1
|
Karimzadeh F, Soltani Fard E, Nadi A, Malekzadeh R, Elahian F, Mirzaei SA. Advances in skin gene therapy: utilizing innovative dressing scaffolds for wound healing, a comprehensive review. J Mater Chem B 2024; 12:6033-6062. [PMID: 38887828 DOI: 10.1039/d4tb00966e] [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: 06/20/2024]
Abstract
The skin, serving as the body's outermost layer, boasts a vast area and intricate structure, functioning as the primary barrier against external threats. Disruptions in the composition and functionality of the skin can lead to a diverse array of skin conditions, such as wounds, burns, and diabetic ulcers, along with inflammatory disorders, infections, and various types of skin cancer. These disorders not only exacerbate concerns regarding skin health and beauty but also have a significant impact on mental well-being. Due to the complexity of these disorders, conventional treatments often prove insufficient, necessitating the exploration of new therapeutic approaches. Researchers develop new therapies by deciphering these intricacies and gaining a thorough understanding of the protein networks and molecular processes in skin. A new window of opportunity has opened up for improving wound healing processes because of recent advancements in skin gene therapy. To enhance skin regeneration and healing, this extensive review investigates the use of novel dressing scaffolds in conjunction with gene therapy approaches. Scaffolds that do double duty as wound protectors and vectors for therapeutic gene delivery are being developed using innovative biomaterials. To improve cellular responses and speed healing, these state-of-the-art scaffolds allow for the targeted delivery and sustained release of genetic material. The most recent developments in gene therapy techniques include RNA interference, CRISPR-based gene editing, and the utilization of viral and non-viral vectors in conjunction with scaffolds, which were reviewed here to overcome skin disorders and wound complications. In the future, there will be rare chances to develop custom methods for skin health care thanks to the combination of modern technology and collaboration among disciplines.
Collapse
Affiliation(s)
- Fatemeh Karimzadeh
- Student Research Committee, Shahrekord University of Medical Sciences, Shahrekord, Iran
- Department of Medical Biotechnology, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran.
| | - Elahe Soltani Fard
- Student Research Committee, Shahrekord University of Medical Sciences, Shahrekord, Iran
- Department of Molecular Medicine, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Akram Nadi
- Stem Cell Biology Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Rahim Malekzadeh
- Student Research Committee, Shahrekord University of Medical Sciences, Shahrekord, Iran
- Department of Medical Biotechnology, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran.
| | - Fatemeh Elahian
- Advanced Technology Cores, Baylor College of Medicine, Houston, Texas, USA
| | - Seyed Abbas Mirzaei
- Department of Medical Biotechnology, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran.
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| |
Collapse
|
2
|
Sun B, Wang L, Guo W, Chen S, Ma Y, Wang D. New treatment methods for myocardial infarction. Front Cardiovasc Med 2023; 10:1251669. [PMID: 37840964 PMCID: PMC10569499 DOI: 10.3389/fcvm.2023.1251669] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Accepted: 08/31/2023] [Indexed: 10/17/2023] Open
Abstract
For a long time, cardiovascular clinicians have focused their research on coronary atherosclerotic cardiovascular disease and acute myocardial infarction due to their high morbidity, high mortality, high disability rate, and limited treatment options. Despite the continuous optimization of the therapeutic methods and pharmacological therapies for myocardial ischemia-reperfusion, the incidence rate of heart failure continues to increase year by year. This situation is speculated to be caused by the current therapies, such as reperfusion therapy after ischemic injury, drugs, rehabilitation, and other traditional treatments, that do not directly target the infarcted myocardium. Consequently, these therapies cannot fundamentally solve the problems of myocardial pathological remodeling and the reduction of cardiac function after myocardial infarction, allowing for the progression of heart failure after myocardial infarction. Coupled with the decline in mortality caused by acute myocardial infarction in recent years, this combination leads to an increase in the incidence of heart failure. As a new promising therapy rising at the beginning of the twenty-first century, cardiac regenerative medicine provides a new choice and hope for the recovery of cardiac function and the prevention and treatment of heart failure after myocardial infarction. In the past two decades, regeneration engineering researchers have explored and summarized the elements, such as cells, scaffolds, and cytokines, required for myocardial regeneration from all aspects and various levels day and night, paving the way for our later scholars to carry out relevant research and also putting forward the current problems and directions for us. Here, we describe the advantages and challenges of cardiac tissue engineering, a contemporary innovative therapy after myocardial infarction, to provide a reference for clinical treatment.
Collapse
Affiliation(s)
- Bingbing Sun
- Department of Critical Care Medicine, The Air Force Characteristic Medical Center, Air Force Medical University, Beijing, China
| | - Long Wang
- Department of General Internal Medicine, Beijing Dawanglu Emergency Hospital, Beijing, China
| | - Wenmin Guo
- Department of Critical Care Medicine, The Air Force Characteristic Medical Center, Air Force Medical University, Beijing, China
| | - Shixuan Chen
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, China
| | - Yujie Ma
- Department of Critical Care Medicine, The Air Force Characteristic Medical Center, Air Force Medical University, Beijing, China
| | - Dongwei Wang
- Department of Cardiac Rehabilitation, Zhengzhou Central Hospital affiliated to Zhengzhou University, Zhengzhou, China
| |
Collapse
|
3
|
Sabahi M, Salehipour A, Bazl MSY, Rezaei N, Mansouri A, Borghei-Razavi H. Local immunotherapy of glioblastoma: A comprehensive review of the concept. J Neuroimmunol 2023; 381:578146. [PMID: 37451079 DOI: 10.1016/j.jneuroim.2023.578146] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 05/24/2023] [Accepted: 07/04/2023] [Indexed: 07/18/2023]
Abstract
Despite advancements in standard treatments, the prognosis of Glioblastoma (GBM) remains poor, prompting research for novel therapies. Immunotherapy is a promising treatment option for GBM, and many immunotherapeutic agents are currently under investigation. Chimeric antigen receptor (CAR) T cells are rapidly evolving in immunotherapy of GBM with many clinical trials showing efficacy of CAR T cells exerting anti-tumor activity following recognition of tumor-associated antigens (TAAs). Exhaustion in CAR T cells can reduce their capacity for long-term persistence and anti-tumor action. Local immunotherapy, which targets the tumor microenvironment and creates a more hospitable immunological environment for CAR T cells, has the potential to reduce CAR T cell exhaustion and increase immunity. Tertiary lymphoid structures (TLS) are ectopic lymphoid-like formations that can develop within the tumor microenvironment or in other non-lymphoid tissues. As a comprehensive local immunotherapy tool, the incorporation of TLS into an implanted biodegradable scaffold has amazing immunotherapeutic potential. The immune response to GBM can be improved even further by strategically inserting a stimulator of interferon genes (STING) agonist into the scaffold. Additionally, the scaffold's addition of glioma stem cells (GSC), which immunotherapeutic approaches may use to target, enhances the removal of cancer cells from their source. Furthermore, it has been demonstrated that GSCs have an impact on TLS formation, which helps to create a favorable tumor microenvironment. Herein, we overview local delivery of a highly specific tandem AND-gate CAR T cell along with above mentioned components. A multifaceted approach that successfully engages the immune system to mount an efficient targeted immune response against GBM is provided by the integration of CAR T cells, TLS, STING agonists, and GSCs within an implantable biodegradable scaffold. This approach offers a promising therapeutic approach for patients with GBM.
Collapse
Affiliation(s)
- Mohammadmahdi Sabahi
- Department of Neurological Surgery, Pauline Braathen Neurological Center, Cleveland Clinic Florida, Weston, FL, USA.
| | - Arash Salehipour
- Neurosurgery Research Group (NRG), Student Research Committee, Hamadan University of Medical Sciences, Hamadan, Iran; Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran.
| | - Mohammad Sajjad Yavari Bazl
- Neurosurgery Research Group (NRG), Student Research Committee, Hamadan University of Medical Sciences, Hamadan, Iran; Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran.
| | - Nima Rezaei
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran; Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Alireza Mansouri
- Department of Neurosurgery, Penn State Milton S. Hershey Medical Center, Hershey, PA, USA.
| | - Hamid Borghei-Razavi
- Department of Neurological Surgery, Pauline Braathen Neurological Center, Cleveland Clinic Florida, Weston, FL, USA.
| |
Collapse
|
4
|
Shi CS, Hu Q, Fang SL, Sun CX, Shao DH. MicroRNA-204-5p Ameliorates Neurological Injury via the EphA4/PI3K/AKT Signaling Pathway in Ischemic Stroke. ACS Chem Neurosci 2023. [PMID: 37196241 DOI: 10.1021/acschemneuro.3c00047] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/19/2023] Open
Abstract
Ischemic stroke has extremely high mortality and disability rates worldwide. miR-204-5p has been reported to be associated with neurological diseases. However, the relationship linking miR-204-5p to ischemic stroke and its molecular mechanism remain unclear. Herein, we found that expression of miR-204-5p was significantly decreased while EphA4 increased in vivo and vitro, which reached the peak at 24 h after cerebral ischemia/reperfusion. Then, we altered miR-204-5p expression in rats by cerebroventricular injection. Our study showed that miR-204-5p overexpression obviously reduced the brain infarction area and neurological score. We successfully cultured neurons to investigate the downstream mechanism. Upregulation of miR-204-5p increased cell viability and suppressed the release of LDH. Moreover, the proportion of apoptotic cells tested by TUNEL and flow cytometry and protein expression of Cleaved Caspase3 and Bax were inhibited. The relative expression of IL-6, TNF-α, and IL-1β was repressed. In contrary, knockdown of miR-204-5p showed the opposite results. Bioinformatics and a dual luciferase assay illustrated that EphA4 was a target gene. Further research studies demonstrated that the neuroprotective effects of miR-204-5p could be partially mitigated by upregulating EphA4. Next, we proved that the miR-204-5p/EphA4 axis furtherly activated the PI3K/AKT pathway. We thoroughly illustrated the role of neuroinflammation and apoptosis. However, whether there are other mechanisms associated with the EphA4/PI3K/AKT pathway needs further investigation. Altogether, the miR-204-5p axis ameliorates neurological injury via the EphA4/PI3K/AKT pathway, which is expected to serve as an effective treatment for ischemic stroke.
Collapse
Affiliation(s)
- Chang-Sheng Shi
- Department of Anesthesiology, The Affiliated People's Hospital of Jiangsu University, No.8 Dian Li Road, Zhenjiang, Jiangsu 212000, China
- Department of Medicine, The University of Jiangsu, No.301 Xue Fu Road, Zhenjiang, Jiangsu 212000, China
| | - Qi Hu
- Department of Anesthesiology, The Affiliated People's Hospital of Jiangsu University, No.8 Dian Li Road, Zhenjiang, Jiangsu 212000, China
| | - Shi-Lei Fang
- Department of Anesthesiology, The Affiliated People's Hospital of Jiangsu University, No.8 Dian Li Road, Zhenjiang, Jiangsu 212000, China
| | - Cai-Xia Sun
- Department of Anesthesiology, The Affiliated People's Hospital of Jiangsu University, No.8 Dian Li Road, Zhenjiang, Jiangsu 212000, China
| | - Dong-Hua Shao
- Department of Anesthesiology, The Affiliated People's Hospital of Jiangsu University, No.8 Dian Li Road, Zhenjiang, Jiangsu 212000, China
| |
Collapse
|
5
|
Yang M, Zhang Y, Li M, Liu X, Darvishi M. The various role of microRNAs in breast cancer angiogenesis, with a special focus on novel miRNA-based delivery strategies. Cancer Cell Int 2023; 23:24. [PMID: 36765409 PMCID: PMC9912632 DOI: 10.1186/s12935-022-02837-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 12/20/2022] [Indexed: 02/12/2023] Open
Abstract
After skin malignancy, breast cancer is the most widely recognized cancer detected in women in the United States. Breast cancer (BCa) can happen in all kinds of people, but it's much more common in women. One in four cases of cancer and one in six deaths due to cancer are related to breast cancer. Angiogenesis is an essential factor in the growth of tumors and metastases in various malignancies. An expanded level of angiogenesis is related to diminished endurance in BCa patients. This function assumes a fundamental part inside the human body, from the beginning phases of life to dangerous malignancy. Various factors, referred to as angiogenic factors, work to make a new capillary. Expanding proof demonstrates that angiogenesis is managed by microRNAs (miRNAs), which are small non-coding RNA with 19-25 nucleotides. MiRNA is a post-transcriptional regulator of gene expression that controls many critical biological processes. Endothelial miRNAs, referred to as angiomiRs, are probably concerned with tumor improvement and angiogenesis via regulation of pro-and anti-angiogenic factors. In this article, we reviewed therapeutic functions of miRNAs in BCa angiogenesis, several novel delivery carriers for miRNA-based therapeutics, as well as CRISPR/Cas9 as a targeted therapy in breast cancer.
Collapse
Affiliation(s)
- Min Yang
- College of Traditional Chinese Medicine, Jilin Agricultural Science and Technology University, Jilin, 132101 China
| | - Ying Zhang
- College of Traditional Chinese Medicine, Jilin Agricultural Science and Technology University, Jilin, 132101 China
| | - Min Li
- College of Traditional Chinese Medicine, Jilin Agricultural Science and Technology University, Jilin, 132101 China
| | - Xinglong Liu
- College of Traditional Chinese Medicine, Jilin Agricultural Science and Technology University, Jilin, 132101 China
| | - Mohammad Darvishi
- Infectious Diseases and Tropical Medicine Research Center (IDTMRC), Department of Aerospace and Subaquatic Medicine, AJA University of Medical Sciences, Tehran, Iran
| |
Collapse
|
6
|
Sainsbury E, Costard L, O'Brien FJ, Curtin CM. Assessment of Cell Cytotoxicity in 3D Biomaterial Scaffolds Following miRNA Transfection. Methods Mol Biol 2023; 2595:203-210. [PMID: 36441464 DOI: 10.1007/978-1-0716-2823-2_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Assessment of cell cytotoxicity following transfection of cells with microRNA (miRNA) is an essential step in the evaluation of basic miRNA functional effects within cells in both 2D and 3D microenvironments. The lactate dehydrogenase (LDH) assay is a colorimetric assay that provides a basic, dependable method for determining cellular cytotoxicity through assessment of the level of plasma membrane damage in a cell population. Here, we describe the overexpression of miRNA in breast cancer cells when cultured in 3D collagen-based biomaterial scaffolds, achieved by Lipofectamine transfection, with subsequent examination of cell cytotoxicity using the LDH assay.
Collapse
Affiliation(s)
- Elizabeth Sainsbury
- Department of Anatomy & Regenerative Medicine, Tissue Engineering Research Group (TERG), Royal College of Surgeons in Ireland (RCSI), Dublin, Ireland
| | - Lara Costard
- Department of Anatomy & Regenerative Medicine, Tissue Engineering Research Group (TERG), Royal College of Surgeons in Ireland (RCSI), Dublin, Ireland
| | - Fergal J O'Brien
- Department of Anatomy & Regenerative Medicine, Tissue Engineering Research Group (TERG), Royal College of Surgeons in Ireland (RCSI), Dublin, Ireland
- Trinity Centre for Biomedical Engineering (TCBE), Trinity College Dublin (TCD), Dublin, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, Ireland
| | - Caroline M Curtin
- Department of Anatomy & Regenerative Medicine, Tissue Engineering Research Group (TERG), Royal College of Surgeons in Ireland (RCSI), Dublin, Ireland.
- Trinity Centre for Biomedical Engineering (TCBE), Trinity College Dublin (TCD), Dublin, Ireland.
- Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, Ireland.
| |
Collapse
|
7
|
Sharma AR, Lee YH, Lee SS. Recent advancements of miRNAs in the treatment of bone diseases and their delivery potential. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2022; 4:100150. [PMID: 36691422 PMCID: PMC9860349 DOI: 10.1016/j.crphar.2022.100150] [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: 08/23/2022] [Revised: 10/26/2022] [Accepted: 12/21/2022] [Indexed: 12/28/2022] Open
Abstract
Advances in understanding miRNAs as endogenous posttranscriptional regulatory units have projected them as novel therapeutics for several untreatable diseases. miRNAs are endogenous non-coding small single-stranded RNA molecules (20-24 nucleotides) with specific gene regulatory functions like repression of mRNA translation by degrading mRNAs. Emerging evidence suggests the role of miRNAs in various stages of bone growth and development. Undoubtedly, due to their critical role in bone remodeling, miRNAs might be projected as a novel approach to treating bone-related diseases. However, the instability associated with miRNAs in their complex environment, such as degradation by nucleases, is a concern. Thus, recent attention is being paid to maintaining the miRNAs' safety and efficacy in the cells. Various efficient delivery systems and chemical modifications of miRNAs are being developed to make them a potential therapeutic option for bone diseases. Here, we have tried to recapitulate the recent advances in the role of miRNAs in bone disease, along with the potential delivery systems for their efficient delivery to the cells.
Collapse
Affiliation(s)
- Ashish Ranjan Sharma
- Corresponding author. Institute for Skeletal Aging & Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon-si, Gangwon-do, 24252, South Korea.
| | | | - Sang-Soo Lee
- Corresponding author. Institute for Skeletal Aging & Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon-si, Gangwon-do, 24252, South Korea.
| |
Collapse
|
8
|
Fu P, Chen H, Ouyang L, Li L, Wang Y, Qian S, Cao Z, Wu K, Chao J, Zheng J. DNA Nanoribbon for Efficient Anti-miRNA Peptide Nucleic Acid Delivery and Synergistic Enhancement of Cancer Cell Apoptosis. Anal Chem 2022; 95:1811-1816. [PMID: 36542541 DOI: 10.1021/acs.analchem.2c04760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Antisense peptide nucleic acid (asPNA), an effective antisense drug, has been employed as a gene therapy agent and a useful tool in molecular biology. Gaining control over the delivery of asPNA to target tissues has been a major hindrance to its wide application in clinical practice. A simple and efficient DNA nanoribbon (DNR)-based drug delivery process has been designed in this study that releases the asPNA agent to inhibit oncogenic microRNAs (miRNAs). Furthermore, we demonstrated how the AS1411 aptamer that binds nucleolin on the cell membranes works as a control mechanism capable of identifying target cancer cells and enhancing the enrichment capacity of DNR. With the biodegradability of DNR, we can efficiently initiate the release of asPNA into the cytoplasm, particularly targeting the intended miR-21 and synergistically increasing programmed cell death 4 (PDCD4) expression to enhance cell apoptosis. We assume that this well-defined delivery mechanism will aid in designing antisense site-specific treatments for various diseases, including cancer.
Collapse
Affiliation(s)
- Pan Fu
- Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, P. R. China
| | - Hao Chen
- Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, P. R. China
| | - Lilin Ouyang
- State Key Laboratory of Organic Electronics and Information Displays and Jiangsu Key Laboratory for Biosensors, Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Lin Li
- Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, P. R. China
| | - Yuhui Wang
- Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, P. R. China
| | - Sihua Qian
- Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, P. R. China
| | - Zhanglei Cao
- Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, P. R. China
| | - Kerong Wu
- Ningbo First Hospital, Ningbo, Zhejiang 315000, P. R. China
| | - Jie Chao
- State Key Laboratory of Organic Electronics and Information Displays and Jiangsu Key Laboratory for Biosensors, Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Jianping Zheng
- Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, P. R. China
| |
Collapse
|
9
|
Xu P, Chang J, Ma G, Liao F, Xu T, Wu Y, Yin Z. MiR-145 inhibits the differentiation and proliferation of bone marrow stromal mesenchymal stem cells by GABARAPL1 in steroid-induced femoral head necrosis. BMC Musculoskelet Disord 2022; 23:1020. [PMID: 36435763 PMCID: PMC9701430 DOI: 10.1186/s12891-022-05928-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 10/25/2022] [Accepted: 10/29/2022] [Indexed: 11/28/2022] Open
Abstract
Steroid-induced osteonecrosis of femoral head (SANFH) involves impaired differentiation of bone marrow mesenchymal stem cells (BMSC), the mechanism of which is regulated by multiple microRNAs. Studies have shown that miR-145 is a key regulatory molecule of BMSC cells, but its mechanism in steroid-induced femur head necrosis remains unclear. The present study mainly explored the specific mechanism of miR-145 involved in SANFH. In this study dexamethasone, a typical glucocorticoid, was used to induce osteogenic differentiation of BMSC cells. Western blot, qPCR, CCK8 and flow cytometry were used to investigate the effects of miR-145 on the proliferation and differentiation of BMSC. The relationship between miR-145 and GABA Type A Receptor Associated Protein Like 1(GABARAPL1) was identified using dual luciferase reports and the effects of the two molecules on BMSC were investigated in vitro. The results showed that miR-145 was up-regulated in SANFH patients, while GABARAPL1 was down-regulated. Inhibition of miR-145 can improve apoptosis and promote proliferation and activation of BMSC. GABARAPL1 is a downstream target gene of miR-145 and is negatively regulated by miR-145. In conclusion, miR-145 regulates the proliferation and differentiation of glucocorticoid-induced BMSC cells through GABARAPL1 and pharmacologically inhibit targeting miR-145 may provide new aspect for the treatment of SANFH.
Collapse
|
10
|
Amini M, Venkatesan JK, Liu W, Leroux A, Nguyen TN, Madry H, Migonney V, Cucchiarini M. Advanced Gene Therapy Strategies for the Repair of ACL Injuries. Int J Mol Sci 2022; 23:ijms232214467. [PMID: 36430947 PMCID: PMC9695211 DOI: 10.3390/ijms232214467] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/07/2022] [Accepted: 11/19/2022] [Indexed: 11/23/2022] Open
Abstract
The anterior cruciate ligament (ACL), the principal ligament for stabilization of the knee, is highly predisposed to injury in the human population. As a result of its poor intrinsic healing capacities, surgical intervention is generally necessary to repair ACL lesions, yet the outcomes are never fully satisfactory in terms of long-lasting, complete, and safe repair. Gene therapy, based on the transfer of therapeutic genetic sequences via a gene vector, is a potent tool to durably and adeptly enhance the processes of ACL repair and has been reported for its workability in various experimental models relevant to ACL injuries in vitro, in situ, and in vivo. As critical hurdles to the effective and safe translation of gene therapy for clinical applications still remain, including physiological barriers and host immune responses, biomaterial-guided gene therapy inspired by drug delivery systems has been further developed to protect and improve the classical procedures of gene transfer in the future treatment of ACL injuries in patients, as critically presented here.
Collapse
Affiliation(s)
- Mahnaz Amini
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, D-66421 Homburg, Germany
| | - Jagadeesh K. Venkatesan
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, D-66421 Homburg, Germany
| | - Wei Liu
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, D-66421 Homburg, Germany
| | - Amélie Leroux
- Laboratoire CSPBAT UMR CNRS 7244, Université Sorbonne Paris Nord, Avenue JB Clément, 93430 Villetaneuse, France
| | - Tuan Ngoc Nguyen
- Laboratoire CSPBAT UMR CNRS 7244, Université Sorbonne Paris Nord, Avenue JB Clément, 93430 Villetaneuse, France
| | - Henning Madry
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, D-66421 Homburg, Germany
| | - Véronique Migonney
- Laboratoire CSPBAT UMR CNRS 7244, Université Sorbonne Paris Nord, Avenue JB Clément, 93430 Villetaneuse, France
| | - Magali Cucchiarini
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, D-66421 Homburg, Germany
- Correspondence: or
| |
Collapse
|
11
|
Trideva Sastri K, Vishal Gupta N, Kannan A, Balamuralidhara V, Ramkishan A. Potential nanocarrier-mediated miRNA-based therapy approaches for multiple sclerosis. Drug Discov Today 2022; 27:103357. [PMID: 36115632 DOI: 10.1016/j.drudis.2022.103357] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 08/16/2022] [Accepted: 09/09/2022] [Indexed: 11/22/2022]
Abstract
Multiple sclerosis (MS) is an autoimmune neuroinflammatory disorder attributed to neurodegeneration and demyelination, resulting in neurological impairment. miRNA has a significant role in biological processes in MS. In this review, we focus on the feasibility of delivering miRNAs through nanoformulations for managing MS. We provide a brief discussion of miRNA synthesis and evidence for miRNA dysregulation in MS. We also highlight formulation strategies and resulting technologies for the effective delivery of miRNAs through nanocarrier systems for achieving high therapeutic benefits.
Collapse
Affiliation(s)
- K Trideva Sastri
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Shivarathreeshwara Nagara, Bannimantap, Mysuru, India
| | - N Vishal Gupta
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Shivarathreeshwara Nagara, Bannimantap, Mysuru, India.
| | - Anbarasu Kannan
- Department of Biochemistry, CSIR-Central Food Technological Research Institute, Mysuru, India
| | - V Balamuralidhara
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Shivarathreeshwara Nagara, Bannimantap, Mysuru, India
| | - A Ramkishan
- Deputy Drugs Controller (India), Central Drugs Standard Control Organization, Directorate General of Health Services, Ministry of Health & Family Welfare, Government of India, India
| |
Collapse
|
12
|
MicroRNA and their implications in dental pulp inflammation: current trends and future perspectives. Odontology 2022:10.1007/s10266-022-00762-0. [DOI: 10.1007/s10266-022-00762-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 10/18/2022] [Indexed: 11/25/2022]
|
13
|
Hosseinpour S, Gomez-Cerezo MN, Cao Y, Lei C, Dai H, Walsh LJ, Ivanovski S, Xu C. A Comparative Study of Mesoporous Silica and Mesoporous Bioactive Glass Nanoparticles as Non-Viral MicroRNA Vectors for Osteogenesis. Pharmaceutics 2022; 14:pharmaceutics14112302. [PMID: 36365121 PMCID: PMC9694756 DOI: 10.3390/pharmaceutics14112302] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/13/2022] [Accepted: 10/20/2022] [Indexed: 11/16/2022] Open
Abstract
Micro-ribonucleic acid (miRNA)-based therapies show advantages for bone regeneration but need efficient intracellular delivery methods. Inorganic nanoparticles such as mesoporous bioactive glass nanoparticles (MBGN) and mesoporous silica nanoparticles (MSN) have received growing interest in the intracellular delivery of nucleic acids. This study explores the capacity of MBGN and MSN for delivering miRNA to bone marrow mesenchymal stem cells (BMSC) for bone regenerative purposes, with a focus on comparing the two in terms of cell viability, transfection efficiency, and osteogenic actions. Spherical MBGN and MSN with a particle size of ~200 nm and small-sized mesopores were prepared using the sol-gel method, and then the surface was modified with polyethyleneimine for miRNA loading and delivery. The results showed miRNA can be loaded into both nanoparticles within 2 h and was released sustainedly for up to 3 days. Confocal laser scanning microscopy and flow cytometry analysis indicated a high transfection efficiency (>64%) of both nanoparticles without statistical difference. Compared with MSN, MBGN showed stronger activation of alkaline phosphatase and activation of osteocalcin genes. This translated to a greater osteogenic effect of MBGN on BMSC, with Alizarin red staining showing greater mineralization compared with the MSN group. These findings show the potential for MBGN to be used in bone tissue engineering.
Collapse
Affiliation(s)
- Sepanta Hosseinpour
- School of Dentistry, The University of Queensland, Herston, QLD 4006, Australia
| | - Maria Natividad Gomez-Cerezo
- School of Dentistry, The University of Queensland, Herston, QLD 4006, Australia
- Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre i+12, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain
| | - Yuxue Cao
- The Pharmacy Australia Centre of Excellence (PACE), The University of Queensland, Woolloongabba, QLD 4102, Australia
| | - Chang Lei
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Huan Dai
- School of Dentistry, The University of Queensland, Herston, QLD 4006, Australia
| | - Laurence J. Walsh
- School of Dentistry, The University of Queensland, Herston, QLD 4006, Australia
| | - Saso Ivanovski
- School of Dentistry, The University of Queensland, Herston, QLD 4006, Australia
| | - Chun Xu
- School of Dentistry, The University of Queensland, Herston, QLD 4006, Australia
- Correspondence:
| |
Collapse
|
14
|
Pouya FD, Rasmi Y, Gazouli M, Zografos E, Nemati M. MicroRNAs as therapeutic targets in breast cancer metastasis. Drug Deliv Transl Res 2022; 12:1029-1046. [PMID: 33987801 DOI: 10.1007/s13346-021-00999-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/29/2021] [Indexed: 12/24/2022]
Abstract
Breast cancer is a complex disease with multiple risk factors involved in its pathogenesis. Among these factors, microRNAs are considered for playing a fundamental role in the development and progression of malignant breast tumors. In recent years, various studies have demonstrated that several microRNAs exhibit increased or decreased expression in metastatic breast cancer, acting as indicators of metastatic potential in body fluids and tissue samples. The identification of these microRNA expression patterns could prove instrumental for the development of novel therapeutic molecules that either mimic or inhibit microRNA action. Additionally, an efficient delivery system mediated by viral vectors, nonviral carriers, or scaffold biomaterials is a prerequisite for implementing microRNA-based therapies; therefore, this review attempts to highlight essential microRNA molecules involved in the metastatic process of breast cancer and discuss recent advances in microRNA-based therapeutic approaches with potential future applications to the treatment sequence of breast cancer.
Collapse
Affiliation(s)
- Fahima Danesh Pouya
- Department of Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Yousef Rasmi
- Department of Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran.
- Cellular and Molecular Research Center, Urmia University of Medical Sciences, Urmia, Iran.
| | - Maria Gazouli
- Laboratory of Biology, Medical School, National and Kapodistrian University of Athens, 11527, Athens, Greece
| | - Eleni Zografos
- Laboratory of Biology, Medical School, National and Kapodistrian University of Athens, 11527, Athens, Greece
| | - Mohadeseh Nemati
- Department of Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
15
|
Kharaghani D, Kurniwan EB, Khan MQ, Yoshiko Y. MiRNA-Nanofiber, the Next Generation of Bioactive Scaffolds for Bone Regeneration: A Review. MICROMACHINES 2021; 12:mi12121472. [PMID: 34945325 PMCID: PMC8707075 DOI: 10.3390/mi12121472] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 11/23/2021] [Accepted: 11/23/2021] [Indexed: 12/19/2022]
Abstract
Scaffold-based bone tissue engineering has been introduced as an alternative treatment option for bone grafting due to limitations in the allograft. Not only physical conditions but also biological conditions such as gene expression significantly impact bone regeneration. Scaffolds in composition with bioactive molecules such as miRNA mimics provide a platform to enhance migration, proliferation, and differentiation of osteoprogenitor cells for bone regeneration. Among scaffolds, fibrous structures showed significant advantages in promoting osteogenic differentiation and bone regeneration via delivering bioactive molecules over the past decade. Here, we reviewed the bone and bone fracture healing considerations for the impact of miRNAs on bone regeneration. We also examined the methods used to improve miRNA mimics uptake by cells, the fabrication of fibrous scaffolds, and the effective delivery of miRNA mimics using fibrous scaffold and their processes for bone development. Finally, we offer our view on the principal challenges of miRNA mimics delivery by nanofibers for bone tissue engineering.
Collapse
Affiliation(s)
- Davood Kharaghani
- Department of Calcified Tissue Biology, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan;
- Correspondence: ; Tel.: +81-82-257-5621
| | - Eben Bashir Kurniwan
- School of Dentistry, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan;
| | - Muhammad Qamar Khan
- Nanotechnology Research Lab, Department of Textile and Clothing, National Textile University, Karachi Campus, Karachi 74900, Pakistan;
| | - Yuji Yoshiko
- Department of Calcified Tissue Biology, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan;
| |
Collapse
|
16
|
Yang M, Weng T, Zhang W, Zhang M, He X, Han C, Wang X. The Roles of Non-coding RNA in the Development and Regeneration of Hair Follicles: Current Status and Further Perspectives. Front Cell Dev Biol 2021; 9:720879. [PMID: 34708037 PMCID: PMC8542792 DOI: 10.3389/fcell.2021.720879] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Accepted: 09/23/2021] [Indexed: 12/12/2022] Open
Abstract
Alopecia is a common problem that affects almost every age group and is considered to be an issue for cosmetic or psychiatric reasons. The loss of hair follicles (HFs) and hair caused by alopecia impairs self-esteem, thermoregulation, tactile sensation and protection from ultraviolet light. One strategy to solve this problem is HF regeneration. Many signalling pathways and molecules participate in the morphology and regeneration of HF, such as Wnt/β-catenin, Sonic hedgehog, bone morphogenetic protein and Notch. Non-coding RNAs (ncRNAs), especially microRNAs and long ncRNAs, have significant modulatory roles in HF development and regeneration via regulation of these signalling pathways. This review provides a comprehensive overview of the status and future prospects of ncRNAs in HF regeneration and could prompt novel ncRNA-based therapeutic strategies.
Collapse
Affiliation(s)
- Min Yang
- Department of Burns & Wound Care Center, Second Affiliated Hospital of Zhejiang University, Hangzhou, China.,Key Laboratory of the Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, Hangzhou, China
| | - Tingting Weng
- Department of Burns & Wound Care Center, Second Affiliated Hospital of Zhejiang University, Hangzhou, China.,Key Laboratory of the Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, Hangzhou, China
| | - Wei Zhang
- Department of Burns & Wound Care Center, Second Affiliated Hospital of Zhejiang University, Hangzhou, China.,Key Laboratory of the Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, Hangzhou, China
| | - Manjia Zhang
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiaojie He
- Department of General Practice, Second Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Chunmao Han
- Department of Burns & Wound Care Center, Second Affiliated Hospital of Zhejiang University, Hangzhou, China.,Key Laboratory of the Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, Hangzhou, China
| | - Xingang Wang
- Department of Burns & Wound Care Center, Second Affiliated Hospital of Zhejiang University, Hangzhou, China.,Key Laboratory of the Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, Hangzhou, China
| |
Collapse
|
17
|
Laird NZ, Acri TM, Tingle K, Salem AK. Gene- and RNAi-activated scaffolds for bone tissue engineering: Current progress and future directions. Adv Drug Deliv Rev 2021; 174:613-627. [PMID: 34015421 PMCID: PMC8217358 DOI: 10.1016/j.addr.2021.05.009] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 04/30/2021] [Accepted: 05/11/2021] [Indexed: 01/02/2023]
Abstract
Large bone defects are usually managed by replacing lost bone with non-biological prostheses or with bone grafts that come from the patient or a donor. Bone tissue engineering, as a field, offers the potential to regenerate bone within these large defects without the need for grafts or prosthetics. Such therapies could provide improved long- and short-term outcomes in patients with critical-sized bone defects. Bone tissue engineering has long relied on the administration of growth factors in protein form to stimulate bone regeneration, though clinical applications have shown that using such proteins as therapeutics can lead to concerning off-target effects due to the large amounts required for prolonged therapeutic action. Gene-based therapies offer an alternative to protein-based therapeutics where the genetic material encoding the desired protein is used and thus loading large doses of protein into the scaffolds is avoided. Gene- and RNAi-activated scaffolds are tissue engineering devices loaded with nucleic acids aimed at promoting local tissue repair. A variety of different approaches to formulating gene- and RNAi-activated scaffolds for bone tissue engineering have been explored, and include the activation of scaffolds with plasmid DNA, viruses, RNA transcripts, or interfering RNAs. This review will discuss recent progress in the field of bone tissue engineering, with specific focus on the different approaches employed by researchers to implement gene-activated scaffolds as a means of facilitating bone tissue repair.
Collapse
Affiliation(s)
- Noah Z Laird
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA, USA
| | - Timothy M Acri
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA, USA
| | - Kelsie Tingle
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA, USA
| | - Aliasger K Salem
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
18
|
Iaquinta MR, Lanzillotti C, Mazziotta C, Bononi I, Frontini F, Mazzoni E, Oton-Gonzalez L, Rotondo JC, Torreggiani E, Tognon M, Martini F. The role of microRNAs in the osteogenic and chondrogenic differentiation of mesenchymal stem cells and bone pathologies. Theranostics 2021; 11:6573-6591. [PMID: 33995677 PMCID: PMC8120225 DOI: 10.7150/thno.55664] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 03/15/2021] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stem cells (MSCs) have been identified in many adult tissues. MSCs can regenerate through cell division or differentiate into adipocytes, osteoblasts and chondrocytes. As a result, MSCs have become an important source of cells in tissue engineering and regenerative medicine for bone tissue and cartilage. Several epigenetic factors are believed to play a role in MSCs differentiation. Among these, microRNA (miRNA) regulation is involved in the fine modulation of gene expression during osteogenic/chondrogenic differentiation. It has been reported that miRNAs are involved in bone homeostasis by modulating osteoblast gene expression. In addition, countless evidence has demonstrated that miRNAs dysregulation is involved in the development of osteoporosis and bone fractures. The deregulation of miRNAs expression has also been associated with several malignancies including bone cancer. In this context, bone-associated circulating miRNAs may be useful biomarkers for determining the predisposition, onset and development of osteoporosis, as well as in clinical applications to improve the diagnosis, follow-up and treatment of cancer and metastases. Overall, this review will provide an overview of how miRNAs activities participate in osteogenic/chondrogenic differentiation, while addressing the role of miRNA regulatory effects on target genes. Finally, the role of miRNAs in pathologies and therapies will be presented.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Fernanda Martini
- Department of Medical Sciences, Section of Experimental Medicine, School of Medicine, University of Ferrara. Ferrara, Italy
| |
Collapse
|
19
|
Radmanesh F, Sadeghi Abandansari H, Ghanian MH, Pahlavan S, Varzideh F, Yakhkeshi S, Alikhani M, Moradi S, Braun T, Baharvand H. Hydrogel-mediated delivery of microRNA-92a inhibitor polyplex nanoparticles induces localized angiogenesis. Angiogenesis 2021; 24:657-676. [PMID: 33742265 DOI: 10.1007/s10456-021-09778-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 03/01/2021] [Indexed: 01/07/2023]
Abstract
Localized stimulation of angiogenesis is an attractive strategy to improve the repair of ischemic or injured tissues. Several microRNAs (miRNAs) such as miRNA-92a (miR-92a) have been reported to negatively regulate angiogenesis in ischemic disease. To exploit the clinical potential of miR-92a inhibitors, safe and efficient delivery needs to be established. Here, we used deoxycholic acid-modified polyethylenimine polymeric conjugates (PEI-DA) to deliver a locked nucleic acid (LNA)-based miR-92a inhibitor (LNA-92a) in vitro and in vivo. The positively charged PEI-DA conjugates condense the negatively charged inhibitors into nano-sized polyplexes (135 ± 7.2 nm) with a positive net charge (34.2 ± 10.6 mV). Similar to the 25 kDa-branched PEI (bPEI25) and Lipofectamine RNAiMAX, human umbilical vein endothelial cells (HUVECs) significantly internalized PEI-DA/LNA-92a polyplexes without any obvious cytotoxicity. Down-regulation of miR-92a following the polyplex-mediated delivery of LNA-92a led to a substantial increase in the integrin subunit alpha 5 (ITGA5), the sirtuin-1 (SIRT1) and Krüppel-like factors (KLF) KLF2/4 expression, formation of capillary-like structures by HUVECs, and migration rate of HUVECs in vitro. Furthermore, PEI-DA/LNA-92a resulted in significantly enhanced capillary density in a chicken chorioallantoic membrane (CAM) model. Localized angiogenesis was substantially induced in the subcutaneous tissues of mice by sustained release of PEI-DA/LNA-92a polyplexes from an in situ forming, biodegradable hydrogel based on clickable poly(ethylene glycol) (PEG) macromers. Our results indicate that PEI-DA conjugates efficiently deliver LNA-92a to improve angiogenesis. Localized delivery of RNA interference (RNAi)-based therapeutics via hydrogel-laden PEI-DA polyplex nanoparticles appears to be a safe and effective approach for different therapeutic targets.
Collapse
Affiliation(s)
- Fatemeh Radmanesh
- Uro-Oncology Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Hamid Sadeghi Abandansari
- Department of Cancer Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Babol, Iran
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mohammad Hossein Ghanian
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Sara Pahlavan
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Fahimeh Varzideh
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Saeed Yakhkeshi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mehdi Alikhani
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Sharif Moradi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Thomas Braun
- Max-Planck Institute for Heart and Lung Research, Department of Cardiac Development and Remodeling, Bad Nauheim, Germany
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
- Department of Developmental Biology, University of Science and Culture, Tehran, Iran.
| |
Collapse
|
20
|
Oieni J, Lolli A, D'Atri D, Kops N, Yayon A, van Osch GJVM, Machluf M. Nano-ghosts: Novel biomimetic nano-vesicles for the delivery of antisense oligonucleotides. J Control Release 2021; 333:28-40. [PMID: 33741386 DOI: 10.1016/j.jconrel.2021.03.018] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 03/08/2021] [Accepted: 03/12/2021] [Indexed: 12/18/2022]
Abstract
Antisense oligonucleotides (ASOs) carry an enormous therapeutic potential in different research areas, however, the lack of appropriate carriers for their delivery to the target tissues is hampering their clinical translation. The present study investigates the application of novel biomimetic nano-vesicles, Nano-Ghosts (NGs), for the delivery of ASOs to human mesenchymal stem cells (MSCs), using a microRNA inhibitor (antimiR) against miR-221 as proof-of-concept. The integration of this approach with a hyaluronic acid-fibrin (HA-FB) hydrogel scaffold is also studied, thus expanding the potential of NGs applications in regenerative medicine. The study shows robust antimiR encapsulation in the NGs using electroporation and the NGs ability to be internalized in MSCs and to deliver their cargo while avoiding endo-lysosomal degradation. This leads to rapid and strong knock-down of miR-221 in hMSCs in vitro, both in 2D and 3D hydrogel culture conditions (>90% and > 80% silencing efficiency, respectively). Finally, in vivo studies performed with an osteochondral defect model demonstrate the NGs ability to effectively deliver antimiR to endogenous cells. Altogether, these results prove that the NGs can operate as stand-alone system or as integrated platform in combination with scaffolds for the delivery of ASOs for a wide range of applications in drug delivery and regenerative medicine.
Collapse
Affiliation(s)
- Jacopo Oieni
- Department of Biotechnology and Food Engineering, Technion - Israel Institute of Technology, Haifa 32000, Israel
| | - Andrea Lolli
- Department of Orthopaedics, Erasmus MC, University Medical Center, Rotterdam 3015GD, the Netherlands; Department of Oral and Maxillofacial Surgery, Erasmus MC, University Medical Center, Rotterdam 3015GD, the Netherlands
| | - Domenico D'Atri
- Department of Biotechnology and Food Engineering, Technion - Israel Institute of Technology, Haifa 32000, Israel
| | - Nicole Kops
- Department of Orthopaedics, Erasmus MC, University Medical Center, Rotterdam 3015GD, the Netherlands
| | - Avner Yayon
- Procore Ltd., Weizmann Science Park, 7 Golda Meir St., Ness Ziona 7414002, Israel
| | - Gerjo J V M van Osch
- Department of Orthopaedics, Erasmus MC, University Medical Center, Rotterdam 3015GD, the Netherlands; Department of Otorhinolaryngology, Head and Neck Surgery, Erasmus MC, University Medical Center, Rotterdam, 3015GD, the Netherlands; Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology, 2628, the Netherlands
| | - Marcelle Machluf
- Department of Biotechnology and Food Engineering, Technion - Israel Institute of Technology, Haifa 32000, Israel.
| |
Collapse
|
21
|
An ultrasensitive biosensor based on three-dimensional nanoporous conducting polymer decorated with gold nanoparticles for microRNA detection. Microchem J 2021. [DOI: 10.1016/j.microc.2020.105780] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
22
|
Kim YS, Mikos AG. Emerging strategies in reprogramming and enhancing the fate of mesenchymal stem cells for bone and cartilage tissue engineering. J Control Release 2021; 330:565-574. [DOI: 10.1016/j.jconrel.2020.12.055] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 12/21/2020] [Accepted: 12/29/2020] [Indexed: 02/06/2023]
|
23
|
Costard LS, Kelly DC, Power RN, Hobbs C, Jaskaniec S, Nicolosi V, Cavanagh BL, Curtin CM, O’Brien FJ. Layered Double Hydroxide as a Potent Non-viral Vector for Nucleic Acid Delivery Using Gene-Activated Scaffolds for Tissue Regeneration Applications. Pharmaceutics 2020; 12:pharmaceutics12121219. [PMID: 33339452 PMCID: PMC7765978 DOI: 10.3390/pharmaceutics12121219] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/02/2020] [Accepted: 12/10/2020] [Indexed: 02/04/2023] Open
Abstract
Nonviral vectors offer a safe alternative to viral vectors for gene therapy applications, albeit typically exhibiting lower transfection efficiencies. As a result, there remains a significant need for the development of a nonviral delivery system with low cytotoxicity and high transfection efficacy as a tool for safe and transient gene delivery. This study assesses MgAl-NO3 layered double hydroxide (LDH) as a nonviral vector to deliver nucleic acids (pDNA, miRNA and siRNA) to mesenchymal stromal cells (MSCs) in 2D culture and using a 3D tissue engineering scaffold approach. Nanoparticles were formulated by complexing LDH with pDNA, microRNA (miRNA) mimics and inhibitors, and siRNA at varying mass ratios of LDH:nucleic acid. In 2D monolayer, pDNA delivery demonstrated significant cytotoxicity issues, and low cellular transfection was deemed to be a result of the poor physicochemical properties of the LDH–pDNA nanoparticles. However, the lower mass ratios required to successfully complex with miRNA and siRNA cargo allowed for efficient delivery to MSCs. Furthermore, incorporation of LDH–miRNA nanoparticles into collagen-nanohydroxyapatite scaffolds resulted in successful overexpression of miRNA in MSCs, demonstrating the development of an efficacious miRNA delivery platform for gene therapy applications in regenerative medicine.
Collapse
Affiliation(s)
- Lara S. Costard
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland (RCSI), 123 St Stephen’s Green, D02 YN77 Dublin, Ireland; (L.S.C.); (D.C.K.); (R.N.P.)
| | - Domhnall C. Kelly
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland (RCSI), 123 St Stephen’s Green, D02 YN77 Dublin, Ireland; (L.S.C.); (D.C.K.); (R.N.P.)
- Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland, Galway (NUI, Galway), H91 TK33 Galway, Ireland
| | - Rachael N. Power
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland (RCSI), 123 St Stephen’s Green, D02 YN77 Dublin, Ireland; (L.S.C.); (D.C.K.); (R.N.P.)
| | - Christopher Hobbs
- Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and Trinity College Dublin (TCD), College Green, D02 PN40 Dublin, Ireland; (C.H.); (S.J.); (V.N.)
- School of Chemistry and Centre for Research on Adaptive Nanostructures and Nanodevices (CRANN), Trinity College Dublin, College Green, D02 PN40 Dublin, Ireland
| | - Sonia Jaskaniec
- Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and Trinity College Dublin (TCD), College Green, D02 PN40 Dublin, Ireland; (C.H.); (S.J.); (V.N.)
- School of Chemistry and Centre for Research on Adaptive Nanostructures and Nanodevices (CRANN), Trinity College Dublin, College Green, D02 PN40 Dublin, Ireland
| | - Valeria Nicolosi
- Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and Trinity College Dublin (TCD), College Green, D02 PN40 Dublin, Ireland; (C.H.); (S.J.); (V.N.)
- School of Chemistry and Centre for Research on Adaptive Nanostructures and Nanodevices (CRANN), Trinity College Dublin, College Green, D02 PN40 Dublin, Ireland
| | - Brenton L. Cavanagh
- Cellular and Molecular Imaging Core, RCSI, 123 St Stephen’s Green, D02 YN77 Dublin, Ireland;
| | - Caroline M. Curtin
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland (RCSI), 123 St Stephen’s Green, D02 YN77 Dublin, Ireland; (L.S.C.); (D.C.K.); (R.N.P.)
- Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and Trinity College Dublin (TCD), College Green, D02 PN40 Dublin, Ireland; (C.H.); (S.J.); (V.N.)
- Trinity Centre for BioMedical Engineering, Trinity Biomedical Sciences Institute, TCD, College Green, D02 PN40 Dublin, Ireland
- Correspondence: (C.M.C.); (F.J.O.); Tel.: +353-1-4028620 (C.M.C.); +353-1-4028533 (F.J.O.)
| | - Fergal J. O’Brien
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland (RCSI), 123 St Stephen’s Green, D02 YN77 Dublin, Ireland; (L.S.C.); (D.C.K.); (R.N.P.)
- Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland, Galway (NUI, Galway), H91 TK33 Galway, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and Trinity College Dublin (TCD), College Green, D02 PN40 Dublin, Ireland; (C.H.); (S.J.); (V.N.)
- Trinity Centre for BioMedical Engineering, Trinity Biomedical Sciences Institute, TCD, College Green, D02 PN40 Dublin, Ireland
- Correspondence: (C.M.C.); (F.J.O.); Tel.: +353-1-4028620 (C.M.C.); +353-1-4028533 (F.J.O.)
| |
Collapse
|
24
|
Singh P, Singh A, Shah S, Vataliya J, Mittal A, Chitkara D. RNA Interference Nanotherapeutics for Treatment of Glioblastoma Multiforme. Mol Pharm 2020; 17:4040-4066. [PMID: 32902291 DOI: 10.1021/acs.molpharmaceut.0c00709] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Nucleic acid therapeutics for RNA interference (RNAi) are gaining attention in the treatment and management of several kinds of the so-called "undruggable" tumors via targeting specific molecular pathways or oncogenes. Synthetic ribonucleic acid (RNAs) oligonucleotides like siRNA, miRNA, shRNA, and lncRNA have shown potential as novel therapeutics. However, the delivery of such oligonucleotides is significantly hampered by their physiochemical (such as hydrophilicity, negative charge, and instability) and biopharmaceutical features (in vivo serum stability, fast renal clearance, interaction with extracellular proteins, and hindrance in cellular internalization) that markedly reduce their biological activity. Recently, several nanocarriers have evolved as suitable non-viral vectors for oligonucleotide delivery, which are known to either complex or conjugate with these oligonucleotides efficiently and also overcome the extracellular and intracellular barriers, thereby allowing access to the tumoral micro-environment for the better and desired outcome in glioblastoma multiforme (GBM). This Review focuses on the up-to-date advancements in the field of RNAi nanotherapeutics utilized for GBM treatment.
Collapse
Affiliation(s)
- Prabhjeet Singh
- Department of Pharmacy, Birla Institute of Technology and Science (BITS) Pilani, Pilani Campus, Vidya Vihar, Pilani - 333 031, Rajasthan, India
| | - Aditi Singh
- Department of Pharmacy, Birla Institute of Technology and Science (BITS) Pilani, Pilani Campus, Vidya Vihar, Pilani - 333 031, Rajasthan, India
| | - Shruti Shah
- Department of Pharmacy, Birla Institute of Technology and Science (BITS) Pilani, Pilani Campus, Vidya Vihar, Pilani - 333 031, Rajasthan, India
| | - Jalpa Vataliya
- Department of Pharmacy, Birla Institute of Technology and Science (BITS) Pilani, Pilani Campus, Vidya Vihar, Pilani - 333 031, Rajasthan, India
| | - Anupama Mittal
- Department of Pharmacy, Birla Institute of Technology and Science (BITS) Pilani, Pilani Campus, Vidya Vihar, Pilani - 333 031, Rajasthan, India
| | - Deepak Chitkara
- Department of Pharmacy, Birla Institute of Technology and Science (BITS) Pilani, Pilani Campus, Vidya Vihar, Pilani - 333 031, Rajasthan, India
| |
Collapse
|
25
|
Mendrek B, Fus-Kujawa A, Teper P, Botor M, Kubacki J, Sieroń AL, Kowalczuk A. Star polymer-based nanolayers with immobilized complexes of polycationic stars and DNA for deposition gene delivery and recovery of intact transfected cells. Int J Pharm 2020; 589:119823. [PMID: 32861771 DOI: 10.1016/j.ijpharm.2020.119823] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 07/31/2020] [Accepted: 08/24/2020] [Indexed: 12/31/2022]
Abstract
We designed a novel thermoresponsive system of nanolayers composed of star poly[oligo(ethylene glycol) methacrylate]s (S-POEGMA) covalently bonded to a solid support and covered with polyplexes of cationic star polymers and plasmid DNA (pDNA). S-POEGMA stars were attached to the solid support via a UV-mediated "grafting to" method. To the best of our knowledge, for the first time, the conformational changes of obtained star nanolayers, occurring with changes in temperature, were studied using a quartz crystal microbalance technique. Next, the polyplexes of star poly[N,N'-dimethylaminoethyl methacrylate-ran-di(ethylene glycol) methacrylate] (S-P(DMAEMA-DEGMA)) with pDNA, exhibiting a phase transition temperature (TCP) in culture medium DMEM, were deposited on S-POEGMA layers when the temperature increased above the TCP of polyplex. The thermoresponsivity of the system was then the main mechanism for controlling the adhesion, proliferation, transfection and detachment of HT-1080 cells. The nanolayers promoted the effective cell culture and delivered nucleic acids into cells, with a transfection efficiency several times higher than that of the control. The detachment of the transfected cells was regulated only by the change of temperature. The studies demonstrated that we obtained a novel and effective system, based on a star polymer architecture, useful for gene delivery and tissue engineering applications.
Collapse
Affiliation(s)
- Barbara Mendrek
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, M. Curie-Sklodowskiej 34, 41-819 Zabrze, Poland
| | - Agnieszka Fus-Kujawa
- Department of Molecular Biology and Genetics, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Medykow 18 Street, 40-752 Katowice, Poland
| | - Paulina Teper
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, M. Curie-Sklodowskiej 34, 41-819 Zabrze, Poland
| | - Malwina Botor
- Department of Molecular Biology and Genetics, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Medykow 18 Street, 40-752 Katowice, Poland
| | - Jerzy Kubacki
- A. Chelkowski Institute of Physics, University of Silesia, Uniwersytecka 4, 40-007 Katowice, Poland; Silesian Center for Education and Interdisciplinary Research, 75 Pulku Piechoty 1A, 41-500 Chorzów, Poland
| | - Aleksander L Sieroń
- Department of Molecular Biology and Genetics, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Medykow 18 Street, 40-752 Katowice, Poland
| | - Agnieszka Kowalczuk
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, M. Curie-Sklodowskiej 34, 41-819 Zabrze, Poland.
| |
Collapse
|
26
|
Small Extracellular Vesicles from Human Fetal Dermal Cells and Their MicroRNA Cargo: KEGG Signaling Pathways Associated with Angiogenesis and Wound Healing. Stem Cells Int 2020; 2020:8889379. [PMID: 32855639 PMCID: PMC7443045 DOI: 10.1155/2020/8889379] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 07/08/2020] [Accepted: 07/21/2020] [Indexed: 02/06/2023] Open
Abstract
The use of cell secreted factors in clinical settings could be an alternative to conventional cell therapy, with the advantage of limiting concerns generally associated with traditional cell transplantation, such as tumorigenicity, immunoreactivity, and carrying of infections. Based on our published data, we predict a potential role for extracellular vesicles (EVs) in contributing to the proangiogenic activity of human fetal dermal cell secretome. Depletion of nanosized EVs from secretome significantly impaired its ability to induce formation of mesh-like structures in vitro. The isolated EVs were characterized for size and concentration by nanoparticle tracking analysis, and for protein markers (Rab5+, Alix+, CD63+, and calnexin−). The microRNA profile of EVs revealed 87 microRNAs significantly upregulated (≥15-fold increase) in fetal compared to adult dermal cell-derived EVs. Interestingly, these upregulated microRNAs included microRNAs with a validated role in angiogenesis according to literature. Moreover, the DIANA-TarBase v7.0 analysis confirmed enrichment in the KEGG signaling pathways associated with angiogenesis and wound healing, with the identification of putative target genes including thrombospondin 1. To validate the in silico data, EVs were also characterized for total protein contents. When tested in in vitro angiogenesis, fetal dermal cell-derived EVs were more effective than their adult counterpart in inducing formation of complete mesh-like structures. Furthermore, treatment of fibroblasts with fetal dermal-derived EVs determined a 4-fold increase of thrombospondin 1 protein amounts compared with the untreated fibroblasts. Finally, visualization of CSFE-labeled EVs in the cytosol of target cells suggested a successful uptake of these particles at 4-8 hours of incubation. We conclude that EVs are important contributors of the proangiogenic effect of fetal dermal cell secretome. Hence, EVs could also serve as vehicle for a successful delivery of microRNAs or other molecules of therapeutic interest to target cells.
Collapse
|
27
|
Borja-Gonzalez M, Casas-Martinez JC, McDonagh B, Goljanek-Whysall K. Aging Science Talks: The role of miR-181a in age-related loss of muscle mass and function. TRANSLATIONAL MEDICINE OF AGING 2020; 4:81-85. [PMID: 32835152 PMCID: PMC7341035 DOI: 10.1016/j.tma.2020.07.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 07/03/2020] [Indexed: 02/07/2023] Open
Affiliation(s)
- Maria Borja-Gonzalez
- School of Medicine, Physiology, National University of Ireland Galway, Galway, H91 W5P7, Ireland
| | - Jose C Casas-Martinez
- School of Medicine, Physiology, National University of Ireland Galway, Galway, H91 W5P7, Ireland
| | - Brian McDonagh
- School of Medicine, Physiology, National University of Ireland Galway, Galway, H91 W5P7, Ireland
| | - Katarzyna Goljanek-Whysall
- School of Medicine, Physiology, National University of Ireland Galway, Galway, H91 W5P7, Ireland
- Institute of Aging and Chronic Disease & The Medical Research Council Versus Arthritis Centre for Integrated Research Into Musculoskeletal Aging, CIMA, University of Liverpool, Liverpool, L7 8TJ, UK
| |
Collapse
|
28
|
Castaño IM, Raftery RM, Chen G, Cavanagh B, Quinn B, Duffy GP, O'Brien FJ, Curtin CM. Rapid bone repair with the recruitment of CD206 +M2-like macrophages using non-viral scaffold-mediated miR-133a inhibition of host cells. Acta Biomater 2020; 109:267-279. [PMID: 32251781 DOI: 10.1016/j.actbio.2020.03.042] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 03/10/2020] [Accepted: 03/27/2020] [Indexed: 01/01/2023]
Abstract
microRNAs offer vast therapeutic potential for multiple disciplines. From a bone perspective, inhibition of miR-133a may offer potential to enhance Runx2 activity and increase bone repair. This study aims to assess the therapeutic capability of antagomiR-133a delivery from collagen-nanohydroxyapatite (coll-nHA) scaffolds following cell-free implantation in rat calvarial defects (7 mm diameter). This is, to the best of our knowledge, the first report of successful in vivo antagomiR uptake in host cells of fully immunocompetent animals without distribution to other off-target tissues. Our results demonstrate the localized release of antagomiR-133a to the implant site at 1 week post-implantation with increased calcium deposits already evident in the antagomiR-133a loaded scaffolds at this early timepoint. This was followed by an approximate 2-fold increase in bone volume versus antagomiR-free scaffolds and a significant 10-fold increase over the empty defect controls, after just 4 weeks. An increase in host CD206+ cells suggests an accelerated pro-remodeling response by M2-like macrophages accompanying bone repair with this treatment. Overall, this non-viral scaffold-mediated antagomiR-133a delivery platform demonstrates capability to accelerate bone repair in vivo - without the addition of exogenous cells - and underlines the role of M2 macrophage-like cells in directing accelerated bone repair. Expanding the repertoire of this platform to deliver alternative miRNAs offers exciting possibilities for a variety of therapeutic indications. STATEMENT OF SIGNIFICANCE: microRNAs, small non-coding RNA molecules involved in gene regulation, may have potential as a new class of bone healing therapeutics as they can enhance the regenerative capacity of bone-forming cells. We developed a collagen-nanohydroxyapatite-microRNA scaffold system to investigate whether miR133a inhibition can enhance osteogenesis in rat MSCs and ultimately accelerate endogenous bone repair by host cells in vivo without pre-seeding cells prior to implantation. Overall, this off-the-shelf, non-viral scaffold-mediated antagomiR-133a delivery platform demonstrates capability to accelerate bone repair in vivo - without the requirement of exogenous cells - and highlights the role of CD206+M2 macrophage-like cells in guiding accelerated bone repair. Translating the repertoire of this platform to deliver alternative miRNAs offers exciting possibilities for a vast myriad of therapeutic indications.
Collapse
|
29
|
Leng Q, Chen L, Lv Y. RNA-based scaffolds for bone regeneration: application and mechanisms of mRNA, miRNA and siRNA. Am J Cancer Res 2020; 10:3190-3205. [PMID: 32194862 PMCID: PMC7053199 DOI: 10.7150/thno.42640] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 01/16/2020] [Indexed: 02/07/2023] Open
Abstract
Globally, more than 1.5 million patients undergo bone graft surgeries annually, and the development of biomaterial scaffolds that mimic natural bone for bone grafting remains a tremendous challenge. In recent decades, due to the improved understanding of the mechanisms of bone remodeling and the rapid development of gene therapy, RNA (including messenger RNA (mRNA), microRNA (miRNA), and short interfering RNA (siRNA)) has attracted increased attention as a new tool for bone tissue engineering due to its unique nature and great potential to cure bone defects. Different types of RNA play roles via a variety of mechanisms in bone-related cells in vivo as well as after synthesis in vitro. In addition, RNAs are delivered to injured sites by loading into scaffolds or systemic administration after combination with vectors for bone tissue engineering. However, the challenge of effectively and stably delivering RNA into local tissue remains to be solved. This review describes the mechanisms of the three types of RNAs and the application of the relevant types of RNA delivery vectors and scaffolds in bone regeneration. The improvements in their development are also discussed.
Collapse
|
30
|
Piperno A, Mazzaglia A, Scala A, Pennisi R, Zagami R, Neri G, Torcasio SM, Rosmini C, Mineo PG, Potara M, Focsan M, Astilean S, Zhou GG, Sciortino MT. Casting Light on Intracellular Tracking of a New Functional Graphene-Based MicroRNA Delivery System by FLIM and Raman Imaging. ACS APPLIED MATERIALS & INTERFACES 2019; 11:46101-46111. [PMID: 31729219 DOI: 10.1021/acsami.9b15826] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The theranostic ability of a new fluorescently labeled cationic cyclodextrin-graphene nanoplatform (GCD@Ada-Rhod) was investigated by studying its intracellular trafficking and its ability to deliver plasmid DNA and microRNA. The nanoplatform was synthesized by both covalent and supramolecular approaches, and its chemical structure, morphology, and colloidal behavior were investigated by TGA, TEM, spectroscopic analysis such as UV-vis, fluorescence emission, DLS, and ζ-potential measurements. The cellular internalization of GCD@Ada-Rhod and its perinuclear localization were assessed by FLIM, Raman imaging, and fluorescence microscopy. Biological experiments with pCMS-EGFP and miRNA-15a evidenced the excellent capability of GCD@Ada-Rhod to deliver both pDNA and microRNA without significant cytotoxicity. The biological results evidenced an unforeseen caveolae-mediated endocytosis internalization pathway (generally expected for particles <200 nm), despite the fact that the GCD@Ada-Rhod size is about 400 nm (by DLS and TEM data). We supposed that the internalization pathway was driven by physical-chemical features of GCD@Ada-Rhod, and the caveolae-mediated uptake enhanced the transfection efficiency, avoiding the lysosomal acid degradation. The cellular effects of internalized miRNA-15a on the oncogene protein BCL-2 were investigated at two different concentrations (N/P = 10 and 5), and a reduction of the BCL-2 level was detected at a low concentration (i.e., N/P = 10). miRNA-15a is considered an ideal cancer therapy molecule due to its activity on multiple transcription factors, and the elucidation of the correlation between the concentration of delivered miRNA-15a and the down-/up-regulation of the BCL-2 level, documented for the first time in this work, could be an important contribution to guide its clinical application.
Collapse
Affiliation(s)
- Anna Piperno
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences , University of Messina , V.le F. Stagno d'Alcontres 31 , 98166 Messina , Italy
| | - Antonino Mazzaglia
- CNR-ISMN c/o Department of Chemical, Biological, Pharmaceutical and Environmental Sciences , University of Messina , V.le F. Stagno d'Alcontres 31 , 98166 Messina , Italy
| | - Angela Scala
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences , University of Messina , V.le F. Stagno d'Alcontres 31 , 98166 Messina , Italy
| | - Rosamaria Pennisi
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences , University of Messina , V.le F. Stagno d'Alcontres 31 , 98166 Messina , Italy
- Shenzhen International Institute for Biomedical Research , Shenzhen , Guangdong 518119 , China
| | - Roberto Zagami
- CNR-ISMN c/o Department of Chemical, Biological, Pharmaceutical and Environmental Sciences , University of Messina , V.le F. Stagno d'Alcontres 31 , 98166 Messina , Italy
| | - Giulia Neri
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences , University of Messina , V.le F. Stagno d'Alcontres 31 , 98166 Messina , Italy
| | - Serena M Torcasio
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences , University of Messina , V.le F. Stagno d'Alcontres 31 , 98166 Messina , Italy
| | - Consolato Rosmini
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences , University of Messina , V.le F. Stagno d'Alcontres 31 , 98166 Messina , Italy
| | - Placido G Mineo
- Department of Chemical Sciences , University of Catania , V.le A. Doria 6 , 95125 Catania , Italy
| | - Monica Potara
- Nanobiophotonics and Laser Microspectroscopy Center, Interdisciplinary Research Institute in Bio-Nano-Sciences , Babes-Bolyai University , T. Laurian Str. 42 , 400271 Cluj-Napoca , Romania
| | - Monica Focsan
- Nanobiophotonics and Laser Microspectroscopy Center, Interdisciplinary Research Institute in Bio-Nano-Sciences , Babes-Bolyai University , T. Laurian Str. 42 , 400271 Cluj-Napoca , Romania
| | - Simion Astilean
- Nanobiophotonics and Laser Microspectroscopy Center, Interdisciplinary Research Institute in Bio-Nano-Sciences , Babes-Bolyai University , T. Laurian Str. 42 , 400271 Cluj-Napoca , Romania
- Department of Biomolecular Physics, Faculty of Physics , Babes-Bolyai University , M Kogalniceanu Str. 1 , 400084 Cluj-Napoca , Romania
| | - Grace Guoying Zhou
- Shenzhen International Institute for Biomedical Research , Shenzhen , Guangdong 518119 , China
| | - Maria Teresa Sciortino
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences , University of Messina , V.le F. Stagno d'Alcontres 31 , 98166 Messina , Italy
| |
Collapse
|
31
|
Moncal KK, Aydin RST, Abu-Laban M, Heo DN, Rizk E, Tucker SM, Lewis GS, Hayes D, Ozbolat IT. Collagen-infilled 3D printed scaffolds loaded with miR-148b-transfected bone marrow stem cells improve calvarial bone regeneration in rats. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 105:110128. [PMID: 31546389 PMCID: PMC6761997 DOI: 10.1016/j.msec.2019.110128] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 08/05/2019] [Accepted: 08/23/2019] [Indexed: 12/12/2022]
Abstract
Differentiation of progenitors in a controlled environment improves the repair of critical-sized calvarial bone defects; however, integrating micro RNA (miRNA) therapy with 3D printed scaffolds still remains a challenge for craniofacial reconstruction. In this study, we aimed to engineer three-dimensional (3D) printed hybrid scaffolds as a new ex situ miR-148b expressing delivery system for osteogenic induction of rat bone marrow stem cells (rBMSCs) in vitro, and also in vivo in critical-sized rat calvarial defects. miR-148b-transfected rBMSCs underwent early differentiation in collagen-infilled 3D printed hybrid scaffolds, expressing significant levels of osteogenic markers compared to non-transfected rBMSCs, as confirmed by gene expression and immunohistochemical staining. Furthermore, after eight weeks of implantation, micro-computed tomography, histology and immunohistochemical staining results indicated that scaffolds loaded with miR-148b-transfected rBMSCs improved bone regeneration considerably compared to the scaffolds loaded with non-transfected rBMSCs and facilitated near-complete repair of critical-sized calvarial defects. In conclusion, our results demonstrate that collagen-infilled 3D printed scaffolds serve as an effective system for miRNA transfected progenitor cells, which has a promising potential for stimulating osteogenesis and calvarial bone repair.
Collapse
Affiliation(s)
- Kazim K Moncal
- Engineering Science and Mechanics, Penn State University, University Park, PA, USA; Huck Institutes of the Life Sciences, Penn State University, University Park, PA, USA
| | - R Seda Tigli Aydin
- Huck Institutes of the Life Sciences, Penn State University, University Park, PA, USA; Department of Biomedical Engineering, Bulent Ecevit University, Incivez, Zonguldak, Turkey
| | - Mohammad Abu-Laban
- Department of Biomedical Engineering, Penn State University, University Park, PA, USA; Materials Research Institute, Penn State University, University Park, PA, USA
| | - Dong N Heo
- Engineering Science and Mechanics, Penn State University, University Park, PA, USA; Huck Institutes of the Life Sciences, Penn State University, University Park, PA, USA
| | - Elias Rizk
- Department of Neurosurgery, Penn State University, Hershey, PA, USA
| | - Scott M Tucker
- Engineering Science and Mechanics, Penn State University, University Park, PA, USA; Department of Orthopaedics and Rehabilitation, Penn State University, Hershey, PA, USA
| | - Gregory S Lewis
- Engineering Science and Mechanics, Penn State University, University Park, PA, USA; Department of Orthopaedics and Rehabilitation, Penn State University, Hershey, PA, USA
| | - Daniel Hayes
- Huck Institutes of the Life Sciences, Penn State University, University Park, PA, USA; Department of Biomedical Engineering, Penn State University, University Park, PA, USA; Materials Research Institute, Penn State University, University Park, PA, USA
| | - Ibrahim T Ozbolat
- Engineering Science and Mechanics, Penn State University, University Park, PA, USA; Huck Institutes of the Life Sciences, Penn State University, University Park, PA, USA; Department of Biomedical Engineering, Penn State University, University Park, PA, USA; Materials Research Institute, Penn State University, University Park, PA, USA.
| |
Collapse
|
32
|
Levingstone TJ, Herbaj S, Dunne NJ. Calcium Phosphate Nanoparticles for Therapeutic Applications in Bone Regeneration. NANOMATERIALS (BASEL, SWITZERLAND) 2019; 9:E1570. [PMID: 31698700 PMCID: PMC6915504 DOI: 10.3390/nano9111570] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 10/19/2019] [Accepted: 11/01/2019] [Indexed: 01/01/2023]
Abstract
Bone injuries and diseases constitute a burden both socially and economically, as the consequences of a lack of effective treatments affect both the patients' quality of life and the costs on the health systems. This impended need has led the research community's efforts to establish efficacious bone tissue engineering solutions. There has been a recent focus on the use of biomaterial-based nanoparticles for the delivery of therapeutic factors. Among the biomaterials being considered to date, calcium phosphates have emerged as one of the most promising materials for bone repair applications due to their osteoconductivity, osteoinductivity and their ability to be resorbed in the body. Calcium phosphate nanoparticles have received particular attention as non-viral vectors for gene therapy, as factors such as plasmid DNAs, microRNAs (miRNA) and silencing RNA (siRNAs) can be easily incorporated on their surface. Calcium phosphate nanoparticles loaded with therapeutic factors have also been delivered to the site of bone injury using scaffolds and hydrogels. This review provides an extensive overview of the current state-of-the-art relating to the design and synthesis of calcium phosphate nanoparticles as carriers for therapeutic factors, the mechanisms of therapeutic factors' loading and release, and their application in bone tissue engineering.
Collapse
Affiliation(s)
- Tanya J. Levingstone
- School of Mechanical and Manufacturing Engineering, Dublin City University, Dublin 9, Ireland; (T.J.L.); (S.H.)
- Centre for Medical Engineering Research, School of Mechanical and Manufacturing Engineering, Dublin City University, Stokes Building, Collins Avenue, Dublin 9, Ireland
- Tissue Engineering Research Group, Royal College of Surgeons in Ireland, Dublin 2, Ireland
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 9, Ireland
| | - Simona Herbaj
- School of Mechanical and Manufacturing Engineering, Dublin City University, Dublin 9, Ireland; (T.J.L.); (S.H.)
- Centre for Medical Engineering Research, School of Mechanical and Manufacturing Engineering, Dublin City University, Stokes Building, Collins Avenue, Dublin 9, Ireland
| | - Nicholas J. Dunne
- School of Mechanical and Manufacturing Engineering, Dublin City University, Dublin 9, Ireland; (T.J.L.); (S.H.)
- Centre for Medical Engineering Research, School of Mechanical and Manufacturing Engineering, Dublin City University, Stokes Building, Collins Avenue, Dublin 9, Ireland
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 9, Ireland
- School of Pharmacy, Queen’s University Belfast, Belfast BT7 1NN, UK
- Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin 2, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland and Trinity College Dublin, Dublin 2, Ireland
| |
Collapse
|
33
|
Gonzalez-Fernandez T, Sikorski P, Leach JK. Bio-instructive materials for musculoskeletal regeneration. Acta Biomater 2019; 96:20-34. [PMID: 31302298 PMCID: PMC6717669 DOI: 10.1016/j.actbio.2019.07.014] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Revised: 06/26/2019] [Accepted: 07/09/2019] [Indexed: 02/06/2023]
Abstract
The prevalence and cost of disorders affecting the musculoskeletal system are predicted to rise significantly in the coming years due to the aging global population and the increase of associated risk factors. Despite being the second largest cause of disability, the clinical options for therapeutic intervention remain limited. The clinical translation of cell-based therapies for the treatment of musculoskeletal disorders faces many challenges including maintenance of cell survival in the harsh in vivo environment and the lack of control over regulating cell phenotype upon implantation. In order to address these challenges, the development of bio-instructive materials to modulate cell behavior has taken center stage as a strategy to increase the therapeutic potential of various cell populations. However, the determination of the necessary cues for a specific application and how these signals should be presented from a biomaterial remains elusive. This review highlights recent biochemical and physical strategies used to engineer bio-instructive materials for the repair of musculoskeletal tissues. There is a particular emphasis on emerging efforts such as the engineering of immunomodulatory and antibacterial materials, as well as the incorporation of these strategies into biofabrication and organ-on-a-chip approaches. STATEMENT OF SIGNIFICANCE: Disorders affecting the musculoskeletal system affect individuals across the lifespan and have a profound effect on mobility and quality of life. While small defects in many tissues can heal successfully, larger defects are often unable to heal or instead heal with inferior quality fibrous tissue and require clinical intervention. Cell-based therapies are a promising option for clinical translation, yet challenges related to maintaining cell survival and instructing cell phenotype upon implantation have limited the success of this approach. Bio-instructive materials provide an exciting opportunity to modulate cell behavior and enhance the efficacy of cell-based approaches for musculoskeletal repair. However, the identification of critical instructive cues and how to present these stimuli is a focus of intense investigation. This review highlights recent biochemical and physical strategies used to engineer bio-instructive materials for the repair of musculoskeletal tissues, while also considering exciting progress in the engineering of immunomodulatory and antibacterial materials.
Collapse
Affiliation(s)
| | - Pawel Sikorski
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, USA; Department of Physics, Norwegian University of Science and Technology, NTNU, Trondheim, Norway
| | - J Kent Leach
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, USA; Department of Orthopaedic Surgery, School of Medicine, UC Davis Health, Sacramento, CA, USA.
| |
Collapse
|
34
|
Hosseinpour S, He Y, Nanda A, Ye Q. MicroRNAs Involved in the Regulation of Angiogenesis in Bone Regeneration. Calcif Tissue Int 2019; 105:223-238. [PMID: 31175386 DOI: 10.1007/s00223-019-00571-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 06/01/2019] [Indexed: 12/11/2022]
Abstract
MicroRNAs (miRNAs) as a newly founded and thriving non-coding endogenous class of molecules which regulate many cellular pathways after transcription have been extensively investigated in regenerative medicine. In this systematic review, we sought to analyze miRNAs-mediated therapeutic approaches for influencing angiogenesis in bone tissue/bone regeneration. An electronic search in MEDLINE, Scopus, EMBASE, Cochrane library, web of science, and google scholar with no time limit were done on English publications. All types of original articles which a miRNA for angiogenesis in bone regeneration were included in our review. In the process of reviewing, we used PRISMA guideline and, SYRCLE's and science in risk assessment and policy tools for analyzing risk of bias. Among 751 initial retrieved records, 16 studies met the inclusion criteria and were fully assessed in this review. 275 miRNAs, one miRNA 195~497 cluster, and one Cysteine-rich 61 short hairpin RNA were differentially expressed during bone regeneration with 24 predicted targets reported in these studies. Among these miRNAs, miRNA-7b, -9, -21, -26a, -27a, -210, -378, -195~497 cluster, -378 and -675 positively promoted both angiogenesis and osteogenesis, whereas miRNA-10a, -222 and -494 inhibited both processes. The most common target was vasculoendothelial growth factor-signaling pathway. Recent evidence has demonstrated that miRNAs actively participated in angio-osteogenic coupling that can improve their therapeutic potentials for the treatment of bone-related diseases and bone regeneration. However, there is still need for further research to unravel the exact mechanisms.
Collapse
Affiliation(s)
- Sepanta Hosseinpour
- School of Dentistry, The University of Queensland, Herston, Brisbane, QLD, 4006, Australia
| | - Yan He
- School of Dentistry, The University of Queensland, Herston, Brisbane, QLD, 4006, Australia
| | - Ashwin Nanda
- School of Dentistry, The University of Queensland, Herston, Brisbane, QLD, 4006, Australia
| | - Qingsong Ye
- School of Dentistry, The University of Queensland, Herston, Brisbane, QLD, 4006, Australia.
| |
Collapse
|
35
|
Kelly DC, Raftery RM, Curtin CM, O'Driscoll CM, O'Brien FJ. Scaffold-Based Delivery of Nucleic Acid Therapeutics for Enhanced Bone and Cartilage Repair. J Orthop Res 2019; 37:1671-1680. [PMID: 31042304 DOI: 10.1002/jor.24321] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 04/09/2019] [Indexed: 02/04/2023]
Abstract
Recent advances in tissue engineering have made progress toward the development of biomaterials capable of the delivery of growth factors, such as bone morphogenetic proteins, in order to promote enhanced tissue repair. However, controlling the release of these growth factors on demand and within the desired localized area is a significant challenge and the associated high costs and side effects of uncontrolled delivery have proven increasingly problematic in clinical orthopedics. Gene therapy may be a valuable tool to avoid the limitations of local delivery of growth factors. Following a series of setbacks in the 1990s, the field of gene therapy is now seeing improvements in safety and efficacy resulting in substantial clinical progress and a resurgence in confidence. Biomaterial scaffold-mediated gene therapy provides a template for cell infiltration and tissue formation while promoting transfection of cells to engineer therapeutic proteins in a sustained but ultimately transient fashion. Additionally, scaffold-mediated delivery of RNA-based therapeutics can silence specific genes associated with orthopedic pathological states. This review will provide an overview of the current state-of-the-art in the field of gene-activated scaffolds and their use within orthopedic tissue engineering applications. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 37:1671-1680, 2019.
Collapse
Affiliation(s)
- Domhnall C Kelly
- Tissue Engineering Research Group (TERG), Department of Anatomy, Royal College of Surgeons in Ireland (RCSI), Dublin, Ireland.,Trinity Centre of Bioengineering (TCBE), Trinity College Dublin (TCD), Dublin, Ireland.,Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, Ireland.,Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI, Galway), Galway, Ireland
| | - Rosanne M Raftery
- Tissue Engineering Research Group (TERG), Department of Anatomy, Royal College of Surgeons in Ireland (RCSI), Dublin, Ireland.,Trinity Centre of Bioengineering (TCBE), Trinity College Dublin (TCD), Dublin, Ireland.,Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, Ireland
| | - Caroline M Curtin
- Tissue Engineering Research Group (TERG), Department of Anatomy, Royal College of Surgeons in Ireland (RCSI), Dublin, Ireland.,Trinity Centre of Bioengineering (TCBE), Trinity College Dublin (TCD), Dublin, Ireland.,Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, Ireland
| | - Caitriona M O'Driscoll
- Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI, Galway), Galway, Ireland.,Pharmacodelivery Group, School of Pharmacy, University College Cork, Cork, Ireland
| | - Fergal J O'Brien
- Tissue Engineering Research Group (TERG), Department of Anatomy, Royal College of Surgeons in Ireland (RCSI), Dublin, Ireland.,Trinity Centre of Bioengineering (TCBE), Trinity College Dublin (TCD), Dublin, Ireland.,Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, Ireland.,Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI, Galway), Galway, Ireland
| |
Collapse
|
36
|
Arriaga MA, Ding MH, Gutierrez AS, Chew SA. The Application of microRNAs in Biomaterial Scaffold-Based Therapies for Bone Tissue Engineering. Biotechnol J 2019; 14:e1900084. [PMID: 31166084 DOI: 10.1002/biot.201900084] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 05/28/2019] [Indexed: 12/13/2022]
Abstract
In recent years, the application of microRNAs (miRNAs) or anti-microRNAs (anti-miRNAs) that can induce expression of the runt-related transcription factor 2 (RUNX2), a master regulator of osteogenesis, has been investigated as a promising alternative bone tissue engineering strategy. In this review, biomaterial scaffold-based applications that have been used to deliver cells expressing miRNAs or anti-miRNAs that induce expression of RUNX2 for bone tissue engineering are discussed. An overview of the components of the scaffold-based therapies including the miRNAs/anti-miRNAs, cell types, gene delivery vectors, and scaffolds that have been applied are provided. To date, there have been nine miRNAs/anti-miRNAs (i.e., miRNA-26a, anti-miRNA-31, anti-miRNA-34a, miRNA-135, anti-miRNA-138, anti-miRNA-146a, miRNA-148b, anti-miRNA-221, and anti-miRNA-335) that have been incorporated into scaffold-based bone tissue engineering applications and investigated in an in vivo bone critical-sized defect model. For all of the biomaterial scaffold-based miRNA therapies that have been developed thus far, cells that are transfected or transduced with the miRNA/anti-miRNA are loaded into the scaffolds and implanted at the site of interest instead of locally delivering the miRNA/anti-miRNAs directly from the scaffolds. Thus, future work may focus on developing biomaterial scaffolds to deliver miRNAs or anti-miRNAs into cells in vivo.
Collapse
Affiliation(s)
- Marco A Arriaga
- Department of Health and Biomedical Sciences, University of Texas Rio Grande Valley, One West University Blvd, Brownsville, TX, 78520, USA
| | - May-Hui Ding
- Department of Health and Biomedical Sciences, University of Texas Rio Grande Valley, One West University Blvd, Brownsville, TX, 78520, USA
| | - Astrid S Gutierrez
- Department of Health and Biomedical Sciences, University of Texas Rio Grande Valley, One West University Blvd, Brownsville, TX, 78520, USA
| | - Sue Anne Chew
- Department of Health and Biomedical Sciences, University of Texas Rio Grande Valley, One West University Blvd, Brownsville, TX, 78520, USA
| |
Collapse
|
37
|
Chen J, Hao P, Zheng T, Zhang Y. miR-628 reduces prostate cancer proliferation and invasion via the FGFR2 signaling pathway. Exp Ther Med 2019; 18:1005-1012. [PMID: 31316598 PMCID: PMC6601141 DOI: 10.3892/etm.2019.7682] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Accepted: 01/31/2019] [Indexed: 12/15/2022] Open
Abstract
Recently, microRNA (miR)-628 was identified as a potential biomarker for several types of cancer, including prostate cancer (PCa). The aim of the present study was to investigate miR-628 expression and its underlying mechanism in PCa cell proliferation and invasion and the fibroblast growth factor receptor 2 (FGFR2) signaling pathway. The serum expression levels of miR-628, prostate-specific antigen, fibroblast growth factor 1, and FGFR2 were examined in patients with PCa. The relative expression levels of miR-628 and FGFR2 were determined by reverse transcription-quantitative polymerase chain reaction in PCa cells following transfection with miR-628-5p mimic or inhibitor. In addition, the protein expression level of FGFR2 was examined by western blot analysis following transfection with miR-628-5p mimic or inhibitor. Following bioinformatics analysis, dual-luciferase reporter assay was used to confirm the direct interaction between miR-628 and FGFR2. The current study demonstrated that the protein expression level of FGFR2 decreased following transfection with miR-628-5p mimic and increased following transfection with miR-628-5p inhibitor. Similarly, the proliferation and invasion of PCa cells were significantly enhanced following transfection with miR-628-5p inhibitor. By contrast, the proliferation and invasion of PCa cells were significantly inhibited following transfection with miR-628 mimic. Therefore, downregulating the expression level of miR-628 may increase the expression level of FGF in PCa, thereby promoting tumor proliferation and invasion. In conclusion, the FGF signaling pathway may be involved in promoting PCa cell proliferation and invasion. miR-628 may be a potential therapeutic target for patients with PCa.
Collapse
Affiliation(s)
- Jun Chen
- Department of Urology, Wuhan Fourth Hospital, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei, Wuhan 430033, P.R. China
| | - Peng Hao
- Department of Urology, Wuhan Fourth Hospital, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei, Wuhan 430033, P.R. China
| | - Tao Zheng
- Department of Urology, Wuhan Fourth Hospital, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei, Wuhan 430033, P.R. China
| | - Yong Zhang
- Department of Urology, Wuhan Fourth Hospital, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei, Wuhan 430033, P.R. China
| |
Collapse
|
38
|
Delivery of miRNA-29b Using R9-LK15, a Novel Cell-Penetrating Peptide, Promotes Osteogenic Differentiation of Bone Mesenchymal Stem Cells. BIOMED RESEARCH INTERNATIONAL 2019; 2019:3032158. [PMID: 31111046 PMCID: PMC6487134 DOI: 10.1155/2019/3032158] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 03/31/2019] [Indexed: 02/06/2023]
Abstract
Delivery of osteogenesis-promoting microRNAs (miRNAs) is a promising approach to enhance bone regeneration. In this study, we generated nanocomplexes comprising the novel cell-penetrating peptide R9-LK15 and miR-29b and investigated their effects on osteogenic differentiation of bone mesenchymal stem cells (BMSCs). R9-LK15/miR-29b nanocomplexes were prepared and characterized. The transfection efficiency, cell viability, and osteogenic differentiation were investigated. The results showed that R9-LK15 maintained the stability of miR-29b in serum for up to 24 h. Moreover, R9-LK15 efficiently delivered miR-29b into BMSCs; the transfection efficiency was ~10-fold higher than that achieved using Lipofectamine 2000. The Cell Counting Kit-8 assay showed that R9-LK15 and R9-LK15/miR-29b nanocomplexes had negligible cytotoxic effects on BMSCs. Delivery of R9-LK15/miR-29b nanocomplexes promoted osteogenic differentiation of BMSCs and extracellular matrix mineralization by upregulating alkaline phosphatase expression and downregulating histone deacetylase-4 expression. In general, we developed a novel miRNA delivery system that has a high transfection efficiency and promotes osteogenic differentiation.
Collapse
|
39
|
Yang C, Yin M, Xu G, Lin W, Chen J, Zhang Y, Feng T, Huang P, Chen C, Yong K. Biodegradable Polymers as a Noncoding miRNA Nanocarrier for Multiple Targeting Therapy of Human Hepatocellular Carcinoma. Adv Healthc Mater 2019; 8:e1801318. [PMID: 30829008 DOI: 10.1002/adhm.201801318] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 01/26/2019] [Indexed: 12/21/2022]
Abstract
Therapeutic strategy based on the restoration of tumor suppressor-microRNAs (miRNAs) is a promising approach for cancer therapy, but the low delivery efficiency of miRNA remains a huge hurdle due to the lack of safe and efficient nonviral carriers. In this work, with the use of newly developed PEGylated biodegradable charged polyester-based vectors (PEG-BCPVs) as the carrier, the miR26a and miR122 codelivering therapeutic strategy (PEG-BCPVs/miR26a/miR122 as the delivery formulation) is successfully developed for efficient treatment of human hepatocellular carcinoma (HCC). In vitro study results show that PEG-BCPVs are capable of effectively facilitating miRNA cellular uptake via a cell endocytosis pathway. Consequently, the restoration of miR26a and miR122 remarkably inhibit the cell growth, migration, invasion, colony formation, and induced apoptosis of HepG2 cells. More importantly, the chemosensitivity of HepG2 to anticancer drug is also considerably enhanced. After treatment with the PEG-BCPV-based miRNA delivery system, the expression of the multiple targeted genes corresponding to miR26a and miR122 in HepG2 cells is greatly downregulated. Accordingly, the newly developed miRNA restoration therapeutic strategy via biodegradable PEG-BCPVs as the carrier should be a promising modality for combating HCC.
Collapse
Affiliation(s)
- Chengbin Yang
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound ImagingSchool of Biomedical EngineeringHealth Science CenterShenzhen University Shenzhen 518060 China
| | - Mingjie Yin
- School of Electrical and Electronic EngineeringNanyang Technological University Singapore 639798 Singapore
| | - Gaixia Xu
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound ImagingSchool of Biomedical EngineeringHealth Science CenterShenzhen University Shenzhen 518060 China
| | - Wei‐Jen Lin
- Department of Fiber and Composite MaterialsFeng Chia University Taichung 40724 Republic of China, Taiwan
| | - Jiajie Chen
- School of MedicineHealth Science CenterShenzhen University Shenzhen 518060 China
| | - Yinling Zhang
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound ImagingSchool of Biomedical EngineeringHealth Science CenterShenzhen University Shenzhen 518060 China
| | - Tao Feng
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound ImagingSchool of Biomedical EngineeringHealth Science CenterShenzhen University Shenzhen 518060 China
| | - Peng Huang
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound ImagingSchool of Biomedical EngineeringHealth Science CenterShenzhen University Shenzhen 518060 China
| | - Chih‐Kuang Chen
- Department of Chemical and Materials EngineeringNational Yunlin University of Science and Technology Yunlin 64002 Republic of China, Taiwan
| | - Ken‐Tye Yong
- School of Electrical and Electronic EngineeringNanyang Technological University Singapore 639798 Singapore
| |
Collapse
|
40
|
Loessner D, Goettig P, Preis S, Felber J, Bronger H, Clements JA, Dorn J, Magdolen V. Kallikrein-related peptidases represent attractive therapeutic targets for ovarian cancer. Expert Opin Ther Targets 2018; 22:745-763. [PMID: 30114962 DOI: 10.1080/14728222.2018.1512587] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Aberrant levels of kallikrein-related peptidases (KLK1-15) have been linked to cancer cell proliferation, invasion and metastasis. In ovarian cancer, the KLK proteolytic network has a crucial role in the tissue and tumor microenvironment. Publically available ovarian cancer genome and expression data from multiple patient cohorts show an upregulation of most KLKs. Areas covered: Here, we review the expression levels of all 15 members of this family in normal and ovarian cancer tissues, categorizing them into highly and moderately or weakly expressed KLKs, and their association with patient prognosis and survival. We summarize their tumor-biological functions determined in cell-based assays and xenograft models, further highlighting their suitability as cancer biomarkers and attractive candidates for drug development. Finally, we discuss some different pharmaceutical approaches, including peptide-based and small molecule inhibitors, cyclic peptides, depsipeptides, engineered natural inhibitors, antibodies, RNA/DNA-based aptamers, prodrugs, miRNA and siRNA. Expert opinion: In light of the results from clinical and tumor-biological studies, together with the available pharmaceutical tools, we suggest KLK4, KLK5, KLK6 and possibly KLK7 as preferred targets for inhibition in ovarian cancer.
Collapse
Affiliation(s)
- Daniela Loessner
- a Barts Cancer Institute , Queen Mary University of London , London , UK.,b Institute of Health and Biomedical Innovation , Queensland University of Technology (QUT) , Brisbane , Australia
| | - Peter Goettig
- c Department of Biosciences , University of Salzburg , Salzburg , Austria
| | - Sarah Preis
- d Department of Obstetrics and Gynecology , Technical University of Munich , Munich , Germany
| | - Johanna Felber
- d Department of Obstetrics and Gynecology , Technical University of Munich , Munich , Germany
| | - Holger Bronger
- d Department of Obstetrics and Gynecology , Technical University of Munich , Munich , Germany
| | - Judith A Clements
- b Institute of Health and Biomedical Innovation , Queensland University of Technology (QUT) , Brisbane , Australia.,e Australian Prostate Cancer Research Centre - Queensland , Queensland University of Technology (QUT), Translational Research Institute , Brisbane , Australia
| | - Julia Dorn
- d Department of Obstetrics and Gynecology , Technical University of Munich , Munich , Germany
| | - Viktor Magdolen
- d Department of Obstetrics and Gynecology , Technical University of Munich , Munich , Germany
| |
Collapse
|
41
|
Controlled Non-Viral Gene Delivery in Cartilage and Bone Repair: Current Strategies and Future Directions. ADVANCED THERAPEUTICS 2018. [DOI: 10.1002/adtp.201800038] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
42
|
Piotto C, Julier Z, Martino MM. Immune Regulation of Tissue Repair and Regeneration via miRNAs-New Therapeutic Target. Front Bioeng Biotechnol 2018; 6:98. [PMID: 30057898 PMCID: PMC6053520 DOI: 10.3389/fbioe.2018.00098] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 06/26/2018] [Indexed: 12/13/2022] Open
Abstract
The importance of immunity in tissue repair and regeneration is now evident. Thus, promoting tissue healing through immune modulation is a growing and promising field. Targeting microRNAs (miRNAs) is an appealing option since they regulate immunity through post-transcriptional gene fine-tuning in immune cells. Indeed, miRNAs are involved in inflammation as well as in its resolution by controlling immune cell phenotypes and functions. In this review, we first discuss the immunoregulatory role of miRNAs during the restoration of tissue homeostasis after injury, focusing mainly on neutrophils, macrophages and T lymphocytes. As tissue examples, we present the immunoregulatory function of miRNAs during the repair and regeneration of the heart, skeletal muscles, skin and liver. Secondly, we discuss recent technological advances for designing therapeutic strategies which target miRNAs. Specifically, we highlight the possible use of miRNAs and anti-miRNAs for promoting tissue regeneration via modulation of the immune system.
Collapse
Affiliation(s)
- Celeste Piotto
- European Molecular Biology Laboratory Australia, Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia
| | - Ziad Julier
- European Molecular Biology Laboratory Australia, Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia
| | - Mikaël M Martino
- European Molecular Biology Laboratory Australia, Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
43
|
Hernandez MJ, Gaetani R, Pieters VM, Ng NW, Chang AE, Martin TR, van Ingen E, Mol EA, Sluijter JPG, Christman KL. Decellularized Extracellular Matrix Hydrogels as a Delivery Platform for MicroRNA and Extracellular Vesicle Therapeutics. ADVANCED THERAPEUTICS 2018; 1. [PMID: 31544132 DOI: 10.1002/adtp.201800032] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
In the last decade, the use of microRNA (miRNA) and extracellular vesicle (EV) therapies has emerged as an alternative approach to mitigate the negative effects of several disease pathologies ranging from cancer to tissue and organ regeneration; however, delivery approaches towards target tissues have not been optimized. To alleviate these challenges, including rapid diffusion upon injection and susceptibility to degradation, porcine-derived decellularized extracellular matrix (ECM) hydrogels are examined as a potential delivery platform for miRNA and EV therapeutics. The incorporation of EVs and miRNA antagonists, including anti-miR and antago-miR, in ECM hydrogels results in a prolonged release as compared to the biologic agents alone. In addition, individual in vitro assessments confirm the bioactivity of the therapeutics upon release from the ECM hydrogels. This work demonstrates the feasibility of encapsulating miRNA and EV therapeutics in ECM hydrogels to enhance delivery and potentially efficacy in later in vivo applications.
Collapse
Affiliation(s)
- Melissa J Hernandez
- Department of Bioengineering, Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Roberto Gaetani
- Department of Bioengineering, Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Vera M Pieters
- Department of Bioengineering, Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Nathan W Ng
- Department of Bioengineering, Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Audrey E Chang
- Department of Bioengineering, Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Taylor R Martin
- Department of Bioengineering, Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Eva van Ingen
- Department of Bioengineering, Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Emma A Mol
- Department of Cardiology, Experimental Cardiology Laboratory, University Medical Center Utrecht, Utrecht, 3584CX, NL
| | - Joost P G Sluijter
- Department of Cardiology, Experimental Cardiology Laboratory, UMC Utrecht Regenerative Medicine Center, University Medical Center Utrecht, Utrecht, 3584CX, NL
| | - Karen L Christman
- Department of Bioengineering, Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
| |
Collapse
|
44
|
Mencía Castaño I, Curtin CM, Duffy GP, O'Brien FJ. Harnessing an Inhibitory Role of miR-16 in Osteogenesis by Human Mesenchymal Stem Cells for Advanced Scaffold-Based Bone Tissue Engineering. Tissue Eng Part A 2018; 25:24-33. [PMID: 29490603 DOI: 10.1089/ten.tea.2017.0460] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
MicroRNA (miRNA) therapeutics is increasingly being developed to either target bone-related diseases such as osteoporosis and osteoarthritis or as the basis for novel bone tissue engineering strategies. A number of miRNAs have been reported as potential osteo-therapeutics but no consensus has yet been established on the optimal target. miR-16 has been studied extensively in nonosteogenic functions and used as functionality reporter target in the development of nonviral miRNA delivery platforms. This study hypothesized that miR-16 may also play an inhibitory role in osteogenesis due to its ability to directly target Smad5 and AcvR2a. This study thus aimed to assess the potential of miR-16 inhibition to increase osteogenesis in human mesenchymal stem cells (hMSCs) using a previously established miRNA delivery platform composed of nanohydroxyapatite (nHA) particles as nonviral vectors in combination with collagen-nHA scaffolds designed specifically for bone repair. Initial results showed that antagomiR-16 delivery efficiently increased the relative levels of both putative targets and Runx2, the key transcription factor for osteogenesis, while also increasing osteocalcin levels. Furthermore, significant increases in mineral calcium deposition by hMSCs were found in both monolayer and most importantly in scaffold-based osteodifferentiation studies, ultimately demonstrating that miR-16 inhibition further enhances the therapeutic potential of a scaffold with known potential for bone repair applications and thus holds significant therapeutic potential as a novel bone tissue engineering strategy. Furthermore, we suggest that harnessing the additional functions known to miR-16 by incorporating either its enhancers or inhibitors to tissue-specific tailored scaffolds provides exciting opportunities for a diverse range of therapeutic indications.
Collapse
Affiliation(s)
- Irene Mencía Castaño
- 1 Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland, Dublin, Ireland.,2 Trinity Centre for Bioengineering, Trinity College Dublin, Dublin, Ireland.,3 Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, Ireland
| | - Caroline M Curtin
- 1 Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland, Dublin, Ireland.,2 Trinity Centre for Bioengineering, Trinity College Dublin, Dublin, Ireland.,3 Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, Ireland
| | - Garry P Duffy
- 1 Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland, Dublin, Ireland.,2 Trinity Centre for Bioengineering, Trinity College Dublin, Dublin, Ireland.,3 Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, Ireland.,4 Department of Anatomy, School of Medicine, College of Medicine Nursing and Health Sciences, National University of Ireland Galway, Ireland
| | - Fergal J O'Brien
- 1 Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland, Dublin, Ireland.,2 Trinity Centre for Bioengineering, Trinity College Dublin, Dublin, Ireland.,3 Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, Ireland
| |
Collapse
|
45
|
Coppin L, Leclerc J, Vincent A, Porchet N, Pigny P. Messenger RNA Life-Cycle in Cancer Cells: Emerging Role of Conventional and Non-Conventional RNA-Binding Proteins? Int J Mol Sci 2018; 19:ijms19030650. [PMID: 29495341 PMCID: PMC5877511 DOI: 10.3390/ijms19030650] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 02/16/2018] [Accepted: 02/19/2018] [Indexed: 02/06/2023] Open
Abstract
Functional specialization of cells and tissues in metazoans require specific gene expression patterns. Biological processes, thus, need precise temporal and spatial coordination of gene activity. Regulation of the fate of messenger RNA plays a crucial role in this context. In the present review, the current knowledge related to the role of RNA-binding proteins in the whole mRNA life-cycle is summarized. This field opens up a new angle for understanding the importance of the post-transcriptional control of gene expression in cancer cells. The emerging role of non-classic RNA-binding proteins is highlighted. The goal of this review is to encourage readers to view, through the mRNA life-cycle, novel aspects of the molecular basis of cancer and the potential to develop RNA-based therapies.
Collapse
Affiliation(s)
- Lucie Coppin
- University of Lille, UMR-S 1172-JPARC-Jean-Pierre Aubert Research Center, F-59000 Lille, France.
- Inserm, UMR-S 1172, Team "Mucins, Epithelial Differentiation and Carcinogenesis", F-59000 Lille, Frances.
- CHU Lille, Service de Biochimie "Hormonologie, Métabolisme-Nutrition, Oncologie", F-59000 Lille, France.
| | - Julie Leclerc
- University of Lille, UMR-S 1172-JPARC-Jean-Pierre Aubert Research Center, F-59000 Lille, France.
- Inserm, UMR-S 1172, Team "Mucins, Epithelial Differentiation and Carcinogenesis", F-59000 Lille, Frances.
- CHU Lille, Service de Biochimie "Hormonologie, Métabolisme-Nutrition, Oncologie", F-59000 Lille, France.
| | - Audrey Vincent
- University of Lille, UMR-S 1172-JPARC-Jean-Pierre Aubert Research Center, F-59000 Lille, France.
- Inserm, UMR-S 1172, Team "Mucins, Epithelial Differentiation and Carcinogenesis", F-59000 Lille, Frances.
- CHU Lille, Service de Biochimie "Hormonologie, Métabolisme-Nutrition, Oncologie", F-59000 Lille, France.
| | - Nicole Porchet
- University of Lille, UMR-S 1172-JPARC-Jean-Pierre Aubert Research Center, F-59000 Lille, France.
- Inserm, UMR-S 1172, Team "Mucins, Epithelial Differentiation and Carcinogenesis", F-59000 Lille, Frances.
- CHU Lille, Service de Biochimie "Hormonologie, Métabolisme-Nutrition, Oncologie", F-59000 Lille, France.
| | - Pascal Pigny
- University of Lille, UMR-S 1172-JPARC-Jean-Pierre Aubert Research Center, F-59000 Lille, France.
- Inserm, UMR-S 1172, Team "Mucins, Epithelial Differentiation and Carcinogenesis", F-59000 Lille, Frances.
- CHU Lille, Service de Biochimie "Hormonologie, Métabolisme-Nutrition, Oncologie", F-59000 Lille, France.
| |
Collapse
|