1
|
Choi B, Lee S, Chung S, Barcelona EE, Hong J, Lee SJ. PLGA nanoparticle-mediated anti-inflammatory gene delivery for the treatment of neuropathic pain. Nanomedicine (Lond) 2025; 20:943-954. [PMID: 40186589 PMCID: PMC12051573 DOI: 10.1080/17435889.2025.2487410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 03/28/2025] [Indexed: 04/07/2025] Open
Abstract
AIM This study aimed to mitigate neuropathic pain behavior in a sciatic nerve transection (SNT)-induced mouse model by delivering anti-inflammatory cytokines - interleukin-4 (IL-4), interleukin-10 (IL-10), and transforming growth factor-beta 1 (TGF-β1) - via poly(d,l-lactic-co-glycolic acid) (PLGA) nanoparticles (NPs). MATERIALS & METHODS Upon gene delivery of IL-4, IL-10, and TGF- β1, the anti-inflammatory effects and induction of microglia M2 polarization were evaluated. Plasmid (IL-4, IL-10, and TGF-β1)-encapsulated PLGA NPs (PLGA@IL-4, PLGA@IL-10, and PLGA@TGF-β1) were synthesized and characterized for size, zeta potential, cellular toxicity, and cellular uptake. The analgesic effect of anti-inflammatory gene delivery using PLGA NPs was then assessed in a mouse model of neuropathic pain. RESULTS Gene delivery of IL-4, IL-10, and TGF-β1 showed a significant anti-inflammatory effect in LPS-treated cells and IL-4 strongly promoted microglia M2 polarization in vitro. PLGA NPs successfully delivered the anti-inflammatory cytokine-coding genes into mouse spinal cord cells, specifically targeting microglia. PLGA@IL-4, PLGA@IL-10, and PLGA@TGF-β1 NPs produced analgesic effects in a SNT-induced mouse neuropathic pain model. Notably, PLGA@IL-4 demonstrated the most effective and remarkably long-lasting analgesic effect, strongly enhancing microglia M2 polarization in spinal cord microglia. CONCLUSION Gene therapy using PLGA NPs for overexpression of anti-inflammatory cytokines could be a promising strategy for the treatment of neuropathic pain.
Collapse
Affiliation(s)
- Boomin Choi
- Department of Neuroscience and Physiology, Dental Research Institute, School of Dentistry, Seoul National University, Seoul, Republic of Korea
| | - Subeen Lee
- Interdisciplinary Program in Neuroscience, College of Natural Science, Seoul National University, Seoul, Republic of Korea
| | - Seohyun Chung
- Department of Neuroscience and Physiology, Dental Research Institute, School of Dentistry, Seoul National University, Seoul, Republic of Korea
| | - Ellane Eda Barcelona
- Department of Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul, Republic of Korea
| | - Jinpyo Hong
- Department of Neuroscience and Physiology, Dental Research Institute, School of Dentistry, Seoul National University, Seoul, Republic of Korea
| | - Sung Joong Lee
- Department of Neuroscience and Physiology, Dental Research Institute, School of Dentistry, Seoul National University, Seoul, Republic of Korea
- Interdisciplinary Program in Neuroscience, College of Natural Science, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
2
|
Shin J, Wu J, Park H, Kim SI, Shin N, Shin HJ, Ren G, Kim JA, Hwang PTJ, Jun HW, Lee SY, Lee S, Kim HG, Kim DW. Microglial pyroptosis drives neuropathic pain and targeting NLRP3 alleviates pain and neuroinflammation. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167737. [PMID: 39971256 DOI: 10.1016/j.bbadis.2025.167737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 01/22/2025] [Accepted: 02/14/2025] [Indexed: 02/21/2025]
Abstract
Neuropathic pain is triggered by nerve damage or disease and involves chronic neuroinflammation driven by activated microglia releasing pro-inflammatory cytokines. PANoptosis, a complex cell death program encompassing apoptosis, pyroptosis, and necroptosis, has emerged as a key player in neuroinflammation. While individual PANoptosis pathway have been linked to pain, its systemic role in neuropathic pain remains unclear. This study explored the involvement of PANoptosis in microglia under neuropathic pain and its potential therapeutic targeting. After spinal nerve ligation (SNL), robust microglia activation and pro-inflammatory cytokines were increased in spinal dorsal horn. To figure out the major PANoptosis under neuropathic pain, bioinformatic analysis and protein analysis were explored by using spinal dorsal horn on 14 days of post injury. The results supported that pyroptosis was the dominant pathway after injury, and we further investigated pyroptosis-related markers on microglia specifically. Notably, pyroptosis marker (caspase-1) was elevated in microglia compared to apoptosis (cleaved caspase-3) and necroptosis (p-RIPK3) markers. This finding highlights microglia pyroptosis as a key driver of neuropathic pain development. To harness this knowledge therapeutically, we employed intrathecal injection of NLRP3 siRNA nanoparticles. NLRP3, a crucial component of the inflammasome complex triggering pyroptosis, served as our target. Strikingly, this intervention effectively alleviated mechanical allodynia, a hallmark of neuropathic pain, alongside reducing microgliosis and dampening microglial pyroptosis. Our findings reveal that microglia pyroptosis plays a key role in neuropathic pain and suggest NLRP3 siRNA nanoparticles as a promising therapeutic avenue for pain management.
Collapse
Affiliation(s)
- Juhee Shin
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon 34126, Republic of Korea
| | - Junhua Wu
- Department of Medical Science, Brain Research Institute, Chungnam National University School of Medicine, Daejeon, Republic of Korea; Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University School of Medicine, Daejeon, Republic of Korea; Department of Neurology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Hyewon Park
- Department of Medical Science, Brain Research Institute, Chungnam National University School of Medicine, Daejeon, Republic of Korea; Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Song I Kim
- Department of Medical Science, Brain Research Institute, Chungnam National University School of Medicine, Daejeon, Republic of Korea; Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Nara Shin
- Department of Medical Science, Brain Research Institute, Chungnam National University School of Medicine, Daejeon, Republic of Korea; Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Hyo Jung Shin
- Department of Medical Science, Brain Research Institute, Chungnam National University School of Medicine, Daejeon, Republic of Korea; Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Guang Ren
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jeong-A Kim
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Patrick T J Hwang
- Rowan-Virtua School of Translational Biomedical Engineering & Sciences, Rowan University, Glassboro, NJ 08028, USA
| | - Ho-Wook Jun
- Department of Biomedical Engineering, University of Alabama at Birmingham, 806 Shelby, 1825 University Boulevard, Birmingham, AL 35294, USA
| | - Sun Yeul Lee
- Department of Anesthesia and Pain Medicine, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Sangkyu Lee
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon 34126, Republic of Korea
| | - Hyeong-Geug Kim
- Nanoglia, Daejeon, Republic of Korea; Department of Biochemistry and Molecular Biology, School of Medicine, Indiana University, Indianapolis, IN 46202, USA.
| | - Dong Woon Kim
- Department of Oral Anatomy and Developmental Biology, Kyung Hee University College of Dentistry, Seoul, Republic of Korea.
| |
Collapse
|
3
|
Keshavarz Shahbaz S, Koushki K, Keshavarz Hedayati S, McCloskey AP, Kesharwani P, Naderi Y, Sahebkar A. Polymer nanotherapeutics: A promising approach toward microglial inhibition in neurodegenerative diseases. Med Res Rev 2024; 44:2793-2824. [PMID: 39031446 DOI: 10.1002/med.22064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 01/30/2024] [Accepted: 07/01/2024] [Indexed: 07/22/2024]
Abstract
Nanoparticles (NPs) that target multiple transport mechanisms facilitate targeted delivery of active therapeutic agents to the central nervous system (CNS) and improve therapeutic transport and efficacy across the blood-brain barrier (BBB). CNS nanotherapeutics mostly target neurons and endothelial cells, however, microglial immune cells are the first line of defense against neuronal damage and brain infections. Through triggering release of inflammatory cytokines, chemokines and proteases, microglia can however precipitate neurological damage-a significant factor in neurodegenerative diseases. Thus, microglial inhibitory agents are attracting much attention among those researching and developing novel treatments for neurodegenerative disorders. The most established inhibitors of microglia investigated to date are resveratrol, curcumin, quercetin, and minocycline. Thus, there is great interest in developing novel agents that can bypass or easily cross the BBB. One such approach is the use of modified-nanocarriers as, or for, delivery of, therapeutic agents to the brain and wider CNS. For microglial inhibition, polymeric NPs are the preferred vehicles for choice. Here, we summarize the immunologic and neuroinflammatory role of microglia, established microglia inhibitor agents, challenges of CNS drug delivery, and the nanotherapeutics explored for microglia inhibition to date. We also discuss applications of the currently considered "most useful" polymeric NPs for microglial-inhibitor drug delivery in CNS-related diseases.
Collapse
Affiliation(s)
- Sanaz Keshavarz Shahbaz
- Cellular and Molecular Research Center, Research Institute for prevention of Non-Communicable Disease, Qazvin University of Medical Sciences, Qazvin, Iran
- USERN Office, Qazvin University of Medical Science, Qazvin, Iran
| | - Khadije Koushki
- Department of Neurosurgery, University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | | | - Alice P McCloskey
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Yazdan Naderi
- Department of Pharmacology, Faculty of Medicine, Qazvin University of Medical Science, Qazvin, Iran
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
4
|
Kim SI, Yang J, Shin J, Shin N, Shin HJ, Lee J, Noh C, Kim DW, Lee SY. Amitriptyline nanoparticle repositioning prolongs the anti-allodynic effect of enhanced microglia targeting. Nanomedicine (Lond) 2024; 19:2099-2112. [PMID: 39229790 PMCID: PMC11485917 DOI: 10.1080/17435889.2024.2390349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 08/06/2024] [Indexed: 09/05/2024] Open
Abstract
Aim: Amitriptyline (AMI) has been used to treat neuropathic pain. However, the clinical outcomes remain unsatisfactory, presumably due to a limited understanding of the underlying molecular mechanisms. Here, we investigated a drug repositioning strategy using a low-dose of AMI encapsulated in poly (D, L lactic-co-glycolic acid) (PLGA) nanoparticles (AMI NPs) for neuropathic pain, since PLGA nanoparticles are known to enhance delivery to microglia.Methods: We evaluated the anti-allodynic effects of AMI and AMI NPs on neuropathic pain by assessing behaviors and inflammatory responses in a rat model of spinal nerve ligation (SNL). While the anti-allodynic effect of AMI (30 μg) drug injection on SNL-induced neuropathic pain persisted for 12 h, AMI NPs significantly alleviated mechanical allodynia for 3 days.Results: Histological and cytokine analyses showed AMI NPs facilitated the reduction of microglial activation and pro-inflammatory mediators in the spinal dorsal horn. This study suggests that AMI NPs can provide a sustained anti-allodynic effect by enhancing the targeting of microglia and regulating the release of pro-inflammatory cytokines from activated microglia.Conclusion: Our findings suggest that the use of microglial-targeted NPs continuously releasing AMI (2 μg) as a drug repositioning strategy offers long-term anti-allodynic effects.
Collapse
Affiliation(s)
- Song I Kim
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea
- Department of Anatomy & Cell Biology, Brain Research Institute, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea
| | - Jiah Yang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX77054, USA
| | - Juhee Shin
- Center for Cognition & Sociality, Institute for Basic Science, Daejeon, Republic of Korea
| | - Nara Shin
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea
- Department of Anatomy & Cell Biology, Brain Research Institute, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea
| | - Hyo Jung Shin
- Department of Anatomy & Cell Biology, Brain Research Institute, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea
| | - Jiyong Lee
- Department of Anesthesia & Pain Medicine, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea
| | - Chan Noh
- Department of Anesthesia & Pain Medicine, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea
| | - Dong Woon Kim
- Department of Oral Anatomy & Developmental Biology, College of Dentistry Kyung Hee University 26, Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea
| | - Sun Yeul Lee
- Department of Anesthesia & Pain Medicine, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea
| |
Collapse
|
5
|
Li H, Guan M, Zhang NN, Wang Y, Liang T, Wu H, Wang C, Sun T, Liu S. Harnessing nanomedicine for modulating microglial states in the central nervous system disorders: Challenges and opportunities. Biomed Pharmacother 2024; 177:117011. [PMID: 38917758 DOI: 10.1016/j.biopha.2024.117011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 05/30/2024] [Accepted: 06/17/2024] [Indexed: 06/27/2024] Open
Abstract
Microglia are essential for maintaining homeostasis and responding to pathological events in the central nervous system (CNS). Their dynamic and multidimensional states in different environments are pivotal factors in various CNS disorders. However, therapeutic modulation of microglial states is challenging due to the intricate balance these cells maintain in the CNS environment and the blood-brain barrier's restriction of drug delivery. Nanomedicine presents a promising avenue for addressing these challenges, offering a method for the targeted and efficient modulation of microglial states. This review covers the challenges faced in microglial therapeutic modulation and potential use of nanoparticle-based drug delivery systems. We provide an in-depth examination of nanoparticle applications for modulating microglial states in a range of CNS disorders, encompassing neurodegenerative and autoimmune diseases, infections, traumatic injuries, stroke, tumors, chronic pain, and psychiatric conditions. This review highlights the recent advancements and future prospects in nanomedicine for microglial modulation, paving the way for future research and clinical applications of therapeutic interventions in CNS disorders.
Collapse
Affiliation(s)
- Haisong Li
- Cancer Center, The First Hospital, Jilin University, Changchun, Jilin, China; Department of Neurosurgery, The First Hospital, Jilin University, Changchun, Jilin, China
| | - Meng Guan
- Cancer Center, The First Hospital, Jilin University, Changchun, Jilin, China; Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China
| | - Ning-Ning Zhang
- State Key Laboratory of Supramolecular Structure and Materials, Jilin University, Changchun, Jilin, China
| | - Yizhuo Wang
- Cancer Center, The First Hospital, Jilin University, Changchun, Jilin, China
| | - Tingting Liang
- Cancer Center, The First Hospital, Jilin University, Changchun, Jilin, China
| | - Haitao Wu
- Cancer Center, The First Hospital, Jilin University, Changchun, Jilin, China
| | - Chang Wang
- Cancer Center, The First Hospital, Jilin University, Changchun, Jilin, China.
| | - Tianmeng Sun
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China; International Center of Future Science, Jilin University, Changchun, Jilin, China; State Key Laboratory of Supramolecular Structure and Materials, Jilin University, Changchun, Jilin, China.
| | - Shuhan Liu
- Cancer Center, The First Hospital, Jilin University, Changchun, Jilin, China; Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China.
| |
Collapse
|
6
|
Shin HJ, Choi SG, Qu F, Yi MH, Lee CH, Kim SR, Kim HG, Beom J, Yi Y, Kim DK, Joe EH, Song HJ, Kim Y, Kim DW. Peptide-mediated targeted delivery of SOX9 nanoparticles into astrocytes ameliorates ischemic brain injury. NANOSCALE 2024; 16:833-847. [PMID: 38093712 DOI: 10.1039/d3nr01318a] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2024]
Abstract
Astrocytes are highly activated following brain injuries, and their activation influences neuronal survival. Additionally, SOX9 expression is known to increase in reactive astrocytes. However, the role of SOX9 in activated astrocytes following ischemic brain damage has not been clearly elucidated yet. Therefore, in the present study, we investigated the role of SOX9 in reactive astrocytes using a poly-lactic-co-glycolic acid (PLGA) nanoparticle plasmid delivery system in a photothrombotic stroke animal model. We designed PLGA nanoparticles to exclusively enhance SOX9 gene expression in glial fibrillary acidic protein (GFAP)-immunoreactive astrocytes. Our observations indicate that PLGA nanoparticles encapsulated with GFAP:SOX9:tdTOM reduce ischemia-induced neurological deficits and infarct volume through the prostaglandin D2 pathway. Thus, the astrocyte-targeting PLGA nanoparticle plasmid delivery system provides a potential opportunity for stroke treatment. Since the only effective treatment currently available is reinstating the blood supply, cell-specific gene therapy using PLGA nanoparticles will open a new therapeutic paradigm for brain injury patients in the future.
Collapse
Affiliation(s)
- Hyo Jung Shin
- Department of Medical Science, Chungnam National University, Daejeon 35015, Korea.
- Department of Anatomy and Cell Biology, Chungnam National University, Daejeon 35015, Korea
- Brain Research Institute, Chungnam National University, Daejeon 35015, Korea
| | - Seung Gyu Choi
- Department of Medical Science, Chungnam National University, Daejeon 35015, Korea.
- Department of Anatomy and Cell Biology, Chungnam National University, Daejeon 35015, Korea
| | - Fengrui Qu
- Department of Chemistry and Biochemistry, The University of Alabama, Tuscaloosa, AL 35487, USA
| | - Min-Hee Yi
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Choong-Hyun Lee
- Department of Pharmacy, College of Pharmacy, Dankook University, Cheonan 31116, Korea
| | - Sang Ryong Kim
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Brain Science and Engineering Institute, Kyungpook National University, Daegu 41566, Korea
| | - Hyeong-Geug Kim
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Jaewon Beom
- Department of Rehabilitation Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Korea
| | - Yoonyoung Yi
- Department of Pediatrics, College of Medicine, Hallym University and Gangdong Sacred Heart Hospital, Seoul 05355, Korea
| | - Do Kyung Kim
- Department of Anatomy, College of Medicine, Konyang University, Daejeon 35365, Republic of Korea
| | - Eun-Hye Joe
- Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, Korea
| | - Hee-Jung Song
- Department of Neurology, Chungnam National University and Sejong Hospital, Sejong 30099, Korea
| | - Yonghyun Kim
- Department of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, AL 35487, USA.
| | - Dong Woon Kim
- Department of Medical Science, Chungnam National University, Daejeon 35015, Korea.
- Department of Anatomy and Cell Biology, Chungnam National University, Daejeon 35015, Korea
- Brain Research Institute, Chungnam National University, Daejeon 35015, Korea
| |
Collapse
|
7
|
Ajeeb R, Clegg JR. Intrathecal delivery of Macromolecules: Clinical status and emerging technologies. Adv Drug Deliv Rev 2023; 199:114949. [PMID: 37286086 DOI: 10.1016/j.addr.2023.114949] [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] [Received: 01/30/2023] [Revised: 05/31/2023] [Accepted: 06/03/2023] [Indexed: 06/09/2023]
Abstract
The proximity and association of cerebrospinal fluid (CSF) and the intrathecal (IT) space with deep targets in the central nervous system (CNS) parenchyma makes IT injection an attractive route of administration for brain drug delivery. However, the extent to which intrathecally administered macromolecules are effective in treating neurological diseases is a question of both clinical debate and technological interest. We present the biological, chemical, and physical properties of the intrathecal space that are relevant to drug absorption, distribution, metabolism, and elimination from CSF. We then analyze the evolution of IT drug delivery in clinical trials over the last 20 years. Our analysis revealed that the percentage of clinical trials assessing IT delivery for the delivery of biologics (i.e., macromolecules, cells) for treatment of chronic conditions (e.g., neurodegeneration, cancer, and metabolic diseases) has steadily increased. Clinical trials exploring cell or macromolecular delivery within the IT space have not evaluated engineering technologies, such as depots, particles, or other delivery systems. Recent pre-clinical studies have evaluated IT macromolecule delivery in small animals, postulating that delivery efficacy can be assisted by external medical devices, micro- or nanoparticles, bulk biomaterials, and viral vectors. Further studies are necessary to evaluate the extent to which engineering technologies and IT administration improve CNS targeting and therapeutic outcome.
Collapse
Affiliation(s)
- Rana Ajeeb
- Stephenson School of Biomedical Engineering, The University of Oklahoma, Norman, OK, United States
| | - John R Clegg
- Stephenson School of Biomedical Engineering, The University of Oklahoma, Norman, OK, United States; Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States; Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States; Institute for Biomedical Engineering, Science, and Technology, University of Oklahoma, Norman, OK, United States.
| |
Collapse
|
8
|
Choi SG, Shin J, Lee KY, Park H, Kim SI, Yi YY, Kim DW, Song HJ, Shin HJ. PINK1 siRNA-loaded poly(lactic-co-glycolic acid) nanoparticles provide neuroprotection in a mouse model of photothrombosis-induced ischemic stroke. Glia 2023; 71:1294-1310. [PMID: 36655313 DOI: 10.1002/glia.24339] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 12/14/2022] [Accepted: 01/05/2023] [Indexed: 01/20/2023]
Abstract
PTEN-induced kinase 1 (PINK1) is a well-known critical marker in the pathway for mitophagy regulation as well as mitochondrial dysfunction. Evidence suggests that mitochondrial dynamics and mitophagy flux play an important role in the development of brain damage from stroke pathogenesis. In this study, we propose a treatment strategy using nanoparticles that can control PINK1. We used a murine photothrombotic ischemic stroke (PTS) model in which clogging of blood vessels is induced with Rose Bengal (RB) to cause brain damage. We targeted PINK1 with poly(lactic-co-glycolic acid) (PLGA)-based nanoparticles loaded with PINK1 siRNA (PINK1 NPs). After characterizing siRNA loading in the nanoparticles, we assessed the efficacy of PINK1 NPs in mice with PTS using immunohistochemistry, 1% 2,3,5-triphenyltetrazolium chloride staining, measurement of motor dysfunction, and Western blot. PINK1 was highly expressed in microglia 24 h after PTS induction. PINK1 siRNA treatment increased phagocytic activity, migration, and expression of an anti-inflammatory state in microglia. In addition, the PLGA nanoparticles were selectively taken up by microglia and specifically regulated PINK1 expression in those cells. Treatment with PINK1 NPs prior to stroke induction reduced expression of mitophagy-inducing factors, infarct volume, and motor dysfunction in mice with photothrombotic ischemia. Experiments with PINK1-knockout mice and microglia depletion with PLX3397 confirmed a decrease in stroke-induced infarct volume and behavioral dysfunction. Application of nanoparticles for PINK1 inhibition attenuates RB-induced photothrombotic ischemic injury by inhibiting microglia responses, suggesting that a nanomedical approach targeting the PINK1 pathway may provide a therapeutic avenue for stroke treatment.
Collapse
Affiliation(s)
- Seung Gyu Choi
- Department of Medical Science, Chungnam National University, Daejeon, Republic of Korea
- Department of Anatomy and Cell Biology, Chungnam National University, Daejeon, Republic of Korea
| | - Juhee Shin
- Department of Medical Science, Chungnam National University, Daejeon, Republic of Korea
- Department of Anatomy and Cell Biology, Chungnam National University, Daejeon, Republic of Korea
| | - Ka Young Lee
- Department of Medical Science, Chungnam National University, Daejeon, Republic of Korea
- Department of Anatomy and Cell Biology, Chungnam National University, Daejeon, Republic of Korea
| | - Hyewon Park
- Department of Medical Science, Chungnam National University, Daejeon, Republic of Korea
- Department of Anatomy and Cell Biology, Chungnam National University, Daejeon, Republic of Korea
| | - Song I Kim
- Department of Medical Science, Chungnam National University, Daejeon, Republic of Korea
- Department of Anatomy and Cell Biology, Chungnam National University, Daejeon, Republic of Korea
| | - Yoon Young Yi
- Department of Pediatrics, College of Medicine, Hallym University and Gangdong Sacred Heart Hospital, Seoul, Republic of Korea
| | - Dong Woon Kim
- Department of Medical Science, Chungnam National University, Daejeon, Republic of Korea
- Department of Anatomy and Cell Biology, Chungnam National University, Daejeon, Republic of Korea
| | - Hee-Jung Song
- Department of Neurology, Chungnam National University Sejong Hospital and College of Medicine, Republic of Korea
| | - Hyo Jung Shin
- Brain Research Institute, Chungnam National University, Daejeon, Republic of Korea
| |
Collapse
|
9
|
DUSP8/TAK1 signaling mediates neuropathic pain through regulating neuroinflammation and neuron death in a spinal nerve ligation (SNL) rat model. Int Immunopharmacol 2022; 113:109284. [DOI: 10.1016/j.intimp.2022.109284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 09/01/2022] [Accepted: 09/22/2022] [Indexed: 11/05/2022]
|
10
|
Tran Q, Pham TL, Shin HJ, Shin J, Shin N, Kwon HH, Park H, Kim SI, Choi SG, Wu J, Ngo VTH, Park JB, Kim DW. Targeting spinal microglia with fexofenadine-loaded nanoparticles prolongs pain relief in a rat model of neuropathic pain. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2022; 44:102576. [PMID: 35714922 DOI: 10.1016/j.nano.2022.102576] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 06/07/2022] [Accepted: 06/08/2022] [Indexed: 06/15/2023]
Abstract
Targeting microglial activation is emerging as a clinically promising drug target for neuropathic pain treatment. Fexofenadine, a histamine receptor 1 antagonist, is a clinical drug for the management of allergic reactions as well as pain and inflammation. However, the effect of fexofenadine on microglial activation and pain behaviors remains elucidated. Here, we investigated nanomedicinal approach that targets more preferentially microglia and long-term analgesics. Fexofenadine significantly abolished histamine-induced microglial activation. The fexofenadine-encapsulated poly(lactic-co-glycolic acid) nanoparticles (Fexo NPs) injection reduced the pain sensitivity of spinal nerve ligation rats in a dose-dependent manner. This alleviation was sustained for 4 days, whereas the effective period by direct fexofenadine injection was 3 h. Moreover, Fexo NPs inhibited microglial activation, inflammatory signaling, cytokine release, and a macrophage phenotype shift towards the alternative activated state in the spinal cord. These results show that Fexo NPs exhibit drug repositioning promise as a long-term treatment modality for neuropathic pain.
Collapse
Affiliation(s)
- Quangdon Tran
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon 35015, Republic of Korea; Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University School of Medicine, Daejeon 35015, Republic of Korea; Molecular Biology Laboratory, Department of Medical Laboratories, Hai Phong International Hospital, Hai Phong City #18000, Viet Nam
| | - Thuy Linh Pham
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon 35015, Republic of Korea; Department of Histology & Embryology, Hai Phong University of Medicine & Pharmacy, Hai Phong 042-12, Viet Nam
| | - Hyo Jung Shin
- Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University School of Medicine, Daejeon 35015, Republic of Korea
| | - Juhee Shin
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon 35015, Republic of Korea
| | - Nara Shin
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon 35015, Republic of Korea
| | - Hyeok Hee Kwon
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon 35015, Republic of Korea
| | - Hyewon Park
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon 35015, Republic of Korea
| | - Song I Kim
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon 35015, Republic of Korea
| | - Seoung Gyu Choi
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon 35015, Republic of Korea
| | - Junhua Wu
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon 35015, Republic of Korea
| | - Van T H Ngo
- Graduate Department of Healthcare Science, Dainam University, Viet Nam
| | - Jin Bong Park
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon 35015, Republic of Korea; Department of Physiology, Brain Research Institute, Chungnam National University School of Medicine, Daejeon 35015, Republic of Korea
| | - Dong Woon Kim
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon 35015, Republic of Korea; Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University School of Medicine, Daejeon 35015, Republic of Korea.
| |
Collapse
|
11
|
Jiang M, Wang Y, Wang J, Feng S, Wang X. The etiological roles of miRNAs, lncRNAs, and circRNAs in neuropathic pain: A narrative review. J Clin Lab Anal 2022; 36:e24592. [PMID: 35808924 PMCID: PMC9396192 DOI: 10.1002/jcla.24592] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 06/24/2022] [Accepted: 06/25/2022] [Indexed: 11/16/2022] Open
Abstract
Background Non‐coding RNAs (ncRNAs) are involved in neuropathic pain development. Herein, we systematically searched for neuropathic pain‐related ncRNAs expression changes, including microRNAs (miRNAs), long non‐coding RNAs (lncRNAs), and circular non‐coding RNAs (circRNAs). Methods We searched two databases, PubMed and GeenMedical, for relevant studies. Results Peripheral nerve injury or noxious stimuli can induce extensive changes in the expression of ncRNAs. For example, higher serum miR‐132‐3p, ‐146b‐5p, and ‐384 was observed in neuropathic pain patients. Either sciatic nerve ligation, dorsal root ganglion (DRG) transaction, or ventral root transection (VRT) could upregulate miR‐21 and miR‐31 while downregulating miR‐668 and miR‐672 in the injured DRG. lncRNAs, such as early growth response 2‐antisense‐RNA (Egr2‐AS‐RNA) and Kcna2‐AS‐RNA, were upregulated in Schwann cells and inflicted DRG after nerve injury, respectively. Dysregulated circRNA homeodomain‐interacting protein kinase 3 (circHIPK3) in serum and the DRG, abnormally expressed lncRNAs X‐inactive specific transcript (XIST), nuclear enriched abundant transcript 1 (NEAT1), small nucleolar RNA host gene 1 (SNHG1), as well as ciRS‐7, zinc finger protein 609 (cirZNF609), circ_0005075, and circAnks1a in the spinal cord were suggested to participate in neuropathic pain development. Dysregulated miRNAs contribute to neuropathic pain via neuroinflammation, autophagy, abnormal ion channel expression, regulating pain‐related mediators, protein kinases, structural proteins, neurotransmission excitatory–inhibitory imbalances, or exosome miRNA‐mediated neuron–glia communication. In addition, lncRNAs and circRNAs are essential in neuropathic pain by acting as antisense RNA and miRNA sponges, epigenetically regulating pain‐related molecules expression, or modulating miRNA processing. Conclusions Numerous dysregulated ncRNAs have been suggested to participate in neuropathic pain development. However, there is much work to be done before ncRNA‐based analgesics can be clinically used for various reasons such as conservation among species, proper delivery, stability, and off‐target effects.
Collapse
Affiliation(s)
- Ming Jiang
- Department of Anesthesiology and Pain Medicine, Nanjing Maternity and Child Health Care Hospital, Women's Hospital of Nanjing Medical University, Nanjing, China
| | - Yelong Wang
- Department of Anesthesiology, Gaochun People's Hospital, Nanjing, China
| | - Jing Wang
- Department of Anesthesiology and Pain Medicine, Nanjing Maternity and Child Health Care Hospital, Women's Hospital of Nanjing Medical University, Nanjing, China
| | - Shanwu Feng
- Department of Anesthesiology and Pain Medicine, Nanjing Maternity and Child Health Care Hospital, Women's Hospital of Nanjing Medical University, Nanjing, China
| | - Xian Wang
- Department of Anesthesiology and Pain Medicine, Nanjing Maternity and Child Health Care Hospital, Women's Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
12
|
da Silva A, Lepetre-Mouelhi S, Couvreur P. Micro- and nanocarriers for pain alleviation. Adv Drug Deliv Rev 2022; 187:114359. [PMID: 35654211 DOI: 10.1016/j.addr.2022.114359] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 05/10/2022] [Accepted: 05/20/2022] [Indexed: 12/28/2022]
Abstract
Acute or chronic pain is a major source of impairment in quality of life and affects a substantial part of the population. To date, pain is alleviated by a limited range of treatments with significant toxicity, increased risk of misuse and inconsistent efficacy, owing, in part, to lack of specificity and/or unfavorable pharmacokinetic properties. Thanks to the unique properties of nanoscaled drug carriers, nanomedicine may enhance drug biodistribution and targeting, thus contributing to improved bioavailability and lower off-target toxicity. After a brief overview of the current situation and the main critical issues regarding pain alleviation, this review will examine the most advanced approaches using nanomedicine of each drug class, from the preclinical stage to approved nanomedicines.
Collapse
|
13
|
Gu Z, Li Y, Zhang L, Chen X, Xu H. Foxp3 attenuates cerebral ischemia/reperfusion injury through microRNA-150-5p-modified NCS1. Exp Cell Res 2021:112942. [PMID: 34822811 DOI: 10.1016/j.yexcr.2021.112942] [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: 06/03/2021] [Revised: 11/12/2021] [Accepted: 11/18/2021] [Indexed: 11/27/2022]
Abstract
OBJECTIVE Cerebral ischemia/reperfusion injury (CI/RI) is a pathological process involving complicated molecular mechanisms. We investigated forkhead box P3 (Foxp3)-related mechanism in CI/RI with particular focus on microRNA (miR)-150-5p/nucleobase cation symporter-1 (NCS1) axis. METHODS A mouse model was constructed by middle cerebral artery occlusion (MCAO) method. Levels of Foxp3, miR-150-5p and NCS1 were assessed in brain tissues of MCAO mice. By determining the neurological behavior function, neurological deficits, brain tissue pathological characteristics, neuronal apoptosis, inflammatory factors, and oxidative stress-related factors, the functional role of Foxp3, miR-150-5p and NCS1 were evaluated in MCAO mice. The feedback loop was analyzed among Foxp3, miR-150-5p and NCS1. RESULTS The level of Foxp3 and NCS1 were reduced and that of miR-150-5p was augmented in MCAO mice. Foxp3 bound to miR-150-5p to target NCS1. Up-regulating Foxp3 or NCS1 or suppressing miR-150-5p improved neurological behavior function and neurological deficits, and reduced brain tissue pathological damage, neuronal apoptosis, inflammatory and oxidative stress reactions in MCAO mice. Silencing miR-150-5p or elevating NCS1 decreased Foxp3 silencing-mediated ischemic injury in MCAO mice. CONCLUSION Foxp3 is neuroprotective in CI/RI through binding to miR-150-5p to promote NCS1 expression.
Collapse
Affiliation(s)
- Zhen Gu
- Department of Neurosurgery, The Affiliated Hospital of Yunnan University, Kunming, 650011, Yunnan, China.
| | - Yajie Li
- Department of Neurosurgery, The Affiliated Hospital of Yunnan University, Kunming, 650011, Yunnan, China
| | - Liang Zhang
- Central Laboratory, The Affiliated Hospital of Yunnan University, Kunming, 650011, Yunnan, China
| | - Xu Chen
- Department of Neurosurgery, The Affiliated Hospital of Yunnan University, Kunming, 650011, Yunnan, China
| | - Hongling Xu
- Department of Neurosurgery, The Affiliated Hospital of Yunnan University, Kunming, 650011, Yunnan, China
| |
Collapse
|
14
|
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
|
15
|
Kim SI, Shin J, Tran Q, Park H, Kwon HH, Shin N, Hwang JA, Shin HJ, Lee J, Lee WH, Lee SY, Kim DW. Application of PLGA nanoparticles to enhance the action of duloxetine on microglia in neuropathic pain. Biomater Sci 2021; 9:6295-6307. [PMID: 34378557 DOI: 10.1039/d1bm00486g] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Duloxetine (DLX) is a selective serotonin and noradrenaline reuptake inhibitor (SNRI) used for the treatment of pain, but it has been reported to show side effects in 10-20% of patients. Its analgesic efficacy in central pain is putatively related to its influence on descending inhibitory neuronal pathways. However, DLX can also affect the activation of microglia. This study was performed to investigate whether PLGA nanoparticles (NPs), which are expected to enhance targeting to microglia, can improve the analgesic efficacy and limit the side effects of DLX. PLGA NPs encapsulating a low dose of DLX (DLX NPs) were synthesized and characterized and their localization was determined. The analgesic and anti-inflammatory effects of DLX NPs were evaluated in a spinal nerve ligation (SNL)-induced neuropathic pain model. The analgesic effect of DLX lasted for only a few hours and disappeared within 1 day. However, DLX NPs alleviated mechanical allodynia, and the effect was maintained for 1 week. DLX NPs were localized to the spinal microglia and suppressed microglial activation, phosphorylation of p38/NF-κB-mediated pathways and the production of inflammatory cytokines in the spinal dorsal horn of SNL rats. We demonstrated that DLX NPs can provide a prolonged analgesic effect by enhanced targeting of microglia. Our observations imply that DLX delivery through nanoparticle encapsulation allows drug repositioning with a prolonged analgesic effect, and reduces the potential side effects of abuse and overdose.
Collapse
Affiliation(s)
- Song I Kim
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea and Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea.
| | - Juhee Shin
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea and Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea.
| | - Quangdon Tran
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea and Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea.
| | - Hyewon Park
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea and Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea.
| | - Hyeok Hee Kwon
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea and Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea.
| | - Nara Shin
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea and Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea.
| | - Jeong-Ah Hwang
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea and Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea.
| | - Hyo Jung Shin
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea and Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea.
| | - Jiyong Lee
- Department of Anesthesia and Pain Medicine, Chungnam National University School of Medicine, Daejeon 35015, Republic of Korea.
| | - Won Hyung Lee
- Department of Anesthesia and Pain Medicine, Chungnam National University School of Medicine, Daejeon 35015, Republic of Korea.
| | - Sun Yeul Lee
- Department of Anesthesia and Pain Medicine, Chungnam National University School of Medicine, Daejeon 35015, Republic of Korea.
| | - Dong Woon Kim
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea and Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea.
| |
Collapse
|
16
|
Zhu FD, Hu YJ, Yu L, Zhou XG, Wu JM, Tang Y, Qin DL, Fan QZ, Wu AG. Nanoparticles: A Hope for the Treatment of Inflammation in CNS. Front Pharmacol 2021; 12:683935. [PMID: 34122112 PMCID: PMC8187807 DOI: 10.3389/fphar.2021.683935] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 04/26/2021] [Indexed: 12/12/2022] Open
Abstract
Neuroinflammation, an inflammatory response within the central nervous system (CNS), is a main hallmark of common neurodegenerative diseases, including Alzheimer’s disease (AD), Parkinson’s disease (PD), and amyotrophic lateral sclerosis (ALS), among others. The over-activated microglia release pro-inflammatory cytokines, which induces neuronal death and accelerates neurodegeneration. Therefore, inhibition of microglia over-activation and microglia-mediated neuroinflammation has been a promising strategy for the treatment of neurodegenerative diseases. Many drugs have shown promising therapeutic effects on microglia and inflammation. However, the blood–brain barrier (BBB)—a natural barrier preventing brain tissue from contact with harmful plasma components—seriously hinders drug delivery to the microglial cells in CNS. As an emerging useful therapeutic tool in CNS-related diseases, nanoparticles (NPs) have been widely applied in biomedical fields for use in diagnosis, biosensing and drug delivery. Recently, many NPs have been reported to be useful vehicles for anti-inflammatory drugs across the BBB to inhibit the over-activation of microglia and neuroinflammation. Therefore, NPs with good biodegradability and biocompatibility have the potential to be developed as an effective and minimally invasive carrier to help other drugs cross the BBB or as a therapeutic agent for the treatment of neuroinflammation-mediated neurodegenerative diseases. In this review, we summarized various nanoparticles applied in CNS, and their mechanisms and effects in the modulation of inflammation responses in neurodegenerative diseases, providing insights and suggestions for the use of NPs in the treatment of neuroinflammation-related neurodegenerative diseases.
Collapse
Affiliation(s)
- Feng-Dan Zhu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Yu-Jiao Hu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, China.,Department of Anesthesia, Southwest Medical University, Luzhou, China
| | - Lu Yu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Xiao-Gang Zhou
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Jian-Ming Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Yong Tang
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Da-Lian Qin
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Qing-Ze Fan
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, China.,Department of Pharmacy, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - An-Guo Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, China.,Department of Pharmacy, Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
17
|
Sokolova V, Kostka K, Shalumon KT, Prymak O, Chen JP, Epple M. Synthesis and characterization of PLGA/HAP scaffolds with DNA-functionalised calcium phosphate nanoparticles for bone tissue engineering. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2020; 31:102. [PMID: 33140175 PMCID: PMC7606283 DOI: 10.1007/s10856-020-06442-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 09/25/2020] [Indexed: 05/06/2023]
Abstract
Porous scaffolds of poly(lactide-co-glycolide) (PLGA; 85:15) and nano-hydroxyapatite (nHAP) were prepared by an emulsion-precipitation procedure from uniform PLGA-nHAP spheres (150-250 µm diameter). These spheres were then thermally sintered at 83 °C to porous scaffolds that can serve for bone tissue engineering or for bone substitution. The base materials PLGA and nHAP and the PLGA-nHAP scaffolds were extensively characterized by X-ray powder diffraction, infrared spectroscopy, thermogravimetry, differential scanning calorimetry, and scanning electron microscopy. The scaffold porosity was about 50 vol% as determined by relating mass and volume of the scaffolds, together with the computed density of the solid phase (PLGA-nHAP). The cultivation of HeLa cells demonstrated their high cytocompatibility. In combination with DNA-loaded calcium phosphate nanoparticles, they showed a good activity of gene transfection with enhanced green fluorescent protein (EGFP) as model protein. This is expected enhance bone growth around an implanted scaffold or inside a scaffold for tissue engineering.
Collapse
Affiliation(s)
- Viktoriya Sokolova
- Inorganic Chemistry and Center for Nanointegration Duisburg-Essen (CeNIDE), University of Duisburg-Essen, Universitaetsstr. 5-7, 45117, Essen, Germany
| | - Kathrin Kostka
- Inorganic Chemistry and Center for Nanointegration Duisburg-Essen (CeNIDE), University of Duisburg-Essen, Universitaetsstr. 5-7, 45117, Essen, Germany
| | - K T Shalumon
- Department of Chemical and Materials Engineering, Chang Gung University, Kweishan, Taoyuan, 333, Taiwan
- Inter University Centre for Nanomaterials and Devices, Cochin University of Science and Technology, Cochin, Kerala, 682022, India
| | - Oleg Prymak
- Inorganic Chemistry and Center for Nanointegration Duisburg-Essen (CeNIDE), University of Duisburg-Essen, Universitaetsstr. 5-7, 45117, Essen, Germany
| | - Jyh-Ping Chen
- Department of Chemical and Materials Engineering, Chang Gung University, Kweishan, Taoyuan, 333, Taiwan.
- Department of Plastic and Reconstructive Surgery and Craniofacial Research Center, Chang Gung Memorial Hospital at Linkou, Collage of Medicine, Chang Gung University, Kwei-San, Taoyuan, 33305, Taiwan.
| | - Matthias Epple
- Inorganic Chemistry and Center for Nanointegration Duisburg-Essen (CeNIDE), University of Duisburg-Essen, Universitaetsstr. 5-7, 45117, Essen, Germany.
| |
Collapse
|
18
|
Phạm TL, Kim DW. Poly(lactic-co-glycolic acid) nanomaterial-based treatment options for pain management: a review. Nanomedicine (Lond) 2020; 15:1897-1913. [PMID: 32757701 DOI: 10.2217/nnm-2020-0114] [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] [Indexed: 12/13/2022] Open
Abstract
Neuropathic pain is one of the most intense types of chronic pain; it constitutes a pervasive complaint throughout the public health system. With few effective treatments, it remains a significant challenge. Commercially available drugs for neuropathic pain are still limited and have disappointing efficacy. Therefore, chronic neuropathic pain imposes a tremendous burden on patients' quality of life. Recently, the introduction and application of nanotechnology in multiple fields has accelerated the development of new drugs. This review highlights the application of poly(lactic-co-glycolic acid) nanomaterial-based vehicles for drug delivery and how they improve the therapeutic outcomes for neuropathic pain treatment. Finally, future developments for pain research and effective management are presented.
Collapse
Affiliation(s)
- Thuỳ Linh Phạm
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
- Department of Anatomy, Brain Research Institute, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
- Department of Histology & Embryology, Hai Phong University of Medicine & Pharmacy Hospital, Hai Phong, 042-12, Vietnam
| | - Dong Woon Kim
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
- Department of Anatomy, Brain Research Institute, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
| |
Collapse
|
19
|
Patel G, Thakur NS, Kushwah V, Patil MD, Nile SH, Jain S, Banerjee UC, Kai G. Liposomal Delivery of Mycophenolic Acid With Quercetin for Improved Breast Cancer Therapy in SD Rats. Front Bioeng Biotechnol 2020; 8:631. [PMID: 32612988 PMCID: PMC7308462 DOI: 10.3389/fbioe.2020.00631] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 05/22/2020] [Indexed: 12/27/2022] Open
Abstract
The present study explores the influence of mycophenolic acid (MPA) in combination therapy with quercetin (QC) (impeding MPA metabolic rate) delivered using the liposomal nanoparticles (LNPs). Mycophenolic acid liposome nanoparticles (MPA-LNPs) and quercetin liposome nanoparticles (QC-LNPs) were individually prepared and comprehensively characterized. The size of prepared MPA-LNPs and QC-LNPs were found to be 183 ± 13 and 157 ± 09.8, respectively. The in vitro studies revealed the higher cellular uptake and cytotoxicity of combined therapy (MPA-LNPs + QC-LNPs) compared to individual ones. Moreover pharmacokinetics studies in female SD-rat shown higher T 1 / 2 value (1.94 fold) of combined therapy compared to MPA. Furthermore, in vivo anticancer activity in combination of MPA-LNPs and QC-LNPs was also significantly higher related to other treatments groups. The combination therapy of liposomes revealed the new therapeutic approach for the treatment of breast cancer.
Collapse
Affiliation(s)
- Gopal Patel
- Laboratory of Medicinal Plant Biotechnology, College of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Pharmaceutical Technology (Biotechnology), National Institute of Pharmaceutical Education and Research, Sahibzada Ajit Singh Nagar, India
| | - Neeraj Singh Thakur
- Department of Pharmaceutical Technology (Biotechnology), National Institute of Pharmaceutical Education and Research, Sahibzada Ajit Singh Nagar, India
| | - Varun Kushwah
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Sahibzada Ajit Singh Nagar, India
| | - Mahesh D. Patil
- Department of Systems Biotechnology, Konkuk University, Seoul, South Korea
| | - Shivraj Hariram Nile
- Laboratory of Medicinal Plant Biotechnology, College of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Sanyog Jain
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Sahibzada Ajit Singh Nagar, India
| | - Uttam Chand Banerjee
- Department of Pharmaceutical Technology (Biotechnology), National Institute of Pharmaceutical Education and Research, Sahibzada Ajit Singh Nagar, India
| | - Guoyin Kai
- Laboratory of Medicinal Plant Biotechnology, College of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
20
|
CX3CR1-Targeted PLGA Nanoparticles Reduce Microglia Activation and Pain Behavior in Rats with Spinal Nerve Ligation. Int J Mol Sci 2020; 21:ijms21103469. [PMID: 32423102 PMCID: PMC7279022 DOI: 10.3390/ijms21103469] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 05/08/2020] [Accepted: 05/12/2020] [Indexed: 02/07/2023] Open
Abstract
Activation of CX3CR1 in microglia plays an important role in the development of neuropathic pain. Here, we investigated whether neuropathic pain could be attenuated in spinal nerve ligation (SNL)-induced rats by reducing microglial activation through the use of poly(D,L-lactic-co-glycolic acid) (PLGA)-encapsulated CX3CR1 small-interfering RNA (siRNA) nanoparticles. After confirming the efficacy and specificity of CX3CR1 siRNA, as evidenced by its anti-inflammatory effects in lipopolysaccharide-stimulated BV2 cells in vitro, PLGA-encapsulated CX3CR1 siRNA nanoparticles were synthesized by sonication using the conventional double emulsion (W/O/W) method and administered intrathecally into SNL rats. CX3CR1 siRNA-treated rats exhibited significant reductions in the activation of microglia in the spinal dorsal horn and a downregulation of proinflammatory mediators, as well as a significant attenuation of mechanical allodynia. These data indicate that the PLGA-encapsulated CX3CR1 siRNA nanoparticles effectively reduce neuropathic pain in SNL-induced rats by reducing microglial activity and the expression of proinflammatory mediators. Therefore, we believe that PLGA-encapsulated CX3CR1 siRNA nanoparticles represent a valuable new treatment option for neuropathic pain.
Collapse
|
21
|
Shin N, Shin HJ, Yi Y, Beom J, Lee W, Lee CH, Kim DW. p66shc siRNA-Encapsulated PLGA Nanoparticles Ameliorate Neuropathic Pain Following Spinal Nerve Ligation. Polymers (Basel) 2020; 12:polym12051014. [PMID: 32365512 PMCID: PMC7284875 DOI: 10.3390/polym12051014] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 04/17/2020] [Accepted: 04/22/2020] [Indexed: 12/17/2022] Open
Abstract
p66shc, a member of the shc adaptor protein family, has been shown to participate in regulation of mitochondrial homeostasis, apoptosis, and autophagosome formation. The present study was performed to investigate whether p66shc siRNA-encapsulated poly(d,l-lactic-co-glycolic acid) nanoparticles (p66shc siRNA-PLGA NPs) can attenuate spinal nerve ligation (SNL)-induced neuropathic pain in rats. The SNL-induced pain behavior was decreased in the p66shc siRNA-PLGA NP-treated group compared with the scrambled siRNA-PLGA NP-treated group. In the L5 spinal cord of the p66shc siRNA-PLGA NP-treated group, expression levels of phosphorylated p66shc, cleaved caspase-3, p62, and PINK1, as well as microglial activation, were also decreased. In addition, p66shc knockdown using p66shc siRNA reduced the expression levels of cleaved caspase-3, p62, and PINK1, as well as proinflammatory mediators in the H2O2-treated HT22 neuronal cells. These results suggest that downregulation of p66shc expression in the spinal cord using p66shc siRNA-PLGA NPs could reduce the SNL-induced neuropathic pain by attenuating the SNL-induced aberrant autophagic, mitophagic, and neuroinflammatory processes in rats.
Collapse
Affiliation(s)
- Nara Shin
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon 35015, Korea
- Department of Anatomy, Brain Research Institute, Chungnam National University College of Medicine, Daejeon 35015, Korea
| | - Hyo Jung Shin
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon 35015, Korea
- Department of Anatomy, Brain Research Institute, Chungnam National University College of Medicine, Daejeon 35015, Korea
| | - Yoonyoung Yi
- Department of Pediatrics, College of Medicine, Hallym University and Gangdong Sacred Heart Hospital, Seoul 05355, Korea
| | - Jaewon Beom
- Department of Rehabilitation Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Korea
| | - Wonhyung Lee
- Department of Anesthesia and Pain Medicine, Chungnam National University Hospital, Daejeon 35015, Korea
| | - Choong-Hyun Lee
- Department of Pharmacy, College of Pharmacy, Dankook University, Cheonan 31116, Korea
- Correspondence: (C.-H.L.); (D.W.K.); Tel.: +82-41-550-1441 (C.-H.L.); +82-42-580-8201 (D.W.K.); Fax: +82-41-559-7899 (C.-H.L.); +82-42-586-4800 (D.W.K.)
| | - Dong Woon Kim
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon 35015, Korea
- Department of Anatomy, Brain Research Institute, Chungnam National University College of Medicine, Daejeon 35015, Korea
- Correspondence: (C.-H.L.); (D.W.K.); Tel.: +82-41-550-1441 (C.-H.L.); +82-42-580-8201 (D.W.K.); Fax: +82-41-559-7899 (C.-H.L.); +82-42-586-4800 (D.W.K.)
| |
Collapse
|
22
|
Phạm TL, Yin Y, Kwon HH, Shin N, Kim SI, Park H, Shin J, Shin HJ, Hwang JA, Song HJ, Kim SR, Lee JH, Hwang PTJ, Jun HW, Kim DW. miRNA 146a-5p-loaded poly(d,l-lactic-co-glycolic acid) nanoparticles impair pain behaviors by inhibiting multiple inflammatory pathways in microglia. Nanomedicine (Lond) 2020; 15:1113-1126. [PMID: 32292108 DOI: 10.2217/nnm-2019-0462] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Aims: We investigated whether miRNA (miR) 146a-5p-loaded nanoparticles (NPs) can attenuate neuropathic pain behaviors in the rat spinal nerve ligation-induced neuropathic pain model by inhibiting activation of the NF-κB and p38 MAPK pathways in spinal microglia. Materials & methods: After NP preparation, miR NPs were assessed for their physical characteristics and then injected intrathecally into the spinal cords of rat spinal nerve ligation rats to test their analgesic effects. Results: miR NPs reduced pain behaviors for 11 days by negatively regulating the inflammatory response in spinal microglia. Conclusion: The anti-inflammatory effects of miR 146a-5p along with nanoparticle-based materials make miR NPs promising tools for treating neuropathic pain.
Collapse
Affiliation(s)
- Thuỳ Linh Phạm
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea.,Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
| | - Yuhua Yin
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea.,Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea.,Department of Anesthesia, Guangzhou Women and Children's Medical Center, Guangzhou, 510623, Guangdong Province, PR China
| | - Hyeok Hee Kwon
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea.,Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
| | - Nara Shin
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea.,Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
| | - Song I Kim
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea.,Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
| | - Hyewon Park
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea.,Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
| | - Juhee Shin
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea.,Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
| | - Hyo Jung Shin
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea.,Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
| | - Jeong-Ah Hwang
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea.,Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
| | - Hee-Jung Song
- Department of Neurology, Brain Research Institute, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
| | - Sang Ryong Kim
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Institute of Life Science & Biotechnology, Brain Science & Engineering Institute, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Joo Hyoung Lee
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Patrick T J Hwang
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Ho-Wook Jun
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Dong Woon Kim
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea.,Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
| |
Collapse
|
23
|
Kuthati Y, Navakanth Rao V, Busa P, Tummala S, Davuluri Venkata Naga G, Wong CS. Scope and Applications of Nanomedicines for the Management of Neuropathic Pain. Mol Pharm 2020; 17:1015-1027. [PMID: 32142287 DOI: 10.1021/acs.molpharmaceut.9b01027] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Neuropathic pain, resulting from the dysfunction of the peripheral and central nervous system, occurs in a variety of pathological conditions including trauma, diabetes, cancer, HIV, surgery, multiple sclerosis, ischemic attack, alcoholism, spinal cord damage, and many others. Despite the availability of various treatment strategies, the percentage of patients achieving adequate pain relief remains low. The clinical failure of most effective drugs is often not due to a lack of drug efficacy but due to the dose-limiting central nervous system (CNS) toxicity of the drugs that preclude dose escalation. There is a need for cross-disciplinary collaborations to meet these challenges. In this regard, the integration of nanotechnology with neuroscience is one of the most important fields. In recent years, promising preclinical research has been reported in this field. This review highlights the current challenges associated with conventional neuropathic pain treatments, the scope for nanomaterials in delivering drugs across the blood-brain barrier, and the state and prospects of nanomaterials for the management of neuropathic pain.
Collapse
Affiliation(s)
- Yaswanth Kuthati
- Department of Anesthesiology, Cathy General Hospital, Taipei 280, Taiwan
| | - Vaikar Navakanth Rao
- Institute of Pharmacology and Toxicology, Tzu Chi University, Hualien 970, Taiwan
| | - Prabhakar Busa
- Department of Life Sciences, National Dong Hwa University, Hualien 97401, Taiwan
| | - Srikrishna Tummala
- Department of Chemistry, National Dong Hwa University, Hualien 97401, Taiwan
| | | | - Chih Shung Wong
- Department of Anesthesiology, Cathy General Hospital, Taipei 280, Taiwan.,Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei 280, Taiwan
| |
Collapse
|
24
|
Zhang M, Cheng J, Zhang Y, Kong H, Wang S, Luo J, Qu H, Zhao Y. Green synthesis of Zingiberis rhizoma-based carbon dots attenuates chemical and thermal stimulus pain in mice. Nanomedicine (Lond) 2020; 15:851-869. [PMID: 32238028 DOI: 10.2217/nnm-2019-0369] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Aim: To evaluate the analgesic activity of Zingiberis rhizoma-based carbon dots (ZR-CDs). Materials & methods: Novel ZR-CDs were prepared via a facile, green pyrolysis method. Microstructure, optical and functional group properties were characterized. Acetic acid writhing, hot-plate and tail-immersion tests were performed using mice to evaluate the analgesic activity of ZR-CDs, followed by a preliminary study on the analgesic mechanism. Results: ZR-CDs with a quantum yield of 5.2% had a diameter ranging from 2.23 to 3.77 nm. Remarkable analgesic effect of ZR-CDs was observed against both thermal and chemical stimulus tests, possibly mediated by an opioid-like mechanism and the regulation of 5-hydroxytryptamine levels. Conclusion: ZR-CDs have a promising potential for biomedical application in relieving pain-related diseases.
Collapse
Affiliation(s)
- Meiling Zhang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, PR China
| | - Jinjun Cheng
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, PR China
| | - Yue Zhang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, PR China
| | - Hui Kong
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, PR China
| | - Suna Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, PR China
| | - Juan Luo
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, PR China
| | - Huihua Qu
- Center of Scientific Experiment, Beijing University of Chinese Medicine, Beijing, 100029, PR China
| | - Yan Zhao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, PR China
| |
Collapse
|
25
|
Shin HJ, Park H, Shin N, Kwon HH, Yin Y, Hwang JA, Kim SI, Kim SR, Kim S, Joo Y, Kim Y, Kim J, Beom J, Kim DW. p47phox siRNA-Loaded PLGA Nanoparticles Suppress ROS/Oxidative Stress-Induced Chondrocyte Damage in Osteoarthritis. Polymers (Basel) 2020; 12:polym12020443. [PMID: 32069893 PMCID: PMC7077645 DOI: 10.3390/polym12020443] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 02/07/2020] [Accepted: 02/10/2020] [Indexed: 12/11/2022] Open
Abstract
Osteoarthritis (OA) is the most common joint disorder that has had an increasing prevalence due to the aging of the population. Recent studies have concluded that OA progression is related to oxidative stress and reactive oxygen species (ROS). ROS are produced at low levels in articular chondrocytes, mainly by the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase, and ROS production and oxidative stress have been found to be elevated in patients with OA. The cartilage of OA-affected rat exhibits a significant induction of p47phox, a cytosolic subunit of the NADPH oxidase, similarly to human osteoarthritis cartilage. Therefore, this study tested whether siRNA p47phox that is introduced with poly (D,L-lactic-co-glycolic acid) (PLGA) nanoparticles (p47phox si_NPs) can alleviate chondrocyte cell death by reducing ROS production. Here, we confirm that p47phox si_NPs significantly attenuated oxidative stress and decreased cartilage damage in mono-iodoacetate (MIA)-induced OA. In conclusion, these data suggest that p47phox si_NPs may be of therapeutic value in the treatment of osteoarthritis.
Collapse
Affiliation(s)
- Hyo Jung Shin
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon 35015, Korea; (H.J.S.); (H.P.); (N.S.); (H.H.K.); (Y.Y.); (J.-A.H.); (S.I.K.)
- Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University College of Medicine, Daejeon 35015, Korea;
| | - Hyewon Park
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon 35015, Korea; (H.J.S.); (H.P.); (N.S.); (H.H.K.); (Y.Y.); (J.-A.H.); (S.I.K.)
- Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University College of Medicine, Daejeon 35015, Korea;
| | - Nara Shin
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon 35015, Korea; (H.J.S.); (H.P.); (N.S.); (H.H.K.); (Y.Y.); (J.-A.H.); (S.I.K.)
- Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University College of Medicine, Daejeon 35015, Korea;
| | - Hyeok Hee Kwon
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon 35015, Korea; (H.J.S.); (H.P.); (N.S.); (H.H.K.); (Y.Y.); (J.-A.H.); (S.I.K.)
- Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University College of Medicine, Daejeon 35015, Korea;
| | - Yuhua Yin
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon 35015, Korea; (H.J.S.); (H.P.); (N.S.); (H.H.K.); (Y.Y.); (J.-A.H.); (S.I.K.)
- Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University College of Medicine, Daejeon 35015, Korea;
| | - Jeong-Ah Hwang
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon 35015, Korea; (H.J.S.); (H.P.); (N.S.); (H.H.K.); (Y.Y.); (J.-A.H.); (S.I.K.)
- Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University College of Medicine, Daejeon 35015, Korea;
| | - Song I Kim
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon 35015, Korea; (H.J.S.); (H.P.); (N.S.); (H.H.K.); (Y.Y.); (J.-A.H.); (S.I.K.)
- Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University College of Medicine, Daejeon 35015, Korea;
| | - Sang Ryong Kim
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Institute of Life Science & Biotechnology, Brain Science and Engineering Institute, Kyungpook National University, Daegu 41566, Korea;
| | - Sooil Kim
- Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University College of Medicine, Daejeon 35015, Korea;
| | - Yongbum Joo
- Department of Orthopedics, Chungnam National University College of Medicine, Daejeon 35015, Korea; (Y.J.); (Y.K.)
| | - Youngmo Kim
- Department of Orthopedics, Chungnam National University College of Medicine, Daejeon 35015, Korea; (Y.J.); (Y.K.)
| | - Jinhyun Kim
- Division of Rheumatology, Department of Internal Medicine, Chungnam National University College of Medicine, Daejeon 35015, Korea;
| | - Jaewon Beom
- Department of Physical Medicine and Rehabilitation, Chung-Ang University Hospital, Chung-Ang University College of Medicine, Seoul 06973, Korea;
| | - Dong Woon Kim
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon 35015, Korea; (H.J.S.); (H.P.); (N.S.); (H.H.K.); (Y.Y.); (J.-A.H.); (S.I.K.)
- Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University College of Medicine, Daejeon 35015, Korea;
- Correspondence:
| |
Collapse
|
26
|
Mycophenolate co-administration with quercetin via lipid-polymer hybrid nanoparticles for enhanced breast cancer management. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2020; 24:102147. [DOI: 10.1016/j.nano.2019.102147] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 11/27/2019] [Accepted: 12/15/2019] [Indexed: 01/31/2023]
|
27
|
Yin Y, Park H, Lee SY, Lee WH, Song HJ, Kim J, Kim DW, Hong J. Analgesic Effect of Toll-like Receptor 4 Antagonistic Peptide 2 on Mechanical Allodynia Induced with Spinal Nerve Ligation in Rats. Exp Neurobiol 2019; 28:352-361. [PMID: 31308795 PMCID: PMC6614064 DOI: 10.5607/en.2019.28.3.352] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 05/11/2019] [Accepted: 05/28/2019] [Indexed: 01/06/2023] Open
Abstract
Neuroinflammation is one of the key mechanisms of neuropathic pain, which is primarily mediated by the Toll-like receptor 4 (TLR4) signaling pathways in microglia. Therefore, TLR4 may be a reasonable target for treatment of neuropathic pain. Here, we examined the analgesic effect of TLR4 antagonistic peptide 2 (TAP2) on neuropathic pain induced by spinal nerve ligation in rats. When lipopolysaccharide (LPS)-stimulated BV2 microglia cells were treated with TAP2 (10 µM), the mRNA levels of proinflammatory mediators, such as tumor necrosis factor (TNF)-α, interleukin (IL)-1β, IL-6, cyclooxygenase (COX)-2, and inducible nitric oxide synthase (iNOS), were markedly decreased by 54-83% as determined by quantitative PCR (qPCR) analysis. Furthermore, when TAP2 (25 nmol in 20 µL PBS) was intrathecally administered to the spinal nerve ligation-induced rats on day 3 after surgery, the mechanical allodynia was markedly decreased for approximately 2 weeks in von Frey filament tests, with a reduction in microglial activation. On immunohistochemical and qPCR analyses, both the level of reactive oxygen species and the gene expression of the proinflammatory mediators, such as TNF-α, IL-1β, IL-6, COX-2, and iNOS, were significantly decreased in the ipsilateral spinal dorsal horn. Finally, the analgesic effect of TAP2 was reproduced in rats with monoiodoacetate-induced osteoarthritic pain. The findings of the present study suggest that TAP2 efficiently mitigates neuropathic pain behavior by suppressing microglial activation, followed by downregulation of neuropathic pain-related factors, such as reactive oxygen species and proinflammatory molecules. Therefore, it may be useful as a new analgesic for treatment of neuropathic pain.
Collapse
Affiliation(s)
- Yuhua Yin
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon 35015, Korea.,Department of Anatomy, Brain Research Institute, Chungnam National University School of Medicine, Daejeon 35015, Korea
| | - Hyewon Park
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon 35015, Korea.,Department of Anatomy, Brain Research Institute, Chungnam National University School of Medicine, Daejeon 35015, Korea
| | - Sun Yeul Lee
- Department of Anesthesia and Pain Medicine, Chungnam National University Hospital, Daejeon 35015, Korea
| | - Won-Hyung Lee
- Department of Anesthesia and Pain Medicine, Chungnam National University Hospital, Daejeon 35015, Korea
| | - Hee-Jung Song
- Department of Neurology, Chungnam National University Hospital, Daejeon 35015, Korea
| | - Jinhyun Kim
- Department of Anesthesia and Pain Medicine, Chungnam National University Hospital, Daejeon 35015, Korea
| | - Dong Woon Kim
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon 35015, Korea.,Department of Anatomy, Brain Research Institute, Chungnam National University School of Medicine, Daejeon 35015, Korea
| | - Jinpyo Hong
- Department of Anatomy, Brain Research Institute, Chungnam National University School of Medicine, Daejeon 35015, Korea
| |
Collapse
|