101
|
Wang L, Zhang D, Ren Y, Guo S, Li J, Ma S, Yao M, Guan F. Injectable hyaluronic acid hydrogel loaded with BMSC and NGF for traumatic brain injury treatment. Mater Today Bio 2022; 13:100201. [PMID: 35024600 PMCID: PMC8733324 DOI: 10.1016/j.mtbio.2021.100201] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 12/21/2021] [Accepted: 12/29/2021] [Indexed: 12/16/2022] Open
Abstract
Injectable hydrogel has the advantage to fill the defective area and thereby shows promise as therapeutic implant or cell/drug delivery vehicle for tissue repair. In this study, an injectable hyaluronic acid hydrogel in situ dual-enzymatically cross-linked by galactose oxidase (GalOx) and horseradish peroxidase (HRP) was synthesized and optimized, and the therapeutic effect of this hydrogel encapsulated with bone mesenchymal stem cells (BMSC) and nerve growth factors (NGF) for traumatic brain injury (TBI) mice was investigated. Results from in vitro experiments showed that either tyramine-modified hyaluronic acid hydrogels (HT) or NGF loaded HT hydrogels (HT/NGF) possessed good biocompatibility. More importantly, the HT hydrogels loaded with BMSC and NGF could facilitate the survival and proliferation of endogenous neural cells probably by neurotrophic factors release and neuroinflammation regulation, and consequently improved the neurological function recovery and accelerated the repair process in a C57BL/6 TBI mice model. All these findings highlight that this injectable, BMSC and NGF-laden HT hydrogel has enormous potential for TBI and other tissue repair therapy.
Collapse
Affiliation(s)
| | | | - Yikun Ren
- School of Life Science, Zhengzhou University, 100 Science Road, Zhengzhou, 450001, PR China
| | - Shen Guo
- School of Life Science, Zhengzhou University, 100 Science Road, Zhengzhou, 450001, PR China
| | - Jinrui Li
- School of Life Science, Zhengzhou University, 100 Science Road, Zhengzhou, 450001, PR China
| | - Shanshan Ma
- School of Life Science, Zhengzhou University, 100 Science Road, Zhengzhou, 450001, PR China
| | - Minghao Yao
- School of Life Science, Zhengzhou University, 100 Science Road, Zhengzhou, 450001, PR China
| | - Fangxia Guan
- School of Life Science, Zhengzhou University, 100 Science Road, Zhengzhou, 450001, PR China
| |
Collapse
|
102
|
Tang R, Langdon WY, Zhang J. Negative regulation of receptor tyrosine kinases by ubiquitination: Key roles of the Cbl family of E3 ubiquitin ligases. Front Endocrinol (Lausanne) 2022; 13:971162. [PMID: 35966060 PMCID: PMC9365936 DOI: 10.3389/fendo.2022.971162] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 07/11/2022] [Indexed: 11/13/2022] Open
Abstract
Receptor tyrosine kinases (RTKs) serve as transmembrane receptors that participate in a broad spectrum of cellular processes including cellular growth, motility, differentiation, proliferation, and metabolism. Hence, elucidating the regulatory mechanisms of RTKs involved in an assortment of diseases such as cancers attracts increasing interest from researchers. Members of the Cbl family ubiquitin ligases (c-Cbl, Cbl-b and Cbl-c in mammals) have emerged as negative regulators of activated RTKs. Upon activation of RTKs by growth factors, Cbl binds to RTKs via its tyrosine kinase binding (TKB) domain and targets them for ubiquitination, thus facilitating their degradation and negative regulation of RTK signaling. RTKs such as epidermal growth factor receptor (EGFR), platelet-derived growth factor receptor (PDGF), fibroblast growth factor receptor (FGFR) and hepatocyte growth factor receptor (HGFR) undergo ubiquitination upon interaction with Cbl family members. In this review, we summarize the current knowledge related to the negative regulation of RTKs by Cbl family proteins.
Collapse
Affiliation(s)
- Rong Tang
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
| | - Wallace Y. Langdon
- School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
| | - Jian Zhang
- Department of Pathology, The University of Iowa, Iowa City, IA, United States
- *Correspondence: Jian Zhang,
| |
Collapse
|
103
|
Kato Y, Uchiumi H, Usami R, Takatsu H, Aoki Y, Yanai S, Endo S, Fukui K. Tocotrienols reach the brain and play roles in the attenuation of body weight gain and improvement of cognitive function in high-fat diet-treated mice. J Clin Biochem Nutr 2021; 69:256-264. [PMID: 34857987 PMCID: PMC8611367 DOI: 10.3164/jcbn.21-10] [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: 01/19/2021] [Accepted: 03/30/2021] [Indexed: 12/14/2022] Open
Abstract
Obesity induces severe disorders such as type 2 diabetes and cardiovascular events, and the number of people with obesity is increasing all over the world. Furthermore, it is possible that obesity increases the risk of cognitive dysfunction via the acceleration of oxidative damage. Tocotrienols, which are part of the vitamin E family, have antioxidant and anti-obesity effects. However, the effects of tocotrienols on high-fat diet-treated mice have not been completely elucidated. In this study, we assessed changes in body weight, spatial reference memory acquisition, liver lipid droplet size, blood brain barrier-related protein expressions and antioxidative defense systems in high-fat diet-treated mice in the presence or absence of tocotrienols. The results showed that tocotrienols significantly inhibited body weight gain and lipid droplet synthesis. Although the amount was very small, it was confirmed that tocotrienols surely reached the brain in the perfused brain. Treatment with tocotrienols was tended to improve cognitive function in the control mice. However, tocotrienols did not modulate blood brain barrier-related protein expressions or antioxidative defense systems. These results indicate that treatment with tocotrienols could be effective for the prevention of obesity and cognitive dysfunction. Further extended research is needed to elucidate the relationship between anti-obesity and antioxidant effects of tocotrienols, especially in the brain.
Collapse
Affiliation(s)
- Yugo Kato
- Molecular Cell Biology Laboratory, Department of Functional Control Systems, Graduate School of Engineering and Science, Shibaura Institute of Technology, Fukasaku 307, Minuma-ku, Saitama 337-8570, Japan
| | - Hayami Uchiumi
- Molecular Cell Biology Laboratory, Department of Bioscience and Engineering, College of Systems Engineering and Science, Shibaura Institute of Technology, Fukasaku 307, Minuma-ku, Saitama 337-8570, Japan
| | - Ryo Usami
- Molecular Cell Biology Laboratory, Department of Bioscience and Engineering, College of Systems Engineering and Science, Shibaura Institute of Technology, Fukasaku 307, Minuma-ku, Saitama 337-8570, Japan
| | - Hirokatsu Takatsu
- Department of Medical Technology, Faculty of Health Sciences, Kyorin University, Shimorenjaku 5-4-1, Mitaka, Tokyo 181-8612, Japan
| | - Yoshinori Aoki
- Mitsubishi-Chemical Foods Corporation, Marunouchi 1-1-1, Chiyoda-ku, Tokyo 100-8251, Japan
| | - Shuichi Yanai
- Aging Neuroscience Research Team, Tokyo Metropolitan Institute of Gerontology, Sakae-cho 35-2, Itabashi-ku, Tokyo 173-0015, Japan
| | - Shogo Endo
- Aging Neuroscience Research Team, Tokyo Metropolitan Institute of Gerontology, Sakae-cho 35-2, Itabashi-ku, Tokyo 173-0015, Japan
| | - Koji Fukui
- Molecular Cell Biology Laboratory, Department of Functional Control Systems, Graduate School of Engineering and Science, Shibaura Institute of Technology, Fukasaku 307, Minuma-ku, Saitama 337-8570, Japan.,Molecular Cell Biology Laboratory, Department of Bioscience and Engineering, College of Systems Engineering and Science, Shibaura Institute of Technology, Fukasaku 307, Minuma-ku, Saitama 337-8570, Japan
| |
Collapse
|
104
|
Fujitani M, Otani Y, Miyajima H. Do Neurotrophins Connect Neurological Disorders and Heart Diseases? Biomolecules 2021; 11:1730. [PMID: 34827728 PMCID: PMC8615910 DOI: 10.3390/biom11111730] [Citation(s) in RCA: 4] [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: 10/06/2021] [Revised: 11/08/2021] [Accepted: 11/16/2021] [Indexed: 12/21/2022] Open
Abstract
Neurotrophins (NTs) are one of the most characterized neurotrophic factor family members and consist of four members in mammals. Growing evidence suggests that there is a complex inter- and bi-directional relationship between central nervous system (CNS) disorders and cardiac dysfunction, so-called "brain-heart axis". Recent studies suggest that CNS disorders, including neurodegenerative diseases, stroke, and depression, affect cardiovascular function via various mechanisms, such as hypothalamic-pituitary-adrenal axis augmentation. Although this brain-heart axis has been well studied in humans and mice, the involvement of NT signaling in the axis has not been fully investigated. In the first half of this review, we emphasize the importance of NTs not only in the nervous system, but also in the cardiovascular system from the embryonic stage to the adult state. In the second half, we discuss the involvement of NTs in the pathogenesis of cardiovascular diseases, and then examine whether an alteration in NTs could serve as the mediator between neurological disorders and heart dysfunction. The further investigation we propose herein could contribute to finding direct evidence for the involvement of NTs in the axis and new treatment for cardiovascular diseases.
Collapse
Affiliation(s)
- Masashi Fujitani
- Department of Anatomy and Neuroscience, Faculty of Medicine, Shimane University, 89-1 Enya-cho, Izumo-shi 693-8501, Shimane, Japan; (Y.O.); (H.M.)
| | | | | |
Collapse
|
105
|
Parker BJ, Rhodes DI, O'Brien CM, Rodda AE, Cameron NR. Nerve guidance conduit development for primary treatment of peripheral nerve transection injuries: A commercial perspective. Acta Biomater 2021; 135:64-86. [PMID: 34492374 DOI: 10.1016/j.actbio.2021.08.052] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 08/19/2021] [Accepted: 08/30/2021] [Indexed: 12/17/2022]
Abstract
Commercial nerve guidance conduits (NGCs) for repair of peripheral nerve discontinuities are of little use in gaps larger than 30 mm, and for smaller gaps they often fail to compete with the autografts that they are designed to replace. While recent research to develop new technologies for use in NGCs has produced many advanced designs with seemingly positive functional outcomes in animal models, these advances have not been translated into viable clinical products. While there have been many detailed reviews of the technologies available for creating NGCs, none of these have focussed on the requirements of the commercialisation process which are vital to ensure the translation of a technology from bench to clinic. Consideration of the factors essential for commercial viability, including regulatory clearance, reimbursement processes, manufacturability and scale up, and quality management early in the design process is vital in giving new technologies the best chance at achieving real-world impact. Here we have attempted to summarise the major components to consider during the development of emerging NGC technologies as a guide for those looking to develop new technology in this domain. We also examine a selection of the latest academic developments from the viewpoint of clinical translation, and discuss areas where we believe further work would be most likely to bring new NGC technologies to the clinic. STATEMENT OF SIGNIFICANCE: NGCs for peripheral nerve repairs represent an adaptable foundation with potential to incorporate modifications to improve nerve regeneration outcomes. In this review we outline the regulatory processes that functionally distinct NGCs may need to address and explore new modifications and the complications that may need to be addressed during the translation process from bench to clinic.
Collapse
Affiliation(s)
- Bradyn J Parker
- Department of Materials Science and Engineering, Monash University, 22 Alliance Lane, Clayton, Victoria 3800, Australia; Commonwealth Scientific and Industrial Research Organisation (CSIRO) Manufacturing, Research Way, Clayton, Victoria 3168, Australia
| | - David I Rhodes
- Department of Materials Science and Engineering, Monash University, 22 Alliance Lane, Clayton, Victoria 3800, Australia; ReNerve Pty. Ltd., Brunswick East 3057, Australia
| | - Carmel M O'Brien
- Commonwealth Scientific and Industrial Research Organisation (CSIRO) Manufacturing, Research Way, Clayton, Victoria 3168, Australia; Australian Regenerative Medicine Institute, Science, Technology, Research and innovation Precinct (STRIP), Monash University, Wellington Road, Clayton, Victoria 3800, Australia
| | - Andrew E Rodda
- Department of Materials Science and Engineering, Monash University, 22 Alliance Lane, Clayton, Victoria 3800, Australia
| | - Neil R Cameron
- Department of Materials Science and Engineering, Monash University, 22 Alliance Lane, Clayton, Victoria 3800, Australia; School of Engineering, University of Warwick, Coventry CV4 7AL, United Kingdom.
| |
Collapse
|
106
|
Togo K, Fukusumi H, Shofuda T, Ohnishi H, Yamazaki H, Hayashi MK, Kawasaki N, Takei N, Nakazawa T, Saito Y, Baba K, Hashimoto H, Sekino Y, Shirao T, Mochizuki H, Kanemura Y. Postsynaptic structure formation of human iPS cell-derived neurons takes longer than presynaptic formation during neural differentiation in vitro. Mol Brain 2021; 14:149. [PMID: 34629097 PMCID: PMC8504131 DOI: 10.1186/s13041-021-00851-1] [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: 02/11/2021] [Accepted: 09/04/2021] [Indexed: 11/10/2022] Open
Abstract
The generation of mature synaptic structures using neurons differentiated from human-induced pluripotent stem cells (hiPSC-neurons) is expected to be applied to physiological studies of synapses in human cells and to pathological studies of diseases that cause abnormal synaptic function. Although it has been reported that synapses themselves change from an immature to a mature state as neurons mature, there are few reports that clearly show when and how human stem cell-derived neurons change to mature synaptic structures. This study was designed to elucidate the synapse formation process of hiPSC-neurons. We propagated hiPSC-derived neural progenitor cells (hiPSC-NPCs) that expressed localized markers of the ventral hindbrain as neurospheres by dual SMAD inhibition and then differentiated them into hiPSC-neurons in vitro. After 49 days of in vitro differentiation, hiPSC-neurons significantly expressed pre- and postsynaptic markers at both the transcript and protein levels. However, the expression of postsynaptic markers was lower than in normal human or normal rat brain tissues, and immunostaining analysis showed that it was relatively modest and was lower than that of presynaptic markers and that its localization in synaptic structures was insufficient. Neurophysiological analysis using a microelectrode array also revealed that no synaptic activity was generated on hiPSC-neurons at 49 days of differentiation. Analysis of subtype markers by immunostaining revealed that most hiPSC-neurons expressed vesicular glutamate transporter 2 (VGLUT2). The presence or absence of NGF, which is required for the survival of cholinergic neurons, had no effect on their cell fractionation. These results suggest that during the synaptogenesis of hiPSC-neurons, the formation of presynaptic structures is not the only requirement for the formation of postsynaptic structures and that the mRNA expression of postsynaptic markers does not correlate with the formation of their mature structures. Technically, we also confirmed a certain level of robustness and reproducibility of our neuronal differentiation method in a multicenter setting, which will be helpful for future research. Synapse formation with mature postsynaptic structures will remain an interesting issue for stem cell-derived neurons, and the present method can be used to obtain early and stable quality neuronal cultures from hiPSC-NPCs.
Collapse
Affiliation(s)
- Kazuyuki Togo
- Department of Neurology, Graduate School of Medicine, Osaka University, Suita, Osaka, 565-0871, Japan.,Division of Stem Cell Research, Department of Biomedical Research and Innovation, Institute for Clinical Research, National Hospital Organization Osaka National Hospital, Osaka, Osaka, 540-0006, Japan
| | - Hayato Fukusumi
- Division of Stem Cell Research, Department of Biomedical Research and Innovation, Institute for Clinical Research, National Hospital Organization Osaka National Hospital, Osaka, Osaka, 540-0006, Japan
| | - Tomoko Shofuda
- Division of Stem Cell Research, Department of Biomedical Research and Innovation, Institute for Clinical Research, National Hospital Organization Osaka National Hospital, Osaka, Osaka, 540-0006, Japan
| | - Hiroshi Ohnishi
- Department of Laboratory Sciences, Gunma University Graduate School of Health Sciences, Maebashi, Gunma, 371-8514, Japan
| | - Hiroyuki Yamazaki
- Department of Neurobiology and Behavior, Gunma University Graduate School of Medicine, Maebashi, Gunma, 371-8511, Japan.,Faculty of Social Welfare, Gunma University of Health and Welfare, Maebashi, Gunma, 371-0823, Japan
| | - Mariko Kato Hayashi
- School of Medicine, International University of Health and Welfare, Narita, Chiba, 286-8686, Japan.,Department of Food Science and Nutrition, Faculty of Food and Health Sciences, Showa Women's University, Setagaya-ku, Tokyo, 154-8533, Japan
| | - Nana Kawasaki
- Laboratory of Biopharmaceutical and Regenerative Sciences, Graduate School of Medical Life Science, Yokohama City University, Yokohama, Kanagawa, 230-0045, Japan
| | - Nobuyuki Takei
- Department of Brain Tumor Biology, Brain Research Institute, Niigata University, Niigata, Niigata, 951-8585, Japan
| | - Takanobu Nakazawa
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, 565-0871, Japan.,Department of Bioscience, Faculty of Life Sciences, Tokyo University of Agriculture, Setagaya-ku, Tokyo, 156-8502, Japan
| | - Yumiko Saito
- Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Hiroshima, 739-8521, Japan
| | - Kousuke Baba
- Department of Neurology, Graduate School of Medicine, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Hitoshi Hashimoto
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, 565-0871, Japan.,Molecular Research Center for Children's Mental Development, United Graduate School of Child Development, Osaka University, Suita, Osaka, 565-0871, Japan.,Division of Bioscience, Institute for Datability Science, Osaka University, Suita, Osaka, 565-0871, Japan.,Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Osaka, 565-0871, Japan.,Department of Molecular Pharmaceutical Sciences, Graduate School of Medicine, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Yuko Sekino
- Endowed Laboratory of Human Cell-Based Drug Discovery, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Tomoaki Shirao
- Department of Neurobiology and Behavior, Gunma University Graduate School of Medicine, Maebashi, Gunma, 371-8511, Japan
| | - Hideki Mochizuki
- Department of Neurology, Graduate School of Medicine, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Yonehiro Kanemura
- Division of Regenerative Medicine, Department of Biomedical Research and Innovation, Institute for Clinical Research, National Hospital Organization Osaka National Hospital, 2-1-14 Hoenzaka, Chuo-ku, Osaka, Osaka, 540-0006, Japan. .,Department of Neurosurgery, National Hospital Organization Osaka National Hospital, Osaka, Osaka, 540-0006, Japan.
| |
Collapse
|
107
|
He B, Sun C, Li H, Yang T, Song H, Wang Y, Wang Y. Primary culture of adult cortical neurons from reptile Gekko japonicus. J Anat 2021; 239:913-919. [PMID: 34047374 PMCID: PMC8450464 DOI: 10.1111/joa.13461] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 05/06/2021] [Accepted: 05/07/2021] [Indexed: 01/26/2023] Open
Abstract
Adult neurons of several reptiles still retain the ability of axonal regeneration in contrast to the low intrinsic regenerative capacity of those in the central nervous system (CNS) in mammals. This feature of the reptilian neurons has provided a perfect model for elucidating the regenerative mechanism lost in the mammalian counterparts. However, little information is available on the primary culture method of adult reptilian neurons, which greatly limits their valuable applications. In the present study, we introduced a simple and easy method for the isolation, culture, and identification of neurons from the cerebral cortex using the adult geckos. The cultured cells were further identified by immunofluorescence using antibodies against neuron-specific markers β-Ⅲ-tubulin and NeuN. The cortical neurons from adult gecko displayed spindle-shaped, bipolar, or multipolar morphology with a plump soma. This primary culture method for adult reptilian neurons will be beneficial for comparative studies of neuronal biology in various vertebrates.
Collapse
Affiliation(s)
- Bingqiang He
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of EducationCo‐innovation Center of NeuroregenerationNantong UniversityNantongPR China
| | - Chunshuai Sun
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of EducationCo‐innovation Center of NeuroregenerationNantong UniversityNantongPR China
| | - Hui Li
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of EducationCo‐innovation Center of NeuroregenerationNantong UniversityNantongPR China
| | - Ting Yang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of EducationCo‐innovation Center of NeuroregenerationNantong UniversityNantongPR China
| | - Honghua Song
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of EducationCo‐innovation Center of NeuroregenerationNantong UniversityNantongPR China
| | - Yongjun Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of EducationCo‐innovation Center of NeuroregenerationNantong UniversityNantongPR China
| | - Yingjie Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of EducationCo‐innovation Center of NeuroregenerationNantong UniversityNantongPR China
| |
Collapse
|
108
|
Mitra S, Gera R, Linderoth B, Lind G, Wahlberg L, Almqvist P, Behbahani H, Eriksdotter M. A Review of Techniques for Biodelivery of Nerve Growth Factor (NGF) to the Brain in Relation to Alzheimer's Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1331:167-191. [PMID: 34453298 DOI: 10.1007/978-3-030-74046-7_11] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
Abstract
Age-dependent progressive neurodegeneration and associated cognitive dysfunction represent a serious concern worldwide. Currently, dementia accounts for the fifth highest cause of death, among which Alzheimer's disease (AD) represents more than 60% of the cases. AD is associated with progressive cognitive dysfunction which affects daily life of the affected individual and associated family. The cognitive dysfunctions are at least partially due to the degeneration of a specific set of neurons (cholinergic neurons) whose cell bodies are situated in the basal forebrain region (basal forebrain cholinergic neurons, BFCNs) but innervate wide areas of the brain. It has been explicitly shown that the delivery of the neurotrophic protein nerve growth factor (NGF) can rescue BFCNs and restore cognitive dysfunction, making NGF interesting as a potential therapeutic substance for AD. Unfortunately, NGF cannot pass through the blood-brain barrier (BBB) and thus peripheral administration of NGF protein is not viable therapeutically. NGF must be delivered in a way which will allow its brain penetration and availability to the BFCNs to modulate BFCN activity and viability. Over the past few decades, various methodologies have been developed to deliver NGF to the brain tissue. In this chapter, NGF delivery methods are discussed in the context of AD.
Collapse
Affiliation(s)
- Sumonto Mitra
- Division of Clinical Geriatrics, NVS Department, Karolinska Institutet, Stockholm, Sweden.
| | - Ruchi Gera
- Division of Clinical Geriatrics, NVS Department, Karolinska Institutet, Stockholm, Sweden
| | - Bengt Linderoth
- Section of Neurosurgery, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Göran Lind
- Section of Neurosurgery, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | | | - Per Almqvist
- Section of Neurosurgery, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Homira Behbahani
- Division of Clinical Geriatrics, NVS Department, Karolinska Institutet, Stockholm, Sweden.,Karolinska Universitets laboratoriet (LNP5), Karolinska University Hospital, Stockholm, Sweden
| | - Maria Eriksdotter
- Division of Clinical Geriatrics, NVS Department, Karolinska Institutet, Stockholm, Sweden.,Theme Aging, Karolinska University Hospital, Huddinge, Sweden
| |
Collapse
|
109
|
Kong M, Zhou T, Xiang B. Expression of the axon guidance factor Slit2 and its receptor Robo1 in patients with Hirschsprung disease: An observational study. Medicine (Baltimore) 2021; 100:e26981. [PMID: 34414975 PMCID: PMC8376357 DOI: 10.1097/md.0000000000026981] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 07/12/2021] [Accepted: 07/27/2021] [Indexed: 02/05/2023] Open
Abstract
ABSTRACT Hirschsprung disease (HD) is a common form of digestive tract malformation in children. However, the pathogenesis of HD is not very clear. This study aimed to investigate the expression of slit guidance ligand 2 (Slit2) and roundabout 1 (Robo1) in patients with HD.From January 2018 to January 2019, 30 colon specimens from children with HD undergoing surgical resection at the Department of Surgery in Qilu Children's Hospital of Shandong University were obtained. These specimens were divided into the normal segment group, the transitional segment group and the spastic segment group. Immunohistochemical staining, Western blotting, and real-time polymerase chain reaction were used to measure the expression of Slit2 and Robo1 in the intestinal walls of normal, transitional, and spastic segments.Immunohistochemical staining and Western blot analyses showed high levels of the Slit2 and Robo1 proteins in normal ganglion cells in children with HD, lower levels in transitional ganglion cells, and no expression in spastic segments, with significant differences between groups (P < .05). Similarly, the real-time polymerase chain reaction results were consistent with the Western blot analysis results.The expression of Slit2 and Robo1 decreases significantly in the spastic segment of the intestinal tract in patients with HD.
Collapse
Affiliation(s)
- Meng Kong
- Department of Pediatric Surgery, Qilu Children's Hospital of Shandong University, Jinan, China
| | - Tao Zhou
- Department of Pediatric Surgery, Dazhou Central Hospital, Dazhou, China
| | - Bo Xiang
- Department of Pediatric Surgery, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
110
|
袁 金, 吴 德, 王 文, 段 军, 许 晓, 唐 久. A prospective randomized controlled study on mouse nerve growth factor in the treatment of global developmental delay in children. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2021; 23:786-790. [PMID: 34511166 PMCID: PMC8428910 DOI: 10.7499/j.issn.1008-8830.2106042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 07/05/2021] [Indexed: 06/13/2023]
Abstract
OBJECTIVES To study the clinical effect of mouse nerve growth factor (mNGF) in the treatment of children with global developmental delay (GDD). METHODS A prospective clinical trial was conducted in 60 children with GDD who were treated in the First Affiliated Hospital of Anhui Medical University between July 2016 and July 2017. These children were randomly divided into two groups: conventional rehabilitation treatment and mNGF treatment group (n=30 each). The children in the conventional rehabilitation treatment group were given neurodevelopmental therapy, and those in the mNGF treatment group were given mNGF treatment in addition to the treatment in the control group. The evaluation results of the Gesell Developmental Scale were compared between the two groups before and after treatment. RESULTS Before treatment and after 1.5 months of treatment, there was no significant difference in the developmental quotient (DQ) of each functional area of the Gesell Developmental Scale between the mNGF treatment and conventional rehabilitation treatment groups (P>0.05). After 3 months of treatment, the mNGF treatment group had significantly higher DQs of gross motor, fine motor, and personal-social interaction than the conventional rehabilitation treatment group (P˂0.05). The incidence rate of transient injection site pain after injection of mNGF was 7% (2/30), and there was no epilepsy or other serious adverse reactions. CONCLUSIONS In children with GDD, routine rehabilitation training combined with mNGF therapy can significantly improve their cognitive, motor, and social abilities.
Collapse
|
111
|
Alastra G, Aloe L, Baldassarro VA, Calzà L, Cescatti M, Duskey JT, Focarete ML, Giacomini D, Giardino L, Giraldi V, Lorenzini L, Moretti M, Parmeggiani I, Sannia M, Tosi G. Nerve Growth Factor Biodelivery: A Limiting Step in Moving Toward Extensive Clinical Application? Front Neurosci 2021; 15:695592. [PMID: 34335170 PMCID: PMC8319677 DOI: 10.3389/fnins.2021.695592] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 06/21/2021] [Indexed: 12/11/2022] Open
Abstract
Nerve growth factor (NGF) was the first-discovered member of the neurotrophin family, a class of bioactive molecules which exerts powerful biological effects on the CNS and other peripheral tissues, not only during development, but also during adulthood. While these molecules have long been regarded as potential drugs to combat acute and chronic neurodegenerative processes, as evidenced by the extensive data on their neuroprotective properties, their clinical application has been hindered by their unexpected side effects, as well as by difficulties in defining appropriate dosing and administration strategies. This paper reviews aspects related to the endogenous production of NGF in healthy and pathological conditions, along with conventional and biomaterial-assisted delivery strategies, in an attempt to clarify the impediments to the clinical application of this powerful molecule.
Collapse
Affiliation(s)
- Giuseppe Alastra
- Interdepartmental Centre for Industrial Research in Health Sciences and Technologies, University of Bologna, Bologna, Italy
| | | | - Vito Antonio Baldassarro
- Interdepartmental Centre for Industrial Research in Health Sciences and Technologies, University of Bologna, Bologna, Italy
| | - Laura Calzà
- Interdepartmental Centre for Industrial Research in Health Sciences and Technologies, University of Bologna, Bologna, Italy
- IRET Foundation, Bologna, Italy
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | | | - Jason Thomas Duskey
- Nanotech Laboratory, TeFarTI Center, Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Maria Letizia Focarete
- Interdepartmental Centre for Industrial Research in Health Sciences and Technologies, University of Bologna, Bologna, Italy
- Department of Chemistry “Giacomo Ciamician”, University of Bologna, Bologna, Italy
| | - Daria Giacomini
- Interdepartmental Centre for Industrial Research in Health Sciences and Technologies, University of Bologna, Bologna, Italy
- Department of Chemistry “Giacomo Ciamician”, University of Bologna, Bologna, Italy
| | - Luciana Giardino
- IRET Foundation, Bologna, Italy
- Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
| | - Valentina Giraldi
- Interdepartmental Centre for Industrial Research in Health Sciences and Technologies, University of Bologna, Bologna, Italy
- Department of Chemistry “Giacomo Ciamician”, University of Bologna, Bologna, Italy
| | - Luca Lorenzini
- Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
| | | | - Irene Parmeggiani
- Nanotech Laboratory, TeFarTI Center, Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Michele Sannia
- Interdepartmental Centre for Industrial Research in Health Sciences and Technologies, University of Bologna, Bologna, Italy
| | - Giovanni Tosi
- Nanotech Laboratory, TeFarTI Center, Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
112
|
LianXia Formula Granule Attenuates Cardiac Sympathetic Remodeling in Rats with Myocardial Infarction via the NGF/TrKA/PI3K/AKT Signaling Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:5536406. [PMID: 34221073 PMCID: PMC8213506 DOI: 10.1155/2021/5536406] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 04/08/2021] [Accepted: 05/04/2021] [Indexed: 01/24/2023]
Abstract
Sympathetic remodeling may cause severe arrhythmia after myocardial infarction (MI). Thus, targeting this process may be an effective strategy for clinical prevention of arrhythmias. LianXia Formula Granule (LXFG) can effectively improve the symptoms of patients with arrhythmia after MI, and modern pharmacological studies have shown that Coptidis Rhizoma and Rhizoma Pinelliae Preparata, the components of LXFG, have antiarrhythmia effects. Here, we investigated whether LXFG can mitigate sympathetic remodeling and suppress arrhythmia and then elucidated its underlying mechanism of action in rats after MI. Sprague-Dawley (SD) rats that had undergone a myocardial infarction model were randomly divided into 6 groups, namely, sham, model, metoprolol, and LXFG groups, with high, medium, and low dosages. We exposed the animals to 30 days of treatment and then evaluated incidence of arrhythmia and arrhythmia scores in vivo using programmed electrical stimulation. Moreover, we determined plasma catecholamines contents via enzyme-linked immunosorbent assay and detected expression of tyrosine hydroxylase (TH) at infarcted border zones via western blot, real-time PCR, and immunohistochemical analyses to assess sympathetic remodeling. Finally, we measured key molecules involved in the NGF/TrKA/PI3K/AKT pathways via western blot and real-time PCR. Compared with the model group, treatment with high dose of LXFG suppressed arrhythmia incidence and arrhythmia scores. In addition, all the LXFG groups significantly decreased protein and mRNA levels of TH, improved the average optical density of TH-positive nerve fibers, and reduced the levels of plasma catecholamines relative to the model group. Meanwhile, expression analysis revealed that key molecules in the NGF/TrKA/PI3K/AKT pathways were downregulated in the LXFG group when compared with model group. Overall, these findings indicate that LXFG suppresses arrhythmia and attenuates sympathetic remodeling in rats after MI. The mechanism is probably regulated by suppression of the NGF/TrKA/PI3K/AKT signaling pathway.
Collapse
|
113
|
Gascon S, Jann J, Langlois-Blais C, Plourde M, Lavoie C, Faucheux N. Peptides Derived from Growth Factors to Treat Alzheimer's Disease. Int J Mol Sci 2021; 22:ijms22116071. [PMID: 34199883 PMCID: PMC8200100 DOI: 10.3390/ijms22116071] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 05/30/2021] [Accepted: 06/01/2021] [Indexed: 12/13/2022] Open
Abstract
Alzheimer's disease (AD) is a devastating neurodegenerative disease characterized by progressive neuron losses in memory-related brain structures. The classical features of AD are a dysregulation of the cholinergic system, the accumulation of amyloid plaques, and neurofibrillary tangles. Unfortunately, current treatments are unable to cure or even delay the progression of the disease. Therefore, new therapeutic strategies have emerged, such as the exogenous administration of neurotrophic factors (e.g., NGF and BDNF) that are deficient or dysregulated in AD. However, their low capacity to cross the blood-brain barrier and their exorbitant cost currently limit their use. To overcome these limitations, short peptides mimicking the binding receptor sites of these growth factors have been developed. Such peptides can target selective signaling pathways involved in neuron survival, differentiation, and/or maintenance. This review focuses on growth factors and their derived peptides as potential treatment for AD. It describes (1) the physiological functions of growth factors in the brain, their neuronal signaling pathways, and alteration in AD; (2) the strategies to develop peptides derived from growth factor and their capacity to mimic the role of native proteins; and (3) new advancements and potential in using these molecules as therapeutic treatments for AD, as well as their limitations.
Collapse
Affiliation(s)
- Suzanne Gascon
- Laboratory of Cell-Biomaterial Biohybrid Systems, Department of Chemical and Biotechnological Engineering, 2500 Boulevard Université, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada; (S.G.); (J.J.)
| | - Jessica Jann
- Laboratory of Cell-Biomaterial Biohybrid Systems, Department of Chemical and Biotechnological Engineering, 2500 Boulevard Université, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada; (S.G.); (J.J.)
| | - Chloé Langlois-Blais
- Département de Pharmacologie-Physiologie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada;
| | - Mélanie Plourde
- Centre de Recherche sur le Vieillissement, Centre Intégré Universitaire de Santé et Services Sociaux de l’Estrie–Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC J1G 1B1, Canada;
- Département de Médecine, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Christine Lavoie
- Département de Pharmacologie-Physiologie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada;
- Institut de Pharmacologie de Sherbrooke, 3001 12th Avenue, N., Sherbrooke, QC J1H 5N4, Canada
- Correspondence: (C.L.); (N.F.); Tel.: +1-819-821-8000 (ext. 72732) (C.L.); +1-819-821-8000 (ext. 61343) (N.F.)
| | - Nathalie Faucheux
- Laboratory of Cell-Biomaterial Biohybrid Systems, Department of Chemical and Biotechnological Engineering, 2500 Boulevard Université, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada; (S.G.); (J.J.)
- Institut de Pharmacologie de Sherbrooke, 3001 12th Avenue, N., Sherbrooke, QC J1H 5N4, Canada
- Correspondence: (C.L.); (N.F.); Tel.: +1-819-821-8000 (ext. 72732) (C.L.); +1-819-821-8000 (ext. 61343) (N.F.)
| |
Collapse
|
114
|
Morton AB, Jacobsen NL, Segal SS. Functionalizing biomaterials to promote neurovascular regeneration following skeletal muscle injury. Am J Physiol Cell Physiol 2021; 320:C1099-C1111. [PMID: 33852364 PMCID: PMC8285637 DOI: 10.1152/ajpcell.00501.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 03/29/2021] [Accepted: 04/06/2021] [Indexed: 12/18/2022]
Abstract
During embryogenesis, blood vessels and nerves develop with similar branching structure in response to shared signaling pathways guiding network growth. With both systems integral to physiological homeostasis, dual targeting of blood vessels and nerves to promote neurovascular regeneration following injury is an emerging therapeutic approach in biomedical engineering. A limitation to this strategy is that the nature of cross talk between emergent vessels and nerves during regeneration in an adult is poorly understood. Following peripheral nerve transection, intraneural vascular cells infiltrate the site of injury to provide a migratory pathway for mobilized Schwann cells of regenerating axons. As Schwann cells demyelinate, they secrete vascular endothelial growth factor, which promotes angiogenesis. Recent advances point to concomitant restoration of neurovascular architecture and function through simultaneous targeting of growth factors and guidance cues shared by both systems during regeneration. In the context of traumatic injury associated with volumetric muscle loss, we consider the nature of biomaterials used to engineer three-dimensional scaffolds, functionalization of scaffolds with molecular signals that guide and promote neurovascular growth, and seeding scaffolds with progenitor cells. Physiological success is defined by each tissue component of the bioconstruct (nerve, vessel, muscle) becoming integrated with that of the host. Advances in microfabrication, cell culture techniques, and progenitor cell biology hold great promise for engineering bioconstructs able to restore organ function after volumetric muscle loss.
Collapse
Affiliation(s)
- Aaron B Morton
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - Nicole L Jacobsen
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - Steven S Segal
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
- Dalton Cardiovascular Research Center, Columbia, Missouri
| |
Collapse
|
115
|
Liu Z, Wu H, Huang S. Role of NGF and its receptors in wound healing (Review). Exp Ther Med 2021; 21:599. [PMID: 33884037 PMCID: PMC8056114 DOI: 10.3892/etm.2021.10031] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 03/08/2021] [Indexed: 12/17/2022] Open
Abstract
Wound healing is an important and complicated process that includes four highly integrated and overlapping phases, haemostasis, inflammation, proliferation and tissue remodelling. Nerve growth factor (NGF) was the first member of a family of neurotrophic factors to be discovered, and is an essential neurotrophic factor for the development and maintenance of the central and peripheral nervous systems. Several studies have proposed that NGF and its receptors, tropomyosin-related kinase receptor 1 and NGF receptor, are involved in the wound healing process, and are important components of the healing of several wounds both in vivo and in vitro. Topical application of NGF significantly promotes the healing of different types of wounds, including diabetic foot ulcers, pressure ulcers and corneal wounds. The present review summarizes the status of NGF and its receptors in current literature, and discusses data obtained in the last few years on the healing action of NGF in cutaneous, corneal and oral wounds.
Collapse
Affiliation(s)
- Zhenxing Liu
- Department of Oral and Maxillofacial Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, P.R. China
| | - Haiwei Wu
- Department of Oral and Maxillofacial Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, P.R. China
| | - Shengyun Huang
- Department of Oral and Maxillofacial Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, P.R. China
| |
Collapse
|
116
|
Moattari CR, Granstein RD. Neuropeptides and neurohormones in immune, inflammatory and cellular responses to ultraviolet radiation. Acta Physiol (Oxf) 2021; 232:e13644. [PMID: 33724698 DOI: 10.1111/apha.13644] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 02/26/2021] [Accepted: 03/02/2021] [Indexed: 12/16/2022]
Abstract
Humans are exposed to varying amounts of ultraviolet radiation (UVR) through sunlight. UVR penetrates into human skin leading to release of neuropeptides, neurotransmitters and neuroendocrine hormones. These messengers released from local sensory nerves, keratinocytes, Langerhans cells (LCs), mast cells, melanocytes and endothelial cells (ECs) modulate local and systemic immune responses, mediate inflammation and promote differing cell biologic effects. In this review, we will focus on both animal and human studies that elucidate the roles of calcitonin gene-related peptide (CGRP), substance P (SP), nerve growth factor (NGF), nitric oxide and proopiomelanocortin (POMC) derivatives in mediating immune and inflammatory effects of exposure to UVR as well as other cell biologic effects of UVR exposure.
Collapse
|
117
|
Luo W, Gong Y, Qiu F, Yuan Y, Jia W, Liu Z, Gao L. NGF nanoparticles enhance the potency of transplanted human umbilical cord mesenchymal stem cells for myocardial repair. Am J Physiol Heart Circ Physiol 2021; 320:H1959-H1974. [PMID: 33769916 DOI: 10.1152/ajpheart.00855.2020] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In this study, we investigated whether human umbilical cord mesenchymal stem cell (hUCMSC) fibrin patches loaded with nerve growth factor (NGF) poly(lactic-co-glycolic acid) (PLGA) nanoparticles could enhance the therapeutic potency of hUCMSCs for myocardial infarction (MI). In vitro, NGF significantly improved the proliferation of hUCMSCs and mitigated cytotoxicity and apoptosis under hypoxic injury. NGF also promoted the paracrine effects of hUCMSCs on angiogenesis and cardiomyocyte protection. The tyrosine kinase A (TrkA) and phosphoinositide 3-kinase (PI3K)-serine/threonine protein kinase (Akt) signaling pathways in hUCMSCs were involved in the NGF-induced protection. NGF PLGA nanoparticles continued to release NGF for at least 1 mo and also exerted a protective effect on hUCMSCs, the same with free NGF. In vivo, we treated MI mice with nothing (MI group), a cell-free fibrin patch with blank PLGA nanoparticles (MI + OP group), a cell-free fibrin patch with NGF nanoparticles (MI + NGF group), and hUCMSC fibrin patches with blank PLGA nanoparticles (MI + MSC group) or NGF PLGA nanoparticles (MSC + NGF group). Among these groups, the MSC + NGF group exhibited the best cardiac contractile function, the smallest infarct size, and the thickest ventricular wall. The application of NGF PLGA nanoparticles significantly improved the retention of transplanted hUCMSCs and enhanced their ability to reduce myocardial apoptosis and promote angiogenesis in the mouse heart after MI. These findings demonstrate the promising therapeutic potential of hUCMSC fibrin cardiac patches loaded with NGF PLGA nanoparticles.NEW & NOTEWORTHY NGF PLGA nanoparticles can exert a protective effect on hUCMSCs and promote the paracrine effects of hUCMSCs on angiogenesis and cardiomyocyte protection through TrkA-PI3K/Akt signaling pathway, the same with free NGF. The application of NGF PLGA nanoparticles in the hUCMSC fibrin cardiac patches can significantly improve the retention of transplanted hUCMSCs and enhance their ability to reduce myocardial apoptosis and promote angiogenesis in the mouse heart after MI.
Collapse
Affiliation(s)
- Wei Luo
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China.,Department of Cardiovascular and Thoracic Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yanshan Gong
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Fan Qiu
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China.,Department of Cardiovascular and Thoracic Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yi Yuan
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wenwen Jia
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zhongmin Liu
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China.,Department of Cardiovascular and Thoracic Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China.,Shanghai Institute of Stem Cell Research and Clinical translation, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Ling Gao
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China.,Shanghai Institute of Stem Cell Research and Clinical translation, Shanghai East Hospital, Tongji University, Shanghai, China
| |
Collapse
|
118
|
Aloe L, Rocco ML, Balzamino BO, Esposito G, Micera A. Retrobulbar administration of purified anti-nerve growth factor in developing rats induces structural and biochemical changes in the retina and cornea. Int J Ophthalmol 2021; 14:209-216. [PMID: 33614448 DOI: 10.18240/ijo.2021.02.05] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 08/26/2020] [Indexed: 12/14/2022] Open
Abstract
AIM To develop an experimental model of endogenous nerve growth factor (NGF) deprivation by retrobulbar administration of purified neutralizing anti-NGF antibodies in young Sprague-Dawley rats and provide further information on NGF expression in the retina and cornea. METHODS Sixty old pathogen-free Sprague Dawley rats (p14, post-natal days) were treated with repeated retrobulbar injections of neutralizing anti-NGF (2 µL, 100 µg/mL, every 3d). After 2wk (p28), retinal and corneal tissues were investigated for morphological, biochemical, and molecular expression of trkANGFR by using Western blotting or immunofluorescence. Rhodopsin as well as protein profile expression were also investigated. RESULTS Chronic retrobulbar neutralizing anti-NGF antibodies changed the distribution of trkANGFR immunoreactivity at retinal level, while no changes were detected for global trkANGFR protein expression. By contrary, the treatment resulted in the increase of corneal trkANGFR expression. Retinal tissues showed a decreased rhodopsin expression as well as reduced number of both rhodopsin expressing and total retinal cells, as observed after single cell extraction. A decreased expression of ICAM-1, IL-17 and IL-13 as well as an increased expression of IL-21 typified retinal extracts. No significant changes were observed for corneal tissues. CONCLUSION The reduced availability of endogenous NGF, as produced by chronic retrobulbar anti-NGF administration, produce a quick response from retinal tissues, with respect to corneal ones, suggesting the presence of early compensatory mechanisms to protect retinal networking.
Collapse
Affiliation(s)
- Luigi Aloe
- Institute of Cell Biology and Neurobiology, CNR, Lazio 00143, Rome, Italy.,Fondazione IRET, Ozzano Emilia, Bologna 40064, Italy
| | - Maria Luisa Rocco
- Fondazione IRET, Ozzano Emilia, Bologna 40064, Italy.,Institute of Translational Pharmacology, CNR, Lazio 00133, Rome, Italy
| | - Bijorn Omar Balzamino
- Research and Development Laboratory for Biochemical, Molecular and Cellular Applications in Ophthalmological Science, IRCCS, Fondazione Bietti, Rome 00182, Italy
| | - Graziana Esposito
- Research and Development Laboratory for Biochemical, Molecular and Cellular Applications in Ophthalmological Science, IRCCS, Fondazione Bietti, Rome 00182, Italy
| | - Alessandra Micera
- Research and Development Laboratory for Biochemical, Molecular and Cellular Applications in Ophthalmological Science, IRCCS, Fondazione Bietti, Rome 00182, Italy
| |
Collapse
|
119
|
Zha K, Yang Y, Tian G, Sun Z, Yang Z, Li X, Sui X, Liu S, Zhao J, Guo Q. Nerve growth factor (NGF) and NGF receptors in mesenchymal stem/stromal cells: Impact on potential therapies. Stem Cells Transl Med 2021; 10:1008-1020. [PMID: 33586908 PMCID: PMC8235142 DOI: 10.1002/sctm.20-0290] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 12/27/2020] [Accepted: 01/12/2021] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) are promising for the treatment of degenerative diseases and traumatic injuries. However, MSC engraftment is not always successful and requires a strong comprehension of the cytokines and their receptors that mediate the biological behaviors of MSCs. The effects of nerve growth factor (NGF) and its two receptors, TrkA and p75NTR, on neural cells are well studied. Increasing evidence shows that NGF, TrkA, and p75NTR are also involved in various aspects of MSC function, including their survival, growth, differentiation, and angiogenesis. The regulatory effect of NGF on MSCs is thought to be achieved mainly through its binding to TrkA. p75NTR, another receptor of NGF, is regarded as a novel surface marker of MSCs. This review provides an overview of advances in understanding the roles of NGF and its receptors in MSCs as well as the effects of MSC‐derived NGF on other cell types, which will provide new insight for the optimization of MSC‐based therapy.
Collapse
Affiliation(s)
- Kangkang Zha
- Medical School of Chinese PLA, Beijing, People's Republic of China.,Institute of Orthopaedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma and War Injuries, PLA, Beijing, People's Republic of China.,School of Medicine, Nankai University, Tianjin, People's Republic of China
| | - Yu Yang
- Department of Othopaedics, Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, People's Republic of China
| | - Guangzhao Tian
- Medical School of Chinese PLA, Beijing, People's Republic of China.,Institute of Orthopaedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma and War Injuries, PLA, Beijing, People's Republic of China.,School of Medicine, Nankai University, Tianjin, People's Republic of China
| | - Zhiqiang Sun
- Medical School of Chinese PLA, Beijing, People's Republic of China.,Institute of Orthopaedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma and War Injuries, PLA, Beijing, People's Republic of China.,School of Medicine, Nankai University, Tianjin, People's Republic of China
| | - Zhen Yang
- Medical School of Chinese PLA, Beijing, People's Republic of China.,Institute of Orthopaedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma and War Injuries, PLA, Beijing, People's Republic of China.,School of Medicine, Nankai University, Tianjin, People's Republic of China
| | - Xu Li
- Musculoskeletal Research Laboratory, Department of Orthopaedics and Traumatology, Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, People's Republic of China
| | - Xiang Sui
- Institute of Orthopaedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma and War Injuries, PLA, Beijing, People's Republic of China
| | - Shuyun Liu
- Institute of Orthopaedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma and War Injuries, PLA, Beijing, People's Republic of China
| | - Jinmin Zhao
- Department of Othopaedics, Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, People's Republic of China
| | - Quanyi Guo
- Institute of Orthopaedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma and War Injuries, PLA, Beijing, People's Republic of China
| |
Collapse
|
120
|
Jaworecka K, Muda-Urban J, Rzepko M, Reich A. Molecular Aspects of Pruritus Pathogenesis in Psoriasis. Int J Mol Sci 2021; 22:ijms22020858. [PMID: 33467067 PMCID: PMC7830783 DOI: 10.3390/ijms22020858] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 01/11/2021] [Accepted: 01/13/2021] [Indexed: 12/23/2022] Open
Abstract
Psoriasis is a chronic, systemic inflammatory disease with a genetic background that involves almost 3% of the general population worldwide. Approximately, 70–90% of patients with psoriasis suffer from pruritus, an unpleasant sensation that provokes a desire to scratch. Despite the enormous progress in understanding the mechanisms that cause psoriasis, the pathogenesis of psoriasis-related pruritus still remains unclear. In order to improve patients’ quality of life, development of more effective and safer antipruritic therapies is necessary. In turn to make it possible, better understanding of complexed and multifactorial pathogenesis of this symptom is needed. In this article we have systematized the current knowledge about pruritus origin in psoriasis.
Collapse
Affiliation(s)
- Kamila Jaworecka
- Department of Dermatology, Institute of Medical Sciences, Medical College of Rzeszow University, PL-35-055 Rzeszow, Poland; (K.J.); (J.M.-U.)
| | - Joanna Muda-Urban
- Department of Dermatology, Institute of Medical Sciences, Medical College of Rzeszow University, PL-35-055 Rzeszow, Poland; (K.J.); (J.M.-U.)
| | - Marian Rzepko
- Institute of Physical Culture Sciences, Medical College of Rzeszow University, PL-35-055 Rzeszow, Poland;
| | - Adam Reich
- Department of Dermatology, Institute of Medical Sciences, Medical College of Rzeszow University, PL-35-055 Rzeszow, Poland; (K.J.); (J.M.-U.)
- Correspondence: ; Tel.: +48-605076722
| |
Collapse
|
121
|
Lee SJ, Wang W, Jin L, Lu X, Gao L, Chen Y, Liu T, Emery D, Vukmanic E, Liu Y, Kaplan HJ, Dean DC. Rod photoreceptor clearance due to misfolded rhodopsin is linked to a DAMP-immune checkpoint switch. J Biol Chem 2021; 296:100102. [PMID: 33214223 PMCID: PMC7949052 DOI: 10.1074/jbc.ra120.016053] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 11/13/2020] [Accepted: 11/19/2020] [Indexed: 01/19/2023] Open
Abstract
Chronic endoplasmic reticulum stress resulting from misfolding of the visual pigment rhodopsin (RHO) can lead to loss of rod photoreceptors, which initiates retinitis pigmentosa, characterized initially by diminished nighttime and peripheral vision. Cone photoreceptors depend on rods for glucose transport, which the neurons use for assembly of visual pigment-rich structures; as such, loss of rods also leads to a secondary loss of cone function, diminishing high-resolution color vision utilized for tasks including reading, driving, and facial recognition. If dysfunctional rods could be maintained to continue to serve this secondary cone preservation function, it might benefit patients with retinitis pigmentosa, but the mechanisms by which rods are removed are not fully established. Using pigs expressing mutant RHO, we find that induction of a danger-associated molecular pattern (DAMP) "eat me" signal on the surface of mutant rods is correlated with targeting the live cells for (PrCR) by retinal myeloid cells. Glucocorticoid therapy leads to replacement of this DAMP with a "don't eat me" immune checkpoint on the rod surface and inhibition of PrCR. Surviving rods then continue to promote glucose transport to cones, maintaining their viability.
Collapse
Affiliation(s)
- Sang Joon Lee
- Department of Ophthalmology and Visual Sciences, University of Louisville Health Sciences Center, Louisville, Kentucky, USA; Department of Ophthalmology, Kosin University College of Medicine, Seo-gu, Busan, Korea
| | - Wei Wang
- Department of Ophthalmology and Visual Sciences, University of Louisville Health Sciences Center, Louisville, Kentucky, USA
| | - Lei Jin
- Department of Ophthalmology and Visual Sciences, University of Louisville Health Sciences Center, Louisville, Kentucky, USA; Department of Ophthalmology, The Third People's Hospital of Dalian, Dalian Medical University, Dalian, China
| | - Xiaoqin Lu
- Department of Ophthalmology and Visual Sciences, University of Louisville Health Sciences Center, Louisville, Kentucky, USA; Department of Medicine, University of Louisville Health Sciences Center, Louisville, Kentucky, USA
| | - Lei Gao
- Department of Ophthalmology and Visual Sciences, University of Louisville Health Sciences Center, Louisville, Kentucky, USA; Department of Hematology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Yao Chen
- Department of Ophthalmology and Visual Sciences, University of Louisville Health Sciences Center, Louisville, Kentucky, USA; Department of Ophthalmology, Xiangya Hospital, Central South University, Changsha, China
| | - Tingting Liu
- Department of Ophthalmology and Visual Sciences, University of Louisville Health Sciences Center, Louisville, Kentucky, USA; Department of Ophthalmology, The Third People's Hospital of Dalian, Dalian Medical University, Dalian, China
| | - Douglas Emery
- Department of Ophthalmology and Visual Sciences, University of Louisville Health Sciences Center, Louisville, Kentucky, USA; Department of Medicine, University of Louisville Health Sciences Center, Louisville, Kentucky, USA
| | - Eric Vukmanic
- Department of Ophthalmology and Visual Sciences, University of Louisville Health Sciences Center, Louisville, Kentucky, USA; Department of Medicine, University of Louisville Health Sciences Center, Louisville, Kentucky, USA
| | - Yongqing Liu
- Department of Ophthalmology and Visual Sciences, University of Louisville Health Sciences Center, Louisville, Kentucky, USA; Department of Medicine, University of Louisville Health Sciences Center, Louisville, Kentucky, USA
| | - Henry J Kaplan
- Department of Ophthalmology and Visual Sciences, University of Louisville Health Sciences Center, Louisville, Kentucky, USA
| | - Douglas C Dean
- Department of Ophthalmology and Visual Sciences, University of Louisville Health Sciences Center, Louisville, Kentucky, USA; Department of Medicine, University of Louisville Health Sciences Center, Louisville, Kentucky, USA.
| |
Collapse
|
122
|
Eftimiadi G, Soligo M, Manni L, Di Giuda D, Calcagni ML, Chiaretti A. Topical delivery of nerve growth factor for treatment of ocular and brain disorders. Neural Regen Res 2021; 16:1740-1750. [PMID: 33510063 PMCID: PMC8328750 DOI: 10.4103/1673-5374.306062] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Neurotrophins are a family of proteins that support neuronal proliferation, survival, and differentiation in the central and peripheral nervous systems, and are regulators of neuronal plasticity. Nerve growth factor is one of the best-described neurotrophins and has advanced to clinical trials for treatment of ocular and brain diseases due to its trophic and regenerative properties. Prior trials over the past few decades have produced conflicting results, which have principally been ascribed to adverse effects of systemic nerve growth factor administration, together with poor penetrance of the blood-brain barrier that impairs drug delivery. Contrastingly, recent studies have revealed that topical ocular and intranasal nerve growth factor administration are safe and effective, suggesting that topical nerve growth factor delivery is a potential alternative to both systemic and invasive intracerebral delivery. The therapeutic effects of local nerve growth factor delivery have been extensively investigated for different ophthalmic diseases, including neurotrophic keratitis, glaucoma, retinitis pigmentosa, and dry eye disease. Further, promising pharmacologic effects were reported in an optic glioma model, which indicated that topically administered nerve growth factor diffused far beyond where it was topically applied. These findings support the therapeutic potential of delivering topical nerve growth factor preparations intranasally for acquired and degenerative brain disorders. Preliminary clinical findings in both traumatic and non-traumatic acquired brain injuries are encouraging, especially in pediatric patients, and clinical trials are ongoing. The present review will focus on the therapeutic effects of both ocular and intranasal nerve growth factor delivery for diseases of the brain and eye.
Collapse
Affiliation(s)
- Gemma Eftimiadi
- Institute of Pediatrics, Fondazione Policlinico Universitario Agostino Gemelli, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Marzia Soligo
- Institute of Translational Pharmacology, Consiglio Nazionale delle Ricerche (CNR), Rome, Italy
| | - Luigi Manni
- Institute of Translational Pharmacology, Consiglio Nazionale delle Ricerche (CNR), Rome, Italy
| | - Daniela Di Giuda
- Institute of Nuclear Medicine, Fondazione Policlinico Universitario Agostino Gemelli, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Maria Lucia Calcagni
- Institute of Nuclear Medicine, Fondazione Policlinico Universitario Agostino Gemelli, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Antonio Chiaretti
- Institute of Pediatrics, Fondazione Policlinico Universitario Agostino Gemelli, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| |
Collapse
|
123
|
Zhang S, Hu B, Liu W, Wang P, Lv X, Chen S, Shao Z. The role of structure and function changes of sensory nervous system in intervertebral disc-related low back pain. Osteoarthritis Cartilage 2021; 29:17-27. [PMID: 33007412 DOI: 10.1016/j.joca.2020.09.002] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 08/18/2020] [Accepted: 09/21/2020] [Indexed: 02/06/2023]
Abstract
Low back pain (LBP) is a common musculoskeletal symptom, which can be developed in multiple clinical diseases. It is widely recognized that intervertebral disc (IVD) degeneration (IVDD) is one of the leading causes of LBP. However, the pathogenesis of IVD-related LBP is still controversial, and the treatment means are also insufficient to date. In recent decades, the role of structure and function changes of sensory nervous system in the induction and the maintenance of LBP is drawing more and more attention. With the progress of IVDD, IVD cell exhaustion and extracellular matrix degradation result in IVD structural damage, while neovascularization, innervation and inflammatory activation further deteriorate the microenvironment of IVD. New nerve ingrowth into degenerated IVD amplifies the impacts of IVD-derived nociceptive molecules on sensory endings. Moreover, IVDD is usually accompanied with disc herniation, which could injure and inflame affected nerves. Under mechanical and pro-inflammatory stimulation, the pain-transmitting pathway exhibits a sensitized function state and ultimately leads to LBP. Hence, relevant pathogenic factors, such as neurotrophins, ion channels, inflammatory factors, etc., are supposed to serve as promising therapeutic targets for LBP. The purpose of this review is to comprehensively summarize the current evidence on 1) the pathological changes of sensory nervous system during IVDD and their association with LBP, and 2) potential therapeutic strategies for LBP targeting relevant pathogenic factors.
Collapse
Affiliation(s)
- S Zhang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - B Hu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - W Liu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - P Wang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - X Lv
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - S Chen
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| | - Z Shao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
124
|
Tan W, Zhang Q, Dong Z, Yan Y, Fu Y, Liu X, Zhao B, Duan X. Phosphatidylcholine Ameliorates LPS-Induced Systemic Inflammation and Cognitive Impairments via Mediating the Gut-Brain Axis Balance. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:14884-14895. [PMID: 33289390 DOI: 10.1021/acs.jafc.0c06383] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Systemic inflammation will cause an imbalance in the steady state of the gut-brain axis. Phosphatidylcholine (PC) is a phospholipid found in egg yolk that has anti-inflammatory and antioxidant properties. The present research proved that PC supplementation (60 mg/kg body weight) for 35 days prevented inflammatory responses and behavioral disturbances in lipopolysaccharide (LPS)-induced mice. PC could regulate the expression of neurotrophic factors and synaptic proteins, which effectively alleviated the nerve damage and synaptic dysfunction caused by LPS. In addition, PC supplementation ameliorated gut barrier damage, altered gut genes, and improved gut health by modulating the cell adhesion molecule (CAM) pathway. Furthermore, PC remodeled the gut microbiome structure in the mice of the LPS group by increasing the relative abundance of Rikenellaceae and Lachnospiraceae. PC also increased short-chain fatty acid (SCFA) production in LPS-induced mice, which in turn ameliorated brain inflammatory responses. In conclusion, PC supplementation may be a nutritional strategy for the prevention of systemic inflammation via the gut-brain axis.
Collapse
Affiliation(s)
- Wen Tan
- College of Food Science and Engineering, Northwest A&F University, 22 Xi-nong Road, Yangling 712100, China
| | - Qinjun Zhang
- College of Food Science and Engineering, Northwest A&F University, 22 Xi-nong Road, Yangling 712100, China
| | - Zhijian Dong
- Xi'an Gaoxin Hospital, Xi'an 710000, Shaanxi, China
| | - Yubin Yan
- College of Food Science and Engineering, Northwest A&F University, 22 Xi-nong Road, Yangling 712100, China
| | - Yukun Fu
- College of Food Science and Engineering, Northwest A&F University, 22 Xi-nong Road, Yangling 712100, China
| | - Xuebo Liu
- College of Food Science and Engineering, Northwest A&F University, 22 Xi-nong Road, Yangling 712100, China
| | - Beita Zhao
- College of Food Science and Engineering, Northwest A&F University, 22 Xi-nong Road, Yangling 712100, China
| | - Xiang Duan
- College of Food Science and Engineering, Northwest A&F University, 22 Xi-nong Road, Yangling 712100, China
| |
Collapse
|
125
|
Fo Y, Zhang C, Chen X, Liu X, Ye T, Guo Y, Qu C, Shi S, Yang B. Chronic sigma-1 receptor activation ameliorates ventricular remodeling and decreases susceptibility to ventricular arrhythmias after myocardial infarction in rats. Eur J Pharmacol 2020; 889:173614. [PMID: 33010304 DOI: 10.1016/j.ejphar.2020.173614] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 09/16/2020] [Accepted: 09/28/2020] [Indexed: 11/24/2022]
Abstract
The present study aimed to assess the effect of sigma-1 receptor (S1R) stimulation on ventricular remodeling and susceptibility to ventricular arrhythmias (VAs) after myocardial infarction (MI) in rats. Wild-type male rats were placed into one of the following four treatment groups. For four weeks, animals in the Sham group and MI group received intraperitoneal (i.p.) injections of 0.9% saline (1 ml/kg/day); those in the MI + F group received fluvoxamine (FLV) (0.3 mg/kg/day); and those in the MI + F + BD group received FLV plus BD1047 (0.3 mg/kg/day). After that, the ventricular electrophysiological parameters were measured via the langendorff system. Ventricular fibrosis quantification was determined with Masson staining. Cardiac function was evaluated by echocardiography. The protein levels of S1R, connexin (Cx)43, Cav1.2, Kv4.2, Kv4.3, tyrosine hydroxylase (TH), nerve growth factor (NGF), growth-associated protein 43 (GAP43) were detected by Western blot assays. Our results indicated that fluvoxamine significantly prolonged the ventricular effective refractory period (ERP), shortened action potential duration (APD), reduced susceptibility to VAs after MI. Masson staining showed a decrease in ventricular fibrosis in the MI + F group. Furthermore, the contents of Cx43, S1R, Cav1.2, Kv4.2, Kv4.3 were increased in the MI + F group compared with the MI group (all P < 0.05). The contents of TH, NGF, GAP43 were reduced in the MI + F group compared with the MI group. (all P < 0.05). However, BD1047 reduces all of these effects of FLV. The results suggest that S1R stimulation reduces susceptibility to VAs and improves cardiac function by improving myocardial fibrosis, lightning sympathetic remodeling, electrical remodeling, gap junction remodeling and upregulating S1R content.
Collapse
Affiliation(s)
- Yuhong Fo
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan, 430060, PR China
| | - Cui Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan, 430060, PR China
| | - Xiuhuan Chen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan, 430060, PR China
| | - Xin Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan, 430060, PR China
| | - Tianxin Ye
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan, 430060, PR China
| | - Yan Guo
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan, 430060, PR China
| | - Chuan Qu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan, 430060, PR China
| | - Shaobo Shi
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan, 430060, PR China.
| | - Bo Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan, 430060, PR China.
| |
Collapse
|
126
|
Xu Y, Chen F. Factors and Molecular Mechanisms Influencing the Protein Synthesis, Degradation and Membrane Trafficking of ASIC1a. Front Cell Dev Biol 2020; 8:596304. [PMID: 33195276 PMCID: PMC7644914 DOI: 10.3389/fcell.2020.596304] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 10/05/2020] [Indexed: 12/31/2022] Open
Abstract
Acid-sensing ion channels (ASICs) are members of the degenerin/epithelial sodium channel superfamily. They are extracellular pH sensors that are activated by protons. Among all ASICs, ASIC1a is one of the most intensively studied isoforms because of its unique ability to be permeable to Ca2+. In addition, it is considered to contribute to various pathophysiological conditions. As a membrane proton receptor, the number of ASIC1a present on the cell surface determines its physiological and pathological functions, and this number partially depends on protein synthesis, degradation, and membrane trafficking processes. Recently, several studies have shown that various factors affect these processes. Therefore, this review elucidated the major factors and underlying molecular mechanisms affecting ASIC1a protein expression and membrane trafficking.
Collapse
Affiliation(s)
- Yayun Xu
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China.,The Key Laboratory of Major Autoimmune Diseases of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China
| | - Feihu Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China.,The Key Laboratory of Major Autoimmune Diseases of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China
| |
Collapse
|
127
|
Rhode SC, Beier JP, Ruhl T. Adipose tissue stem cells in peripheral nerve regeneration-In vitro and in vivo. J Neurosci Res 2020; 99:545-560. [PMID: 33070351 DOI: 10.1002/jnr.24738] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/16/2020] [Accepted: 09/30/2020] [Indexed: 12/12/2022]
Abstract
After peripheral nerve injury, Schwann cells (SCs) are crucially involved in several steps of the subsequent regenerative processes, such as the Wallerian degeneration. They promote lysis and phagocytosis of myelin, secrete numbers of neurotrophic factors and cytokines, and recruit macrophages for a biological debridement. However, nerve injuries with a defect size of >1 cm do not show proper tissue regeneration and require a surgical nerve gap reconstruction. To find a sufficient alternative to the current gold standard-the autologous nerve transplant-several cell-based therapies have been developed and were experimentally investigated. One approach aims on the use of adipose tissue stem cells (ASCs). These are multipotent mesenchymal stromal cells that can differentiate into multiple phenotypes along the mesodermal lineage, such as osteoblasts, chondrocytes, and myocytes. Furthermore, ASCs also possess neurotrophic features, that is, they secrete neurotrophic factors like the nerve growth factor, brain-derived neurotrophic factor, neurotrophin-3, ciliary neurotrophic factor, glial cell-derived neurotrophic factor, and artemin. They can also differentiate into the so-called Schwann cell-like cells (SCLCs). These cells share features with naturally occurring SCs, as they also promote nerve regeneration in the periphery. This review gives a comprehensive overview of the use of ASCs in peripheral nerve regeneration and peripheral nerve tissue engineering both in vitro and in vivo. While the sustainability of differentiation of ASCs to SCLCs in vivo is still questionable, ASCs used with different nerve conduits, such as hydrogels or silk fibers, have been shown to promote nerve regeneration.
Collapse
Affiliation(s)
- Sophie Charlotte Rhode
- Department of Plastic Surgery, Hand Surgery and Burn Center, University Hospital RWTH Aachen, Aachen, Germany
| | - Justus Patrick Beier
- Department of Plastic Surgery, Hand Surgery and Burn Center, University Hospital RWTH Aachen, Aachen, Germany
| | - Tim Ruhl
- Department of Plastic Surgery, Hand Surgery and Burn Center, University Hospital RWTH Aachen, Aachen, Germany
| |
Collapse
|
128
|
Popova NK, Kulikov AV, Naumenko VS. Spaceflight and brain plasticity: Spaceflight effects on regional expression of neurotransmitter systems and neurotrophic factors encoding genes. Neurosci Biobehav Rev 2020; 119:396-405. [PMID: 33086127 DOI: 10.1016/j.neubiorev.2020.10.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 07/14/2020] [Accepted: 10/13/2020] [Indexed: 12/13/2022]
Abstract
The critical problem of space exploration is the effect of long-term space travel on brain functioning. Current information concerning the effects of actual spaceflight on the brain was obtained on rats and mice flown on five missions of Soviet/Russian biosatellites, NASA Neurolab Mission STS90, and International Space Station (ISS). The review provides converging lines of evidence that: 1) long-term spaceflight affects both principle regulators of brain neuroplasticity - neurotransmitters (5-HT and DA) and neurotrophic factors (CDNF, GDNF but not BDNF); 2) 5-HT- (5-HT2A receptor and MAO A) and especially DA-related genes (TH, MAO A, COMT, D1 receptor, CDNF and GDNF) belong to the risk neurogenes; 3) brain response to spaceflight is region-specific. Substantia nigra, striatum and hypothalamus are highly sensitive to the long-term spaceflight: in these brain areas spaceflight decreased the expression of both DA-related and neurotrophic factors genes. Since DA system is involved in the regulation of movement and cognition the data discussed in the review could explain dysfunction of locomotion and behavior of astronauts and direct further investigations to the DA system.
Collapse
Affiliation(s)
- Nina K Popova
- Institute of Cytology and Genetics, Siberian Division of Russian Academy of Sciences, Novosibirsk, 630090, Russia.
| | - Alexander V Kulikov
- Institute of Cytology and Genetics, Siberian Division of Russian Academy of Sciences, Novosibirsk, 630090, Russia
| | - Vladimir S Naumenko
- Institute of Cytology and Genetics, Siberian Division of Russian Academy of Sciences, Novosibirsk, 630090, Russia.
| |
Collapse
|
129
|
Rascher M, Wittstein K, Winter B, Rupcic Z, Wolf-Asseburg A, Stadler M, Köster RW. Erinacine C Activates Transcription from a Consensus ETS DNA Binding Site in Astrocytic Cells in Addition to NGF Induction. Biomolecules 2020; 10:E1440. [PMID: 33066380 PMCID: PMC7602259 DOI: 10.3390/biom10101440] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 10/08/2020] [Accepted: 10/13/2020] [Indexed: 11/16/2022] Open
Abstract
Medicinal mushrooms of the genus Hericium are known to produce secondary metabolites with homeostatic properties for the central nervous system. We and others have recently demonstrated that among these metabolites cyathane diterpenoids and in particular erinacine C possess potent neurotrophin inducing properties in astrocytic cells. Yet, the signaling events downstream of erinacine C induced neurotrophin acitivity in neural-like adrenal phaeochromocytoma cells (PC12) cells have remained elusive. Similar, signaling events activated by erinacine C in astrocytic cells are unknown. Using a combination of genetic and pharmacological inhibitors we show that erinacine C induced neurotrophic activity mediates PC12 cell differentiation via the TrkA receptor and likely its associated PLCγ-, PI3K-, and MAPK/ERK pathways. Furthermore, a small library of transcriptional activation reporters revealed that erinacine C induces transcriptional activation mediated by DNA consensus binding sites of selected conserved transcription factor families. Among these, transcription is activated from an ETS consensus in a concentration dependent manner. Interestingly, induced ETS-consensus transcription occurs in parallel and independent of neurotrophin induction. This finding helps to explain the many pleiotropic functions of cyathane diterpenoids. Moreover, our studies provide genetic access to cyathane diterpenoid functions in astrocytic cells and help to mechanistically understand the action of cyathanes in glial cells.
Collapse
Affiliation(s)
- Monique Rascher
- Division of Cellular and Molecular Biology, Zoological Institute, Technische Universität Braunschweig, Spielmannstraße 7, 38106 Braunschweig, Germany; (M.R.); (B.W.); (A.W.-A.)
- Department Microbial Drugs, Helmholtz Centre for Infection Research GmbH and Institute of Microbiology, Technische Universität Braunschweig, Inhoffenstraße 7, 38124 Braunschweig, Germany; (K.W.); (Z.R.)
- German Centre for Infection Research (DZIF), Technische Universität Braunschweig, Partner Site Hannover-Braunschweig, 38124 Braunschweig, Germany
| | - Kathrin Wittstein
- Department Microbial Drugs, Helmholtz Centre for Infection Research GmbH and Institute of Microbiology, Technische Universität Braunschweig, Inhoffenstraße 7, 38124 Braunschweig, Germany; (K.W.); (Z.R.)
- German Centre for Infection Research (DZIF), Technische Universität Braunschweig, Partner Site Hannover-Braunschweig, 38124 Braunschweig, Germany
| | - Barbara Winter
- Division of Cellular and Molecular Biology, Zoological Institute, Technische Universität Braunschweig, Spielmannstraße 7, 38106 Braunschweig, Germany; (M.R.); (B.W.); (A.W.-A.)
| | - Zeljka Rupcic
- Department Microbial Drugs, Helmholtz Centre for Infection Research GmbH and Institute of Microbiology, Technische Universität Braunschweig, Inhoffenstraße 7, 38124 Braunschweig, Germany; (K.W.); (Z.R.)
- German Centre for Infection Research (DZIF), Technische Universität Braunschweig, Partner Site Hannover-Braunschweig, 38124 Braunschweig, Germany
| | - Alexandra Wolf-Asseburg
- Division of Cellular and Molecular Biology, Zoological Institute, Technische Universität Braunschweig, Spielmannstraße 7, 38106 Braunschweig, Germany; (M.R.); (B.W.); (A.W.-A.)
| | - Marc Stadler
- Department Microbial Drugs, Helmholtz Centre for Infection Research GmbH and Institute of Microbiology, Technische Universität Braunschweig, Inhoffenstraße 7, 38124 Braunschweig, Germany; (K.W.); (Z.R.)
- German Centre for Infection Research (DZIF), Technische Universität Braunschweig, Partner Site Hannover-Braunschweig, 38124 Braunschweig, Germany
- Institute of Microbiology, Technische Universität Braunschweig, Spielmannstraße 7, 38106 Braunschweig, Germany
| | - Reinhard W. Köster
- Division of Cellular and Molecular Biology, Zoological Institute, Technische Universität Braunschweig, Spielmannstraße 7, 38106 Braunschweig, Germany; (M.R.); (B.W.); (A.W.-A.)
| |
Collapse
|
130
|
Xiao S, Zhang F, Zheng Y, Liu Z, Wang D, Wei Z, Deng C. Synergistic effect of nanofat and mouse nerve-growth factor for promotion of sensory recovery in anterolateral thigh free flaps. Stem Cells Transl Med 2020; 10:181-189. [PMID: 33043628 PMCID: PMC7848322 DOI: 10.1002/sctm.20-0226] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 08/18/2020] [Accepted: 09/12/2020] [Indexed: 12/12/2022] Open
Abstract
Anterolateral thigh (ALT) free flaps are widely used for reconstruction, but poor sensory recovery of the flap tissue can cause unsatisfactory outcomes and poor function. Adipose‐derived mesenchymal stem cells (ADSCs) promote neural regeneration, but the clinical use of stem‐cell therapy has been limited by lack of regulatory approval. Nanofat is an autologous product that is prepared mechanically from harvested fat. It is enriched in ADSCs and does not contain any exogenous substances. The developmental and adult neurobiology of nerve‐growth factor (NGF) are well investigated, and mouse (m)NGF has been used to promote recovery following peripheral nerve injury. We investigated the promotion of nanofat and mNGF as either mono‐ or combined therapy on the sensory recovery of ALT free flaps. We found that nanofat and mNGF had a synergistic effect on sensory recovery that was associated with stimulation of angiogenesis and neurogenesis. Nanofat combined with mNGF was better at promoting neural regeneration and improving sensory recovery than treatment with either agent alone. The results provide a theoretical rationale for further study of the clinical use of nanofat combined with mNGF to promote the sensory recovery of ALT free flaps.
Collapse
Affiliation(s)
- Shune Xiao
- Department of Plastic SurgeryAffiliated Hospital of Zunyi Medical UniversityZunyiPeople's Republic of China
| | - Fengling Zhang
- Department of Plastic SurgeryAffiliated Hospital of Zunyi Medical UniversityZunyiPeople's Republic of China
| | - Yongjian Zheng
- Department of Plastic SurgeryAffiliated Hospital of Zunyi Medical UniversityZunyiPeople's Republic of China
| | - Zhiyuan Liu
- Department of Plastic SurgeryAffiliated Hospital of Zunyi Medical UniversityZunyiPeople's Republic of China
| | - Dali Wang
- Department of Plastic SurgeryAffiliated Hospital of Zunyi Medical UniversityZunyiPeople's Republic of China
| | - Zairong Wei
- Department of Plastic SurgeryAffiliated Hospital of Zunyi Medical UniversityZunyiPeople's Republic of China
| | - Chengliang Deng
- Department of Plastic SurgeryAffiliated Hospital of Zunyi Medical UniversityZunyiPeople's Republic of China
| |
Collapse
|
131
|
Liu X, Yang X, Zhan C, Zhang Y, Hou J, Yin X. Perineural Invasion in Adenoid Cystic Carcinoma of the Salivary Glands: Where We Are and Where We Need to Go. Front Oncol 2020; 10:1493. [PMID: 33014792 PMCID: PMC7461905 DOI: 10.3389/fonc.2020.01493] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 07/13/2020] [Indexed: 12/25/2022] Open
Abstract
Adenoid cystic carcinoma of the salivary gland (SACC) is a rare malignant tumors of the head and neck region, but it is one of the most common malignant tumors that are prone to perineural invasion (PNI) of the head and neck. The prognosis of patients with SACC is strongly associated with the presence of perineural spread (PNS). Although many contributing factors have been reported, the mechanisms underlying the preferential destruction of the blood-nerve barrier (BNB) by tumors and the infiltration of the tumor microenvironment by nerve fibers in SACC, have received little research attention. This review summarizes the current knowledge concerning the characteristics of SACC in relation to the PNI, and then highlights the interplay between components of the tumor microenvironment and perineural niche, as well as their contributions to the PNI. Finally, we provide new insights into the possible mechanisms underlying the pathogenesis of PNI, with particular emphasis on the role of extracellular vesicles that may serve as an attractive entry point in future studies.
Collapse
Affiliation(s)
- Xiaohao Liu
- Department of Oral and Maxillofacial Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaojun Yang
- Department of Oral and Maxillofacial Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Chaoning Zhan
- Department of Oral and Maxillofacial Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yan Zhang
- Department of Oral and Maxillofacial Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jin Hou
- Department of Oral and Maxillofacial Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xuemin Yin
- Department of Oral and Maxillofacial Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
132
|
Chernov AN, Alaverdian DA, Glotov OS, Talabaev MV, Urazov SP, Shcherbak SG, Renieri A, Frullanti E, Shamova O. Related expression of TRKA and P75 receptors and the changing copy number of MYC-oncogenes determine the sensitivity of brain tumor cells to the treatment of the nerve growth factor in combination with cisplatin and temozolomide. Drug Metab Pers Ther 2020; 0:/j/dmdi.ahead-of-print/dmdi-2020-0109/dmdi-2020-0109.xml. [PMID: 32887179 DOI: 10.1515/dmdi-2020-0109] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 07/29/2020] [Indexed: 02/07/2023]
Abstract
Objectives Oncological diseases are an urgent medical and social problem. The chemotherapy induces not only the death of the tumor cells but also contributes to the development of their multidrug resistance and death of the healthy cells and tissues. In this regard, the search for the new pharmacological substances with anticancer activity against drug-resistant tumors is of utmost importance. In the present study we primarily investigated the correlation between the expression of TrkA and p75 receptors with the nerve growth factor (NGF) and cisplatin or temozolomide sensitivity of anaplastic astrocytoma (AA), glioblastoma (GB) and medulloblastoma (MB) cell cultures. We then evaluated the changing of copy numbers of MYCC and MYCN and its correlation with cytotoxicity index (CI) in MB cells under NGF exposition. Methods The primary cell cultures were obtained from the tumor biopsy samples of the patients with AA (n=5), GB (n=7) or MB (n=25) prior to radiotherapy and chemotherapy. The cytotoxicity effect of NGF and its combinations with cisplatin or temozolomide, the relative expression of TrkA and p75 receptors, its correlations with CI in AA, GB and MB primary cell cultures were studied by trypan blue cytotoxicity assay and immunofluorescence staining respectively. The effect of NGF on MYCC and MYCN copy numbers in MB cell cultures was studied by fluorescence in situ hybridization. Results We found that the expression of TrkA and p75 receptors (p=0.03) and its ratio (p=0.0004) depends on the sensitivity of AA and GB cells to treatment with NGF and its combinations with cisplatin or temozolomide. NGF reduces (p<0.05) the quantity of MB cells with six or eight copies of MYCN and three or eight copies of MYCC. Besides, NGF increases (p<0.05) the quantity of MB cells containing two copies of both oncogenes. The negative correlation (r=-0.65, p<0.0001) is established between MYCC average copy numbers and CI of NGF in MB cells. Conclusions The relative expression of NGF receptors (TrkA/p75) and its correlation with CI of NGF and its combinations in AA and GB cells point to the mechanism involving a cell death signaling pathway. NGF downregulates (p<0.05) some increased copy numbers of MYCC and MYCN in the human MB cell cultures, and upregulates normal two copies of both oncogenes (p<0.05).
Collapse
Affiliation(s)
- Alexandr N Chernov
- Department of Clinics and Genetics Investigations, Saint Petersburg City Hospital No40 of Resort District, Saint Petersburg, Russian Federation
- Laboratory of Design and Synthesis of Biologically Active Peptides, Department of General Pathology and Pathophysiology, Institute of Experimental Medicine, Saint-Petersburg, Russian Federation
| | - Diana A Alaverdian
- Medical Genetics, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Oleg S Glotov
- Department of Clinics and Genetics Investigations, Saint Petersburg City Hospital No40 of Resort District, Saint Petersburg, Russian Federation
| | - Michael V Talabaev
- Department of Pediatric Neurosurgery, Republican Center for Neurology and Neurosurgery, Minsk, The Republic of Belarus
| | - Stanislav P Urazov
- Department of Clinics and Genetics Investigations, Saint Petersburg City Hospital No40 of Resort District, Saint Petersburg, Russian Federation
| | - Sergei G Shcherbak
- Department of Clinics and Genetics Investigations, Saint Petersburg City Hospital No40 of Resort District, Saint Petersburg, Russian Federation
| | - Alessandra Renieri
- Medical Genetics, Department of Medical Biotechnologies, University of Siena, Siena, Italy
- Genetica Medica, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | - Elisa Frullanti
- Medical Genetics, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Olga Shamova
- Laboratory of Design and Synthesis of Biologically Active Peptides, Department of General Pathology and Pathophysiology, Institute of Experimental Medicine, Saint-Petersburg, Russian Federation
| |
Collapse
|
133
|
SEYİTHANOĞLU M, İNANÇ Y, BAVLİ S. Üriner Sistem Semptomları Olan Multipl Sklerozlu Hastalarda Üriner Sinir Büyüme Faktörü Düzeyleri. KAHRAMANMARAŞ SÜTÇÜ İMAM ÜNIVERSITESI TIP FAKÜLTESI DERGISI 2020. [DOI: 10.17517/ksutfd.793595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
|
134
|
Chernov AN, Alaverdian DA, Glotov OS, Talabaev MV, Urazov SP, Shcherbak SG, Renieri A, Frullanti E, Shamova O. Related expression of TRKA and P75 receptors and the changing copy number of MYC-oncogenes determine the sensitivity of brain tumor cells to the treatment of the nerve growth factor in combination with cisplatin and temozolomide. Drug Metab Pers Ther 2020; 35:dmpt-2020-0109. [PMID: 34704697 DOI: 10.1515/dmpt-2020-0109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 07/29/2020] [Indexed: 12/24/2022]
Abstract
OBJECTIVES Oncological diseases are an urgent medical and social problem. The chemotherapy induces not only the death of the tumor cells but also contributes to the development of their multidrug resistance and death of the healthy cells and tissues. In this regard, the search for the new pharmacological substances with anticancer activity against drug-resistant tumors is of utmost importance. In the present study we primarily investigated the correlation between the expression of TrkA and p75 receptors with the nerve growth factor (NGF) and cisplatin or temozolomide sensitivity of anaplastic astrocytoma (AA), glioblastoma (GB) and medulloblastoma (MB) cell cultures. We then evaluated the changing of copy numbers of MYCC and MYCN and its correlation with cytotoxicity index (CI) in MB cells under NGF exposition. METHODS The primary cell cultures were obtained from the tumor biopsy samples of the patients with AA (n=5), GB (n=7) or MB (n=25) prior to radiotherapy and chemotherapy. The cytotoxicity effect of NGF and its combinations with cisplatin or temozolomide, the relative expression of TrkA and p75 receptors, its correlations with CI in AA, GB and MB primary cell cultures were studied by trypan blue cytotoxicity assay and immunofluorescence staining respectively. The effect of NGF on MYCC and MYCN copy numbers in MB cell cultures was studied by fluorescence in situ hybridization. RESULTS We found that the expression of TrkA and p75 receptors (p=0.03) and its ratio (p=0.0004) depends on the sensitivity of AA and GB cells to treatment with NGF and its combinations with cisplatin or temozolomide. NGF reduces (p<0.05) the quantity of MB cells with six or eight copies of MYCN and three or eight copies of MYCC. Besides, NGF increases (p<0.05) the quantity of MB cells containing two copies of both oncogenes. The negative correlation (r=-0.65, p<0.0001) is established between MYCC average copy numbers and CI of NGF in MB cells. CONCLUSIONS The relative expression of NGF receptors (TrkA/p75) and its correlation with CI of NGF and its combinations in AA and GB cells point to the mechanism involving a cell death signaling pathway. NGF downregulates (p<0.05) some increased copy numbers of MYCC and MYCN in the human MB cell cultures, and upregulates normal two copies of both oncogenes (p<0.05).
Collapse
Affiliation(s)
- Alexandr N Chernov
- Department of Clinics and Genetics Investigations, Saint Petersburg City Hospital No40 of Resort District, Saint Petersburg, Russian Federation.,Laboratory of Design and Synthesis of Biologically Active Peptides, Department of General Pathology and Pathophysiology, Institute of Experimental Medicine, Saint-Petersburg, Russian Federation
| | - Diana A Alaverdian
- Medical Genetics, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Oleg S Glotov
- Department of Clinics and Genetics Investigations, Saint Petersburg City Hospital No40 of Resort District, Saint Petersburg, Russian Federation
| | - Michael V Talabaev
- Department of Pediatric Neurosurgery, Republican Center for Neurology and Neurosurgery, Minsk, The Republic of Belarus
| | - Stanislav P Urazov
- Department of Clinics and Genetics Investigations, Saint Petersburg City Hospital No40 of Resort District, Saint Petersburg, Russian Federation
| | - Sergei G Shcherbak
- Department of Clinics and Genetics Investigations, Saint Petersburg City Hospital No40 of Resort District, Saint Petersburg, Russian Federation
| | - Alessandra Renieri
- Medical Genetics, Department of Medical Biotechnologies, University of Siena, Siena, Italy.,Genetica Medica, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | - Elisa Frullanti
- Medical Genetics, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Olga Shamova
- Laboratory of Design and Synthesis of Biologically Active Peptides, Department of General Pathology and Pathophysiology, Institute of Experimental Medicine, Saint-Petersburg, Russian Federation
| |
Collapse
|
135
|
Lahiani A, Haham-Geula D, Lankri D, Cornell-Kennon S, Schaefer EM, Tsvelikhovsky D, Lazarovici P. Neurotropic activity and safety of methylene-cycloalkylacetate (MCA) derivative 3-(3-allyl-2-methylenecyclohexyl) propanoic acid. ACS Chem Neurosci 2020; 11:2577-2589. [PMID: 32667774 PMCID: PMC7497641 DOI: 10.1021/acschemneuro.0c00255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 07/15/2020] [Indexed: 11/30/2022] Open
Abstract
Polyneuropathy is a disease involving multiple peripheral nerves injuries. Axon regrowth remains the major prerequisite for plasticity, regeneration, circuit formation, and eventually functional recovery and therefore, regulation of neurite outgrowth might be a candidate for treating polyneuropathies. In a recent study, we synthesized and established the methylene-cycloalkylacetate (MCAs) pharmacophore as a lead for the development of a neurotropic drug (inducing neurite/axonal outgrowth) using the PC12 neuronal model. In the present study we extended the characterizations of the in vitro neurotropic effect of the derivative 3-(3-allyl-2-methylenecyclohexyl) propanoic acid (MCA-13) on dorsal root ganglia and spinal cord neuronal cultures and analyzed its safety properties using blood biochemistry and cell counting, acute toxicity evaluation in mice and different in vitro "off-target" pharmacological evaluations. This MCA derivative deserves further preclinical mechanistic pharmacological characterizations including therapeutic efficacy in in vivo animal models of polyneuropathies, toward development of a clinically relevant neurotropic drug.
Collapse
Affiliation(s)
- Adi Lahiani
- The
Institute for Drug Research, Division of Pharmacology, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel
| | - Dikla Haham-Geula
- The
Institute for Drug Research, Division of Pharmacology, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel
| | - David Lankri
- The
Institute for Drug Research, Division of Medicinal Chemistry, School
of Pharmacy, Faculty of Medicine, The Hebrew
University of Jerusalem, Jerusalem 9112102, Israel
| | - Susan Cornell-Kennon
- AssayQuant
Technologies Inc. 260
Cedar Hill Street, Marlboro, Massachusetts 01752, United States
| | - Erik M. Schaefer
- AssayQuant
Technologies Inc. 260
Cedar Hill Street, Marlboro, Massachusetts 01752, United States
| | - Dmitry Tsvelikhovsky
- The
Institute for Drug Research, Division of Medicinal Chemistry, School
of Pharmacy, Faculty of Medicine, The Hebrew
University of Jerusalem, Jerusalem 9112102, Israel
| | - Philip Lazarovici
- The
Institute for Drug Research, Division of Pharmacology, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel
| |
Collapse
|
136
|
Chou LY, Chao YM, Peng YC, Lin HC, Wu YL. Glucosamine Enhancement of BDNF Expression and Animal Cognitive Function. Molecules 2020; 25:molecules25163667. [PMID: 32806562 PMCID: PMC7465318 DOI: 10.3390/molecules25163667] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 08/10/2020] [Accepted: 08/11/2020] [Indexed: 12/19/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF) is an important factor for memory consolidation and cognitive function. Protein kinase A (PKA) signaling interacts significantly with BDNF-provoked downstream signaling. Glucosamine (GLN), a common dietary supplement, has been demonstrated to perform a variety of beneficial physiological functions. In the current study, an in vivo model of 7-week-old C57BL/6 mice receiving daily intraperitoneal injection of GLN (0, 3, 10 and 30 mg/animal) was subjected to the novel object recognition test in order to determine cognitive performance. GLN significantly increased cognitive function. In the hippocampus GLN elevated tissue cAMP concentrations and CREB phosphorylation, and upregulated the expression of BDNF, CREB5 and the BDNF receptor TrkB, but it reduced PDE4B expression. With the in vitro model in the HT22 hippocampal cell line, GLN exposure significantly increased protein and mRNA levels of BDNF and CREB5 and induced cAMP responsive element (CRE) reporter activity; the GLN-mediated BDNF expression and CRE reporter induction were suppressed by PKA inhibitor H89. Our current findings suggest that GLN can exert a cognition-enhancing function and this may act at least in part by upregulating the BDNF levels via a cAMP/PKA/CREB-dependent pathway.
Collapse
Affiliation(s)
- Lien-Yu Chou
- Department of Physiology, School of Medicine, National Yang-Ming University, Taipei 11221, Taiwan; (L.-Y.C.), (Y.-M.C.); (H.-C.L.)
| | - Yu-Ming Chao
- Department of Physiology, School of Medicine, National Yang-Ming University, Taipei 11221, Taiwan; (L.-Y.C.), (Y.-M.C.); (H.-C.L.)
| | - Yen-Chun Peng
- Department of Internal Medicine, Taichung Veterans General Hospital, Taichung 40705, Taiwan;
| | - Hui-Ching Lin
- Department of Physiology, School of Medicine, National Yang-Ming University, Taipei 11221, Taiwan; (L.-Y.C.), (Y.-M.C.); (H.-C.L.)
| | - Yuh-Lin Wu
- Department of Physiology, School of Medicine, National Yang-Ming University, Taipei 11221, Taiwan; (L.-Y.C.), (Y.-M.C.); (H.-C.L.)
- Correspondence: ; Tel.: +886-2-2826-7081; Fax: +886-2-2826-4049
| |
Collapse
|
137
|
Yang J, Wu S, Hou L, Zhu D, Yin S, Yang G, Wang Y. Therapeutic Effects of Simultaneous Delivery of Nerve Growth Factor mRNA and Protein via Exosomes on Cerebral Ischemia. MOLECULAR THERAPY. NUCLEIC ACIDS 2020; 21:512-522. [PMID: 32682291 PMCID: PMC7365960 DOI: 10.1016/j.omtn.2020.06.013] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 05/26/2020] [Accepted: 06/15/2020] [Indexed: 12/20/2022]
Abstract
Stroke is the leading neurological cause of death and disability all over the world, with few effective drugs. Nerve growth factor (NGF) is well known for its multifaceted neuroprotective functions post-ischemia. However, the lack of an efficient approach to systemically deliver bioactive NGF into ischemic region hinders its clinical application. In this study, we engineered the exosomes with RVG peptide on the surface for neuron targeting and loaded NGF into exosomes simultaneously, with the resultant exosomes denoted as NGF@ExoRVG. By systemic administration of NGF@ExoRVG, NGF was efficiently delivered into ischemic cortex, with a burst release of encapsulated NGF protein and de novo NGF protein translated from the delivered mRNA. Moreover, NGF@ExoRVG was found to be highly stable for preservation and function efficiently for a long time in vivo. Functional study revealed that the delivered NGF reduced inflammation by reshaping microglia polarization, promoted cell survival, and increased the population of doublecortin-positive cells, a marker of neuroblast. The results of our study suggest the potential therapeutic effects of NGF@ExoRVG for stroke. Moreover, the strategy proposed in our study may shed light on the clinical application of other neurotrophic factors for central nervous system diseases.
Collapse
Affiliation(s)
- Jialei Yang
- China National Clinical Research Center for Neurological Diseases, Beijing 100070, China; Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China; Department of Neurology, PLA Rocket Force Characteristic Medical Center, Beijing, China; Beijing Institute of Biotechnology, Beijing, China
| | - Shipo Wu
- Beijing Institute of Biotechnology, Beijing, China
| | - Lihua Hou
- Beijing Institute of Biotechnology, Beijing, China
| | - Danni Zhu
- Beijing Institute of Biotechnology, Beijing, China
| | - Shimin Yin
- Department of Neurology, PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Guodong Yang
- Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, Shaanxi 710032, China.
| | - Yongjun Wang
- China National Clinical Research Center for Neurological Diseases, Beijing 100070, China; Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China.
| |
Collapse
|
138
|
Future avenues for Alzheimer's disease detection and therapy: liquid biopsy, intracellular signaling modulation, systems pharmacology drug discovery. Neuropharmacology 2020; 185:108081. [PMID: 32407924 DOI: 10.1016/j.neuropharm.2020.108081] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 03/01/2020] [Accepted: 03/30/2020] [Indexed: 12/20/2022]
Abstract
When Alzheimer's disease (AD) disease-modifying therapies will be available, global healthcare systems will be challenged by a large-scale demand for clinical and biological screening. Validation and qualification of globally accessible, minimally-invasive, and time-, cost-saving blood-based biomarkers need to be advanced. Novel pathophysiological mechanisms (and related candidate biomarkers) - including neuroinflammation pathways (TREM2 and YKL-40), axonal degeneration (neurofilament light chain protein), synaptic dysfunction (neurogranin, synaptotagmin, α-synuclein, and SNAP-25) - may be integrated into an expanding pathophysiological and biomarker matrix and, ultimately, integrated into a comprehensive blood-based liquid biopsy, aligned with the evolving ATN + classification system and the precision medicine paradigm. Liquid biopsy-based diagnostic and therapeutic algorithms are increasingly employed in Oncology disease-modifying therapies and medical practice, showing an enormous potential for AD and other brain diseases as well. For AD and other neurodegenerative diseases, newly identified aberrant molecular pathways have been identified as suitable therapeutic targets and are currently investigated by academia/industry-led R&D programs, including the nerve-growth factor pathway in basal forebrain cholinergic neurons, the sigma1 receptor, and the GTPases of the Rho family. Evidence for a clinical long-term effect on cognitive function and brain health span of cholinergic compounds, drug candidates for repositioning programs, and non-pharmacological multidomain interventions (nutrition, cognitive training, and physical activity) is developing as well. Ultimately, novel pharmacological paradigms, such as quantitative systems pharmacology-based integrative/explorative approaches, are gaining momentum to optimize drug discovery and accomplish effective pathway-based strategies for precision medicine. This article is part of the special issue on 'The Quest for Disease-Modifying Therapies for Neurodegenerative Disorders'.
Collapse
|
139
|
Anti-Aging Effects of GDF11 on Skin. Int J Mol Sci 2020; 21:ijms21072598. [PMID: 32283613 PMCID: PMC7177281 DOI: 10.3390/ijms21072598] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 04/02/2020] [Accepted: 04/07/2020] [Indexed: 12/24/2022] Open
Abstract
Human skin is composed of three layers: the epidermis, the dermis, and the hypodermis. The epidermis has four major cell layers made up of keratinocytes in varying stages of progressive differentiation. Skin aging is a multi-factorial process that affects every phase of its biology and function. The expression profiles of inflammation-related genes analyzed in resident immune cells demonstrated that these cells have a strong ability to regenerate adult skin stem cells and to produce endogenous substances such as growth differentiation factor 11 (GDF11). GDF11 appears to be the key to progenitor proliferation and/or differentiation. The preservation of youthful phenotypes has been tied to the presence of GDF11 in different human tissues, and, in the skin, this factor inhibits inflammatory responses. The protective role of GDF11 depends on a multi-factorial process implicating various types of skin cells such as keratinocytes, fibroblasts and inflammatory cells. GDF11 should be further studied for the purpose of developing novel therapies for the treatment of skin diseases.
Collapse
|
140
|
Liu X, Lovell P, Cruz DR, Houser L, LaGrange CA, Mohs AM. Novel Intra-Operative Peripheral Nerve Agent for Fluorescence Guided Imaging. PROCEEDINGS OF SPIE--THE INTERNATIONAL SOCIETY FOR OPTICAL ENGINEERING 2020; 11225:112250J. [PMID: 40007602 PMCID: PMC11853179 DOI: 10.1117/12.2545107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2025]
Abstract
Nerve injuries that significantly affect a patient's quality of life are a common complication of major surgeries. Fluorescence-guided surgery (FGS) has become increasingly popular because it allows physicians to position their instruments precisely during surgeries to spare nerves, for example, in radical prostatectomies. Visualization of nerves during oncological surgeries is an unmet clinical need that is under investigation. Here, we address this unmet need with a contrast agent that is selective for peripheral nerves. Our contrast agent combines an existing near infrared (NIR) dye that fluoresces in the 800 region with a naturally-occurring protein of the human nervous system, nerve growth factor (NGF) - a combination termed Nervelight™. Due to the fact that exogenously administered NGF localizes to the distal ends of nerves due to guidance by high affinity receptors, our contrast agent binds specifically to, then is endocytosed, and is transported up the nerve via retrograde axonal transport. In the clinical setting during nerve sparing surgeries, the area in question would be incised, and the surgeon could intra-operatively apply the agent to at-risk nerves before removing the tumor. In preliminary studies, after we directly applied the contrast agent to the nerve of interest, the targeted nerve was clearly labeled by this fluorescent imaging agent. In these experiments, visualization was obtained after 10 minutes. Other studies suggest that nerves may be seen for the duration of at least one hour and likely longer. These results suggest that Nervelight™ can serve as a fluorescence-guided surgical tool that will improve the visualization of at-risk nerves during radical prostatectomies, and possibly other oncological surgeries.
Collapse
Affiliation(s)
- Xiang Liu
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Paul Lovell
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Denzel Ryan Cruz
- University of Illinois at Urbana- Champaign, Champaign, IL 61820, USA
| | - Lucas Houser
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Chad A LaGrange
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Department of Urology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Aaron M Mohs
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
141
|
Liu H, Tan N, Xu D, Li CY, Xian GJ. NGF and CNTF expression and regulation mechanism by miRNA in acute paralytic strabismus. Int Ophthalmol 2020; 40:975-984. [PMID: 31925656 DOI: 10.1007/s10792-019-01270-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 12/30/2019] [Indexed: 11/24/2022]
Abstract
BACKGROUND Nerve growth factor (NGF) and ciliary neurotrophic factor (CNTF) are well-known neurotrophic factors and widely used in the clinical treatment for its promotion effect on peripheral nerve regeneration. And they were also recommended for the acute paralytic strabismus treatment. However, whether the NGF and CNTF have protective effect for the extraocular muscles of acute paralytic strabismus patients is still poorly understood. PURPOSE In this study, we want to evaluate the biological function of NGF and CNTF on the extraocular muscle cells and reveale the regulation mechanism behind it. METHODS Firstly, the relative expression of ngf and cntf was assessed by quantitative real-time RT-PCR. Then, the influence of NGF and CNTF on the extraocular muscle cell proliferation was determined by CCK-8. The inflammatory response in muscle cells after NGF and CNTF treatment was evaluated by ELISA and ROS detection. In addition to this, the up-stream regulation of the ngf and cntf expression was also studied. The TargetScan was used for the predication of potential miRNAs targeting with ngf and cntf 30-UTR, which is soon confirmed by luciferase activity assay. RESULTS all the results in this research indicated that NGF and CNTF could promote the muscle cell proliferation and inhibit the inflammatory levels, then exert protective effect on the muscle cell function. RESULTS All the results in this research indicated that NGF and CNTF could promote the muscle cell proliferation and inhibit the inflammatory levels, then exert protective effect on the muscle cell function. CONCLUSION It was conceivable that let 7-5p was the up-stream regulator of ngf and cntf, and let 7-5p might serve as a potential molecular target for acute paralytic strabismus treatment.
Collapse
Affiliation(s)
- Hua Liu
- Department of Ophthalmology, Daping Hospital, Army Medical Center of PLA, Chongqing, China
| | - Nian Tan
- Department of Ophthalmology, Daping Hospital, Army Medical Center of PLA, Chongqing, China
| | - Duo Xu
- Department of Ophthalmology, Daping Hospital, Army Medical Center of PLA, Chongqing, China.
| | - Chong-Yi Li
- Department of Ophthalmology, Daping Hospital, Army Medical Center of PLA, Chongqing, China
| | - Guang-Jun Xian
- Department of Ophthalmology, Daping Hospital, Army Medical Center of PLA, Chongqing, China
| |
Collapse
|
142
|
Hah YS, Yoo WS, Seo SW, Chung I, Kim HA, Cho HY, Kim SJ. Reduced NGF Level Promotes Epithelial-Mesenchymal Transition in Human Lens Epithelial Cells Exposed to High Dexamethasone Concentrations. Curr Eye Res 2019; 45:686-695. [PMID: 31751158 DOI: 10.1080/02713683.2019.1695844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Purpose: To investigate the protective effects of nerve growth factor (NGF) against steroid-induced cataract formation in dexamethasone (Dex)-treated human lens epithelial B-3 (HLE-B3) cells and the possible molecular mechanisms underlying this protection.Materials and Methods: HLE-B3 cells were treated with Dex, and cell viability was assessed using the Cell Counting Kit-8 (CCK-8) assay. The levels of expression of NGF, fibronectin, α-smooth muscle actin (α-SMA), and E-cadherin mRNAs were measured by real-time quantitative polymerase chain reaction (qPCR), and the levels of NGF, fibronectin, α-SMA, E-cadherin, tropomyosin receptor kinase A (TrkA), and Akt proteins were measured by Western blot analysis. Gene expression profiles of growth factors in Dex-treated HLE-B3 cells were determined by PCR arrays. In addition, anterior capsule tissue was obtained during cataract surgery, and the specimens were also examined expressions of NGF.Results: NGF was expressed in HLE-B3 cells and also in lens epithelial cells of anterior lens capsules. Dex treatment of HLE-B3 cells increased their expression of epithelial-mesenchymal transition (EMT) markers and migration activity, while markedly downregulating the expression of NGF. NGF treatment significantly reduced the expression of α-SMA and fibronectin, as well as cell proliferation. The decreased phosphorylation of p38 MAPK and Akt induced by Dex treatment was significantly reversed by treatment with NGF.Conclusion: NGF/TrkA may repress EMT by targeting the p38 MAPK and pAkt pathways in Dex-treated HLE-B3 cells. NGF may be a novel therapeutic target for patients with steroid-induced cataract.
Collapse
Affiliation(s)
- Young-Sool Hah
- Biomedical Research Institute, Gyeongsang National University Hospital, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju, Korea
| | - Woong-Sun Yoo
- Department of Ophthalmology, Institute of Health Sciences, Gyeongsang National University School of Medicine, Gyeongsang National University Hospital, Jinju, Korea
| | - Seong-Wook Seo
- Department of Ophthalmology, Institute of Health Sciences, Gyeongsang National University School of Medicine, Gyeongsang National University Hospital, Jinju, Korea
| | - Inyoung Chung
- Department of Ophthalmology, Institute of Health Sciences, Gyeongsang National University School of Medicine, Gyeongsang National University Hospital, Jinju, Korea
| | - Hyun-A Kim
- Department of Ophthalmology, Institute of Health Sciences, Gyeongsang National University School of Medicine, Gyeongsang National University Hospital, Jinju, Korea
| | - Hee Young Cho
- Biomedical Research Institute, Gyeongsang National University Hospital, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju, Korea
| | - Seong-Jae Kim
- Department of Ophthalmology, Institute of Health Sciences, Gyeongsang National University School of Medicine, Gyeongsang National University Hospital, Jinju, Korea
| |
Collapse
|
143
|
Magnetic Resonance Imaging as a Biomarker in Rodent Peripheral Nerve Injury Models Reveals an Age-Related Impairment of Nerve Regeneration. Sci Rep 2019; 9:13508. [PMID: 31534149 PMCID: PMC6751200 DOI: 10.1038/s41598-019-49850-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 08/24/2019] [Indexed: 12/12/2022] Open
Abstract
Assessment of myelin integrity in peripheral nerve injuries and pathologies has largely been limited to post-mortem analysis owing to the difficulty in obtaining biopsies without affecting nerve function. This is further encumbered by the small size of the tissue and its location. Therefore, the development of robust, non-invasive methods is highly attractive. In this study, we used magnetic resonance imaging (MRI) techniques, including magnetization transfer ratio (MTR), to longitudinally and non-invasively characterize both the sciatic nerve crush and lysolecithin (LCP) demyelination models of peripheral nerve injury in rodents. Electrophysiological, gene expression and histological assessments complemented the extensive MRI analyses in young and aged animals. In the nerve crush model, MTR analysis indicated a slower recovery in regions distal to the site of injury in aged animals, as well as incomplete recovery at six weeks post-crush when analyzing across the entire nerve surface. Similar regional impairments were also found in the LCP demyelination model. This research underlines the power of MTR for the study of peripheral nerve injury in small tissues such as the sciatic nerve of rodents and contributes new knowledge to the effect of aging on recovery after injury. A particular advantage of the approach is the translational potential to human neuropathies.
Collapse
|
144
|
Houlton J, Abumaria N, Hinkley SFR, Clarkson AN. Therapeutic Potential of Neurotrophins for Repair After Brain Injury: A Helping Hand From Biomaterials. Front Neurosci 2019; 13:790. [PMID: 31427916 PMCID: PMC6688532 DOI: 10.3389/fnins.2019.00790] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 07/15/2019] [Indexed: 12/17/2022] Open
Abstract
Stroke remains the leading cause of long-term disability with limited options available to aid in recovery. Significant effort has been made to try and minimize neuronal damage following stroke with use of neuroprotective agents, however, these treatments have yet to show clinical efficacy. Regenerative interventions have since become of huge interest as they provide the potential to restore damaged neural tissue without being limited by a narrow therapeutic window. Neurotrophins, such as brain-derived neurotrophic factor (BDNF), and their high affinity receptors are actively produced throughout the brain and are involved in regulating neuronal activity and normal day-to-day function. Furthermore, neurotrophins are known to play a significant role in both protection and recovery of function following neurodegenerative diseases such as stroke and traumatic brain injury (TBI). Unfortunately, exogenous administration of these neurotrophins is limited by a lack of blood-brain-barrier (BBB) permeability, poor half-life, and rapid degradation. Therefore, we have focused this review on approaches that provide a direct and sustained neurotrophic support using pharmacological therapies and mimetics, physical activity, and potential drug delivery systems, including discussion around advantages and limitations for use of each of these systems. Finally, we discuss future directions of biomaterial drug-delivery systems, including the incorporation of heparan sulfate (HS) in conjunction with neurotrophin-based interventions.
Collapse
Affiliation(s)
- Josh Houlton
- Brain Health Research Centre, Department of Anatomy, University of Otago, Dunedin, New Zealand
| | - Nashat Abumaria
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institute of Brain Science, Fudan University, Shanghai, China
- Department of Laboratory Animal Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Simon F. R. Hinkley
- The Ferrier Research Institute, Victoria University of Wellington, Petone, New Zealand
| | - Andrew N. Clarkson
- Brain Health Research Centre, Department of Anatomy, University of Otago, Dunedin, New Zealand
| |
Collapse
|
145
|
Matteucci A, Ricceri L, Fabbri A, Fortuna A, Travaglione S, Guidotti M, Martinelli A, Villa M, Pricci F, Maroccia Z, Campana G, Malchiodi-Albedi F, Fiorentini C, Loizzo S. Eye Drop Instillation of the Rac1 Modulator CNF1 Attenuates Retinal Gliosis and Ameliorates Visual Performance in a Rat Model of Hypertensive Retinopathy. Neuroscience 2019; 411:119-129. [PMID: 31128161 DOI: 10.1016/j.neuroscience.2019.05.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 05/08/2019] [Accepted: 05/09/2019] [Indexed: 12/15/2022]
Abstract
In hypertensive retinopathy, the retinal damage due to high blood pressure is accompanied by increased expression of Glial Fibrillary Acidic Protein (GFAP), which indicates a role of neuroinflammatory processes in such a retinopathy. Proteins belonging to the Rho GTPase family, particularly Rac1, are involved in the activation of Müller glia and in the progression of photoreceptor degeneration, and may thus represent a novel candidate for therapeutic intervention following central nervous system inflammation. In this paper, we have observed that topical administration as eye drops of Cytotoxic Necrotizing Factor 1 (CNF1), a Rho GTPase modulator, surprisingly improves electrophysiological and behavioral visual performances in aged spontaneously hypertensive rats. Furthermore, such functional improvement is accompanied by a reduction of Rac1 activity and retinal GFAP expression. Our results suggest that Rac1 inhibition through CNF1 topical administration may represent a new strategy to target retinal gliosis.
Collapse
Affiliation(s)
- Andrea Matteucci
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Laura Ricceri
- Centre for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Alessia Fabbri
- Italian Center for Global Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Andrea Fortuna
- Italian Center for Global Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Sara Travaglione
- Italian Center for Global Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Marco Guidotti
- Department of Veterinary Public Health and Food Safety, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Andrea Martinelli
- National Centre for Animal Research and Welfare, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Marika Villa
- Department of Cardiovascular, Dysmetabolic and Endocrine-Metabolic Diseases, and Ageing, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Flavia Pricci
- Department of Cardiovascular, Dysmetabolic and Endocrine-Metabolic Diseases, and Ageing, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Zaira Maroccia
- Italian Center for Global Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Gabriele Campana
- Department of Pharmacy and Biotechnology, University of Bologna, Via Zamboni, 33, Bologna, Italy
| | - Fiorella Malchiodi-Albedi
- Centre for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Carla Fiorentini
- Italian Center for Global Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy; Association for Research on Integrative Oncology Therapies (ARTOI), Rome, Italy
| | - Stefano Loizzo
- Italian Center for Global Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy.
| |
Collapse
|