1
|
Song J, Wu Y, Chen Y, Sun X, Zhang Z. Epigenetic regulatory mechanism of macrophage polarization in diabetic wound healing (Review). Mol Med Rep 2025; 31:2. [PMID: 39422035 PMCID: PMC11551531 DOI: 10.3892/mmr.2024.13367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Accepted: 09/24/2024] [Indexed: 10/19/2024] Open
Abstract
Diabetic wounds represent a significant complication of diabetes and present a substantial challenge to global public health. Macrophages are crucial effector cells that play a pivotal role in the pathogenesis of diabetic wounds, through their polarization into distinct functional phenotypes. The field of epigenetics has emerged as a rapidly advancing research area, as this phenomenon has the potential to markedly affect gene expression, cellular differentiation, tissue development and susceptibility to disease. Understanding epigenetic mechanisms is crucial to further exploring disease pathogenesis. A growing body of scientific evidence has highlighted the pivotal role of epigenetics in the regulation of macrophage phenotypes. Various epigenetic mechanisms, such as DNA methylation, histone modification and non‑coding RNAs, are involved in the modulation of macrophage phenotype differentiation in response to the various environmental stimuli present in diabetic wounds. The present review provided an overview of the various changes that take place in macrophage phenotypes and functions within diabetic wounds and discussed the emerging role of epigenetic modifications in terms of regulating macrophage plasticity in diabetic wounds. It is hoped that this synthesis of information will facilitate the elucidation of diabetic wound pathogenesis and the identification of potential therapeutic targets.
Collapse
Affiliation(s)
- Jielin Song
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 300000, P.R. China
| | - Yuqing Wu
- The First Clinical Medical College, Guangdong Pharmaceutical University, Guangzhou, Guangdong 510000, P.R. China
| | - Yunli Chen
- The First Clinical Medical College, Guangdong Pharmaceutical University, Guangzhou, Guangdong 510000, P.R. China
| | - Xu Sun
- Department of Traditional Chinese Medicine Surgery, The Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300000, P.R. China
| | - Zhaohui Zhang
- Department of Traditional Chinese Medicine Surgery, The Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300000, P.R. China
| |
Collapse
|
2
|
Fawzy MP, Hassan HAFM, Sedky NK, Nafie MS, Youness RA, Fahmy SA. Revolutionizing cancer therapy: nanoformulation of miRNA-34 - enhancing delivery and efficacy for various cancer immunotherapies: a review. NANOSCALE ADVANCES 2024:d4na00488d. [PMID: 39309515 PMCID: PMC11414826 DOI: 10.1039/d4na00488d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 09/12/2024] [Indexed: 09/25/2024]
Abstract
Despite recent advancements in cancer therapies, challenges such as severe toxic effects, non-selective targeting, resistance to chemotherapy and radiotherapy, and recurrence of metastatic tumors persist. Consequently, there has been considerable effort to explore innovative anticancer compounds, particularly in immunotherapy, which offer the potential for enhanced biosafety and efficacy in cancer prevention and treatment. One such avenue of exploration involves the miRNA-34 (miR-34) family, known for its ability to inhibit tumorigenesis across various cancers. Dysregulation of miR-34 has been observed in several human cancers, and it is recognized as a tumor suppressor microRNA due to its synergistic interaction with the well-established tumor suppressor p53. However, challenges have arisen with the therapeutic application of miR-34a. These include its susceptibility to degradation by RNase in serum, limiting its ability to penetrate capillary endothelium and reach target cells, as well as reports of immunoreactive adverse reactions. Furthermore, unexpected side effects may occur, such as the accumulation of therapeutic miRNAs in healthy tissues due to interactions with serum proteins on nano-vector surfaces, nanoparticle breakdown in the bloodstream due to shearing stress, and unsuccessful extravasation of nanocarriers to target cells owing to interstitial fluid pressure. Despite these challenges, miR-34a remains a promising candidate for cancer therapy, and other members of the miR-34 family have also shown potential in inhibiting tumor cell proliferation. While the in vivo applications of miR-34b/c are limited, they warrant further exploration for oncotherapy. Recently, procedures utilizing nanoparticles have been developed to address the challenges associated with the clinical use of miR-34, demonstrating efficacy both in vitro and in vivo. This review highlights emerging trends in nanodelivery systems for miR-34 targeting cancer cells, offering insights into novel nanoformulations designed to enhance the anticancer therapeutic activity and targeting precision of miR-34. As far as current knowledge extends, no similar recent review comprehensively addresses the diverse nanoformulations aimed at optimizing the therapeutic potential of miR-34 in anticancer strategies.
Collapse
Affiliation(s)
- Marola Paula Fawzy
- Department of Chemistry, School of Life and Medical Sciences, University of Hertfordshire Hosted by Global Academic Foundation R5 New Garden City, New Capital Cairo 11835 Egypt
| | - Hatem A F M Hassan
- Medway School of Pharmacy, University of Kent Central Avenue, Chatham Maritime Canterbury ME44TB UK
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University 11562 Cairo Egypt
| | - Nada K Sedky
- Department of Biochemistry, School of Life and Medical Sciences, University of Hertfordshire Hosted by Global Academic Foundation R5 New Garden City, New Administrative Capital Cairo 11835 Egypt
| | - Mohamed S Nafie
- Department of Chemistry, College of Sciences, University of Sharjah (P.O. 27272) Sharjah United Arab Emirates (UAE)
- Chemistry Department, Faculty of Science, Suez Canal University (P.O. 41522) Ismailia Egypt
| | - Rana A Youness
- Molecular Genetics and Biochemistry Department, Molecular Genetics Research Team (MGRT), Faculty of Biotechnology, German International University (GIU) 11835 Cairo Egypt
| | - Sherif Ashraf Fahmy
- Department of Chemistry, School of Life and Medical Sciences, University of Hertfordshire Hosted by Global Academic Foundation R5 New Garden City, New Capital Cairo 11835 Egypt
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg Robert-Koch-Str. 4 35037 Marburg Germany
| |
Collapse
|
3
|
Wang H. A Review of Nanotechnology in microRNA Detection and Drug Delivery. Cells 2024; 13:1277. [PMID: 39120308 PMCID: PMC11311607 DOI: 10.3390/cells13151277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 07/27/2024] [Accepted: 07/28/2024] [Indexed: 08/10/2024] Open
Abstract
MicroRNAs (miRNAs) are small, non-coding RNAs that play a crucial role in regulating gene expression. Dysfunction in miRNAs can lead to various diseases, including cancers, neurological disorders, and cardiovascular conditions. To date, approximately 2000 miRNAs have been identified in humans. These small molecules have shown promise as disease biomarkers and potential therapeutic targets. Therefore, identifying miRNA biomarkers for diseases and developing effective miRNA drug delivery systems are essential. Nanotechnology offers promising new approaches to addressing scientific and medical challenges. Traditional miRNA detection methods include next-generation sequencing, microarrays, Northern blotting, and reverse transcription-quantitative polymerase chain reaction (RT-qPCR). Nanotechnology can serve as an effective alternative to Northern blotting and RT-qPCR for miRNA detection. Moreover, nanomaterials exhibit unique properties that differ from larger counterparts, enabling miRNA therapeutics to more effectively enter target cells, reduce degradation in the bloodstream, and be released in specific tissues or cells. This paper reviews the application of nanotechnology in miRNA detection and drug delivery systems. Given that miRNA therapeutics are still in the developing stages, nanotechnology holds great promise for accelerating miRNA therapeutics development.
Collapse
Affiliation(s)
- Hsiuying Wang
- Institute of Statistics, National Yang Ming Chiao Tung University, Hsinchu 300093, Taiwan
| |
Collapse
|
4
|
Gao X, You X, Wang G, Liu M, Ye L, Meng Y, Luo G, Xu D, Liu M. MiR-320 inhibits PRRSV replication by targeting PRRSV ORF6 and porcine CEBPB. Vet Res 2024; 55:61. [PMID: 38750508 PMCID: PMC11097481 DOI: 10.1186/s13567-024-01309-7] [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: 11/12/2023] [Accepted: 02/23/2024] [Indexed: 05/18/2024] Open
Abstract
Porcine reproductive and respiratory syndrome (PRRS), a highly contagious disease caused by Porcine reproductive and respiratory syndrome virus (PRRSV), results in huge economic losses to the world pig industry. MiRNAs have been reported to be involved in regulation of viral infection. In our study, miR-320 was one of 21 common differentially expressed miRNAs of Meishan, Pietrain, and Landrace pig breeds at 9-h post-infection (hpi). Bioinformatics and experiments found that PRRSV replication was inhibited by miR-320 through directly targeting PRRSV ORF6. In addition, the expression of CCAAT enhancer binding protein beta (CEBPB) was also inhibited by miR-320 by targeting the 3' UTR of CEBPB, which significantly promotes PRRSV replication. Intramuscular injection of pEGFP-N1-miR-320 verified that miR-320 significantly inhibited the replication of PRRSV and alleviated the symptoms caused by PRRSV in piglets. Taken together, miR-320 have significant roles in the infection and may be promising therapeutic target for PRRS.
Collapse
Affiliation(s)
- Xiaoxiao Gao
- Colleges of Animal Science and Technology/College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Xiangbin You
- Colleges of Animal Science and Technology/College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471023, China
| | - Guowei Wang
- Colleges of Animal Science and Technology/College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Mengtian Liu
- Colleges of Animal Science and Technology/College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Longlong Ye
- Colleges of Animal Science and Technology/College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Yufeng Meng
- Colleges of Animal Science and Technology/College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
- Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, 430070, China
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan, 430070, China
| | - Gan Luo
- Colleges of Animal Science and Technology/College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
- Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, 430070, China
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan, 430070, China
| | - Dequan Xu
- Colleges of Animal Science and Technology/College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
- Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, 430070, China
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan, 430070, China
| | - Min Liu
- Colleges of Animal Science and Technology/College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China.
| |
Collapse
|
5
|
Wang R, Yan Q, Liu X, Wu J. Unraveling lipid metabolism reprogramming for overcoming drug resistance in melanoma. Biochem Pharmacol 2024; 223:116122. [PMID: 38467377 DOI: 10.1016/j.bcp.2024.116122] [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: 01/17/2024] [Revised: 02/27/2024] [Accepted: 03/07/2024] [Indexed: 03/13/2024]
Abstract
Cutaneous melanoma is the deadliest form of skin cancer, and its incidence is continuing to increase worldwide in the last decades. Traditional therapies for melanoma can easily cause drug resistance, thus the treatment of melanoma remains a challenge. Various studies have focused on reversing the drug resistance. As tumors grow and progress, cancer cells face a constantly changing microenvironment made up of different nutrients, metabolites, and cell types. Multiple studies have shown that metabolic reprogramming of cancer is not static, but a highly dynamic process. There is a growing interest in exploring the relationship between melanoma andmetabolic reprogramming, one of which may belipid metabolism. This review frames the recent research progresses on lipid metabolism in melanoma.In addition, we emphasize the dynamic ability of metabolism during tumorigenesis as a target for improving response to different therapies and for overcoming drug resistance in melanoma.
Collapse
Affiliation(s)
- Ruilong Wang
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Qin Yan
- Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xiao Liu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China.
| | - Jinfeng Wu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
6
|
Singh RR, Mondal I, Janjua T, Popat A, Kulshreshtha R. Engineered smart materials for RNA based molecular therapy to treat Glioblastoma. Bioact Mater 2024; 33:396-423. [PMID: 38059120 PMCID: PMC10696434 DOI: 10.1016/j.bioactmat.2023.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 10/19/2023] [Accepted: 11/14/2023] [Indexed: 12/08/2023] Open
Abstract
Glioblastoma (GBM) is an aggressive malignancy of the central nervous system (CNS) that remains incurable despite the multitude of improvements in cancer therapeutics. The conventional chemo and radiotherapy post-surgery have only been able to improve the prognosis slightly; however, the development of resistance and/or tumor recurrence is almost inevitable. There is a pressing need for adjuvant molecular therapies that can successfully and efficiently block tumor progression. During the last few decades, non-coding RNAs (ncRNAs) have emerged as key players in regulating various hallmarks of cancer including that of GBM. The levels of many ncRNAs are dysregulated in cancer, and ectopic modulation of their levels by delivering antagonists or overexpression constructs could serve as an attractive option for cancer therapy. The therapeutic potential of several types of ncRNAs, including miRNAs, lncRNAs, and circRNAs, has been validated in both in vitro and in vivo models of GBM. However, the delivery of these RNA-based therapeutics is highly challenging, especially to the tumors of the brain as the blood-brain barrier (BBB) poses as a major obstacle, among others. Also, since RNA is extremely fragile in nature, careful considerations must be met while designing a delivery agent. In this review we have shed light on how ncRNA therapy can overcome the limitations of its predecessor conventional therapy with an emphasis on smart nanomaterials that can aide in the safe and targeted delivery of nucleic acids to treat GBM. Additionally, critical gaps that currently exist for successful transition from viral to non-viral vector delivery systems have been identified. Finally, we have provided a perspective on the future directions, potential pathways, and target areas for achieving rapid clinical translation of, RNA-based macromolecular therapy to advance the effective treatment of GBM and other related diseases.
Collapse
Affiliation(s)
- Ravi Raj Singh
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi, New Delhi, India
- School of Pharmacy, The University of Queensland, Brisbane, QLD, 4072, Australia
- University of Queensland –IIT Delhi Academy of Research (UQIDAR)
| | - Indranil Mondal
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi, New Delhi, India
| | - Taskeen Janjua
- School of Pharmacy, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Amirali Popat
- School of Pharmacy, The University of Queensland, Brisbane, QLD, 4072, Australia
- Department of Functional Materials and Catalysis, Faculty of Chemistry, University of Vienna, Währinger Straße 42, 1090 Vienna, Austria
| | - Ritu Kulshreshtha
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi, New Delhi, India
| |
Collapse
|
7
|
Chen J, Zhang X, Cross R, Ahn Y, Huskin G, Evans W, Hwang PT, Kim JA, Brott BC, Jo H, Yoon YS, Jun HW. Atherosclerotic three-layer nanomatrix vascular sheets for high-throughput therapeutic evaluation. Biomaterials 2024; 305:122450. [PMID: 38169190 PMCID: PMC10843643 DOI: 10.1016/j.biomaterials.2023.122450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 12/15/2023] [Accepted: 12/22/2023] [Indexed: 01/05/2024]
Abstract
In vitro atherosclerosis models are essential to evaluate therapeutics before in vivo and clinical studies, but significant limitations remain, such as the lack of three-layer vascular architecture and limited atherosclerotic features. Moreover, no scalable 3D atherosclerosis model is available for making high-throughput assays for therapeutic evaluation. Herein, we report an in vitro 3D three-layer nanomatrix vascular sheet with critical atherosclerosis multi-features (VSA), including endothelial dysfunction, monocyte recruitment, macrophages, extracellular matrix remodeling, smooth muscle cell phenotype transition, inflammatory cytokine secretion, foam cells, and calcification initiation. Notably, we present the creation of high-throughput functional assays with VSAs and the use of these assays for evaluating therapeutics for atherosclerosis treatment. The therapeutics include conventional drugs (statin and sirolimus), candidates for treating atherosclerosis (curcumin and colchicine), and potential gene therapy (miR-146a-loaded liposomes). The high efficiency and flexibility of the scalable VSA functional assays should facilitate drug discovery and development for atherosclerosis.
Collapse
Affiliation(s)
- Jun Chen
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, AL, USA; Endomimetics, LLC., Birmingham, AL, USA
| | - Xixi Zhang
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Robbie Cross
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Yujin Ahn
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Gillian Huskin
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Will Evans
- Augusta University/University of Georgia Medical Partnership, Athens, GA, USA
| | | | - Jeong-A Kim
- Department of Medicine, Division of Endocrinology and Metabolism, UAB Comprehensive Diabetes Center, Birmingham, AL, USA
| | - Brigitta C Brott
- Endomimetics, LLC., Birmingham, AL, USA; Department of Medicine and Division of Cardiovascular Disease, The University of Alabama at Birmingham, AL, USA
| | - Hanjoong Jo
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Young-Sup Yoon
- Division of Cardiology, School of Medicine, Emory University, Atlanta, GA, USA
| | - Ho-Wook Jun
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, AL, USA; Endomimetics, LLC., Birmingham, AL, USA.
| |
Collapse
|
8
|
Gherman LM, Chiroi P, Nuţu A, Bica C, Berindan-Neagoe I. Profiling canine mammary tumors: A potential model for studying human breast cancer. Vet J 2024; 303:106055. [PMID: 38097103 DOI: 10.1016/j.tvjl.2023.106055] [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: 05/24/2023] [Revised: 12/05/2023] [Accepted: 12/08/2023] [Indexed: 12/24/2023]
Abstract
Despite all clinical progress recorded in the last decades, human breast cancer (HBC) remains a major challenge worldwide both in terms of its incidence and its management. Canine mammary tumors (CMTs) share similarities with HBC and represent an alternative model for HBC. The utility of the canine model in studying HBC relies on their common features, include spontaneous development, subtype classification, mutational profile, alterations in gene expression profile, and incidence/prevalence. This review describes the similarities between CMTs and HBC regarding genomic landscape, microRNA expression alteration, methylation, and metabolomic changes occurring during mammary gland carcinogenesis. The primary purpose of this review is to highlight the advantages of using the canine model as a translational animal model for HBC research and to investigate the challenges and limitations of this approach.
Collapse
Affiliation(s)
- Luciana-Madalina Gherman
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; Experimental Center of Iuliu Hatieganu University of Medicine and Pharmacy Cluj-Napoca, 400349 Cluj-Napoca, Romania
| | - Paul Chiroi
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
| | - Andreea Nuţu
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
| | - Cecilia Bica
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania.
| | - Ioana Berindan-Neagoe
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
| |
Collapse
|
9
|
Wang X, Gong W, Li R, Li L, Wang J. Preparation of genetically or chemically engineered exosomes and their therapeutic effects in bone regeneration and anti-inflammation. Front Bioeng Biotechnol 2024; 12:1329388. [PMID: 38314353 PMCID: PMC10834677 DOI: 10.3389/fbioe.2024.1329388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Accepted: 01/11/2024] [Indexed: 02/06/2024] Open
Abstract
The treatment of bone or cartilage damage and inflammation-related diseases has been a long-standing research hotspot. Traditional treatments such as surgery and cell therapy have only displayed limited efficacy because they can't avoid potential deterioration and ensure cell activity. Recently, exosomes have become a favorable tool for various tissue reconstruction due to their abundant content of proteins, lipids, DNA, RNA and other substances, which can promote bone regeneration through osteogenesis, angiogenesis and inflammation modulation. Besides, exosomes are also promising delivery systems because of stability in the bloodstream, immune stealth capacity, intrinsic cell-targeting property and outstanding intracellular communication. Despite having great potential in therapeutic delivery, exosomes still show some limitations in clinical studies, such as inefficient targeting ability, low yield and unsatisfactory therapeutic effects. In order to overcome the shortcomings, increasing studies have prepared genetically or chemically engineered exosomes to improve their properties. This review focuses on different methods of preparing genetically or chemically engineered exosomes and the therapeutic effects of engineering exosomes in bone regeneration and anti-inflammation, thereby providing some references for future applications of engineering exosomes.
Collapse
Affiliation(s)
- Xinyue Wang
- School of Stomatology, Lanzhou University, Lanzhou, China
| | - Weitao Gong
- School of Stomatology, Lanzhou University, Lanzhou, China
| | - Rongrong Li
- School of Stomatology, Lanzhou University, Lanzhou, China
| | - Lin Li
- School of Stomatology, Lanzhou University, Lanzhou, China
| | - Jing Wang
- School of Stomatology, Lanzhou University, Lanzhou, China
- Clinical Research Center for Oral Diseases, Lanzhou, China
| |
Collapse
|
10
|
Anajafi S, Paryan M, Khoshnazar A, Soleimani M, Mohammadi-Yeganeh S. miRNAs Delivery for Cancer-associated Fibroblasts' Activation and Drug Resistance in Cancer Microenvironment. Endocr Metab Immune Disord Drug Targets 2024; 24:333-347. [PMID: 37612874 DOI: 10.2174/1871530323666230823094556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 05/25/2023] [Accepted: 07/06/2023] [Indexed: 08/25/2023]
Abstract
Cancer-associated fibroblasts (CAFs) as a major component of cancer stroma contribute to diverse procedures of most solid tumors and might be a targeted cancer therapy approach. Their specified features, related signaling pathways, distinct biomarkers, and sub-populations need to be deciphered. There is a need for CAF extraction or induction for in vitro investigations. Some miRNAs could activate CAF-like phenotype and they also interfere in CAF-mediated drug resistance, aggressiveness, and metastatic behaviors of several cancer cell types. Due to the complex relevance of miRNA and CAFs, these non-coding oligonucleotides may serve as attractive scope for anti-cancer targeted therapies, but the lack of an efficient delivery system is still a major hurdle. Here, we have summarized the investigated information on CAF features, isolation, and induction procedures, and highlighted the miRNA-CAF communications, providing special insight into nano-delivery systems.
Collapse
Affiliation(s)
- Sara Anajafi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahdi Paryan
- Department of Research and Development, Production and Research Complex, Pasteur Institute of Iran, Tehran, Iran
| | - Amineh Khoshnazar
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Masoud Soleimani
- Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Samira Mohammadi-Yeganeh
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
11
|
Zhou X, Ye C, Jiang L, Zhu X, Zhou F, Xia M, Chen Y. The bone mesenchymal stem cell-derived exosomal miR-146a-5p promotes diabetic wound healing in mice via macrophage M1/M2 polarization. Mol Cell Endocrinol 2024; 579:112089. [PMID: 37863468 DOI: 10.1016/j.mce.2023.112089] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 10/10/2023] [Accepted: 10/12/2023] [Indexed: 10/22/2023]
Abstract
A diabetic wound is a refractory disease that afflicts patients globally. MicroRNA-146a-5p (miR-146a-5p) is reported to represent a potential therapeutic target for diabetic wounds. However, microRNA easily degrades in the wound microenvironment. This study extracted bone marrow mesenchymal stem cell (BMSC)-derived exosomes (EXO). Electroporation technology was used to load miR-146a-5p into EXO (labeled as EXO-miR-146a). The endothelial cells (human umbilical vein endothelial cells [HUVECs]) and macrophages were cocultured in transwell chambers in the presence of high glucose. Cell proliferation, migration, and angiogenesis were measured with cell counting kit 8, scratch, and tube forming assays, respectively. Flow cytometry was introduced to validate the biomarker of macrophages and BMSCs. The expression level of macrophage polarization-related proteins and tumor necrosis factor receptor-associated factor 6 (TRAF6) was assessed with western blotting analysis. The full-thickness skin wound model was developed to verify the in vitro results. EXO-miR-146a promoted the proliferation, migration, and angiogenesis of HUVECs in the hyperglycemic state by suppressing the TRAF6 expression in vitro. Additionally, EXO-miR-146a treatment facilitated M2 but inhibited M1 macrophage polarization. Furthermore, EXO-miR-146a enhances reepithelialization, angiogenesis, and M2 macrophage polarization, thereby accelerating diabetic wound healing in vivo. The EXO-miR-146a facilitated M2 macrophage polarization, proliferation, migration, and angiogenesis of HUVECs through TRAF6, thereby ameliorating intractable diabetic wound healing. These results established the basis for using EXO to deliver drugs and revealed mediators for diabetic wound treatment.
Collapse
Affiliation(s)
- Xijie Zhou
- Department of Hand and Microsurgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
| | - Chenhao Ye
- Department of Hand and Microsurgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
| | - Liangfu Jiang
- Department of Hand and Microsurgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
| | - Xuwei Zhu
- Department of Hand and Microsurgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
| | - Feiya Zhou
- Department of Hand and Microsurgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
| | - Meizi Xia
- Department of Nephrology, Wenzhou Geriatric Hospital, Wenzhou, 325000, China.
| | - Yiheng Chen
- Department of Hand and Microsurgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
| |
Collapse
|
12
|
Andone BA, Handrea-Dragan IM, Botiz I, Boca S. State-of-the-art and future perspectives in infertility diagnosis: Conventional versus nanotechnology-based assays. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2023; 54:102709. [PMID: 37717928 DOI: 10.1016/j.nano.2023.102709] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 08/27/2023] [Accepted: 09/07/2023] [Indexed: 09/19/2023]
Abstract
According to the latest World Health Organization statistics, around 50 to 80 million people worldwide suffer from infertility, amongst which male factors are responsible for around 20 to 30 % of all infertility cases while 50 % were attributed to the female ones. As it is becoming a recurrent health problem worldwide, clinicians require more accurate methods for the improvement of both diagnosis and treatment schemes. By emphasizing the potential use of innovative methods for the rapid identification of the infertility causes, this review presents the news from this dynamic domain and highlights the benefits brought by emerging research fields. A systematic description of the standard techniques used in clinical protocols for diagnosing infertility in both genders is firstly provided, followed by the presentation of more accurate and comprehensive nanotechnology-related analysis methods such as nanoscopic-resolution imaging, biosensing approaches and assays that employ nanomaterials in their design. Consequently, the implementation of nanotechnology related tools in clinical practice, as recently demonstrated in the selection of spermatozoa, the detection of key proteins in the fertilization process or the testing of DNA integrity or the evaluation of oocyte quality, might confer excellent advantages both for improving the assessment of infertility, and for the success of the fertilization process.
Collapse
Affiliation(s)
- Bianca-Astrid Andone
- Interdisciplinary Research Institute in Bio-Nano-Sciences, Babes-Bolyai University, 42 T. Laurian Str., 400271 Cluj-Napoca, Romania; Faculty of Physics, Babes-Bolyai University, 1 M. Kogalniceanu Str., 400084 Cluj-Napoca, Romania
| | - Iuliana M Handrea-Dragan
- Interdisciplinary Research Institute in Bio-Nano-Sciences, Babes-Bolyai University, 42 T. Laurian Str., 400271 Cluj-Napoca, Romania; Faculty of Physics, Babes-Bolyai University, 1 M. Kogalniceanu Str., 400084 Cluj-Napoca, Romania
| | - Ioan Botiz
- Interdisciplinary Research Institute in Bio-Nano-Sciences, Babes-Bolyai University, 42 T. Laurian Str., 400271 Cluj-Napoca, Romania
| | - Sanda Boca
- Interdisciplinary Research Institute in Bio-Nano-Sciences, Babes-Bolyai University, 42 T. Laurian Str., 400271 Cluj-Napoca, Romania; National Institute for Research and Development of Isotopic and Molecular Technologies, 67-103 Donat Str., 400293 Cluj-Napoca, Romania.
| |
Collapse
|
13
|
Wang J, Yao M, Zou J, Ding W, Sun M, Zhuge Y, Gao F. pH-Sensitive Nanoparticles for Colonic Delivery Anti-miR-301a in Mouse Models of Inflammatory Bowel Diseases. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:2797. [PMID: 37887947 PMCID: PMC10610125 DOI: 10.3390/nano13202797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 10/13/2023] [Accepted: 10/16/2023] [Indexed: 10/28/2023]
Abstract
Though the anti-miR-301a (anti-miR) is a promising treatment strategy for inflammatory bowel disease (IBD), the degradability and the poor targeting of the intestine are a familiar issue. This study aimed to develop a multifunctional oral nanoparticle delivery system loaded with anti-miR for improving the targeting ability and the therapeutic efficacy. The HA-CS/ES100/PLGA nanoparticles (HCeP NPs) were prepared using poly (lactic-co-glycolic acid) copolymer (PLGA), enteric material Eudragit®S100 (ES100), chitosan (CS), and hyaluronic acid (HA). The toxicity of nanoparticles was investigated via the Cell Counting Kit-8, and the cellular uptake and inflammatory factors of nanoparticles were further studied. Moreover, we documented the colon targeting and pharmacodynamic properties of nanoparticles. The nanoparticles with uniform particle size exhibited pH-sensitive release, favorable gene protection, and storage stability. Cytology experiments showed that anti-miR@HCeP NPs improved the cellular uptake through HA and reduced pro-inflammatory factors. Administering anti-miR@HCeP NPs orally to IBD mice markedly reduced their pro-inflammatory factors levels and disease activity indices. We also confirmed that anti-miR@HCeP NPs mostly accumulated in the colon site, and effectively repaired the intestinal barrier, as well as relieved intestinal inflammation. The above nanoparticle is a candidate of the treatment for IBD due to its anti-inflammatory properties.
Collapse
Affiliation(s)
- Junshan Wang
- Department of Gastroenterology, Chongming Branch of Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 202157, China
| | - Min Yao
- Shanghai Frontier Science Research Base of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China; (M.Y.); (J.Z.); (W.D.); (M.S.)
| | - Jiafeng Zou
- Shanghai Frontier Science Research Base of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China; (M.Y.); (J.Z.); (W.D.); (M.S.)
| | - Wenxing Ding
- Shanghai Frontier Science Research Base of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China; (M.Y.); (J.Z.); (W.D.); (M.S.)
| | - Mingyue Sun
- Shanghai Frontier Science Research Base of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China; (M.Y.); (J.Z.); (W.D.); (M.S.)
| | - Ying Zhuge
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China;
| | - Feng Gao
- Shanghai Frontier Science Research Base of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China; (M.Y.); (J.Z.); (W.D.); (M.S.)
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
- Shanghai Key Laboratory of Functional Materials Chemistry, East China University of Science and Technology, Shanghai 200237, China
- Optogenetics and Synthetic Biology Interdisciplinary Research Center, State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| |
Collapse
|
14
|
Garrido-Cano I, Adam-Artigues A, Lameirinhas A, Blandez JF, Candela-Noguera V, Lluch A, Bermejo B, Sancenón F, Cejalvo JM, Martínez-Máñez R, Eroles P. Delivery of miR-200c-3p Using Tumor-Targeted Mesoporous Silica Nanoparticles for Breast Cancer Therapy. ACS APPLIED MATERIALS & INTERFACES 2023; 15:38323-38334. [PMID: 37549382 PMCID: PMC10436244 DOI: 10.1021/acsami.3c07541] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 07/25/2023] [Indexed: 08/09/2023]
Abstract
Despite advances in breast cancer treatment, it remains the leading cause of cancer-related death in women worldwide. In this context, microRNAs have emerged as potential therapeutic targets but still present some limitations for in vivo applications. Particularly, miR-200c-3p is a well-known tumor suppressor microRNA that inhibits tumor progression and metastasis in breast cancer through downregulating ZEB1 and ZEB2. Based on the above, we describe the design and validation of a nanodevice using mesoporous silica nanoparticles for miR-200c-3p delivery for breast cancer treatment. We demonstrate the biocompatibility of the synthesized nanodevices as well as their ability to escape from endosomes/lysosomes and inhibit tumorigenesis, invasion, migration, and proliferation of tumor cells in vitro. Moreover, tumor targeting and effective delivery of miR-200c-3p from the nanoparticles in vivo are confirmed in an orthotopic breast cancer mouse model, and the therapeutic efficacy is also evidenced by a decrease in tumor size and lung metastasis, while showing no signs of toxicity. Overall, our results provide evidence that miR-200c-3p-loaded nanoparticles are a potential strategy for breast cancer therapy and a safe and effective system for tumor-targeted delivery of microRNAs.
Collapse
Affiliation(s)
- Iris Garrido-Cano
- Biomedical
Research Institute INCLIVA, Valencia 46010, Spain
- Instituto
Interuniversitario de Investigación de Reconocimiento Molecular
y Desarrollo Tecnológico (IDM), Universitat Politècnica
de València, Universitat de València, Valencia 46010, Spain
| | | | - Ana Lameirinhas
- Biomedical
Research Institute INCLIVA, Valencia 46010, Spain
| | - Juan F. Blandez
- Instituto
Interuniversitario de Investigación de Reconocimiento Molecular
y Desarrollo Tecnológico (IDM), Universitat Politècnica
de València, Universitat de València, Valencia 46010, Spain
- CIBER
de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid 28029, Spain
- Unidad
Mixta de Investigación en Nanomedicina y Sensores, Universitat Politècnica de València,
IIS La Fe, Valencia 46026, Spain
| | - Vicente Candela-Noguera
- Instituto
Interuniversitario de Investigación de Reconocimiento Molecular
y Desarrollo Tecnológico (IDM), Universitat Politècnica
de València, Universitat de València, Valencia 46010, Spain
| | - Ana Lluch
- Biomedical
Research Institute INCLIVA, Valencia 46010, Spain
- Centro
de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid 28029, Spain
- Universitat
de València, Valencia 46010, Spain
- Clinical
Oncology Department, Hospital Clínico
Universitario de Valencia, Valencia 46010, Spain
| | - Begoña Bermejo
- Biomedical
Research Institute INCLIVA, Valencia 46010, Spain
- Centro
de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid 28029, Spain
- Clinical
Oncology Department, Hospital Clínico
Universitario de Valencia, Valencia 46010, Spain
| | - Felix Sancenón
- Instituto
Interuniversitario de Investigación de Reconocimiento Molecular
y Desarrollo Tecnológico (IDM), Universitat Politècnica
de València, Universitat de València, Valencia 46010, Spain
- CIBER
de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid 28029, Spain
- Unidad
Mixta de Investigación en Nanomedicina y Sensores, Universitat Politècnica de València,
IIS La Fe, Valencia 46026, Spain
- Unidad
Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades
y Nanomedicina. Universitat Politècnica de Valencia, Centro de Investigación Príncipe Felipe, Valencia 46012, Spain
| | - Juan Miguel Cejalvo
- Biomedical
Research Institute INCLIVA, Valencia 46010, Spain
- Centro
de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid 28029, Spain
- Clinical
Oncology Department, Hospital Clínico
Universitario de Valencia, Valencia 46010, Spain
| | - Ramón Martínez-Máñez
- Instituto
Interuniversitario de Investigación de Reconocimiento Molecular
y Desarrollo Tecnológico (IDM), Universitat Politècnica
de València, Universitat de València, Valencia 46010, Spain
- CIBER
de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid 28029, Spain
- Unidad
Mixta de Investigación en Nanomedicina y Sensores, Universitat Politècnica de València,
IIS La Fe, Valencia 46026, Spain
- Unidad
Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades
y Nanomedicina. Universitat Politècnica de Valencia, Centro de Investigación Príncipe Felipe, Valencia 46012, Spain
| | - Pilar Eroles
- Biomedical
Research Institute INCLIVA, Valencia 46010, Spain
- Centro
de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid 28029, Spain
- Universitat
de València, Valencia 46010, Spain
| |
Collapse
|
15
|
Bravo-Vázquez LA, Méndez-García A, Rodríguez AL, Sahare P, Pathak S, Banerjee A, Duttaroy AK, Paul S. Applications of nanotechnologies for miRNA-based cancer therapeutics: current advances and future perspectives. Front Bioeng Biotechnol 2023; 11:1208547. [PMID: 37576994 PMCID: PMC10416113 DOI: 10.3389/fbioe.2023.1208547] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 07/18/2023] [Indexed: 08/15/2023] Open
Abstract
MicroRNAs (miRNAs) are short (18-25 nt), non-coding, widely conserved RNA molecules responsible for regulating gene expression via sequence-specific post-transcriptional mechanisms. Since the human miRNA transcriptome regulates the expression of a number of tumor suppressors and oncogenes, its dysregulation is associated with the clinical onset of different types of cancer. Despite the fact that numerous therapeutic approaches have been designed in recent years to treat cancer, the complexity of the disease manifested by each patient has prevented the development of a highly effective disease management strategy. However, over the past decade, artificial miRNAs (i.e., anti-miRNAs and miRNA mimics) have shown promising results against various cancer types; nevertheless, their targeted delivery could be challenging. Notably, numerous reports have shown that nanotechnology-based delivery of miRNAs can greatly contribute to hindering cancer initiation and development processes, representing an innovative disease-modifying strategy against cancer. Hence, in this review, we evaluate recently developed nanotechnology-based miRNA drug delivery systems for cancer therapeutics and discuss the potential challenges and future directions, such as the promising use of plant-made nanoparticles, phytochemical-mediated modulation of miRNAs, and nanozymes.
Collapse
Affiliation(s)
| | | | - Alma L. Rodríguez
- Tecnologico de Monterrey, School of Engineering and Sciences, Querétaro, México
| | - Padmavati Sahare
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, México
| | - Surajit Pathak
- Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Chennai, India
| | - Antara Banerjee
- Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Chennai, India
| | - Asim K. Duttaroy
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Sujay Paul
- Tecnologico de Monterrey, School of Engineering and Sciences, Querétaro, México
| |
Collapse
|
16
|
Timofeeva AM, Paramonik AP, Sedykh SS, Nevinsky GA. Milk Exosomes: Next-Generation Agents for Delivery of Anticancer Drugs and Therapeutic Nucleic Acids. Int J Mol Sci 2023; 24:10194. [PMID: 37373342 DOI: 10.3390/ijms241210194] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/05/2023] [Accepted: 06/14/2023] [Indexed: 06/29/2023] Open
Abstract
Exosomes are nanovesicles 40-120 nm in diameter secreted by almost all cell types and providing humoral intercellular interactions. Given the natural origin and high biocompatibility, the potential for loading various anticancer molecules and therapeutic nucleic acids inside, and the surface modification possibility for targeted delivery, exosomes are considered to be a promising means of delivery to cell cultures and experimental animal organisms. Milk is a unique natural source of exosomes available in semi-preparative and preparative quantities. Milk exosomes are highly resistant to the harsh conditions of the gastrointestinal tract. In vitro studies have demonstrated that milk exosomes have an affinity to epithelial cells, are digested by cells by endocytosis mechanism, and can be used for oral delivery. With milk exosome membranes containing hydrophilic and hydrophobic components, exosomes can be loaded with hydrophilic and lipophilic drugs. This review covers a number of scalable protocols for isolating and purifying exosomes from human, cow, and horse milk. Additionally, it considers passive and active methods for drug loading into exosomes, as well as methods for modifying and functionalizing the surface of milk exosomes with specific molecules for more efficient and specific delivery to target cells. In addition, the review considers various approaches to visualize exosomes and determine cellular localization and bio-distribution of loaded drug molecules in tissues. In conclusion, we outline new challenges for studying milk exosomes, a new generation of targeted delivery agents.
Collapse
Affiliation(s)
- Anna M Timofeeva
- SB RAS Institute of Chemical Biology and Fundamental Medicine, 630090 Novosibirsk, Russia
- Faculty of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Anastasia P Paramonik
- SB RAS Institute of Chemical Biology and Fundamental Medicine, 630090 Novosibirsk, Russia
- Faculty of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Sergey S Sedykh
- SB RAS Institute of Chemical Biology and Fundamental Medicine, 630090 Novosibirsk, Russia
- Faculty of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Georgy A Nevinsky
- SB RAS Institute of Chemical Biology and Fundamental Medicine, 630090 Novosibirsk, Russia
- Faculty of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| |
Collapse
|
17
|
Capolla S, Argenziano M, Bozzer S, D’Agaro T, Bittolo T, De Leo L, Not T, Busato D, Dal Bo M, Toffoli G, Cavalli R, Gattei V, Bomben R, Macor P. Targeted chitosan nanobubbles as a strategy to down-regulate microRNA-17 into B-cell lymphoma models. Front Immunol 2023; 14:1200310. [PMID: 37359561 PMCID: PMC10285521 DOI: 10.3389/fimmu.2023.1200310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 05/26/2023] [Indexed: 06/28/2023] Open
Abstract
Introduction MicroRNAs represent interesting targets for new therapies because their altered expression influences tumor development and progression. miR-17 is a prototype of onco-miRNA, known to be overexpressed in B-cell non-Hodgkin lymphoma (B-NHL) with peculiar clinic-biological features. AntagomiR molecules have been largely studied to repress the regulatory functions of up-regulated onco-miRNAs, but their clinical use is mainly limited by their rapid degradation, kidney elimination and poor cellular uptake when injected as naked oligonucleotides. Methods To overcome these problems, we exploited CD20 targeted chitosan nanobubbles (NBs) for a preferential and safe delivery of antagomiR17 to B-NHL cells. Results Positively charged 400 nm-sized nanobubbles (NBs) represent a stable and effective nanoplatform for antagomiR encapsulation and specific release into B-NHL cells. NBs rapidly accumulated in tumor microenvironment, but only those conjugated with a targeting system (antiCD20 antibodies) were internalized into B-NHL cells, releasing antagomiR17 in the cytoplasm, both in vitro and in vivo. The result is the down-regulation of miR-17 level and the reduction in tumor burden in a human-mouse B-NHL model, without any documented side effects. Discussion Anti-CD20 targeted NBs investigated in this study showed physico-chemical and stability properties suitable for antagomiR17 delivery in vivo and represent a useful nanoplatform to address B-cell malignancies or other cancers through the modification of their surface with specific targeting antibodies.
Collapse
Affiliation(s)
- Sara Capolla
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Monica Argenziano
- Department of Scienza e Tecnologia del Farmaco, University of Turin, Turin, Italy
| | - Sara Bozzer
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Tiziana D’Agaro
- Clinical and Experimental Onco-Hematology Unit, Centro di Riferimento Oncologico di Aviano (CRO)-Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Aviano, Italy
| | - Tamara Bittolo
- Clinical and Experimental Onco-Hematology Unit, Centro di Riferimento Oncologico di Aviano (CRO)-Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Aviano, Italy
| | - Luigina De Leo
- Department of Pediatrics, Institute for Maternal and Child Health, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Burlo Garofolo, Trieste, Italy
| | - Tarcisio Not
- Department of Pediatrics, Institute for Maternal and Child Health, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Burlo Garofolo, Trieste, Italy
| | - Davide Busato
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO)-Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Aviano, Italy
| | - Michele Dal Bo
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO)-Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Aviano, Italy
| | - Giuseppe Toffoli
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO)-Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Aviano, Italy
| | - Roberta Cavalli
- Department of Scienza e Tecnologia del Farmaco, University of Turin, Turin, Italy
| | - Valter Gattei
- Clinical and Experimental Onco-Hematology Unit, Centro di Riferimento Oncologico di Aviano (CRO)-Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Aviano, Italy
| | - Riccardo Bomben
- Clinical and Experimental Onco-Hematology Unit, Centro di Riferimento Oncologico di Aviano (CRO)-Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Aviano, Italy
| | - Paolo Macor
- Department of Life Sciences, University of Trieste, Trieste, Italy
| |
Collapse
|
18
|
Iqbal MJ, Javed Z, Sadia H, Mehmood S, Akbar A, Zahid B, Nadeem T, Roshan S, Varoni EM, Iriti M, Gürer ES, Sharifi-Rad J, Calina D. Targeted therapy using nanocomposite delivery systems in cancer treatment: highlighting miR34a regulation for clinical applications. Cancer Cell Int 2023; 23:84. [PMID: 37149609 PMCID: PMC10164299 DOI: 10.1186/s12935-023-02929-3] [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: 01/23/2023] [Accepted: 04/18/2023] [Indexed: 05/08/2023] Open
Abstract
The clinical application of microRNAs in modern therapeutics holds great promise to uncover molecular limitations and conquer the unbeatable castle of cancer metastasis. miRNAs play a decisive role that regulating gene expression at the post-transcription level while controlling both the stability and translation capacity of mRNAs. Specifically, miR34a is a master regulator of the tumor suppressor gene, cancer progression, stemness, and drug resistance at the cell level in p53-dependent and independent signaling. With changing, trends in nanotechnology, in particular with the revolution in the field of nanomedicine, nano drug delivery systems have emerged as a prominent strategy in clinical practices coupled with miR34a delivery. Recently, it has been observed that forced miR34a expression in human cancer cell lines and model organisms limits cell proliferation and metastasis by targeting several signaling cascades, with various studies endorsing that miR34a deregulation in cancer cells modulates apoptosis and thus requires targeted nano-delivery systems for cancer treatment. In this sense, the present review aims to provide an overview of the clinical applications of miR34a regulation in targeted therapy of cancer.
Collapse
Affiliation(s)
| | - Zeeshan Javed
- Centre for Applied Molecular Biology, University of the Punjab, Lahore, Pakistan
| | | | - Sajid Mehmood
- Department of Biochemistry, Islam Medical and Dental College, Sialkot, Pakistan
| | - Ali Akbar
- Department of Microbiology, University of Balochistan Quetta, Quetta, Pakistan
| | - Benish Zahid
- Department of Pathobiology, KBCMA, CVAS, Sub Campus University of Veterinary and Animal Sciences, Narowal, Pakistan
| | - Tariq Nadeem
- National Centre of Excellence in Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Sadia Roshan
- Department of Zoology, University of Gujrat, Gujrat, Pakistan
| | - Elena Maria Varoni
- Dipartimento di Scienze Biomediche, Chirurgiche ed Odontoiatriche, Università degli Studi di Milano, Milan, Italy
| | - Marcello Iriti
- Department of Agricultural and Environmental Sciences, Università degli Studi di Milano, Milan, Italy
| | - Eda Sönmez Gürer
- Faculty of Pharmacy, Department of Pharmacognosy, Sivas Cumhuriyet University, Sivas, Turkey
| | | | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, Craiova, 200349, Romania.
| |
Collapse
|
19
|
Ho D, Lynd TO, Jun C, Shin J, Millican RC, Estep BK, Chen J, Zhang X, Brott BC, Kim DW, Sherwood JA, Hwang PTJ. MiR-146a encapsulated liposomes reduce vascular inflammatory responses through decrease of ICAM-1 expression, macrophage activation, and foam cell formation. NANOSCALE 2023; 15:3461-3474. [PMID: 36723042 DOI: 10.1039/d2nr03280e] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Vascular insults can create an inflammatory cascade involving endothelial cell, smooth muscle cell, and macrophage activation which can eventually lead to vascular disease such as atherosclerosis. Several studies have identified microRNA 146a's (miR-146a) anti-inflammatory potential based on its role in regulating the nuclear factor kappa beta (NF-κβ) pathway. Therefore, in this study, we introduced exogenous miR-146a encapsulated by liposomes to lipopolysaccharide (LPS) stimulated vascular cells and macrophages to reduce inflammatory responses. First, the miR-146a encapsulated liposomes showed uniform size (radius 96.4 ± 4.22 nm) and round shape, long term stability (at least two months), high encapsulation efficiency (69.73 ± 0.07%), and were well transfected to human aortic endothelial cells (HAECs), human aortic smooth muscle cells (SMCs), and human differentiated monocytes (U937 cells). In addition, we demonstrated that miR-146a encapsulated liposomes reduced vascular inflammation responses in HAECs and SMCs through inhibition of ICAM-1 expression and decreased monocyte adhesion. In macrophages, miR-146a liposome treatment demonstrated decreased production of proinflammatory cytokines, tumor necrosis factor-alpha (TNF-α) and interleukin-1 beta (IL-1β), as well as reduced oxidized low-density lipoprotein (ox-LDL) uptake and foam cell formation. Thus, based on these results, miR-146a encapsulated liposomes may be promising for reducing vascular inflammation by targeting its multiple associated mediators.
Collapse
Affiliation(s)
- Donald Ho
- Department of Pediatric Dentistry, University of Alabama at Birmingham, AL, 35294, USA
| | - Tyler O Lynd
- Department of Biomedical Engineering, University of Alabama at Birmingham, AL, 35294, USA
| | - Claire Jun
- School of Arts and Sciences, University of Pennsylvania, PA, 19104, USA
| | - Juhee Shin
- Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
| | | | - Benjamin K Estep
- Department of Biomedical Engineering, University of Alabama at Birmingham, AL, 35294, USA
| | - Jun Chen
- Department of Biomedical Engineering, University of Alabama at Birmingham, AL, 35294, USA
| | - Xixi Zhang
- Department of Biomedical Engineering, University of Alabama at Birmingham, AL, 35294, USA
| | - Brigitta C Brott
- Endomimetics, LLC, Birmingham, AL, 35242, USA.
- Department of Medicine and Division of Cardiovascular Disease, University of Alabama at Birmingham, AL, 35233, USA
| | - Dong Woon Kim
- Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
| | | | - Patrick T J Hwang
- Department of Biomedical Engineering, University of Alabama at Birmingham, AL, 35294, USA
- Endomimetics, LLC, Birmingham, AL, 35242, USA.
| |
Collapse
|
20
|
Traber GM, Yu AM. RNAi-Based Therapeutics and Novel RNA Bioengineering Technologies. J Pharmacol Exp Ther 2023; 384:133-154. [PMID: 35680378 PMCID: PMC9827509 DOI: 10.1124/jpet.122.001234] [Citation(s) in RCA: 106] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 05/10/2022] [Accepted: 05/12/2022] [Indexed: 01/26/2023] Open
Abstract
RNA interference (RNAi) provides researchers with a versatile means to modulate target gene expression. The major forms of RNAi molecules, genome-derived microRNAs (miRNAs) and exogenous small interfering RNAs (siRNAs), converge into RNA-induced silencing complexes to achieve posttranscriptional gene regulation. RNAi has proven to be an adaptable and powerful therapeutic strategy where advancements in chemistry and pharmaceutics continue to bring RNAi-based drugs into the clinic. With four siRNA medications already approved by the US Food and Drug Administration (FDA), several RNAi-based therapeutics continue to advance to clinical trials with functions that closely resemble their endogenous counterparts. Although intended to enhance stability and improve efficacy, chemical modifications may increase risk of off-target effects by altering RNA structure, folding, and biologic activity away from their natural equivalents. Novel technologies in development today seek to use intact cells to yield true biologic RNAi agents that better represent the structures, stabilities, activities, and safety profiles of natural RNA molecules. In this review, we provide an examination of the mechanisms of action of endogenous miRNAs and exogenous siRNAs, the physiologic and pharmacokinetic barriers to therapeutic RNA delivery, and a summary of the chemical modifications and delivery platforms in use. We overview the pharmacology of the four FDA-approved siRNA medications (patisiran, givosiran, lumasiran, and inclisiran) as well as five siRNAs and several miRNA-based therapeutics currently in clinical trials. Furthermore, we discuss the direct expression and stable carrier-based, in vivo production of novel biologic RNAi agents for research and development. SIGNIFICANCE STATEMENT: In our review, we summarize the major concepts of RNA interference (RNAi), molecular mechanisms, and current state and challenges of RNAi drug development. We focus our discussion on the pharmacology of US Food and Drug Administration-approved RNAi medications and those siRNAs and miRNA-based therapeutics that entered the clinical investigations. Novel approaches to producing new true biological RNAi molecules for research and development are highlighted.
Collapse
Affiliation(s)
| | - Ai-Ming Yu
- Department of Biochemistry and Molecular Medicine, University of California (UC) Davis School of Medicine, Sacramento, California
| |
Collapse
|
21
|
Shah AM, Giacca M. Small non-coding RNA therapeutics for cardiovascular disease. Eur Heart J 2022; 43:4548-4561. [PMID: 36106499 PMCID: PMC9659475 DOI: 10.1093/eurheartj/ehac463] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 07/29/2022] [Accepted: 08/11/2022] [Indexed: 01/07/2023] Open
Abstract
Novel bio-therapeutic agents that harness the properties of small, non-coding nucleic acids hold great promise for clinical applications. These include antisense oligonucleotides that inhibit messenger RNAs, microRNAs (miRNAs), or long non-coding RNAs; positive effectors of the miRNA pathway (short interfering RNAs and miRNA mimics); or small RNAs that target proteins (i.e. aptamers). These new therapies also offer exciting opportunities for cardiovascular diseases and promise to move the field towards more precise approaches based on disease mechanisms. There have been substantial advances in developing chemical modifications to improve the in vivo pharmacological properties of antisense oligonucleotides and reduce their immunogenicity. Carrier methods (e.g. RNA conjugates, polymers, and lipoplexes) that enhance cellular uptake of RNA therapeutics and stability against degradation by intracellular nucleases are also transforming the field. A number of small non-coding RNA therapies for cardiovascular indications are now approved. Moreover, there is a large pipeline of therapies in clinical development and an even larger list of putative therapies emerging from pre-clinical studies. Progress in this area is reviewed herein along with the hurdles that need to be overcome to allow a broader clinical translation.
Collapse
Affiliation(s)
- Ajay M Shah
- King’s College London, British Heart Foundation Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine and Sciences, The James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Mauro Giacca
- King’s College London, British Heart Foundation Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine and Sciences, The James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| |
Collapse
|
22
|
Li H, Zha S, Li H, Liu H, Wong KL, All AH. Polymeric Dendrimers as Nanocarrier Vectors for Neurotheranostics. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2203629. [PMID: 36084240 DOI: 10.1002/smll.202203629] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 08/01/2022] [Indexed: 06/15/2023]
Abstract
Dendrimers are polymers with well-defined 3D branched structures that are vastly utilized in various neurotheranostics and biomedical applications, particularly as nanocarrier vectors. Imaging agents can be loaded into dendrimers to improve the accuracy of diagnostic imaging processes. Likewise, combining pharmaceutical agents and anticancer drugs with dendrimers can enhance their solubility, biocompatibility, and efficiency. Practically, by modifying ligands on the surface of dendrimers, effective therapeutic and diagnostic platforms can be constructed and implemented for targeted delivery. Dendrimer-based nanocarriers also show great potential in gene delivery. Since enzymes can degrade genetic materials during their blood circulation, dendrimers exhibit promising packaging and delivery alternatives, particularly for central nervous system (CNS) treatments. The DNA and RNA encapsulated in dendrimers represented by polyamidoamine that are used for targeted brain delivery, via chemical-structural adjustments and appropriate generation, significantly improve the correlation between transfection efficiency and cytotoxicity. This article reports a comprehensive review of dendrimers' structures, synthesis processes, and biological applications. Recent progress in diagnostic imaging processes and therapeutic applications for cancers and other CNS diseases are presented. Potential challenges and future directions in the development of dendrimers, which provide the theoretical basis for their broader applications in healthcare, are also discussed.
Collapse
Affiliation(s)
- Hengde Li
- Department of Chemistry, Hong Kong Baptist University, 224 Waterloo Road, Kowloon, Hong Kong SAR, P. R. China
| | - Shuai Zha
- Department of Chemistry, Hong Kong Baptist University, 224 Waterloo Road, Kowloon, Hong Kong SAR, P. R. China
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR, P. R. China
| | - Haolan Li
- Department of Chemistry, Hong Kong Baptist University, 224 Waterloo Road, Kowloon, Hong Kong SAR, P. R. China
| | - Haitao Liu
- Department of Chemistry, Hong Kong Baptist University, 224 Waterloo Road, Kowloon, Hong Kong SAR, P. R. China
| | - Ka-Leung Wong
- Department of Chemistry, Hong Kong Baptist University, 224 Waterloo Road, Kowloon, Hong Kong SAR, P. R. China
| | - Angelo H All
- Department of Chemistry, Hong Kong Baptist University, 224 Waterloo Road, Kowloon, Hong Kong SAR, P. R. China
| |
Collapse
|
23
|
Xu J, Zheng Y, Wang L, Liu Y, Wang X, Li Y, Chi G. miR-124: A Promising Therapeutic Target for Central Nervous System Injuries and Diseases. Cell Mol Neurobiol 2022; 42:2031-2053. [PMID: 33886036 DOI: 10.1007/s10571-021-01091-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Accepted: 04/12/2021] [Indexed: 02/07/2023]
Abstract
Central nervous system injuries and diseases, such as ischemic stroke, spinal cord injury, neurodegenerative diseases, glioblastoma, multiple sclerosis, and the resulting neuroinflammation often lead to death or long-term disability. MicroRNAs are small, non-coding, single-stranded RNAs that regulate posttranscriptional gene expression in both physiological and pathological cellular processes, including central nervous system injuries and disorders. Studies on miR-124, one of the most abundant microRNAs in the central nervous system, have shown that its dysregulation is related to the occurrence and development of pathology within the central nervous system. Herein, we review the molecular regulatory functions, underlying mechanisms, and effective delivery methods of miR-124 in the central nervous system, where it is involved in pathological conditions. The review also provides novel insights into the therapeutic target potential of miR-124 in the treatment of human central nervous system injuries or diseases.
Collapse
Affiliation(s)
- Jinying Xu
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, 130000, People's Republic of China
| | - Yangyang Zheng
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, 130000, People's Republic of China
| | - Liangjia Wang
- Clinical Medical College, Jilin University, Changchun, 130000, People's Republic of China
| | - Yining Liu
- Clinical Medical College, Jilin University, Changchun, 130000, People's Republic of China
| | - Xishu Wang
- Clinical Medical College, Jilin University, Changchun, 130000, People's Republic of China
| | - Yulin Li
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, 130000, People's Republic of China.
| | - Guangfan Chi
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, 130000, People's Republic of China.
| |
Collapse
|
24
|
Panda M, Kalita E, Singh S, Kumar K, Rao A, Prajapati VK. MiRNA-SARS-CoV-2 dialogue and prospective anti-COVID-19 therapies. Life Sci 2022; 305:120761. [PMID: 35787998 PMCID: PMC9249409 DOI: 10.1016/j.lfs.2022.120761] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 06/24/2022] [Accepted: 06/28/2022] [Indexed: 02/08/2023]
Abstract
COVID-19 is a highly transmissible disease caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), affects 226 countries and continents, and has resulted in >6.2 million deaths worldwide. Despite the efforts of all scientific institutions worldwide to identify potential therapeutics, no specific drug has been approved by the FDA to treat the COVID-19 patient. SARS-CoV-2 variants of concerns make the potential of publicly known therapeutics to respond to and detect disease onset highly improbable. The quest for universal therapeutics pointed to the ability of RNA-based molecules to shield and detect the adverse effects of the COVID-19 illness. One such candidate, miRNA (microRNA), works on regulating the differential expression of the target gene post-transcriptionally. The prime focus of this review is to report the critical miRNA molecule and their regular expression in patients with COVID-19 infection and associated comorbidities. Viral and host miRNAs control the etiology of COVID-19 infection throughout the life cycle and host inflammatory response, where host miRNAs are identified as a double-edged showing as a proviral and antiviral response. The review also covered the role of viral miRNAs in mediating host cell signaling expression during disease pathology. Studying molecular interactions between the host and the SARS-CoV-2 virus during COVID-19 pathogenesis offers the chance to use miRNA-based therapeutics to reduce the severity of the illness. By utilizing an appropriate delivery vehicle, these small non-coding RNA could be envisioned as a promising biomarker in designing a practical RNAi-based treatment approach of clinical significance.
Collapse
Affiliation(s)
- Mamta Panda
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Bandarsindri, Kishangarh, Ajmer 305817, Rajasthan, India
| | - Elora Kalita
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Bandarsindri, Kishangarh, Ajmer 305817, Rajasthan, India
| | - Satyendra Singh
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Bandarsindri, Kishangarh, Ajmer 305817, Rajasthan, India
| | - Ketan Kumar
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Bandarsindri, Kishangarh, Ajmer 305817, Rajasthan, India
| | - Abhishek Rao
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Bandarsindri, Kishangarh, Ajmer 305817, Rajasthan, India
| | - Vijay Kumar Prajapati
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Bandarsindri, Kishangarh, Ajmer 305817, Rajasthan, India.
| |
Collapse
|
25
|
Kara G, Arun B, Calin GA, Ozpolat B. miRacle of microRNA-Driven Cancer Nanotherapeutics. Cancers (Basel) 2022; 14:3818. [PMID: 35954481 PMCID: PMC9367393 DOI: 10.3390/cancers14153818] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 07/28/2022] [Accepted: 08/04/2022] [Indexed: 12/23/2022] Open
Abstract
MicroRNAs (miRNAs) are non-protein-coding RNA molecules 20-25 nucleotides in length that can suppress the expression of genes involved in numerous physiological processes in cells. Accumulating evidence has shown that dysregulation of miRNA expression is related to the pathogenesis of various human diseases and cancers. Thus, stragegies involving either restoring the expression of tumor suppressor miRNAs or inhibiting overexpressed oncogenic miRNAs hold potential for targeted cancer therapies. However, delivery of miRNAs to tumor tissues is a challenging task. Recent advances in nanotechnology have enabled successful tumor-targeted delivery of miRNA therapeutics through newly designed nanoparticle-based carrier systems. As a result, miRNA therapeutics have entered human clinical trials with promising results, and they are expected to accelerate the transition of miRNAs from the bench to the bedside in the next decade. Here, we present recent perspectives and the newest developments, describing several engineered natural and synthetic novel miRNA nanocarrier formulations and their key in vivo applications and clinical trials.
Collapse
Affiliation(s)
- Goknur Kara
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
- Department of Chemistry, Biochemistry Division, Ordu University, Ordu 52200, Turkey
| | - Banu Arun
- Department of Breast Medical Oncology, MD Anderson Cancer Center, The University of Texas, Houston, TX 77030, USA
| | - George A. Calin
- Department of Translational Molecular Pathology, MD Anderson Cancer Center, The University of Texas, Houston, TX 77030, USA
| | - Bulent Ozpolat
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
- Houston Methodist Neal Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
26
|
New Insights into the Functions of MicroRNAs in Cardiac Fibrosis: From Mechanisms to Therapeutic Strategies. Genes (Basel) 2022; 13:genes13081390. [PMID: 36011301 PMCID: PMC9407613 DOI: 10.3390/genes13081390] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/16/2022] [Accepted: 08/03/2022] [Indexed: 02/06/2023] Open
Abstract
Cardiac fibrosis is a significant global health problem associated with almost all types of heart disease. Extensive cardiac fibrosis reduces tissue compliance and contributes to adverse outcomes, such as cardiomyocyte hypertrophy, cardiomyocyte apoptosis, and even heart failure. It is mainly associated with pathological myocardial remodeling, characterized by the excessive deposition of extracellular matrix (ECM) proteins in cardiac parenchymal tissues. In recent years, a growing body of evidence demonstrated that microRNAs (miRNAs) have a crucial role in the pathological development of cardiac fibrosis. More than sixty miRNAs have been associated with the progression of cardiac fibrosis. In this review, we summarized potential miRNAs and miRNAs-related regulatory mechanisms for cardiac fibrosis and discussed the potential clinical application of miRNAs in cardiac fibrosis.
Collapse
|
27
|
Sargazi S, Siddiqui B, Qindeel M, Rahdar A, Bilal M, Behzadmehr R, Mirinejad S, Pandey S. Chitosan nanocarriers for microRNA delivery and detection: A preliminary review with emphasis on cancer. Carbohydr Polym 2022; 290:119489. [DOI: 10.1016/j.carbpol.2022.119489] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 04/04/2022] [Accepted: 04/12/2022] [Indexed: 02/08/2023]
|
28
|
Role of Nano-miRNAs in Diagnostics and Therapeutics. Int J Mol Sci 2022; 23:ijms23126836. [PMID: 35743278 PMCID: PMC9223810 DOI: 10.3390/ijms23126836] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/15/2022] [Accepted: 06/17/2022] [Indexed: 12/26/2022] Open
Abstract
MicroRNAs (miRNA) are key regulators of gene expression, controlling different biological processes such as cellular development, differentiation, proliferation, metabolism, and apoptosis. The relationships between miRNA expression and the onset and progression of different diseases, such as tumours, cardiovascular and rheumatic diseases, and neurological disorders, are well known. A nanotechnology-based approach could match miRNA delivery and detection to move beyond the proof-of-concept stage. Different kinds of nanotechnologies can have a major impact on the diagnosis and treatment of miRNA-related diseases such as cancer. Developing novel methodologies aimed at clinical practice represents a big challenge for the early diagnosis of specific diseases. Within this context, nanotechnology represents a wide emerging area at the forefront of research over the last two decades, whose potential has yet to be fully attained. Nanomedicine, derived from nanotechnology, can exploit the unique properties of nanometer-sized particles for diagnostic and therapeutic purposes. Through nanomedicine, specific treatment to counteract only cancer-cell proliferation will be improved, while leaving healthy cells intact. In this review, we dissect the properties of different nanocarriers and their roles in the early detection and treatment of cancer.
Collapse
|
29
|
Li G, Zong X, Cheng Y, Xu J, Deng J, Huang Y, Ma C, Fu Q. miR-223-3p contributes to suppressing NLRP3 inflammasome activation in Streptococcus equi ssp. zooepidemicus infection. Vet Microbiol 2022; 269:109430. [PMID: 35427992 DOI: 10.1016/j.vetmic.2022.109430] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 03/21/2022] [Accepted: 04/03/2022] [Indexed: 11/20/2022]
Abstract
Streptococcus equi subsp. zooepidemicus (SEZ) is an essential pathogen in a range of species, causing a worldwide variety of diseases, such as meningitis, endocarditis, and septicaemia. Studies have shown that microRNAs (miRNAs), which regulate target genes at the post-transcriptional level, play an important regulatory role in the organism. In this study, the infection of J774A.1 murine macrophages with SEZ up-regulated NLRP3 inflammasome and downstream pathways accompanied by miR-223-3p down-regulation. Through computational prediction and experimental confirmation, we have shown that miR-223-3p directly targets the NLRP3 mRNA. Consequently, overexpression of miR-223-3p suppressed NLRP3 inflammasome activation and downstream pathways in response to SEZ infection. The miR-223-3p inhibitor exhibited the opposite effect, causing hyperactivation of NLRP3 inflammation activation and downstream pathways. Additionally, we further demonstrated that miRNA-223-3p inhibited the secretion of IL-1β and IL-18 by regulating the NLRP3/caspase-1 pathway. Furthermore, intravenous administration of miR-223-3p significantly decreased inflammation in mice in response to SEZ. In conclusion, our results demonstrated that miR-223-3p contributes to suppressing the NLRP3 inflammasome activation in SEZ infection, contributing novel evidence to identify a therapeutic target for treating SEZ.
Collapse
Affiliation(s)
- Guochao Li
- School of Life Science and Engineering, Foshan University, Guangdong 528225, China
| | - Xueqing Zong
- School of Life Science and Engineering, Foshan University, Guangdong 528225, China
| | - Yun Cheng
- School of Life Science and Engineering, Foshan University, Guangdong 528225, China
| | - Jianqi Xu
- School of Life Science and Engineering, Foshan University, Guangdong 528225, China
| | - Jingfei Deng
- School of Life Science and Engineering, Foshan University, Guangdong 528225, China
| | - Yunfei Huang
- School of Life Science and Engineering, Foshan University, Guangdong 528225, China; Foshan University Veterinary Teaching Hospital, Foshan University, Guangdong 528225, China
| | - Chunquan Ma
- School of Life Science and Engineering, Foshan University, Guangdong 528225, China; Foshan University Veterinary Teaching Hospital, Foshan University, Guangdong 528225, China
| | - Qiang Fu
- School of Life Science and Engineering, Foshan University, Guangdong 528225, China; Foshan University Veterinary Teaching Hospital, Foshan University, Guangdong 528225, China.
| |
Collapse
|
30
|
miRNAs in Cancer (Review of Literature). Int J Mol Sci 2022; 23:ijms23052805. [PMID: 35269947 PMCID: PMC8910953 DOI: 10.3390/ijms23052805] [Citation(s) in RCA: 115] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 02/27/2022] [Accepted: 03/02/2022] [Indexed: 02/07/2023] Open
Abstract
MicroRNAs (miRNAs) are short, noncoding, single-stranded RNA molecules that regulate gene expression at the post-transcriptional level by binding to mRNAs. miRNAs affect the course of processes of fundamental importance for the proper functioning of the organism. These processes include cell division, proliferation, differentiation, cell apoptosis and the formation of blood vessels. Altered expression of individual miRNAs has been shown in numerous cancers, which may indicate the oncogenic or suppressor potential of the molecules in question. This paper discusses the current knowledge about the possibility of using miRNA as a diagnostic marker and a potential target in modern anticancer therapies.
Collapse
|
31
|
Manikkath J, Jishnu PV, Wich PR, Manikkath A, Radhakrishnan R. Nanoparticulate strategies for the delivery of miRNA mimics and inhibitors in anticancer therapy and its potential utility in oral submucous fibrosis. Nanomedicine (Lond) 2022; 17:181-195. [PMID: 35014880 DOI: 10.2217/nnm-2021-0381] [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] [Indexed: 12/12/2022] Open
Abstract
MicroRNAs (miRNAs) are naturally occurring noncoding RNAs with multiple functionalities. They are dysregulated in several conditions and can serve as disease biomarkers, therapeutic targets and therapeutic agents. Translation of miRNA therapeutics to the clinic poses several challenges related to the safe and effective delivery of these agents to the site of action. Nanoparticulate carriers hold promise in this area by enhancing targeting efficiency and reducing off-target effects. This paper reviews recent advances in the delivery strategies of miRNAs in anticancer therapy, with a focus on lipid-based, polymeric, inorganic platforms, cell membrane-derived vesicles and bacterial minicells. Additionally, this review explores the potentiality of miRNAs in the treatment of oral submucous fibrosis, a potentially premalignant condition of the oral cavity with no definitive treatment to date.
Collapse
Affiliation(s)
- Jyothsna Manikkath
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Padacherri Vethil Jishnu
- Department of Cell & Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Peter R Wich
- School of Chemical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Aparna Manikkath
- Arthur A. Dugoni School of Dentistry, University of the Pacific, San Francisco, CA 94103, USA
| | - Raghu Radhakrishnan
- Department of Oral Pathology, Manipal College of Dental Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| |
Collapse
|
32
|
Yan C, Chen J, Wang C, Yuan M, Kang Y, Wu Z, Li W, Zhang G, Machens HG, Rinkevich Y, Chen Z, Yang X, Xu X. Milk exosomes-mediated miR-31-5p delivery accelerates diabetic wound healing through promoting angiogenesis. Drug Deliv 2022; 29:214-228. [PMID: 34985397 PMCID: PMC8741248 DOI: 10.1080/10717544.2021.2023699] [Citation(s) in RCA: 114] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The refractory diabetic wound has remained a worldwide challenge as one of the major health problems. The impaired angiogenesis phase during diabetic wound healing partly contributes to the pathological process. MicroRNA (miRNA) is an essential regulator of gene expression in crucial biological processes and is a promising nucleic acid drug in therapeutic fields of the diabetic wound. However, miRNA therapies have limitations due to lacking an effective delivery system. In the present study, we found a significant reduction of miR-31-5p expression in the full-thickness wounds of diabetic mice compared to normal mice. Further, miR-31-5p has been proven to promote the proliferation, migration, and angiogenesis of endothelial cells. Thus, we conceived the idea of exogenously supplementing miR-31-5p mimics to treat the diabetic wound. We used milk-derived exosomes as a novel system for miR-31-5p delivery and successfully encapsulated miR-31-5p mimics into milk exosomes through electroporation. Then, we proved that the miR-31-5p loaded in exosomes achieved higher cell uptake and was able to resist degradation. Moreover, our miRNA-exosomal formulation demonstrated dramatically improved endothelial cell functions in vitro, together with the promotion of angiogenesis and enhanced diabetic wound healing in vivo. Collectively, our data showed the feasibility of milk exosomes as a scalable, biocompatible, and cost-effective delivery system to enhance the bioavailability and efficacy of miRNAs.
Collapse
Affiliation(s)
- Chengqi Yan
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Chen
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cheng Wang
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Meng Yuan
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Kang
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zihan Wu
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenqing Li
- Department of Hand and Foot Surgery, Huazhong University of Science and Technology, Union Shenzhen Hospital, Shenzhen, China
| | - Guolei Zhang
- Department of Hand and Foot Surgery, Huazhong University of Science and Technology, Union Shenzhen Hospital, Shenzhen, China
| | - Hans-Günther Machens
- Department of Plastic and Hand Surgery, Technical University of Munich, Munich, Germany
| | - Yuval Rinkevich
- Institute of Lung Biology and Disease, Helmholtz Zentrum München, Munich, Germany.,Institute of Regenerative Biology and Medicine, Helmholtz Zentrum München, Munich, Germany
| | - Zhenbing Chen
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaofan Yang
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiang Xu
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
33
|
Bautista-Becerril B, Pérez-Dimas G, Sommerhalder-Nava PC, Hanono A, Martínez-Cisneros JA, Zarate-Maldonado B, Muñoz-Soria E, Aquino-Gálvez A, Castillejos-López M, Juárez-Cisneros A, Lopez-Gonzalez JS, Camarena A. miRNAs, from Evolutionary Junk to Possible Prognostic Markers and Therapeutic Targets in COVID-19. Viruses 2021; 14:41. [PMID: 35062245 PMCID: PMC8781105 DOI: 10.3390/v14010041] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Accepted: 12/21/2021] [Indexed: 01/08/2023] Open
Abstract
The COVID-19 pandemic has been a public health issue around the world in the last few years. Currently, there is no specific antiviral treatment to fight the disease. Thus, it is essential to highlight possible prognostic predictors that could identify patients with a high risk of developing complications. Within this framework, miRNA biomolecules play a vital role in the genetic regulation of various genes, principally, those related to the pathophysiology of the disease. Here, we review the interaction of host and viral microRNAs with molecular and cellular elements that could potentiate the main pulmonary, cardiac, renal, circulatory, and neuronal complications in COVID-19 patients. miR-26a, miR-29b, miR-21, miR-372, and miR-2392, among others, have been associated with exacerbation of the inflammatory process, increasing the risk of a cytokine storm. In addition, increased expression of miR-15b, -199a, and -491 are related to the prognosis of the disease, and miR-192 and miR-323a were identified as clinical predictors of mortality in patients admitted to the intensive care unit. Finally, we address miR-29, miR-122, miR-155, and miR-200, among others, as possible therapeutic targets. However, more studies are required to confirm these findings.
Collapse
Affiliation(s)
- Brandon Bautista-Becerril
- Laboratorio HLA, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (B.B.-B.); (A.J.-C.)
- Escuela Superior de Medicina, Departamento de Posgrado, Instituto Politécnico Nacional, Mexico City 11340, Mexico; (G.P.-D.); (E.M.-S.)
| | - Guillermo Pérez-Dimas
- Escuela Superior de Medicina, Departamento de Posgrado, Instituto Politécnico Nacional, Mexico City 11340, Mexico; (G.P.-D.); (E.M.-S.)
| | - Paola C. Sommerhalder-Nava
- Facultad de Ciencias de la Salud, Universidad Anáhuac México Norte, Mexico City 52786, Mexico; (P.C.S.-N.); (A.H.); (B.Z.-M.)
| | - Alejandro Hanono
- Facultad de Ciencias de la Salud, Universidad Anáhuac México Norte, Mexico City 52786, Mexico; (P.C.S.-N.); (A.H.); (B.Z.-M.)
| | | | - Bárbara Zarate-Maldonado
- Facultad de Ciencias de la Salud, Universidad Anáhuac México Norte, Mexico City 52786, Mexico; (P.C.S.-N.); (A.H.); (B.Z.-M.)
| | - Evangelina Muñoz-Soria
- Escuela Superior de Medicina, Departamento de Posgrado, Instituto Politécnico Nacional, Mexico City 11340, Mexico; (G.P.-D.); (E.M.-S.)
| | - Arnoldo Aquino-Gálvez
- Laboratorio de Biología Molecular, Departamento de Fibrosis Pulmonar, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico;
| | - Manuel Castillejos-López
- Departamento de Epidemiología Hospitalaria e Infectología, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico;
| | - Armida Juárez-Cisneros
- Laboratorio HLA, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (B.B.-B.); (A.J.-C.)
| | - Jose S. Lopez-Gonzalez
- Laboratorio de Cáncer Pulmonar, Departamento de Enfermedades Crónico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico;
| | - Angel Camarena
- Laboratorio HLA, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (B.B.-B.); (A.J.-C.)
| |
Collapse
|
34
|
Paluszkiewicz P, Martuszewski A, Zaręba N, Wala K, Banasik M, Kepinska M. The Application of Nanoparticles in Diagnosis and Treatment of Kidney Diseases. Int J Mol Sci 2021; 23:ijms23010131. [PMID: 35008556 PMCID: PMC8745391 DOI: 10.3390/ijms23010131] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/16/2021] [Accepted: 12/21/2021] [Indexed: 12/12/2022] Open
Abstract
Nanomedicine is currently showing great promise for new methods of diagnosing and treating many diseases, particularly in kidney disease and transplantation. The unique properties of nanoparticles arise from the diversity of size effects, used to design targeted nanoparticles for specific cells or tissues, taking renal clearance and tubular secretion mechanisms into account. The design of surface particles on nanoparticles offers a wide range of possibilities, among which antibodies play an important role. Nanoparticles find applications in encapsulated drug delivery systems containing immunosuppressants and other drugs, in imaging, gene therapies and many other branches of medicine. They have the potential to revolutionize kidney transplantation by reducing and preventing ischemia-reperfusion injury, more efficiently delivering drugs to the graft site while avoiding systemic effects, accurately localizing and visualising the diseased site and enabling continuous monitoring of graft function. So far, there are known nanoparticles with no toxic effects on human tissue, although further studies are still needed to confirm their safety.
Collapse
Affiliation(s)
- Patrycja Paluszkiewicz
- Department of Emergency Medical Service, Wroclaw Medical University, Bartla 5, 50-367 Wroclaw, Poland;
| | - Adrian Martuszewski
- Department of Population Health, Division of Environmental Health and Occupational Medicine, Wroclaw Medical University, Mikulicza-Radeckiego 7, 50-368 Wroclaw, Poland;
| | - Natalia Zaręba
- Department of Pharmaceutical Biochemistry, Division of Biomedical and Environmental Analysis, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211a, 50-556 Wrocław, Poland;
| | - Kamila Wala
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland;
| | - Mirosław Banasik
- Department of Nephrology and Transplantation Medicine, Wroclaw Medical University, Borowska 213, 50-556 Wroclaw, Poland
- Correspondence: (M.B.); (M.K.); Tel.: +48-71-733-2500 (M.B.); +48-71-784-0171 (M.K.)
| | - Marta Kepinska
- Department of Pharmaceutical Biochemistry, Division of Biomedical and Environmental Analysis, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211a, 50-556 Wrocław, Poland;
- Correspondence: (M.B.); (M.K.); Tel.: +48-71-733-2500 (M.B.); +48-71-784-0171 (M.K.)
| |
Collapse
|
35
|
Challenges for the Development of Extracellular Vesicle-Based Nucleic Acid Medicines. Cancers (Basel) 2021; 13:cancers13236137. [PMID: 34885247 PMCID: PMC8656933 DOI: 10.3390/cancers13236137] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/29/2021] [Accepted: 12/01/2021] [Indexed: 12/12/2022] Open
Abstract
Nucleic acid drugs, such as siRNAs, antisense oligonucleotides, and miRNAs, exert their therapeutic effects by causing genetic changes in cells. However, there are various limitations in their delivery to target organs and cells, making their application to cancer treatment difficult. Extracellular vesicles (EVs) are lipid bilayer particles that are released from most cells, are stable in the blood, and have low immunogenicity. Methods using EVs to deliver nucleic acid drugs to target organs are rapidly being developed that take advantage of these properties. There are two main methods for loading nucleic acid drugs into EVs. One is to genetically engineer the parent cell and load the target gene into the EV, and the other is to isolate EVs and then load them with the nucleic acid drug. Target organ delivery methods include passive targeting using the enhanced permeation and retention effect of EVs and active targeting in which EVs are modified with antibodies, peptides, or aptamers to enhance their accumulation in tumors. In this review, we summarize the advantages of EVs as a drug delivery system for nucleic acid drugs, the methods of loading nucleic acid drugs into EVs, and the targeting of EVs to target organs.
Collapse
|
36
|
Johnson TF, Jones K, Iacoviello F, Turner S, Jackson NB, Zourna K, Welsh JH, Shearing PR, Hoare M, Bracewell DG. Liposome Sterile Filtration Characterization via X-ray Computed Tomography and Confocal Microscopy. MEMBRANES 2021; 11:membranes11110905. [PMID: 34832134 PMCID: PMC8620169 DOI: 10.3390/membranes11110905] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 11/16/2021] [Accepted: 11/19/2021] [Indexed: 12/12/2022]
Abstract
Two high resolution, 3D imaging techniques were applied to visualize and characterize sterilizing grade dual-layer filtration of liposomes, enabling membrane structure to be related with function and performance. Two polyethersulfone membranes with nominal retention ratings of 650 nm and 200 nm were used to filter liposomes of an average diameter of 143 nm and a polydispersity index of 0.1. Operating conditions including differential pressure were evaluated. X-ray computed tomography at a pixel size of 63 nm was capable of resolving the internal geometry of each membrane. The respective asymmetry and symmetry of the upstream and downstream membranes could be measured, with pore network modeling used to identify pore sizes as a function of distance through the imaged volume. Reconstructed 3D digital datasets were the basis of tortuous flow simulation through each porous structure. Confocal microscopy visualized liposome retention within each membrane using fluorescent dyes, with bacterial challenges also performed. It was found that increasing pressure drop from 0.07 MPa to 0.21 MPa resulted in differing fluorescent retention profiles in the upstream membrane. These results highlighted the capability for complementary imaging approaches to deepen understanding of liposome sterilizing grade filtration.
Collapse
Affiliation(s)
- Thomas F. Johnson
- Department of Biochemical Engineering, University College London, Bernard Katz, London WC1E 6BT, UK; (T.F.J.); (M.H.)
| | - Kyle Jones
- Pall Corporation 5 Harbourgate Business Park, Southampton Road, Portsmouth PO6 4BQ, UK; (K.J.); (S.T.); (N.B.J.); (K.Z.); (J.H.W.)
| | - Francesco Iacoviello
- Electrochemical Innovation Laboratory, Department of Chemical Engineering, University College London, Torrington Place, London WC1E 7JE, UK; (F.I.); (P.R.S.)
| | - Stephen Turner
- Pall Corporation 5 Harbourgate Business Park, Southampton Road, Portsmouth PO6 4BQ, UK; (K.J.); (S.T.); (N.B.J.); (K.Z.); (J.H.W.)
| | - Nigel B. Jackson
- Pall Corporation 5 Harbourgate Business Park, Southampton Road, Portsmouth PO6 4BQ, UK; (K.J.); (S.T.); (N.B.J.); (K.Z.); (J.H.W.)
| | - Kalliopi Zourna
- Pall Corporation 5 Harbourgate Business Park, Southampton Road, Portsmouth PO6 4BQ, UK; (K.J.); (S.T.); (N.B.J.); (K.Z.); (J.H.W.)
| | - John H. Welsh
- Pall Corporation 5 Harbourgate Business Park, Southampton Road, Portsmouth PO6 4BQ, UK; (K.J.); (S.T.); (N.B.J.); (K.Z.); (J.H.W.)
| | - Paul R. Shearing
- Electrochemical Innovation Laboratory, Department of Chemical Engineering, University College London, Torrington Place, London WC1E 7JE, UK; (F.I.); (P.R.S.)
| | - Mike Hoare
- Department of Biochemical Engineering, University College London, Bernard Katz, London WC1E 6BT, UK; (T.F.J.); (M.H.)
| | - Daniel G. Bracewell
- Department of Biochemical Engineering, University College London, Bernard Katz, London WC1E 6BT, UK; (T.F.J.); (M.H.)
- Correspondence: ; Tel.: +44-20-7679-2374
| |
Collapse
|
37
|
miRNA Delivery by Nanosystems: State of the Art and Perspectives. Pharmaceutics 2021; 13:pharmaceutics13111901. [PMID: 34834316 PMCID: PMC8619868 DOI: 10.3390/pharmaceutics13111901] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 10/25/2021] [Accepted: 11/04/2021] [Indexed: 01/09/2023] Open
Abstract
MicroRNAs (miRNAs) are short (~21-23 nucleotides), non-coding endogenous RNA molecules that modulate gene expression at the post-transcriptional level via the endogenous RNA interference machinery of the cell. They have emerged as potential biopharmaceuticals candidates for the treatment of various diseases, including cancer, cardiovascular and metabolic diseases. However, in order to advance miRNAs therapeutics into clinical settings, their delivery remains a major challenge. Different types of vectors have been investigated to allow the delivery of miRNA in the diseased tissue. In particular, non-viral delivery systems have shown important advantages such as versatility, low cost, easy fabrication and low immunogenicity. Here, we present a general overview of the main types of non-viral vectors developed for miRNA delivery, with their advantages, limitations and future perspectives.
Collapse
|
38
|
Bartsch B, Goody PR, Hosen MR, Nehl D, Mohammadi N, Zietzer A, Düsing P, Pfeifer A, Nickenig G, Jansen F. NcRNAs in Vascular and Valvular Intercellular Communication. Front Mol Biosci 2021; 8:749681. [PMID: 34805273 PMCID: PMC8602872 DOI: 10.3389/fmolb.2021.749681] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 09/23/2021] [Indexed: 12/05/2022] Open
Abstract
Non-coding RNAs have been shown to be important biomarkers and mediators of many different disease entities, including cardiovascular (CV) diseases like atherosclerosis, aneurysms, and valvulopathies. Growing evidence suggests a central role of ncRNAs as regulators of different pathological pathways involved in endothelial dysfunction, cardiovascular inflammation, cell differentiation, and calcification. This review will discuss the role of protein-bound and extracellular vesicular-bound ncRNAs as biomarkers of vascular and valvular diseases, their role as intercellular communicators, and regulators of disease pathways and also highlights possible treatment strategies.
Collapse
Affiliation(s)
- Benedikt Bartsch
- Department of Internal Medicine II, Heart Center Bonn, University Hospital Bonn, Bonn, Germany
| | - Philip Roger Goody
- Department of Internal Medicine II, Heart Center Bonn, University Hospital Bonn, Bonn, Germany
| | - Mohammed Rabiul Hosen
- Department of Internal Medicine II, Heart Center Bonn, University Hospital Bonn, Bonn, Germany
| | - Denise Nehl
- Department of Internal Medicine II, Heart Center Bonn, University Hospital Bonn, Bonn, Germany
| | - Neda Mohammadi
- Institute of Pharmacology and Toxicology, University Hospital Bonn, Bonn, Germany
| | - Andreas Zietzer
- Department of Internal Medicine II, Heart Center Bonn, University Hospital Bonn, Bonn, Germany
| | - Philip Düsing
- Department of Internal Medicine II, Heart Center Bonn, University Hospital Bonn, Bonn, Germany
| | - Alexander Pfeifer
- Institute of Pharmacology and Toxicology, University Hospital Bonn, Bonn, Germany
| | - Georg Nickenig
- Department of Internal Medicine II, Heart Center Bonn, University Hospital Bonn, Bonn, Germany
| | - Felix Jansen
- Department of Internal Medicine II, Heart Center Bonn, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
39
|
Zeng X, Li Z, Zhu C, Xu L, Sun Y, Han S. Research progress of nanocarriers for gene therapy targeting abnormal glucose and lipid metabolism in tumors. Drug Deliv 2021; 28:2329-2347. [PMID: 34730054 PMCID: PMC8567922 DOI: 10.1080/10717544.2021.1995081] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
In recent years, the incidence of various types of tumors has gradually increased, and it has also been found that there is a certain correlation between abnormal glucose and lipid metabolism and tumors. Glycolipid metabolism can promote tumor progression through multiple pathways, and the expression of related genes also directly or indirectly affects tumor metabolism, metastasis, invasion, and apoptosis. There has been much research on targeted drug delivery systems designed for abnormal glucose and lipid metabolism due to their accuracy and efficiency when used for tumor therapy. In addition, gene mutations have become an important factor in tumorigenesis. For this reason, gene therapy consisting of drugs designed for certain specifically expressed genes have been transfected into target cells to express or silence the corresponding proteins. Targeted gene drug vectors that achieve their corresponding therapeutic purposes are also rapidly developing. The genes related to glucose and lipid metabolism are considered as the target, and a corresponding gene drug carrier is constructed to influence and interfere with the expression of related genes, so as to block the tumorigenesis process and inhibit tumor growth. Designing drugs that target genes related to glucose and lipid metabolism within tumors is considered to be a promising strategy for the treatment of tumor diseases. This article summarizes the chemical drugs/gene drug delivery systems and the corresponding methods used in recent years for the treatment of abnormal glucose and lipid metabolism of tumors, and provides a theoretical basis for the development of glucolipid metabolism related therapeutic methods.
Collapse
Affiliation(s)
- Xianhu Zeng
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao, China
| | - Zhipeng Li
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao, China
| | - Chunrong Zhu
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao, China
| | - Lisa Xu
- School of Public Health, Qingdao University, Qingdao, China
| | - Yong Sun
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao, China
| | - Shangcong Han
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao, China
| |
Collapse
|
40
|
Hussen BM, Abdullah ST, Rasul MF, Salihi A, Ghafouri-Fard S, Hidayat HJ, Taheri M. MicroRNAs: Important Players in Breast Cancer Angiogenesis and Therapeutic Targets. Front Mol Biosci 2021; 8:764025. [PMID: 34778378 PMCID: PMC8582349 DOI: 10.3389/fmolb.2021.764025] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 10/13/2021] [Indexed: 12/11/2022] Open
Abstract
The high incidence of breast cancer (BC) is linked to metastasis, facilitated by tumor angiogenesis. MicroRNAs (miRNAs or miRs) are small non-coding RNA molecules that have an essential role in gene expression and are significantly linked to the tumor development and angiogenesis process in different types of cancer, including BC. There's increasing evidence showed that various miRNAs play a significant role in disease processes; specifically, they are observed and over-expressed in a wide range of diseases linked to the angiogenesis process. However, more studies are required to reach the best findings and identify the link among miRNA expression, angiogenic pathways, and immune response-related genes to find new therapeutic targets. Here, we summarized the recent updates on miRNA signatures and their cellular targets in the development of breast tumor angiogenetic and discussed the strategies associated with miRNA-based therapeutic targets as anti-angiogenic response.
Collapse
Affiliation(s)
- Bashdar Mahmud Hussen
- Department of Pharmacognosy, College of Pharmacy, Hawler Medical University, Erbil, Iraq
| | - Sara Tharwat Abdullah
- Department of Pharmacology and Toxicology, College of Pharmacy, Hawler Medical University, Erbil, Iraq
| | - Mohammed Fatih Rasul
- Department of Medical Analysis, Faculty of Science, Tishk International University-Erbil, Erbil, Iraq
| | - Abbas Salihi
- Department of Biology, College of Science, Salahaddin University-Erbil, Erbil, Iraq
- Center of Research and Strategic Studies, Lebanese French University, Erbil, Iraq
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hazha Jamal Hidayat
- Department of Biology, College of Education, Salahaddin University-Erbil, Erbil, Iraq
| | - Mohammad Taheri
- Skull Base Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Institute of Human Genetics, Jena University Hospital, Jena, Germany
| |
Collapse
|
41
|
Cao C, Shu X. Suppression of circ_0008932 inhibits tumor growth and metastasis in osteosarcoma by targeting miR-145-5p. Exp Ther Med 2021; 22:1106. [PMID: 34504560 PMCID: PMC8383749 DOI: 10.3892/etm.2021.10540] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Accepted: 06/17/2021] [Indexed: 12/21/2022] Open
Abstract
Osteosarcoma (OS) is a common type of primary malignant tumor. Although the pathogenesis of OS has been extensively studied, the underlying molecular mechanisms have remained to be fully elucidated. Accumulating evidence has revealed that dysregulation of various circular RNAs (circRNAs) is associated with tumorigenesis and recent studies have indicated that circRNA circ_0008932 is aberrantly expressed in tumors. In the present study, the expression and detailed function of circ_0008932 in OS were elucidated. The levels of circ_0008932 in OS samples and cell lines were examined using reverse transcription-quantitative PCR. A cell model with circ_0008932 knockdown was generated using specific small interfering RNA (si-circ_0008932). Cell viability was determined by a Cell Counting Kit-8 assay, the cell migratory/invasive capacity was evaluated using Transwell assays and cell apoptosis was assessed by flow cytometry. The results suggested that circ_0008932 was upregulated in most primary OS tumors, suggesting that circ_0008932 is associated with the development of OS. In the in vitro assays, si-circ_0008932 inhibited the proliferation, migration and invasion of OS cells, while apoptosis was promoted. A luciferase reporter assay revealed that circ_0008932 may downregulate microRNA (miR)-145-5p through direct binding. Furthermore, the expression of miR-145-5p was negatively correlated with circ_0008932 levels in OS specimens. In addition, further functional studies indicated that miR-145-5p inhibitors eliminated the effects caused by si-circ_0008932 in OS cells. In comparison, the changes in the biological behavior of OS cells transfected with si-circ_0008932 were enhanced by miR-145-5p. In summary, circ_0008932 may be a novel oncogenic factor during the progression and development of OS by targeting miR-145-5p; more importantly, circ_0008932 may be a potential therapeutic target for OS.
Collapse
Affiliation(s)
- Chenggang Cao
- Department of Orthopedics, Chongqing Traditional Chinese Medicine Hospital/Chongqing First People's Hospital, Chongqing 400011, P.R. China
| | - Xiaolei Shu
- Department of Radiation Oncology, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing 400030, P.R. China
| |
Collapse
|
42
|
MicroRNA-375: potential cancer suppressor and therapeutic drug. Biosci Rep 2021; 41:229736. [PMID: 34494089 PMCID: PMC8458691 DOI: 10.1042/bsr20211494] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 08/31/2021] [Accepted: 09/06/2021] [Indexed: 02/06/2023] Open
Abstract
MiR-375 is a conserved noncoding RNA that is known to be involved in tumor cell proliferation, migration, and drug resistance. Previous studies have shown that miR-375 affects the epithelial-mesenchymal transition (EMT) of human tumor cells via some key transcription factors, such as Yes-associated protein 1 (YAP1), Specificity protein 1 (SP1) and signaling pathways (Wnt signaling pathway, nuclear factor κB (NF-κB) pathway and transforming growth factor β (TGF-β) signaling pathway) and is vital for the development of cancer. Additionally, recent studies have identified microRNA (miRNA) delivery system carriers for improved in vivo transportation of miR-375 to specific sites. Here, we discussed the role of miR-375 in different types of cancers, as well as molecular mechanisms, and analyzed the potential of miR-375 as a molecular biomarker and therapeutic target to improve the efficiency of clinical diagnosis of cancer.
Collapse
|
43
|
Application of Non-Viral Vectors in Drug Delivery and Gene Therapy. Polymers (Basel) 2021; 13:polym13193307. [PMID: 34641123 PMCID: PMC8512075 DOI: 10.3390/polym13193307] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/15/2021] [Accepted: 09/18/2021] [Indexed: 12/13/2022] Open
Abstract
Vectors and carriers play an indispensable role in gene therapy and drug delivery. Non-viral vectors are widely developed and applied in clinical practice due to their low immunogenicity, good biocompatibility, easy synthesis and modification, and low cost of production. This review summarized a variety of non-viral vectors and carriers including polymers, liposomes, gold nanoparticles, mesoporous silica nanoparticles and carbon nanotubes from the aspects of physicochemical characteristics, synthesis methods, functional modifications, and research applications. Notably, non-viral vectors can enhance the absorption of cargos, prolong the circulation time, improve therapeutic effects, and provide targeted delivery. Additional studies focused on recent innovation of novel synthesis techniques for vector materials. We also elaborated on the problems and future research directions in the development of non-viral vectors, which provided a theoretical basis for their broad applications.
Collapse
|
44
|
Zhang R, Liu P, Zhang X, Ye Y, Yu J. Lin28A promotes the proliferation and stemness of lung cancer cells via the activation of mitogen-activated protein kinase pathway dependent on microRNA let-7c. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:982. [PMID: 34277782 PMCID: PMC8267304 DOI: 10.21037/atm-21-2124] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 06/04/2021] [Indexed: 12/20/2022]
Abstract
Background Among patients with lung cancer, metastatic and relapsed cases account for the largest proportion of disease-associated deaths. Tumor metastasis and relapse are believed to originate from cancer stem cells (CSCs), which have the capacity to be highly proliferative and invasive. In our previous studies, we established a conditional basement membrane extract-based (BME-based) 3-dimensional (3D) culture system to mimic the tumor growth environment in vivo and further amplified lung cancer stem cells (LCSCs) in our system. However, the molecular mechanisms of LCSC amplification and development in our 3D culture system have not been fully uncovered. Method We established the conditional 3D culture system to amplify LCSCs in other lung cancer cell lines, followed by examining the expression of Lin28A and let-7 microRNAs in them. We also explored the expression of Lin28A and let-7 microRNAs in LCSCs from clinical lung cancer tissue samples and even analyzed the correlation of Lin28A/let-7c and patients’ survival outcomes. We further constructed A549 cells either knockdown of Lin28A or overexpression of let-7c, followed by investigating stemness marker gene expression, and stemness phenotypes including mammosphere culture, cell migration and invasion in vitro, as well as tumorigenicity in vivo. Results Here, we observed that Lin28A/let-7c was dysregulated in LCSCs in both the 3D culture system and lung cancer tissues. Further, the abnormal expression of Lin28A/let-7c was correlated with poor survival outcomes. Via the construction of A549 cells with let-7c over-expression, we found that let-7c inhibited the maintenance of LCSC properties, while the results of Lin28A knockdown showed that Lin28A played a critical role in the enrichment and proliferation of LCSCs via mitogen-activated protein kinase (MAPK) signaling pathway. Importantly, we found that LCSCs with knockdown of Lin28A or over-expression of let-7c exhibited inhibited carcinogenesis and disrupted expansion in vivo. Conclusions Our study uncovered the functions and mechanisms of the Lin28A/let-7c/MAPK signaling pathway in promoting the proliferation and cancer stemness of LCSCs, which might be a potential therapeutic target for reducing and even eliminating LCSCs in the future.
Collapse
Affiliation(s)
- Rui Zhang
- Cancer Molecular Diagnostics Core, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Pengpeng Liu
- Cancer Molecular Diagnostics Core, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Xiao Zhang
- Cancer Molecular Diagnostics Core, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Yingnan Ye
- Cancer Molecular Diagnostics Core, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Jinpu Yu
- Cancer Molecular Diagnostics Core, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China
| |
Collapse
|
45
|
Bai Z, Zhou Q, Zhu H, Ye X, Wu P, Ma L. QTMP, a Novel Thiourea Polymer, Causes DNA Damage to Exert Anticancer Activity and Overcome Multidrug Resistance in Colorectal Cancer Cells. Front Oncol 2021; 11:667689. [PMID: 34123833 PMCID: PMC8194350 DOI: 10.3389/fonc.2021.667689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Accepted: 05/07/2021] [Indexed: 11/13/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most common malignancies, and multidrug resistance (MDR) severely restricts the effectiveness of various anticancer drugs. Therefore, the development of novel anticancer drugs for the treatment of CRC patients with MDR is necessary. Quaternized thiourea main-chain polymer (QTMP) is a self-assembled nanoparticle with good water solubility. Notably, QTMP is not a P-glycoprotein (P-gp) substrate, and it exhibits potent cytotoxic activity against CRC cells, including HCT116/DDP and P-gp-mediated multidrug-resistant Caco2 cells. QTMP also exhibits a strong anticancer activity against SW480 cells in vivo. Interestingly, reactive oxygen species (ROS) and reactive nitrogen species (RNS) production were increased in a concentration-dependent manner in QTMP-treated HCT116, SW480 and Caco2 cells. Importantly, QTMP causes DNA damage in these CRC cells via direct insertion into the DNA or regulation of ROS and/or RNS production. QTMP also induces caspase-dependent apoptosis via overproduction of ROS and RNS. Therefore, QTMP is a promising anticancer therapeutic agent for patients with CRC, including those cancer cells with P-gp-mediated MDR. The present study also indicates that the design and synthesis of anticancer drugs based on thiourea polymers is promising and valuable, thereby offering a new strategy to address MDR, and provides reference resources for further investigations of thiourea polymers.
Collapse
Affiliation(s)
- Zhaoshi Bai
- Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & the Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Qing Zhou
- Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & the Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Huayun Zhu
- Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & the Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Xinyue Ye
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Pingping Wu
- Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & the Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Lingman Ma
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
46
|
Kang T, Ni JS, Li T, Wang J, Li Z, Li Y, Zha M, Zhang C, Wu X, Guo H, Xi L, Li K. Efficient and precise delivery of microRNA by photoacoustic force generated from semiconducting polymer-based nanocarriers. Biomaterials 2021; 275:120907. [PMID: 34090050 DOI: 10.1016/j.biomaterials.2021.120907] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 05/16/2021] [Accepted: 05/20/2021] [Indexed: 12/14/2022]
Abstract
One major challenge in miRNA-based therapy is to explore facile delivery strategies, which can facilitate the efficient and precise accumulation of intrinsically instable microRNAs (miRNAs) at targeted tumor sites. To address this critical issue, for the first time we demonstrate that a near-infrared (NIR) pulse laser can guide efficient delivery of miRNAs mediated by a NIR-absorbing and photoacoustic active semiconducting polymer (SP) nanocarrier, which can generate photoacoustic radiation force to intravascularly overcome the endothelial barriers. Importantly, we demonstrate an ultrafast delivery of miRNA (miR-7) to tumor tissues under the irradiation of pulse laser in 20 min, showing a 5-fold boosted efficiency in comparison to the traditional passive targeting strategy. The delivered miR-7 acts as a sensitizer of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and synergizes with TRAIL-inducing compound (TIC), leading to sustained TRAIL upregulation for effective tumor suppression in mice. As such, our results indicate that the NIR-absorbing semiconducting polymer-mediated nanocarrier platform can significantly enhance the targeted delivery efficiency of therapeutic miRNAs to tumors, resulting in potent tumor growth inhibition.
Collapse
Affiliation(s)
- Tianyi Kang
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Department of Biomedical Engineering, Southern University of Science and Technology (SUSTech), Shenzhen, 518055, China
| | - Jen-Shyang Ni
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Department of Biomedical Engineering, Southern University of Science and Technology (SUSTech), Shenzhen, 518055, China
| | - Tingting Li
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Department of Biomedical Engineering, Southern University of Science and Technology (SUSTech), Shenzhen, 518055, China
| | - Jun Wang
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Department of Biomedical Engineering, Southern University of Science and Technology (SUSTech), Shenzhen, 518055, China
| | - Zeshun Li
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Department of Biomedical Engineering, Southern University of Science and Technology (SUSTech), Shenzhen, 518055, China
| | - Yaxi Li
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Department of Biomedical Engineering, Southern University of Science and Technology (SUSTech), Shenzhen, 518055, China
| | - Menglei Zha
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Department of Biomedical Engineering, Southern University of Science and Technology (SUSTech), Shenzhen, 518055, China
| | - Chen Zhang
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Department of Biomedical Engineering, Southern University of Science and Technology (SUSTech), Shenzhen, 518055, China
| | - Xue Wu
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Department of Biomedical Engineering, Southern University of Science and Technology (SUSTech), Shenzhen, 518055, China
| | - Heng Guo
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Department of Biomedical Engineering, Southern University of Science and Technology (SUSTech), Shenzhen, 518055, China
| | - Lei Xi
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Department of Biomedical Engineering, Southern University of Science and Technology (SUSTech), Shenzhen, 518055, China
| | - Kai Li
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Department of Biomedical Engineering, Southern University of Science and Technology (SUSTech), Shenzhen, 518055, China.
| |
Collapse
|
47
|
Li F, Liang Y, Wang M, Xu X, Zhao F, Wang X, Sun Y, Chen W. Multifunctional nanoplatforms as cascade-responsive drug-delivery carriers for effective synergistic chemo-photodynamic cancer treatment. J Nanobiotechnology 2021; 19:140. [PMID: 34001157 PMCID: PMC8130269 DOI: 10.1186/s12951-021-00876-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 04/28/2021] [Indexed: 12/19/2022] Open
Abstract
Synergistic chemo-photodynamic therapy has garnered attention in the field of cancer treatment. Here, a pH cascade-responsive micellar nanoplatform with nucleus-targeted ability, for effective synergistic chemo-photodynamic cancer treatment, was fabricated. In this micellar nanoplatform, 5-(4-carboxyphenyl)-10,15,20-triphenylporphyrin (Por), a photodynamic therapy (PDT) agent was utilized for carrying the novel anticancer drug GNA002 to construct a hydrophobic core, and cyclic RGD peptide (cRGD)-modified polyethylene glycol (PEG) (cRGD-PEG) connected the cell-penetrating peptide hexaarginine (R6) through a pH-responsive hydrazone bond (cRGD-PEG-N = CH-R6) to serve as a hydrophilic shell for increasing blood circulation time. After passively accumulating in tumor sites, the self-assembled GNA002-loaded nanoparticles were actively internalized into cancer cells via the cRGD ligands. Once phagocytosed by lysosomes, the acidity-triggered detachment of the cRGD-PEG shell led to the formation of R6-coated secondary nanoparticles and subsequent R6-mediated nucleus-targeted drug delivery. Combined with GNA002-induced nucleus-specific chemotherapy, reactive oxygen species produced by Por under 532-nm laser irradiation achieved a potent synergistic chemo-photodynamic cancer treatment. Moreover, our in vitro and in vivo anticancer investigations revealed high cancer-suppression efficacy of this ideal multifunctional nanoplatform, indicating that it could be a promising candidate for synergistic anticancer therapy.![]()
Collapse
Affiliation(s)
- Fan Li
- Department of Oral and Maxillofacial Head & Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.,Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai, 200011, China
| | - Yan Liang
- Department of Pharmaceutics, Qingdao University School of Pharmacy, Qingdao, 266021, China
| | - Miaochen Wang
- Department of Oral and Maxillofacial Head & Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.,Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai, 200011, China
| | - Xing Xu
- Department of Oral and Maxillofacial Head & Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.,Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai, 200011, China
| | - Fen Zhao
- Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai, 200011, China
| | - Xu Wang
- Department of Oral and Maxillofacial Head & Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China. .,Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai, 200011, China.
| | - Yong Sun
- Department of Pharmaceutics, Qingdao University School of Pharmacy, Qingdao, 266021, China.
| | - Wantao Chen
- Department of Oral and Maxillofacial Head & Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China. .,Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai, 200011, China.
| |
Collapse
|
48
|
Cenariu D, Zimta AA, Munteanu R, Onaciu A, Moldovan CS, Jurj A, Raduly L, Moldovan A, Florea A, Budisan L, Pop LA, Magdo L, Albu MT, Tonea RB, Muresan MS, Ionescu C, Petrut B, Buiga R, Irimie A, Gulei D, Berindan-Neagoe I. Hsa-miR-125b Therapeutic Role in Colon Cancer Is Dependent on the Mutation Status of the TP53 Gene. Pharmaceutics 2021; 13:664. [PMID: 34066331 PMCID: PMC8148199 DOI: 10.3390/pharmaceutics13050664] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 04/27/2021] [Accepted: 05/01/2021] [Indexed: 12/13/2022] Open
Abstract
Colon cancer is the third most common cancer type worldwide and is highly dependent on DNA mutations that progressively appear and accumulate in the normal colon epithelium. Mutations in the TP53 gene appear in approximately half of these patients and have significant implications in disease progression and response to therapy. miR-125b-5p is a controversial microRNA with a dual role in cancer that has been reported to target specifically TP53 in colon adenocarcinomas. Our study investigated the differential therapeutic effect of miR-125b-5p replacement in colon cancer based on the TP53 mutation status of colon cancer cell lines. In TP53 mutated models, miR-125b-5p overexpression slows cancer cells' malignant behavior by inhibiting the invasion/migration and colony formation capacity via direct downregulation of mutated TP53. In TP53 wild type cells, the exogenous modulation of miR-125b-5p did not significantly affect the molecular and phenotypic profile. In conclusion, our data show that miR-125b-5p has an anti-cancer effect only in TP53 mutated colon cancer cells, explaining partially the dual behavior of this microRNA in malignant pathologies.
Collapse
Affiliation(s)
- Diana Cenariu
- MEDFUTURE—Research Center for Advanced Medicine, “Iuliu-Hatieganu” University of Medicine and Pharmacy, Marinescu 23 Street/Louis Pasteur 4–6 Street, 400337 Cluj-Napoca, Romania; (D.C.); (A.-A.Z.); (R.M.); (A.O.); (C.S.M.); (A.M.)
| | - Alina-Andreea Zimta
- MEDFUTURE—Research Center for Advanced Medicine, “Iuliu-Hatieganu” University of Medicine and Pharmacy, Marinescu 23 Street/Louis Pasteur 4–6 Street, 400337 Cluj-Napoca, Romania; (D.C.); (A.-A.Z.); (R.M.); (A.O.); (C.S.M.); (A.M.)
| | - Raluca Munteanu
- MEDFUTURE—Research Center for Advanced Medicine, “Iuliu-Hatieganu” University of Medicine and Pharmacy, Marinescu 23 Street/Louis Pasteur 4–6 Street, 400337 Cluj-Napoca, Romania; (D.C.); (A.-A.Z.); (R.M.); (A.O.); (C.S.M.); (A.M.)
| | - Anca Onaciu
- MEDFUTURE—Research Center for Advanced Medicine, “Iuliu-Hatieganu” University of Medicine and Pharmacy, Marinescu 23 Street/Louis Pasteur 4–6 Street, 400337 Cluj-Napoca, Romania; (D.C.); (A.-A.Z.); (R.M.); (A.O.); (C.S.M.); (A.M.)
| | - Cristian Silviu Moldovan
- MEDFUTURE—Research Center for Advanced Medicine, “Iuliu-Hatieganu” University of Medicine and Pharmacy, Marinescu 23 Street/Louis Pasteur 4–6 Street, 400337 Cluj-Napoca, Romania; (D.C.); (A.-A.Z.); (R.M.); (A.O.); (C.S.M.); (A.M.)
| | - Ancuta Jurj
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 23 Marinescu Street, 400337 Cluj-Napoca, Romania; (A.J.); (L.R.); (L.B.); (L.A.P.); (I.B.-N.)
| | - Lajos Raduly
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 23 Marinescu Street, 400337 Cluj-Napoca, Romania; (A.J.); (L.R.); (L.B.); (L.A.P.); (I.B.-N.)
| | - Alin Moldovan
- MEDFUTURE—Research Center for Advanced Medicine, “Iuliu-Hatieganu” University of Medicine and Pharmacy, Marinescu 23 Street/Louis Pasteur 4–6 Street, 400337 Cluj-Napoca, Romania; (D.C.); (A.-A.Z.); (R.M.); (A.O.); (C.S.M.); (A.M.)
| | - Adrian Florea
- Department of Cell and Molecular Biology, Faculty of Medicine, “Iuliu Haţieganu” University of Medicine and Pharmacy, 6 Louis Pasteur St., 400349 Cluj-Napoca, Romania;
| | - Liviuta Budisan
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 23 Marinescu Street, 400337 Cluj-Napoca, Romania; (A.J.); (L.R.); (L.B.); (L.A.P.); (I.B.-N.)
| | - Laura Ancuta Pop
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 23 Marinescu Street, 400337 Cluj-Napoca, Romania; (A.J.); (L.R.); (L.B.); (L.A.P.); (I.B.-N.)
| | - Lorand Magdo
- Faculty of Medicine, “Iuliu-Hatieganu” University of Medicine and Pharmacy, 8 Victor Babes Street, 400012 Cluj-Napoca, Romania; (L.M.); (M.T.A.); (R.B.T.)
| | - Mihai Tudor Albu
- Faculty of Medicine, “Iuliu-Hatieganu” University of Medicine and Pharmacy, 8 Victor Babes Street, 400012 Cluj-Napoca, Romania; (L.M.); (M.T.A.); (R.B.T.)
| | - Rares Bogdan Tonea
- Faculty of Medicine, “Iuliu-Hatieganu” University of Medicine and Pharmacy, 8 Victor Babes Street, 400012 Cluj-Napoca, Romania; (L.M.); (M.T.A.); (R.B.T.)
| | - Mihai-Stefan Muresan
- 5th Surgical Department, Municipal Hospital, 11 Tăbăcarilor Street, 400139 Cluj-Napoca, Romania; (M.-S.M.); (C.I.)
- Surgical and Gynecological Oncology Department, Prof. Dr. Ion Chiricuta” Oncology Institute, Republicii 34–36 Street, 400015 Cluj-Napoca, Romania
- Department of Surgery V, “Iuliu Hatieganu” University of Medicine and Pharmacy, 8 Victor Babes Street, 400012 Cluj-Napoca, Romania
| | - Calin Ionescu
- 5th Surgical Department, Municipal Hospital, 11 Tăbăcarilor Street, 400139 Cluj-Napoca, Romania; (M.-S.M.); (C.I.)
- Department of Surgery V, “Iuliu Hatieganu” University of Medicine and Pharmacy, 8 Victor Babes Street, 400012 Cluj-Napoca, Romania
| | - Bogdan Petrut
- Department of Urology, “Prof. Dr. Ion Chiricuta” Oncology Institute, Republicii 34–36 Street, 400015 Cluj-Napoca, Romania;
- Department of Urology, “Iuliu-Hatieganu” University of Medicine and Pharmacy, 8 Victor Babes Street, 400012 Cluj-Napoca, Romania
| | - Rares Buiga
- Department of Pathology, “Prof. Dr. Ion Chiricuta” Oncology Institute, Republicii 34–36 Street, 400015 Cluj-Napoca, Romania;
- Department of Pathology, “Iuliu-Hatieganu” University of Medicine and Pharmacy, 8 Victor Babes Street, 400012 Cluj-Napoca, Romania
| | - Alexandru Irimie
- 11th Department of Surgical Oncology and Gynaecological Oncology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 8 Victor Babes Street, 400012 Cluj-Napoca, Romania;
- Department of Surgery, The Oncology Institute “Prof. Dr. Ion Chiricuta”, 34–36 Republicii Street, 400015 Cluj-Napoca, Romania
| | - Diana Gulei
- MEDFUTURE—Research Center for Advanced Medicine, “Iuliu-Hatieganu” University of Medicine and Pharmacy, Marinescu 23 Street/Louis Pasteur 4–6 Street, 400337 Cluj-Napoca, Romania; (D.C.); (A.-A.Z.); (R.M.); (A.O.); (C.S.M.); (A.M.)
| | - Ioana Berindan-Neagoe
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 23 Marinescu Street, 400337 Cluj-Napoca, Romania; (A.J.); (L.R.); (L.B.); (L.A.P.); (I.B.-N.)
- Department of Functional Genomics and Experimental Pathology, “Prof. Dr. Ion Chiricuta” Oncology Institute, 34–36 Republicii Street, 400015 Cluj-Napoca, Romania
| |
Collapse
|
49
|
Cui X, Song K, Lu X, Feng W, Di W. Liposomal Delivery of MicroRNA-7 Targeting EGFR to Inhibit the Growth, Invasion, and Migration of Ovarian Cancer. ACS OMEGA 2021; 6:11669-11678. [PMID: 34056322 PMCID: PMC8153987 DOI: 10.1021/acsomega.1c00992] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 04/06/2021] [Indexed: 05/03/2023]
Abstract
Ovarian cancer is highly aggressive and has high rates of recurrence and metastasis. Due to the limited effects of current treatments, it is necessary to conduct research and develop new treatment options. The application of gene therapy in tumor therapy is gradually increasing and has exciting prospects. MicroRNA-7 (miR-7) has been reported to inhibit the growth, invasion, and metastasis of a variety of solid tumors. Cationic liposomes are safe and effective gene delivery systems for transfection in vivo and in vitro. To realize the application of miR-7 in the treatment of ovarian cancer, cationic liposomes were prepared with 1,2-dioleoyl-3-trimethylammonium-propane, 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine, and cholesterol. The miR-7 liposomes had a suitable particle size, potential, and a high cellular uptake rate. MiR-7 encapsulated by liposomes could be effectively delivered to ovarian cancer cells and successfully targeted to the tumor site in a mouse xenograft model of ovarian cancer. In vitro and in vivo experiments revealed that the miR-7 liposomes had a significant ability to inhibit the growth, invasion, and migration of ovarian cancer, probably by inhibiting the expression of the epidermal growth factor receptor. Our studies of miR-7 liposomes demonstrated a safe and efficient microRNA delivery system for the gene therapy of ovarian cancer.
Collapse
Affiliation(s)
- Xiaojuan Cui
- Department
of Obstetrics and Gynecology, Key Laboratory of Gynecologic Oncology,
and State Key Laboratory of Oncogenes and Related Genes, Shanghai
Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
- Department
of Obstetrics and Gynecology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China
| | - Keqi Song
- Department
of Obstetrics and Gynecology, Key Laboratory of Gynecologic Oncology,
and State Key Laboratory of Oncogenes and Related Genes, Shanghai
Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Xiaolan Lu
- Department
of Obstetrics and Gynecology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China
| | - Weiwei Feng
- Department
of Obstetrics and Gynecology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China
- . Phone: +86-21-64370045. Fax: +86-21-64370045
| | - Wen Di
- Department
of Obstetrics and Gynecology, Key Laboratory of Gynecologic Oncology,
and State Key Laboratory of Oncogenes and Related Genes, Shanghai
Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
- . Phone: +86-21-68383829. Fax: +86-21-68383829
| |
Collapse
|
50
|
Solé C, Lawrie CH. MicroRNAs in Metastasis and the Tumour Microenvironment. Int J Mol Sci 2021; 22:4859. [PMID: 34064331 PMCID: PMC8125549 DOI: 10.3390/ijms22094859] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 04/27/2021] [Accepted: 04/30/2021] [Indexed: 12/12/2022] Open
Abstract
Metastasis is the process whereby cancer cells migrate from the primary tumour site to colonise the surrounding or distant tissue or organ. Metastasis is the primary cause of cancer-related mortality and approximately half of all cancer patients present at diagnosis with some form of metastasis. Consequently, there is a clear need to better understand metastasis in order to develop new tools to combat this process. MicroRNAs (miRNAs) regulate gene expression and play an important role in cancer development and progression including in the metastatic process. Particularly important are the roles that miRNAs play in the interaction between tumour cells and non-tumoral cells of the tumour microenvironment (TME), a process mediated largely by circulating miRNAs contained primarily in extracellular vesicles (EVs). In this review, we outline the accumulating evidence for the importance of miRNAs in the communication between tumour cells and the cells of the TME in the context of the pre-metastatic and metastatic niche.
Collapse
Affiliation(s)
- Carla Solé
- Molecular Oncology Group, Biodonostia Research Institute, 20014 San Sebastian, Spain;
| | - Charles Henderson Lawrie
- Molecular Oncology Group, Biodonostia Research Institute, 20014 San Sebastian, Spain;
- IKERBASQUE, Basque Foundation for Science, 48009 Bilbao, Spain
- Radcliffe Department of Medicine, University of Oxford, Oxford OX4 3DU, UK
| |
Collapse
|