1
|
Mao S, Li J, Huang J, Lv L, Zhang Q, Cheng Q, Liu X, Bi Z, Yao J. Therapeutic potential of microRNA-506 in cancer treatment: mechanisms and therapeutic implications. Front Oncol 2025; 15:1524763. [PMID: 40248198 PMCID: PMC12003368 DOI: 10.3389/fonc.2025.1524763] [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: 11/08/2024] [Accepted: 03/17/2025] [Indexed: 04/19/2025] Open
Abstract
Cancer is a complex and highly lethal disease marked by unchecked cell proliferation, aggressive behavior, and a strong tendency to metastasize. Despite significant advancements in cancer diagnosis and treatment, challenges such as early detection difficulties, drug resistance, and adverse effects of radiotherapy or chemotherapy continue to threaten patient survival. MicroRNAs (miRNAs) have emerged as critical regulators in cancer biology, with miR-506 being extensively studied and recognized for its tumor-suppressive effects across multiple cancer types. This review examines the regulatory mechanisms of miR-506 in common cancers, focusing on its role in the competing endogenous RNA (ceRNA) network and its effects on cancer cell proliferation, apoptosis, and migration. We also discuss the potential of miR-506 as a therapeutic target and its role in overcoming drug resistance in cancer treatment. Overall, these insights underscore the therapeutic potential of miR-506 and its promise in developing novel cancer therapies.
Collapse
Affiliation(s)
- Shuzhen Mao
- Department of Pharmacy, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Junyan Li
- Department of Pathology, Second People’s Hospital of Ningyang, Taian, Shandong, China
| | - Jiahui Huang
- Jining Key Laboratory of Pharmacology, School of Basic Medicine, Jining Medical University, Jining, Shandong, China
| | - Lili Lv
- Department of Pathology, Second People’s Hospital of Ningyang, Taian, Shandong, China
| | - Qilian Zhang
- Department of Pathology, People’s Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Qing Cheng
- Jining Key Laboratory of Pharmacology, School of Basic Medicine, Jining Medical University, Jining, Shandong, China
| | - Xiaojing Liu
- Jining Key Laboratory of Pharmacology, School of Basic Medicine, Jining Medical University, Jining, Shandong, China
| | - Zhiwei Bi
- Jining Key Laboratory of Pharmacology, School of Basic Medicine, Jining Medical University, Jining, Shandong, China
| | - Jing Yao
- Jining Key Laboratory of Pharmacology, School of Basic Medicine, Jining Medical University, Jining, Shandong, China
| |
Collapse
|
2
|
Seong J, Kim M, Yoo J, Mack DL, Lee JH, Joo J. Sustained Release of HIF-2α Inhibitors Using Biodegradable Porous Silicon Carriers for Enhanced Immunogenic Cell Death of Malignant Merkel Cell Carcinoma. ACS APPLIED MATERIALS & INTERFACES 2025; 17:7449-7461. [PMID: 39834008 DOI: 10.1021/acsami.4c19961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Merkel cell carcinoma (MCC) is a rare but aggressive neuroendocrine skin cancer with limited treatment options, often associated with Merkel cell polyomavirus (MCPyV) and marked by hypoxic tumor microenvironments that promote resistance to therapies. Belzutifan, an FDA-approved hypoxia-inducible factor-2α (HIF-2α) inhibitor, has shown promise in inhibiting tumor growth; however, its clinical efficacy is hindered by its low solubility, rapid clearance, and limited bioavailability. In this study, we present a strategy using porous silicon (pSi) microparticles and nanoparticles as carriers for the sustained delivery of benzoate to MCC cells. The pSi carriers were engineered to securely encapsulate and gradually release belzutifan, overcoming the limitations of free drug administration. Microparticles provided sustained extracellular release, while nanoparticles enabled efficient intracellular delivery, enhancing HIF-2α inhibition. Moreover, the use of biodegradable silicon particles enables long-term consistent release of belzutifan over 10 days in vitro with a single dose administration in the tumor microenvironment, while free belzutifan is rapidly deactivated within 1 day postadministration. In vitro studies demonstrated significant immunogenic cell death (ICD) in MCC cells, marked by the cytosolic localization of HMGB1 and elevated expression of pro-inflammatory cytokines as well as strong upregulation of TLR9. Particularly, the increased TLR9 expression in both MCC cell lines with pSi carrier treatment reinforces immune activation through toll-like receptor signaling, enhancing both innate and adaptive immune responses within the tumor microenvironment. These findings indicate that pSi carriers not only enhance belzutifan's stability and release profile but also amplify antitumor immune responses within the tumor microenvironment. Our results suggest that belzutifan-loaded pSi carriers offer a potent and targeted therapeutic strategy for MCC, potentially addressing key challenges in cancer immunotherapy by combining HIF-2α inhibition with robust immune activation. This platform highlights the universal utility of pSi-based delivery systems to advance MCC treatment with implications for broader cancer therapies.
Collapse
Affiliation(s)
- Juyoung Seong
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
- Institute for Stem Cell and Regenerative Medicine (ISCRM), University of Washington, Seattle, Washington 98109, United States
| | - Minju Kim
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Jounghyun Yoo
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - David L Mack
- Institute for Stem Cell and Regenerative Medicine (ISCRM), University of Washington, Seattle, Washington 98109, United States
- Department of Bioengineering, University of Washington, Seattle, Washington 98195, United States
- Department of Rehabilitation Medicine, University of Washington, Seattle, Washington 98195, United States
| | - Jung Hyun Lee
- Institute for Stem Cell and Regenerative Medicine (ISCRM), University of Washington, Seattle, Washington 98109, United States
- Department of Dermatology, School of Medicine, University of Washington, Seattle, Washington 98109, United States
| | - Jinmyoung Joo
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
- Graduate School of Health Science and Technology, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
- Center for Genomic Integrity, Institute for Basic Science, Ulsan 44919, Republic of Korea
| |
Collapse
|
3
|
Yan J, Siwakoti P, Shaw S, Bose S, Kokil G, Kumeria T. Porous silicon and silica carriers for delivery of peptide therapeutics. Drug Deliv Transl Res 2024; 14:3549-3567. [PMID: 38819767 PMCID: PMC11499345 DOI: 10.1007/s13346-024-01609-7] [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] [Accepted: 04/15/2024] [Indexed: 06/01/2024]
Abstract
Peptides have gained tremendous popularity as biological therapeutic agents in recent years due to their favourable specificity, diversity of targets, well-established screening methods, ease of production, and lower cost. However, their poor physiological and storage stability, pharmacokinetics, and fast clearance have limited their clinical translation. Novel nanocarrier-based strategies have shown promise in overcoming these issues. In this direction, porous silicon (pSi) and mesoporous silica nanoparticles (MSNs) have been widely explored as potential carriers for the delivery of peptide therapeutics. These materials possess several advantages, including large surface areas, tunable pore sizes, and adjustable pore architectures, which make them attractive carriers for peptide delivery systems. In this review, we cover pSi and MSNs as drug carriers focusing on their use in peptide delivery. The review provides a brief overview of their fabrication, surface modification, and interesting properties that make them ideal peptide drug carriers. The review provides a systematic account of various studies that have utilised these unique porous carriers for peptide delivery describing significant in vitro and in vivo results. We have also provided a critical comparison of the two carriers in terms of their physicochemical properties and short-term and long-term biocompatibility. Lastly, we have concluded the review with our opinion of this field and identified key areas for future research for clinical translation of pSi and MSN-based peptide therapeutic formulations.
Collapse
Affiliation(s)
- Jiachen Yan
- School of Materials Science and Engineering, The University of New South Wales, Sydney, NSW, 2052, Australia
| | - Prakriti Siwakoti
- School of Materials Science and Engineering, The University of New South Wales, Sydney, NSW, 2052, Australia
- Australian Centre for Nanomedicine, The University of New South Wales, Sydney, NSW, 2052, Australia
| | - Siuli Shaw
- Centre for Medical Biotechnology, Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh, 201301, India
| | - Sudeep Bose
- Centre for Medical Biotechnology, Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh, 201301, India
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University, Noida, Uttar Pradesh, 201301, India
| | - Ganesh Kokil
- School of Materials Science and Engineering, The University of New South Wales, Sydney, NSW, 2052, Australia.
- Australian Centre for Nanomedicine, The University of New South Wales, Sydney, NSW, 2052, Australia.
| | - Tushar Kumeria
- School of Materials Science and Engineering, The University of New South Wales, Sydney, NSW, 2052, Australia.
- Australian Centre for Nanomedicine, The University of New South Wales, Sydney, NSW, 2052, Australia.
- School of Pharmacy, The University of Queensland, Woolloongabba, QLD, 4102, Australia.
| |
Collapse
|
4
|
Putri HMAR, Novianti PW, Pradjatmo H, Haryana SM. MicroRNA‑mediated approaches in ovarian cancer therapy: A comprehensive systematic review. Oncol Lett 2024; 28:491. [PMID: 39185494 PMCID: PMC11342411 DOI: 10.3892/ol.2024.14624] [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: 12/21/2023] [Accepted: 07/05/2024] [Indexed: 08/27/2024] Open
Abstract
Ovarian cancer (OC) poses a significant health risk to women worldwide, with late diagnoses and chemotherapy resistance leading to high mortality rates. Despite several histological subtypes, the primary challenge remains the subtle nature of its symptoms, resulting in advanced-stage diagnosis and reduced treatment success rates. With platinum-based therapies showing relative efficacy but limited survival enhancements, the emergence of chemotherapy resistance during recurrence remains a critical obstacle. Precision medicine development has aimed to address these challenges in the context of the molecular diversity of OC. The present review explored the landscape of microRNA (miRNA)-mediated approaches in OC treatment. miRNAs have emerged as regulators of gene expression, serving as both oncogenes and tumor suppressors in OC. Dysregulated miRNAs are associated with disease progression and chemotherapy resistance, underscoring their significance in diagnosis and tailored treatment strategies. The present review extracted 295 publications from the PUBMED database. Out of the 73 eligible studies, 55 miRNAs were assessed. A total of three of these miRNAs were not associated with any disease or cancer, whilst eight were associated with OC, albeit also associated with other diseases. The present review encompassed three dimensions: i) The role of miRNAs in treatment efficacy; ii) the use of miRNAs to enhance therapy outcomes; and iii) adjunctive strategies for improved treatment results. Furthermore, it offered insights into potential avenues for improving OC treatment using miRNA-based approaches.
Collapse
Affiliation(s)
| | | | - Heru Pradjatmo
- Department of Obstetrics and Gynecology, Faculty of Medicine, Public Health and Nursing, Gadjah Mada University, Depok, Yogyakarta 55281, Indonesia
- Department of Obstetrics and Gynecology, Sardjito Hospital, Depok, Yogyakarta 55281, Indonesia
| | - Sofia Mubarika Haryana
- Department of Histology and Cell Biology, Faculty of Medicine, Public Health and Nursing, Gadjah Mada University, Depok, Yogyakarta 55281, Indonesia
| |
Collapse
|
5
|
Shin S, Jo H, Agura T, Jeong S, Ahn H, Kim Y, Kang JS. Use of surface-modified porous silicon nanoparticles to deliver temozolomide with enhanced pharmacokinetic and therapeutic efficacy for intracranial glioblastoma in mice. J Mater Chem B 2024; 12:9335-9344. [PMID: 39171683 DOI: 10.1039/d4tb00631c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2024]
Abstract
Glioblastoma (GBM) is one of the most common and fatal primary brain tumors, with a 5-year survival rate of 7.2%. The standard treatment for GBM involves surgical resection followed by chemoradiotherapy, and temozolomide (TMZ) is currently the only approved chemotherapeutic agent for the treatment of GBM. However, hydrolytic instability and insufficient drug accumulation are major challenges that limit the effectiveness of TMZ chemotherapy. To overcome these limitations, we have developed a drug delivery platform utilizing porous silicon nanoparticles (pSiNPs) to improve the stability and blood-brain barrier penetration of TMZ. The pSiNPs are synthesized via electrochemical etching and functionalized with octadecane. The octadecyl-modified pSiNP (pSiNP-C18) demonstrates the superiority of loading efficiency, in vivo stability, and brain accumulation of TMZ. Treatment of intracranial tumor-bearing mice with TMZ-loaded pSiNP-C18 results in a decreased tumor burden and a corresponding increase in survival compared with equivalent free-drug dosing. Furthermore, the mice treated with TMZ-loaded nanoparticles do not exhibit in vivo toxicity, thus underscoring the preclinical potential of the pSiNP-based platform for the delivery of therapeutic agents to gliomas.
Collapse
Affiliation(s)
- Seulgi Shin
- Laboratory of Vitamin C and Antioxidant Immunology, Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea.
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul 03080, Republic of Korea
- Department of Research and Development, N therapeutics Co., Ltd, Seoul 08813, Republic of Korea
| | - Hyejung Jo
- Laboratory of Vitamin C and Antioxidant Immunology, Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea.
| | - Tomoyo Agura
- Laboratory of Vitamin C and Antioxidant Immunology, Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea.
| | - Seoyoun Jeong
- Laboratory of Vitamin C and Antioxidant Immunology, Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea.
| | - Hyovin Ahn
- Laboratory of Vitamin C and Antioxidant Immunology, Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea.
| | - Yejin Kim
- Laboratory of Vitamin C and Antioxidant Immunology, Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea.
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul 03080, Republic of Korea
| | - Jae Seung Kang
- Laboratory of Vitamin C and Antioxidant Immunology, Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea.
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul 03080, Republic of Korea
- Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
6
|
Shrestha S, Tieu T, Wojnilowicz M, Voelcker NH, Forsythe JS, Frith JE. Delivery of miRNAs Using Porous Silicon Nanoparticles Incorporated into 3D Hydrogels Enhances MSC Osteogenesis by Modulation of Fatty Acid Signaling and Silicon Degradation. Adv Healthc Mater 2024; 13:e2400171. [PMID: 38657207 DOI: 10.1002/adhm.202400171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 04/10/2024] [Indexed: 04/26/2024]
Abstract
Strategies incorporating mesenchymal stromal cells (MSC), hydrogels and osteoinductive signals offer promise for bone repair. Osteoinductive signals such as growth factors face challenges in clinical translation due to their high cost, low stability and immunogenicity leading to interest in microRNAs as a simple, inexpensive and powerful alternative. The selection of appropriate miRNA candidates and their efficient delivery must be optimised to make this a reality. This study evaluated pro-osteogenic miRNAs and used porous silicon nanoparticles modified with polyamidoamine dendrimers (PAMAM-pSiNP) to deliver these to MSC encapsulated within gelatin-PEG hydrogels. miR-29b-3p, miR-101-3p and miR-125b-5p are strongly pro-osteogenic and are shown to target FASN and ELOVL4 in the fatty acid biosynthesis pathway to modulate MSC osteogenesis. Hydrogel delivery of miRNA:PAMAM-pSiNP complexes enhanced transfection compared to 2D. The osteogenic potential of hBMSC in hydrogels with miR125b:PAMAM-pSiNP complexes is evaluated. Importantly, a dual-effect on osteogenesis occurred, with miRNAs increasing expression of alkaline phosphatase (ALP) and Runt-related transcription factor 2 (RUNX2) whilst the pSiNPs enhanced mineralisation, likely via degradation into silicic acid. Overall, this work presents insights into the role of miRNAs and fatty acid signalling in osteogenesis, providing future targets to improve bone formation and a promising system to enhance bone tissue engineering.
Collapse
Affiliation(s)
- Surakshya Shrestha
- Department of Materials Science and Engineering, Monash University, Clayton, VIC, 3800, Australia
| | - Terence Tieu
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, Clayton, VIC, 3168, Australia
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
- Commonwealth Scientific and Industrial Research Organisation (CSIRO) Manufacturing, Bayview Avenue, Clayton, VIC, 3168, Australia
| | - Marcin Wojnilowicz
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
- Commonwealth Scientific and Industrial Research Organisation (CSIRO) Manufacturing, Bayview Avenue, Clayton, VIC, 3168, Australia
| | - Nicolas H Voelcker
- Department of Materials Science and Engineering, Monash University, Clayton, VIC, 3800, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, Clayton, VIC, 3168, Australia
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
- Commonwealth Scientific and Industrial Research Organisation (CSIRO) Manufacturing, Bayview Avenue, Clayton, VIC, 3168, Australia
- ARC Training Centre for Cell and Tissue Engineering Technologies, Monash University, Clayton, VIC, 3800, Australia
| | - John S Forsythe
- Department of Materials Science and Engineering, Monash University, Clayton, VIC, 3800, Australia
- ARC Training Centre for Cell and Tissue Engineering Technologies, Monash University, Clayton, VIC, 3800, Australia
| | - Jessica E Frith
- Department of Materials Science and Engineering, Monash University, Clayton, VIC, 3800, Australia
- ARC Training Centre for Cell and Tissue Engineering Technologies, Monash University, Clayton, VIC, 3800, Australia
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, 3800, Australia
| |
Collapse
|
7
|
Nair MG, Mavatkar AD, Naidu CM, V. P. S, C. E. A, Rajarajan S, Sahoo S, Mohan G, Jaikumar VS, Ramesh RS, B. S. S, Jolly MK, Maliekal TT, Prabhu JS. Elucidating the Role of MicroRNA-18a in Propelling a Hybrid Epithelial-Mesenchymal Phenotype and Driving Malignant Progression in ER-Negative Breast Cancer. Cells 2024; 13:821. [PMID: 38786043 PMCID: PMC11119613 DOI: 10.3390/cells13100821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 04/19/2024] [Accepted: 04/29/2024] [Indexed: 05/25/2024] Open
Abstract
Epigenetic alterations that lead to differential expression of microRNAs (miRNAs/miR) are known to regulate tumour cell states, epithelial-mesenchymal transition (EMT) and the progression to metastasis in breast cancer. This study explores the key contribution of miRNA-18a in mediating a hybrid E/M cell state that is pivotal to the malignant transformation and tumour progression in the aggressive ER-negative subtype of breast cancer. The expression status and associated effects of miR-18a were evaluated in patient-derived breast tumour samples in combination with gene expression data from public datasets, and further validated in in vitro and in vivo breast cancer model systems. The clinical relevance of the study findings was corroborated against human breast tumour specimens (n = 446 patients). The down-regulated expression of miR-18a observed in ER-negative tumours was found to drive the enrichment of hybrid epithelial/mesenchymal (E/M) cells with luminal attributes, enhanced traits of migration, stemness, drug-resistance and immunosuppression. Further analysis of the miR-18a targets highlighted possible hypoxia-inducible factor 1-alpha (HIF-1α)-mediated signalling in these tumours. This is a foremost report that validates the dual role of miR-18a in breast cancer that is subtype-specific based on hormone receptor expression. The study also features a novel association of low miR-18a levels and subsequent enrichment of hybrid E/M cells, increased migration and stemness in a subgroup of ER-negative tumours that may be attributed to HIF-1α mediated signalling. The results highlight the possibility of stratifying the ER-negative disease into clinically relevant groups by analysing miRNA signatures.
Collapse
Affiliation(s)
- Madhumathy G. Nair
- Division of Molecular Medicine, St. John’s Research Institute, St. John’s Medical College, Bangalore 560034, Karnataka, India
| | - Apoorva D. Mavatkar
- Division of Molecular Medicine, St. John’s Research Institute, St. John’s Medical College, Bangalore 560034, Karnataka, India
| | - Chandrakala M. Naidu
- Division of Molecular Medicine, St. John’s Research Institute, St. John’s Medical College, Bangalore 560034, Karnataka, India
| | - Snijesh V. P.
- Division of Molecular Medicine, St. John’s Research Institute, St. John’s Medical College, Bangalore 560034, Karnataka, India
| | - Anupama C. E.
- Division of Molecular Medicine, St. John’s Research Institute, St. John’s Medical College, Bangalore 560034, Karnataka, India
| | - Savitha Rajarajan
- Division of Molecular Medicine, St. John’s Research Institute, St. John’s Medical College, Bangalore 560034, Karnataka, India
| | - Sarthak Sahoo
- Department of Bioengineering, Indian Institute of Science (Bangalore), Bengaluru 560012, Karnataka, India
| | - Gayathri Mohan
- Cancer Research, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram 695014, Kerala, India
| | - Vishnu Sunil Jaikumar
- Animal Research Facility, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram 695014, Kerala, India
| | - Rakesh S. Ramesh
- Department of Surgical Oncology, St. John’s Medical College and Hospital, Bangalore 560034, Karnataka, India
| | - Srinath B. S.
- Department of Surgical Oncology, Sri Shankara Cancer Hospital and Research Centre, Bangalore 560004, Karnataka, India
| | - Mohit Kumar Jolly
- Department of Bioengineering, Indian Institute of Science (Bangalore), Bengaluru 560012, Karnataka, India
| | - Tessy Thomas Maliekal
- Cancer Research, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram 695014, Kerala, India
| | - Jyothi S. Prabhu
- Division of Molecular Medicine, St. John’s Research Institute, St. John’s Medical College, Bangalore 560034, Karnataka, India
| |
Collapse
|
8
|
Broc B, Varini K, Sonnette R, Pecqueux B, Benoist F, Masse M, Mechioukhi Y, Ferracci G, Temsamani J, Khrestchatisky M, Jacquot G, Lécorché P. LDLR-Mediated Targeting and Productive Uptake of siRNA-Peptide Ligand Conjugates In Vitro and In Vivo. Pharmaceutics 2024; 16:548. [PMID: 38675209 PMCID: PMC11054735 DOI: 10.3390/pharmaceutics16040548] [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: 02/13/2024] [Revised: 03/29/2024] [Accepted: 04/02/2024] [Indexed: 04/28/2024] Open
Abstract
Small RNA molecules such as microRNA and small interfering RNA (siRNA) have become promising therapeutic agents because of their specificity and their potential to modulate gene expression. Any gene of interest can be potentially up- or down-regulated, making RNA-based technology the healthcare breakthrough of our era. However, the functional and specific delivery of siRNAs into tissues of interest and into the cytosol of target cells remains highly challenging, mainly due to the lack of efficient and selective delivery systems. Among the variety of carriers for siRNA delivery, peptides have become essential candidates because of their high selectivity, stability, and conjugation versatility. Here, we describe the development of molecules encompassing siRNAs against SOD1, conjugated to peptides that target the low-density lipoprotein receptor (LDLR), and their biological evaluation both in vitro and in vivo.
Collapse
Affiliation(s)
- Baptiste Broc
- Vect-Horus S.A.S, Faculté des Sciences Médicales et Paramédicales Secteur Timone, 13385 Marseille, France
- Aix-Marseille Univ, CNRS, INP, Inst Neurophysiopathol, 13005 Marseille, France
| | - Karine Varini
- Vect-Horus S.A.S, Faculté des Sciences Médicales et Paramédicales Secteur Timone, 13385 Marseille, France
| | - Rose Sonnette
- Vect-Horus S.A.S, Faculté des Sciences Médicales et Paramédicales Secteur Timone, 13385 Marseille, France
| | - Belinda Pecqueux
- Vect-Horus S.A.S, Faculté des Sciences Médicales et Paramédicales Secteur Timone, 13385 Marseille, France
| | - Florian Benoist
- Vect-Horus S.A.S, Faculté des Sciences Médicales et Paramédicales Secteur Timone, 13385 Marseille, France
| | - Maxime Masse
- Vect-Horus S.A.S, Faculté des Sciences Médicales et Paramédicales Secteur Timone, 13385 Marseille, France
| | - Yasmine Mechioukhi
- Vect-Horus S.A.S, Faculté des Sciences Médicales et Paramédicales Secteur Timone, 13385 Marseille, France
| | - Géraldine Ferracci
- Aix-Marseille Univ, CNRS, INP, Inst Neurophysiopathol, 13005 Marseille, France
| | - Jamal Temsamani
- Vect-Horus S.A.S, Faculté des Sciences Médicales et Paramédicales Secteur Timone, 13385 Marseille, France
| | | | - Guillaume Jacquot
- Vect-Horus S.A.S, Faculté des Sciences Médicales et Paramédicales Secteur Timone, 13385 Marseille, France
| | - Pascaline Lécorché
- Vect-Horus S.A.S, Faculté des Sciences Médicales et Paramédicales Secteur Timone, 13385 Marseille, France
| |
Collapse
|
9
|
Jiang NJ, Yin YN, Lin J, Li WY, Long DR, Mei L. MicroRNA-21 in gynecological cancers: From molecular pathogenesis to clinical significance. Pathol Res Pract 2023; 248:154630. [PMID: 37393665 DOI: 10.1016/j.prp.2023.154630] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 06/16/2023] [Accepted: 06/18/2023] [Indexed: 07/04/2023]
Abstract
Ovarian, cervical, and endometrial cancers are the three most common gynecological cancer types (GCs). They hold a significant position as the leading causes of mortality among women with cancer-related death. However, GCs are often diagnosed late, severely limiting the efficacy of current treatment options. Thus, there is an urgent, unmet need for innovative experimentation to enhance the clinical treatment of GC patients. MicroRNAs (miRNAs) are a large and varied class of short noncoding RNAs (22 nucleotides in length) that have been shown to play essential roles in various biological processes involved in development. Recent research has shown that miR-211 influences tumorigenesis and cancer formation, adding to our knowledge of the miR-21 dysregulation in GCs. Furthermore, current research that sheds light on the crucial functions of miR-21 may provide supporting evidence for its potential prognostic, diagnostic, and therapeutic applications in the context of GCs. This review will thus focus on the most recent findings concerning miR-21 expression, miR-21 target genes, and the processes behind GCs. In addition, the latest findings that support miR-21's potential use as a non-invasive biomarker and therapeutic agent for detecting and treating cancer will be elucidated in this review. The roles played by various lncRNA/circRNA-miRNA-mRNA axis in GCs are also comprehensively summarized and described in this study, along with any possible implications for how these regulatory networks may contribute to the pathogenesis of GCs. Also, it is crucial to recognize the complexity of the processes involved in tumour therapeutic resistance as a significant obstacle in treating GCs. Furthermore, this review provides an overview of the current state of knowledge regarding the functional significance miR-21 in therapeutic resistance within the context of GCs.
Collapse
Affiliation(s)
- Ni-Jie Jiang
- Department of Gynecology and Obstetrics Nursing, West China Second University Hospital Sichuan University, Chengdu, 610041, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children Sichuan University, Ministry of Education, Chengdu, 610041, China
| | - Ya-Nan Yin
- Department of Gynecology and Obstetrics Nursing, West China Second University Hospital Sichuan University, Chengdu, 610041, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children Sichuan University, Ministry of Education, Chengdu, 610041, China
| | - Jiao Lin
- Department of Gynecology and Obstetrics Nursing, West China Second University Hospital Sichuan University, Chengdu, 610041, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children Sichuan University, Ministry of Education, Chengdu, 610041, China
| | - Wen-Yuan Li
- West China Nursing School, Sichuan University, Chengdu, 610041, China
| | - De-Rong Long
- Department of Gynecology and Obstetrics Nursing, West China Second University Hospital Sichuan University, Chengdu, 610041, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children Sichuan University, Ministry of Education, Chengdu, 610041, China
| | - Ling Mei
- Key Laboratory of Birth Defects and Related Diseases of Women and Children Sichuan University, Ministry of Education, Chengdu, 610041, China; Department of Gynecology and Obstetrics, West China Second Hospital of Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
10
|
Zhang J, Ding H, Zhang F, Xu Y, Liang W, Huang L. New trends in diagnosing and treating ovarian cancer using nanotechnology. Front Bioeng Biotechnol 2023; 11:1160985. [PMID: 37082219 PMCID: PMC10110946 DOI: 10.3389/fbioe.2023.1160985] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 03/22/2023] [Indexed: 04/07/2023] Open
Abstract
Ovarian cancer stands as the fifth most prevalent cancer among women, causing more mortalities than any other disease of the female reproductive system. There are numerous histological subtypes of ovarian cancer, each of which has distinct clinical characteristics, risk factors, cell origins, molecular compositions, and therapeutic options. Typically, it is identified at a late stage, and there is no efficient screening method. Standard therapies for newly diagnosed cancer are cytoreductive surgery and platinum-based chemotherapy. The difficulties of traditional therapeutic procedures encourage researchers to search for other approaches, such as nanotechnology. Due to the unique characteristics of matter at the nanoscale, nanomedicine has emerged as a potent tool for creating novel drug carriers that are more effective and have fewer adverse effects than traditional treatments. Nanocarriers including liposomes, dendrimers, polymer nanoparticles, and polymer micelles have unique properties in surface chemistry, morphology, and mechanism of action that can distinguish between malignant and normal cells, paving the way for targeted drug delivery. In contrast to their non-functionalized counterparts, the development of functionalized nano-formulations with specific ligands permits selective targeting of ovarian cancers and ultimately increases the therapeutic potential. This review focuses on the application of various nanomaterials to the treatment and diagnosis of ovarian cancer, their advantages over conventional treatment methods, and the effective role of controlled drug delivery systems in the therapy of ovarian cancer.
Collapse
Affiliation(s)
- Juan Zhang
- Department of Gynecology, Shaoxing Maternity and Child Healthcare Hospital, Shaoxing, China
- Obstetrics and Gynecology Hospital of Shaoxing University, Shaoxing, China
| | - Haigang Ding
- Department of Gynecology, Shaoxing Maternity and Child Healthcare Hospital, Shaoxing, China
- Obstetrics and Gynecology Hospital of Shaoxing University, Shaoxing, China
| | - Feng Zhang
- Department of Gynecology, Shaoxing Maternity and Child Healthcare Hospital, Shaoxing, China
- Obstetrics and Gynecology Hospital of Shaoxing University, Shaoxing, China
| | - Yan Xu
- Intensive Care Unit, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Wenqing Liang
- Medical Research Center, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
- *Correspondence: Liping Huang, ; Wenqing Liang,
| | - Liping Huang
- Department of Medical Oncology, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
- *Correspondence: Liping Huang, ; Wenqing Liang,
| |
Collapse
|
11
|
Bhatia A, Upadhyay AK, Sharma S. miRNAs are now starring in "No Time to Die: Overcoming the chemoresistance in cancer". IUBMB Life 2023; 75:238-256. [PMID: 35678612 DOI: 10.1002/iub.2652] [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: 03/15/2022] [Accepted: 05/04/2022] [Indexed: 12/24/2022]
Abstract
Cancer is a leading cause of death globally, with about 19.3 million new cases reported each year. Current therapies for cancer management include-chemotherapy, radiotherapy, and surgery. However, they are loaded with side effects and tend to cause toxicity in the patient's body posttreatment, ultimately hindering the response towards the treatment building up resistance. This is where noncoding RNAs such as miRNAs help provide us with a helping hand for taming the chemoresistance and providing potential holistic cancer management. MicroRNAs are promising targets for anticancer therapy as they perform critical regulatory roles in various signaling cascades related to cell proliferation, apoptosis, migration, and invasion. Combining miRNAs and anticancer drugs and devising a combination therapy has managed cancer well in various independent studies. This review aims to provide insights into how miRNAs play a mechanistic role in cancer development and progression and regulate drug resistance in various types of cancers. Furthermore, next-generation novel therapies using miRNAs in combination with anticancer treatments in multiple cancers have been put forth and how they improve the efficacy of the treatments. Exemplary studies currently in the preclinical and clinical models have been summarized. Ultimately, we briefly talk through the challenges that come forward with it and minimize them.
Collapse
Affiliation(s)
- Anmol Bhatia
- Department of Biotechnology, Thapar Institute of Engineering & Technology, Patiala, India
| | - Atul Kumar Upadhyay
- Department of Biotechnology, Thapar Institute of Engineering & Technology, Patiala, India
| | - Siddharth Sharma
- Department of Biotechnology, Thapar Institute of Engineering & Technology, Patiala, India
| |
Collapse
|
12
|
Porous silicon surface modification via a microwave-induced in situ cyclic disulfide (S-S) cleavage and Si-S bond formation. Colloids Surf B Biointerfaces 2023; 222:113055. [PMID: 36463610 DOI: 10.1016/j.colsurfb.2022.113055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 11/15/2022] [Accepted: 11/22/2022] [Indexed: 11/27/2022]
Abstract
Porous silicon (pSi) materials have gained a great deal of attention from various research fields, and their surface-functionalization is one of the critical points for their applications. In this study, a new surface modification method of Si-H-terminated pSi materials via microwave-induced Si-S bond formation is disclosed. The silicon hydride (Si-H) functionality on the pSi surface could react with the 5-membered cyclic disulfide (S-S) compound (DL-α-lipoic acid in this study) by microwave-induced in situ S-S bond cleavage and Si-S bond formation. This surface chemistry is fast responsive (<10 min) and more efficient than other methods such as vortexing, heating stirring, or ultrasonication. The reaction maintains the primary porous structure of pSi materials including pSi wafer, pSi rugate filer, and pSi nanoparticles. An additional functional group such as carboxylic acid is demonstrated to be readily introducible on the pSi surface for further applications. Overall, this study has successfully demonstrated the porous silicon surface modification via a microwave-induced in situ cyclic disulfide (S-S) cleavage and Si-S bond formation.
Collapse
|
13
|
Current Update on Nanotechnology-Based Approaches in Ovarian Cancer Therapy. Reprod Sci 2023; 30:335-349. [PMID: 35585292 DOI: 10.1007/s43032-022-00968-1] [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: 01/13/2022] [Accepted: 05/06/2022] [Indexed: 10/18/2022]
Abstract
Ovarian cancer is one of the leading causes of cancer-related deaths among women. The drawbacks of conventional therapeutic strategies encourage researchers to look for alternative strategies, including nanotechnology. Nanotechnology is one of the upcoming domains of science that is rechanneled towards targeted cancer therapy and diagnosis. Nanocarriers such as dendrimers, liposomes, polymer micelles, and polymer nanoparticles present distinct surface characteristics in morphology, surface chemistry, and mode of action that help differentiate normal and malignant cells, which paves the way for target-specific drug delivery. Similarly, nanoparticles have been strategically utilized as efficacious vehicles to deliver drugs that alter the epigenetic modifications in epigenetic therapy. Some studies suggest that the use of specialized target-modified nanoparticles in siRNA-based nanotherapy prevents internalization and improves the antitumor activity of siRNA by ensuring unrestrained entry of siRNA into the tumor vasculature and efficient intracellular delivery of siRNA. Moreover, research findings highlight the significance of utilizing nanoparticles as depots for photosensitive drugs in photodynamic therapy. The applicability of nanoparticles is further extended to medical imaging. They serve as contrast agents in combination with conventional imaging modalities such as MRI, CT, and fluorescence-based imaging to produce vivid and enhanced images of tumors. Therefore, this review aims to explore and delve deeper into the advent of various nanotechnology-based therapeutic and imaging techniques that provide non-invasive and effective means to tackle ovarian cancers.
Collapse
|
14
|
Kim J, Um H, Kim NH, Kim D. Potential Alzheimer's disease therapeutic nano-platform: Discovery of amyloid-beta plaque disaggregating agent and brain-targeted delivery system using porous silicon nanoparticles. Bioact Mater 2023; 24:497-506. [PMID: 36685808 PMCID: PMC9841037 DOI: 10.1016/j.bioactmat.2023.01.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 12/04/2022] [Accepted: 01/05/2023] [Indexed: 01/11/2023] Open
Abstract
There has been a lot of basic and clinical research on Alzheimer's disease (AD) over the last 100 years, but its mechanisms and treatments have not been fully clarified. Despite some controversies, the amyloid-beta hypothesis is one of the most widely accepted causes of AD. In this study, we disclose a new amyloid-beta plaque disaggregating agent and an AD brain-targeted delivery system using porous silicon nanoparticles (pSiNPs) as a therapeutic nano-platform to overcome AD. We hypothesized that the negatively charged sulfonic acid functional group could disaggregate plaques and construct a chemical library. As a result of the in vitro assay of amyloid plaques and library screening, we confirmed that 6-amino-2-naphthalenesulfonic acid (ANA) showed the highest efficacy for plaque disaggregation as a hit compound. To confirm the targeted delivery of ANA to the AD brain, a nano-platform was created using porous silicon nanoparticles (pSiNPs) with ANA loaded into the pore of pSiNPs and biotin-polyethylene glycol (PEG) surface functionalization. The resulting nano-formulation, named Biotin-CaCl2-ANA-pSiNPs (BCAP), delivered a large amount of ANA to the AD brain and ameliorated memory impairment of the AD mouse model through the disaggregation of amyloid plaques in the brain. This study presents a new bioactive small molecule for amyloid plaque disaggregation and its promising therapeutic nano-platform for AD brain-targeted delivery.
Collapse
Affiliation(s)
- Jaehoon Kim
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Hyeji Um
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Na Hee Kim
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Dokyoung Kim
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea,Medical Research Center for Bioreaction to Reactive Oxygen Species and Biomedical Science Institute, Kyung Hee University, Seoul, 02447, Republic of Korea,Department of Anatomy and Neurobiology, College of Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea,Center for Converging Humanities, Kyung Hee University, Seoul, 02447, Republic of Korea,KHU-KIST Department of Converging Science and Technology, Kyung Hee University, Seoul, 02447, Republic of Korea,UC San Diego Materials Research Science and Engineering Center, 9500 Gilman Drive La Jolla, CA, 92093, USA,Corresponding author. Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea.
| |
Collapse
|
15
|
Vijayakumar S, Nasr SH, Davis JE, Wang E, Zuidema JM, Lu YS, Lo YH, Sicklick JK, Sailor MJ, Ray P. Anti-KIT DNA aptamer-conjugated porous silicon nanoparticles for the targeted detection of gastrointestinal stromal tumors. NANOSCALE 2022; 14:17700-17713. [PMID: 36416809 PMCID: PMC9744628 DOI: 10.1039/d2nr03905b] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Evaluation of Gastrointestinal Stromal Tumors (GIST) during initial clinical staging, surgical intervention, and postoperative management can be challenging. Current imaging modalities (e.g., PET and CT scans) lack sensitivity and specificity. Therefore, advanced clinical imaging modalities that can provide clinically relevant images with high resolution would improve diagnosis. KIT is a tyrosine kinase receptor overexpressed on GIST. Here, the application of a specific DNA aptamer targeting KIT, decorated onto a fluorescently labeled porous silicon nanoparticle (pSiNP), is used for the in vitro & in vivo imaging of GIST. This nanoparticle platform provides high-fidelity GIST imaging with minimal cellular toxicity. An in vitro analysis shows greater than 15-fold specific KIT protein targeting compared to the free KIT aptamer, while in vivo analyses of GIST-burdened mice that had been injected intravenously (IV) with aptamer-conjugated pSiNPs show extensive nanoparticle-to-tumor signal co-localization (>90% co-localization) compared to control particles. This provides an effective platform for which aptamer-conjugated pSiNP constructs can be used for the imaging of KIT-expressing cancers or for the targeted delivery of therapeutics.
Collapse
Affiliation(s)
- Sanahan Vijayakumar
- Materials Science and Engineering Program, University of California, San Diego, La Jolla, California, 92093, USA.
| | - Seyedmehdi H Nasr
- Department of Chemistry & Biochemistry, University of California, San Diego, La Jolla, California, 92093, USA
| | - Jacob E Davis
- Department of Surgery, Division of Surgical Oncology, University of California, San Diego, San Diego, California, 92093, USA.
- Moores Cancer Center, University of California, San Diego, La Jolla, California, 92093, USA
| | - Edward Wang
- Materials Science and Engineering Program, University of California, San Diego, La Jolla, California, 92093, USA.
| | - Jonathan M Zuidema
- Department of Chemistry & Biochemistry, University of California, San Diego, La Jolla, California, 92093, USA
- Department of Neurosciences, University of California, San Diego, San Diego, California, 92093, USA
| | - Yi-Sheng Lu
- Materials Science and Engineering Program, University of California, San Diego, La Jolla, California, 92093, USA.
| | - Yu-Hwa Lo
- Materials Science and Engineering Program, University of California, San Diego, La Jolla, California, 92093, USA.
- Department of Electrical & Computer Engineering, University of California, San Diego, La Jolla, California, 92093, USA
| | - Jason K Sicklick
- Department of Surgery, Division of Surgical Oncology, University of California, San Diego, San Diego, California, 92093, USA.
- Moores Cancer Center, University of California, San Diego, La Jolla, California, 92093, USA
- Department of Pharmacology, University of California, San Diego, San Diego, California, 92093, USA
| | - Michael J Sailor
- Materials Science and Engineering Program, University of California, San Diego, La Jolla, California, 92093, USA.
- Department of Chemistry & Biochemistry, University of California, San Diego, La Jolla, California, 92093, USA
| | - Partha Ray
- Department of Surgery, Division of Surgical Oncology, University of California, San Diego, San Diego, California, 92093, USA.
- Moores Cancer Center, University of California, San Diego, La Jolla, California, 92093, USA
| |
Collapse
|
16
|
Kandettu A, Adiga D, Devi V, Suresh PS, Chakrabarty S, Radhakrishnan R, Kabekkodu SP. Deregulated miRNA clusters in ovarian cancer: Imperative implications in personalized medicine. Genes Dis 2022; 9:1443-1465. [PMID: 36157483 PMCID: PMC9485269 DOI: 10.1016/j.gendis.2021.12.026] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 12/04/2021] [Accepted: 12/31/2021] [Indexed: 11/25/2022] Open
Abstract
Ovarian cancer (OC) is one of the most common and fatal types of gynecological cancer. OC is usually detected at the advanced stages of the disease, making it highly lethal. miRNAs are single-stranded, small non-coding RNAs with an approximate size ranging around 22 nt. Interestingly, a considerable proportion of miRNAs are organized in clusters with miRNA genes placed adjacent to one another, getting transcribed together to result in miRNA clusters (MCs). MCs comprise two or more miRNAs that follow the same orientation during transcription. Abnormal expression of the miRNA cluster has been identified as one of the key drivers in OC. MC exists both as tumor-suppressive and oncogenic clusters and has a significant role in OC pathogenesis by facilitating cancer cells to acquire various hallmarks. The present review summarizes the regulation and biological function of MCs in OC. The review also highlights the utility of abnormally expressed MCs in the clinical management of OC.
Collapse
Affiliation(s)
- Amoolya Kandettu
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Divya Adiga
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Vasudha Devi
- Department of Pharmacology, Centre for Cardiovascular Pharmacology, Melaka Manipal Medical College, Manipal Academy of Higher Education, Manipal Campus, Manipal, Karnataka 576104, India
| | - Padmanaban S. Suresh
- School of Biotechnology, National Institute of Technology, Calicut, Kerala 673601, India
| | - Sanjiban Chakrabarty
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
- Center for DNA Repair and Genome Stability (CDRGS), Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Raghu Radhakrishnan
- Department of Oral Pathology, Manipal College of Dental Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Shama Prasada Kabekkodu
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
- Center for DNA Repair and Genome Stability (CDRGS), Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| |
Collapse
|
17
|
Katopodi T, Petanidis S, Tsavlis D, Anestakis D, Charalampidis C, Chatziprodromidou I, Eskitzis P, Zarogoulidis P, Kosmidis C, Matthaios D, Porpodis K. Engineered multifunctional nanocarriers for controlled drug delivery in tumor immunotherapy. Front Oncol 2022; 12:1042125. [PMID: 36338748 PMCID: PMC9634039 DOI: 10.3389/fonc.2022.1042125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 10/06/2022] [Indexed: 11/25/2022] Open
Abstract
The appearance of chemoresistance in cancer is a major issue. The main barriers to conventional tumor chemotherapy are undesirable toxic effects and multidrug resistance. Cancer nanotherapeutics were developed to get around the drawbacks of conventional chemotherapy. Through clinical evaluation of thoughtfully developed nano delivery systems, cancer nanotherapeutics have recently offered unmatched potential to comprehend and combat drug resistance and toxicity. In different design approaches, including passive targeting, active targeting, nanomedicine, and multimodal nanomedicine combination therapy, were successful in treating cancer in this situation. Even though cancer nanotherapy has achieved considerable technological development, tumor biology complexity and heterogeneity and a lack of full knowledge of nano-bio interactions remain important hurdles to future clinical translation and commercialization. The recent developments and advancements in cancer nanotherapeutics utilizing a wide variety of nanomaterial-based platforms to overcome cancer treatment resistance are covered in this article. Additionally, an evaluation of different nanotherapeutics-based approaches to cancer treatment, such as tumor microenvironment targeted techniques, sophisticated delivery methods for the precise targeting of cancer stem cells, as well as an update on clinical studies are discussed. Lastly, the potential for cancer nanotherapeutics to overcome tumor relapse and the therapeutic effects and targeted efficacies of modern nanosystems are analyzed.
Collapse
Affiliation(s)
- Theodora Katopodi
- Department of Medicine, Laboratory of Medical Biology and Genetics, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Savvas Petanidis
- Department of Medicine, Laboratory of Medical Biology and Genetics, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Drosos Tsavlis
- Department of Medicine, Laboratory of Experimental Physiology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Doxakis Anestakis
- Department of Histology, Medical School, University of Cyprus, Nicosia, Cyprus
| | | | | | | | - Paul Zarogoulidis
- Third Department of Surgery, “AHEPA“ University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Christoforos Kosmidis
- Third Department of Surgery, “AHEPA“ University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | | | - Konstantinos Porpodis
- Pulmonary Department-Oncology Unit, “G. Papanikolaou” General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
18
|
miR-18a-5p and ATM Expression in Esophageal Squamous Cell Carcinoma and Their Correlations with Clinicopathological Features. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:5260608. [PMID: 36267307 PMCID: PMC9578886 DOI: 10.1155/2022/5260608] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 09/23/2022] [Accepted: 09/29/2022] [Indexed: 11/17/2022]
Abstract
Objective. To investigate miR-18a-5p and ataxia telangiectasia muted (ATM) expression in esophageal squamous cell carcinoma (ESCC) and their correlations with clinicopathological features. Methods. The subjects of this study were 62 ESCC patients (research group, RG) and 57 healthy controls (control group, CG) presented to our hospital between July 2019 and April 2020. Peripheral blood (PB) miR-18a-5p and ATM levels in these participants were quantified via qRT-PCR, and the correlations of the two genes with ESCC patients’ clinicopathological characteristics were investigated. In addition, a two-year follow-up was performed on ESCC patients to understand their survival, so as to further determine the prognostic utility of miR-18a-5p and ATM in ESCC. Factors influencing patient outcomes were identified by COX analysis. Results. PB miR-18a-5p expression was higher in RG compared with CG, while ATM was lower, suggesting an inverse connection between the two genes in ESCC (
). miR-18a-5p and ATM levels were determined to be strongly linked to TNM stage, differentiation degree, and lymph node metastasis in ESCC patients (
and
). The patients who succumbed to the disease exhibited higher miR-18a-5p and lower ATM than the survival (
). ROC analysis suggested favorable evaluation effects of miR-18a-5p and ATM on the occurrence and prognostic death of ESCC (
). Further, these two genes were identified by the COX analysis to be factors independently affecting the prognosis of ESCC. Conclusion. miR-18a-5p is highly expressed in ESCC, and ATM is underexpressed, both of which are closely linked to the pathological process of ESCC and have a good evaluation effect on the occurrence and prognosis of ESCC, which may become a breakthrough in future diagnosis and treatment of ESCC.
Collapse
|
19
|
Amaldoss MJN, Yang JL, Koshy P, Unnikrishnan A, Sorrell CC. Inorganic nanoparticle-based advanced cancer therapies: promising combination strategies. Drug Discov Today 2022; 27:103386. [PMID: 36182068 DOI: 10.1016/j.drudis.2022.103386] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 08/15/2022] [Accepted: 09/24/2022] [Indexed: 11/17/2022]
Abstract
Inorganic nanoparticles for drug delivery in cancer treatment offer many potential advantages because they can maximize therapeutic effect through targeting ligands while minimizing off-target side-effects through drug adsorption and infiltration. Although inorganic nanoparticles were introduced as drug carriers, they have emerged as having the capacity for combined therapeutic capabilities, including anticancer effects through cytotoxicity, suppression of oncogenes and cancer cell signaling pathway inhibition. The most promising advanced strategies for cancer therapy are as synergistic platforms for RNA interference (siRNA, miRNA, shRNA) and as synergistic drug delivery agents for the inhibition of cancer cell signaling pathways. The present work summarizes relevant current work, the promise of which is suggested by a projected compound annual growth rate of ∼20% for drug delivery alone.
Collapse
Affiliation(s)
- Maria John Newton Amaldoss
- Adult Cancer Program, Lowy Cancer Research Centre, Prince of Wales Clinical School, UNSW Sydney, Sydney, NSW 2052, Australia; School of Materials Science and Engineering, UNSW Sydney, Sydney, NSW 2052, Australia.
| | - Jia-Lin Yang
- Adult Cancer Program, Lowy Cancer Research Centre, Prince of Wales Clinical School, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Pramod Koshy
- School of Materials Science and Engineering, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Ashwin Unnikrishnan
- Adult Cancer Program, Lowy Cancer Research Centre, Prince of Wales Clinical School, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Charles C Sorrell
- School of Materials Science and Engineering, UNSW Sydney, Sydney, NSW 2052, Australia
| |
Collapse
|
20
|
Farmani AR, Salmeh MA, Golkar Z, Moeinzadeh A, Ghiasi FF, Amirabad SZ, Shoormeij MH, Mahdavinezhad F, Momeni S, Moradbeygi F, Ai J, Hardy JG, Mostafaei A. Li-Doped Bioactive Ceramics: Promising Biomaterials for Tissue Engineering and Regenerative Medicine. J Funct Biomater 2022; 13:162. [PMID: 36278631 PMCID: PMC9589997 DOI: 10.3390/jfb13040162] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/18/2022] [Accepted: 09/20/2022] [Indexed: 12/03/2022] Open
Abstract
Lithium (Li) is a metal with critical therapeutic properties ranging from the treatment of bipolar depression to antibacterial, anticancer, antiviral and pro-regenerative effects. This element can be incorporated into the structure of various biomaterials through the inclusion of Li chloride/carbonate into polymeric matrices or being doped in bioceramics. The biocompatibility and multifunctionality of Li-doped bioceramics present many opportunities for biomedical researchers and clinicians. Li-doped bioceramics (capable of immunomodulation) have been used extensively for bone and tooth regeneration, and they have great potential for cartilage/nerve regeneration, osteochondral repair, and wound healing. The synergistic effect of Li in combination with other anticancer drugs as well as the anticancer properties of Li underline the rationale that bioceramics doped with Li may be impactful in cancer treatments. The role of Li in autophagy may explain its impact in regenerative, antiviral, and anticancer research. The combination of Li-doped bioceramics with polymers can provide new biomaterials with suitable flexibility, especially as bio-ink used in 3D printing for clinical applications of tissue engineering. Such Li-doped biomaterials have significant clinical potential in the foreseeable future.
Collapse
Affiliation(s)
- Ahmad Reza Farmani
- Tissue Engineering and Applied Cell Sciences Department, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran 14166-34793, Iran
- Tissue Engineering Department, School of Advanced Technologies in Medicine, Fasa University of Medical Sciences, Fasa 74615-168, Iran
- Students’ Scientific Research Center, Tehran University of Medical Sciences, Tehran 14166-34793, Iran
| | - Mohammad Ali Salmeh
- Department of Biotechnology, School of Chemical Engineering, College of Engineering, University of Tehran, Tehran 14155-6619, Iran
| | - Zahra Golkar
- Department of Midwifery, Firoozabad Branch, Islamic Azad University, Firoozabad 74715-117, Iran
| | - Alaa Moeinzadeh
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 14496-14535, Iran
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran 14496-14535, Iran
| | - Farzaneh Farid Ghiasi
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 14496-14535, Iran
| | - Sara Zamani Amirabad
- Department of Chemical Engineering, Faculty of Engineering, Yasouj University, Yasouj 75918-74934, Iran
| | - Mohammad Hasan Shoormeij
- Emergency Medicine Department, Shariati Hospital, Tehran University of Medical Sciences, Tehran 14166-34793, Iran
| | - Forough Mahdavinezhad
- Anatomy Department, School of Medicine, Tehran University of Medical Sciences, Tehran 14166-34793, Iran
- Department of Infertility, Velayat Hospital, Qazvin University of Medical Sciences, Qazvin 34199-15315, Iran
| | - Simin Momeni
- Chemistry Department, Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz 83151-61355, Iran
| | - Fatemeh Moradbeygi
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz 71348-14336, Iran
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz 71348-14336, Iran
| | - Jafar Ai
- Tissue Engineering and Applied Cell Sciences Department, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran 14166-34793, Iran
| | - John G. Hardy
- Department of Chemistry, Faraday Building, Lancaster University, Lancaster LA1 4YB, UK
- Materials Science Institute, Lancaster University, Lancaster LA1 4YW, UK
| | - Amir Mostafaei
- Department of Mechanical, Materials, and Aerospace Engineering, Illinois Institute of Technology, 10 W 32nd Street, Chicago, IL 60616, USA
| |
Collapse
|
21
|
Waggoner LE, Kang J, Zuidema JM, Vijayakumar S, Hurtado AA, Sailor MJ, Kwon EJ. Porous Silicon Nanoparticles Targeted to the Extracellular Matrix for Therapeutic Protein Delivery in Traumatic Brain Injury. Bioconjug Chem 2022; 33:1685-1697. [PMID: 36017941 PMCID: PMC9492643 DOI: 10.1021/acs.bioconjchem.2c00305] [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] [Indexed: 11/29/2022]
Abstract
Traumatic brain injury (TBI) is a major cause of disability and death among children and young adults in the United States, yet there are currently no treatments that improve the long-term brain health of patients. One promising therapeutic for TBI is brain-derived neurotrophic factor (BDNF), a protein that promotes neurogenesis and neuron survival. However, outstanding challenges to the systemic delivery of BDNF are its instability in blood, poor transport into the brain, and short half-life in circulation and brain tissue. Here, BDNF is encapsulated into an engineered, biodegradable porous silicon nanoparticle (pSiNP) in order to deliver bioactive BDNF to injured brain tissue after TBI. The pSiNP carrier is modified with the targeting ligand CAQK, a peptide that binds to extracellular matrix components upregulated after TBI. The protein cargo retains bioactivity after release from the pSiNP carrier, and systemic administration of the CAQK-modified pSiNPs results in effective delivery of the protein cargo to injured brain regions in a mouse model of TBI. When administered after injury, the CAQK-targeted pSiNP delivery system for BDNF reduces lesion volumes compared to free BDNF, supporting the hypothesis that pSiNPs mediate therapeutic protein delivery after systemic administration to improve outcomes in TBI.
Collapse
Affiliation(s)
- Lauren E. Waggoner
- Department of Nanoengineering, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Jinyoung Kang
- Department of Nanoengineering, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Jonathan M. Zuidema
- Department of Chemistry & Biochemistry, University of California, San Diego, La Jolla, CA, 92093, USA
- Department of Neuroscience, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Sanahan Vijayakumar
- Department of Chemistry & Biochemistry, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Alan A. Hurtado
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Michael J. Sailor
- Department of Nanoengineering, University of California, San Diego, La Jolla, CA, 92093, USA
- Department of Chemistry & Biochemistry, University of California, San Diego, La Jolla, CA, 92093, USA
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Ester J. Kwon
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, 92093, USA
| |
Collapse
|
22
|
Saburi A, Kahrizi MS, Naghsh N, Etemadi H, İlhan A, Adili A, Ghoreishizadeh S, Tamjidifar R, Akbari M, Ercan G. A comprehensive survey into the role of microRNAs in ovarian cancer chemoresistance; an updated overview. J Ovarian Res 2022; 15:81. [PMID: 35799305 PMCID: PMC9264529 DOI: 10.1186/s13048-022-01012-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 06/23/2022] [Indexed: 11/30/2022] Open
Abstract
Ovarian cancer (OC), a frequent malignant tumor that affects women, is one of the leading causes of cancer-related death in this group of individuals. For the treatment of ovarian cancer, systemic chemotherapy with platinum-based drugs or taxanes is the first-line option. However, drug resistance developed over time during chemotherapy medications worsens the situation. Since uncertainty exists for the mechanism of chemotherapy resistance in ovarian cancer, there is a need to investigate and overcome this problem. miRNAs are engaged in various signaling pathways that contribute to the chemotherapeutic resistance of ovarian cancer. In the current study, we have tried to shed light on the mechanisms by which microRNAs contribute to the drug resistance of ovarian cancer and the use of some microRNAs to combat this chemoresistance, leading to the worse outcome of ovarian cancer patients treated with systemic chemotherapeutics.
Collapse
Affiliation(s)
- Ahmad Saburi
- Department of Biology, Faculty of Basic Sciences, Gonbad Kavous University, Gonbad Kavous, Iran
| | | | - Navid Naghsh
- Department of Pharmacy, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Hasti Etemadi
- Department of Biotechnology, Rajiv Gandhi Institute of IT and Biotechnology, Bharati Vidyapeeth University, Pune, India
| | - Ahmet İlhan
- Department of Medical Biochemistry, Faculty of Medicine, Cukurova University, Adana, Turkey
| | - Ali Adili
- Senior Adult Oncology Department, Moffitt Cancer Center, University of South Florida, Tampa, Florida USA
- Department of Oncology, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Rozita Tamjidifar
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Medical Biochemistry, Faculty of Medicine, Ege University, Izmir, 35100 Turkey
| | - Morteza Akbari
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Gülinnaz Ercan
- Department of Medical Biochemistry, Faculty of Medicine, Ege University, Izmir, 35100 Turkey
- Department of Stem Cell, Institute of Health Sciences, Ege University, Izmir, 35100 Turkey
| |
Collapse
|
23
|
Ma Y, Mao J, Qin H, Liang P, Huang W, Liu C, Gao J. Nano-Metal-Organic Framework Decorated With Pt Nanoparticles as an Efficient Theranostic Nanoprobe for CT/MRI/PAI Imaging-Guided Radio-Photothermal Synergistic Cancer Therapy. Front Bioeng Biotechnol 2022; 10:927461. [PMID: 35875484 PMCID: PMC9298652 DOI: 10.3389/fbioe.2022.927461] [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: 04/24/2022] [Accepted: 05/23/2022] [Indexed: 11/21/2022] Open
Abstract
The multifunctional theranostic nanoplatforms, which can realize changing the contrasts of medical images and enhance cancer therapies simultaneously, have attracted tremendous attention from chemists and medicine in past decades. Herein, a nanoscale metal-organic framework-based material was first synthesized and then decorated with platinum (NMOF545@Pt) successfully for multimodal imaging-guided synergistic cancer therapy. The obtained NMOF545@Pt is advantageous in shortening the longitudinal relaxation time (T1), enhancing photoacoustic effects, and elevating X-ray absorption efficiently. Thus, the enchantments of tripe imaging modalities, computed tomography (CT)/magnetic resonance imaging (MRI)/photoacoustic imaging (PAI), were realized with NMOF545@Pt administration simultaneously and can be cleared from the mice. Meanwhile, in vitro and in vivo experiments demonstrate that the synthesized NMOF545@Pt can dramatically increase photothermal therapy (PTT) and radiotherapy (RT) efficacy. Convincing evidence proves that tumor growth can be wholly inhibited without noticeable side effects or organ damage. The results demonstrated the promise of multifunctional nanocomposites NMOF545@Pt to improve biomedical imaging and synergistic tumor treatments.
Collapse
Affiliation(s)
- Yingjian Ma
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Jing Mao
- School of Materials Science and Engineering, Zhengzhou University, Zhengzhou, China
| | - Haojie Qin
- Forensic Medicine School of Henan University of Science and Technology, Luoyang, China
| | - Pan Liang
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wenpeng Huang
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chenchen Liu
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jianbo Gao
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
24
|
Wang W, Jo H, Park S, Kim H, Kim SI, Han Y, Lee J, Seol A, Kim J, Lee M, Lee C, Dhanasekaran DN, Ahn T, Song YS. Integrated analysis of ascites and plasma extracellular vesicles identifies a miRNA-based diagnostic signature in ovarian cancer. Cancer Lett 2022; 542:215735. [DOI: 10.1016/j.canlet.2022.215735] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 05/05/2022] [Accepted: 05/06/2022] [Indexed: 01/03/2023]
|
25
|
|
26
|
Chaix A, Griveau A, Defforge T, Grimal V, Le Borgne B, Gautier G, Eyer J. Cell penetrating peptide decorated magnetic porous silicon nanorods for glioblastoma therapy and imaging. RSC Adv 2022; 12:11708-11714. [PMID: 35432942 PMCID: PMC9008514 DOI: 10.1039/d2ra00508e] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 03/30/2022] [Indexed: 01/19/2023] Open
Abstract
Glioblastoma multiforme (GBM) is the most malignant primary brain tumor of the central nervous system. Despite advances in therapy, it remains largely untreatable, in part due to the low permeability of chemotherapeutic drugs across the blood-brain barrier (BBB) which significantly compromises their effectiveness. To circumvent the lack of drug efficiency, we designed multifunctional nanoparticles based on porous silicon. Herein, we propose an innovative synthesis technique for porous silicon nanorods (pSiNRs) with three-dimensional (3D) shape-controlled nanostructure. In order to achieve an efficient administration and improved treatment against GBM cells, a porous silicon nanoplatform is designed with magnetic guidance, fluorescence tracking and a cell-penetrating peptide (CPP). A NeuroFilament Light (NFL) subunit derived 24 amino acid tubulin binding site peptide called NFL-TBS.40-63 peptide or NFL-peptide was reported to preferentially target human GBM cells compared to healthy cells. Motivated by this approach, we investigated the use of magnetic-pSiNRs covered with superparamagnetic iron oxide nanoparticles (SPIONs) for magnetic guidance, then decorated with the NFL-peptide to facilitate targeting and enhance internalization into human GBM cells. Unexpectedly, under confocal microscope imaging, the internalized multifunctional nanoparticles in GBM cells induce a remarkable exaltation of green fluorescence instead of the red native fluorescence from the dye due to a possible Förster resonance energy transfer (FRET). In addition, we showed that the uptake of NFL-peptide decorated magnetic-pSiNRs was preferential towards human GBM cells. This study presents the fabrication of magnetic-pSiNRs decorated with the NFL-peptide, which act as a remarkable candidate to treat brain tumors. This is supported by in vitro results and confocal imaging.
Collapse
Affiliation(s)
- Arnaud Chaix
- GREMAN UMR-CNRS 7347, INSA Centre Val de Loire, Université de Tours Tours France
| | - Audrey Griveau
- MINT, INSERM, CNRS, SFR-ICAT, UNIV Angers 49000 Angers France
| | - Thomas Defforge
- GREMAN UMR-CNRS 7347, INSA Centre Val de Loire, Université de Tours Tours France
| | - Virginie Grimal
- GREMAN UMR-CNRS 7347, INSA Centre Val de Loire, Université de Tours Tours France
| | - Brice Le Borgne
- GREMAN UMR-CNRS 7347, INSA Centre Val de Loire, Université de Tours Tours France
| | - Gaël Gautier
- GREMAN UMR-CNRS 7347, INSA Centre Val de Loire, Université de Tours Tours France
| | - Joël Eyer
- MINT, INSERM, CNRS, SFR-ICAT, UNIV Angers 49000 Angers France
| |
Collapse
|
27
|
Fu C, Wang X, Xue F, Zhu P, Zhou W, Ge S, Yu J. Laser ablative TiO 2 and tremella-like CuInS 2 nanocomposites for robust and ultrasensitive photoelectrochemical sensing of let-7a. Mikrochim Acta 2022; 189:145. [PMID: 35296924 DOI: 10.1007/s00604-022-05178-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Accepted: 01/05/2022] [Indexed: 10/18/2022]
Abstract
A photoelectrochemical (PEC) biosensor based on a multiple signal amplification strategy was established for highly sensitive detection of microRNA (miRNA). TiO2 was prepared on the surface of titanium sheet by laser etching to improve its stability and photoelectrical properties, and CuInS2-sensitized TiO2 was used to form a superior photoelectrical layer, which realized the initial signal amplification. The electron donor dopamine (DA) was modified to H2 as a signal regulator, which effectively increased the photocurrent signal. To further amplify the signal, an enzyme-free hybridization reaction was implemented. When target let-7a and fuel-DNA (F-DNA) were present, the base of H1 specifically recognized let-7a and forced dopamine@AuNPs-H2 away from the electrode surface. Subsequently, the end base of H1 specifically recognized F-DNA, and let-7a was replaced and recycled to participate in the next cycle. Enzyme-free circulation, as a multifunctional amplification method, ensured the recycling of target molecules. This PEC sensor for let-7a detection showed an excellent linear response from 0.5 to 1000 pM with a detection limit of 0.12 pM. The intra-batch RSD was 3.8% and the recovery was 87.74-108.1%. The sensor was further used for clinical biomolecular monitoring of miRNA, showing excellent quantitative detection capability.
Collapse
Affiliation(s)
- Cuiping Fu
- Collaborative Innovation Center of Technology and Equipment for Biological Diagnosis and Therapy in Universities of Shandong, Institute for Advanced Interdisciplinary Research, University of Jinan, Jinan, 250022, People's Republic of China
| | - Xuefeng Wang
- Collaborative Innovation Center of Technology and Equipment for Biological Diagnosis and Therapy in Universities of Shandong, Institute for Advanced Interdisciplinary Research, University of Jinan, Jinan, 250022, People's Republic of China
| | - Fumin Xue
- Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250014, People's Republic of China
| | - Peihua Zhu
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan, 250022, People's Republic of China.
| | - Weijia Zhou
- Collaborative Innovation Center of Technology and Equipment for Biological Diagnosis and Therapy in Universities of Shandong, Institute for Advanced Interdisciplinary Research, University of Jinan, Jinan, 250022, People's Republic of China.
| | - Shenguang Ge
- Collaborative Innovation Center of Technology and Equipment for Biological Diagnosis and Therapy in Universities of Shandong, Institute for Advanced Interdisciplinary Research, University of Jinan, Jinan, 250022, People's Republic of China.
| | - Jinghua Yu
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan, 250022, People's Republic of China
| |
Collapse
|
28
|
Wu L, Zhou W, Lin L, Chen A, Feng J, Qu X, Zhang H, Yue J. Delivery of therapeutic oligonucleotides in nanoscale. Bioact Mater 2022; 7:292-323. [PMID: 34466734 PMCID: PMC8379367 DOI: 10.1016/j.bioactmat.2021.05.038] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 04/28/2021] [Accepted: 05/22/2021] [Indexed: 02/07/2023] Open
Abstract
Therapeutic oligonucleotides (TOs) represent one of the most promising drug candidates in the targeted cancer treatment due to their high specificity and capability of modulating cellular pathways that are not readily druggable. However, efficiently delivering of TOs to cancer cellular targets is still the biggest challenge in promoting their clinical translations. Emerging as a significant drug delivery vector, nanoparticles (NPs) can not only protect TOs from nuclease degradation and enhance their tumor accumulation, but also can improve the cell uptake efficiency of TOs as well as the following endosomal escape to increase the therapeutic index. Furthermore, targeted and on-demand drug release of TOs can also be approached to minimize the risk of toxicity towards normal tissues using stimuli-responsive NPs. In the past decades, remarkable progresses have been made on the TOs delivery based on various NPs with specific purposes. In this review, we will first give a brief introduction on the basis of TOs as well as the action mechanisms of several typical TOs, and then describe the obstacles that prevent the clinical translation of TOs, followed by a comprehensive overview of the recent progresses on TOs delivery based on several various types of nanocarriers containing lipid-based nanoparticles, polymeric nanoparticles, gold nanoparticles, porous nanoparticles, DNA/RNA nanoassembly, extracellular vesicles, and imaging-guided drug delivery nanoparticles.
Collapse
Affiliation(s)
- Lei Wu
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, 510006, Guangdong, China
| | - Wenhui Zhou
- Pharmaceutical Sciences Laboratory and Turku Bioscience Centre, Åbo Akademi University, Turku, 20520, Finland
- Southern Medical University Affiliated Fengxian Hospital, Shanghai, 201499, China
| | - Lihua Lin
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, 510006, Guangdong, China
| | - Anhong Chen
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, 510006, Guangdong, China
| | - Jing Feng
- Southern Medical University Affiliated Fengxian Hospital, Shanghai, 201499, China
| | - Xiangmeng Qu
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, 510006, Guangdong, China
| | - Hongbo Zhang
- Pharmaceutical Sciences Laboratory and Turku Bioscience Centre, Åbo Akademi University, Turku, 20520, Finland
| | - Jun Yue
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, 510006, Guangdong, China
| |
Collapse
|
29
|
Sempere LF, Azmi AS, Moore A. microRNA-based diagnostic and therapeutic applications in cancer medicine. WILEY INTERDISCIPLINARY REVIEWS. RNA 2021; 12:e1662. [PMID: 33998154 PMCID: PMC8519065 DOI: 10.1002/wrna.1662] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 04/10/2021] [Accepted: 04/12/2021] [Indexed: 01/18/2023]
Abstract
It has been almost two decades since the first link between microRNAs and cancer was established. In the ensuing years, this abundant class of short noncoding regulatory RNAs has been studied in virtually all cancer types. This tremendously large body of research has generated innovative technological advances for detection of microRNAs in tissue and bodily fluids, identified the diagnostic, prognostic, and/or predictive value of individual microRNAs or microRNA signatures as potential biomarkers for patient management, shed light on regulatory mechanisms of RNA-RNA interactions that modulate gene expression, uncovered cell-autonomous and cell-to-cell communication roles of specific microRNAs, and developed a battery of viral and nonviral delivery approaches for therapeutic intervention. Despite these intense and prolific research efforts in preclinical and clinical settings, there are a limited number of microRNA-based applications that have been incorporated into clinical practice. We review recent literature and ongoing clinical trials that highlight most promising approaches and standing challenges to translate these findings into viable microRNA-based clinical tools for cancer medicine. This article is categorized under: RNA in Disease and Development > RNA in Disease.
Collapse
Affiliation(s)
- Lorenzo F. Sempere
- Department of Radiology, Precision Health ProgramMichigan State UniversityEast LansingMichiganUSA
| | - Asfar S. Azmi
- Department of OncologyWayne State University School of MedicineDetroitMichiganUSA
- Karmanos Cancer InstituteDetroitMichiganUSA
| | - Anna Moore
- Departments of Radiology and Physiology, Precision Health ProgramMichigan State UniversityEast LansingMichiganUSA
| |
Collapse
|
30
|
Neri M, Kang J, Zuidema JM, Gasparello J, Finotti A, Gambari R, Sailor MJ, Bertucci A, Corradini R. Tuning the Loading and Release Properties of MicroRNA-Silencing Porous Silicon Nanoparticles by Using Chemically Diverse Peptide Nucleic Acid Payloads. ACS Biomater Sci Eng 2021; 8:4123-4131. [PMID: 34468123 PMCID: PMC9554869 DOI: 10.1021/acsbiomaterials.1c00431] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Peptide nucleic acids (PNAs) are a class of artificial oligonucleotide mimics that have garnered much attention as precision biotherapeutics for their efficient hybridization properties and their exceptional biological and chemical stability. However, the poor cellular uptake of PNA is a limiting factor to its more extensive use in biomedicine; encapsulation in nanoparticle carriers has therefore emerged as a strategy for internalization and delivery of PNA in cells. In this study, we demonstrate that PNA can be readily loaded into porous silicon nanoparticles (pSiNPs) following a simple salt-based trapping procedure thus far employed only for negatively charged synthetic oligonucleotides. We show that the ease and versatility of PNA chemistry also allows for producing PNAs with different net charge, from positive to negative, and that the use of differently charged PNAs enables optimization of loading into pSiNPs. Differently charged PNA payloads determine different release kinetics and allow modulation of the temporal profile of the delivery process. In vitro silencing of a set of specific microRNAs using a pSiNP-PNA delivery platform demonstrates the potential for biomedical applications.
Collapse
Affiliation(s)
- Martina Neri
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parco Area delle Scienze 17/A, 43124 Parma, Italy
| | - Jinyoung Kang
- Department of Nanoengineering, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Jonathan M Zuidema
- Department of Chemistry and Biochemistry and Department of Neurosciences, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Jessica Gasparello
- Department of Life Sciences and Biotechnology, University of Ferrara, Via Fossato di Mortara 74, 44121 Ferrara, Italy
| | - Alessia Finotti
- Department of Life Sciences and Biotechnology, University of Ferrara, Via Fossato di Mortara 74, 44121 Ferrara, Italy
| | - Roberto Gambari
- Department of Life Sciences and Biotechnology, University of Ferrara, Via Fossato di Mortara 74, 44121 Ferrara, Italy
| | - Michael J Sailor
- Department of Chemistry and Biochemistry, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Alessandro Bertucci
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parco Area delle Scienze 17/A, 43124 Parma, Italy
| | - Roberto Corradini
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parco Area delle Scienze 17/A, 43124 Parma, Italy
| |
Collapse
|
31
|
Yin L, Wang Y. Long non-coding RNA NEAT1 facilitates the growth, migration, and invasion of ovarian cancer cells via the let-7 g/MEST/ATGL axis. Cancer Cell Int 2021; 21:437. [PMID: 34416900 PMCID: PMC8379830 DOI: 10.1186/s12935-021-02018-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 06/10/2021] [Indexed: 12/14/2022] Open
Abstract
Background/Aim Growing evidence indicates a significant role of long non-coding RNA (lncRNA) nuclear-enriched abundant transcript 1 (NEAT1) in ovarian cancer, a frequently occurring malignant tumor in women; however, the possible effects of an interplay of NEAT1 with microRNA (miRNA or miR) let-7 g in ovarian cancer are not known. The current study aimed to investigate the role of the NEAT1/let-7 g axis in the growth, migration, and invasion of ovarian cancer cells and explore underlying mechanisms. Methods NEAT1 expression levels were examined in clinical tissue samples and cell lines. The relationships between NEAT1, let-7 g, and MEST were then analyzed. Gain- or loss-of-function approaches were used to manipulate NEAT1 and let-7 g. The effects of NEAT1 on cell proliferation, migration, invasion, and apoptosis were evaluated. Mouse xenograft models of ovarian cancer cells were established to verify the function of NEAT1 in vivo. Results NEAT1 expression was elevated while let-7 g was decreased in ovarian cancer clinical tissue samples and cell lines. A negative correlation existed between NEAT1 and let-7 g, whereby NEAT1 competitively bound to let-7 g and consequently down-regulate let-7 g expression. By this mechanism, the growth, migration, and invasion of ovarian cancer cells were stimulated. In addition, let-7 g targeted mesoderm specific transcript (MEST) and inhibited its expression, leading to promotion of adipose triglyceride lipase (ATGL) expression and inhibition of ovarian cancer cell growth, migration, and invasion. However, the effect of let-7 g was abolished by overexpression of MEST. Furthermore, silencing of NEAT1 decreased the xenograft tumor growth by decreasing MEST while up-regulating let-7 g and ATGL. Conclusions Cumulatively, the findings demonstrated that NEAT1 could promote malignant phenotypes of ovarian cancer cells by regulating the let-7 g/MEST/ATGL signaling axis. Therefore, NEAT1 can be regarded as an important molecular target and biomarker for ovarian cancer. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-021-02018-3.
Collapse
Affiliation(s)
- Lili Yin
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Heping District, Shenyang, Liaoning Province, 110004, P.R. China
| | - Yu Wang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Heping District, Shenyang, Liaoning Province, 110004, P.R. China.
| |
Collapse
|
32
|
Xie W, Sun H, Li X, Lin F, Wang Z, Wang X. Ovarian cancer: epigenetics, drug resistance, and progression. Cancer Cell Int 2021; 21:434. [PMID: 34404407 PMCID: PMC8369623 DOI: 10.1186/s12935-021-02136-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Accepted: 08/03/2021] [Indexed: 03/05/2023] Open
Abstract
Ovarian cancer (OC) is one of the most common malignant tumors in women. OC is associated with the activation of oncogenes, the inactivation of tumor suppressor genes, and the activation of abnormal cell signaling pathways. Moreover, epigenetic processes have been found to play an important role in OC tumorigenesis. Epigenetic processes do not change DNA sequences but regulate gene expression through DNA methylation, histone modification, and non-coding RNA. This review comprehensively considers the importance of epigenetics in OC, with a focus on microRNA and long non-coding RNA. These types of RNA are promising molecular markers and therapeutic targets that may support precision medicine in OC. DNA methylation inhibitors and histone deacetylase inhibitors may be useful for such targeting, with a possible novel approach combining these two therapies. Currently, the clinical application of such epigenetic approaches is limited by multiple obstacles, including the heterogeneity of OC, insufficient sample sizes in reported studies, and non-optimized methods for detecting potential tumor markers. Nonetheless, the application of epigenetic approaches to OC patient diagnosis, treatment, and prognosis is a promising area for future clinical investigation.
Collapse
Affiliation(s)
- Weiwei Xie
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University School of Medicine Xinhua Hospital, 1665 Kongjiang Road, Yangpu District, Shanghai, China
| | - Huizhen Sun
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University School of Medicine Xinhua Hospital, 1665 Kongjiang Road, Yangpu District, Shanghai, China
| | - Xiaoduan Li
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Feikai Lin
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University School of Medicine Xinhua Hospital, 1665 Kongjiang Road, Yangpu District, Shanghai, China
| | - Ziliang Wang
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University School of Medicine Xinhua Hospital, 1665 Kongjiang Road, Yangpu District, Shanghai, China.
| | - Xipeng Wang
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University School of Medicine Xinhua Hospital, 1665 Kongjiang Road, Yangpu District, Shanghai, China.
| |
Collapse
|
33
|
Kolanthai E, Fu Y, Kumar U, Babu B, Venkatesan AK, Liechty KW, Seal S. Nanoparticle mediated RNA delivery for wound healing. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2021; 14:e1741. [PMID: 34369096 DOI: 10.1002/wnan.1741] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 07/05/2021] [Accepted: 07/07/2021] [Indexed: 12/19/2022]
Abstract
Wound healing is a complicated physiological process that comprises various steps, including hemostasis, inflammation, proliferation, and remodeling. The wound healing process is significantly affected by coexisting disease states such as diabetes, immunosuppression, or vascular disease. It can also be impacted by age, repeated injury, or hypertrophic scarring. These comorbidities can affect the rate of wound closure, the quality of wound closure, and tissues' function at the affected sites. There are limited options to improve the rate or quality of wound healing, creating a significant unmet need. Advances in nucleic acid research and the human genome project have developed potential novel approaches to address these outstanding requirements. In particular, the use of microRNA, short hairpin RNA, and silencing RNA is unique in their abilities as key regulators within the physiologic machinery of the cell. Although this innovative therapeutic approach using ribonucleic acid (RNA) is an attractive approach, the application as a therapeutic remains a challenge due to site-specific delivery, off-target effects, and RNA degradation obstacles. An ideal delivery system is essential for successful gene delivery. An ideal delivery system should result in high bioactivity, inhibit rapid dilution, controlled release, allow specific activation timings facilitating physiological stability, and minimize multiple dosages. Currently, these goals can be achieved by inorganic nanoparticle (NP) (e.g., cerium oxide, gold, silica, etc.) based delivery systems. This review focuses on providing insight into the preeminent research carried out on various RNAs and their delivery through NPs for effective wound healing. This article is categorized under: Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Therapeutic Approaches and Drug Discovery > Emerging Technologies Biology-Inspired Nanomaterials > Nucleic Acid-Based Structures.
Collapse
Affiliation(s)
- Elayaraja Kolanthai
- Advanced Materials Processing and Analysis Center, Department of Materials Science and Engineering, University of Central Florida, Orlando, Florida, USA
| | - Yifei Fu
- Advanced Materials Processing and Analysis Center, Department of Materials Science and Engineering, University of Central Florida, Orlando, Florida, USA
| | - Udit Kumar
- Advanced Materials Processing and Analysis Center, Department of Materials Science and Engineering, University of Central Florida, Orlando, Florida, USA
| | - Balaashwin Babu
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida, USA
| | | | - Kenneth W Liechty
- Laboratory for Fetal and Regenerative Biology, Department of Surgery, University of Colorado Denver School of Medicine and Children's Hospital Colorado, Aurora, Colorado, USA
| | - Sudipta Seal
- Advanced Materials Processing and Analysis Center, Department of Materials Science and Engineering, University of Central Florida, Orlando, Florida, USA.,College of Medicine, Nanoscience Technology Center, Biionix Cluster, University of Central Florida, Orlando, Florida, USA
| |
Collapse
|
34
|
Jung Y, Kim D. Recent advances in hybrid system of porous silicon nanoparticles and biocompatible polymers for biomedical applications. Biomed Eng Lett 2021; 11:171-181. [PMID: 34350046 PMCID: PMC8316517 DOI: 10.1007/s13534-021-00194-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 06/08/2021] [Accepted: 06/09/2021] [Indexed: 11/28/2022] Open
Abstract
Hybrid systems of nanoparticles and polymers have emerged as a new material in the biomedical field. To date, various kinds of hybrid systems have been introduced and applied to drug delivery, regenerative medicine, therapeutics, disease diagnosis, and medical implantation. Among them, the hybridization of nanostructured porous silicon nanoparticles (pSiNPs) and biocompatible polymers has been highlighted due to its unique biological and physicochemical properties. This review focuses on the recent advances in the hybrid systems of pSiNPs and biocompatible polymers from an engineering aspect and its biomedical applications. Representative hybrid formulations, (i) Polymer-coated pSiNPs, (ii) pSiNPs-embedded polymeric nanofibers, are outlined along with their preparation methods, biomedical applications, and future perspectives. We believe this review provides insight into a new hybrid system of pSiNPs and biocompatible polymers as a promising nano-platform for further biomedical applications. Recently developed and representative hybrid systems of porous silicon nanoparticles and biocompatible polymers and their biomedical applications are introduced.
Collapse
Affiliation(s)
- Yuna Jung
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, 02447 Republic of Korea
| | - Dokyoung Kim
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, 02447 Republic of Korea
- Department of Anatomy and Neurobiology, College of Medicine, Kyung Hee University, Seoul, 02447 Republic of Korea
- Center for Converging Humanities, Kyung Hee University, Seoul, 02447 Republic of Korea
| |
Collapse
|
35
|
Tieu T, Wei Y, Cifuentes‐Rius A, Voelcker NH. Overcoming Barriers: Clinical Translation of siRNA Nanomedicines. ADVANCED THERAPEUTICS 2021. [DOI: 10.1002/adtp.202100108] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Terence Tieu
- Parkville Campus 381 Royal Parade Monash Institute of Pharmaceutical Sciences Monash University Parkville VIC 3052 Australia
- CSIRO Manufacturing Bayview Avenue Clayton VIC 3168 Australia
| | - Yingkai Wei
- Parkville Campus 381 Royal Parade Monash Institute of Pharmaceutical Sciences Monash University Parkville VIC 3052 Australia
| | - Anna Cifuentes‐Rius
- Parkville Campus 381 Royal Parade Monash Institute of Pharmaceutical Sciences Monash University Parkville VIC 3052 Australia
| | - Nicolas H. Voelcker
- Parkville Campus 381 Royal Parade Monash Institute of Pharmaceutical Sciences Monash University Parkville VIC 3052 Australia
- CSIRO Manufacturing Bayview Avenue Clayton VIC 3168 Australia
- Melbourne Centre for Nanofabrication 151 Wellington Road Victorian Node of the Australian National Fabrication Facility Clayton VIC 3168 Australia
| |
Collapse
|
36
|
Kang RH, Park J, Kim J, Chowdhury T, Oh JH, Kim J, Shin J, Kim M, Park CK, Lee S, Lee JY, Kim D. A Deep Dive: SIWV Tetra-Peptide Enhancing the Penetration of Nanotherapeutics into the Glioblastoma. ACS Biomater Sci Eng 2021; 8:4163-4174. [PMID: 34196517 DOI: 10.1021/acsbiomaterials.1c00653] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Glioblastoma multiforme (GBM) is the most aggressive malignant tumor. It is difficult to regulate GBM using conventional chemotherapy-based methods due to its anatomical structure specificity, low drug targeting ability, and limited penetration depth capability to reach the tumor interior. Numerous approaches have been proposed to overcome such issues, including nanoparticle-based drug delivery system (DDS) with the development of GBM site targeting and penetration depth enhancing moieties (e.g., peptides, sugars, proteins, etc.). In this study, we prepared four different types of nanoparticles, which are based on porous silicon nanoparticles (pSiNPs) incorporating polyethylene glycol (PEG), iRGD peptide (well-known cancer targeting peptide), and SIWV tetra-peptide (a recently disclosed GBM-targeting peptide), and analyzed their deep-tumor penetration abilities in cell spheroids, in GBM patient-derived tumoroids, and in GBM xenograft mice. We found that SIWV tetra-peptide significantly enhanced the penetration depth of pSiNPs, and its therapeutic formulation (temozolomide-loaded/SIWV-functionalized pSiNPs) showed a higher anticancer efficacy compared with other formulations. These findings hold great promise for the development of nanotherapeutics and peptide-conjugated drugs for GBM.
Collapse
Affiliation(s)
- Rae Hyung Kang
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Jinju Park
- Neural Development and Anomaly Laboratory, Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Jieun Kim
- Neural Development and Anomaly Laboratory, Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Tamrin Chowdhury
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Ji Hyeon Oh
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Jaehoon Kim
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Jaeha Shin
- R&D Center, Pensees Inc., Seoul 04043, Republic of Korea
| | - Minji Kim
- R&D Center, Pensees Inc., Seoul 04043, Republic of Korea
| | - Chul-Kee Park
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Sungjun Lee
- R&D Center, Pensees Inc., Seoul 04043, Republic of Korea
| | - Ji Yeoun Lee
- Neural Development and Anomaly Laboratory, Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea.,Division of Pediatric Neurosurgery, Seoul National University Children's Hospital, Seoul 03080, Republic of Korea
| | - Dokyoung Kim
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea.,Department of Anatomy and Neurobiology, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea.,Center for Converging Humanities, Kyung Hee University, Seoul 02447, Republic of Korea.,Medical Research Center for Bioreaction to Reactive Oxygen Species and Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea.,KHU-KIST Department of Converging Science and Technology, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
37
|
Zhao J, Zhang L, Qi Y, Liao K, Wang Z, Wen M, Zhou D. NIR Laser Responsive Nanoparticles for Ovarian Cancer Targeted Combination Therapy with Dual-Modal Imaging Guidance. Int J Nanomedicine 2021; 16:4351-4369. [PMID: 34234430 PMCID: PMC8254569 DOI: 10.2147/ijn.s299376] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 06/01/2021] [Indexed: 01/04/2023] Open
Abstract
Purpose Multifunctional nanoparticles with targeted therapeutic function and diagnostic-imaging are of great interest in the domain of precision therapy. NIR laser responsive nanoparticles (PLGA-PEG-FA encapsulating Bi2S3, PFP, and Dox (designed as FBPD NPs)) are synthesized for ovarian cancer targeted combination therapy with CT/PA dual-modal imaging guidance (PA: photoacoustic; CT: X-ray computed tomography). Methods and Results The FBPD NPS prepared by the double emulsification method revealed excellent dispersity, great stability, outstanding optical properties. The temperature of FBPD NPs increased rapidly after laser irradiation, inducing liquid-to-gas conversion of perfluoropentane (PFP), and promoting the release of Dox up to 86.7%. These FBPD NPs demonstrated their outstanding imaging capability for both PA and CT imaging both in vitro and in vivo, providing the potential for therapeutic guidance and monitoring. Assisted by folic acid, these nanoparticles could highly enrich in ovarian tumor tissue and the accumulation peaked at 3 h after intravenous administration. The desirable photothermal-conversion efficiency of the nanoparticles combined with chemotherapy achieved highly efficient therapy, which was demonstrated both in vitro and in vivo. Conclusion We successfully constructed multifunctional theranostic FBPD NPs for highly efficient PTT/chemotherapy combined therapy with dual CT/PA imaging guidance/monitoring. The unique nanoparticles with multiple abilities pave an emerging way toward precise treatment of ovarian cancer.
Collapse
Affiliation(s)
- Jiawen Zhao
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Liang Zhang
- Department of Ultrasound, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Yingjie Qi
- Department of Intensive Care Unit (ICU), Dianjiang People's Hospital of Chongqing, Chongqing, People's Republic of China
| | - Kui Liao
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Zhigang Wang
- Institute of Ultrasound Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Ming Wen
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Di Zhou
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| |
Collapse
|
38
|
Reda El Sayed S, Cristante J, Guyon L, Denis J, Chabre O, Cherradi N. MicroRNA Therapeutics in Cancer: Current Advances and Challenges. Cancers (Basel) 2021; 13:cancers13112680. [PMID: 34072348 PMCID: PMC8198729 DOI: 10.3390/cancers13112680] [Citation(s) in RCA: 108] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/21/2021] [Accepted: 05/24/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Cancer is a complex disease associated with deregulation of numerous genes. In addition, redundant cellular pathways limit efficiency of monotarget drugs in cancer therapy. MicroRNAs are a class of gene expression regulators, which often function by targeting multiple genes. This feature makes them a double-edged sword (a) as attractive targets for anti-tumor therapy and concomitantly (b) as risky targets due to their potential side effects on healthy tissues. As for conventional antitumor drugs, nanocarriers have been developed to circumvent the problems associated with miRNA delivery to tumors. In this review, we highlight studies that have established the pre-clinical proof-of concept of miRNAs as relevant therapeutic targets in oncology. Particular attention was brought to new strategies based on nanovectorization of miRNAs as well as to the perspectives for their applications. Abstract The discovery of microRNAs (miRNAs) in 1993 has challenged the dogma of gene expression regulation. MiRNAs affect most of cellular processes from metabolism, through cell proliferation and differentiation, to cell death. In cancer, deregulated miRNA expression leads to tumor development and progression by promoting acquisition of cancer hallmark traits. The multi-target action of miRNAs, which enable regulation of entire signaling networks, makes them attractive tools for the development of anti-cancer therapies. Hence, supplementing downregulated miRNA by synthetic oligonucleotides or silencing overexpressed miRNAs through artificial antagonists became a common strategy in cancer research. However, the ultimate success of miRNA therapeutics will depend on solving pharmacokinetic and targeted delivery issues. The development of a number of nanocarrier-based platforms holds significant promises to enhance the cell specific controlled delivery and safety profile of miRNA-based therapies. In this review, we provide among the most comprehensive assessments to date of promising nanomedicine platforms that have been tested preclinically, pertaining to the treatment of selected solid tumors including lung, liver, breast, and glioblastoma tumors as well as endocrine malignancies. The future challenges and potential applications in clinical oncology are discussed.
Collapse
Affiliation(s)
- Soha Reda El Sayed
- University Grenoble Alpes, INSERM, CEA, Interdisciplinary Research Institute of Grenoble (IRIG), Biology and Biotechnologies for Health UMR_1292, F-38000 Grenoble, France; (S.R.E.S.); (J.C.); (L.G.); (J.D.); (O.C.)
| | - Justine Cristante
- University Grenoble Alpes, INSERM, CEA, Interdisciplinary Research Institute of Grenoble (IRIG), Biology and Biotechnologies for Health UMR_1292, F-38000 Grenoble, France; (S.R.E.S.); (J.C.); (L.G.); (J.D.); (O.C.)
- Centre Hospitalier Universitaire Grenoble Alpes, Service d’Endocrinologie, F-38000 Grenoble, France
| | - Laurent Guyon
- University Grenoble Alpes, INSERM, CEA, Interdisciplinary Research Institute of Grenoble (IRIG), Biology and Biotechnologies for Health UMR_1292, F-38000 Grenoble, France; (S.R.E.S.); (J.C.); (L.G.); (J.D.); (O.C.)
| | - Josiane Denis
- University Grenoble Alpes, INSERM, CEA, Interdisciplinary Research Institute of Grenoble (IRIG), Biology and Biotechnologies for Health UMR_1292, F-38000 Grenoble, France; (S.R.E.S.); (J.C.); (L.G.); (J.D.); (O.C.)
| | - Olivier Chabre
- University Grenoble Alpes, INSERM, CEA, Interdisciplinary Research Institute of Grenoble (IRIG), Biology and Biotechnologies for Health UMR_1292, F-38000 Grenoble, France; (S.R.E.S.); (J.C.); (L.G.); (J.D.); (O.C.)
- Centre Hospitalier Universitaire Grenoble Alpes, Service d’Endocrinologie, F-38000 Grenoble, France
| | - Nadia Cherradi
- University Grenoble Alpes, INSERM, CEA, Interdisciplinary Research Institute of Grenoble (IRIG), Biology and Biotechnologies for Health UMR_1292, F-38000 Grenoble, France; (S.R.E.S.); (J.C.); (L.G.); (J.D.); (O.C.)
- Correspondence: ; Tel.: +33-(0)4-38783501; Fax: +33-(0)4-38785058
| |
Collapse
|
39
|
Pudakalakatti S, Enriquez JS, McCowan C, Ramezani S, Davis JS, Zacharias NM, Bourgeois D, Constantinou PE, Harrington DA, Carson D, Farach-Carson MC, Bhattacharya PK. Hyperpolarized MRI with silicon micro and nanoparticles: Principles and applications. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2021; 13:e1722. [PMID: 33982426 DOI: 10.1002/wnan.1722] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 04/18/2021] [Accepted: 04/19/2021] [Indexed: 11/08/2022]
Abstract
Silicon-based micro and nanoparticles are ideally suited for use as biomedical imaging agents because of their biocompatibility, biodegradability, and simple surface chemistry that facilitates drug loading and targeting. A method to hyperpolarize silicon particles using dynamic nuclear polarization (DNP), which increases magnetic resonance (MR) imaging signals by several orders-of-magnitude through enhanced nuclear spin alignment, was developed to allow silicon particles to function as contrast agents for in vivo magnetic resonance imaging. In this review, we describe the application of the DNP technique to silicon particles and nanoparticles for background-free real-time molecular MR imaging. This review provides a summary of the state-of-the-science in silicon particle hyperpolarization with a detailed protocol for hyperpolarizing silicon particles. This information will foster awareness and spur interest in this emerging area of nanoimaging and provide a path to new developments and discoveries to further advance the field. This article is categorized under: Diagnostic Tools > In Vivo Nanodiagnostics and Imaging Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Therapeutic Approaches and Drug Discovery > Emerging Technologies.
Collapse
Affiliation(s)
- Shivanand Pudakalakatti
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - José S Enriquez
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.,MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas, USA
| | - Caitlin McCowan
- Department of Electrical and Computer Engineering, Rice University, Houston, Texas, USA.,Department of Diagnostic and Biomedical Sciences, The University of Texas Health Science Center, School of Dentistry, Houston, Texas, USA
| | - Saleh Ramezani
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas, USA.,Department of Diagnostic and Biomedical Sciences, The University of Texas Health Science Center, School of Dentistry, Houston, Texas, USA
| | - Jennifer S Davis
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Niki M Zacharias
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas, USA.,Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Dontrey Bourgeois
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.,Department of Statistics, Rice University, Houston, Texas, USA
| | - Pamela E Constantinou
- Department of BioSciences, Rice University, Houston, Texas, USA.,Sheikh Ahmed Center for Pancreatic Cancer Research, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Daniel A Harrington
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas, USA.,Department of Diagnostic and Biomedical Sciences, The University of Texas Health Science Center, School of Dentistry, Houston, Texas, USA.,Department of BioSciences, Rice University, Houston, Texas, USA
| | - Daniel Carson
- Department of BioSciences, Rice University, Houston, Texas, USA
| | - Mary C Farach-Carson
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas, USA.,Department of Diagnostic and Biomedical Sciences, The University of Texas Health Science Center, School of Dentistry, Houston, Texas, USA.,Department of BioSciences, Rice University, Houston, Texas, USA
| | - Pratip K Bhattacharya
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.,MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas, USA
| |
Collapse
|
40
|
Shafiei N, Nasrollahzadeh M, Iravani S. Green Synthesis of Silica and Silicon Nanoparticles and Their Biomedical and Catalytic Applications. COMMENT INORG CHEM 2021. [DOI: 10.1080/02603594.2021.1904912] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Nasrin Shafiei
- Department of Chemistry, Faculty of Science, University of Qom, Qom, Iran
| | | | - Siavash Iravani
- Faculty of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
41
|
Dwivedi SKD, Rao G, Dey A, Mukherjee P, Wren JD, Bhattacharya R. Small Non-Coding-RNA in Gynecological Malignancies. Cancers (Basel) 2021; 13:1085. [PMID: 33802524 PMCID: PMC7961667 DOI: 10.3390/cancers13051085] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 02/18/2021] [Accepted: 02/25/2021] [Indexed: 12/12/2022] Open
Abstract
Gynecologic malignancies, which include cancers of the cervix, ovary, uterus, vulva, vagina, and fallopian tube, are among the leading causes of female mortality worldwide, with the most prevalent being endometrial, ovarian, and cervical cancer. Gynecologic malignancies are complex, heterogeneous diseases, and despite extensive research efforts, the molecular mechanisms underlying their development and pathology remain largely unclear. Currently, mechanistic and therapeutic research in cancer is largely focused on protein targets that are encoded by about 1% of the human genome. Our current understanding of 99% of the genome, which includes noncoding RNA, is limited. The discovery of tens of thousands of noncoding RNAs (ncRNAs), possessing either structural or regulatory functions, has fundamentally altered our understanding of genetics, physiology, pathophysiology, and disease treatment as they relate to gynecologic malignancies. In recent years, it has become clear that ncRNAs are relatively stable, and can serve as biomarkers for cancer diagnosis and prognosis, as well as guide therapy choices. Here we discuss the role of small non-coding RNAs, i.e., microRNAs (miRs), P-Element induced wimpy testis interacting (PIWI) RNAs (piRNAs), and tRNA-derived small RNAs in gynecological malignancies, specifically focusing on ovarian, endometrial, and cervical cancer.
Collapse
Affiliation(s)
- Shailendra Kumar Dhar Dwivedi
- Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (S.K.D.D.); (A.D.)
| | - Geeta Rao
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (G.R.); (P.M.)
| | - Anindya Dey
- Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (S.K.D.D.); (A.D.)
| | - Priyabrata Mukherjee
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (G.R.); (P.M.)
- Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Jonathan D. Wren
- Biochemistry and Molecular Biology Department, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
- Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Resham Bhattacharya
- Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (S.K.D.D.); (A.D.)
- Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Department of Cell Biology, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
42
|
Zafar A, Alruwaili NK, Imam SS, Alharbi KS, Afzal M, Alotaibi NH, Yasir M, Elmowafy M, Alshehri S. Novel nanotechnology approaches for diagnosis and therapy of breast, ovarian and cervical cancer in female: A review. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2020.102198] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
43
|
Pan H, Ding Y, Jiang Y, Wang X, Rao J, Zhang X, Yu H, Hou Q, Li T. LncRNA LIFR-AS1 promotes proliferation and invasion of gastric cancer cell via miR-29a-3p/COL1A2 axis. Cancer Cell Int 2021; 21:7. [PMID: 33407453 PMCID: PMC7789183 DOI: 10.1186/s12935-020-01644-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 11/09/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND LncRNA was known to be closely associated with the progression of human tumors. The role of lncRNA LIFR-AS1 in the pathogenesis and progression of gastric tumor is still unclear. The aim of this study was to investigate the function of LIFR-AS1 and the underlying mechanism in the pathogenesis and progression of gastric cancer. METHODS QRT-PCR was used to evaluate the expression of LIFR-AS1, miR-29a-3p and COL1A2 in gastric tumor tissues and cells. Western blotting was used to evaluate the protein expression of COL1A2 in gastric tumor cells. CCK-8 assay, transwell assay and flow cytometry were used to evaluate the roles of LIFR-AS1, miR-29a-3p and COL1A2 in cell proliferation, invasion, migration and apoptosis. The relationship among LIFR-AS1, miR-29a-3p and COL1A2 was assessed by bioinformatics analyses and luciferase reporter assay. RESULTS The expression levels of LIFR-AS1 were significantly increased in gastric tumor tissues and cells, while the expression levels of miR-29a-3p were decreased. The expression of miR-29a-3p was negatively correlated with the expression of LIFR-AS1 in gastric cancer tumor tissues. Knocking down of LIFR-AS1 inhibited proliferation, invasion and migration of gastric tumor cells, and induced apoptosis of gastric tumor cells. Bioinformatics analyses and integrated experiments revealed that LIFR-AS1 elevated the expression of COL1A2 through sponging miR-29a-3p, which further resulted in the progression of gastric tumor. CONCLUSION LIFR-AS1 plays an important role as a competing endogenous RNA in gastric tumor pathogenesis and may be a potential target for the diagnosis and treatment of gastric tumor.
Collapse
Affiliation(s)
- Haiyan Pan
- School of Public Health, Guangdong Medical University, Dongguan, 523808, Guangdong, People's Republic of China
| | - Yuanlin Ding
- School of Public Health, Guangdong Medical University, Dongguan, 523808, Guangdong, People's Republic of China
| | - Yugang Jiang
- Department of gastrointestinal Surgery, Shandong Provincial Hospital, Jinan, 250021, Shandong, People's Republic of China
| | - Xingjie Wang
- School of Public Health, Guangdong Medical University, Dongguan, 523808, Guangdong, People's Republic of China
| | - Jiawei Rao
- School of Public Health, Guangdong Medical University, Dongguan, 523808, Guangdong, People's Republic of China
| | - Xingshan Zhang
- School of Public Health, Guangdong Medical University, Dongguan, 523808, Guangdong, People's Republic of China
| | - Haibing Yu
- School of Public Health, Guangdong Medical University, Dongguan, 523808, Guangdong, People's Republic of China
| | - Qinghua Hou
- College of Ocean and Meteorology, Guangdong Ocean University, Zhanjiang, 524088, Guangdong, People's Republic of China
| | - Tao Li
- Department of Chemotherapy, The People's Hospital of Gaozhou, Gaozhou, 525200, Guangdong, People's Republic of China.
| |
Collapse
|
44
|
Volpi S, Cancelli U, Neri M, Corradini R. Multifunctional Delivery Systems for Peptide Nucleic Acids. Pharmaceuticals (Basel) 2020; 14:14. [PMID: 33375595 PMCID: PMC7823687 DOI: 10.3390/ph14010014] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 12/21/2020] [Accepted: 12/23/2020] [Indexed: 02/07/2023] Open
Abstract
The number of applications of peptide nucleic acids (PNAs)-oligonucleotide analogs with a polyamide backbone-is continuously increasing in both in vitro and cellular systems and, parallel to this, delivery systems able to bring PNAs to their targets have been developed. This review is intended to give to the readers an overview on the available carriers for these oligonucleotide mimics, with a particular emphasis on newly developed multi-component- and multifunctional vehicles which boosted PNA research in recent years. The following approaches will be discussed: (a) conjugation with carrier molecules and peptides; (b) liposome formulations; (c) polymer nanoparticles; (d) inorganic porous nanoparticles; (e) carbon based nanocarriers; and (f) self-assembled and supramolecular systems. New therapeutic strategies enabled by the combination of PNA and proper delivery systems are discussed.
Collapse
Affiliation(s)
| | | | | | - Roberto Corradini
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, 43124 Parma, Italy; (S.V.); (U.C.); (M.N.)
| |
Collapse
|
45
|
Kimna C, Lieleg O. Molecular micromanagement: DNA nanotechnology establishes spatio-temporal control for precision medicine. BIOPHYSICS REVIEWS 2020; 1:011305. [PMID: 38505628 PMCID: PMC10903406 DOI: 10.1063/5.0033378] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 12/08/2020] [Indexed: 03/21/2024]
Abstract
Current advances in DNA nanotechnology pinpoint exciting perspectives for the design of customized, patient-specific treatments. This advance is made possible by the exceptionally high precision and specificity that are typical for DNA base pairing on the one hand and our growing ability to harness those features in synthetic, DNA-based constructs on the other hand. Modern medicine may soon benefit from recent developments in this field, especially regarding the targeted delivery of drugs and the rational interference of synthetic DNA strands with cellular oligonucleotides. In this Review, we summarize selected examples from the area of DNA nanotechnology, where the development of precisely controlled, advanced functional mechanisms was achieved. To demonstrate the high versatility of these rationally designed structures, we categorize the dynamic DNA-based materials suggested for precision medicine according to four fundamental tasks: "hold & release," "heal," "detect & measure," as well as "guide & direct." In all the biomedical applications we highlight, DNA strands not only constitute structural building blocks but allow for creating stimuli-responsive objects, serve as an active cargo, or act as molecular control/guidance tools. Moreover, we discuss several issues that need to be considered when DNA-based structures are designed for applications in the field of precision medicine. Even though the majority of DNA-based objects have not been used in clinical settings yet, recent progress regarding the stability, specificity, and control over the dynamic behavior of synthetic DNA structures has advanced greatly. Thus, medical applications of those nanoscopic objects should be feasible in the near future.
Collapse
|
46
|
Kelly IB, Fletcher RB, McBride JR, Weiss SM, Duvall CL. Tuning Composition of Polymer and Porous Silicon Composite Nanoparticles for Early Endosome Escape of Anti-microRNA Peptide Nucleic Acids. ACS APPLIED MATERIALS & INTERFACES 2020; 12:39602-39611. [PMID: 32805967 PMCID: PMC8356247 DOI: 10.1021/acsami.0c05827] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Porous silicon nanoparticles (PSNPs) offer tunable pore structure and easily modified surface chemistry, enabling high loading capacity for drugs with diverse chemicophysical properties. While PSNPs are also cytocompatible and degradable, PSNP integration into composite structures can be a useful approach to enhance carrier colloidal stability, drug-cargo loading stability, and endosome escape. Here, we explored PSNP polymer composites formed by coating of oxidized PSNPs with a series of poly[ethylene glycol-block-(dimethylaminoethyl methacrylate-co-butyl methacrylate)] (PEG-DB) diblock copolymers with varied molar ratios of dimethylaminoethyl methacrylate (D) and butyl methacrylate (B) in the random copolymer block. We screened and developed PSNP composites specifically toward intracellular delivery of microRNA inhibitory peptide nucleic acids (PNA). While a copolymer with 50 mol % B (50B) is optimal for early endosome escape in free polymer form, its pH switch was suppressed when it was formed into 50B polymer-coated PSNP composites (50BCs). We demonstrate that a lower mol % B (30BC) is the ideal PEG-DB composition for PSNP/PEG-DB nanocomposites based on having both the highest endosome disruption potential and miR-122 inhibitory activity. At a 1 mM PNA dose, 30BCs facilitated more potent inhibition of miR-122 in comparison to 40BC (p = 0.0095), 50BC (p < 0.0001), or an anti-miR-122 oligonucleotide delivered with the commercial transfection reagent Fugene 6. Using a live cell galectin 8-based endosome disruption reporter, 30BCs had greater endosomal escape than 40BCs and 50BCs within 2 h after treatment, suggesting that rapid endosome escape correlates with higher intracellular bioactivity. This study provides new insight on the polymer structure-dependent effects on stability, endosome escape, and cargo intracellular bioavailability for endosomolytic polymer-coated PSNPs.
Collapse
Affiliation(s)
- Isom B Kelly
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - R Brock Fletcher
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - James R McBride
- Vanderbilt Institute of Nanoscale Science and Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Sharon M Weiss
- Department of Electrical Engineering and Computer Science, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Craig L Duvall
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| |
Collapse
|
47
|
Kim KH, Park YJ, Jang HJ, Lee SJ, Lee S, Yun BS, Lee SW, Rho MC. Rugosic acid A, derived from Rosa rugosa Thunb., is novel inhibitory agent for NF-κB and IL-6/STAT3 axis in acute lung injury model. Phytother Res 2020; 34:3200-3210. [PMID: 32779813 PMCID: PMC7404680 DOI: 10.1002/ptr.6767] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 05/13/2020] [Accepted: 05/23/2020] [Indexed: 12/20/2022]
Abstract
Rosa rugosa Thunb., is as a medicinal plant known for anti‐diabetic, and anti‐inflammatory activities. However, the specific active compounds responsible for the individual pharmacological effects of in R. rugosa extract (95% EtOH) remain unknown. Here, we hypothesized that terpenoid structure, the most abundant constituents in R. rugosa extract, are responsible for its anti‐inflammatory activity. We investigated the phytochemical substituents (compounds 1–13) and newly purified 11‐methoxy polisin A, and 13‐methoxy bisaborosaol F using NMR and ESI‐MS and to screened their effects on NO production in LPS‐induced macrophages. Rugosic acid A (RA) induced to ameliorate NO production, iNOS, and pro‐inflammatory cytokines associated with the NF‐κB. And, RA suppressed IL‐6 secretion and IL‐6‐mediated STAT3 activation in LPS‐mediated inflammation. In addition, RA was evaluated in LPS‐mediated acute lung injury (ALI) model similar to acute pneumonia. Our results suggested that RA was suppressed to translocate nuclear NF‐κB and IL‐6‐mediated STAT3 activation. Finally, RA led to amelioration of ALI by decreasing myeloperoxidase (MPO) and inhibiting phosphorylation of NF‐κB and STAT3. Our group originally found that R. rugosa extract had new methoxy compounds and RA may be alternative natural agent for acute pneumonia similar to severe acute respiratory syndrome by coronavirus.
Collapse
Affiliation(s)
- Kang-Hoon Kim
- Immunoregulatory Material Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup-si, Jeonbuk, Republic of Korea
| | - Ye-Ji Park
- Immunoregulatory Material Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup-si, Jeonbuk, Republic of Korea.,Division of Biotechnology and Advanced Institute of Environment and Bioscience, College of Environmental and Bioresource Sciences, Jeonbuk National University, Iksan-si, Republic of Korea
| | - Hyun-Jae Jang
- Immunoregulatory Material Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup-si, Jeonbuk, Republic of Korea
| | - Seung-Jae Lee
- Immunoregulatory Material Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup-si, Jeonbuk, Republic of Korea
| | - Soyoung Lee
- Immunoregulatory Material Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup-si, Jeonbuk, Republic of Korea
| | - Bong-Sik Yun
- Division of Biotechnology and Advanced Institute of Environment and Bioscience, College of Environmental and Bioresource Sciences, Jeonbuk National University, Iksan-si, Republic of Korea
| | - Seung Woong Lee
- Immunoregulatory Material Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup-si, Jeonbuk, Republic of Korea
| | - Mun-Chual Rho
- Immunoregulatory Material Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup-si, Jeonbuk, Republic of Korea
| |
Collapse
|
48
|
Kwon Y, Kim M, Kim Y, Jung HS, Jeoung D. Exosomal MicroRNAs as Mediators of Cellular Interactions Between Cancer Cells and Macrophages. Front Immunol 2020; 11:1167. [PMID: 32595638 PMCID: PMC7300210 DOI: 10.3389/fimmu.2020.01167] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 05/12/2020] [Indexed: 02/06/2023] Open
Abstract
Tumor microenvironment consists of cancer cells and various stromal cells such as endothelial cells, cancer-associated fibroblasts (CAFs), myeloid-derived suppressor cells (MDSCs), neutrophils, macrophages, and other innate and adaptive immune cells. Of these innate immune cells, macrophages are an extremely heterogeneous population, and display both pro-inflammatory and anti-inflammatory functions. While M1 macrophages (classically activated macrophages) display anti-tumoral and pro-inflammatory functions, M2 macrophages display pro-tumoral and anti-inflammatory functions. Cellular interactions and molecular factors in the tumor microenvironment affect the polarization of macrophages. We review molecules and immune cells that influence the polarization status of macrophages. Tumor-associated macrophages (TAMs) generally express M2 phenotype, and mediate many processes that include tumor initiation, angiogenesis, and metastasis. A high number of TAMs has been associated with the poor prognosis of cancers. MicroRNAs (miRNAs) have been known to regulate cellular interactions that involve cancer cells and macrophages. Tumor-derived exosomes play critical roles in inducing the M1 or M2-like polarization of macrophages. The roles of exosomal miRNAs from tumor cells in the polarization of macrophages are also discussed and the targets of these miRNAs are presented. We review the effects of exosomal miRNAs from TAMs on cancer cell invasion, growth, and anti-cancer drug resistance. The relevance of exosomal microRNAs (miRNAs) as targets for the development of anti-cancer drugs is discussed. We review recent progress in the development of miRNA therapeutics aimed at elevating or decreasing levels of miRNAs.
Collapse
Affiliation(s)
- Yoojung Kwon
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, Chuncheon, South Korea
| | - Misun Kim
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, Chuncheon, South Korea
| | - Youngmi Kim
- Institute of New Frontier Research, College of Medicine, Hallym University, Chuncheon, South Korea
| | - Hyun Suk Jung
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, Chuncheon, South Korea
| | - Dooil Jeoung
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, Chuncheon, South Korea
| |
Collapse
|
49
|
Xie M, Zhu Y, Xu S, Xu G, Xiong R, Sun X, Liu C. A nanoplatform with tumor-targeted aggregation and drug-specific release characteristics for photodynamic/photothermal combined antitumor therapy under near-infrared laser irradiation. NANOSCALE 2020; 12:11497-11509. [PMID: 32427255 DOI: 10.1039/d0nr00123f] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Due to their high biocompatibility, high spatial resolution, chromatographic capability, and adjustable size and morphology, magnetic nanoparticles have become the most promising nanomaterials for clinical application in noninvasive imaging and drug delivery for the treatment of malignant tumors. Herein, a novel magnetic nanoparticle coated with calcium carbonate was prepared and loaded with near-infrared drugs to be used as a multifunctional theranostic nanoplatform for the diagnosis and treatment of malignant tumors. Then, these drug-loaded nanoparticles were used for combined photodynamic/photothermal therapy by intravenous administration that was simultaneously guided by fluorescence/MR imaging. Due to the targeted induction of the external magnetic field and tumor response degradation of the calcium carbonate layer, the nanoprobe demonstrated excellent tumor targeting and greatly improved drug aggregation at the tumor site. Finally, single wavelength-mediated photothermal/photodynamic therapy was applied to liver cancer model mice, ultimately achieving an exciting antitumor therapeutic effect. This study may promote further exploration of nanoplatforms based on magnetic nanoparticles for clinical application in the treatment of malignant tumors.
Collapse
Affiliation(s)
- Mingran Xie
- Department of Thoracic Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China.
| | - Yongfu Zhu
- Department of Oncology, Anhui University of Tradational Chinese Medicine, Hefei 230000, China
| | - Shibin Xu
- Department of Thoracic Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China.
| | - Guangwen Xu
- Department of Thoracic Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China.
| | - Ran Xiong
- Department of Thoracic Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China.
| | - Xiaohui Sun
- Department of Thoracic Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China.
| | - Changqing Liu
- Department of Thoracic Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China.
| |
Collapse
|
50
|
Liu Z, Deng Y, Li T, Zhu F, Zhou X, He Y. The opposite functions of miR-24 in the osteogenesis and adipogenesis of adipose-derived mesenchymal stem cells are mediated by the HOXB7/β-catenin complex. FASEB J 2020; 34:9034-9050. [PMID: 32413244 DOI: 10.1096/fj.202000006rr] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Revised: 04/07/2020] [Accepted: 04/09/2020] [Indexed: 12/28/2022]
Abstract
Adipose-derived mesenchymal stem cells (ADMSCs) used in combination with nanoparticles or scaffolds represent promising candidates for bone engineering. Compared to bone marrow-derived MSCs (BMMSCs), ADMSCs show a relatively low capacity for osteogenesis. In the current study, miR-24 was identified as an osteogenesis- and adipogenesis-related miRNA that performs opposing roles (inhibition in osteogenesis and promotion in adipogenesis) during these two differentiation processes. Through bioinformatics analysis and luciferase reporter assays, homeobox protein Hox-B7 (HOXB7) was identified as a potential novel downstream target of miR-24 that contains a miR-24 binding site in the 3'-UTR of its mRNA. Overexpression of HOXB7 could partly halt the inhibitory effect of miR-24 on osteogenesis, and downregulation of HOXB7 could also partly suppress the positive effect of miR-24 on adipogenesis. Furthermore, immunoprecipitation experiments found that HOXB7 and β-catenin formed a functional complex that acted as an essential modulator during osteogenesis and adipogenesis of ADMSCs. After transfecting ADMSCs with an MSNs-PEI-miR-24 agomir or antagomir and loading the cells onto gelatin-chitosan scaffolds, the compounds were assessed for their abilities to repair the critical-sized calvarial defects in rats. Comprehensive evaluation, including micro-CT, sequential fluorescent labeling, and immunohistochemistry analysis, revealed that silencing miR-24 distinctly promoted in vivo bone remolding, whereas overexpression of miR-24 significantly repressed bone formation. Taken together, our findings demonstrated opposite roles for the miR-24/HOXB7/β-catenin signaling pathway in the osteogenesis and adipogenesis of ADMSCs, which may provide a novel mechanism for determining the balance between these two biological processes.
Collapse
Affiliation(s)
- Zhonglong Liu
- Department of Oral Maxillofacial & Head and Neck Oncology, Shanghai Ninth People's Hospital Affiliated to, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center of Stomatology, Shanghai, China
| | - Yiwen Deng
- Department of Oral Mucosal Diseases, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, National Clinical Research Center of Stomatology, Shanghai, China
| | - Tao Li
- Department of Orthopedics, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fengshuo Zhu
- Department of Oral Maxillofacial & Head and Neck Oncology, Shanghai Ninth People's Hospital Affiliated to, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center of Stomatology, Shanghai, China
| | - Xiaojun Zhou
- Department of Orthopedics, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yue He
- Department of Oral Maxillofacial & Head and Neck Oncology, Shanghai Ninth People's Hospital Affiliated to, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center of Stomatology, Shanghai, China
| |
Collapse
|