1
|
Xu X, Liu Y, Lan M, Liu F, Xia H, Zeng J. Suppression of SRC protein kinase activity alleviates the severity of aganglionosis by impairing CAV1/FLNA expression. Sci Prog 2025; 108:368504251336287. [PMID: 40296549 PMCID: PMC12041699 DOI: 10.1177/00368504251336287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
ObjectiveHirschsprung's disease (HSCR) is a rare congenital disorder attributed to the defects of enteric neural crest cells. We aim to identify characteristic phosphorylation proteins and preliminarily explore underlying related action mechanisms in HSCR.MethodsColon samples from HSCR patients underwent proteomic and phosphoproteomic sequencing to identify differentially expressed phosphoproteins (DEPPs) and proteins (DEPs). Interaction network construction and analysis of correlations with upstream phosphorylating kinases were employed to pinpoint core proteins. HSCR rat models were established through enema administration of Benzalkonium chloride and evaluated by measuring colon cross-sectional area, colon weight, AchE, and PGP9.5 levels. Histopathological damage was assessed via hematoxylin and eosin staining. Protein expression was analyzed using western blotting. Furthermore, the impact of SRC kinase in HSCR was investigated utilizing an SRC-specific inhibitor in HSCR rat models.ResultsA total of 5725 DEPPs were identified, with SRC kinase emerging as a key regulatory protein. In the HSCR rat model, SRC expression was elevated along with increased pCAV1 and FLNA levels. Notably, inhibition of SRC protein kinase activity by 1-(tert-butyl)-3-(4-chlorophenyl)-1H-pyrazolo[3,4-d] pyrimidin-4-amine (PP2) led to reduced colon cross-sectional area and weight, an increase in the number of colonic ganglion cells, heightened AchE levels, enhanced PGP9.5 expression, and slight enlargement of the crypt, thereby alleviating HSCR symptoms in rats. Additionally, SRC kinase inhibition following PP2 treatment decreased the expression of pCAV1 and FLNA.ConclusionsInhibition of SRC kinase activity may potentially reduce CAV1/FLNA expression, ultimately alleviating the severity of HSCR in rats.
Collapse
Affiliation(s)
- Xiaogang Xu
- Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Yanqing Liu
- Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, China
| | - Menglong Lan
- Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, China
| | - Fei Liu
- Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, China
| | - Huimin Xia
- Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Jixiao Zeng
- Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, China
| |
Collapse
|
2
|
Xiang TY, Zhou XH, Xie XL, Liu SQ, Gui MJ, Li H, Huang DX, Liu XL, Hou ZG. Learning Motor Cues in Brain-Muscle Modulation. IEEE TRANSACTIONS ON CYBERNETICS 2025; 55:86-98. [PMID: 39423087 DOI: 10.1109/tcyb.2024.3415369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2024]
Abstract
Current studies for brain-muscle modulation often analyze selected properties in electrophysiological signals, leading to a partial understanding. This article proposes a cross-modal generative model that converts brain activities measured by electroencephalography (EEG) to corresponding muscular responses recorded by electromyography (EMG). Examining the generation process in the model highlights how the motor cue, representing implicit motor information hidden within brain activities, modulates the interaction between brain and muscle systems. The proposed model employs a two-stage generation process to bridge the semantic gap in cross-modal signals. Initially, the shared movement-related information between EEG and EMG signals is extracted using a contrastive learning framework. These shared representations act as conditional vectors in the subsequent EMG generation stage based on generative adversarial networks (GANs). Experiments on a self-collected multimodal electrophysiological signal data set show the algorithm's superiority over existing time series generative methods in cross-modal EMG generation. Further insights derived from the model's inference process underscore the brain's strategy for muscle control during movements. This research provides a data-driven approach for the neuroscience community, offering a comprehensive perspective of brain-muscular modulation.
Collapse
|
3
|
Bonsor M, Ammar O, Schnoegl S, Wanker EE, Silva Ramos E. Polyglutamine disease proteins: Commonalities and differences in interaction profiles and pathological effects. Proteomics 2024; 24:e2300114. [PMID: 38615323 DOI: 10.1002/pmic.202300114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 03/25/2024] [Accepted: 03/27/2024] [Indexed: 04/16/2024]
Abstract
Currently, nine polyglutamine (polyQ) expansion diseases are known. They include spinocerebellar ataxias (SCA1, 2, 3, 6, 7, 17), spinal and bulbar muscular atrophy (SBMA), dentatorubral-pallidoluysian atrophy (DRPLA), and Huntington's disease (HD). At the root of these neurodegenerative diseases are trinucleotide repeat mutations in coding regions of different genes, which lead to the production of proteins with elongated polyQ tracts. While the causative proteins differ in structure and molecular mass, the expanded polyQ domains drive pathogenesis in all these diseases. PolyQ tracts mediate the association of proteins leading to the formation of protein complexes involved in gene expression regulation, RNA processing, membrane trafficking, and signal transduction. In this review, we discuss commonalities and differences among the nine polyQ proteins focusing on their structure and function as well as the pathological features of the respective diseases. We present insights from AlphaFold-predicted structural models and discuss the biological roles of polyQ-containing proteins. Lastly, we explore reported protein-protein interaction networks to highlight shared protein interactions and their potential relevance in disease development.
Collapse
Affiliation(s)
- Megan Bonsor
- Department of Neuroproteomics, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Orchid Ammar
- Department of Neuroproteomics, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Sigrid Schnoegl
- Department of Neuroproteomics, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Erich E Wanker
- Department of Neuroproteomics, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Eduardo Silva Ramos
- Department of Neuroproteomics, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| |
Collapse
|
4
|
Hirunagi T, Nakatsuji H, Sahashi K, Yamamoto M, Iida M, Tohnai G, Kondo N, Yamada S, Murakami A, Noda S, Adachi H, Sobue G, Katsuno M. Exercise attenuates polyglutamine-mediated neuromuscular degeneration in a mouse model of spinal and bulbar muscular atrophy. J Cachexia Sarcopenia Muscle 2024; 15:159-172. [PMID: 37937369 PMCID: PMC10834330 DOI: 10.1002/jcsm.13344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 09/04/2023] [Accepted: 09/11/2023] [Indexed: 11/09/2023] Open
Abstract
BACKGROUND Spinal and bulbar muscular atrophy (SBMA) is a hereditary neuromuscular disorder caused by the expansion of trinucleotide cytosine-adenine-guanine (CAG) repeats, which encodes a polyglutamine (polyQ) tract in the androgen receptor (AR) gene. Recent evidence suggests that, in addition to motor neuron degeneration, defective skeletal muscles are also the primary contributors to the pathogenesis in SBMA. While benefits of physical exercise have been suggested in SBMA, underlying mechanism remains elusive. METHODS We investigated the effect of running exercise in a transgenic mouse model of SBMA carrying human AR with 97 expanded CAGs (AR97Q). We assigned AR97Q mice to exercise and sedentary control groups, and mice in the exercise group received 1-h forced running wheel (5 m/min) 5 days a week for 4 weeks during the early stage of the disease. Motor function (grip strength and rotarod performance) and survival of each group were analysed, and histopathological and biological features in skeletal muscles and motor neurons were evaluated. RESULTS AR97Q mice in the exercise group showed improvement in motor function (~40% and ~50% increase in grip strength and rotarod performance, respectively, P < 0.05) and survival (median survival 23.6 vs. 16.7 weeks, P < 0.05) with amelioration of neuronal and muscular histopathology (~1.4-fold and ~2.8-fold increase in motor neuron and muscle fibre size, respectively, P < 0.001) compared to those in the sedentary group. Nuclear accumulation of polyQ-expanded AR in skeletal muscles and motor neurons was suppressed in the mice with exercise compared to the sedentary mice (~50% and ~30% reduction in 1C2-positive cells in skeletal muscles and motor neurons, respectively, P < 0.05). We found that the exercise activated 5'-adenosine monophosphate-activated protein kinase (AMPK) signalling and inhibited mammalian target of rapamycin pathway that regulates protein synthesis in skeletal muscles of SBMA mice. Pharmacological activation of AMPK inhibited protein synthesis and reduced polyQ-expanded AR proteins in C2C12 muscle cells. CONCLUSIONS Our findings suggest the therapeutic potential of exercise-induced effect via AMPK activation in SBMA.
Collapse
Affiliation(s)
- Tomoki Hirunagi
- Department of NeurologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Hideaki Nakatsuji
- Department of NeurologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Kentaro Sahashi
- Department of NeurologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Mikiyasu Yamamoto
- Department of NeurologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Madoka Iida
- Department of NeurologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Genki Tohnai
- Department of NeurologyNagoya University Graduate School of MedicineNagoyaJapan
- Aichi Medical UniversityNagakuteJapan
| | - Naohide Kondo
- Department of NeurologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Shinichiro Yamada
- Department of NeurologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Ayuka Murakami
- Department of NeurologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Seiya Noda
- Department of NeurologyNagoya University Graduate School of MedicineNagoyaJapan
- Department of NeurologyNational Hospital Organization Suzuka HospitalSuzukaJapan
| | - Hiroaki Adachi
- Department of NeurologyUniversity of Occupational and Environmental Health School of MedicineKitakyushuJapan
| | - Gen Sobue
- Aichi Medical UniversityNagakuteJapan
| | - Masahisa Katsuno
- Department of NeurologyNagoya University Graduate School of MedicineNagoyaJapan
- Department of Clinical Research EducationNagoya University Graduate School of MedicineNagoyaJapan
| |
Collapse
|
5
|
Inhibition of Oncogenic Src Ameliorates Silica-Induced Pulmonary Fibrosis via PI3K/AKT Pathway. Int J Mol Sci 2023; 24:ijms24010774. [PMID: 36614217 PMCID: PMC9821169 DOI: 10.3390/ijms24010774] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 12/27/2022] [Accepted: 12/28/2022] [Indexed: 01/03/2023] Open
Abstract
Silicosis is a refractory disease. Previous studies indicate that damaged alveolar epithelial cells act as a driver in pulmonary fibrosis. Our results show that epithelial cells that acquire the mesenchymal phenotype are associated with the pathogenesis of silicosis. c-Src kinase, a non-receptor tyrosine kinase, has been shown to be a positive regulator of organ fibrosis, but specific mechanisms remain unclear and rarely researched in silicosis. The activated Phosphatidylinositol-3 kinases/AKT(PI3K/AKT) pathway promotes fibrosis. We aimed to determine whether c-Src regulates fibrosis via the PI3K/AKT signaling pathway in the development of silicosis. C57/BL mice were intratracheally perfused with 10 mg silica suspension to establish a model of silicosis. In vivo, silica particles induced lung fibrosis. The profibrotic cytokine transforming growth factor-β1 (TGF-β1) exhibited a high expression in pulmonary fibrosis. The phosphorylated c-Src protein was increased and the PI3K/AKT pathway was activated in model lung tissue. In vitro, silica increased the expression of TGF-β1- and TGF-β1-induced mesenchymal phenotype and fibrosis in a mouse epithelial cells line. siRNA-Src inhibited the c-Src, the phosphorylation of the PI3K/AKT pathway, and the mesenchymal phenotype induced by TGF-β1. LY294002, a specific inhibitor of PI3K, suppressed the phosphorylation of PI3K/AKT but did not affect Src activation. SU6656, a selective Src inhibitor, attenuated fibrosis in silicosis model. In summary, c-Src promotes fibrosis via the PI3K/AKT pathway in silica-induced lung fibrosis, and Src kinase inhibitors are potentially effective for silicosis treatment.
Collapse
|
6
|
Yamada S, Hashizume A, Hijikata Y, Inagaki T, Ito D, Kishimoto Y, Kinoshita F, Hirakawa A, Shimizu S, Nakamura T, Katsuno M. Mexiletine in spinal and bulbar muscular atrophy: a randomized controlled trial. Ann Clin Transl Neurol 2022; 9:1702-1714. [PMID: 36208052 PMCID: PMC9639628 DOI: 10.1002/acn3.51667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 09/09/2022] [Accepted: 09/12/2022] [Indexed: 11/22/2022] Open
Abstract
Objective Patients with spinal and bulbar muscular atrophy (SBMA) often experience muscular weakness under cold exposure. Methods In our previously conducted observational study, we assessed nerve conduction and grip strength to examine the effect of cold exposure on motor function, based on which we conducted a randomized controlled trial to evaluate the efficacy and safety of mexiletine hydrochloride in SBMA (MEXPRESS). Results In the observational study, 51 consecutive patients with SBMA and 18 healthy controls (HCs) were enrolled. Of the patients with SBMA, 88.0% experienced cold paresis. Patients with SBMA exhibited greater prolongation of ulnar nerve distal latency under cold (SBMA, 5.6 ± 1.1 msec; HC, 4.3 ± 0.6 msec; p <0.001); the change in the distal latencies between room temperature and cold exposure conditions correlated with the change in grip power. In the MEXPRESS trial, 20 participants took mexiletine or lactose, three times a day for 4 weeks with a crossover design. There was no difference in distal latencies at room temperature and under cold exposure between mexiletine and placebo groups as the primary endpoint. However, tongue pressure and 10‐sec grip and release test under cold exposure were improved in the mexiletine group. There were no serious adverse events throughout the study period. Interpretation Cold paresis is common and associated with prolongation of distal latency in SBMA. The results of the phase II clinical trial revealed that mexiletine showed short‐term safety, but it did not restore cold exposure‐induced prolongation of distal latency.
Collapse
Affiliation(s)
- Shinichiro Yamada
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Atsushi Hashizume
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Department of Clinical Research Education, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yasuhiro Hijikata
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tomonori Inagaki
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Daisuke Ito
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoshiyuki Kishimoto
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Fumie Kinoshita
- Department of Advanced Medicine, Nagoya University Hospital, Nagoya, Japan
| | - Akihiro Hirakawa
- Department of Clinical Biostatistics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Shinobu Shimizu
- Department of Advanced Medicine, Nagoya University Hospital, Nagoya, Japan
| | - Tomohiko Nakamura
- First Department of Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Masahisa Katsuno
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Department of Clinical Research Education, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
7
|
Ogura Y, Sahashi K, Hirunagi T, Iida M, Miyata T, Katsuno M. Mid1 is associated with androgen-dependent axonal vulnerability of motor neurons in spinal and bulbar muscular atrophy. Cell Death Dis 2022; 13:601. [PMID: 35821212 PMCID: PMC9276699 DOI: 10.1038/s41419-022-05001-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 05/31/2022] [Accepted: 06/07/2022] [Indexed: 01/21/2023]
Abstract
Spinal and bulbar muscular atrophy (SBMA) is an adult-onset hereditary neurodegenerative disease caused by the expansions of CAG repeats in the androgen receptor (AR) gene. Androgen-dependent nuclear accumulation of pathogenic AR protein causes degeneration of lower motor neurons, leading to progressive muscle weakness and atrophy. While the successful induction of SBMA-like pathology has been achieved in mouse models, mechanisms underlying motor neuron vulnerability remain unclear. In the present study, we performed a transcriptome-based screening for genes expressed exclusively in motor neurons and dysregulated in the spinal cord of SBMA mice. We found upregulation of Mid1 encoding a microtubule-associated RNA binding protein which facilitates the translation of CAG-expanded mRNAs. Based on the finding that lower motor neurons begin expressing Mid1 during embryonic stages, we developed an organotypic slice culture system of the spinal cord obtained from SBMA mouse fetuses to study the pathogenic role of Mid1 in SBMA motor neurons. Impairment of axonal regeneration arose in the spinal cord culture in SBMA mice in an androgen-dependent manner, but not in mice with non-CAG-expanded AR, and was either exacerbated or ameliorated by Mid1 overexpression or knockdown, respectively. Hence, an early Mid1 expression confers vulnerability to motor neurons, at least by inducing axonogenesis defects, in SBMA.
Collapse
Affiliation(s)
- Yosuke Ogura
- grid.27476.300000 0001 0943 978XDepartment of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kentaro Sahashi
- grid.27476.300000 0001 0943 978XDepartment of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tomoki Hirunagi
- grid.27476.300000 0001 0943 978XDepartment of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Madoka Iida
- grid.27476.300000 0001 0943 978XDepartment of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takaki Miyata
- grid.27476.300000 0001 0943 978XDepartment of Anatomy and Cell Biology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masahisa Katsuno
- grid.27476.300000 0001 0943 978XDepartment of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan ,grid.27476.300000 0001 0943 978XDepartment of Clinical Research Education, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
8
|
Imai Y, Iida M, Kanie K, Katsuno M, Kato R. Label-free morphological sub-population cytometry for sensitive phenotypic screening of heterogenous neural disease model cells. Sci Rep 2022; 12:9296. [PMID: 35710681 PMCID: PMC9203459 DOI: 10.1038/s41598-022-12250-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 04/20/2022] [Indexed: 11/20/2022] Open
Abstract
Label-free image analysis has several advantages with respect to the development of drug screening platforms. However, the evaluation of drug-responsive cells based exclusively on morphological information is challenging, especially in cases of morphologically heterogeneous cells or a small subset of drug-responsive cells. We developed a novel label-free cell sub-population analysis method called “in silico FOCUS (in silico analysis of featured-objects concentrated by anomaly discrimination from unit space)” to enable robust phenotypic screening of morphologically heterogeneous spinal and bulbar muscular atrophy (SBMA) model cells. This method with the anomaly discrimination concept can sensitively evaluate drug-responsive cells as morphologically anomalous cells through in silico cytometric analysis. As this algorithm requires only morphological information of control cells for training, no labeling or drug administration experiments are needed. The responses of SBMA model cells to dihydrotestosterone revealed that in silico FOCUS can identify the characteristics of a small sub-population with drug-responsive phenotypes to facilitate robust drug response profiling. The phenotype classification model confirmed with high accuracy the SBMA-rescuing effect of pioglitazone using morphological information alone. In silico FOCUS enables the evaluation of delicate quality transitions in cells that are difficult to profile experimentally, including primary cells or cells with no known markers.
Collapse
Affiliation(s)
- Yuta Imai
- Department of Basic Medicinal Sciences, Graduate School of Pharmaceutical Sciences, Nagoya University, Tokai National Higher Education and Research System, Furocho, Chikusa-ku, Nagoya, Aichi, 464-8601, Japan
| | - Madoka Iida
- Department of Neurology, Nagoya University Graduate School of Medicine, Tokai National Higher Education and Research System, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan
| | - Kei Kanie
- Department of Basic Medicinal Sciences, Graduate School of Pharmaceutical Sciences, Nagoya University, Tokai National Higher Education and Research System, Furocho, Chikusa-ku, Nagoya, Aichi, 464-8601, Japan
| | - Masahisa Katsuno
- Department of Neurology, Nagoya University Graduate School of Medicine, Tokai National Higher Education and Research System, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan.,Institute of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University, Tokai National Higher Education and Research System, Furocho, Chikusa-ku, Nagoya, Aichi, 464-8601, Japan.,Department of Clinical Research Education, Nagoya University Graduate School of Medicine, Tokai National Higher Education and Research System, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan.,Institute for Glyco-Core Research (iGCORE), Nagoya University, Tokai National Higher Education and Research System, Furocho, Chikusa-ku, Nagoya, Aichi, 464-8601, Japan
| | - Ryuji Kato
- Department of Basic Medicinal Sciences, Graduate School of Pharmaceutical Sciences, Nagoya University, Tokai National Higher Education and Research System, Furocho, Chikusa-ku, Nagoya, Aichi, 464-8601, Japan. .,Institute of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University, Tokai National Higher Education and Research System, Furocho, Chikusa-ku, Nagoya, Aichi, 464-8601, Japan. .,Institute for Glyco-Core Research (iGCORE), Nagoya University, Tokai National Higher Education and Research System, Furocho, Chikusa-ku, Nagoya, Aichi, 464-8601, Japan.
| |
Collapse
|
9
|
Tanaka Y, Murata M, Tanegashima K, Oda Y, Ito T. Nectin cell adhesion molecule 4 regulates angiogenesis through Src signaling and serves as a novel therapeutic target in angiosarcoma. Sci Rep 2022; 12:4031. [PMID: 35256687 PMCID: PMC8901754 DOI: 10.1038/s41598-022-07727-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 02/15/2022] [Indexed: 12/15/2022] Open
Abstract
Angiosarcoma is a rare, life-threatening soft tissue sarcoma with malignant endothelial cells that is mainly found in the skin. Multidisciplinary approaches are used to treat patients with unresectable metastasized lesions; considering the cellular origin of angiosarcoma, anti-angiogenic therapy has also been used recently. However, these treatments have limited efficacy, and the survival rate remains low. Thus, more effective treatments need to be developed. Nectin cell adhesion molecule 4 (NECTIN4) is highly expressed in malignant tumors and promotes tumor progression. Thus, NECTIN4 is expected to be a novel therapeutic target for cancer. However, the significance of NECTIN4 in angiosarcoma remains unknown. Using immunohistochemistry, we investigated NECTIN4 expression in 74 tissue samples from angiosarcoma patients, finding variable NECTIN4 expression. In addition, we investigated NECTIN4 expression and function in human angiosarcoma cell lines. NECTIN4 expression was higher in angiosarcoma cells than normal endothelial cells, and angiosarcoma cells were sensitive to monomethyl auristatin E, the cytotoxic part of a NECTIN4-targetting antibody-drug conjugate. NECTIN4 knockdown inhibited the proliferation and angiogenesis of angiosarcoma cells, and Src kinase signaling was shown to be involved in NECTIN4 function, at least in part. NECTIN4-targeted therapy has the potential to be a novel treatment strategy for angiosarcoma.
Collapse
Affiliation(s)
- Yuka Tanaka
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka City, Fukuoka, 812-8582, Japan
| | - Maho Murata
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka City, Fukuoka, 812-8582, Japan
| | - Keiko Tanegashima
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka City, Fukuoka, 812-8582, Japan
| | - Yoshinao Oda
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takamichi Ito
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka City, Fukuoka, 812-8582, Japan.
| |
Collapse
|
10
|
Clenbuterol-sensitive delayed outward potassium currents in a cell model of spinal and bulbar muscular atrophy. Pflugers Arch 2021; 473:1213-1227. [PMID: 34021780 DOI: 10.1007/s00424-021-02559-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 03/09/2021] [Accepted: 03/23/2021] [Indexed: 10/21/2022]
Abstract
Spinal and bulbar muscular atrophy (SBMA) is a neuromuscular disease caused by polyglutamine (polyQ) expansions in the androgen receptor (AR) gene. SBMA is characterized by selective dysfunction and degeneration of motor neurons in the brainstem and spinal cord through still unclear mechanisms in which ion channel modulation might play a central role as for other neurodegenerative diseases. The beta2-adrenergic agonist clenbuterol was observed to ameliorate the SBMA phenotype in mice and patient-derived myotubes. However, the underlying molecular mechanism has yet to be clarified. Here, we unveil that ionic current alterations induced by the expression of polyQ-expanded AR in motor neuron-derived MN-1 cells are attenuated by the administration of clenbuterol. Our combined electrophysiological and pharmacological approach allowed us to reveal that clenbuterol modifies delayed outward potassium currents. Overall, we demonstrated that the protection provided by clenbuterol restores the normal function through the modulation of KV2-type outward potassium currents, possibly contributing to the protective effect on motor neuron toxicity in SBMA.
Collapse
|
11
|
Molecular pathogenesis of spinal bulbar muscular atrophy (Kennedy's disease) and avenues for treatment. Curr Opin Neurol 2021; 33:629-634. [PMID: 32773451 DOI: 10.1097/wco.0000000000000856] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW The aim of this study was to illustrate the current understanding and avenues for developing treatment in spinal and bulbar muscular atrophy (SBMA), an inherited neuromuscular disorder caused by a CAG trinucleotide repeat expansion in the androgen receptor (AR) gene. RECENT FINDINGS Important advances have been made in characterizing the molecular mechanism of the disease, including the disruption of protein homeostasis, intracellular trafficking and signalling pathways. Biomarkers such as MRI quantification of muscle volume and fat fraction have been used to track disease progression, and will be useful in future clinical studies. Therapies tested and under development have been based on diverse strategies, including targeting mutant AR gene expression, stability and activity, and pathways that mitigate disease toxicity. SUMMARY We provide an overview of the recent advances in understanding the SBMA disease mechanism and highlight efforts to translate these insights into well tolerated and effective therapy.
Collapse
|
12
|
Selective suppression of polyglutamine-expanded protein by lipid nanoparticle-delivered siRNA targeting CAG expansions in the mouse CNS. MOLECULAR THERAPY. NUCLEIC ACIDS 2021; 24:1-10. [PMID: 33738134 PMCID: PMC7937577 DOI: 10.1016/j.omtn.2021.02.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 02/09/2021] [Indexed: 12/11/2022]
Abstract
Polyglutamine (polyQ) diseases are inherited neurodegenerative disorders caused by expansion of cytosine-adenine-guanine (CAG)-trinucleotide repeats in causative genes. These diseases include spinal and bulbar muscular atrophy (SBMA), Huntington’s disease, dentatorubral-pallidoluysian atrophy, and spinocerebellar ataxias. Targeting expanded CAG repeats is a common therapeutic approach to polyQ diseases, but concomitant silencing of genes with normal CAG repeats may lead to toxicity. Previous studies have shown that CAG repeat-targeting small interfering RNA duplexes (CAG-siRNAs) have the potential to selectively suppress mutant proteins in in vitro cell models of polyQ diseases. However, in vivo application of these siRNAs has not yet been investigated. In this study, we demonstrate that an unlocked nucleic acid (UNA)-modified CAG-siRNA shows high selectivity for polyQ-expanded androgen receptor (AR) inhibition in in vitro cell models and that lipid nanoparticle (LNP)-mediated delivery of the CAG-siRNA selectively suppresses mutant AR in the central nervous system of an SBMA mouse model. In addition, a subcutaneous injection of the LNP-delivered CAG-siRNA efficiently suppresses mutant AR in the skeletal muscle of the SBMA mouse model. These results support the therapeutic potential of LNP-delivered UNA-modified CAG-siRNAs for selective suppression of mutant proteins in SBMA and other polyQ diseases.
Collapse
|
13
|
Nagashima T, Hadiwidjaja S, Ohsumi S, Murata A, Hisada T, Kato R, Okada Y, Honda H, Shimizu K. In Vitro Model of Human Skeletal Muscle Tissues with Contractility Fabricated by Immortalized Human Myogenic Cells. ACTA ACUST UNITED AC 2020; 4:e2000121. [DOI: 10.1002/adbi.202000121] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 10/04/2020] [Indexed: 12/12/2022]
Affiliation(s)
- Takunori Nagashima
- Department of Biomolecular Engineering Graduate School of Engineering Nagoya University Nagoya 464‐8603 Japan
| | - Stacy Hadiwidjaja
- Department of Biomolecular Engineering Graduate School of Engineering Nagoya University Nagoya 464‐8603 Japan
| | - Saki Ohsumi
- Department of Biomolecular Engineering Graduate School of Engineering Nagoya University Nagoya 464‐8603 Japan
| | - Akari Murata
- Department of Biomolecular Engineering Graduate School of Engineering Nagoya University Nagoya 464‐8603 Japan
| | - Takumi Hisada
- Department of Basic Medicinal Sciences Graduate School of Pharmaceutical Sciences Nagoya University Nagoya 464‐8601 Japan
| | - Ryuji Kato
- Department of Basic Medicinal Sciences Graduate School of Pharmaceutical Sciences Nagoya University Nagoya 464‐8601 Japan
| | - Yohei Okada
- Department of Neurology Aichi Medical University School of Medicine Aichi 480‐1195 Japan
| | - Hiroyuki Honda
- Department of Biomolecular Engineering Graduate School of Engineering Nagoya University Nagoya 464‐8603 Japan
| | - Kazunori Shimizu
- Department of Biomolecular Engineering Graduate School of Engineering Nagoya University Nagoya 464‐8603 Japan
| |
Collapse
|
14
|
Hashizume A, Fischbeck KH, Pennuto M, Fratta P, Katsuno M. Disease mechanism, biomarker and therapeutics for spinal and bulbar muscular atrophy (SBMA). J Neurol Neurosurg Psychiatry 2020; 91:1085-1091. [PMID: 32934110 DOI: 10.1136/jnnp-2020-322949] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 07/10/2020] [Accepted: 07/27/2020] [Indexed: 12/13/2022]
Abstract
Spinal and bulbar muscular atrophy (SBMA) is a hereditary neuromuscular disorder caused by CAG trinucleotide expansion in the gene encoding the androgen receptor (AR). In the central nervous system, lower motor neurons are selectively affected, whereas pathology of patients and animal models also indicates involvement of skeletal muscle including loss of fast-twitch type 2 fibres and increased slow-twitch type 1 fibres, together with a glycolytic-to-oxidative metabolic switch. Evaluation of muscle and fat using MRI, in addition to biochemical indices such as serum creatinine level, are promising biomarkers to track the disease progression. The serum level of creatinine starts to decrease before the onset of muscle weakness, followed by the emergence of hand tremor, a prodromal sign of the disease. Androgen-dependent nuclear accumulation of the polyglutamine-expanded AR is an essential step in the pathogenesis, providing therapeutic opportunities via hormonal manipulation and gene silencing with antisense oligonucleotides. Animal studies also suggest that hyperactivation of Src, alteration of autophagy and a mitochondrial deficit underlie the neuromuscular degeneration in SBMA and provide alternative therapeutic targets.
Collapse
MESH Headings
- 5-alpha Reductase Inhibitors/therapeutic use
- Adipose Tissue/diagnostic imaging
- Adrenergic beta-Agonists/therapeutic use
- Autophagy
- Biomarkers
- Bulbo-Spinal Atrophy, X-Linked/diagnostic imaging
- Bulbo-Spinal Atrophy, X-Linked/metabolism
- Bulbo-Spinal Atrophy, X-Linked/physiopathology
- Bulbo-Spinal Atrophy, X-Linked/therapy
- Clenbuterol/therapeutic use
- Creatinine/metabolism
- Dutasteride/therapeutic use
- Glycolysis
- Humans
- Insulin-Like Growth Factor I/analogs & derivatives
- Leuprolide/therapeutic use
- Magnetic Resonance Imaging
- Mitochondria/metabolism
- Muscle Fibers, Fast-Twitch/metabolism
- Muscle Fibers, Fast-Twitch/pathology
- Muscle Fibers, Slow-Twitch/metabolism
- Muscle Fibers, Slow-Twitch/pathology
- Muscle, Skeletal/diagnostic imaging
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Oligonucleotides, Antisense/therapeutic use
- Oxidation-Reduction
- RNAi Therapeutics
- Receptors, Androgen/genetics
- Receptors, Androgen/metabolism
- Trinucleotide Repeat Expansion
Collapse
Affiliation(s)
- Atsushi Hashizume
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Kenneth H Fischbeck
- Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Maria Pennuto
- Department of Biomedical Sciences (DBS), University of Padova, Padova, Italy
- Veneto Institute of Molecular Medicine (VIMM), Padova, Italy
| | - Pietro Fratta
- Depatment of Neuromuscular Diseases, University College London Institute of Neurology, London, UK
- MRC Centre for Neuromuscular Diseases, University College London Institute of Neurology, London, UK
| | - Masahisa Katsuno
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| |
Collapse
|