1
|
Ciechanowska A, Mika J. CC Chemokine Family Members' Modulation as a Novel Approach for Treating Central Nervous System and Peripheral Nervous System Injury-A Review of Clinical and Experimental Findings. Int J Mol Sci 2024; 25:3788. [PMID: 38612597 PMCID: PMC11011591 DOI: 10.3390/ijms25073788] [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: 02/05/2024] [Revised: 03/18/2024] [Accepted: 03/27/2024] [Indexed: 04/14/2024] Open
Abstract
Despite significant progress in modern medicine and pharmacology, damage to the nervous system with various etiologies still poses a challenge to doctors and scientists. Injuries lead to neuroimmunological changes in the central nervous system (CNS), which may result in both secondary damage and the development of tactile and thermal hypersensitivity. In our review, based on the analysis of many experimental and clinical studies, we indicate that the mechanisms occurring both at the level of the brain after direct damage and at the level of the spinal cord after peripheral nerve damage have a common immunological basis. This suggests that there are opportunities for similar pharmacological therapeutic interventions in the damage of various etiologies. Experimental data indicate that after CNS/PNS damage, the levels of 16 among the 28 CC-family chemokines, i.e., CCL1, CCL2, CCL3, CCL4, CCL5, CCL6, CCL7, CCL8, CCL9, CCL11, CCL12, CCL17, CCL19, CCL20, CCL21, and CCL22, increase in the brain and/or spinal cord and have strong proinflammatory and/or pronociceptive effects. According to the available literature data, further investigation is still needed for understanding the role of the remaining chemokines, especially six of them which were found in humans but not in mice/rats, i.e., CCL13, CCL14, CCL15, CCL16, CCL18, and CCL23. Over the past several years, the results of studies in which available pharmacological tools were used indicated that blocking individual receptors, e.g., CCR1 (J113863 and BX513), CCR2 (RS504393, CCX872, INCB3344, and AZ889), CCR3 (SB328437), CCR4 (C021 and AZD-2098), and CCR5 (maraviroc, AZD-5672, and TAK-220), has beneficial effects after damage to both the CNS and PNS. Recently, experimental data have proved that blockades exerted by double antagonists CCR1/3 (UCB 35625) and CCR2/5 (cenicriviroc) have very good anti-inflammatory and antinociceptive effects. In addition, both single (J113863, RS504393, SB328437, C021, and maraviroc) and dual (cenicriviroc) chemokine receptor antagonists enhanced the analgesic effect of opioid drugs. This review will display the evidence that a multidirectional strategy based on the modulation of neuronal-glial-immune interactions can significantly improve the health of patients after CNS and PNS damage by changing the activity of chemokines belonging to the CC family. Moreover, in the case of pain, the combined administration of such antagonists with opioid drugs could reduce therapeutic doses and minimize the risk of complications.
Collapse
Affiliation(s)
| | - Joanna Mika
- Department of Pain Pharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, 12 Smetna Str., 31-343 Kraków, Poland;
| |
Collapse
|
2
|
Johnbosco C, Karbaat L, Korthagen NM, Warmink K, Koerselman M, Coeleveld K, Becker M, van Loo B, Zoetebier B, Both S, Weinans H, Karperien M, Leijten J. Microencapsulated stem cells reduce cartilage damage in a material dependent manner following minimally invasive intra-articular injection in an OA rat model. Mater Today Bio 2023; 22:100791. [PMID: 37731960 PMCID: PMC10507156 DOI: 10.1016/j.mtbio.2023.100791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 08/05/2023] [Accepted: 09/04/2023] [Indexed: 09/22/2023] Open
Abstract
Osteoarthritis (OA) is a degenerative disease of the joints for which no curative treatment exists. Intra-articular injection of stem cells is explored as a regenerative approach, but rapid clearance of cells from the injection site limits the therapeutic outcome. Microencapsulation of mesenchymal stem cells (MSCs) can extend the retention time of MSCs, but the outcomes of the few studies currently performed are conflicting. We hypothesize that the composition of the micromaterial's shell plays a deciding factor in the treatment outcome of intra-articular MSC injection. To this end, we microencapsulate MSCs using droplet microfluidic generators in flow-focus mode using various polymers and polymer concentrations. We demonstrate that polymer composition and concentration potently alter the metabolic activity as well as the secretome of MSCs. Moreover, while microencapsulation consistently prolongs the retention time of MSC injected in rat joints, distinct biodistribution within the joint is demonstrated for the various microgel formulations. Furthermore, intra-articular injections of pristine and microencapsulated MSC in OA rat joints show a strong material-dependent effect on the reduction of cartilage degradation and matrix loss. Collectively, this study highlights that micromaterial composition and concentration are key deciding factors for the therapeutic outcome of intra-articular injections of microencapsulated stem cells to treat degenerative joint diseases.
Collapse
Affiliation(s)
- Castro Johnbosco
- Department of Developmental BioEngineering, TechMed Centre, University of Twente, the Netherlands
| | - Lisanne Karbaat
- Department of Developmental BioEngineering, TechMed Centre, University of Twente, the Netherlands
| | - Nicoline M. Korthagen
- Faculty of Veterinary Sciences Department of equine sciences, University of Utrecht, the Netherlands
- Department of Orthopaedics, University Medical Centre Utrecht, the Netherlands
| | - Kelly Warmink
- Department of Orthopaedics, University Medical Centre Utrecht, the Netherlands
| | - Michelle Koerselman
- Department of Developmental BioEngineering, TechMed Centre, University of Twente, the Netherlands
| | - Katja Coeleveld
- Department of Rheumatology & Clinical Immunology, University Medical Centre Utrecht, the Netherlands
| | - Malin Becker
- Department of Developmental BioEngineering, TechMed Centre, University of Twente, the Netherlands
| | - Bas van Loo
- Department of Developmental BioEngineering, TechMed Centre, University of Twente, the Netherlands
| | - Bram Zoetebier
- Department of Developmental BioEngineering, TechMed Centre, University of Twente, the Netherlands
| | - Sanne Both
- Department of Developmental BioEngineering, TechMed Centre, University of Twente, the Netherlands
| | - Harrie Weinans
- Department of Orthopaedics, University Medical Centre Utrecht, the Netherlands
| | - Marcel Karperien
- Department of Developmental BioEngineering, TechMed Centre, University of Twente, the Netherlands
| | - Jeroen Leijten
- Department of Developmental BioEngineering, TechMed Centre, University of Twente, the Netherlands
| |
Collapse
|
3
|
Lenders V, Koutsoumpou X, Phan P, Soenen SJ, Allegaert K, de Vleeschouwer S, Toelen J, Zhao Z, Manshian BB. Modulation of engineered nanomaterial interactions with organ barriers for enhanced drug transport. Chem Soc Rev 2023; 52:4672-4724. [PMID: 37338993 DOI: 10.1039/d1cs00574j] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/22/2023]
Abstract
The biomedical use of nanoparticles (NPs) has been the focus of intense research for over a decade. As most NPs are explored as carriers to alter the biodistribution, pharmacokinetics and bioavailability of associated drugs, the delivery of these NPs to the tissues of interest remains an important topic. To date, the majority of NP delivery studies have used tumor models as their tool of interest, and the limitations concerning tumor targeting of systemically administered NPs have been well studied. In recent years, the focus has also shifted to other organs, each presenting their own unique delivery challenges to overcome. In this review, we discuss the recent advances in leveraging NPs to overcome four major biological barriers including the lung mucus, the gastrointestinal mucus, the placental barrier, and the blood-brain barrier. We define the specific properties of these biological barriers, discuss the challenges related to NP transport across them, and provide an overview of recent advances in the field. We discuss the strengths and shortcomings of different strategies to facilitate NP transport across the barriers and highlight some key findings that can stimulate further advances in this field.
Collapse
Affiliation(s)
- Vincent Lenders
- Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Herestraat 49, B3000 Leuven, Belgium.
| | - Xanthippi Koutsoumpou
- Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Herestraat 49, B3000 Leuven, Belgium.
| | - Philana Phan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Stefaan J Soenen
- Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Herestraat 49, B3000 Leuven, Belgium.
- NanoHealth and Optical Imaging Group, Department of Imaging and Pathology, KU Leuven, Herestraat 49, B3000 Leuven, Belgium
| | - Karel Allegaert
- Department of Hospital Pharmacy, Erasmus MC University Medical Center, CN Rotterdam, 3015, The Netherlands
- Clinical Pharmacology and Pharmacotherapy, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, B3000 Leuven, Belgium
- Leuven Child and Youth Institute, KU Leuven, 3000 Leuven, Belgium
- Woman and Child, Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
| | - Steven de Vleeschouwer
- Department of Neurosurgery, University Hospitals Leuven, Leuven, Belgium
- Laboratory of Experimental Neurosurgery and Neuroanatomy, Department of Neurosciences, KU Leuven, Leuven, Belgium
- Leuven Brain Institute (LBI), KU Leuven, Leuven, Belgium
| | - Jaan Toelen
- Leuven Child and Youth Institute, KU Leuven, 3000 Leuven, Belgium
- Woman and Child, Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
- Department of Pediatrics, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Zongmin Zhao
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Bella B Manshian
- Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Herestraat 49, B3000 Leuven, Belgium.
| |
Collapse
|
4
|
Mohammed FS, Omay SB, Sheth KN, Zhou J. Nanoparticle-based drug delivery for the treatment of traumatic brain injury. Expert Opin Drug Deliv 2023; 20:55-73. [PMID: 36420918 PMCID: PMC9983310 DOI: 10.1080/17425247.2023.2152001] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 10/10/2022] [Accepted: 11/22/2022] [Indexed: 11/25/2022]
Abstract
INTRODUCTION Traumatic brain injuries (TBIs) impact the breadth of society and remain without any approved pharmacological treatments. Despite successful Phase II clinical trials, the failure of many Phase III clinical trials may be explained by insufficient drug targeting and retention, preventing the proper attainment of an observable dosage threshold. To address this challenge, nanoparticles can be functionalized to protect pharmacological payloads, improve targeted drug delivery to sites of injury, and can be combined with supportive scaffolding to improve secondary outcomes. AREAS COVERED This review briefly covers the pathophysiology of TBIs and their subtypes, the current pre-clinical and clinical management strategies, explores the common models of focal, diffuse, and mixed traumatic brain injury employed in experimental animals, and surveys the existing literature on nanoparticles developed to treat TBIs. EXPERT OPINION Nanoparticles are well suited to improve secondary outcomes as their multifunctionality and customizability enhance their potential for efficient targeted delivery, payload protection, increased brain penetration, low off-target toxicity, and biocompatibility in both acute and chronic timescales.
Collapse
Affiliation(s)
- Farrah S. Mohammed
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut, USA
| | - Sacit Bulent Omay
- Department of Neurosurgery, Yale University, New Haven, Connecticut, USA
| | - Kevin N. Sheth
- Department of Neurosurgery, Yale University, New Haven, Connecticut, USA
- Department of Neurology, Yale University, New Haven, Connecticut, USA
| | - Jiangbing Zhou
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut, USA
- Department of Neurosurgery, Yale University, New Haven, Connecticut, USA
| |
Collapse
|
5
|
Rasiah PK, Jha KA, Gentry J, Del Mar NA, Townsend T, Torgbe KE, Reiner A, Gangaraju R. A Long-Term Safety and Efficacy Report on Intravitreal Delivery of Adipose Stem Cells and Secretome on Visual Deficits After Traumatic Brain Injury. Transl Vis Sci Technol 2022; 11:1. [PMID: 36180031 PMCID: PMC9547363 DOI: 10.1167/tvst.11.10.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 08/30/2022] [Indexed: 11/24/2022] Open
Abstract
Purpose We compared intravitreal injection of human adipose stem cell concentrated conditioned media (ASC-CCM) to injection of live ASCs for their long-term safety and effectiveness against the visual deficits of mild traumatic brain injury (mTBI). Methods We first tested different intravitreal ASC doses for safety. Other C57BL/6 mice then received focal cranial blast mTBI and were injected with the safe ASC dose (1000 cells/eye), ASC-CCM (∼200 ng protein/eye), or saline solution. At five and 10 months after blast injury, visual, molecular, and histological assessments evaluated treatment efficacy. Histological evaluation of eyes and other organs at 10 months after blast injury assessed safety. Results Human ASCs at 1000 cells/eye were found to be safe, with >10,000 cells causing retinal damage. Blast-injured mice showed significant vision deficits compared to sham blast mice up to 10 months. Blast mice receiving ASC or ASC-CCM showed improved vision at five months but marginal effects at 10 months, correlated with changes in glial fibrillary acidic protein and proinflammatory gene expression in retina. Immunostaining for human IgG failed to detect ASCs in retina. Peripheral organs examined histologically at 10 months after blast injury were normal. Conclusions Intravitreal injection of ASCs or ASC-CCM is safe and effective against the visual deficits of mTBI. Considering the unimproved glial response and the risk of retinal damage with live cells, our studies suggest that ASC-CCM has better safety and effectiveness than live cells for the treatment of visual dysfunction in mTBI. Translational Relevance This study demonstrates the safety and efficacy of mesenchymal stem cell-based therapeutics, supporting them for phase 1 clinical studies.
Collapse
Affiliation(s)
- Pratheepa Kumari Rasiah
- Department of Ophthalmology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Kumar Abhiram Jha
- Department of Ophthalmology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Jordy Gentry
- Department of Ophthalmology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Nobel A. Del Mar
- Department of Anatomy & Neurobiology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Tanisha Townsend
- Department of Pathology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Kwame E. Torgbe
- Department of Pathology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Anton Reiner
- Department of Anatomy & Neurobiology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Rajashekhar Gangaraju
- Department of Ophthalmology, University of Tennessee Health Science Center, Memphis, TN, USA
- Department of Anatomy & Neurobiology, University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
6
|
Palan F, Chatterjee B. Dendrimers in the context of targeting central nervous system disorders. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103474] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
7
|
Hac NEF, Gold DR. Neuro-Visual and Vestibular Manifestations of Concussion and Mild TBI. Curr Neurol Neurosci Rep 2022; 22:219-228. [PMID: 35235169 DOI: 10.1007/s11910-022-01184-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/04/2022] [Indexed: 11/27/2022]
Abstract
PURPOSE OF REVIEW Mild traumatic brain injury, or concussion, is a major cause of disability. Vestibular and visual dysfunction following concussion is common and can negatively affect patients' well-being and prolong recovery. Etiologies of visual and vestibular symptoms are numerous, including ocular, neuro-ophthalmic, otologic, and neuro-vestibular conditions. Some etiologies are benign and may be treatable, while others are potentially vision or life-threatening, making a focused history and examination essential. This review offers an approach to the evaluation and treatment of the most common neuro-visual and vestibular impairments that may result from concussion. RECENT FINDINGS Treatment of concussion including exercise, computerized programs, transcranial magnetic stimulation, gene therapy, stem cell therapy, and nanoparticles has shown promise. Many novel therapies are in the pipework for visual and vestibular recovery after concussion; however, the treatment mainstay remains therapy and evaluation for co-existing diseases.
Collapse
Affiliation(s)
- Nicholas E F Hac
- Department of Neurology, The Johns Hopkins School of Medicine, 600 N Wolfe St, Path 2-210, Baltimore, MD, 21287, USA.
| | - Daniel R Gold
- Department of Neurology, The Johns Hopkins School of Medicine, 600 N Wolfe St, Path 2-210, Baltimore, MD, 21287, USA
| |
Collapse
|
8
|
Kim CK, Sachdev PS, Braidy N. Recent Neurotherapeutic Strategies to Promote Healthy Brain Aging: Are we there yet? Aging Dis 2022; 13:175-214. [PMID: 35111369 PMCID: PMC8782556 DOI: 10.14336/ad.2021.0705] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 07/05/2021] [Indexed: 12/21/2022] Open
Abstract
Owing to the global exponential increase in population ageing, there is an urgent unmet need to develop reliable strategies to slow down and delay the ageing process. Age-related neurodegenerative diseases are among the main causes of morbidity and mortality in our contemporary society and represent a major socio-economic burden. There are several controversial factors that are thought to play a causal role in brain ageing which are continuously being examined in experimental models. Among them are oxidative stress and brain inflammation which are empirical to brain ageing. Although some candidate drugs have been developed which reduce the ageing phenotype, their clinical translation is limited. There are several strategies currently in development to improve brain ageing. These include strategies such as caloric restriction, ketogenic diet, promotion of cellular nicotinamide adenine dinucleotide (NAD+) levels, removal of senescent cells, 'young blood' transfusions, enhancement of adult neurogenesis, stem cell therapy, vascular risk reduction, and non-pharmacological lifestyle strategies. Several studies have shown that these strategies can not only improve brain ageing by attenuating age-related neurodegenerative disease mechanisms, but also maintain cognitive function in a variety of pre-clinical experimental murine models. However, clinical evidence is limited and many of these strategies are awaiting findings from large-scale clinical trials which are nascent in the current literature. Further studies are needed to determine their long-term efficacy and lack of adverse effects in various tissues and organs to gain a greater understanding of their potential beneficial effects on brain ageing and health span in humans.
Collapse
Affiliation(s)
- Chul-Kyu Kim
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, Australia
| | - Perminder S Sachdev
- Neuropsychiatric Institute, Euroa Centre, Prince of Wales Hospital, Sydney, Australia
| | - Nady Braidy
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, Australia
| |
Collapse
|
9
|
Chis AA, Dobrea CM, Rus LL, Frum A, Morgovan C, Butuca A, Totan M, Juncan AM, Gligor FG, Arseniu AM. Dendrimers as Non-Viral Vectors in Gene-Directed Enzyme Prodrug Therapy. Molecules 2021; 26:5976. [PMID: 34641519 PMCID: PMC8512881 DOI: 10.3390/molecules26195976] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 09/19/2021] [Accepted: 09/29/2021] [Indexed: 01/02/2023] Open
Abstract
Gene-directed enzyme prodrug therapy (GDEPT) has been intensively studied as a promising new strategy of prodrug delivery, with its main advantages being represented by an enhanced efficacy and a reduced off-target toxicity of the active drug. In recent years, numerous therapeutic systems based on GDEPT strategy have entered clinical trials. In order to deliver the desired gene at a specific site of action, this therapeutic approach uses vectors divided in two major categories, viral vectors and non-viral vectors, with the latter being represented by chemical delivery agents. There is considerable interest in the development of non-viral vectors due to their decreased immunogenicity, higher specificity, ease of synthesis and greater flexibility for subsequent modulations. Dendrimers used as delivery vehicles offer many advantages, such as: nanoscale size, precise molecular weight, increased solubility, high load capacity, high bioavailability and low immunogenicity. The aim of the present work was to provide a comprehensive overview of the recent advances regarding the use of dendrimers as non-viral carriers in the GDEPT therapy.
Collapse
Affiliation(s)
| | | | | | - Adina Frum
- Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (A.A.C.); (C.M.D.); (L.-L.R.); (A.B.); (M.T.); (A.M.J.); (F.G.G.); (A.M.A.)
| | - Claudiu Morgovan
- Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (A.A.C.); (C.M.D.); (L.-L.R.); (A.B.); (M.T.); (A.M.J.); (F.G.G.); (A.M.A.)
| | | | | | | | | | | |
Collapse
|
10
|
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: 28] [Impact Index Per Article: 9.3] [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
|