1
|
Yang Q, Lu D, Wu J, Liang F, Wang H, Yang J, Zhang G, Wang C, Yang Y, Zhu L, Sun X. Nanoparticles for the treatment of spinal cord injury. Neural Regen Res 2025; 20:1665-1680. [PMID: 39104097 PMCID: PMC11688544 DOI: 10.4103/nrr.nrr-d-23-01848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 03/06/2024] [Accepted: 04/09/2024] [Indexed: 08/07/2024] Open
Abstract
Spinal cord injuries lead to significant loss of motor, sensory, and autonomic functions, presenting major challenges in neural regeneration. Achieving effective therapeutic concentrations at injury sites has been a slow process, partly due to the difficulty of delivering drugs effectively. Nanoparticles, with their targeted delivery capabilities, biocompatibility, and enhanced bioavailability over conventional drugs, are garnering attention for spinal cord injury treatment. This review explores the current mechanisms and shortcomings of existing treatments, highlighting the benefits and progress of nanoparticle-based approaches. We detail nanoparticle delivery methods for spinal cord injury, including local and intravenous injections, oral delivery, and biomaterial-assisted implantation, alongside strategies such as drug loading and surface modification. The discussion extends to how nanoparticles aid in reducing oxidative stress, dampening inflammation, fostering neural regeneration, and promoting angiogenesis. We summarize the use of various types of nanoparticles for treating spinal cord injuries, including metallic, polymeric, protein-based, inorganic non-metallic, and lipid nanoparticles. We also discuss the challenges faced, such as biosafety, effectiveness in humans, precise dosage control, standardization of production and characterization, immune responses, and targeted delivery in vivo. Additionally, we explore future directions, such as improving biosafety, standardizing manufacturing and characterization processes, and advancing human trials. Nanoparticles have shown considerable progress in targeted delivery and enhancing treatment efficacy for spinal cord injuries, presenting significant potential for clinical use and drug development.
Collapse
Affiliation(s)
- Qiwei Yang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
| | - Di Lu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
| | - Jiuping Wu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Fuming Liang
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Huayi Wang
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Junjie Yang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Ganggang Zhang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Chen Wang
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yanlian Yang
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Ling Zhu
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xinzhi Sun
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| |
Collapse
|
2
|
Petillo E, Veneruso V, Gragnaniello G, Brochier L, Frigerio E, Perale G, Rossi F, Cardia A, Orro A, Veglianese P. Targeted therapy and deep learning insights into microglia modulation for spinal cord injury. Mater Today Bio 2024; 27:101117. [PMID: 38975239 PMCID: PMC11225820 DOI: 10.1016/j.mtbio.2024.101117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 05/31/2024] [Accepted: 06/08/2024] [Indexed: 07/09/2024] Open
Abstract
Spinal cord injury (SCI) is a devastating condition that can cause significant motor and sensory impairment. Microglia, the central nervous system's immune sentinels, are known to be promising therapeutic targets in both SCI and neurodegenerative diseases. The most effective way to deliver medications and control microglial inflammation is through nanovectors; however, because of the variability in microglial morphology and the lack of standardized techniques, it is still difficult to precisely measure their activation in preclinical models. This problem is especially important in SCI, where the intricacy of the glia response following traumatic events necessitates the use of a sophisticated method to automatically discern between various microglial cell activation states that vary over time and space as the secondary injury progresses. We address this issue by proposing a deep learning-based technique for quantifying microglial activation following drug-loaded nanovector treatment in a preclinical SCI model. Our method uses a convolutional neural network to segment and classify microglia based on morphological characteristics. Our approach's accuracy and efficiency are demonstrated through evaluation on a collection of histology pictures from injured and intact spinal cords. This robust computational technique has potential for analyzing microglial activation across various neuropathologies and demonstrating the usefulness of nanovectors in modifying microglia in SCI and other neurological disorders. It has the ability to speed development in this crucial sector by providing a standardized and objective way to compare therapeutic options.
Collapse
Affiliation(s)
- Emilia Petillo
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, via Mario Negri 2, Milano 20156, Italy
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, via Mancinelli 7, Milano 20131, Italy
| | - Valeria Veneruso
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, via Mario Negri 2, Milano 20156, Italy
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, via Buffi 13, Lugano 6900, Switzerland
| | - Gianluca Gragnaniello
- Department of Biomedical Sciences, Italian National Research Council, Institute of Biomedical Technologies, Segrate 20054, Italy
| | - Lorenzo Brochier
- Department of Biomedical Sciences, Italian National Research Council, Institute of Biomedical Technologies, Segrate 20054, Italy
| | - Enrico Frigerio
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, via Mario Negri 2, Milano 20156, Italy
| | - Giuseppe Perale
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, via Buffi 13, Lugano 6900, Switzerland
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Donaueschingenstrasse 13, 1200 Vienna, Austria
- Regenera GmbH, Modecenterstrasse 22/D01, 1030 Vienna, Austria
| | - Filippo Rossi
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, via Mancinelli 7, Milano 20131, Italy
| | - Andrea Cardia
- Department of Neurosurgery, Neurocenter of the Southern Switzerland, Regional Hospital of Lugano, Ente Ospedaliero Cantonale (EOC), Lugano, Switzerland
| | - Alessandro Orro
- Department of Biomedical Sciences, Italian National Research Council, Institute of Biomedical Technologies, Segrate 20054, Italy
| | - Pietro Veglianese
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, via Mario Negri 2, Milano 20156, Italy
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, via Buffi 13, Lugano 6900, Switzerland
| |
Collapse
|
3
|
Wan Y, Lin Y, Tan X, Gong L, Lei F, Wang C, Sun X, Du X, Zhang Z, Jiang J, Liu Z, Wang J, Zhou X, Wang S, Zhou X, Jing P, Zhong Z. Injectable Hydrogel To Deliver Bone Mesenchymal Stem Cells Preloaded with Azithromycin To Promote Spinal Cord Repair. ACS NANO 2024; 18:8934-8951. [PMID: 38483284 DOI: 10.1021/acsnano.3c12402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/27/2024]
Abstract
Spinal cord injury is a disease that causes severe damage to the central nervous system. Currently, there is no cure for spinal cord injury. Azithromycin is commonly used as an antibiotic, but it can also exert anti-inflammatory effects by down-regulating M1-type macrophage genes and up-regulating M2-type macrophage genes, which may make it effective for treating spinal cord injury. Bone mesenchymal stem cells possess tissue regenerative capabilities that may help promote the repair of the injured spinal cord. In this study, our objective was to explore the potential of promoting repair in the injured spinal cord by delivering bone mesenchymal stem cells that had internalized nanoparticles preloaded with azithromycin. To achieve this objective, we formulated azithromycin into nanoparticles along with a trans-activating transcriptional activator, which should enhance nanoparticle uptake by bone mesenchymal stem cells. These stem cells were then incorporated into an injectable hydrogel. The therapeutic effects of this formulation were analyzed in vitro using a mouse microglial cell line and a human neuroblastoma cell line, as well as in vivo using a rat model of spinal cord injury. The results showed that the formulation exhibited anti-inflammatory and neuroprotective effects in vitro as well as therapeutic effects in vivo. These results highlight the potential of a hydrogel containing bone mesenchymal stem cells preloaded with azithromycin and trans-activating transcriptional activator to mitigate spinal cord injury and promote tissue repair.
Collapse
Affiliation(s)
- Yujie Wan
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
- Ultrasound Medicine Department, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Yan Lin
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xie Tan
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Lingyi Gong
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Fei Lei
- Department of Spine Surgery, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Changguang Wang
- DataRevive USA, LLC, 30 W Gude Drive, Rockville, Maryland 20850, United States
| | - Xiaoduan Sun
- Department of Pharmacy, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xingjie Du
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Zhirong Zhang
- West China School of Pharmacy, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jun Jiang
- Department of Thyroid and Vascular Surgery, the Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Zhongbing Liu
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Jingxuan Wang
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xiaoling Zhou
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Shuzao Wang
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xiangyu Zhou
- Department of Thyroid and Vascular Surgery, the Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Pei Jing
- Department of Pharmacy, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Zhirong Zhong
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
- Central Nervous System Drug Key Laboratory of Sichuan Province, Luzhou, Sichuan 646000, China
| |
Collapse
|
4
|
Wang W, Yong J, Marciano P, O’Hare Doig R, Mao G, Clark J. The Translation of Nanomedicines in the Contexts of Spinal Cord Injury and Repair. Cells 2024; 13:569. [PMID: 38607008 PMCID: PMC11011097 DOI: 10.3390/cells13070569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Revised: 03/16/2024] [Accepted: 03/20/2024] [Indexed: 04/13/2024] Open
Abstract
PURPOSE OF THIS REVIEW Manipulating or re-engineering the damaged human spinal cord to achieve neuro-recovery is one of the foremost challenges of modern science. Addressing the restricted permission of neural cells and topographically organised neural tissue for self-renewal and spontaneous regeneration, respectively, is not straightforward, as exemplified by rare instances of translational success. This review assembles an understanding of advances in nanomedicine for spinal cord injury (SCI) and related clinical indications of relevance to attempts to design, engineer, and target nanotechnologies to multiple molecular networks. RECENT FINDINGS Recent research provides a new understanding of the health benefits and regulatory landscape of nanomedicines based on a background of advances in mRNA-based nanocarrier vaccines and quantum dot-based optical imaging. In relation to spinal cord pathology, the extant literature details promising advances in nanoneuropharmacology and regenerative medicine that inform the present understanding of the nanoparticle (NP) biocompatibility-neurotoxicity relationship. In this review, the conceptual bases of nanotechnology and nanomaterial chemistry covering organic and inorganic particles of sizes generally less than 100 nm in diameter will be addressed. Regarding the centrally active nanotechnologies selected for this review, attention is paid to NP physico-chemistry, functionalisation, delivery, biocompatibility, biodistribution, toxicology, and key molecular targets and biological effects intrinsic to and beyond the spinal cord parenchyma. SUMMARY The advance of nanotechnologies for the treatment of refractory spinal cord pathologies requires an in-depth understanding of neurobiological and topographical principles and a consideration of additional complexities involving the research's translational and regulatory landscapes.
Collapse
Affiliation(s)
- Wenqian Wang
- School of Chemical Engineering, University of New South Wales (UNSW), Kensington, NSW 2052, Australia; (W.W.); (J.Y.); (G.M.)
| | - Joel Yong
- School of Chemical Engineering, University of New South Wales (UNSW), Kensington, NSW 2052, Australia; (W.W.); (J.Y.); (G.M.)
| | - Paul Marciano
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia; (P.M.); (R.O.D.)
- Neil Sachse Centre for Spinal Cord Research, Lifelong Health Theme, South Australian Health and Medical Research Institute, North Terrace, Adelaide, SA 5000, Australia
| | - Ryan O’Hare Doig
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia; (P.M.); (R.O.D.)
- Neil Sachse Centre for Spinal Cord Research, Lifelong Health Theme, South Australian Health and Medical Research Institute, North Terrace, Adelaide, SA 5000, Australia
| | - Guangzhao Mao
- School of Chemical Engineering, University of New South Wales (UNSW), Kensington, NSW 2052, Australia; (W.W.); (J.Y.); (G.M.)
| | - Jillian Clark
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia; (P.M.); (R.O.D.)
- Neil Sachse Centre for Spinal Cord Research, Lifelong Health Theme, South Australian Health and Medical Research Institute, North Terrace, Adelaide, SA 5000, Australia
| |
Collapse
|
5
|
Yin P, Liang W, Han B, Yang Y, Sun D, Qu X, Hai Y, Luo D. Hydrogel and Nanomedicine-Based Multimodal Therapeutic Strategies for Spinal Cord Injury. SMALL METHODS 2024; 8:e2301173. [PMID: 37884459 DOI: 10.1002/smtd.202301173] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/13/2023] [Indexed: 10/28/2023]
Abstract
Spinal cord injury (SCI) is a severe neurodegenerative disease caused by mechanical and biological factors, manifesting as a loss of motor and sensory functions. Inhibition of injury expansion and even reversal of injury in the acute damage stage of SCI are important strategies for treating this disease. Hydrogels and nanoparticle (NP)-based drugs are the most effective, widely studied, and clinically valuable therapeutic strategies in the field of repair and regeneration. Hydrogels are 3D flow structures that fill the pathological gaps in SCI and provide a microenvironment similar to that of the spinal cord extracellular matrix for nerve cell regeneration. NP-based drugs can easily penetrate the blood-spinal cord barrier, target SCI lesions, and are noninvasive. Hydrogels and NPs as drug carriers can be loaded with various drugs and biological therapeutic factors for slow release in SCI lesions. They help drugs function more efficiently by exerting anti-inflammatory, antioxidant, and nerve regeneration effects to promote the recovery of neurological function. In this review, the use of hydrogels and NPs as drug carriers and the role of both in the repair of SCI are discussed to provide a multimodal strategic reference for nerve repair and regeneration after SCI.
Collapse
Affiliation(s)
- Peng Yin
- Department of Orthopedic Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 101400, China
- Joint Laboratory for Research & Treatment of Spinal Cord Injury in Spinal Deformity, Laboratory for Clinical Medicine, Capital Medical University, Beijing, 100069, China
- Clinical Center for Spinal Deformity, Capital Medical University, Beijing, 100069, China
| | - Weishi Liang
- Department of Orthopedic Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 101400, China
- Joint Laboratory for Research & Treatment of Spinal Cord Injury in Spinal Deformity, Laboratory for Clinical Medicine, Capital Medical University, Beijing, 100069, China
- Clinical Center for Spinal Deformity, Capital Medical University, Beijing, 100069, China
| | - Bo Han
- Department of Orthopedic Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 101400, China
- Joint Laboratory for Research & Treatment of Spinal Cord Injury in Spinal Deformity, Laboratory for Clinical Medicine, Capital Medical University, Beijing, 100069, China
- Clinical Center for Spinal Deformity, Capital Medical University, Beijing, 100069, China
| | - Yihan Yang
- Department of Orthopedic Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
- Joint Laboratory for Research & Treatment of Spinal Cord Injury in Spinal Deformity, Laboratory for Clinical Medicine, Capital Medical University, Beijing, 100069, China
- Clinical Center for Spinal Deformity, Capital Medical University, Beijing, 100069, China
| | - Duan Sun
- Department of Orthopedic Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
- Joint Laboratory for Research & Treatment of Spinal Cord Injury in Spinal Deformity, Laboratory for Clinical Medicine, Capital Medical University, Beijing, 100069, China
- Clinical Center for Spinal Deformity, Capital Medical University, Beijing, 100069, China
| | - Xianjun Qu
- Joint Laboratory for Research & Treatment of Spinal Cord Injury in Spinal Deformity, Laboratory for Clinical Medicine, Capital Medical University, Beijing, 100069, China
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Yong Hai
- Department of Orthopedic Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
- Joint Laboratory for Research & Treatment of Spinal Cord Injury in Spinal Deformity, Laboratory for Clinical Medicine, Capital Medical University, Beijing, 100069, China
- Clinical Center for Spinal Deformity, Capital Medical University, Beijing, 100069, China
| | - Dan Luo
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 101400, China
| |
Collapse
|
6
|
Rahmanian M, Ghahremani A, Kesharwani P, Oroojalian F, Sahebkar A. Nanomedicine innovations in spinal cord injury management: Bridging the gap. ENVIRONMENTAL RESEARCH 2023; 235:116563. [PMID: 37423366 DOI: 10.1016/j.envres.2023.116563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 06/24/2023] [Accepted: 07/04/2023] [Indexed: 07/11/2023]
Abstract
Spinal cord injury (SCI) has devastating effects on a person's physical, social, and professional well-being. It is a life-altering neurological condition that significantly impacts individuals and their caregivers on a socioeconomic level. Recent advancements in medical therapy have greatly improved the diagnosis, stability, survival rates, and overall well-being of SCI patients. However, there are still limited options available for enhancing neurological outcomes in these patients. The complex pathophysiology of SCI, along with the numerous biochemical and physiological changes that occur in the damaged spinal cord, contribute to this gradual improvement. Currently, there are no therapies that offer the possibility of recovery for SCI, although several therapeutic approaches are being developed. However, these therapies are still in the early stages and have not yet demonstrated effectiveness in repairing the damaged fibers, which hinders cellular regeneration and the full restoration of motor and sensory functions. Considering the importance of nanotechnology and tissue engineering in treating neural tissue injuries, this review focuses on the latest advancements in nanotechnology for SCI therapy and tissue healing. It examines research articles from the PubMed database that specifically address SCI in the field of tissue engineering, with an emphasis on nanotechnology as a therapeutic approach. The review evaluates the biomaterials used for treating this condition and the techniques employed to create nanostructured biomaterials.
Collapse
Affiliation(s)
- Mohsen Rahmanian
- School of Medicine, North Khorasan University of Medical Sciences, Bojnord, Iran
| | - Amirali Ghahremani
- Department of Neurology, North Khorasan University of Medical Sciences, Bojnord, Iran
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India; Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India.
| | - Fatemeh Oroojalian
- Department of Advanced Technologies, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran; Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran.
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
7
|
Gao J, Khang MK, Liao Z, Webb K, Detloff MR, Lee JS. Rolipram-loaded PgP nanoparticle reduces secondary injury and enhances motor function recovery in a rat moderate contusion SCI model. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2023; 53:102702. [PMID: 37574117 DOI: 10.1016/j.nano.2023.102702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 07/30/2023] [Accepted: 08/02/2023] [Indexed: 08/15/2023]
Abstract
Spinal cord injury (SCI) results in immediate axonal damage and cell death, as well as a prolonged secondary injury consist of a cascade of pathophysiological processes. One important aspect of secondary injury is activation of phosphodiesterase 4 (PDE4) that leads to reduce cAMP levels in the injured spinal cord. We have developed an amphiphilic copolymer, poly (lactide-co-glycolide)-graft-polyethylenimine (PgP) that can deliver Rolipram, the PDE4 inhibitor. The objective of this work was to investigate the effect of rolipram loaded PgP (Rm-PgP) on secondary injury and motor functional recovery in a rat moderate contusion SCI model. We observed that Rm-PgP can increase cAMP level at the lesion site, and reduce secondary injury such as the inflammatory response by macrophages/microglia, astrogliosis by activated astrocytes and apoptosis as well as improve neuronal survival at 4 weeks post-injury (WPI). We also observed that Rm-PgP can improve motor functional recovery after SCI over 4 WPI.
Collapse
Affiliation(s)
- Jun Gao
- Drug Design Delivery and Development (4D) Laboratory, Department of Bioengineering, Clemson University, Clemson, SC, USA
| | - Min Kyung Khang
- Drug Design Delivery and Development (4D) Laboratory, Department of Bioengineering, Clemson University, Clemson, SC, USA
| | - Zhen Liao
- Drug Design Delivery and Development (4D) Laboratory, Department of Bioengineering, Clemson University, Clemson, SC, USA.
| | - Ken Webb
- MicroEnvironmental Laboratory, Department of Bioengineering, Clemson University, Clemson, SC, USA.
| | - Megan Ryan Detloff
- Department of Neurobiology & Anatomy, Marion Murray Spinal Cord Research Center, College of Medicine, Drexel University, Philadelphia, PA 19129, USA.
| | - Jeoung Soo Lee
- Drug Design Delivery and Development (4D) Laboratory, Department of Bioengineering, Clemson University, Clemson, SC, USA.
| |
Collapse
|
8
|
NPC transplantation rescues sci-driven cAMP/EPAC2 alterations, leading to neuroprotection and microglial modulation. Cell Mol Life Sci 2022; 79:455. [PMID: 35904607 PMCID: PMC9338125 DOI: 10.1007/s00018-022-04494-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 07/07/2022] [Accepted: 07/17/2022] [Indexed: 11/17/2022]
Abstract
Neural progenitor cell (NPC) transplantation represents a promising treatment strategy for spinal cord injury (SCI); however, the underlying therapeutic mechanisms remain incompletely understood. We demonstrate that severe spinal contusion in adult rats causes transcriptional dysregulation, which persists from early subacute to chronic stages of SCI and affects nearly 20,000 genes in total tissue extracts. Functional analysis of this dysregulated transcriptome reveals the significant downregulation of cAMP signalling components immediately after SCI, involving genes such as EPAC2 (exchange protein directly activated by cAMP), PKA, BDNF, and CAMKK2. The ectopic transplantation of spinal cord-derived NPCs at acute or subacute stages of SCI induces a significant transcriptional impact in spinal tissue, as evidenced by the normalized expression of a large proportion of SCI-affected genes. The transcriptional modulation pattern driven by NPC transplantation includes the rescued expression of cAMP signalling genes, including EPAC2. We also explore how the sustained in vivo inhibition of EPAC2 downstream signalling via the intrathecal administration of ESI-05 for 1 week impacts therapeutic mechanisms involved in the NPC-mediated treatment of SCI. NPC transplantation in SCI rats in the presence and absence of ESI-05 administration prompts increased rostral cAMP levels; however, NPC and ESI-05 treated animals exhibit a significant reduction in EPAC2 mRNA levels compared to animals receiving only NPCs treatment. Compared with transplanted animals, NPCs + ESI-05 treatment increases the scar area (as shown by GFAP staining), polarizes microglia into an inflammatory phenotype, and increases the magnitude of the gap between NeuN + cells across the lesion. Overall, our results indicate that the NPC-associated therapeutic mechanisms in the context of SCI involve the cAMP pathway, which reduces inflammation and provides a more neuropermissive environment through an EPAC2-dependent mechanism.
Collapse
|
9
|
Zarepour A, Bal Öztürk A, Koyuncu Irmak D, Yaşayan G, Gökmen A, Karaöz E, Zarepour A, Zarrabi A, Mostafavi E. Combination Therapy Using Nanomaterials and Stem Cells to Treat Spinal Cord Injuries. Eur J Pharm Biopharm 2022; 177:224-240. [PMID: 35850168 DOI: 10.1016/j.ejpb.2022.07.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/29/2022] [Accepted: 07/08/2022] [Indexed: 02/07/2023]
Abstract
As a part of the central nervous system, the spinal cord (SC) provides most of the communications between the brain and other parts of the body. Any damage to SC interrupts this communication, leading to serious problems, which may remain for the rest of their life. Due to its significant impact on patients' quality of life and its exorbitant medical costs, SC injury (SCI) is known as one of the most challengeable diseases in the world. Thus, it is critical to introduce highly translatable therapeutic platforms for SCI treatment. So far, different strategies have been introduced, among which utilizing various types of stem cells is one of the most interesting ones. The capability of stem cells to differentiate into several types of cell lines makes them promising candidates for the regeneration of injured tissues. One of the other interesting and novel strategies for SCI treatment is the application of nanomaterials, which could appear as a carrier for therapeutic agents or as a platform for culturing the cells. Combining these two approaches, stem cells and nanomaterials, could provide promising therapeutic strategies for SCI management. Accordingly, in this review we have summarized some of the recent advancements in which the applications of different types of stem cells and nanomaterials, alone and in combination forms, were evaluated for SCI treatment.
Collapse
Affiliation(s)
- Arezou Zarepour
- Radiology Department, Kashan University of Medical Sciences, Kashan, Isfahan, Iran
| | - Ayça Bal Öztürk
- Department of Stem Cell and Tissue Engineering, Institute of Health Sciences, Istinye University, Istanbul, Turkey; Department of Analytical Chemistry, Faculty of Pharmacy, Istinye University, Zeytinburnu, Turkey
| | | | - Gökçen Yaşayan
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Yeditepe University, Istanbul, Turkey
| | - Aylin Gökmen
- Molecular Biology and Genetics Department, Faculty of Engineering and Natural Sciences, Bahcesehir University, Besiktas, Istanbul, Turkey
| | - Erdal Karaöz
- Liv Hospital, Center for Regenerative Medicine and Stem Cell Manufacturing (LivMedCell), İstanbul, Turkey
| | - Atefeh Zarepour
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Sariyer, Istanbul 34396, Turkey
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Sariyer, Istanbul 34396, Turkey.
| | - Ebrahim Mostafavi
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
10
|
Applications of Various Types of Nanomaterials for the Treatment of Neurological Disorders. NANOMATERIALS 2022; 12:nano12132140. [PMID: 35807977 PMCID: PMC9268720 DOI: 10.3390/nano12132140] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 06/08/2022] [Accepted: 06/19/2022] [Indexed: 02/07/2023]
Abstract
Neurological disorders (NDs) are recognized as one of the major health concerns globally. According to the World Health Organization (WHO), neurological disorders are one of the main causes of mortality worldwide. Neurological disorders include Alzheimer’s disease, Parkinson′s disease, Huntington′s disease, Amyotrophic lateral sclerosis, Frontotemporal dementia, Prion disease, Brain tumor, Spinal cord injury, and Stroke. These diseases are considered incurable diseases because no specific therapies are available to cross the blood-brain barrier (BBB) and reach the brain in a significant amount for the pharmacological effect in the brain. There is a need for the development of strategies that can improve the efficacy of drugs and circumvent BBB. One of the promising approaches is the use of different types of nano-scale materials. These nano-based drugs have the ability to increase the therapeutic effect, reduce toxicity, exhibit good stability, targeted delivery, and drug loading capacity. Different types and shapes of nanomaterials have been widely used for the treatment of neurological disorders, including quantum dots, dendrimers, metallic nanoparticles, polymeric nanoparticles, carbon nanotubes, liposomes, and micelles. These nanoparticles have unique characteristics, including sensitivity, selectivity, and the ability to cross the BBB when used in nano-sized particles, and are widely used for imaging studies and treatment of NDs. In this review, we briefly summarized the recent literature on the use of various nanomaterials and their mechanism of action for the treatment of various types of neurological disorders.
Collapse
|
11
|
Future Treatment of Neuropathic Pain in Spinal Cord Injury: The Challenges of Nanomedicine, Supplements or Opportunities? Biomedicines 2022; 10:biomedicines10061373. [PMID: 35740395 PMCID: PMC9219608 DOI: 10.3390/biomedicines10061373] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 05/28/2022] [Accepted: 06/08/2022] [Indexed: 12/12/2022] Open
Abstract
Neuropathic pain (NP) is a common chronic condition that severely affects patients with spinal cord injuries (SCI). It impairs the overall quality of life and is considered difficult to treat. Currently, clinical management of NP is often limited to drug therapy, primarily with opioid analgesics that have limited therapeutic efficacy. The persistence and intractability of NP following SCI and the potential health risks associated with opioids necessitate improved treatment approaches. Nanomedicine has gained increasing attention in recent years for its potential to improve therapeutic efficacy while minimizing toxicity by providing sensitive and targeted treatments that overcome the limitations of conventional pain medications. The current perspective begins with a brief discussion of the pathophysiological mechanisms underlying NP and the current pain treatment for SCI. We discuss the most frequently used nanomaterials in pain diagnosis and treatment as well as recent and ongoing efforts to effectively treat pain by proactively mediating pain signals following SCI. Although nanomedicine is a rapidly growing field, its application to NP in SCI is still limited. Therefore, additional work is required to improve the current treatment of NP following SCI.
Collapse
|
12
|
Zhou G, Wang Z, Han S, Chen X, Li Z, Hu X, Li Y, Gao J. Multifaceted Roles of cAMP Signaling in the Repair Process of Spinal Cord Injury and Related Combination Treatments. Front Mol Neurosci 2022; 15:808510. [PMID: 35283731 PMCID: PMC8904388 DOI: 10.3389/fnmol.2022.808510] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 01/26/2022] [Indexed: 01/03/2023] Open
Abstract
Spinal cord injury (SCI) results in multiple pathophysiological processes, including blood–spinal cord barrier disruption, hemorrhage/ischemia, oxidative stress, neuroinflammation, scar formation, and demyelination. These responses eventually lead to severe tissue destruction and an inhibitory environment for neural regeneration.cAMP signaling is vital for neurite outgrowth and axonal guidance. Stimulating intracellular cAMP activity significantly promotes neuronal survival and axonal regrowth after SCI.However, neuronal cAMP levels in adult CNS are relatively low and will further decrease after injury. Targeting cAMP signaling has become a promising strategy for neural regeneration over the past two decades. Furthermore, studies have revealed that cAMP signaling is involved in the regulation of glial cell function in the microenvironment of SCI, including macrophages/microglia, reactive astrocytes, and oligodendrocytes. cAMP-elevating agents in the post-injury milieu increase the cAMP levels in both neurons and glial cells and facilitate injury repair through the interplay between neurons and glial cells and ultimately contribute to better morphological and functional outcomes. In recent years, combination treatments associated with cAMP signaling have been shown to exert synergistic effects on the recovery of SCI. Agents carried by nanoparticles exhibit increased water solubility and capacity to cross the blood–spinal cord barrier. Implanted bioscaffolds and injected hydrogels are potential carriers to release agents locally to avoid systemic side effects. Cell transplantation may provide permissive matrices to synergize with the cAMP-enhanced growth capacity of neurons. cAMP can also induce the oriented differentiation of transplanted neural stem/progenitor cells into neurons and increase the survival rate of cell grafts. Emerging progress focused on cAMP compartmentation provides researchers with new perspectives to understand the complexity of downstream signaling, which may facilitate the clinical translation of strategies targeting cAMP signaling for SCI repair.
Collapse
Affiliation(s)
- Gang Zhou
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Zhiyan Wang
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Shiyuan Han
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xiaokun Chen
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Zhimin Li
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xianghui Hu
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yongning Li
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Department of International Medical Service, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jun Gao
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- *Correspondence: Jun Gao
| |
Collapse
|
13
|
Shabani Z, Rahbarghazi R, Karimipour M, Ghadiri T, Salehi R, Sadigh‐Eteghad S, Farhoudi M. Transplantation of bioengineered Reelin-loaded PLGA/PEG micelles can accelerate neural tissue regeneration in photothrombotic stroke model of mouse. Bioeng Transl Med 2021; 7:e10264. [PMID: 35111956 PMCID: PMC8780906 DOI: 10.1002/btm2.10264] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 09/20/2021] [Accepted: 10/16/2021] [Indexed: 01/09/2023] Open
Abstract
Ischemic stroke is characterized by extensive neuronal loss, glial scar formation, neural tissue degeneration that leading to profound changes in the extracellular matrix, neuronal circuitry, and long-lasting functional disabilities. Although transplanted neural stem cells (NSCs) can recover some of the functional deficit after stroke, retrieval is not complete and repair of lost tissue is negligible. Therefore, the current challenge is to use the combination of NSCs with suitably enriched biomaterials to retain these cells within the infarct cavity and accelerate the formation of a de novo tissue. This study aimed to test the regenerative potential of polylactic-co-glycolic acid-polyethylene glycol (PLGA-PEG) micelle biomaterial enriched with Reelin and embryonic NSCs on photothrombotic stroke model of mice to gain appropriate methods in tissue engineering. For this purpose, two sets of experiments, either in vitro or in vivo models, were performed. In vitro analyses exhibited PLGA-PEG plus Reelin-induced proliferation rate (Ki-67+ NSCs) and neurite outgrowth (axonization and dendritization) compared to PLGA-PEG + NSCs and Reelin + NSCs groups (p < 0.05). Besides, neural differentiation (Map-2+ cells) was high in NSCs cultured in the presence of Reelin-loaded PLGA-PEG micelles (p < 0.05). Double immunofluorescence staining showed that Reelin-loaded PLGA-PEG micelles increased the number of migrating neural progenitor cells (DCX+ cells) and mature neurons (NeuN+ cells) around the lesion site compared to the groups received PLGA-PEG and Reelin alone after 1 month (p < 0.05). Immunohistochemistry results showed that the PLGA/PEG plus Reelin significantly decreased the astrocytic gliosis and increased local angiogenesis (vWF-positive cells) relative to the other groups. These changes led to the reduction of cavity size in the Reelin-loaded PLGA-PEG+NSCs group. Neurobehavioral tests indicated Reelin-loaded PLGA-PEG+NSCs promoted neurological outcome and functional recovery (p < 0.05). These results indicated that Reelin-loaded PLGA-PEG is capable of promoting NSCs dynamic growth, neuronal differentiation, and local angiogenesis following ischemic injury via providing a desirable microenvironment. These features can lead to neural tissue regeneration and functional recovery.
Collapse
Affiliation(s)
- Zahra Shabani
- Neurosciences Research Center (NSRC)Tabriz University of Medical SciencesTabrizIran,Department of Neurosciences, Faculty of Advanced Medical SciencesTabriz University of Medical SciencesTabrizIran
| | - Reza Rahbarghazi
- Stem Cell Research CenterTabriz University of Medical SciencesTabrizIran,Department of Applied Cell Sciences, Faculty of Advanced Medical SciencesTabriz University of Medical SciencesTabrizIran
| | - Mohammad Karimipour
- Department of Neurosciences, Faculty of Advanced Medical SciencesTabriz University of Medical SciencesTabrizIran,Department of Anatomical Sciences, Faculty of MedicineTabriz University of Medical SciencesTabrizIran
| | - Tahereh Ghadiri
- Department of Neurosciences, Faculty of Advanced Medical SciencesTabriz University of Medical SciencesTabrizIran
| | - Roya Salehi
- Department of Medical Nanotechnology, Faculty of Advanced Medical SciencesTabriz University of Medical SciencesTabrizIran
| | - Saeed Sadigh‐Eteghad
- Neurosciences Research Center (NSRC)Tabriz University of Medical SciencesTabrizIran
| | - Mehdi Farhoudi
- Neurosciences Research Center (NSRC)Tabriz University of Medical SciencesTabrizIran
| |
Collapse
|
14
|
Gao J, Khang M, Liao Z, Detloff M, Lee JS. Therapeutic targets and nanomaterial-based therapies for mitigation of secondary injury after spinal cord injury. Nanomedicine (Lond) 2021; 16:2013-2028. [PMID: 34402308 PMCID: PMC8411395 DOI: 10.2217/nnm-2021-0113] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 06/29/2021] [Indexed: 12/31/2022] Open
Abstract
Spinal cord injury (SCI) and the resulting neurological trauma commonly result in complete or incomplete neurological dysfunction and there are few effective treatments for primary SCI. However, the following secondary SCI, including the changes of microvasculature, inflammatory response and oxidative stress around the injury site, may provide promising therapeutic targets. The advances of nanomaterials hold promise for delivering therapeutics to alleviate secondary SCI and promote functional recovery. In this review, we highlight recent achievements of nanomaterial-based therapy, specifically targeting blood-spinal cord barrier disruption, mitigation of the inflammatory response and lightening of oxidative stress after spinal cord injury.
Collapse
Affiliation(s)
- Jun Gao
- Department of Bioengineering, Drug Design, Development & Delivery (4D) Laboratory, Clemson University, Clemson, SC 29634, USA
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| | - Minkyung Khang
- Department of Bioengineering, Drug Design, Development & Delivery (4D) Laboratory, Clemson University, Clemson, SC 29634, USA
| | - Zhen Liao
- Department of Bioengineering, Drug Design, Development & Delivery (4D) Laboratory, Clemson University, Clemson, SC 29634, USA
| | - Megan Detloff
- Department of Neurobiology & Anatomy, Drexel University, Philadelphia, PA 19129, USA
| | - Jeoung Soo Lee
- Department of Bioengineering, Drug Design, Development & Delivery (4D) Laboratory, Clemson University, Clemson, SC 29634, USA
| |
Collapse
|
15
|
Mousa AH, Agha Mohammad S, Rezk HM, Muzaffar KH, Alshanberi AM, Ansari SA. Nanoparticles in traumatic spinal cord injury: therapy and diagnosis. F1000Res 2021. [DOI: 10.12688/f1000research.55472.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Nanotechnology has been previously employed for constructing drug delivery vehicles, biosensors, solar cells, lubricants and as antimicrobial agents. The advancement in synthesis procedure makes it possible to formulate nanoparticles (NPs) with precise control over physico-chemical and optical properties that are desired for specific clinical or biological applications. The surface modification technology has further added impetus to the specific applications of NPs by providing them with desirable characteristics. Hence, nanotechnology is of paramount importance in numerous biomedical and industrial applications due to their biocompatibility and stability even in harsh environments. Traumatic spinal cord injuries (TSCIs) are one of the major traumatic injuries that are commonly associated with severe consequences to the patient that may reach to the point of paralysis. Several processes occurring at a biochemical level which exacerbate the injury may be targeted using nanotechnology. This review discusses possible nanotechnology-based approaches for the diagnosis and therapy of TSCI, which have a bright future in clinical practice.
Collapse
|
16
|
Maher A, El Sayed N, Nafea H, Gad M. Rolipram rescues memory consolidation deficits caused by sleep deprivation: Implication of the cAMP/PKA and cAMP/Epac pathways. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2021; 21:631-639. [PMID: 34397335 DOI: 10.2174/1871527320666210816105144] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 07/16/2021] [Accepted: 07/20/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Over the last few years, the number of people suffering from sleeping disorders has increased significantly despite negative effects on cognition and an association with brain inflammation. OBJECTIVES We assessed memory deficits caused by sleep deprivation (SD) to determine the therapeutic effect of phosphodiesterase 4 (PDE4) inhibitors on SD-induced memory deficits and to investigate whether the modulation of memory deficits by PDE4 inhibitors is mediated by a protein kinase A (PKA)-independent pathway in conjunction with a PKA-dependent pathway. METHODS Adult male mice were divided into four groups. Three SD groups were deprived of Rapid eye movement (REM) sleep for 12 h a day for six consecutive days. They were tested daily in the Morris water maze to evaluate learning and memory. One of the SD groups was injected with a PDE4 inhibitor, rolipram (1 mg/kg ip), whereas another had rolipram co-administered with chlorogenic acid (CHA, 20 mg/kg ip), an inhibitor of PKA. After 6 days, the mice were sacrificed, and the hippocampi were evaluated for cyclic AMP (cAMP) and nuclear factor Nrf-2 levels. The hippocampal expression of PKA, phosphorylated cAMP response element-binding protein (CREB), and phosphorylated glycogen synthase 3β (Ser389) were also evaluated. RESULTS SD caused a significant decrease in cAMP levels in the brain and had a detrimental effect on learning and memory. The administration of rolipram or rolipram+CHA resulted in an improvement in cognitive function. CONCLUSION The present study provides evidence that restoration of memory with PDE4 inhibitors occurs through a dual mechanism involving the PKA and Epac pathways.
Collapse
Affiliation(s)
- Ahmed Maher
- Biochemistry Department, Faculty of Pharmacy, October University for Modern Sciences and Arts (MSA), Cairo. Egypt
| | - Nesrine El Sayed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University. Egypt
| | - Heba Nafea
- Biochemistry Department, Faculty of Pharmacy and Biotechnology, German University in Cairo (GUC), Cairo. Egypt
| | - Mohamed Gad
- Biochemistry Department, Faculty of Pharmacy and Biotechnology, German University in Cairo (GUC), Cairo. Egypt
| |
Collapse
|
17
|
Mbugua SN, Njenga LW, Odhiambo RA, Wandiga SO, Onani MO. Beyond DNA-targeting in Cancer Chemotherapy. Emerging Frontiers - A Review. Curr Top Med Chem 2021; 21:28-47. [PMID: 32814532 DOI: 10.2174/1568026620666200819160213] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 07/07/2020] [Accepted: 07/08/2020] [Indexed: 12/14/2022]
Abstract
Modern anti-cancer drugs target DNA specifically for rapid division of malignant cells. One downside of this approach is that they also target other rapidly dividing healthy cells, such as those involved in hair growth leading to serious toxic side effects and hair loss. Therefore, it would be better to develop novel agents that address cellular signaling mechanisms unique to cancerous cells, and new research is now focussing on such approaches. Although the classical chemotherapy area involving DNA as the set target continues to produce important findings, nevertheless, a distinctly discernible emerging trend is the divergence from the cisplatin operation model that uses the metal as the primary active center of the drug. Many successful anti-cancer drugs present are associated with elevated toxicity levels. Cancers also develop immunity against most therapies and the area of cancer research can, therefore, be seen as an area with a high unaddressed need. Hence, ongoing work into cancer pathogenesis is important to create accurate preclinical tests that can contribute to the development of innovative drugs to manage and treat cancer. Some of the emergent frontiers utilizing different approaches include nanoparticles delivery, use of quantum dots, metal complexes, tumor ablation, magnetic hypothermia and hyperthermia by use of Superparamagnetic Iron oxide Nanostructures, pathomics and radiomics, laser surgery and exosomes. This review summarizes these new approaches in good detail, giving critical views with necessary comparisons. It also delves into what they carry for the future, including their advantages and disadvantages.
Collapse
Affiliation(s)
- Simon N Mbugua
- Department of Chemistry, University of Nairobi, P.O. Box 30197-00100, Nairobi, Kenya
| | - Lydia W Njenga
- Department of Chemistry, University of Nairobi, P.O. Box 30197-00100, Nairobi, Kenya
| | - Ruth A Odhiambo
- Department of Chemistry, University of Nairobi, P.O. Box 30197-00100, Nairobi, Kenya
| | - Shem O Wandiga
- Department of Chemistry, University of Nairobi, P.O. Box 30197-00100, Nairobi, Kenya
| | - Martin O Onani
- Organometallics and Nanomaterials, Department of Chemistry, University of the Western Cape, Private Bag X17, Bellville, 7535, South Africa
| |
Collapse
|
18
|
Cao Y, Jiang C, Lin H, Chen Z. Silencing of Long Noncoding RNA Growth Arrest-Specific 5 Alleviates Neuronal Cell Apoptosis and Inflammatory Responses Through Sponging microRNA-93 to Repress PTEN Expression in Spinal Cord Injury. Front Cell Neurosci 2021; 15:646788. [PMID: 34054430 PMCID: PMC8163226 DOI: 10.3389/fncel.2021.646788] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Accepted: 04/20/2021] [Indexed: 02/01/2023] Open
Abstract
A secondary injury induced by a spinal cord injury (SCI) remains the main cause of devastating neural dysfunction; therefore, it has been the subject of focused research for many years. Long noncoding RNA (lncRNA) has been found to participate in the SCI process, and this finding presents a high potential for diagnosis and treatment; however, the role of lncRNA in a secondary injury induced by SCI remains unclear. The aim of this study was to investigate the regulatory effect of lncRNA growth arrest–specific transcript 5 (GAS5) in secondary injury during SCI. The SCI mice model and hypoxic cellular model were established to research the roles of lncRNA GAS5 during SCI. Reverse transcription quantitative polymerase chain reaction (qRT-PCR) was conducted to determine the expression levels of microR-93 (miR-93) and lncRNA GAS5. Western blot analysis of the apoptosis regulator protein and terminal deoxynucleotidyl transferase dUTP nick end labeling assay was conducted to evaluate neuron cell apoptosis. Basso, Beattie, and Bresnahan (BBB) scores were calculated to assess neurological function. Flow cytometry was used to determine neuron cell apoptosis. The associations among GAS5, miR-93, and the phosphatase and tensin homolog (PTEN) were disclosed using RNA immunoprecipitation (RIP) assay, RNA pulldown assay, and dual-luciferase reporter assay. QRT-PCR demonstrated that GAS5 was significantly upregulated in both the SCI mice and hypoxic cellular models. GAS5 knockdown suppressed neuron cell apoptosis and inflammatory response in the SCI mice model. Further studies have indicated that GAS5 functions as a competing endogenous RNA (ceRNA) by sponging miR-93 in neuronal cells. In addition, PTEN was a target of miR-93, and GAS5 knockdown exhibited its anti-apoptotic and anti-inflammatory effects through the miR-93/PTEN axis. These findings suggest that the GAS5/miR-93/PTEN axis may be a promising therapeutic target for SCI.
Collapse
Affiliation(s)
- Yuanwu Cao
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chang Jiang
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Haodong Lin
- Department of Orthopedic Surgery, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zixian Chen
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
19
|
Zimmermann R, Vieira Alves Y, Sperling LE, Pranke P. Nanotechnology for the Treatment of Spinal Cord Injury. TISSUE ENGINEERING PART B-REVIEWS 2020; 27:353-365. [PMID: 33135599 DOI: 10.1089/ten.teb.2020.0188] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Spinal cord injury (SCI) affects the central nervous system (CNS) and there is currently no treatment with the potential for rehabilitation. Although several clinical treatments have been developed, they are still at an early stage and have not shown success in repairing the broken fiber, which prevents cellular regeneration and integral restoration of motor and sensory functions. Considering the importance of nanotechnology and tissue engineering for neural tissue injuries, this review focuses on the latest advances in nanotechnology for SCI treatment and tissue repair. The PubMed database was used for the bibliographic survey. Initial research using the following keywords "tissue engineering and spinal cord injury" revealed 970 articles published in the last 10 years. The articles were further analyzed, excluding those not related to SCI or with results that did not pertain to the field of interest, including the reviews. It was observed that a total of 811 original articles used the quoted keywords. When the word "treatment" was added, 662 articles were found and among them, 529 were original ones. Finally, when the keywords "Nanotechnology and spinal cord injury" were used, 102 articles were found, 65 being original articles. A search concerning the biomaterials used for SCI found 700 articles with 589 original articles. A total of 107 articles were included in the discussion of this review and some are used for the theoretical framework. Recent progress in nanotechnology and tissue engineering has shown promise for repairing CNS damage. A variety of in vivo animal testing for SCI has been used with or without cells and some of these in vivo studies have shown successful results. However, there is no translation to humans using nanotechnology for SCI treatment, although there is one ongoing trial that employs a tissue engineering approach, among other technologies. The first human surgical scaffold implantation will elucidate the possibility of this use for further clinical trials. This review concludes that even though tissue engineering and nanotechnology are being investigated as a possibility for SCI treatment, tests with humans are still in the theoretical stage. Impact statement Thousands of people are affected by spinal cord injury (SCI) per year in the world. This type of lesion is one of the most severe conditions that can affect humans and usually causes permanent loss of strength, sensitivity, and motor function below the injury site. This article reviews studies on the PubMed database, assessing the publications on SCI in the study field of tissue engineering, focusing on the use of nanotechnology for the treatment of SCI. The review makes an evaluation of the biomaterials used for the treatment of this condition and the techniques applied for the production of nanostructured biomaterials.
Collapse
Affiliation(s)
- Rafaela Zimmermann
- Hematology and Stem Cell Laboratory, Faculty of Pharmacy, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Yuri Vieira Alves
- Hematology and Stem Cell Laboratory, Faculty of Pharmacy, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Laura E Sperling
- Hematology and Stem Cell Laboratory, Faculty of Pharmacy, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Health School, Faculty of Medicine, UNISINOS, São Leopoldo, Brazil
| | - Patricia Pranke
- Hematology and Stem Cell Laboratory, Faculty of Pharmacy, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Stem Cell Research Institute, Porto Alegre, Brazil
| |
Collapse
|
20
|
Recent progress in therapeutic drug delivery systems for treatment of traumatic CNS injuries. Future Med Chem 2020; 12:1759-1778. [PMID: 33028091 DOI: 10.4155/fmc-2020-0178] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Most therapeutics for the treatment of traumatic central nervous system injuries, such as traumatic brain injury and spinal cord injury, encounter various obstacles in reaching the target tissue and exerting pharmacological effects, including physiological barriers like the blood-brain barrier and blood-spinal cord barrier, instability rapid elimination from the injured tissue or cerebrospinal fluid and off-target toxicity. For central nervous system delivery, nano- and microdrug delivery systems are regarded as the most suitable and promising carriers. In this review, the pathophysiology and biomarkers of traumatic central nervous system injuries (traumatic brain injury and spinal cord injury) are introduced. Furthermore, various drug delivery systems, novel combinatorial therapies and advanced therapies for the treatment of traumatic brain injury and spinal cord injury are emphasized.
Collapse
|
21
|
Betala J, Bae S, Langan EM, LaBerge M, Lee JS. Combinatorial therapy of sirolimus and heparin by nanocarrier inhibits restenosis after balloon angioplasty ex vivo. Nanomedicine (Lond) 2020; 15:1205-1220. [PMID: 32340540 DOI: 10.2217/nnm-2020-0028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: To develop poly(lactide-co-glycolide)-graft-polyethylenimine (PgP) as a dual drug-delivery carrier for sirolimus (SR) and heparin (Hep) to inhibit restenosis after balloon angioplasty. Materials & methods: SR was loaded in the hydrophobic core and negatively charged Hep complexed with the positively charged hydrophilic shell of PgP. SR- and Hep-loaded PgP was tested on rat aortic smooth muscle cells in vitro and injured porcine coronary arteries after balloon angioplasty ex vivo. Results & conclusion: SR and Hep loading efficiency in PgP were approximately 37 and 82%, respectively. SR- and Hep-loaded PgP treatment decreased smooth muscle cell proliferation up to 14 days post-treatment and decreased proliferation, collagen deposition and neointimal thickness and increased patency in porcine coronary arteries after balloon angioplasty ex vivo.
Collapse
Affiliation(s)
- Jayesh Betala
- Department of Bioengineering, Clemson University, SC 29634, USA
| | - Sooneon Bae
- Department of Bioengineering, Clemson University, SC 29634, USA
| | - Eugene M Langan
- Department of Vascular Surgery, Greenville Health System, Greenville, SC 29615, USA
| | - Martine LaBerge
- Department of Bioengineering, Clemson University, SC 29634, USA
| | - Jeoung Soo Lee
- Department of Bioengineering, Clemson University, SC 29634, USA
| |
Collapse
|
22
|
Khan TI, Hemalatha S, Waseem M. Promising Role of Nano-Encapsulated Drugs for Spinal Cord Injury. Mol Neurobiol 2020; 57:1978-1985. [PMID: 31900861 DOI: 10.1007/s12035-019-01862-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 12/15/2019] [Indexed: 12/18/2022]
Abstract
Nanomaterials have been utilized for the drug delivery in the central nervous system (CNS), and many research investigators are currently focussing on this specified area. There has been a lot of advancement in the nanoparticle-mediated drug delivery to the brain. Neuronal injuries including spinal cord injury (SCI) and their targeted therapies are still in its infancy on this planet. SCI has been known to cause axonal damage followed by the loss of communication between CNS and other non-neuronal systems. SCI has been critically associated with prolonged inflammation, sensory dysfunction, and motor impairment in SCI patients. There has been a critical crosstalk in SCI and blood brain barriers (BBBs) for drug absorption and distribution in patients. There is a paucity of possible therapies for proper intervention of SCI due to selective permeability of the drugs across BBB. Nanomaterials are contemplated in the drug delivery system for SCI. In addition, self-assembled nanomicelles, lipid nanoparticles, and other co-polymers have now been explored for neuronal injuries. This review focuses on the promising approach and/or role of nanodrug delivery to target SCI in both in vitro and in vivo models.
Collapse
Affiliation(s)
- Tasneem Ismail Khan
- School of Life Sciences, B.S. Abdur Rahman Crescent Institute of Science and Technology, Chennai, 600048, India
| | - S Hemalatha
- School of Life Sciences, B.S. Abdur Rahman Crescent Institute of Science and Technology, Chennai, 600048, India
| | - Mohammad Waseem
- School of Life Sciences, B.S. Abdur Rahman Crescent Institute of Science and Technology, Chennai, 600048, India.
| |
Collapse
|
23
|
Suicide Gene Therapy By Amphiphilic Copolymer Nanocarrier for Spinal Cord Tumor. NANOMATERIALS 2019; 9:nano9040573. [PMID: 30965667 PMCID: PMC6523721 DOI: 10.3390/nano9040573] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 03/30/2019] [Accepted: 03/30/2019] [Indexed: 02/06/2023]
Abstract
Spinal cord tumors (SCT) are uncommon neoplasms characterized by irregular growth of tissue inside the spinal cord that can result in non-mechanical back pain. Current treatments for SCT include surgery, radiation therapy, and chemotherapy, but these conventional therapies have many limitations. Suicide gene therapy using plasmid encoding herpes simplex virus-thymidine kinase (pHSV-TK) and ganciclovir (GCV) has been an alternative approach to overcome the limitations of current therapies. However, there is a need to develop a carrier that can deliver both pHSV-TK and GCV for improving therapeutic efficacy. Our group developed a cationic, amphiphilic copolymer, poly (lactide-co-glycolide) -graft-polyethylenimine (PgP), and demonstrated its efficacy as a drug and gene carrier in both cell culture studies and animal models. In this study, we evaluated PgP as a gene carrier and demonstrate that PgP can efficiently deliver reporter genes, pGFP in rat glioma (C6) cells in vitro, and pβ-gal in a rat T5 SCT model in vivo. We also show that PgP/pHSV-TK with GCV treatment showed significantly higher anticancer activity in C6 cells compared to PgP/pHSV-TK without GCV treatment. Finally, we demonstrate that PgP/pHSV-TK with GCV treatment increases the suicide effect and apoptosis of tumor cells and reduces tumor size in a rat T5 SCT model.
Collapse
|
24
|
Gao Y, Vijayaraghavalu S, Stees M, Kwon BK, Labhasetwar V. Evaluating accessibility of intravenously administered nanoparticles at the lesion site in rat and pig contusion models of spinal cord injury. J Control Release 2019; 302:160-168. [PMID: 30930216 DOI: 10.1016/j.jconrel.2019.03.026] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 03/13/2019] [Accepted: 03/27/2019] [Indexed: 01/17/2023]
Abstract
In spinal cord injury (SCI), timely therapeutic intervention is critical to inhibit the post-injury rapidly progressing degeneration of spinal cord. Towards that objective, we determined the accessibility of intravenously administered biodegradable nanoparticles (NPs) as a drug delivery system to the lesion site in rat and pig contusion models of SCI. Poly (d,l-lactide co-glycolide, PLGA)-based NPs loaded with a near-infrared dye as a marker for NPs were used. To analyze and quantify localization of NPs to the lesion site, we mapped the entire spinal cord, segment-by-segment, for the signal count. Our objectives were to determine the NP dose effect and duration of retention of NPs at the lesion site, and the time window post-SCI within which NPs localize at the lesion site. We hypothesized that breakdown of the blood-spinal cord barrier following contusion injury could lead to more specific localization of NPs at the lesion site. The mapping data showed a dose-dependent increase and significantly greater localization of NPs at the lesion site than in the remaining uninjured segment of the spinal cord. Further, NPs were seen to be retained at the lesion site for more than a week. With delayed post-SCI administration, localization of NPs at the lesion site was reduced but still localize even at four weeks post-injury administration. Interestingly, in uninjured animals (sham control), greater accumulation of NPs was seen in the thoracic and lumbar enlargement regions of the spinal cord, which in animals with SCI changed to the lesion site, indicating drastic post-injury hemodynamic changes in the spinal cord. Similar to the rat results, pig contusion model of SCI showed greater NP localization at the lesion site. In conclusion, NPs could potentially be explored as a carrier for delivery of therapeutics to the lesion site to minimize the impact of post-SCI response.
Collapse
Affiliation(s)
- Yue Gao
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Sivakumar Vijayaraghavalu
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Melinda Stees
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Brian K Kwon
- Department of Orthopedics, International Collaboration of Repair Discoveries (ICORD), University of British Columbia, Vancouver, BC, Canada
| | - Vinod Labhasetwar
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA.
| |
Collapse
|
25
|
Chen KS, McGinley LM, Kashlan ON, Hayes JM, Bruno ES, Chang JS, Mendelson FE, Tabbey MA, Johe K, Sakowski SA, Feldman EL. Targeted intraspinal injections to assess therapies in rodent models of neurological disorders. Nat Protoc 2019; 14:331-349. [PMID: 30610242 DOI: 10.1038/s41596-018-0095-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Despite decades of research, pharmacological therapies for spinal cord motor pathologies are limited. Alternatives using macromolecular, viral, or cell-based therapies show early promise. However, introducing these substances into the spinal cord, past the blood-brain barrier, without causing injury is challenging. We describe a technique for intraspinal injection targeting the lumbar ventral horn in rodents. This technique preserves motor performance and has a proven track record of translation into phase 1 and 2 clinical trials in amyotrophic lateral sclerosis (ALS) patients. The procedure, in brief, involves exposure of the thoracolumbar spine and dissection of paraspinous muscles over the target vertebrae. Following laminectomy, the spine is affixed to a stereotactic frame, permitting precise and reproducible injection throughout the lumbar spine. We have used this protocol to inject various stem cell types, primarily human spinal stem cells (HSSCs); however, the injection is adaptable to any candidate therapeutic cell, virus, or macromolecule product. In addition to a detailed procedure, we provide stereotactic coordinates that assist in targeting of the lumbar spine and instructional videos. The protocol takes ~2 h per animal.
Collapse
Affiliation(s)
- Kevin S Chen
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| | - Lisa M McGinley
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | - Osama N Kashlan
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| | - John M Hayes
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | | | - Josh S Chang
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | - Faye E Mendelson
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | - Maegan A Tabbey
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | | | | | - Eva L Feldman
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
26
|
Nanodelivery systems for overcoming limited transportation of therapeutic molecules through the blood-brain barrier. Future Med Chem 2018; 10:2659-2674. [PMID: 30499740 DOI: 10.4155/fmc-2018-0208] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Due to the impermeable structure and barrier function of the blood-brain barrier (BBB), the delivery of therapeutic molecules into the CNS is extremely limited. Nanodelivery systems are regarded as the most effective and versatile carriers for the CNS, as they can transport cargo molecules across the BBB via various mechanisms. This review emphasizes the multi-functionalization strategies of nanodelivery systems and combinatorial approaches for the delivery of therapeutic drugs and genes into the CNS. The characteristics and functions of the BBB and underlying mechanisms of molecular translocation across the BBB are also described.
Collapse
|
27
|
Zhang H, Wang W, Li N, Li P, Liu M, Pan J, Wang D, Li J, Xiong Y, Xia L. LncRNA DGCR5 suppresses neuronal apoptosis to improve acute spinal cord injury through targeting PRDM5. Cell Cycle 2018; 17:1992-2000. [PMID: 30146926 DOI: 10.1080/15384101.2018.1509622] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Spinal cord injury (SCI) usually results in neurological damage. DGCR5 is closely related to neurological disorders, and this study aims to explore its role in neuronal apoptosis in acute SCI. The ASCI model was established in rats, and the Basso, Beattie, and Bresnahan (BBB) scoring was used to assess the neurological function. The expression of RNA and protein was quantified by quantitative real-time PCR (qRT-PCR) and western blotting, respectively. The oxygenglucose deprivation (OGD) was performed upon neurons and apoptosis was evaluated by flow cytometry. The interaction and binding between DGCR5 and PRDM5 was detected with RNA pull-down and RIP assay, respectively. DGCR5 was down-regulated in ASCI model rat and in neurons treated with hypoxia. Over-expression of DGCR5 inhibited neuronal apoptosis. Interaction between DGCR5 negatively regulated PRDM5 protein expression by binding and interacting with it. DGCR5 inhibited neuronal apoptosis through PRDM5. Over-expressed DGCR5 ameliorated ASCI in rat. DGCR5 suppresses neuronal apoptosis through directly binding and negatively regulating PRDM5, and thereby ameliorating ASCI.
Collapse
Affiliation(s)
- Huafeng Zhang
- a Department of Orthopedics , the First Affiliated Hospital of Zhengzhou University , Zhengzhou , China
| | - Wengang Wang
- a Department of Orthopedics , the First Affiliated Hospital of Zhengzhou University , Zhengzhou , China
| | - Ning Li
- a Department of Orthopedics , the First Affiliated Hospital of Zhengzhou University , Zhengzhou , China
| | - Peng Li
- a Department of Orthopedics , the First Affiliated Hospital of Zhengzhou University , Zhengzhou , China
| | - Ming Liu
- a Department of Orthopedics , the First Affiliated Hospital of Zhengzhou University , Zhengzhou , China
| | - Junwei Pan
- a Department of Orthopedics , the First Affiliated Hospital of Zhengzhou University , Zhengzhou , China
| | - Dan Wang
- a Department of Orthopedics , the First Affiliated Hospital of Zhengzhou University , Zhengzhou , China
| | - Junwei Li
- a Department of Orthopedics , the First Affiliated Hospital of Zhengzhou University , Zhengzhou , China
| | - Yuanyuan Xiong
- b Department of Hematology , the Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital , Zhengzhou , China
| | - Lei Xia
- a Department of Orthopedics , the First Affiliated Hospital of Zhengzhou University , Zhengzhou , China
| |
Collapse
|
28
|
Jia ZQ, Li SQ, Qiao WQ, Xu WZ, Xing JW, Liu JT, Song H, Gao ZY, Xing BW, He XJ. Ebselen protects mitochondrial function and oxidative stress while inhibiting the mitochondrial apoptosis pathway after acute spinal cord injury. Neurosci Lett 2018; 678:110-117. [PMID: 29733976 DOI: 10.1016/j.neulet.2018.05.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Revised: 04/20/2018] [Accepted: 05/03/2018] [Indexed: 10/17/2022]
Abstract
Ebselen is a fat-soluble small molecule and organic selenium compound that regulates the activity of glutathione peroxidase to alleviate mitochondrial oxidative stress and improve mitochondrial function. In the present study, we aimed to investigate the effects of ebselen on mitochondrial oxidative stress response, mitochondrial apotosis, and motor behaviors after spinal cord injury (SCI). We found that ebselen significantly increased the BBB score in motor behavior, thus suggesting a rescue effect of ebselen on motor function after SCI in rats. Meanwhile, we revealed that ebselen can increase glutathione (GSH) content as well as superoxide dismutase (SOD) and catalase (CAT) activities after SCI-this suggests ebselen has an antioxidant effect. Furthermore, the ATP content and Na+-K+-ATPase activity in mitochondria were increased by ebselen after SCI, while the mitochondrial membrane potential (MMP) was decreased by ebselen. The Cytochrome C and Smac release from mitochondria were reduced by ebselen after SCI, thus indicating improved membrane permeability by ebselen. Moreover, the alterations in caspase-3, Bax and Bcl-2 protein expression, as well as the proportion of cell apoptosis were improved by ebselen treatment, which together suggested that ebselen has an inhibitory effect on mitochondrial apotosis pathways after SCI. Taken together, our results suggest that ebselen can inhibit secondary damage caused by spinal cord injury. Indeed it plays a neuroprotective role in spinal cord injury perhaps by improving mitochondrial function and inhibiting the mitochondrial apoptosis pathway.
Collapse
Affiliation(s)
- Zhi-Qiang Jia
- Department of Orthopedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xian 710004, PR China; Department of Spinal Surgery, The Second Affiliated Hospital of Henan University of Science and Technology, Luoyang 471000, PR China.
| | - San-Qiang Li
- Medical College, Henan University of Science and Technology, Luoyang 471003, PR China
| | - Wei-Qiang Qiao
- Department of Breast Surgery, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, PR China
| | - Wen-Zhong Xu
- Department of Spinal Surgery, The Second Affiliated Hospital of Henan University of Science and Technology, Luoyang 471000, PR China
| | - Jian-Wu Xing
- Department of Spinal Surgery, The Second Affiliated Hospital of Henan University of Science and Technology, Luoyang 471000, PR China
| | - Jian-Tao Liu
- Department of Orthopedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xian 710004, PR China
| | - Hui Song
- Department of Orthopedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xian 710004, PR China
| | - Zhong-Yang Gao
- Department of Orthopedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xian 710004, PR China
| | - Bing-Wen Xing
- Medical College, Henan University of Science and Technology, Luoyang 471003, PR China
| | - Xi-Jing He
- Department of Orthopedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xian 710004, PR China.
| |
Collapse
|
29
|
Wang P, Xie ZD, Xie CN, Lin CW, Wang JL, Xuan LN, Zhang CW, Wang Y, Huang ZH, Teng HL. AMP-activated protein kinase-dependent induction of autophagy by erythropoietin protects against spinal cord injury in rats. CNS Neurosci Ther 2018; 24:1185-1195. [PMID: 29656591 DOI: 10.1111/cns.12856] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 03/13/2018] [Accepted: 03/13/2018] [Indexed: 12/16/2022] Open
Abstract
AIMS Autophagy has been regarded as a promising therapeutic target for spinal cord injury (SCI). Erythropoietin (EPO) has been demonstrated to exhibit neuroprotective effects in the central nervous system (CNS); however, the molecular mechanisms of its protection against SCI remain unknown. This study aims to investigate whether the neuroprotective effects of EPO on SCI are mediated by autophagy via AMP-activated protein kinase (AMPK) signaling pathways. METHODS Functional assessment and Nissl staining were used to investigate the effects of EPO on SCI. Expressions of proteins were detected by Western blot and immunohistochemistry. RESULTS Treatment with EPO significantly reduced the loss of motor neurons and improved the functional recovery following SCI. Erythropoietin significantly enhanced the SCI-induced autophagy through activating AMPK and inactivating mTOR signaling. The inhibitor of AMPK, compound C, could block the EPO-induced autophagy and beneficial action on SCI, whereas the activator of AMPK, metformin, could mimic the effects of EPO. In the in vitro studies, EPO enhanced the hypoxia-induced autophagy in an AMPK-dependent manner. CONCLUSIONS The AMPK-dependent induction of autophagy contributes to the neuroprotection of EPO on SCI.
Collapse
Affiliation(s)
- Peng Wang
- Department of Spine Surgery, Wenzhou Medical University First Affiliated Hospital, Wenzhou, Zhejiang, China.,Department of Emergency Medicine, Wenzhou Medical University Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou, Zhejiang, China
| | - Zhong-Dong Xie
- Department of Gastrointestinal Surgery, Wenzhou Medical University First Affiliated Hospital, Wenzhou, Zhejiang, China
| | - Chang-Nan Xie
- Department of Spine Surgery, Wenzhou Medical University First Affiliated Hospital, Wenzhou, Zhejiang, China.,Institute of Neuroscience and Institute of Hypoxia Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Chao-Wei Lin
- Department of Spine Surgery, Wenzhou Medical University First Affiliated Hospital, Wenzhou, Zhejiang, China
| | - Ji-Li Wang
- Department of Pathology, Zhejiang University First Affiliated Hospital, Hangzhou, Zhejiang, China
| | - Li-Na Xuan
- Institute of Neuroscience and Institute of Hypoxia Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Chun-Wu Zhang
- Department of Spine Surgery, Wenzhou Medical University First Affiliated Hospital, Wenzhou, Zhejiang, China
| | - Yu Wang
- Department of Spine Surgery, Wenzhou Medical University First Affiliated Hospital, Wenzhou, Zhejiang, China
| | - Zhi-Hui Huang
- Institute of Neuroscience and Institute of Hypoxia Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Hong-Lin Teng
- Department of Spine Surgery, Wenzhou Medical University First Affiliated Hospital, Wenzhou, Zhejiang, China
| |
Collapse
|