoring. DNA amplification reactions present an advantageous strategy to enhance the performance of biosensing and biotherapy platforms. Non-enzymatic DNA amplification reaction (NEDAR), such as hybridization chain reaction and catalytic hairpin assembly, operate via strand displacement. NEDAR presents distinct advantages over traditional enzymatic DNA amplification reactions, including simplified procedures, milder reaction conditions, higher specificity, enhanced controllability, and excellent versatility. Consequently, research focusing on NEDAR-based biosensing and biotherapy has garnered significant attention. NEDAR demonstrates high efficacy in detecting multiple types of biomarkers, including nucleic acids, small molecules, and proteins, with high sensitivity and specificity, enabling the parallel detection of multiple targets. Besides, NEDAR can strengthen drug therapy, cellular behavior control, and cell encapsulation. Moreover, NEDAR holds promise for constructing assembled diagnosis-treatment nanoplatforms in the forms of pure DNA nanostructures and hybrid nanomaterials, which offer utility in disease monitoring and precise treatment. Thus, this paper aims to comprehensively elucidate the reaction mechanism of NEDAR and review the substantial advancements in NEDAR-based diagnosis and treatment over the past five years, encompassing NEDAR-based design strategies, applications, and prospects.
Collapse
Affiliation(s)
- Ye Chen
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Zhiqiang Liu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Bowen Zhang
- Department of Prosthodontics, Tianjin Stomatological Hospital, School of Medicine, Nankai University, Tianjin, 300041, P. R. China
- Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin, 300041, P. R. China
| | - Haoyan Wu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Xiaoying Lv
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Yuxin Zhang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Yunfeng Lin
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
- Sichuan Provincial Engineering Research Center of Oral Biomaterials, Chengdu, Sichuan, 610041, P. R. China
- National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| |
Collapse
|
9
|
Kaupbayeva B, Tsoy A, Safarova (Yantsen) Y, Nurmagambetova A, Murata H, Matyjaszewski K, Askarova S. Unlocking Genome Editing: Advances and Obstacles in CRISPR/Cas Delivery Technologies. J Funct Biomater 2024; 15:324. [PMID: 39590528 PMCID: PMC11595195 DOI: 10.3390/jfb15110324] [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: 09/24/2024] [Revised: 10/18/2024] [Accepted: 10/22/2024] [Indexed: 11/28/2024] Open
Abstract
CRISPR/Cas9 (clustered regularly interspaced short palindromic repeats associated with protein 9) was first identified as a component of the bacterial adaptive immune system and subsequently engineered into a genome-editing tool. The key breakthrough in this field came with the realization that CRISPR/Cas9 could be used in mammalian cells to enable transformative genetic editing. This technology has since become a vital tool for various genetic manipulations, including gene knockouts, knock-in point mutations, and gene regulation at both transcriptional and post-transcriptional levels. CRISPR/Cas9 holds great potential in human medicine, particularly for curing genetic disorders. However, despite significant innovation and advancement in genome editing, the technology still possesses critical limitations, such as off-target effects, immunogenicity issues, ethical considerations, regulatory hurdles, and the need for efficient delivery methods. To overcome these obstacles, efforts have focused on creating more accurate and reliable Cas9 nucleases and exploring innovative delivery methods. Recently, functional biomaterials and synthetic carriers have shown great potential as effective delivery vehicles for CRISPR/Cas9 components. In this review, we attempt to provide a comprehensive survey of the existing CRISPR-Cas9 delivery strategies, including viral delivery, biomaterials-based delivery, synthetic carriers, and physical delivery techniques. We underscore the urgent need for effective delivery systems to fully unlock the power of CRISPR/Cas9 technology and realize a seamless transition from benchtop research to clinical applications.
Collapse
Affiliation(s)
- Bibifatima Kaupbayeva
- Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Astana 010000, Kazakhstan
| | - Andrey Tsoy
- Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Astana 010000, Kazakhstan
| | - Yuliya Safarova (Yantsen)
- Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Astana 010000, Kazakhstan
| | | | - Hironobu Murata
- Chemistry Department, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Krzysztof Matyjaszewski
- Chemistry Department, Carnegie Mellon University, Pittsburgh, PA 15213, USA
- Department of Molecular Physics, Faculty of Chemistry, Lodz University of Technology, 90-924 Łódź, Poland
| | - Sholpan Askarova
- Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Astana 010000, Kazakhstan
| |
Collapse
|
10
|
Di Bella MA, Taverna S. Extracellular Vesicles: Diagnostic and Therapeutic Applications in Cancer. BIOLOGY 2024; 13:716. [PMID: 39336143 PMCID: PMC11446462 DOI: 10.3390/biology13090716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/09/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024]
Abstract
In recent years, knowledge of cell-released extracellular vesicle (EV) functions has undergone rapid growth. EVs are membrane vesicles loaded with proteins, nucleic acids, lipids, and bioactive molecules. Once released into the extracellular space, EVs are delivered to target cells that may go through modifications in physiological or pathological conditions. EVs are nano shuttles with a crucial role in promoting short- and long-distance cell-cell communication. Comprehension of the mechanism that regulates this process is a benefit for both medicine and basic science. Currently, EVs attract immense interest in precision and nanomedicine for their potential use in diagnosis, prognosis, and therapies. This review reports the latest advances in EV studies, focusing on the nature and features of EVs and on conventional and emerging methodologies used for their separation, characterization, and visualization. By searching an extended portion of the relevant literature, this work aims to give a summary of advances in nanomedical applications of EVs. Moreover, concerns that require further studies before translation to clinical applications are discussed.
Collapse
Affiliation(s)
- Maria Antonietta Di Bella
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, 90133 Palermo, Italy
| | - Simona Taverna
- Institute of Translational Pharmacology (IFT), National Research Council (CNR), 90146 Palermo, Italy
| |
Collapse
|
11
|
Yao X, He D, Wei P, Niu Z, Chen H, Li L, Fu P, Wang Y, Lou S, Qian S, Zheng J, Zuo G, Wang K. DNA Nanomaterial-Empowered Surface Engineering of Extracellular Vesicles. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2306852. [PMID: 38041689 DOI: 10.1002/adma.202306852] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 11/30/2023] [Indexed: 12/03/2023]
Abstract
Extracellular vesicles (EVs) are cell-secreted biological nanoparticles that are critical mediators of intercellular communication. They contain diverse bioactive components, which are promising diagnostic biomarkers and therapeutic agents. Their nanosized membrane-bound structures and innate ability to transport functional cargo across major biological barriers make them promising candidates as drug delivery vehicles. However, the complex biology and heterogeneity of EVs pose significant challenges for their controlled and actionable applications in diagnostics and therapeutics. Recently, DNA molecules with high biocompatibility emerge as excellent functional blocks for surface engineering of EVs. The robust Watson-Crick base pairing of DNA molecules and the resulting programmable DNA nanomaterials provide the EV surface with precise structural customization and adjustable physical and chemical properties, creating unprecedented opportunities for EV biomedical applications. This review focuses on the recent advances in the utilization of programmable DNA to engineer EV surfaces. The biology, function, and biomedical applications of EVs are summarized and the state-of-the-art achievements in EV isolation, analysis, and delivery based on DNA nanomaterials are introduced. Finally, the challenges and new frontiers in EV engineering are discussed.
Collapse
Affiliation(s)
- Xuxiang Yao
- Cixi Biomedical Research Institute, Wenzhou Medical University, Ningbo, 315300, P. R. China
| | - Dongdong He
- Cixi Biomedical Research Institute, Wenzhou Medical University, Ningbo, 315300, P. R. China
| | - Pengyao Wei
- Cixi Biomedical Research Institute, Wenzhou Medical University, Ningbo, 315300, P. R. China
| | - Zitong Niu
- Cixi Biomedical Research Institute, Wenzhou Medical University, Ningbo, 315300, P. R. China
| | - Hao Chen
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315300, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Lin Li
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315300, P. R. China
| | - Pan Fu
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315300, P. R. China
| | - Yiting Wang
- College of Chemistry, Jilin Normal University, Siping, 136000, P. R. China
| | - Saiyun Lou
- Second Clinical Medicine Faculty, Zhejiang Chinese Medical University, Hangzhou, 310000, P. R. China
- Ningbo Second Hospital, Ningbo, 315010, P. R. China
| | - Sihua Qian
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315300, P. R. China
| | - Jianping Zheng
- Cixi Biomedical Research Institute, Wenzhou Medical University, Ningbo, 315300, P. R. China
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315300, P. R. China
| | - Guokun Zuo
- Cixi Biomedical Research Institute, Wenzhou Medical University, Ningbo, 315300, P. R. China
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315300, P. R. China
| | - Kaizhe Wang
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315300, P. R. China
| |
Collapse
|
12
|
Maiullari F, Milan M, Chirivì M, Ceraolo MG, Bousselmi S, Fratini N, Galbiati M, Fortunato O, Costantini M, Brambilla F, Mauri P, Di Silvestre D, Calogero A, Sciarra T, Rizzi R, Bearzi C. Enhancing neovascularization post-myocardial infarction through injectable hydrogel functionalized with endothelial-derived EVs. Biofabrication 2024; 16:045009. [PMID: 38986455 DOI: 10.1088/1758-5090/ad6190] [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: 01/19/2024] [Accepted: 07/10/2024] [Indexed: 07/12/2024]
Abstract
Over the past three decades, cell therapy development has fallen short of expectations, with many cellular sources demonstrating a 'Janus effect' and raising safety concerns. Extracellular vesicles (EVs), supported by advanced technologies, present a promising avenue in regenerative medicine, offering benefits such as immune tolerance and avoidance of negative aspects associated with cell transplants. Our previous research showcased enhanced and organized subcutaneous vascularization using three-dimensional bioprinted patches containing HUVEC-derived EVs in immunodeficient animal models. In this context, stress conditions on the cells of origin further boosted the EVs' neoangiogenic potential. Since neovascularization is the first regenerative target requiring restoration, the present study aims to complement our previous work by employing an injectable gelatin methacrylate (GelMA) hydrogel functionalized with HUVEC-derived EVs in a pathological condition of acute myocardial infarction. This bioactive hydrogel resulted in reduced fibrosis, improved contractility, and promoted angiogenesis, showing promise in countering tissue deterioration and addressing vascular deficits. Moreover, the molecular characterization of EVs through miRNome and proteomic analyses further supports their potential as bio-additives for hydrogel functionalization. This cell-free approach mitigates immune rejection and oncogenic risks, offering innovative therapeutic advantages.
Collapse
Affiliation(s)
- Fabio Maiullari
- Fondazione Istituto Nazionale di Genetica Molecolare 'Romeo ed Enrica Invernizzi', Via Francesco Sforza, 35, 20122 Milan, Italy
- PhD Program in Cellular and Molecular Biology, Department of Biology, University of Rome 'Tor Vergata', Via della RicercaScientifica, 1, 00133 Rome, Italy
| | - Marika Milan
- Fondazione Istituto Nazionale di Genetica Molecolare 'Romeo ed Enrica Invernizzi', Via Francesco Sforza, 35, 20122 Milan, Italy
- Neurology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Maila Chirivì
- Neurology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Department of Molecular Medicine, Sapienza University, Viale Regina Elena, 324, 00161 Rome, Italy
| | - Maria Grazia Ceraolo
- Fondazione Istituto Nazionale di Genetica Molecolare 'Romeo ed Enrica Invernizzi', Via Francesco Sforza, 35, 20122 Milan, Italy
- Neurology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Salma Bousselmi
- PhD Program in Cellular and Molecular Biology, Department of Biology, University of Rome 'Tor Vergata', Via della RicercaScientifica, 1, 00133 Rome, Italy
- Neurology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Nicole Fratini
- Fondazione Istituto Nazionale di Genetica Molecolare 'Romeo ed Enrica Invernizzi', Via Francesco Sforza, 35, 20122 Milan, Italy
- Department of Molecular Medicine, Sapienza University, Viale Regina Elena, 324, 00161 Rome, Italy
| | - Matteo Galbiati
- Fondazione Istituto Nazionale di Genetica Molecolare 'Romeo ed Enrica Invernizzi', Via Francesco Sforza, 35, 20122 Milan, Italy
- Institute for Biomedical Technologies, National Research Council, Via Fratelli Cervi, 93, 20054 Segrate, Milan, Italy
| | - Orazio Fortunato
- Tumor Genomics Unit, Department of Research, IRCCS Fondazione Istituto Nazionale dei Tumori, 20133 Milan, Italy
| | - Marco Costantini
- Institute of Physical Chemistry-Polish Academy of Sciences, MarcinaKasprzaka 44/52, 01-224 Warsaw, Poland
| | - Francesca Brambilla
- Institute for Biomedical Technologies, National Research Council, Via Fratelli Cervi, 93, 20054 Segrate, Milan, Italy
| | - Pierluigi Mauri
- Institute for Biomedical Technologies, National Research Council, Via Fratelli Cervi, 93, 20054 Segrate, Milan, Italy
| | - Dario Di Silvestre
- Institute for Biomedical Technologies, National Research Council, Via Fratelli Cervi, 93, 20054 Segrate, Milan, Italy
| | - Antonella Calogero
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, C.so della Repubblica 79, 04100 Latina, Italy
| | - Tommaso Sciarra
- Joint Veteran Center, Scientific Department, Army Medical Center, 00184 Rome, Italy
| | - Roberto Rizzi
- Fondazione Istituto Nazionale di Genetica Molecolare 'Romeo ed Enrica Invernizzi', Via Francesco Sforza, 35, 20122 Milan, Italy
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, C.so della Repubblica 79, 04100 Latina, Italy
| | - Claudia Bearzi
- Fondazione Istituto Nazionale di Genetica Molecolare 'Romeo ed Enrica Invernizzi', Via Francesco Sforza, 35, 20122 Milan, Italy
- Institute for Biomedical Technologies, National Research Council, Via Fratelli Cervi, 93, 20054 Segrate, Milan, Italy
| |
Collapse
|
13
|
Bahadorani M, Nasiri M, Dellinger K, Aravamudhan S, Zadegan R. Engineering Exosomes for Therapeutic Applications: Decoding Biogenesis, Content Modification, and Cargo Loading Strategies. Int J Nanomedicine 2024; 19:7137-7164. [PMID: 39050874 PMCID: PMC11268655 DOI: 10.2147/ijn.s464249] [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: 03/04/2024] [Accepted: 06/20/2024] [Indexed: 07/27/2024] Open
Abstract
Exosomes emerge from endosomal invagination and range in size from 30 to 200 nm. Exosomes contain diverse proteins, lipids, and nucleic acids, which can indicate the state of various physiological and pathological processes. Studies have revealed the remarkable clinical potential of exosomes in diagnosing and prognosing multiple diseases, including cancer, cardiovascular disorders, and neurodegenerative conditions. Exosomes also have the potential to be engineered and deliver their cargo to a specific target. However, further advancements are imperative to optimize exosomes' diagnostic and therapeutic capabilities for practical implementation in clinical settings. This review highlights exosomes' diagnostic and therapeutic applications, emphasizing their engineering through simple incubation, biological, and click chemistry techniques. Additionally, the loading of therapeutic agents onto exosomes, utilizing passive and active strategies, and exploring hybrid and artificial exosomes are discussed.
Collapse
Affiliation(s)
- Mehrnoosh Bahadorani
- Department of Nanoengineering, Joint School of Nanoscience & Nanoengineering, North Carolina Agriculture and Technical State University, Greensboro, NC, USA
| | - Mahboobeh Nasiri
- Department of Nanoengineering, Joint School of Nanoscience & Nanoengineering, North Carolina Agriculture and Technical State University, Greensboro, NC, USA
| | - Kristen Dellinger
- Department of Nanoengineering, Joint School of Nanoscience & Nanoengineering, North Carolina Agriculture and Technical State University, Greensboro, NC, USA
| | - Shyam Aravamudhan
- Department of Nanoengineering, Joint School of Nanoscience & Nanoengineering, North Carolina Agriculture and Technical State University, Greensboro, NC, USA
| | - Reza Zadegan
- Department of Nanoengineering, Joint School of Nanoscience & Nanoengineering, North Carolina Agriculture and Technical State University, Greensboro, NC, USA
| |
Collapse
|
14
|
Li W, Liu S, Wang Z, Gou L, Ou Y, Zhu X, Zhou Y, Zhang T, Liu J, Zheng X, Berggren PO, Liu J, Zheng X. Programmable DNA Scaffolds Enable Orthogonal Engineering of Cell Membrane-Based Nanovesicles for Therapeutic Development. NANO LETTERS 2024. [PMID: 38856668 DOI: 10.1021/acs.nanolett.4c02193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Cell membrane-based nanovesicles (CMNVs) play pivotal roles in biomolecular transportation in living organisms and appear as attractive bioinformed nanomaterials for theranostic applications. However, the current surface-engineering technologies are limited in flexibility and orthogonality, making it challenging to simultaneously display multiple different ligands on the CMNV surface in a precisely controlled manner. Here, we developed a DNA scaffold-programmed approach to orthogonally engineer CMNVs with versatile ligands. The designed DNA scaffolds can rapidly anchor onto the CMNV surface, and their unique sequences and hybridized properties enable independent control of the loading of multiple different types of biomolecules on the CMNVs. As a result, the orthogonal engineering of CMNVs with a renal targeted peptide and a therapeutic protein at controlled ratios demonstrated an enhanced renal targeting and repair potential in vivo. This study highlights that a DNA scaffold-programmed platform can provide a potent means for orthogonal and flexible surface engineering of CMNVs for diverse therapeutic purposes.
Collapse
Affiliation(s)
- Wei Li
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Shuyun Liu
- NHC Key Laboratory of Transplant Engineering and Immunology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhenghao Wang
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu 610041, China
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, SE-17176 Stockholm, Sweden
| | - Liping Gou
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yiran Ou
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xinyue Zhu
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ye Zhou
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Tianci Zhang
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jiaye Liu
- Department of Thyroid and Parathyroid Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu 610041, China
| | - Xiaowei Zheng
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu 610041, China
- Department of Molecular Medicine and Surgery, Karolinska Institutet, SE-17177 Stockholm, Sweden
| | - Per-Olof Berggren
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu 610041, China
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, SE-17176 Stockholm, Sweden
| | - Jingping Liu
- NHC Key Laboratory of Transplant Engineering and Immunology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiaofeng Zheng
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu 610041, China
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu 610041, China
| |
Collapse
|
15
|
Wang S, Jia Z, Dai M, Feng X, Tang C, Liu L, Cao L. Advances in natural and synthetic macromolecules with stem cells and extracellular vesicles for orthopedic disease treatment. Int J Biol Macromol 2024; 268:131874. [PMID: 38692547 DOI: 10.1016/j.ijbiomac.2024.131874] [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: 10/15/2023] [Revised: 04/16/2024] [Accepted: 04/24/2024] [Indexed: 05/03/2024]
Abstract
Serious orthopedic disorders resulting from myriad diseases and impairments continue to pose a considerable challenge to contemporary clinical care. Owing to its limited regenerative capacity, achieving complete bone tissue regeneration and complete functional restoration has proven challenging with existing treatments. By virtue of cellular regenerative and paracrine pathways, stem cells are extensively utilized in the restoration and regeneration of bone tissue; however, low survival and retention after transplantation severely limit their therapeutic effect. Meanwhile, biomolecule materials provide a delivery platform that improves stem cell survival, increases retention, and enhances therapeutic efficacy. In this review, we present the basic concepts of stem cells and extracellular vesicles from different sources, emphasizing the importance of using appropriate expansion methods and modification strategies. We then review different types of biomolecule materials, focusing on their design strategies. Moreover, we summarize several forms of biomaterial preparation and application strategies as well as current research on biomacromolecule materials loaded with stem cells and extracellular vesicles. Finally, we present the challenges currently impeding their clinical application for the treatment of orthopedic diseases. The article aims to provide researchers with new insights for subsequent investigations.
Collapse
Affiliation(s)
- Supeng Wang
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China; Jiujiang City Key Laboratory of Cell Therapy, The First Hospital of Jiujiang City, Jiujiang 332000, China; Ningxia Medical University, Ningxia 750004, China
| | - Zhiqiang Jia
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China
| | - Minghai Dai
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China
| | - Xujun Feng
- Jiujiang City Key Laboratory of Cell Therapy, The First Hospital of Jiujiang City, Jiujiang 332000, China
| | - Chengxuan Tang
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China
| | - Liangle Liu
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China.
| | - Lingling Cao
- Jiujiang City Key Laboratory of Cell Therapy, The First Hospital of Jiujiang City, Jiujiang 332000, China.
| |
Collapse
|
16
|
Bai Z, Zhao Y, Cui C, Yan J, Qin D, Tong J, Peng H, Liu Y, Sun L, Wu X, Li B, Li X. Multifaceted Materials for Enhanced Osteogenesis and Antimicrobial Properties on Bioplastic Polyetheretherketone Surfaces: A Review. ACS OMEGA 2024; 9:17784-17807. [PMID: 38680314 PMCID: PMC11044237 DOI: 10.1021/acsomega.4c00923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 03/05/2024] [Accepted: 03/12/2024] [Indexed: 05/01/2024]
Abstract
Implant-associated infections and the increasing number of bone implants loosening and falling off after implantation have become urgent global challenges, hence the need for intelligent alternative solutions to combat implant loosening and falling off. The application of polyetheretherketone (PEEK) in biomedical and medical therapy has aroused great interest, especially because its elastic modulus close to bone provides an effective alternative to titanium implants, thereby preventing the possibility of bone implants loosening and falling off due to the mismatch of elastic modulus. In this Review, we provide a comprehensive overview of recent advances in surface modifications to prevent bone binding deficiency and bacterial infection after implantation of bone implants, starting with inorganics for surface modification, followed by organics that can effectively promote bone integration and antimicrobial action. In addition, surface modifications derived from cells and related products of biological activity have been proposed, and there is increasing evidence of clinical potential. Finally, the advantages and future challenges of surface strategies against medical associated poor osseointegration and infection are discussed, with promising prospects for developing novel osseointegration and antimicrobial PEEK materials.
Collapse
Affiliation(s)
- Ziyang Bai
- Shanxi
Medical University School and Hospital of Stomatology, Taiyuan, Shanxi 030001, China
- Shanxi
Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, Shanxi 030001, China
| | - Yifan Zhao
- Shanxi
Medical University School and Hospital of Stomatology, Taiyuan, Shanxi 030001, China
- Shanxi
Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, Shanxi 030001, China
| | - Chenying Cui
- Shanxi
Medical University School and Hospital of Stomatology, Taiyuan, Shanxi 030001, China
- Shanxi
Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, Shanxi 030001, China
| | - Jingyu Yan
- Shanxi
Medical University School and Hospital of Stomatology, Taiyuan, Shanxi 030001, China
- Shanxi
Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, Shanxi 030001, China
| | - Danlei Qin
- Shanxi
Medical University School and Hospital of Stomatology, Taiyuan, Shanxi 030001, China
- Shanxi
Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, Shanxi 030001, China
| | - Jiahui Tong
- Shanxi
Medical University School and Hospital of Stomatology, Taiyuan, Shanxi 030001, China
- Shanxi
Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, Shanxi 030001, China
| | - Hongyi Peng
- Shanxi
Medical University School and Hospital of Stomatology, Taiyuan, Shanxi 030001, China
- Shanxi
Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, Shanxi 030001, China
| | - Yingyu Liu
- Shanxi
Medical University School and Hospital of Stomatology, Taiyuan, Shanxi 030001, China
- Shanxi
Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, Shanxi 030001, China
| | - Lingxiang Sun
- Shanxi
Medical University School and Hospital of Stomatology, Taiyuan, Shanxi 030001, China
- Shanxi
Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, Shanxi 030001, China
| | - Xiuping Wu
- Shanxi
Medical University School and Hospital of Stomatology, Taiyuan, Shanxi 030001, China
- Shanxi
Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, Shanxi 030001, China
| | - Bing Li
- Shanxi
Medical University School and Hospital of Stomatology, Taiyuan, Shanxi 030001, China
- Shanxi
Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, Shanxi 030001, China
| | - Xia Li
- Shanxi
Medical University School and Hospital of Stomatology, Taiyuan, Shanxi 030001, China
- Shanxi
Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, Shanxi 030001, China
| |
Collapse
|
17
|
Goldbloom-Helzner L, Bains H, Wang A. Approaches to Characterize and Quantify Extracellular Vesicle Surface Conjugation Efficiency. Life (Basel) 2024; 14:511. [PMID: 38672781 PMCID: PMC11051464 DOI: 10.3390/life14040511] [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: 03/15/2024] [Revised: 04/09/2024] [Accepted: 04/11/2024] [Indexed: 04/28/2024] Open
Abstract
Extracellular vesicles (EVs) are cell-secreted nanovesicles that play an important role in long-range cell-cell communication. Although EVs pose a promising alternative to cell-based therapy, targeted in vivo delivery still falls short. Many studies have explored the surface modification of EVs to enhance their targeting capabilities. However, to our knowledge, there are no standardized practices to confirm the successful surface modification of EVs or calculate the degree of conjugation on EV surfaces (conjugation efficiency). These pieces of information are essential in the reproducibility of targeted EV therapeutics and the determination of optimized conjugation conditions for EVs to see significant therapeutic effects in vitro and in vivo. This review will discuss the vast array of techniques adopted, technologies developed, and efficiency definitions made by studies that have calculated EV/nanoparticle surface conjugation efficiency and how differences between studies may contribute to differently reported conjugation efficiencies.
Collapse
Affiliation(s)
- Leora Goldbloom-Helzner
- Center for Surgical Bioengineering, Department of Surgery, School of Medicine, UC Davis Health, Sacramento, CA 95817, USA;
- Department of Biomedical Engineering, UC Davis, Davis, CA 95616, USA
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA 95817, USA
| | - Harjn Bains
- Department of Biomedical Engineering, UC Davis, Davis, CA 95616, USA
| | - Aijun Wang
- Center for Surgical Bioengineering, Department of Surgery, School of Medicine, UC Davis Health, Sacramento, CA 95817, USA;
- Department of Biomedical Engineering, UC Davis, Davis, CA 95616, USA
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA 95817, USA
| |
Collapse
|
18
|
Han P, Raveendran N, Liu C, Basu S, Jiao K, Johnson N, Moran CS, Ivanovski S. 3D bioprinted small extracellular vesicles from periodontal cells enhance mesenchymal stromal cell function. BIOMATERIALS ADVANCES 2024; 158:213770. [PMID: 38242057 DOI: 10.1016/j.bioadv.2024.213770] [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: 09/15/2023] [Revised: 01/04/2024] [Accepted: 01/10/2024] [Indexed: 01/21/2024]
Abstract
Recent research indicates that combining 3D bioprinting and small extracellular vesicles (sEVs) offers a promising 'cell-free' regenerative medicine approach for various tissue engineering applications. Nonetheless, the majority of existing research has focused on bioprinting of sEVs sourced from cell lines. There remains a notable gap in research regarding the bioprinting of sEVs derived from primary human periodontal cells and their potential impact on ligamentous and osteogenic differentiation. Here, we investigated the effect of 3D bioprinted periodontal cell sEVs constructs on the differentiation potential of human buccal fat pad-derived mesenchymal stromal cells (hBFP-MSCs). Periodontal cell-derived sEVs were enriched by size exclusion chromatography (SEC) with particle-shaped morphology, and characterized by being smaller than 200 nm in size and CD9/CD63/CD81 positive, from primary human periodontal ligament cells (hPDLCs) and human gingival fibroblasts (hGFs). The sEVs were then 3D bioprinted in 10 % gelatin methacryloyl (GelMA) via microextrusion bioprinting. Release of sEVs from bioprinted constructs was determined by DiO-labelling and confocal imaging, and CD9 ELISA. Attachment and ligament/osteogenic/cementogenic differentiation of hBFP-MSCs was assessed on bioprinted GelMA, without and with sEVs (GelMA/hPDLCs-sEVs and GelMA/hGFs-sEVs), scaffolds. hBFP-MSCs seeded on the bioprinted sEVs constructs spread well with significantly enhanced focal adhesion, mechanotransduction associated gene expression, and ligament and osteogenesis/cementogenesis differentiation markers in GelMA/hPDLCs-sEVs, compared to GelMA/hGFs-sEVs and GelMA groups. A 2-week osteogenic and ligamentous differentiation showed enhanced ALP staining, calcium formation and toluidine blue stained cells in hBFP-MSCs on bioprinted GelMA/hPDLCs-sEVs constructs compared to the other two groups. The proof-of-concept data from this study supports the notion that 3D bioprinted GelMA/hPDLCs-sEVs scaffolds promote cell attachment, as well as ligamentous, osteogenic and cementogenic differentiation, of hBFP-MSCs in vitro.
Collapse
Affiliation(s)
- Pingping Han
- The University of Queensland, School of Dentistry, Center for Orofacial Regeneration, Rehabilitation and Reconstruction (COR3), Brisbane, QLD 4006, Australia; The University of Queensland, School of Dentistry, Brisbane, QLD 4006, Australia.
| | - Nimal Raveendran
- The University of Queensland, School of Dentistry, Center for Orofacial Regeneration, Rehabilitation and Reconstruction (COR3), Brisbane, QLD 4006, Australia; The University of Queensland, School of Dentistry, Brisbane, QLD 4006, Australia
| | - Chun Liu
- The University of Queensland, School of Dentistry, Center for Orofacial Regeneration, Rehabilitation and Reconstruction (COR3), Brisbane, QLD 4006, Australia; The University of Queensland, School of Dentistry, Brisbane, QLD 4006, Australia
| | - Saraswat Basu
- The University of Queensland, School of Dentistry, Center for Orofacial Regeneration, Rehabilitation and Reconstruction (COR3), Brisbane, QLD 4006, Australia; The University of Queensland, School of Dentistry, Brisbane, QLD 4006, Australia
| | - Kexin Jiao
- The University of Queensland, School of Dentistry, Center for Orofacial Regeneration, Rehabilitation and Reconstruction (COR3), Brisbane, QLD 4006, Australia; The University of Queensland, School of Dentistry, Brisbane, QLD 4006, Australia
| | - Nigel Johnson
- The University of Queensland, School of Dentistry, Brisbane, QLD 4006, Australia
| | - Corey S Moran
- The University of Queensland, School of Dentistry, Center for Orofacial Regeneration, Rehabilitation and Reconstruction (COR3), Brisbane, QLD 4006, Australia; The University of Queensland, School of Dentistry, Brisbane, QLD 4006, Australia
| | - Sašo Ivanovski
- The University of Queensland, School of Dentistry, Center for Orofacial Regeneration, Rehabilitation and Reconstruction (COR3), Brisbane, QLD 4006, Australia; The University of Queensland, School of Dentistry, Brisbane, QLD 4006, Australia.
| |
Collapse
|
19
|
Ren X, Xu R, Xu C, Su J. Harnessing exosomes for targeted therapy: strategy and application. BIOMATERIALS TRANSLATIONAL 2024; 5:46-58. [PMID: 39220669 PMCID: PMC11362351 DOI: 10.12336/biomatertransl.2024.01.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 01/22/2024] [Accepted: 02/06/2024] [Indexed: 09/04/2024]
Abstract
Exosomes, nanoscopic extracellular vesicles produced by cells, are pivotal in mediating intracellular communication by transporting nucleic acids, proteins, lipids, and other bioactive molecules, thereby influencing physiological and pathological states. Their endogenous origin and inherent diversity confer distinct advantages over synthetic vehicles like liposomes and nanoparticles in diagnostic and therapeutic applications. Despite their potential, the clinical utility of exosomes is hampered by challenges such as limited storage stability, yield, purity, and targeting efficiency. This review focuses on exosomes as targeted therapeutic agents, examining their biogenesis, classification, isolation, and characterisation, while also addressing the current limitations in yield, purity, and targeting. We delve into the literature to propose optimisation strategies that can enhance their therapeutic efficacy and accelerate the translation of exosome-based therapies into clinical practice.
Collapse
Affiliation(s)
- Xiaoxiang Ren
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, China
| | - Ruixue Xu
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT, USA
| | - Chenjie Xu
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Jiacan Su
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, China
- Department of Orthopedic, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, China
| |
Collapse
|
20
|
Wang L, Wei X, He X, Xiao S, Shi Q, Chen P, Lee J, Guo X, Liu H, Fan Y. Osteoinductive Dental Pulp Stem Cell-Derived Extracellular Vesicle-Loaded Multifunctional Hydrogel for Bone Regeneration. ACS NANO 2024; 18:8777-8797. [PMID: 38488479 DOI: 10.1021/acsnano.3c11542] [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: 03/27/2024]
Abstract
Stem cell-derived extracellular vesicles (EVs) show great potential for promoting bone tissue regeneration. However, normal EVs (Nor-EVs) have a limited ability to direct tissue-specific regeneration. Therefore, it is necessary to optimize the osteogenic capacity of EV-based systems for repairing extensive bone defects. Herein, we show that hydrogels loaded with osteoinductive dental pulp stem cell-derived EVs (Ost-EVs) enhanced bone tissue remodeling, resulting in a 2.23 ± 0.25-fold increase in the expression of bone morphogenetic protein 2 (BMP2) compared to the hydrogel control group. Moreover, Ost-EVs led to a higher expression of alkaline phosphatase (ALP) (1.88 ± 0.16 of Ost-EVs relative to Nor-EVs) and the formation of orange-red calcium nodules (1.38 ± 0.10 of Ost-EVs relative to Nor-EVs) in vitro. RNA sequencing revealed that Ost-EVs showed significantly high miR-1246 expression. An ideal hydrogel implant should also adhere to surrounding moist tissues. In this study, we were drawn to mussel-inspired adhesive modification, where the hydrogel carrier was crafted from hyaluronic acid (HA) and polyethylene glycol derivatives, showcasing impressive tissue adhesion, self-healing capabilities, and the ability to promote bone growth. The modified HA (mHA) hydrogel was also responsive to environmental stimuli, making it an effective carrier for delivering EVs. In an ectopic osteogenesis animal model, the Ost-EV/hydrogel system effectively alleviated inflammation, accelerated revascularization, and promoted tissue mineralization. We further used a rat femoral condyle defect model to evaluate the in situ osteogenic ability of the Ost-EVs/hydrogel system. Collectively, our results suggest that Ost-EVs combined with biomaterial-based hydrogels hold promising potential for treating bone defects.
Collapse
Affiliation(s)
- Li Wang
- Key Laboratory for Biomechanics and Mechanobiology (Beihang University) of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, P.R. China
| | - Xinbo Wei
- Key Laboratory for Biomechanics and Mechanobiology (Beihang University) of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, P.R. China
| | - Xi He
- Key Laboratory for Biomechanics and Mechanobiology (Beihang University) of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, P.R. China
| | - Shengzhao Xiao
- Key Laboratory for Biomechanics and Mechanobiology (Beihang University) of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, P.R. China
- Department of Orthodontics, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| | - Qiusheng Shi
- Key Laboratory for Biomechanics and Mechanobiology (Beihang University) of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, P.R. China
| | - Peng Chen
- Department of Ultrasound, The Third Medical Center, Chinese PLA General Hospital, Beijing 100039, P.R. China
| | - Jesse Lee
- Arova Biosciences, Inc., Life Sciences Innovation Hub, Calgary Alberta T2L 1Y8, Canada
| | - Ximin Guo
- Department of Advanced Interdisciplinary Studies, Institute of Basic Medical Sciences, Beijing 100850, P.R. China
| | - Haifeng Liu
- Key Laboratory for Biomechanics and Mechanobiology (Beihang University) of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, P.R. China
| | - Yubo Fan
- Key Laboratory for Biomechanics and Mechanobiology (Beihang University) of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, P.R. China
| |
Collapse
|
21
|
Zeng B, Li Y, Xia J, Xiao Y, Khan N, Jiang B, Liang Y, Duan L. Micro Trojan horses: Engineering extracellular vesicles crossing biological barriers for drug delivery. Bioeng Transl Med 2024; 9:e10623. [PMID: 38435823 PMCID: PMC10905561 DOI: 10.1002/btm2.10623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 10/05/2023] [Accepted: 11/09/2023] [Indexed: 03/05/2024] Open
Abstract
The biological barriers of the body, such as the blood-brain, placental, intestinal, skin, and air-blood, protect against invading viruses and bacteria while providing necessary physical support. However, these barriers also hinder the delivery of drugs to target tissues, reducing their therapeutic efficacy. Extracellular vesicles (EVs), nanostructures with a diameter ranging from 30 nm to 10 μm secreted by cells, offer a potential solution to this challenge. These natural vesicles can effectively pass through various biological barriers, facilitating intercellular communication. As a result, artificially engineered EVs that mimic or are superior to the natural ones have emerged as a promising drug delivery vehicle, capable of delivering drugs to almost any body part to treat various diseases. This review first provides an overview of the formation and cross-species uptake of natural EVs from different organisms, including animals, plants, and bacteria. Later, it explores the current clinical applications, perspectives, and challenges associated with using engineered EVs as a drug delivery platform. Finally, it aims to inspire further research to help bioengineered EVs effectively cross biological barriers to treat diseases.
Collapse
Affiliation(s)
- Bin Zeng
- Graduate SchoolGuangxi University of Chinese MedicineNanningGuangxiChina
- Department of Orthopedics, Shenzhen Intelligent Orthopaedics and Biomedical Innovation Platform, Guangdong Artificial Intelligence Biomedical Innovation Platform, Shenzhen Second People's Hospitalthe First Affiliated Hospital of Shenzhen UniversityShenzhenGuangdongChina
| | - Ying Li
- Department of Orthopedics, Shenzhen Intelligent Orthopaedics and Biomedical Innovation Platform, Guangdong Artificial Intelligence Biomedical Innovation Platform, Shenzhen Second People's Hospitalthe First Affiliated Hospital of Shenzhen UniversityShenzhenGuangdongChina
| | - Jiang Xia
- Department of ChemistryThe Chinese University of Hong Kong, ShatinHong Kong SARChina
| | - Yin Xiao
- School of Medicine and Dentistry & Menzies Health Institute Queensland, SouthportGold CoastQueenslandAustralia
| | - Nawaz Khan
- Department of Orthopedics, Shenzhen Intelligent Orthopaedics and Biomedical Innovation Platform, Guangdong Artificial Intelligence Biomedical Innovation Platform, Shenzhen Second People's Hospitalthe First Affiliated Hospital of Shenzhen UniversityShenzhenGuangdongChina
| | - Bin Jiang
- Graduate SchoolGuangxi University of Chinese MedicineNanningGuangxiChina
- R&D Division, Eureka Biotech Inc, PhiladelphiaPennsylvaniaUSA
| | - Yujie Liang
- Department of Child and Adolescent Psychiatry, Shenzhen Kangning HospitalShenzhen Mental Health Center, Shenzhen Key Laboratory for Psychological Healthcare and Shenzhen Institute of Mental HealthShenzhenGuangdongChina
| | - Li Duan
- Graduate SchoolGuangxi University of Chinese MedicineNanningGuangxiChina
- Department of Orthopedics, Shenzhen Intelligent Orthopaedics and Biomedical Innovation Platform, Guangdong Artificial Intelligence Biomedical Innovation Platform, Shenzhen Second People's Hospitalthe First Affiliated Hospital of Shenzhen UniversityShenzhenGuangdongChina
| |
Collapse
|
22
|
Liao Y, Zhang Z, Ouyang L, Mi B, Liu G. Engineered Extracellular Vesicles in Wound Healing: Design, Paradigms, and Clinical Application. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2307058. [PMID: 37806763 DOI: 10.1002/smll.202307058] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/20/2023] [Indexed: 10/10/2023]
Abstract
The severe quality of life and economic burden imposed by non-healing skin wounds, infection risks, and treatment costs are affecting millions of patients worldwide. To mitigate these challenges, scientists are relentlessly seeking effective treatment measures. In recent years, extracellular vesicles (EVs) have emerged as a promising cell-free therapy strategy, attracting extensive attention from researchers. EVs mediate intercellular communication, possessing excellent biocompatibility and stability. These features make EVs a potential tool for treating a plethora of diseases, including those related to wound repair. However, there is a growing focus on the engineering of EVs to overcome inherent limitations such as low production, relatively fixed content, and targeting capabilities of natural EVs. This engineering could improve both the effectiveness and specificity of EVs in wound repair treatments. In light of this, the present review will introduce the latest progress in the design methods and experimental paradigms of engineered EVs applied in wound repair. Furthermore, it will comprehensively analyze the current clinical research status and prospects of engineered EVs within this field.
Collapse
Affiliation(s)
- Yuheng Liao
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Zhenhe Zhang
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Lizhi Ouyang
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Bobin Mi
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Guohui Liu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
| |
Collapse
|
23
|
Doerfler R, Yerneni S, Newby A, Chaudhary N, Shu A, Fein K, Hofstatter Azambuja J, Whitehead KA. Characterization and comparison of human and mouse milk cells. PLoS One 2024; 19:e0297821. [PMID: 38295101 PMCID: PMC10830055 DOI: 10.1371/journal.pone.0297821] [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: 08/07/2023] [Accepted: 01/12/2024] [Indexed: 02/02/2024] Open
Abstract
Recent data has characterized human milk cells with unprecedented detail and provided insight into cell populations. While such analysis of freshly expressed human milk has been possible, studies of cell functionality within the infant have been limited to animal models. One commonly used animal model for milk research is the mouse; however, limited data are available describing the composition of mouse milk. In particular, the maternal cells of mouse milk have not been previously characterized in detail, in part due to the difficulty in collecting sufficient volumes of mouse milk. In this study, we have established a method to collect high volumes of mouse milk, isolate cells, and compare the cell counts and types to human milk. Surprisingly, we found that mouse milk cell density is three orders of magnitude higher than human milk. The cell types present in the milk of mice and humans are similar, broadly consisting of mammary epithelial cells and immune cells. These results provide a basis of comparison for mouse and human milk cells and will inform the most appropriate uses of mouse models for the study of human phenomena.
Collapse
Affiliation(s)
- Rose Doerfler
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, United States of America
| | - Saigopalakrishna Yerneni
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, United States of America
| | - Alexandra Newby
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, United States of America
| | - Namit Chaudhary
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, United States of America
| | - Ashley Shu
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, United States of America
| | - Katherine Fein
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, United States of America
| | - Juliana Hofstatter Azambuja
- Department of Pediatrics, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Kathryn A. Whitehead
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, United States of America
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, United States of America
| |
Collapse
|
24
|
Ho YS, Cheng TC, Guo P. Targeted Delivery of Potent Chemical Drugs and RNAi to Drug-Resistant Breast Cancer Using RNA-Nanotechnology and RNA-Ligand Displaying Extracellular vesicles. RNA NANOMED 2024; 1:16-43. [PMID: 40125243 PMCID: PMC11927007 DOI: 10.59566/isrnn.2024.0101016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
This review describes a new technology to treat breast-cancer-drug-resistance by targeting the ABC as the multi-homo-subunit ATPase, enlightening by the Christmas-lighting budge with serial circuit and the asymmetrical homo-hexamer of the phi29 DNA packaging motor with sequential revolving mechanism. Chemotherapeutics has been widely used in breast cancer treatments, but drug resistance has raised a serious concern. RNA therapeutics has emerged as the third milestone in pharmaceutical drug development. RNA nanoparticles are dynamic, mild, and deformative, resulting in spontaneous, rapid, and efficient accumulation in tumor vasculature after IV injection. Their negative charge and favorable size bypass the nonspecific targeting of vital organs and normal cells. This motile and deformable nature also led to the fast passing of glomerular filters and their movement into the urine for rapid body clearance for those non-tumor-accumulated nanoparticles, resulting in undetectable toxicity. Extracellular vesicles have shown potential as a delivery system for RNAi and chemotherapeutic drugs in vivo, contributing to the efficacy of cancer remission. However, the lack of cell-targeting ligands on extracellular vesicles and the nonspecific entry into healthy cells has led to safety concerns. This review addresses how to apply RNA nanotechnology and RNA-ligand displaying extracellular vesicles for specific delivery to breast cancer. The particular focus is on using and combining the RNA and extracellular vesicle technology to deal with breast cancer drug resistance. The targeting capabilities and drug safety can be improved through extracellular vesicle engineering techniques, such as affixing ligands on the extracellular vesicle surface utilizing arrow-tail RNA nanoparticles, ultimately addressing off-target effects and toxicity. Using RNA ligands for specific targeting and the efficient membrane fusion of extracellular vesicles has enabled the development of ligand-displayed extracellular vesicles capable of delivering both RNAi and chemical drugs to cells with precision, effectively inhibiting tumor growth. The negative charge inherent in the vesicles results in electrostatic repulsion, reducing non-specific binding to healthy cells that contain negatively charged lipid membranes. By leveraging the principles of RNA nanotechnology, the engineering of extracellular vesicles offers a promising avenue for addressing breast cancer drug resistance. This review also discusses applying the series of circuit mechanisms in Christmas-decorating-lighting to develop effective therapeutics to combat breast cancer chemoresistance by targeting the ABC drug transporter and breast cancer surface receptors.
Collapse
Affiliation(s)
- Yuan Soon Ho
- Institute of Biochemistry and Molecular Biology, College of Life Sciences, China Medical University, Taichung, Taiwan
| | - Tzu-Chun Cheng
- Institute of Biochemistry and Molecular Biology, College of Life Sciences, China Medical University, Taichung, Taiwan
| | - Peixuan Guo
- Division of Pharmaceutics and Pharmacology, College of Pharmacy; Center for RNA Nanotechnology and Nanomedicine; James Comprehensive Cancer Center, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| |
Collapse
|
25
|
Zhang Y, Luo J, Gui X, Zheng Y, Schaar E, Liu G, Shi J. Bioengineered nanotechnology for nucleic acid delivery. J Control Release 2023; 364:124-141. [PMID: 37879440 PMCID: PMC10838211 DOI: 10.1016/j.jconrel.2023.10.034] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 10/15/2023] [Accepted: 10/21/2023] [Indexed: 10/27/2023]
Abstract
Nucleic acid-based therapy has emerged as a promising therapeutic approach for treating various diseases, such as genetic disorders, cancers, and viral infections. Diverse nucleic acid delivery systems have been reported, and some, including lipid nanoparticles, have exhibited clinical success. In parallel, bioengineered nucleic acid delivery nanocarriers have also gained significant attention due to their flexible functional design and excellent biocompatibility. In this review, we summarize recent advances in bioengineered nucleic acid delivery nanocarriers, focusing on exosomes, cell membrane-derived nanovesicles, protein nanocages, and virus-like particles. We highlight their unique features, advantages for nucleic acid delivery, and biomedical applications. Furthermore, we discuss the challenges that bioengineered nanocarriers face towards clinical translation and the possible avenues for their further development. This review ultimately underlines the potential of bioengineered nanotechnology for the advancement of nucleic acid therapy.
Collapse
Affiliation(s)
- Yang Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China; Center for Nanomedicine and Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Jing Luo
- Department of Urology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Xiran Gui
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Yating Zheng
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Eric Schaar
- Center for Nanomedicine and Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Gang Liu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China.
| | - Jinjun Shi
- Center for Nanomedicine and Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
26
|
Zhang M, Wan L, Li R, Li X, Zhu T, Lu H. Engineered exosomes for tissue regeneration: from biouptake, functionalization and biosafety to applications. Biomater Sci 2023; 11:7247-7267. [PMID: 37794789 DOI: 10.1039/d3bm01169k] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/06/2023]
Abstract
Exosomes are increasingly recognized as important effector molecules that regulate intercellular signaling pathways. Notably, certain types of exosomes can induce therapeutic responses, including cell proliferation, angiogenesis, and tissue repair. The use of exosomes in therapy is a hot spot in current research, especially in regenerative medicine. Despite the therapeutic potential, problems have hindered their success in clinical applications. These shortcomings include low concentration, poor targeting and limited loading capability. To fully realize their therapeutic potential, certain modifications are needed in native exosomes. In the present review, we summarize the exosome modification and functionalization strategies. In addition, we provide an overview of potential clinical applications and highlight the issues associated with the biosafety and biocompatibility of engineered exosomes in applications.
Collapse
Affiliation(s)
- Mu Zhang
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China.
| | - Lei Wan
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China.
| | - Ruiqi Li
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China.
| | - Xiaoling Li
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China.
| | - Taifu Zhu
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China.
| | - Haibin Lu
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China.
- The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, 510900, China
| |
Collapse
|
27
|
Tiwari P, Yadav K, Shukla RP, Gautam S, Marwaha D, Sharma M, Mishra PR. Surface modification strategies in translocating nano-vesicles across different barriers and the role of bio-vesicles in improving anticancer therapy. J Control Release 2023; 363:290-348. [PMID: 37714434 DOI: 10.1016/j.jconrel.2023.09.016] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 09/03/2023] [Accepted: 09/06/2023] [Indexed: 09/17/2023]
Abstract
Nanovesicles and bio-vesicles (BVs) have emerged as promising tools to achieve targeted cancer therapy due to their ability to overcome many of the key challenges currently being faced with conventional chemotherapy. These challenges include the diverse and often complex pathophysiology involving the progression of cancer, as well as the various biological barriers that circumvent therapeutic molecules reaching their target site in optimum concentration. The scientific evidence suggests that surface-functionalized nanovesicles and BVs camouflaged nano-carriers (NCs) both can bypass the established biological barriers and facilitate fourth-generation targeting for the improved regimen of treatment. In this review, we intend to emphasize the role of surface-functionalized nanovesicles and BVs camouflaged NCs through various approaches that lead to an improved internalization to achieve improved and targeted oncotherapy. We have explored various strategies that have been employed to surface-functionalize and biologically modify these vesicles, including the use of biomolecule functionalized target ligands such as peptides, antibodies, and aptamers, as well as the targeting of specific receptors on cancer cells. Further, the utility of BVs, which are made from the membranes of cells such as mesenchymal stem cells (MSCs), white blood cells (WBCs), red blood cells (RBCs), platelets (PLTs) as well as cancer cells also been investigated. Lastly, we have discussed the translational challenges and limitations that these NCs can encounter and still need to be overcome in order to fully realize the potential of nanovesicles and BVs for targeted cancer therapy. The fundamental challenges that currently prevent successful cancer therapy and the necessity of novel delivery systems are in the offing.
Collapse
Affiliation(s)
- Pratiksha Tiwari
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India
| | - Krishna Yadav
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India
| | - Ravi Prakash Shukla
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India
| | - Shalini Gautam
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India
| | - Disha Marwaha
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India
| | - Madhu Sharma
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India
| | - Prabhat Ranjan Mishra
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India; Academy of Scientific and Innovation Research (AcSIR), Ghaziabad 201002, U.P., India.
| |
Collapse
|
28
|
Wang K, Wei Y, Xie X, Li Q, Liu X, Wang L, Li J, Wu J, Fan C. DNA-Programmed Stem Cell Niches via Orthogonal Extracellular Vesicle-Cell Communications. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2302323. [PMID: 37463346 DOI: 10.1002/adma.202302323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 07/13/2023] [Accepted: 07/17/2023] [Indexed: 07/20/2023]
Abstract
Extracellular vesicles (EVs) are natural carriers for intercellular transfer of bioactive molecules, which are harnessed for wide biomedical applications. However, a facile yet general approach to engineering interspecies EV-cell communications is still lacking. Here, the use of DNA to encode the heterogeneous interfaces of EVs and cells in a manner free of covalent or genetic modifications is reported, which enables orthogonal EV-cell interkingdom interactions in complex environments. Cholesterol-modified DNA strands and tetrahedral DNA frameworks are employed with complementary sequences to serve as artificial ligands and receptors docking on EVs and living cells, respectively, which can mediate specific yet efficient cellular internalization of EVs via Watson-Crick base pairing. It is shown that based on this system, human cells can adopt EVs derived from the mouse, watermelon, and Escherichia coli. By implementing several EV-cell circuits, it shows that this DNA-programmed system allows orthogonal EV-cell communications in complex environments. This study further demonstrates efficient delivery of EVs with bioactive contents derived from feeder cells toward monkey female germline stem cells (FGSCs), which enables self-renewal and stemness maintenance of the FGSCs without feeder cells. This system may provide a universal platform to customize intercellular exchanges of materials and signals across species and kingdoms.
Collapse
Affiliation(s)
- Kaizhe Wang
- School of Chemistry and Chemical Engineering, New Cornerstone Science Laboratory, Frontiers Science Center for Transformative Molecules, Zhangjiang Institute for Advanced Study and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Ningbo Cixi Institute of BioMedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315300, China
| | - Yuhan Wei
- School of Chemistry and Chemical Engineering, New Cornerstone Science Laboratory, Frontiers Science Center for Transformative Molecules, Zhangjiang Institute for Advanced Study and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xiaodong Xie
- School of Chemistry and Chemical Engineering, New Cornerstone Science Laboratory, Frontiers Science Center for Transformative Molecules, Zhangjiang Institute for Advanced Study and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Qian Li
- School of Chemistry and Chemical Engineering, New Cornerstone Science Laboratory, Frontiers Science Center for Transformative Molecules, Zhangjiang Institute for Advanced Study and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xiaoguo Liu
- School of Chemistry and Chemical Engineering, New Cornerstone Science Laboratory, Frontiers Science Center for Transformative Molecules, Zhangjiang Institute for Advanced Study and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Lihua Wang
- Institute of Materiobiology, Department of Chemistry, College of Science, Shanghai University, Shanghai, 200444, China
- The Interdisciplinary Research Center, Shanghai Synchrotron Radiation Facility, Zhangjiang Laboratory, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai, 201210, China
| | - Jiang Li
- Institute of Materiobiology, Department of Chemistry, College of Science, Shanghai University, Shanghai, 200444, China
- The Interdisciplinary Research Center, Shanghai Synchrotron Radiation Facility, Zhangjiang Laboratory, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai, 201210, China
| | - Ji Wu
- Key Laboratory for the Genetics of Developmental & Neuropsychiatric Disorders (Ministry of Education), Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Chunhai Fan
- School of Chemistry and Chemical Engineering, New Cornerstone Science Laboratory, Frontiers Science Center for Transformative Molecules, Zhangjiang Institute for Advanced Study and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acids Chemistry and Nanomedicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| |
Collapse
|
29
|
Wang W, Xu Z, Liu M, Cai M, Liu X. Prospective applications of extracellular vesicle-based therapies in regenerative medicine: implications for the use of dental stem cell-derived extracellular vesicles. Front Bioeng Biotechnol 2023; 11:1278124. [PMID: 37936823 PMCID: PMC10627172 DOI: 10.3389/fbioe.2023.1278124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 10/06/2023] [Indexed: 11/09/2023] Open
Abstract
In the 21st century, research on extracellular vesicles (EVs) has made remarkable advancements. Recently, researchers have uncovered the exceptional biological features of EVs, highlighting their prospective use as therapeutic targets, biomarkers, innovative drug delivery systems, and standalone therapeutic agents. Currently, mesenchymal stem cells stand out as the most potent source of EVs for clinical applications in tissue engineering and regenerative medicine. Owing to their accessibility and capability of undergoing numerous differentiation inductions, dental stem cell-derived EVs (DSC-EVs) offer distinct advantages in the field of tissue regeneration. Nonetheless, it is essential to note that unmodified EVs are currently unsuitable for use in the majority of clinical therapeutic scenarios. Considering the high feasibility of engineering EVs, it is imperative to modify these EVs to facilitate the swift translation of theoretical knowledge into clinical practice. The review succinctly presents the known biotherapeutic effects of odontogenic EVs and the underlying mechanisms. Subsequently, the current state of functional cargo loading for engineered EVs is critically discussed. For enhancing EV targeting and in vivo circulation time, the review highlights cutting-edge engineering solutions that may help overcome key obstacles in the clinical application of EV therapeutics. By presenting innovative concepts and strategies, this review aims to pave the way for the adaptation of DSC-EVs in regenerative medicine within clinical settings.
Collapse
Affiliation(s)
- Wenhao Wang
- School of Stomatology, Jinan University, Guangzhou, China
- Center of Stomatology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Zinan Xu
- School of Stomatology, Jinan University, Guangzhou, China
- Center of Stomatology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Minyi Liu
- Center of Stomatology, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Clinical Research Platform for Interdiscipline, Jinan University, Guangzhou, China
| | - Mingxiang Cai
- School of Stomatology, Jinan University, Guangzhou, China
- Center of Stomatology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Xiangning Liu
- School of Stomatology, Jinan University, Guangzhou, China
- Center of Stomatology, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Clinical Research Platform for Interdiscipline, Jinan University, Guangzhou, China
| |
Collapse
|
30
|
Koerselman M, Morshuis LCM, Karperien M. The use of peptides, aptamers, and variable domains of heavy chain only antibodies in tissue engineering and regenerative medicine. Acta Biomater 2023; 170:1-14. [PMID: 37517622 DOI: 10.1016/j.actbio.2023.07.045] [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: 01/27/2023] [Revised: 07/07/2023] [Accepted: 07/25/2023] [Indexed: 08/01/2023]
Abstract
Over the years, much research has been focused on the use of small molecules such as peptides or aptamers or more recently on the use of variable antigen-binding domain of heavy chain only antibodies in the field of tissue engineering and regenerative medicine. The use of these molecules originated as an alternative for the larger conventional antibodies, of which most drawbacks are derived from their size and complex structure. In the field of tissue engineering and regenerative medicine, biological functionalities are often conjugated to biomaterials in order to (re-)create an in vivo like situation, especially when bioinert biomaterials are used. Those biomaterials are functionalized with these functionalities for instance for the purpose of cell attachment or cell targeting for targeted drug delivery but also for local enrichment or blocking of ligands such as growth factors or cytokines on the biomaterial surface. In this review, we further refer to peptides, aptamers, and variable antigen-binding domain of heavy chain only antibodies as biological functionalities. Here, we compare these biological functionalities within the field of tissue engineering and regenerative medicine and give an overview of recent work in which these biological functionalities have been explored. We focus on the previously mentioned purposes of the biological functionalities. We will compare structural differences, possible modifications and (chemical) conjugation strategies. In addition, we will provide an overview of biologicals that are, or have been, involved in clinical trials. Finally, we will highlight the challenges of each of these biologicals. STATEMENT OF SIGNIFICANCE: In the field of tissue engineering there is broad application of functionalized biomaterials for cell attachment, targeted drug delivery and local enrichment or blocking of growth factors. This was previously mostly done via conventional antibodies, but their large size and complex structure impose various challenges with respect of retaining biological functionality. Peptides, aptamers and VHHs may provide an alternative solution for the use of conventional antibodies. This review discusses the use of these molecules for biological functionalization of biomaterials. For each of the molecules, their characteristics, conjugation possibilities and current use in research and clinical trials is described. Furthermore, this review sets out the benefits and challenges of using these types of molecules for different fields of application.
Collapse
Affiliation(s)
- Michelle Koerselman
- Department of Developmental BioEngineering, TechMed Institute, University of Twente, The Netherlands. Drienerlolaan 5, 7522 NB, Enschede, the Netherlands
| | - Lisanne C M Morshuis
- Department of Developmental BioEngineering, TechMed Institute, University of Twente, The Netherlands. Drienerlolaan 5, 7522 NB, Enschede, the Netherlands
| | - Marcel Karperien
- Department of Developmental BioEngineering, TechMed Institute, University of Twente, The Netherlands. Drienerlolaan 5, 7522 NB, Enschede, the Netherlands.
| |
Collapse
|
31
|
Zhang M, Xing J, Zhao S, Chen H, Yin X, Zhu X. Engineered extracellular vesicles in female reproductive disorders. Biomed Pharmacother 2023; 166:115284. [PMID: 37572637 DOI: 10.1016/j.biopha.2023.115284] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 07/28/2023] [Accepted: 08/04/2023] [Indexed: 08/14/2023] Open
Abstract
Biologically active and nanoscale extracellular vesicles (EVs) participate in a variety of cellular physiological and pathological processes in a cell-free manner. Unlike cells, EVs not only do not cause acute immune rejection, but are much smaller and have a low risk of tumorigenicity or embolization. Because of their unique advantages, EVs show promise in applications in the diagnosis and treatment of reproductive disorders. As research broadens, engineering strategies for EVs have been developed, and engineering strategies for EVs have substantially improved their application potential while circumventing the defects of natural EVs, driving EVs toward clinical applications. In this paper, we will review the engineering strategies of EVs, as well as their regulatory effects and mechanisms on reproductive disorders (including premature ovarian insufficiency (POI), polycystic ovarian syndrome (PCOS), recurrent spontaneous abortion (RSA), intrauterine adhesion (IUA), and endometriosis (EMS)) and their application prospects. This work provides new ideas for the treatment of female reproductive disorders by engineering EVs.
Collapse
Affiliation(s)
- Mengxue Zhang
- Reproductive Medicine Center, The Fourth Affiliated Hospital of Jiangsu University, 20 Zhengdong Road, Zhenjiang, Jiangsu 212001, PR China; Institute of Reproductive Sciences, Jiangsu University, 20 Zhengdong Road, Zhenjiang, Jiangsu 212001, PR China; Department of Central Laboratory, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, China
| | - Jie Xing
- Reproductive Medicine Center, The Fourth Affiliated Hospital of Jiangsu University, 20 Zhengdong Road, Zhenjiang, Jiangsu 212001, PR China; Institute of Reproductive Sciences, Jiangsu University, 20 Zhengdong Road, Zhenjiang, Jiangsu 212001, PR China; Department of Central Laboratory, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, China
| | - Shijie Zhao
- Reproductive Medicine Center, The Fourth Affiliated Hospital of Jiangsu University, 20 Zhengdong Road, Zhenjiang, Jiangsu 212001, PR China; Institute of Reproductive Sciences, Jiangsu University, 20 Zhengdong Road, Zhenjiang, Jiangsu 212001, PR China; Department of Central Laboratory, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, China
| | - Hui Chen
- Department of Central Laboratory, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, China
| | - Xinming Yin
- Department of Central Laboratory, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, China
| | - Xiaolan Zhu
- Reproductive Medicine Center, The Fourth Affiliated Hospital of Jiangsu University, 20 Zhengdong Road, Zhenjiang, Jiangsu 212001, PR China; Institute of Reproductive Sciences, Jiangsu University, 20 Zhengdong Road, Zhenjiang, Jiangsu 212001, PR China.
| |
Collapse
|
32
|
Roerig J, Schulz-Siegmund M. Standardization Approaches for Extracellular Vesicle Loading with Oligonucleotides and Biologics. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2301763. [PMID: 37287374 DOI: 10.1002/smll.202301763] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 05/13/2023] [Indexed: 06/09/2023]
Abstract
Extracellular vesicles (EVs) are widely recognized for their potential as drug delivery systems. EVs are membranous nanoparticles shed from cells. Among their natural features are their ability to shield cargo molecules against degradation and enable their functional internalization into target cells. Especially biological or bio-inspired large molecules (LMs), like nucleic acids, proteins, peptides, and others, may profit from encapsulation in EVs for drug delivery purposes. In the last years, a variety of loading protocols are explored for different LMs. The lack of standardization in the EV drug delivery field has impeded their comparability so far. Currently, the first reporting frameworks and workflows for EV drug loading are proposed. The aim of this review is to summarize these evolving standardization approaches and set recently developed methods into context. This will allow for enhanced comparability of future work on EV drug loading with LMs.
Collapse
Affiliation(s)
- Josepha Roerig
- Pharmaceutical Technology, Institute of Pharmacy, Medical Faculty, Leipzig University, 04317, Leipzig, Germany
| | - Michaela Schulz-Siegmund
- Pharmaceutical Technology, Institute of Pharmacy, Medical Faculty, Leipzig University, 04317, Leipzig, Germany
| |
Collapse
|
33
|
Gubu A, Zhang X, Lu A, Zhang B, Ma Y, Zhang G. Nucleic acid amphiphiles: Synthesis, properties, and applications. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 33:144-163. [PMID: 37456777 PMCID: PMC10345231 DOI: 10.1016/j.omtn.2023.05.022] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/18/2023]
Abstract
Nucleic acid amphiphiles, referring to nucleic acids modified with large hydrophobic groups, have been widely used in programmable bioengineering. Since nucleic acids are intrinsically hydrophilic, the hydrophobic groups endow nucleic acid amphiphiles with unique properties, such as self-assembling, interactions with artificial or biological membranes, and transmembrane transport. Importantly, the hybridization or target binding capability of oligonucleotide itself supplies nucleic acid amphiphiles with excellent programmability. As a result, this type of molecule has attracted considerable attention in academic studies and has enormous potential for further applications. For a comprehensive understanding of nucleic acid amphiphiles, we review the reported research on nucleic acid amphiphiles from their molecular design to final applications, in which we summarize the synthetic strategies for nucleic acid amphiphiles and draw much attention to their unique properties in different contexts. Finally, a summary of the applications of nucleic acid amphiphiles in drug development, bioengineering, and bioanalysis are critically discussed.
Collapse
Affiliation(s)
- Amu Gubu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Aptacure Therapeutics Limited, Kowloon, Hong Kong SAR, China
| | - Xueli Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences and Chemical Biology Center, Peking University, No. 38 Xueyuan Road, Beijing, China
| | - Aiping Lu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tsai, Hong Kong 999077, China
- Institute of Precision Medicine and Innovative Drug Discovery, HKBU Institute for Research and Continuing Education, Shenzhen 518000, China
| | - Baoting Zhang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yuan Ma
- Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tsai, Hong Kong 999077, China
- Institute of Precision Medicine and Innovative Drug Discovery, HKBU Institute for Research and Continuing Education, Shenzhen 518000, China
| | - Ge Zhang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tsai, Hong Kong 999077, China
- Institute of Precision Medicine and Innovative Drug Discovery, HKBU Institute for Research and Continuing Education, Shenzhen 518000, China
| |
Collapse
|
34
|
Sareen N, Srivastava A, Alagarsamy KN, Lionetti V, Dhingra S. Stem cells derived exosomes and biomaterials to modulate autophagy and mend broken hearts. Biochim Biophys Acta Mol Basis Dis 2023:166806. [PMID: 37437748 DOI: 10.1016/j.bbadis.2023.166806] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/29/2023] [Accepted: 07/09/2023] [Indexed: 07/14/2023]
Abstract
Autophagy maintains cellular homeostasis and plays a crucial role in managing pathological conditions including ischemic myocardial injury leading to heart failure (HF). Despite treatments, no intervention can replace lost cardiomyocytes. Stem cell therapy offers potential for post-myocardial infarction repair but struggles with poor cell retention due to immune rejection. In the search for effective therapies, stem cell-derived extracellular vesicles (EVs), especially exosomes, have emerged as promising tools. These tiny bioactive molecule carriers play vital roles in intercellular communication and tissue engineering. They offer numerous therapeutic benefits including modulating immune responses, promoting tissue repair, and boosting angiogenesis. Additionally, biomaterials provide a conducive 3D microenvironment for cell, exosome, and biomolecule delivery, and enhance heart muscle strength, making it a comprehensive cardiac repair strategy. In this regard, the current review delves into the intricate application of extracellular vesicles (EVs) and biomaterials for managing autophagy in the heart muscle during cardiac injury. Central to our investigation is the exploration of how these elements interact within the context of cardiac repair and regeneration. Additionally, this review also casts light on the formidable challenges that plague this field, such as the issues of safety, efficacy, controlled delivery, and acceptance of these therapeutic strategies for effective clinical translation. Addressing these challenges is crucial for unlocking the full therapeutic potential of EV and biomaterial-based therapies and ensuring their successful translation from bench to bedside.
Collapse
Affiliation(s)
- Niketa Sareen
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, Rady Faculty of Health Science, University of Manitoba, Winnipeg R2H2A6, MB, Canada; Unit of Translational Critical Care Medicine, Institute of Life Sciences, Scuola Superiore Sant'Anna, 56124 Pisa, Italy
| | - Abhay Srivastava
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, Rady Faculty of Health Science, University of Manitoba, Winnipeg R2H2A6, MB, Canada
| | - Keshav Narayan Alagarsamy
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, Rady Faculty of Health Science, University of Manitoba, Winnipeg R2H2A6, MB, Canada
| | - Vincenzo Lionetti
- Unit of Translational Critical Care Medicine, Institute of Life Sciences, Scuola Superiore Sant'Anna, 56124 Pisa, Italy
| | - Sanjiv Dhingra
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, Rady Faculty of Health Science, University of Manitoba, Winnipeg R2H2A6, MB, Canada.
| |
Collapse
|
35
|
Wei J, Wang Z, Han T, Chen J, Ou Y, Wei L, Zhu X, Wang K, Yan Z, Han YP, Zheng X. Extracellular vesicle-mediated intercellular and interorgan crosstalk of pancreatic islet in health and diabetes. Front Endocrinol (Lausanne) 2023; 14:1170237. [PMID: 37305058 PMCID: PMC10248434 DOI: 10.3389/fendo.2023.1170237] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 05/10/2023] [Indexed: 06/13/2023] Open
Abstract
Diabetes mellitus (DM) is a systemic metabolic disease with high mortality and morbidity. Extracellular vesicles (EVs) have emerged as a novel class of signaling molecules, biomarkers and therapeutic agents. EVs-mediated intercellular and interorgan crosstalk of pancreatic islets plays a crucial role in the regulation of insulin secretion of β-cells and insulin action in peripheral insulin target tissues, maintaining glucose homeostasis under physiological conditions, and it's also involved in pathological changes including autoimmune response, insulin resistance and β-cell failure associated with DM. In addition, EVs may serve as biomarkers and therapeutic agents that respectively reflect the status and improve function and viability of pancreatic islets. In this review, we provide an overview of EVs, discuss EVs-mediated intercellular and interorgan crosstalk of pancreatic islet under physiological and diabetic conditions, and summarize the emerging applications of EVs in the diagnosis and treatment of DM. A better understanding of EVs-mediated intercellular and interorgan communication of pancreatic islets will broaden and enrich our knowledge of physiological homeostasis maintenance as well as the development, diagnosis and treatment of DM.
Collapse
Affiliation(s)
- Junlun Wei
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu, China
| | - Zhenghao Wang
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu, China
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institute, Stockholm, Sweden
| | - Tingrui Han
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu, China
| | - Jiaoting Chen
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu, China
| | - Yiran Ou
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu, China
| | - Lan Wei
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu, China
| | - Xinyue Zhu
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu, China
| | - Ke Wang
- Department of Vascular Surgery, University Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhe Yan
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu, China
| | - Yuan-Ping Han
- The Center for Growth, Metabolism and Aging, The College of Life Sciences, Sichuan University, Chengdu, China
| | - Xiaofeng Zheng
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
36
|
Jiao K, Liu C, Basu S, Raveendran N, Nakano T, Ivanovski S, Han P. Bioprinting extracellular vesicles as a "cell-free" regenerative medicine approach. EXTRACELLULAR VESICLES AND CIRCULATING NUCLEIC ACIDS 2023; 4:218-239. [PMID: 39697984 PMCID: PMC11648406 DOI: 10.20517/evcna.2023.19] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 05/08/2023] [Accepted: 05/16/2023] [Indexed: 12/20/2024]
Abstract
Regenerative medicine involves the restoration of tissue or organ function via the regeneration of these structures. As promising regenerative medicine approaches, either extracellular vesicles (EVs) or bioprinting are emerging stars to regenerate various tissues and organs (i.e., bone and cardiac tissues). Emerging as highly attractive cell-free, off-the-shelf nanotherapeutic agents for tissue regeneration, EVs are bilayered lipid membrane particles that are secreted by all living cells and play a critical role as cell-to-cell communicators through an exchange of EV cargos of protein, genetic materials, and other biological components. 3D bioprinting, combining 3D printing and biology, is a state-of-the-art additive manufacturing technology that uses computer-aided processes to enable simultaneous patterning of 3D cells and tissue constructs in bioinks. Although developing an effective system for targeted EVs delivery remains challenging, 3D bioprinting may offer a promising means to improve EVs delivery efficiency with controlled loading and release. The potential application of 3D bioprinted EVs to regenerate tissues has attracted attention over the past few years. As such, it is timely to explore the potential and associated challenges of utilizing 3D bioprinted EVs as a novel "cell-free" alternative regenerative medicine approach. In this review, we describe the biogenesis and composition of EVs, and the challenge of isolating and characterizing small EVs - sEVs (< 200 nm). Common 3D bioprinting techniques are outlined and the issue of bioink printability is explored. After applying the following search strategy in PubMed: "bioprinted exosomes" or "3D bioprinted extracellular vesicles", eight studies utilizing bioprinted EVs were found that have been included in this scoping review. Current studies utilizing bioprinted sEVs for various in vitro and in vivo tissue regeneration applications, including angiogenesis, osteogenesis, immunomodulation, chondrogenesis and myogenesis, are discussed. Finally, we explore the current challenges and provide an outlook on possible refinements for bioprinted sEVs applications.
Collapse
Affiliation(s)
- Kexin Jiao
- The University of Queensland, Faculty of Health and Behavioural Sciences, School of Dentistry, Center for Oral-facial Regeneration, Rehabilitation and Reconstruction (COR3), Epigenetics nanodiagnostic and therapeutic group, Brisbane 4006, QLD, Australia
- The University of Queensland, Faculty of Health and Behavioural Sciences, School of Dentistry, Brisbane 4006, QLD, Australia
| | - Chun Liu
- The University of Queensland, Faculty of Health and Behavioural Sciences, School of Dentistry, Center for Oral-facial Regeneration, Rehabilitation and Reconstruction (COR3), Epigenetics nanodiagnostic and therapeutic group, Brisbane 4006, QLD, Australia
| | - Saraswat Basu
- The University of Queensland, Faculty of Health and Behavioural Sciences, School of Dentistry, Center for Oral-facial Regeneration, Rehabilitation and Reconstruction (COR3), Epigenetics nanodiagnostic and therapeutic group, Brisbane 4006, QLD, Australia
| | - Nimal Raveendran
- The University of Queensland, Faculty of Health and Behavioural Sciences, School of Dentistry, Center for Oral-facial Regeneration, Rehabilitation and Reconstruction (COR3), Epigenetics nanodiagnostic and therapeutic group, Brisbane 4006, QLD, Australia
- The University of Queensland, Faculty of Health and Behavioural Sciences, School of Dentistry, Brisbane 4006, QLD, Australia
| | - Tamaki Nakano
- Hokkaido University, Institute for Catalysis (ICAT), N21 W10, Kita-ku, Sapporo 001-0021, Japan
| | - Sašo Ivanovski
- The University of Queensland, Faculty of Health and Behavioural Sciences, School of Dentistry, Center for Oral-facial Regeneration, Rehabilitation and Reconstruction (COR3), Epigenetics nanodiagnostic and therapeutic group, Brisbane 4006, QLD, Australia
- The University of Queensland, Faculty of Health and Behavioural Sciences, School of Dentistry, Brisbane 4006, QLD, Australia
| | - Pingping Han
- The University of Queensland, Faculty of Health and Behavioural Sciences, School of Dentistry, Center for Oral-facial Regeneration, Rehabilitation and Reconstruction (COR3), Epigenetics nanodiagnostic and therapeutic group, Brisbane 4006, QLD, Australia
- The University of Queensland, Faculty of Health and Behavioural Sciences, School of Dentistry, Brisbane 4006, QLD, Australia
| |
Collapse
|
37
|
Uddin N, Binzel DW, Shu D, Fu TM, Guo P. Targeted delivery of RNAi to cancer cells using RNA-ligand displaying exosome. Acta Pharm Sin B 2023; 13:1383-1399. [PMID: 37139430 PMCID: PMC10149909 DOI: 10.1016/j.apsb.2022.11.019] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 09/27/2022] [Accepted: 10/13/2022] [Indexed: 11/18/2022] Open
Abstract
Exosome is an excellent vesicle for in vivo delivery of therapeutics, including RNAi and chemical drugs. The extremely high efficiency in cancer regression can partly be attributed to its fusion mechanism in delivering therapeutics to cytosol without endosome trapping. However, being composed of a lipid-bilayer membrane without specific recognition capacity for aimed-cells, the entry into nonspecific cells can lead to potential side-effects and toxicity. Applying engineering approaches for targeting-capacity to deliver therapeutics to specific cells is desirable. Techniques with chemical modification in vitro and genetic engineering in cells have been reported to decorate exosomes with targeting ligands. RNA nanoparticles have been used to harbor tumor-specific ligands displayed on exosome surface. The negative charge reduces nonspecific binding to vital cells with negatively charged lipid-membrane due to the electrostatic repulsion, thus lowering the side-effect and toxicity. In this review, we focus on the uniqueness of RNA nanoparticles for exosome surface display of chemical ligands, small peptides or RNA aptamers, for specific cancer targeting to deliver anticancer therapeutics, highlighting recent advances in targeted delivery of siRNA and miRNA that overcomes the previous RNAi delivery roadblocks. Proper understanding of exosome engineering with RNA nanotechnology promises efficient therapies for a wide range of cancer subtypes.
Collapse
Affiliation(s)
- Nasir Uddin
- Center for RNA Nanobiotechnology and Nanomedicine, Division of Pharmaceutics and Pharmacology, College of Pharmacy, the Ohio State University, Columbus, OH 43210, USA
- Dorothy M. Davis Heart and Lung Research Institute, the Ohio State University, Columbus, OH 43210, USA
- James Comprehensive Cancer Center, College of Medicine, the Ohio State University, Columbus, OH 43210, USA
| | - Daniel W. Binzel
- Center for RNA Nanobiotechnology and Nanomedicine, Division of Pharmaceutics and Pharmacology, College of Pharmacy, the Ohio State University, Columbus, OH 43210, USA
- Dorothy M. Davis Heart and Lung Research Institute, the Ohio State University, Columbus, OH 43210, USA
- James Comprehensive Cancer Center, College of Medicine, the Ohio State University, Columbus, OH 43210, USA
| | - Dan Shu
- Center for RNA Nanobiotechnology and Nanomedicine, Division of Pharmaceutics and Pharmacology, College of Pharmacy, the Ohio State University, Columbus, OH 43210, USA
- Dorothy M. Davis Heart and Lung Research Institute, the Ohio State University, Columbus, OH 43210, USA
- James Comprehensive Cancer Center, College of Medicine, the Ohio State University, Columbus, OH 43210, USA
| | - Tian-Min Fu
- Department of Biological Chemistry & Pharmacology, College of Medicine, the Ohio State University, Columbus, OH 43210, USA
| | - Peixuan Guo
- Center for RNA Nanobiotechnology and Nanomedicine, Division of Pharmaceutics and Pharmacology, College of Pharmacy, the Ohio State University, Columbus, OH 43210, USA
- Dorothy M. Davis Heart and Lung Research Institute, the Ohio State University, Columbus, OH 43210, USA
- James Comprehensive Cancer Center, College of Medicine, the Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
38
|
Melamed JR, Yerneni SS, Arral ML, LoPresti ST, Chaudhary N, Sehrawat A, Muramatsu H, Alameh MG, Pardi N, Weissman D, Gittes GK, Whitehead KA. Ionizable lipid nanoparticles deliver mRNA to pancreatic β cells via macrophage-mediated gene transfer. SCIENCE ADVANCES 2023; 9:eade1444. [PMID: 36706177 PMCID: PMC9882987 DOI: 10.1126/sciadv.ade1444] [Citation(s) in RCA: 84] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 12/27/2022] [Indexed: 05/19/2023]
Abstract
Systemic messenger RNA (mRNA) delivery to organs outside the liver, spleen, and lungs remains challenging. To overcome this issue, we hypothesized that altering nanoparticle chemistry and administration routes may enable mRNA-induced protein expression outside of the reticuloendothelial system. Here, we describe a strategy for delivering mRNA potently and specifically to the pancreas using lipid nanoparticles. Our results show that delivering lipid nanoparticles containing cationic helper lipids by intraperitoneal administration produces robust and specific protein expression in the pancreas. Most resultant protein expression occurred within insulin-producing β cells. Last, we found that pancreatic mRNA delivery was dependent on horizontal gene transfer by peritoneal macrophage exosome secretion, an underappreciated mechanism that influences the delivery of mRNA lipid nanoparticles. We anticipate that this strategy will enable gene therapies for intractable pancreatic diseases such as diabetes and cancer.
Collapse
Affiliation(s)
- Jilian R. Melamed
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - Mariah L. Arral
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Samuel T. LoPresti
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Namit Chaudhary
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Anuradha Sehrawat
- Department of Pediatric Surgery, Department of Surgery, Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224, USA
| | - Hiromi Muramatsu
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - Norbert Pardi
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Drew Weissman
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - George K. Gittes
- Department of Pediatric Surgery, Department of Surgery, Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224, USA
| | - Kathryn A. Whitehead
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| |
Collapse
|
39
|
Fan Y, Wu W, Xie N, Huang Y, Wu H, Zhang J, Guo X, Ding S, Guo B. Biocompatible engineered erythrocytes as plasmonic sensor initiators for high-sensitive screening of non-small cell lung cancer-derived exosomal miRNA in an integrated system. Biosens Bioelectron 2023; 219:114802. [PMID: 36270080 DOI: 10.1016/j.bios.2022.114802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 08/20/2022] [Accepted: 10/09/2022] [Indexed: 11/06/2022]
Abstract
The development of a holistic solution is crucial for exosome-based liquid biopsy, which is a significant advancement from basic research to clinical application for the early and non-invasive diagnosis of disease. However, the challenge of current technology is how to facilitate the separation and quickly connect with the detection. Herein, employing miRNA-155 as a target, a novel integrated concentration and determination system of exosomes (ICDSE) was fabricated for the targeted enrichment of exosomes from the plasma of patients with non-small cell lung cancer and instant downstream analysis of exosomal miRNA. This ICDSE consists of aptamer-engineered erythrocytes with a supramolecular plasmonic biosensor. The streamlined analytical procedure benefited from engineered erythrocytes as an interlinkage, needlessly removing them before extracting total RNA due to the lack of nuclei. With the assistance of a specific aptamer and simple centrifugation, engineered erythrocytes achieved exceptional enrichment of exosomes within 30 min. The LOD of the biosensor for miR-155 approached 2.03 fM due to the substantially high refractive index of the quadruplet supramolecular dendrimer and zirconium metal-organic framework. Impressively, the developed ICDSE successfully isolated and evaluated exosomes from clinical specimens of patients with NSCLC, which can also be promising for other diseases with identifiable exosome signatures. Thus, our ICDSE is expected to have widespread biomedical applications, as it is economical and well-accepted.
Collapse
Affiliation(s)
- Yunpeng Fan
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China; Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, 400021, China
| | - Wenwen Wu
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Ning Xie
- Department of Laboratory Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, China; Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China; Department of Medical Laboratory Science, Research Center of Translational Medicine, North Sichuan Medical College, Nanchong, Sichuan, 637000, China
| | - Yi Huang
- Department of Laboratory Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, China; Department of Medical Laboratory Science, Research Center of Translational Medicine, North Sichuan Medical College, Nanchong, Sichuan, 637000, China
| | - Haiping Wu
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Juan Zhang
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, 400021, China
| | - Xiaolan Guo
- Department of Laboratory Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, China; Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China; Department of Medical Laboratory Science, Research Center of Translational Medicine, North Sichuan Medical College, Nanchong, Sichuan, 637000, China
| | - Shijia Ding
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China.
| | - Bin Guo
- Department of Laboratory Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, China; Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China; Department of Medical Laboratory Science, Research Center of Translational Medicine, North Sichuan Medical College, Nanchong, Sichuan, 637000, China.
| |
Collapse
|
40
|
Wu M, Wang M, Jia H, Wu P. Extracellular vesicles: emerging anti-cancer drugs and advanced functionalization platforms for cancer therapy. Drug Deliv 2022; 29:2513-2538. [PMID: 35915054 PMCID: PMC9347476 DOI: 10.1080/10717544.2022.2104404] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Increasing evidences show that unmodified extracellular vesicles (EVs) derived from various cells can effectively inhibit the malignant progression of different types of tumors by delivering the bioactive molecules. Therefore, EVs are expected to be developed as emerging anticancer drugs. Meanwhile, unmodified EVs as an advanced and promising nanocarrier that is frequently used in targeted delivery therapeutic cargos and personalized reagents for the treatment and diagnosis of cancer. To improve the efficacy of EV-based treatments, researchers are trying to engineering EVs as an emerging nanomedicine translational therapy platform through biological, physical and chemical approaches, which can be broaden and altered to enhance their therapeutic capability. EVs loaded with therapeutic components such as tumor suppressor drugs, siRNAs, proteins, peptides, and conjugates exhibit significantly enhanced anti-tumor effects. Moreover, the design and preparation of tumor-targeted modified EVs greatly enhance the specificity and effectiveness of tumor therapy, and these strategies are expected to become novel ideas for tumor precision medicine. This review will focus on reviewing the latest research progress of functionalized EVs, clarifying the superior biological functions and powerful therapeutic potential of EVs, for researchers to explore new design concepts based on EVs and build next-generation nanomedicine therapeutic platforms.
Collapse
Affiliation(s)
- Manling Wu
- Department of Clinical Laboratory, The First Affiliated Hospital of UST C, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, P.R. China
- Anhui Provincial Children’s Hospital, Hefei, Anhui, P.R. China
| | - Min Wang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, P.R. China
| | - Haoyuan Jia
- Department of Clinical Laboratory, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, Jiangsu, P.R. China
| | - Peipei Wu
- Department of Clinical Laboratory, The First Affiliated Hospital of UST C, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, P.R. China
- Anhui Provincial Children’s Hospital, Hefei, Anhui, P.R. China
| |
Collapse
|
41
|
Ma S, Zhang Y, Li S, Li A, Li Y, Pei D. Engineering exosomes for bone defect repair. Front Bioeng Biotechnol 2022; 10:1091360. [PMID: 36568296 PMCID: PMC9768454 DOI: 10.3389/fbioe.2022.1091360] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 11/28/2022] [Indexed: 12/12/2022] Open
Abstract
Currently, bone defect repair is still an intractable clinical problem. Numerous treatments have been performed, but their clinical results are unsatisfactory. As a key element of cell-free therapy, exosome is becoming a promising tool of bone regeneration in recent decades, because of its promoting osteogenesis and osteogenic differentiation function in vivo and in vitro. However, low yield, weak activity, inefficient targeting ability, and unpredictable side effects of natural exosomes have limited the clinical application. To overcome the weakness, various approaches have been applied to produce engineering exosomes by regulating their production and function at present. In this review, we will focus on the engineering exosomes for bone defect repair. By summarizing the exosomal cargos affecting osteogenesis, the strategies of engineering exosomes and properties of exosome-integrated biomaterials, this work will provide novel insights into exploring advanced engineering exosome-based cell-free therapy for bone defect repair.
Collapse
Affiliation(s)
| | | | | | | | - Ye Li
- *Correspondence: Ye Li, ; Dandan Pei,
| | | |
Collapse
|
42
|
Feng L, Guo L, Tanaka Y, Su L. Tumor-Derived Small Extracellular Vesicles Involved in Breast Cancer Progression and Drug Resistance. Int J Mol Sci 2022; 23:ijms232315236. [PMID: 36499561 PMCID: PMC9736664 DOI: 10.3390/ijms232315236] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/28/2022] [Accepted: 12/01/2022] [Indexed: 12/09/2022] Open
Abstract
Breast cancer is one of the most serious and terrifying threats to the health of women. Recent studies have demonstrated that interaction among cancer cells themselves and those with other cells, including immune cells, in a tumor microenvironment potentially and intrinsically regulate and determine cancer progression and metastasis. Small extracellular vesicles (sEVs), a type of lipid-bilayer particles derived from cells, with a size of less than 200 nm, are recognized as one form of important mediators in cell-to-cell communication. sEVs can transport a variety of bioactive substances, including proteins, RNAs, and lipids. Accumulating evidence has revealed that sEVs play a crucial role in cancer development and progression, with a significant impact on proliferation, invasion, and metastasis. In addition, sEVs systematically coordinate physiological and pathological processes, such as coagulation, vascular leakage, and stromal cell reprogramming, to bring about premetastatic niche formation and to determine metastatic organ tropism. There are a variety of oncogenic factors in tumor-derived sEVs that mediate cellular communication between local stromal cells and distal microenvironment, both of which are important in cancer progression and metastasis. Tumor-derived sEVs contain substances that are similar to parental tumor cells, and as such, sEVs could be biomarkers in cancer progression and potential therapeutic targets, particularly for predicting and preventing future metastatic development. Here, we review the mechanisms underlying the regulation by tumor-derived sEVs on cancer development and progression, including proliferation, metastasis, drug resistance, and immunosuppression, which coordinately shape the pro-metastatic microenvironment. In addition, we describe the application of sEVs to the development of cancer biomarkers and potential therapeutic modalities and discuss how they can be engineered and translated into clinical practice.
Collapse
Affiliation(s)
- Lingyun Feng
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Lijuan Guo
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Yoshimasa Tanaka
- Center for Medical Innovation, Nagasaki University, 1-7-1, Sakamoto, Nagasaki 852-8588, Japan
- Correspondence: (Y.T.); (L.S.); Tel.: +81-95-819-7063 (Y.T.); +86-27-8779-2024 (L.S.); Fax: +81-95-819-2189 (Y.T.); +86-27-8779-2072 (L.S.)
| | - Li Su
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Correspondence: (Y.T.); (L.S.); Tel.: +81-95-819-7063 (Y.T.); +86-27-8779-2024 (L.S.); Fax: +81-95-819-2189 (Y.T.); +86-27-8779-2072 (L.S.)
| |
Collapse
|
43
|
Huang Y, Zhong L, Li X, Wu P, He J, Tang C, Tang Z, Su J, Feng Z, Wang B, Ma Y, Peng H, Bai Z, Zhong Y, Liang Y, Lu W, Luo R, Li J, Li H, Deng Z, Lan X, Liu Z, Zhang K, Zhao Y. In Situ Silver-Based Electrochemical Oncolytic Bioreactor. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2109973. [PMID: 35998517 DOI: 10.1002/adma.202109973] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 07/13/2022] [Indexed: 06/15/2023]
Abstract
In this study, it is shown for the first time that a reduced graphene oxide (rGO) carrier has a 20-fold higher catalysis rate than graphene oxide in Ag+ reduction. Based on this, a tumor microenvironment-enabled in situ silver-based electrochemical oncolytic bioreactor (SEOB) which switched Ag+ prodrugs into in situ therapeutic silver nanoparticles with and above 95% transition rate is constructed to inhibit the growths of various tumors. In this SEOB-enabled intratumoral nanosynthetic medicine, intratumoral H2 O2 and rGO act as the reductant and the catalyst, respectively. Chelation of aptamers to the SEOB-unlocked prodrugs increases the production of silver nanoparticles in tumor cells, especially in the presence of Vitamin C, which is broken down in tumor cells to supply massive amounts of H2 O2 . Consequently, apoptosis and pyroptosis are induced to cooperatively contribute to the considerably-elevated anti-tumor effects on subcutaneous HepG2 and A549 tumors and orthotopic implanted HepG2 tumors in livers of nude mice. The specific aptamer targeting and intratumoral silver nanoparticle production guarantee excellent biosafety since it fails to elicit tissue damages in monkeys, which greatly increases the clinical translation potential of the SEOB system.
Collapse
Affiliation(s)
- Yong Huang
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, 530021, China
| | - Liping Zhong
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, 530021, China
| | - Xiaotong Li
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, 530021, China
| | - Pan Wu
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, 530021, China
| | - Jian He
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, 530021, China
| | - Chao Tang
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, 530021, China
| | - Zhiping Tang
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, 530021, China
| | - Jing Su
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, 530021, China
| | - Zhenbo Feng
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, 530021, China
| | - Bing Wang
- Department of Spine Surgery, The Second Xiangya Hospital of Central South University, Changsha, 410011, China
| | - Yun Ma
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, 530021, China
| | - Hongmei Peng
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, 530021, China
| | - Zhihao Bai
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, 530021, China
| | - Yi Zhong
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, 530021, China
| | - Ying Liang
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, 530021, China
| | - Wenxi Lu
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, 530021, China
| | - Ruiyu Luo
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, 530021, China
| | - Jinghua Li
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, 530021, China
| | - Haiping Li
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, 530021, China
| | - Zhiming Deng
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, 530021, China
| | - Xianli Lan
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, 530021, China
| | - Ziqun Liu
- Department of Spine Surgery, The Second Xiangya Hospital of Central South University, Changsha, 410011, China
| | - Kun Zhang
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, 530021, China
| | - Yongxiang Zhao
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, 530021, China
| |
Collapse
|
44
|
Song H, Chen X, Hao Y, Wang J, Xie Q, Wang X. Nanoengineering facilitating the target mission: targeted extracellular vesicles delivery systems design. J Nanobiotechnology 2022; 20:431. [PMID: 36175866 PMCID: PMC9524104 DOI: 10.1186/s12951-022-01638-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 09/04/2022] [Indexed: 11/10/2022] Open
Abstract
Precision medicine has put forward the proposition of "precision targeting" for modern drug delivery systems. Inspired by techniques from biology, pharmaceutical sciences, and nanoengineering, numerous targeted drug delivery systems have been developed in recent decades. But the large-scale applications of these systems are limited due to unsatisfactory targeting efficiency, cytotoxicity, easy removability, and instability. As such, the natural endogenous cargo delivery vehicle-extracellular vesicles (EVs)-have sparked significant interest for its unique inherent targeting properties, biocompatibility, transmembrane ability, and circulatory stability. The membranes of EVs are enriched for receptors or ligands that interact with target cells, which endows them with inherent targeting mission. However, most of the natural therapeutic EVs face the fate of being cleared by macrophages, resulting in off-target. Therefore, the specificity of natural EVs delivery systems urgently needs to be further improved. In this review, we comprehensively summarize the inherent homing mechanisms of EVs and the effects of the donor cell source and administration route on targeting specificity. We then go over nanoengineering techniques that modify EVs for improving specific targeting, such as source cell alteration and modification of EVs surface. We also highlight the auxiliary strategies to enhance specificity by changing the external environment, such as magnetic and photothermal. Furthermore, contemporary issues such as the lack of a gold standard for assessing targeting efficiency are discussed. This review will provide new insights into the development of precision medicine delivery systems.
Collapse
Affiliation(s)
- Haoyue Song
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, China.,Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, China
| | - Xiaohang Chen
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, China.,Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, China
| | - Yujia Hao
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, China.,Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, China
| | - Jia Wang
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, China.,Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, China
| | - Qingpeng Xie
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, China.,Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, China
| | - Xing Wang
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, China. .,Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, China.
| |
Collapse
|
45
|
Engineered extracellular vesicles: Regulating the crosstalk between the skeleton and immune system. ENGINEERED REGENERATION 2022. [DOI: 10.1016/j.engreg.2022.06.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
|
46
|
Extracellular vesicles as an emerging drug delivery system for cancer treatment: Current strategies and recent advances. Biomed Pharmacother 2022; 153:113480. [DOI: 10.1016/j.biopha.2022.113480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 07/22/2022] [Accepted: 07/26/2022] [Indexed: 11/19/2022] Open
|
47
|
Yerneni SS, Yalcintas EP, Smith JD, Averick S, Campbell PG, Ozdoganlar OB. Skin-targeted delivery of extracellular vesicle-encapsulated curcumin using dissolvable microneedle arrays. Acta Biomater 2022; 149:198-212. [PMID: 35809788 DOI: 10.1016/j.actbio.2022.06.046] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 06/29/2022] [Accepted: 06/30/2022] [Indexed: 12/14/2022]
Abstract
Therapeutic benefits of curcumin for inflammatory diseases have been demonstrated. However, curcumin's potential as a clinical therapeutic has been hindered due to its low solubility and stability in vivo. We hypothesized that a hybrid curcumin carrier that incorporates albumin-binding and extracellular vesicle (EV) encapsulation could effectively address the current challenges of curcumin delivery. We further postulated that using dissolvable microneedle arrays (dMNAs) for local delivery of curcumin-albumin-EVs (CA-EVs) could effectively control skin inflammation in vivo. Mild sonication was used to encapsulate curcumin and albumin into EVs, and the resulting CA-EVs were integrated into tip-loaded dMNAs. In vitro and in vivo studies were performed to assess the stability, cellular uptake, and anti-inflammatory bioactivity of dMNA-delivered CA-EVs. Curcumin in CA-EVs exhibited at least five-fold higher stability in vitro than naïve curcumin or curcumin-EVs without albumin. Incorporating CA-EVs into dMNAs did not alter their cellular uptake or anti-inflammatory bioactivity. The dMNA embedded CA-EVs retained their bioactivity when stored at room temperature for at least 12 months. In rat and mice models, dMNA delivered CA-EVs suppressed and significantly reduced lipopolysaccharide and Imiquimod-triggered inflammation. We conclude that dMNA delivery of CA-EVs has the potential to become an effective local-delivery strategy for inflammatory skin diseases. STATEMENT OF SIGNIFICANCE: We introduce and evaluate a skin-targeted delivery system for curcumin that synergistically combines albumin association, extracellular-vesicle encapsulation, and dissolvable microneedle arrays (dMNAs) . In vitro, curcumin-albumin encapsulated extracellular vesicles (CA-EVs) inhibit and reverse the LPS-triggered expression of inflammatory transcription factor NF-κB. The integration of CA-EVs into dMNAs does not affect them physically or functionally. Importantly, dMNAs extend EV storage stability for at least 12 months at room temperature with minimal loss in their bioactivity. We demonstrate that dMNA delivered CA-EVs effectively block and reverse skin inflammation in vivo in mouse and rat models.
Collapse
Affiliation(s)
| | - Ezgi P Yalcintas
- Department of Mechanical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Jason D Smith
- Engineering Research Accelerator, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Saadyah Averick
- Neuroscience Institute, Allegheny Health Network, Allegheny General Hospital, Pittsburgh, PA, USA.; Neuroscience Disruptive Research Lab, Allegheny General Hospital, Pittsburgh, PA, USA
| | - Phil G Campbell
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA; Engineering Research Accelerator, Carnegie Mellon University, Pittsburgh, PA, USA.
| | - O Burak Ozdoganlar
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA; Department of Mechanical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA; Department of Material Science and Engineering, Carnegie Mellon University, Pittsburgh, PA, USA.
| |
Collapse
|
48
|
Gulati R, Nandi D, Sarkar K, Venkataraman P, Ramkumar KM, Ranjan P, Janardhanan R. Exosomes as Theranostic Targets: Implications for the Clinical Prognosis of Aggressive Cancers. Front Mol Biosci 2022; 9:890768. [PMID: 35813829 PMCID: PMC9260243 DOI: 10.3389/fmolb.2022.890768] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 05/20/2022] [Indexed: 12/12/2022] Open
Abstract
Exosomes are extracellular vesicles produced by various cell types and extensively distributed in physiological fluids. Because of their significant role in cancer progression, they have been a focal point for the novel cancer therapy approach. Exosomes are highly efficient at transporting proteins, RNAs, and small drugs into cancer cells for therapeutic purposes. In addition to their prominent role as potential biomarkers for transporting targeted information from their progenitor cells, exosomes have also emerged as a new avenue for developing more effective clinical diagnostics and therapeutic techniques, also known as exosome theranostics. Lipids, proteins, and nucleic acids transported by exosomes were investigated as potential biomarkers for cancer diagnosis, prognosis, and future cancer treatment targets. The unique mechanism of exosomes and their therapeutic as well as diagnostic uses, also known as theranostic applications of exosomes in malignancies, are discussed in this review.
Collapse
Affiliation(s)
- Richa Gulati
- Department of Medical Research, Faculty of Medicine and Health Sciences, SRM Institute of Science and Technology, Kattankulathur, India
| | - Dhruva Nandi
- Department of Medical Research, Faculty of Medicine and Health Sciences, SRM Institute of Science and Technology, Kattankulathur, India
| | - Koustav Sarkar
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, India
| | - P. Venkataraman
- Department of Medical Research, Faculty of Medicine and Health Sciences, SRM Institute of Science and Technology, Kattankulathur, India
| | - K. M. Ramkumar
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, India
| | - Priya Ranjan
- Bhubaneswar Institute of Technology, Rourkela, India
| | - Rajiv Janardhanan
- Department of Medical Research, Faculty of Medicine and Health Sciences, SRM Institute of Science and Technology, Kattankulathur, India
- *Correspondence: Rajiv Janardhanan,
| |
Collapse
|
49
|
Yerneni SS, Lathwal S, Cuthbert J, Kapil K, Szczepaniak G, Jeong J, Das SR, Campbell PG, Matyjaszewski K. Controlled Release of Exosomes Using Atom Transfer Radical Polymerization-Based Hydrogels. Biomacromolecules 2022; 23:1713-1722. [PMID: 35302760 DOI: 10.1021/acs.biomac.1c01636] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Exosomes are 30-200 nm sized extracellular vesicles that are increasingly recognized as potential drug delivery vehicles. However, exogenous exosomes are rapidly cleared from the blood upon intravenous delivery, which limits their therapeutic potential. Here, we report bioactive exosome-tethered poly(ethylene oxide)-based hydrogels for the localized delivery of therapeutic exosomes. Using cholesterol-modified DNA tethers, the lipid membrane of exosomes was functionalized with initiators to graft polymers in the presence of additional initiators and crosslinker using photoinduced atom transfer radical polymerization (ATRP). This strategy of tethering exosomes within the hydrogel network allowed their controlled release over a period of 1 month, which was much longer than physically entrapped exosomes. Exosome release profile was tuned by varying the crosslinking density of the polymer network and the use of photocleavable tethers allowed stimuli-responsive release of exosomes. The therapeutic potential of the hydrogels was assessed by evaluating the osteogenic potential of bone morphogenetic protein 2-loaded exosomes on C2C12 and MC3T3-E1 cells. Thus, ATRP-based exosome-tethered hydrogels represent a tunable platform with improved efficacy and an extended-release profile.
Collapse
Affiliation(s)
- Saigopalakrishna S Yerneni
- Department of Biomedical Engineering and Engineering Research Accelerator, Carnegie Mellon University, 5000 Forbes Avenue, Pittsburgh, Pennsylvania 15213, United States
| | - Sushil Lathwal
- Department of Chemistry, Carnegie Mellon University, 4400 Fifth Avenue, Pittsburgh, Pennsylvania 15213, United States.,The Center for Nucleic Acids Science & Technology, Carnegie Mellon University, 4400 Fifth Avenue, Pittsburgh, Pennsylvania 15213, United States
| | - Julia Cuthbert
- Department of Chemistry, Carnegie Mellon University, 4400 Fifth Avenue, Pittsburgh, Pennsylvania 15213, United States
| | - Kriti Kapil
- Department of Chemistry, Carnegie Mellon University, 4400 Fifth Avenue, Pittsburgh, Pennsylvania 15213, United States
| | - Grzegorz Szczepaniak
- Department of Chemistry, Carnegie Mellon University, 4400 Fifth Avenue, Pittsburgh, Pennsylvania 15213, United States
| | - Jaepil Jeong
- Department of Chemistry, Carnegie Mellon University, 4400 Fifth Avenue, Pittsburgh, Pennsylvania 15213, United States.,The Center for Nucleic Acids Science & Technology, Carnegie Mellon University, 4400 Fifth Avenue, Pittsburgh, Pennsylvania 15213, United States
| | - Subha R Das
- Department of Chemistry, Carnegie Mellon University, 4400 Fifth Avenue, Pittsburgh, Pennsylvania 15213, United States.,The Center for Nucleic Acids Science & Technology, Carnegie Mellon University, 4400 Fifth Avenue, Pittsburgh, Pennsylvania 15213, United States
| | - Phil G Campbell
- Department of Biomedical Engineering and Engineering Research Accelerator, Carnegie Mellon University, 5000 Forbes Avenue, Pittsburgh, Pennsylvania 15213, United States
| | - Krzysztof Matyjaszewski
- Department of Chemistry, Carnegie Mellon University, 4400 Fifth Avenue, Pittsburgh, Pennsylvania 15213, United States
| |
Collapse
|
50
|
Lv H, Wang T, Ma F, Zhang K, Gao T, Pei R, Zhang Y. Aptamer-functionalized targeted siRNA delivery system for tumor immunotherapy. Biomed Mater 2022; 17. [PMID: 35147520 DOI: 10.1088/1748-605x/ac5415] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 02/09/2022] [Indexed: 11/11/2022]
Abstract
Programmed death ligand 1 (PD-L1) overexpressed on the surface of tumor cells is one of the reasons for tumor immune escape. Reducing PD-L1 expression has been proved to be an effective strategy to facilitate immune system activation and inhibit tumor progression. RNA interference (RNAi) is a promising technology for gene regulation in tumor therapy. In this study, we constructed a targeted siRNA delivery system NPs@apt to transfect PD-L1 siRNA into human non-small-cell lung carcinoma cell line (A549) for inhibiting tumor immune evasion. NPs@apt was prepared by compressing PD-L1 siRNA with cationic Lipofectamine 2000, fusing with erythrocyte membrane-derived nanovesicles, and further modifying with targeting AS1411 aptamer. The introduction of erythrocyte membrane endowed the siRNA delivery system with lower cytotoxicity and the ability to escape from the phagocytosis of macrophages. The stability of NPs@apt and the protection to loaded siRNA were confirmed.In vitrostudies after NPs@apt treatment demonstrated that PD-L1 siRNA was selectively delivered into A549 cells, and further resulted in PD-L1 gene knockdown, T cell activation and tumor cell growth inhibition. This study offered an alternative strategy for specific siRNA transfection for improving anti-tumor immunity.
Collapse
Affiliation(s)
- Haiyin Lv
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei 230026, People's Republic of China.,CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, People's Republic of China
| | - Tengfei Wang
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, People's Republic of China
| | - Fanshu Ma
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, People's Republic of China
| | - Kunchi Zhang
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, People's Republic of China
| | - Tian Gao
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei 230026, People's Republic of China.,CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, People's Republic of China
| | - Renjun Pei
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei 230026, People's Republic of China.,CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, People's Republic of China
| | - Ye Zhang
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, People's Republic of China
| |
Collapse
|