1
|
Hassanzadeh-Khanmiri M, Moshari A, Kheradmand R, Haghgouei T, Homaei M, Charsouei S, Mobed A. Nanomedicine: a cost-effective and powerful platform for managing neurodegenerative diseases. Metab Brain Dis 2025; 40:142. [PMID: 40067468 DOI: 10.1007/s11011-025-01564-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Accepted: 02/28/2025] [Indexed: 03/26/2025]
Abstract
Neurodegenerative diseases (NDDs) are characterized by the chronic and progressive deterioration of the structure and function of the nervous system, imposing a significant burden on patients, their families, and society. These diseases have a gradual onset and continually worsen, making early diagnosis challenging. Current drugs on the market struggle to effectively cross the blood-brain barrier (BBB), leading to poor outcomes and limited therapeutic success. Consequently, there is an urgent need for new diagnostic tools and treatment strategies. To address these challenges, nanotechnology-based drug delivery systems-such as liposomes, micelles, dendrimers, and solid lipid nanoparticles (SLNs)-have emerged as promising solutions. This study provides a comprehensive review of recent advances in nanomedicine and nanotechnology-based platforms, alongside an exploration of ND mechanisms. The authors conducted a systematic literature search across relevant databases such as PubMed, Scopus, and Web of Science, focusing on peer-reviewed articles, reviews, and clinical studies published within the last 5 to 10 years. Additionally, this paper addresses the challenges faced by nanomedicines and delivery systems, offering insights into future directions in the field and the need for further research to establish their clinical viability as alternatives to current therapies.
Collapse
Affiliation(s)
| | - Amirreza Moshari
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Reza Kheradmand
- Division of Pharmacology and Physiology, Department of Basic Sciences, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Tannaz Haghgouei
- Division of Pharmacology and Physiology, Department of Basic Sciences, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Maryam Homaei
- Department of Physiology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Division of Pharmacology and Physiology, Department of Basic Sciences, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
- Tabriz Neuroscience Research Center (NRSC), Neurology Department, Tabriz University of Medical Sciences, Tabriz, Iran
- Social Determinants of Health Research Center, Health Management and Safety Promotion, Tabriz, Iran
| | - Saeid Charsouei
- Tabriz Neuroscience Research Center (NRSC), Neurology Department, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Ahmad Mobed
- Social Determinants of Health Research Center, Health Management and Safety Promotion, Tabriz, Iran.
| |
Collapse
|
2
|
Cai X, Xie Z, Zhao J, Lu W, Zhu Z, Chen M, Huang Z, Ying Y, Fu Y, Xu J, Zhu S. FGF20 promotes spinal cord injury repair by inhibiting the formation of necrotic corpuscle P-MLKL/P-RIP1/P-RIP3 in neurons. J Cell Mol Med 2024; 28:e70109. [PMID: 39676730 DOI: 10.1111/jcmm.70109] [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: 11/23/2023] [Revised: 09/11/2024] [Accepted: 09/13/2024] [Indexed: 12/17/2024] Open
Abstract
The disruption of the local microenvironment subsequent to spinal cord injury (SCI) leads to a substantial loss of neurons in the affected region, which is a major contributing factor to impaired motor function recovery in patients. Fibroblast growth factor 20 (FGF20) is a neurotrophic factor that plays a crucial role in neuronal development and homeostasis. In this study, the recombinant human FGF20 (rhFGF20) was found to mitigate the process of necroptosis in a mouse model of SCI, thereby reducing neural functional deficits and promoting SCI repair. FGF20 protein was injected into the SCI mice via intraperitoneal injection. Using the BMS scale and inclined plane test, we found that FGF20 significantly promoted the recovery of motor function. The Nissl staining revealed the level of neuronal survival within the region of injury. The expression changes of NeuN, GAP43, NF200 and GFAP indicated that FGF20 has the nerve repair ability to delay the formation of glial scar. Through fluorescence detection of Ace-Tubulin and Tyr-Tubulin, FGF20 was revealed to promote the polymerization of axon-regenerated microtubules. Furthermore, FGF20 was also found to reduce the expression levels of necroptosis induced by SCI. These data suggest that FGF20 may exert a neuroprotective effect by inhibiting injury-induced necroptosis, thereby facilitating functional recovery following SCI. Moreover, systemic administration of FGF20 holds promise as a potential therapeutic strategy for repairing the damaged spinal cord. The discovery paves the way for a novel avenue of growth factor research in the field of SCI.
Collapse
Affiliation(s)
- Xiong Cai
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhenwen Xie
- The First Clinical School of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Juan Zhao
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Wenjie Lu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhongwei Zhu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Min Chen
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhiyang Huang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yibo Ying
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yining Fu
- The First Clinical School of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jiake Xu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
| | - Sipin Zhu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
3
|
Zhao K, Xu J, Zhao B. Panoramic RNA expression of fibroblast growth factors in human glioblastoma tissues and the impact on the survival of patients. Oncol Lett 2024; 28:317. [PMID: 38807663 PMCID: PMC11130607 DOI: 10.3892/ol.2024.14450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 04/26/2024] [Indexed: 05/30/2024] Open
Abstract
Fibroblast growth factors (FGFs) have a key role in various critical steps of tumor growth and progression through effects on angiogenesis, inflammation and the growth and invasion of malignant cells. Nevertheless, the role of the FGF family in human glioblastoma (GBM) has been rarely studied. The objective of the present study was to assess the RNA expression of all FGF family members in tissues obtained from patients with GBM and to analyze the association between FGF expression and the survival of these patients. For this, the RNA expression of FGF family members in the malignant and proximal tissues of 12 patients with GBM was determined by analyzing high-throughput RNA transcriptome sequencing data uploaded to the National Center for Biotechnology Information database. The relationship between FGF genes and the survival of patients with GBM and glioma was also respectively studied by analyzing data from The Cancer Genome Atlas database using the Gene Expression Profiling Interactive Analysis tool. The results showed that the expression of FGF1, FGF17, FGF20 and FGF22 in GBM tissues was lower than that in adjacent tissues, with a difference of >2 times. Analysis of the overall survival of patients with GBM indicated there were no significant relationships between the expression of FGF1, FGF17, FGF20, FGF22 and overall survival. Analysis of the overall survival of patients with glioma showed that glioma patients with low FGF1 expression achieved a longer survival time than patients with high FGF1 expression; however, high expression of FGF17 and FGF22 indicated a longer survival time. In summary, the results of the present study demonstrated the panoramic expression of FGF family members in patients with GBM, and indicated that FGF1, FGF17 and FGF22 did not affect the survival of patients with GBM, but had a notable influence on the survival of patients with glioma.
Collapse
Affiliation(s)
- Kun Zhao
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Jiakun Xu
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Beichuan Zhao
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| |
Collapse
|
4
|
Cai H, Liu D, Xue WW, Ma L, Xie HT, Ning K. Lipid-based nanoparticles for drug delivery in Parkinson's disease. Transl Neurosci 2024; 15:20220359. [PMID: 39654878 PMCID: PMC11627081 DOI: 10.1515/tnsci-2022-0359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 10/01/2024] [Accepted: 10/07/2024] [Indexed: 12/12/2024] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder that predominantly affects dopaminergic neurons in the substantia nigra and ventral tegmental area, resulting in symptoms such as tremors, muscle rigidity, bradykinesia, and potential cognitive and affective disturbances. The effective delivery of pharmacological agents to the central nervous system is hindered by various factors, including the restrictive properties of the blood‒brain barrier and blood‒spinal cord barrier, as well as the physicochemical characteristics of the drugs. Traditional drug delivery methods may not provide the therapeutic concentrations necessary for functional restoration in PD patients. However, lipid-based nanoparticles (NPs) offer new possibilities for enhancing the bioavailability of established treatment regimens and developing innovative therapies that can modify the course of the disease. This review provides a concise overview of recent advances in lipid-based NP strategies aimed at mitigating specific pathological mechanisms relevant to PD progression. This study also explores the potential applications of nanotechnological innovations in the development of advanced treatment modalities for individuals with PD.
Collapse
Affiliation(s)
- Han Cai
- Guangdong Celconta Biotechnology Co., Ltd, 9 Xincheng Road, Songshan Lake Park, Dongguan, Guangdong, PR China
| | - Dong Liu
- Guangdong Celconta Biotechnology Co., Ltd, 9 Xincheng Road, Songshan Lake Park, Dongguan, Guangdong, PR China
| | - Wei-Wei Xue
- Guangdong Celconta Biotechnology Co., Ltd, 9 Xincheng Road, Songshan Lake Park, Dongguan, Guangdong, PR China
- School of Biological Engineering, Dalian Polytechnic University, Dalian, 116034, China
| | - Liya Ma
- Guangdong Celconta Biotechnology Co., Ltd, 9 Xincheng Road, Songshan Lake Park, Dongguan, Guangdong, PR China
| | - Hai-Tao Xie
- Guangdong Celconta Biotechnology Co., Ltd, 9 Xincheng Road, Songshan Lake Park, Dongguan, Guangdong, PR China
| | - Ke Ning
- Guangdong Celconta Biotechnology Co., Ltd, 9 Xincheng Road, Songshan Lake Park, Dongguan, Guangdong, PR China
- Sheffield Institute of Translational Neuroscience (SITraN), University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
5
|
Muksuris K, Scarisbrick DM, Mahoney JJ, Cherkasova MV. Noninvasive Neuromodulation in Parkinson's Disease: Insights from Animal Models. J Clin Med 2023; 12:5448. [PMID: 37685514 PMCID: PMC10487610 DOI: 10.3390/jcm12175448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 08/14/2023] [Accepted: 08/17/2023] [Indexed: 09/10/2023] Open
Abstract
The mainstay treatments for Parkinson's Disease (PD) have been limited to pharmacotherapy and deep brain stimulation. While these interventions are helpful, a new wave of research is investigating noninvasive neuromodulation methods as potential treatments. Some promising avenues have included transcranial magnetic stimulation (TMS), transcranial direct current stimulation (tDCS), electroconvulsive therapy (ECT), and focused ultrasound (FUS). While these methods are being tested in PD patients, investigations in animal models of PD have sought to elucidate their therapeutic mechanisms. In this rapid review, we assess the available animal literature on these noninvasive techniques and discuss the possible mechanisms mediating their therapeutic effects based on these findings.
Collapse
Affiliation(s)
- Katherine Muksuris
- Department of Psychology, West Virginia University, Morgantown, WV 26506, USA
| | - David M. Scarisbrick
- Department of Behavioral Medicine and Psychiatry, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA
| | - James J. Mahoney
- Department of Behavioral Medicine and Psychiatry, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA
| | - Mariya V. Cherkasova
- Department of Psychology, West Virginia University, Morgantown, WV 26506, USA
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA
| |
Collapse
|
6
|
Michna A, Pomorska A, Ozcan O. Biocompatible Macroion/Growth Factor Assemblies for Medical Applications. Biomolecules 2023; 13:biom13040609. [PMID: 37189357 DOI: 10.3390/biom13040609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 03/24/2023] [Accepted: 03/26/2023] [Indexed: 03/31/2023] Open
Abstract
Growth factors are a class of proteins that play a role in the proliferation (the increase in the number of cells resulting from cell division) and differentiation (when a cell undergoes changes in gene expression becoming a more specific type of cell) of cells. They can have both positive (accelerating the normal healing process) and negative effects (causing cancer) on disease progression and have potential applications in gene therapy and wound healing. However, their short half-life, low stability, and susceptibility to degradation by enzymes at body temperature make them easily degradable in vivo. To improve their effectiveness and stability, growth factors require carriers for delivery that protect them from heat, pH changes, and proteolysis. These carriers should also be able to deliver the growth factors to their intended destination. This review focuses on the current scientific literature concerning the physicochemical properties (such as biocompatibility, high affinity for binding growth factors, improved bioactivity and stability of the growth factors, protection from heat, pH changes or appropriate electric charge for growth factor attachment via electrostatic interactions) of macroions, growth factors, and macroion-growth factor assemblies, as well as their potential uses in medicine (e.g., diabetic wound healing, tissue regeneration, and cancer therapy). Specific attention is given to three types of growth factors: vascular endothelial growth factors, human fibroblast growth factors, and neurotrophins, as well as selected biocompatible synthetic macroions (obtained through standard polymerization techniques) and polysaccharides (natural macroions composed of repeating monomeric units of monosaccharides). Understanding the mechanisms by which growth factors bind to potential carriers could lead to more effective delivery methods for these proteins, which are of significant interest in the diagnosis and treatment of neurodegenerative and civilization diseases, as well as in the healing of chronic wounds.
Collapse
|
7
|
Zhong YX, Liao JC, Liu X, Tian H, Deng LR, Long L. Low intensity focused ultrasound: a new prospect for the treatment of Parkinson's disease. Ann Med 2023; 55:2251145. [PMID: 37634059 PMCID: PMC10461511 DOI: 10.1080/07853890.2023.2251145] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 07/17/2023] [Accepted: 08/20/2023] [Indexed: 08/28/2023] Open
Abstract
Background: As a chronic and progressive neurodegenerative disease, Parkinson's disease (PD) still lacks effective and safe targeted drug therapy. Low-intensity focused ultrasound (LIFU), a new method to stimulate the brain and open the blood-brain barrier (BBB), has been widely concerned by PD researchers due to its non-invasive characteristics.Methods: PubMed was searched for the past 10 years using the terms 'focused ultrasound', 'transcranial ultrasound', 'pulse ultrasound', and 'Parkinson's disease'. Relevant citations were selected from the authors' references. After excluding articles describing high-intensity focused ultrasound or non-Parkinson's disease applications, we found more than 100 full-text analyses for pooled analysis.Results: Current preclinical studies have shown that LIFU could improve PD motor symptoms by regulating microglia activation, increasing neurotrophic factors, reducing oxidative stress, and promoting nerve repair and regeneration, while LIFU combined with microbubbles (MBs) can promote drugs to cross the BBB, which may become a new direction of PD treatment. Therefore, finding an efficient drug carrier system is the top priority of applying LIFU with MBs to deliver drugs.Conclusions: This article aims to review neuro-modulatory effect of LIFU and the possible biophysical mechanism in the treatment of PD, summarize the latest progress in delivering vehicles with MBs, and discuss its advantages and limitations.
Collapse
Affiliation(s)
- Yun-Xiao Zhong
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jin-Chi Liao
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xv Liu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Hao Tian
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Li-Ren Deng
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Ling Long
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
8
|
Prakash N. Developmental pathways linked to the vulnerability of adult midbrain dopaminergic neurons to neurodegeneration. Front Mol Neurosci 2022; 15:1071731. [PMID: 36618829 PMCID: PMC9815185 DOI: 10.3389/fnmol.2022.1071731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 12/05/2022] [Indexed: 12/24/2022] Open
Abstract
The degeneration of dopaminergic and other neurons in the aging brain is considered a process starting well beyond the infantile and juvenile period. In contrast to other dopamine-associated neuropsychiatric disorders, such as schizophrenia and drug addiction, typically diagnosed during adolescence or young adulthood and, thus, thought to be rooted in the developing brain, Parkinson's Disease (PD) is rarely viewed as such. However, evidences have accumulated suggesting that several factors might contribute to an increased vulnerability to death of the dopaminergic neurons at an already very early (developmental) phase in life. Despite the remarkable ability of the brain to compensate such dopamine deficits, the early loss or dysfunction of these neurons might predispose an individual to suffer from PD because the critical threshold of dopamine function will be reached much earlier in life, even if the time-course and strength of naturally occurring and age-dependent dopaminergic cell death is not markedly altered in this individual. Several signaling and transcriptional pathways required for the proper embryonic development of the midbrain dopaminergic neurons, which are the most affected in PD, either continue to be active in the adult mammalian midbrain or are reactivated at the transition to adulthood and under neurotoxic conditions. The persistent activity of these pathways often has neuroprotective functions in adult midbrain dopaminergic neurons, whereas the reactivation of silenced pathways under pathological conditions can promote the survival and even regeneration of these neurons in the lesioned or aging brain. This article summarizes our current knowledge about signaling and transcription factors involved in midbrain dopaminergic neuron development, whose reduced gene dosage or signaling activity are implicated in a lower survival rate of these neurons in the postnatal or aging brain. It also discusses the evidences supporting the neuroprotection of the midbrain dopaminergic system after the external supply or ectopic expression of some of these secreted and nuclear factors in the adult and aging brain. Altogether, the timely monitoring and/or correction of these signaling and transcriptional pathways might be a promising approach to a much earlier diagnosis and/or prevention of PD.
Collapse
|
9
|
Gu Z, Yin Z, Song P, Wu Y, He Y, Zhu M, Wu Z, Zhao S, Huang H, Wang H, Tong C, Qi Z. Safety and biodistribution of exosomes derived from human induced pluripotent stem cells. Front Bioeng Biotechnol 2022; 10:949724. [PMID: 36091443 PMCID: PMC9461140 DOI: 10.3389/fbioe.2022.949724] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Accepted: 07/27/2022] [Indexed: 12/03/2022] Open
Abstract
As a new cell-free therapy, exosomes have provided new ideas for the treatment of various diseases. Human induced pluripotent stem cells (hiPSCs) cannot be used in clinical trials because of tumorigenicity, but the exosomes derived from hiPSCs may combine the advantages of iPSC pluripotency and the nanoscale size of exosomes while avoiding tumorigenicity. Currently, the safety and biodistribution of hiPSC-exosomes in vivo are unclear. Here, we investigated the effects of hiPSC-exosomes on hemolysis, DNA damage, and cytotoxicity through cell experiments. We also explored the safety of vein injection of hiPSC-exosomes in rabbits and rats. Differences in organ distribution after nasal administration were compared in normal and Parkinson’s disease model mice. This study may provide support for clinical therapy and research of intravenous and nasal administration of hiPSC-exosomes.
Collapse
Affiliation(s)
- Zhewei Gu
- Medical College, Guangxi University, Nanning, China
| | - Zhiyu Yin
- Medical College, Guangxi University, Nanning, China
| | - Pengbo Song
- Medical College, Guangxi University, Nanning, China
| | - Ying Wu
- Medical College, Guangxi University, Nanning, China
| | - Ying He
- Medical College, Guangxi University, Nanning, China
| | - Maoshu Zhu
- Medical College, Guangxi University, Nanning, China
| | - Zhengxin Wu
- Medical College, Guangxi University, Nanning, China
| | - Sicheng Zhao
- Medical College, Guangxi University, Nanning, China
| | - Hongri Huang
- GuangXi TaiMeiRenSheng Biotechnology Co., LTD., Nanning, China
| | - Huihuang Wang
- GuangXi TaiMeiRenSheng Biotechnology Co., LTD., Nanning, China
| | - Cailing Tong
- Biotechcomer Co., Ltd., Xiamen, China
- *Correspondence: Cailing Tong, ; Zhongquan Qi,
| | - Zhongquan Qi
- Medical College, Guangxi University, Nanning, China
- *Correspondence: Cailing Tong, ; Zhongquan Qi,
| |
Collapse
|
10
|
Ornitz DM, Itoh N. New developments in the biology of fibroblast growth factors. WIREs Mech Dis 2022; 14:e1549. [PMID: 35142107 PMCID: PMC10115509 DOI: 10.1002/wsbm.1549] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 11/08/2021] [Accepted: 11/09/2021] [Indexed: 01/28/2023]
Abstract
The fibroblast growth factor (FGF) family is composed of 18 secreted signaling proteins consisting of canonical FGFs and endocrine FGFs that activate four receptor tyrosine kinases (FGFRs 1-4) and four intracellular proteins (intracellular FGFs or iFGFs) that primarily function to regulate the activity of voltage-gated sodium channels and other molecules. The canonical FGFs, endocrine FGFs, and iFGFs have been reviewed extensively by us and others. In this review, we briefly summarize past reviews and then focus on new developments in the FGF field since our last review in 2015. Some of the highlights in the past 6 years include the use of optogenetic tools, viral vectors, and inducible transgenes to experimentally modulate FGF signaling, the clinical use of small molecule FGFR inhibitors, an expanded understanding of endocrine FGF signaling, functions for FGF signaling in stem cell pluripotency and differentiation, roles for FGF signaling in tissue homeostasis and regeneration, a continuing elaboration of mechanisms of FGF signaling in development, and an expanding appreciation of roles for FGF signaling in neuropsychiatric diseases. This article is categorized under: Cardiovascular Diseases > Molecular and Cellular Physiology Neurological Diseases > Molecular and Cellular Physiology Congenital Diseases > Stem Cells and Development Cancer > Stem Cells and Development.
Collapse
Affiliation(s)
- David M Ornitz
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Nobuyuki Itoh
- Kyoto University Graduate School of Pharmaceutical Sciences, Sakyo, Kyoto, Japan
| |
Collapse
|
11
|
Rahman MM, Islam MR, Akash S, Harun-Or-Rashid M, Ray TK, Rahaman MS, Islam M, Anika F, Hosain MK, Aovi FI, Hemeg HA, Rauf A, Wilairatana P. Recent advancements of nanoparticles application in cancer and neurodegenerative disorders: At a glance. Biomed Pharmacother 2022; 153:113305. [PMID: 35717779 DOI: 10.1016/j.biopha.2022.113305] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 06/06/2022] [Accepted: 06/13/2022] [Indexed: 11/28/2022] Open
Abstract
Nanoscale engineering is one of the innovative approaches to heal multitudes of ailments, such as varieties of malignancies, neurological problems, and infectious illnesses. Therapeutics for neurodegenerative diseases (NDs) may be modified in aspect because of their ability to stimulate physiological response while limiting negative consequences by interfacing and activating possible targets. Nanomaterials have been extensively studied and employed for cancerous therapeutic strategies since nanomaterials potentially play a significant role in medical transportation. When compared to conventional drug delivery, nanocarriers drug delivery offers various benefits, such as excellent reliability, bioactivity, improved penetration and retention impact, as well as precise targeting and administering. Upregulation of drug efflux transporters, dysfunctional apoptotic mechanisms, and a hypoxic atmosphere are all elements that lead to cancer treatment sensitivity in humans. It has been possible to target these pathways using nanoparticles and increase the effectiveness of multidrug resistance treatments. As innovative strategies of tumor chemoresistance are uncovered, nanomaterials are being developed to target specific pathways of tumor resilience. Scientists have recently begun investigating the function of nanoparticles in immunotherapy, a field that is becoming increasingly useful in the care of malignancies. Nanoscale therapeutics have been explored in this scientific literature and represent the most current approaches to neurodegenerative illnesses and cancer therapy. In addition, current findings and various biomedical nanomaterials' future promise for tissue regeneration, prospective medication design, and the synthesis of novel delivery approaches have been emphasized.
Collapse
Affiliation(s)
- Md Mominur Rahman
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, 1207 Dhaka, Bangladesh
| | - Md Rezaul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, 1207 Dhaka, Bangladesh
| | - Shopnil Akash
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, 1207 Dhaka, Bangladesh
| | - Md Harun-Or-Rashid
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, 1207 Dhaka, Bangladesh
| | - Tanmay Kumar Ray
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, 1207 Dhaka, Bangladesh
| | - Md Saidur Rahaman
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, 1207 Dhaka, Bangladesh
| | - Mahfuzul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, 1207 Dhaka, Bangladesh
| | - Fazilatunnesa Anika
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, 1207 Dhaka, Bangladesh
| | - Md Kawser Hosain
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, 1207 Dhaka, Bangladesh
| | - Farjana Islam Aovi
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, 1207 Dhaka, Bangladesh
| | - Hassan A Hemeg
- Department of Medical Laboratory Technology, College of Applied Medical Sciences, Taibah University, P.O. Box 344, Al-Madinah Al-Monawra 41411, Saudi Arabia
| | - Abdur Rauf
- Department of Chemistry, University of Swabi, Anbar, Khyber Pakhtunkhwa, Pakistan.
| | - Polrat Wilairatana
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand.
| |
Collapse
|
12
|
Cheng G, Liu Y, Ma R, Cheng G, Guan Y, Chen X, Wu Z, Chen T. Anti-Parkinsonian Therapy: Strategies for Crossing the Blood-Brain Barrier and Nano-Biological Effects of Nanomaterials. NANO-MICRO LETTERS 2022; 14:105. [PMID: 35426525 PMCID: PMC9012800 DOI: 10.1007/s40820-022-00847-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 03/12/2022] [Indexed: 05/08/2023]
Abstract
Parkinson's disease (PD), a neurodegenerative disease that shows a high incidence in older individuals, is becoming increasingly prevalent. Unfortunately, there is no clinical cure for PD, and novel anti-PD drugs are therefore urgently required. However, the selective permeability of the blood-brain barrier (BBB) poses a huge challenge in the development of such drugs. Fortunately, through strategies based on the physiological characteristics of the BBB and other modifications, including enhancement of BBB permeability, nanotechnology can offer a solution to this problem and facilitate drug delivery across the BBB. Although nanomaterials are often used as carriers for PD treatment, their biological activity is ignored. Several studies in recent years have shown that nanomaterials can improve PD symptoms via their own nano-bio effects. In this review, we first summarize the physiological features of the BBB and then discuss the design of appropriate brain-targeted delivery nanoplatforms for PD treatment. Subsequently, we highlight the emerging strategies for crossing the BBB and the development of novel nanomaterials with anti-PD nano-biological effects. Finally, we discuss the current challenges in nanomaterial-based PD treatment and the future trends in this field. Our review emphasizes the clinical value of nanotechnology in PD treatment based on recent patents and could guide researchers working in this area in the future.
Collapse
Affiliation(s)
- Guowang Cheng
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang, 330004, People's Republic of China
| | - Yujing Liu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, People's Republic of China
| | - Rui Ma
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, People's Republic of China
| | - Guopan Cheng
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, People's Republic of China
| | - Yucheng Guan
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, People's Republic of China
| | - Xiaojia Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, 999078, People's Republic of China
| | - Zhenfeng Wu
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang, 330004, People's Republic of China.
| | - Tongkai Chen
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, People's Republic of China.
| |
Collapse
|
13
|
Chen K, Rao Z, Dong S, Chen Y, Wang X, Luo Y, Gong F, Li X. Roles of the fibroblast growth factor signal transduction system in tissue injury repair. BURNS & TRAUMA 2022; 10:tkac005. [PMID: 35350443 PMCID: PMC8946634 DOI: 10.1093/burnst/tkac005] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 12/13/2021] [Indexed: 12/13/2022]
Abstract
Following injury, tissue autonomously initiates a complex repair process, resulting in either partial recovery or regeneration of tissue architecture and function in most organisms. Both the repair and regeneration processes are highly coordinated by a hierarchy of interplay among signal transduction pathways initiated by different growth factors, cytokines and other signaling molecules under normal conditions. However, under chronic traumatic or pathological conditions, the reparative or regenerative process of most tissues in different organs can lose control to different extents, leading to random, incomplete or even flawed cell and tissue reconstitution and thus often partial restoration of the original structure and function, accompanied by the development of fibrosis, scarring or even pathogenesis that could cause organ failure and death of the organism. Ample evidence suggests that the various combinatorial fibroblast growth factor (FGF) and receptor signal transduction systems play prominent roles in injury repair and the remodeling of adult tissues in addition to embryonic development and regulation of metabolic homeostasis. In this review, we attempt to provide a brief update on our current understanding of the roles, the underlying mechanisms and clinical application of FGFs in tissue injury repair.
Collapse
Affiliation(s)
| | | | - Siyang Dong
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- Department of breast surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Yajing Chen
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Xulan Wang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Yongde Luo
- Correspondence. Xiaokun Li, ; Fanghua Gong, ; Yongde Luo,
| | - Fanghua Gong
- Correspondence. Xiaokun Li, ; Fanghua Gong, ; Yongde Luo,
| | - Xiaokun Li
- Correspondence. Xiaokun Li, ; Fanghua Gong, ; Yongde Luo,
| |
Collapse
|
14
|
Liu Y, Deng J, Liu Y, Li W, Nie X. FGF, Mechanism of Action, Role in Parkinson's Disease, and Therapeutics. Front Pharmacol 2021; 12:675725. [PMID: 34234672 PMCID: PMC8255968 DOI: 10.3389/fphar.2021.675725] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 06/09/2021] [Indexed: 12/17/2022] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disease associated with severe disability and adverse effects on life quality. In PD, motor dysfunction can occur, such as quiescence, muscle stiffness, and postural instability. PD is also associated with autonomic nervous dysfunction, sleep disorders, psychiatric symptoms, and other non-motor symptoms. Degeneration of dopaminergic neurons in the substantia nigra compact (SNPC), Lewy body, and neuroinflammation are the main pathological features of PD. The death or dysfunction of dopaminergic neurons in the dense part of the substantia nigra leads to dopamine deficiency in the basal ganglia and motor dysfunction. The formation of the Lewy body is associated with the misfolding of α-synuclein, which becomes insoluble and abnormally aggregated. Astrocytes and microglia mainly cause neuroinflammation, and the activation of a variety of pro-inflammatory transcription factors and regulatory proteins leads to the degeneration of dopaminergic neurons. At present, PD is mainly treated with drugs that increase dopamine concentration or directly stimulate dopamine receptors. Fibroblast growth factor (FGF) is a family of cellular signaling proteins strongly associated with neurodegenerative diseases such as PD. FGF and its receptor (FGFR) play an essential role in the development and maintenance of the nervous system as well as in neuroinflammation and have been shown to improve the survival rate of dopaminergic neurons. This paper summarized the mechanism of FGF and its receptors in the pathological process of PD and related signaling pathways, involving the development and protection of dopaminergic neurons in SNPC, α-synuclein aggregation, mitochondrial dysfunction, and neuroinflammation. It provides a reference for developing drugs to slow down or prevent the potential of PD.
Collapse
Affiliation(s)
- Yiqiu Liu
- College of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Junyu Deng
- College of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Ye Liu
- College of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Wei Li
- College of Pharmacy, Zunyi Medical University, Zunyi, China
- Joint International Research Laboratory of Ethnomedicine of Chinese Ministry of Education, College of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Xuqiang Nie
- College of Pharmacy, Zunyi Medical University, Zunyi, China
- Joint International Research Laboratory of Ethnomedicine of Chinese Ministry of Education, College of Pharmacy, Zunyi Medical University, Zunyi, China
- Key Lab of the Basic Pharmacology of the Ministry of Education, College of Pharmacy, Zunyi Medical University, Zunyi, China
| |
Collapse
|
15
|
Chen J, Wang X, Hu J, Du J, Dordoe C, Zhou Q, Huang W, Guo R, Han F, Guo K, Ye S, Lin L, Li X. FGF20 Protected Against BBB Disruption After Traumatic Brain Injury by Upregulating Junction Protein Expression and Inhibiting the Inflammatory Response. Front Pharmacol 2021; 11:590669. [PMID: 33568994 PMCID: PMC7868342 DOI: 10.3389/fphar.2020.590669] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 12/14/2020] [Indexed: 12/11/2022] Open
Abstract
Disruption of the blood-brain barrier (BBB) and the cerebral inflammatory response occurring after traumatic brain injury (TBI) facilitate further brain damage, which leads to long-term complications of TBI. Fibroblast growth factor 20 (FGF20), a neurotrophic factor, plays important roles in brain development and neuronal homeostasis. The aim of the current study was to assess the protective effects of FGF20 on TBI via BBB maintenance. In the present study, recombinant human FGF20 (rhFGF20) reduced neurofunctional deficits, brain edema, Evans blue extravasation and neuroinflammation in a TBI mouse model. In an in vitro TNF-α-induced human brain microvascular endothelial cell (HBMEC) model of BBB disruption, rhFGF20 reduced paracellular permeability and increased trans-endothelial electrical resistance (TEER). Both in the TBI mouse model and in vitro, rhFGF20 increased the expression of proteins composing in BBB-associated tight junctions (TJs) and adherens junctions (AJs), and decreased the inflammatory response, which protected the BBB integrity. Notably, rhFGF20 preserved BBB function by activating the AKT/GSK3β pathway and inhibited the inflammatory response by regulating the JNK/NFκB pathway. Thus, FGF20 is a potential candidate treatment for TBI that protects the BBB by upregulating junction protein expression and inhibiting the inflammatory response.
Collapse
Affiliation(s)
- Jun Chen
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Xue Wang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Jian Hu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Jingting Du
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Confidence Dordoe
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Qiulin Zhou
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Wenting Huang
- School of the First Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Ruili Guo
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Fanyi Han
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Kaiming Guo
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Shasha Ye
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Li Lin
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Xiaokun Li
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
- Research Units of Clinical Translation of Cell Growth Factors and Diseases Research, Chinese Academy of Medical Science, Wenzhou, China
| |
Collapse
|
16
|
Jarrin S, Hakami A, Newland B, Dowd E. Growth Factor Therapy for Parkinson's Disease: Alternative Delivery Systems. JOURNAL OF PARKINSON'S DISEASE 2021; 11:S229-S236. [PMID: 33896851 PMCID: PMC8543245 DOI: 10.3233/jpd-212662] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 03/31/2021] [Indexed: 12/30/2022]
Abstract
Despite decades of research and billions in global investment, there remains no preventative or curative treatment for any neurodegenerative condition, including Parkinson's disease (PD). Arguably, the most promising approach for neuroprotection and neurorestoration in PD is using growth factors which can promote the growth and survival of degenerating neurons. However, although neurotrophin therapy may seem like the ideal approach for neurodegenerative disease, the use of growth factors as drugs presents major challenges because of their protein structure which creates serious hurdles related to accessing the brain and specific targeting of affected brain regions. To address these challenges, several different delivery systems have been developed, and two major approaches-direct infusion of the growth factor protein into the target brain region and in vivo gene therapy-have progressed to clinical trials in patients with PD. In addition to these clinically evaluated approaches, a range of other delivery methods are in various degrees of development, each with their own unique potential. This review will give a short overview of some of these alternative delivery systems, with a focus on ex vivo gene therapy and biomaterial-aided protein and gene delivery, and will provide some perspectives on their potential for clinical development and translation.
Collapse
Affiliation(s)
- Sarah Jarrin
- Pharmacology & Therapeutics and Galway Neuroscience Centre, National University of Ireland, Galway, Ireland
| | - Abrar Hakami
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK
| | - Ben Newland
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK
| | - Eilís Dowd
- Pharmacology & Therapeutics and Galway Neuroscience Centre, National University of Ireland, Galway, Ireland
| |
Collapse
|
17
|
Ballard C, Aarsland D, Cummings J, O'Brien J, Mills R, Molinuevo JL, Fladby T, Williams G, Doherty P, Corbett A, Sultana J. Drug repositioning and repurposing for Alzheimer disease. Nat Rev Neurol 2020; 16:661-673. [PMID: 32939050 PMCID: PMC8291993 DOI: 10.1038/s41582-020-0397-4] [Citation(s) in RCA: 113] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/24/2020] [Indexed: 12/12/2022]
Abstract
Drug repositioning and repurposing can enhance traditional drug development efforts and could accelerate the identification of new treatments for individuals with Alzheimer disease (AD) dementia and mild cognitive impairment. Transcriptional profiling offers a new and highly efficient approach to the identification of novel candidates for repositioning and repurposing. In the future, novel AD transcriptional signatures from cells isolated at early stages of disease, or from human neurons or microglia that carry mutations that increase the risk of AD, might be used as probes to identify additional candidate drugs. Phase II trials assessing repurposed agents must consider the best target population for a specific candidate therapy as well as the mechanism of action of the treatment. In this Review, we highlight promising compounds to prioritize for clinical trials in individuals with AD, and discuss the value of Delphi consensus methodology and evidence-based reviews to inform this prioritization process. We also describe emerging work, focusing on the potential value of transcript signatures as a cost-effective approach to the identification of novel candidates for repositioning.
Collapse
Affiliation(s)
- Clive Ballard
- College of Medicine and Health, University of Exeter, Exeter, UK.
| | - Dag Aarsland
- Institute of Psychiatry Psychology and Neuroscience, King's College London, London, UK
- SESAM (Regional Center for Elderly Medicine and Interaction), University Hospital Stavanger, Stavanger, Norway
| | - Jeffrey Cummings
- Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV, USA
| | - John O'Brien
- School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Roger Mills
- Institute of Psychiatry Psychology and Neuroscience, King's College London, London, UK
- Vincere Consulting, LLC, San Diego, CA, USA
| | | | - Tormod Fladby
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Gareth Williams
- Institute of Psychiatry Psychology and Neuroscience, King's College London, London, UK
| | - Pat Doherty
- Institute of Psychiatry Psychology and Neuroscience, King's College London, London, UK
| | - Anne Corbett
- College of Medicine and Health, University of Exeter, Exeter, UK
| | - Janet Sultana
- Department of Biomedical and Dental Sciences and Morpho-functional Imaging, University of Messina, Messina, Italy
| |
Collapse
|
18
|
Particulate systems for improving therapeutic efficacy of pharmaceuticals against central nervous system-related diseases. J Taiwan Inst Chem Eng 2020. [DOI: 10.1016/j.jtice.2020.09.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
19
|
Chen HX, Liang FC, Gu P, Xu BL, Xu HJ, Wang WT, Hou JY, Xie DX, Chai XQ, An SJ. Exosomes derived from mesenchymal stem cells repair a Parkinson's disease model by inducing autophagy. Cell Death Dis 2020; 11:288. [PMID: 32341347 PMCID: PMC7184757 DOI: 10.1038/s41419-020-2473-5] [Citation(s) in RCA: 178] [Impact Index Per Article: 35.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Revised: 04/07/2020] [Accepted: 04/08/2020] [Indexed: 12/17/2022]
Abstract
Parkinson's disease (PD) is a progressively debilitating neurodegenerative condition that leads to motor and cognitive dysfunction. At present, clinical treatment can only improve symptoms, but cannot effectively protect dopaminergic neurons. Several reports have demonstrated that human umbilical cord mesenchymal stem cells (hucMSCs) afford neuroprotection, while their application is limited because of their uncontrollable differentiation and other reasons. Stem cells communicate with cells through secreted exosomes (Exos), the present study aimed to explore whether Exos secreted by hucMSCs could function instead of hucMSCs. hucMSCs were successfully isolated and characterized, and shown to contribute to 6-hydroxydopamine (6-OHDA)-stimulated SH-SY5Y cell proliferation; hucMSC-derived Exos were also involved in this process. The Exos were purified and identified, and then labeled with PKH 26, it was found that the Exos could be efficiently taken up by SH-SY5Y cells after 12 h of incubation. Pretreatment with Exos promoted 6-OHDA-stimulated SH-SY5Y cells to proliferate and inhibited apoptosis by inducing autophagy. Furthermore, Exos reached the substantia nigra through the blood-brain barrier (BBB) in vivo, relieved apomorphine-induced asymmetric rotation, reduced substantia nigra dopaminergic neuron loss and apoptosis, and upregulated the level of dopamine in the striatum. These results demonstrate that hucMSCs-Exos have a treatment capability for PD and can traverse the BBB, indicating their potential for the effective treatment of PD.
Collapse
Affiliation(s)
- Hong-Xu Chen
- Department of Neurology, the First Hospital of Hebei Medical University, No. 89 Donggang Road, Yuhua District, Shi Jiazhuang, 050031, Hebei, China
- Hebei Provincial Engineering Laboratory of Plant Bioreactor Preparation Technology, No. 326 Xinshi south Road, Qiaoxi District, Shi Jiazhuang, 050090, Hebei, China
| | - Fu-Chao Liang
- Hebei Provincial Engineering Laboratory of Plant Bioreactor Preparation Technology, No. 326 Xinshi south Road, Qiaoxi District, Shi Jiazhuang, 050090, Hebei, China
- Research Center, Hebei University of Chinese Medicine, No. 326 Xinshi south Road, Qiaoxi District, Shi Jiazhuang, 050090, Hebei, China
| | - Ping Gu
- Department of Neurology, the First Hospital of Hebei Medical University, No. 89 Donggang Road, Yuhua District, Shi Jiazhuang, 050031, Hebei, China
| | - Bian-Ling Xu
- Hebei Provincial Engineering Laboratory of Plant Bioreactor Preparation Technology, No. 326 Xinshi south Road, Qiaoxi District, Shi Jiazhuang, 050090, Hebei, China
- Department of Gynecology, the Second Hospital of Hebei Medical University, No. 215, HePing West Road, Shi Jiazhuang, 050000, Hebei, China
| | - Hong-Jun Xu
- Hebei Provincial Engineering Laboratory of Plant Bioreactor Preparation Technology, No. 326 Xinshi south Road, Qiaoxi District, Shi Jiazhuang, 050090, Hebei, China
- Research Center, Hebei University of Chinese Medicine, No. 326 Xinshi south Road, Qiaoxi District, Shi Jiazhuang, 050090, Hebei, China
| | - Wen-Ting Wang
- Department of Neurology, the First Hospital of Hebei Medical University, No. 89 Donggang Road, Yuhua District, Shi Jiazhuang, 050031, Hebei, China
| | - Jia-Yang Hou
- Hebei Provincial Engineering Laboratory of Plant Bioreactor Preparation Technology, No. 326 Xinshi south Road, Qiaoxi District, Shi Jiazhuang, 050090, Hebei, China
- Research Center, Hebei University of Chinese Medicine, No. 326 Xinshi south Road, Qiaoxi District, Shi Jiazhuang, 050090, Hebei, China
| | - Dong-Xiao Xie
- Department of orthopaedic, Third hospital of Hebei Medical University, Shi Jiazhuang, 050000, Hebei, China
| | - Xi-Qing Chai
- Department of Neurology, the First Hospital of Hebei Medical University, No. 89 Donggang Road, Yuhua District, Shi Jiazhuang, 050031, Hebei, China.
| | - Sheng-Jun An
- Hebei Provincial Engineering Laboratory of Plant Bioreactor Preparation Technology, No. 326 Xinshi south Road, Qiaoxi District, Shi Jiazhuang, 050090, Hebei, China.
- Research Center, Hebei University of Chinese Medicine, No. 326 Xinshi south Road, Qiaoxi District, Shi Jiazhuang, 050090, Hebei, China.
| |
Collapse
|
20
|
Teixeira MI, Lopes CM, Amaral MH, Costa PC. Current insights on lipid nanocarrier-assisted drug delivery in the treatment of neurodegenerative diseases. Eur J Pharm Biopharm 2020; 149:192-217. [PMID: 31982574 DOI: 10.1016/j.ejpb.2020.01.005] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 10/16/2019] [Accepted: 01/08/2020] [Indexed: 12/12/2022]
Abstract
The central nervous system (CNS) is vulnerable to pathologic processes that lead to the development of neurodegenerative disorders like Alzheimer's, Parkinson's and Huntington's diseases, Multiple sclerosis or Amyotrophic lateral sclerosis. These are chronic and progressive pathologies characterized by the loss of neurons and the formation of misfolded proteins. Additionally, neurodegenerative diseases are accompanied by a structural and functional dysfunction of the blood-brain barrier (BBB). Although serving as a protection for the CNS, the existence of physiological barriers, especially the BBB, limits the access of several therapeutic agents to the brain, constituting a major hindrance in neurotherapeutics advancement. In this regard, nanotechnology-based approaches have arisen as a promising strategy to not only improve drug targeting to the brain, but also to increase bioavailability. Lipid nanocarriers such as liposomes, solid lipid nanoparticles (SLN), nanostructured lipid carriers (NLC), microemulsions and nanoemulsions, have already proven their potential for enhancing brain transport, crossing more easily into the CNS and allowing the administration of medicines that could benefit the treatment of neurological pathologies. Given the socioeconomic impact of such conditions and the advent of nanotechnology that inevitably leads to more effective and superior therapeutics for their management, it is imperative to constantly update on the current knowledge of these topics. Herein, we provide insight on the BBB and the pathophysiology of the main neurodegenerative disorders. Moreover, this review seeks to highlight the several approaches that can be used to improve the delivery of therapeutic agents to the CNS, while also offering an extensive overview of the latest efforts regarding the use of lipid-based nanocarriers in the management of neurodegenerative diseases.
Collapse
Affiliation(s)
- M I Teixeira
- UCIBIO, REQUIMTE, MEDTECH, Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge de Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| | - C M Lopes
- FP-ENAS/CEBIMED, Fernando Pessoa Energy, Environment and Health Research Unit/Biomedical Research Centre, Faculty of Health Sciences, Fernando Pessoa University, Rua Carlos da Maia, 296, 4200-150 Porto, Portugal
| | - M H Amaral
- UCIBIO, REQUIMTE, MEDTECH, Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge de Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - P C Costa
- UCIBIO, REQUIMTE, MEDTECH, Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge de Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| |
Collapse
|
21
|
Fletcher EJR, Jamieson AD, Williams G, Doherty P, Duty S. Targeted repositioning identifies drugs that increase fibroblast growth factor 20 production and protect against 6-hydroxydopamine-induced nigral cell loss in rats. Sci Rep 2019; 9:8336. [PMID: 31171821 PMCID: PMC6554393 DOI: 10.1038/s41598-019-44803-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 05/21/2019] [Indexed: 12/21/2022] Open
Abstract
Endogenous fibroblast growth factor 20 (FGF20) supports maintenance of dopaminergic neurones within the nigrostriatal pathway. Moreover, direct intracerebral infusion of FGF20 protects against nigrostriatal tract loss in the 6-hydroxydopamine lesion rat model of Parkinson’s disease. Increasing endogenous FGF20 production might provide a less-invasive, more translational way of providing such protection. Accordingly, we adopted a targeted repositioning approach to screen for candidate FDA-approved drugs with potential to enhance endogenous FGF20 production in brain. In silico interrogation of the Broad Institute’s Connectivity Map database (CMap), revealed 50 candidate drugs predicted to increase FGF20 transcription, 16 of which had profiles favourable for use in Parkinson’s disease. Of these, 11 drugs were found to significantly elevate FGF20 protein production in MCF-7 cells, between two- and four-fold. Four drugs were selected for examination in vivo. Following oral dosing in rats for 7 days, salbutamol and triflusal, but not dimethadione or trazodone, significantly elevated FGF20 levels in the nigrostriatal tract. Preliminary examination in the unilateral 6-hydroxydopamine-lesioned rat revealed a modest but significant protection against nigral cell loss with both drugs. Our data demonstrate the power of targeted repositioning as a method to identify existing drugs that may combat disease progression in Parkinson’s by boosting FGF20 levels.
Collapse
Affiliation(s)
- Edward J R Fletcher
- King's College London, Institute of Psychiatry, Psychology & Neuroscience, Wolfson Centre for Age-Related Diseases, Guy's Campus, London, SE1 1UL, UK
| | - Aran D Jamieson
- King's College London, Institute of Psychiatry, Psychology & Neuroscience, Wolfson Centre for Age-Related Diseases, Guy's Campus, London, SE1 1UL, UK
| | - Gareth Williams
- King's College London, Institute of Psychiatry, Psychology & Neuroscience, Wolfson Centre for Age-Related Diseases, Guy's Campus, London, SE1 1UL, UK
| | - Patrick Doherty
- King's College London, Institute of Psychiatry, Psychology & Neuroscience, Wolfson Centre for Age-Related Diseases, Guy's Campus, London, SE1 1UL, UK
| | - Susan Duty
- King's College London, Institute of Psychiatry, Psychology & Neuroscience, Wolfson Centre for Age-Related Diseases, Guy's Campus, London, SE1 1UL, UK.
| |
Collapse
|
22
|
Lee EJ, Fomenko A, Lozano AM. Magnetic Resonance-Guided Focused Ultrasound : Current Status and Future Perspectives in Thermal Ablation and Blood-Brain Barrier Opening. J Korean Neurosurg Soc 2018; 62:10-26. [PMID: 30630292 PMCID: PMC6328789 DOI: 10.3340/jkns.2018.0180] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 11/13/2018] [Indexed: 02/06/2023] Open
Abstract
Magnetic resonance-guided focused ultrasound (MRgFUS) is an emerging new technology with considerable potential to treat various neurological diseases. With refinement of ultrasound transducer technology and integration with magnetic resonance imaging guidance, transcranial sonication of precise cerebral targets has become a therapeutic option. Intensity is a key determinant of ultrasound effects. High-intensity focused ultrasound can produce targeted lesions via thermal ablation of tissue. MRgFUS-mediated stereotactic ablation is non-invasive, incision-free, and confers immediate therapeutic effects. Since the US Food and Drug Administration approval of MRgFUS in 2016 for unilateral thalamotomy in medication-refractory essential tremor, studies on novel indications such as Parkinson's disease, psychiatric disease, and brain tumors are underway. MRgFUS is also used in the context of blood-brain barrier (BBB) opening at low intensities, in combination with intravenously-administered microbubbles. Preclinical studies show that MRgFUS-mediated BBB opening safely enhances the delivery of targeted chemotherapeutic agents to the brain and improves tumor control as well as survival. In addition, BBB opening has been shown to activate the innate immune system in animal models of Alzheimer's disease. Amyloid plaque clearance and promotion of neurogenesis in these studies suggest that MRgFUS-mediated BBB opening may be a new paradigm for neurodegenerative disease treatment in the future. Here, we review the current status of preclinical and clinical trials of MRgFUS-mediated thermal ablation and BBB opening, described their mechanisms of action, and discuss future prospects.
Collapse
Affiliation(s)
- Eun Jung Lee
- Toronto Western Research Institute, University Health Network, Toronto, Canada
| | - Anton Fomenko
- Toronto Western Research Institute, University Health Network, Toronto, Canada
| | - Andres M Lozano
- Toronto Western Research Institute, University Health Network, Toronto, Canada.,Division of Neurosurgery, Department of Surgery, Toronto Western Hospital, University of Toronto, Toronto, Canada
| |
Collapse
|