1
|
Hou Y, Guo X, Ran J, Lu X, Xie C. Conductive polyphenol microneedles coupled with electroacupuncture to accelerate wound healing and alleviate depressive-like behaviors in diabetes. Bioact Mater 2025; 44:516-530. [PMID: 39584064 PMCID: PMC11583732 DOI: 10.1016/j.bioactmat.2024.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 11/01/2024] [Accepted: 11/01/2024] [Indexed: 11/26/2024] Open
Abstract
Inflammation and depression are serious complications of diabetes that interact to form a feedback loop and may hinder diabetic wound healing. They share a common pathophysiological basis of abnormal interactions between diabetic wounds and the brain. Here, we propose a strategy combining electroacupuncture (EA) stimulation of the Dazhui acupoint (GV14) with polyphenol-mediated conductive hydrogel microneedles to promote diabetic wound healing and alleviate depression through local wound-brain interactions. The conductive microneedles comprised methacrylated gelatin, dopamine (DA), DA-modified poly(3,4-ethylenedioxythiophene), and Lycium barbarum polysaccharide. EA at GV14 activated the vagus-adrenal axis to inhibit systemic inflammation while DA coupled electrical signals for long-term inhibition of local wound inflammation. EA at GV14 was also found to elevate 5-hydroxytryptamine levels in rats with diabetic wounds, consequently mitigating depressive-like behaviors. Additionally, the polyphenol-mediated conductive hydrogel microneedles, and coupled with EA stimulation promoted healing of wound tissue and peripheral nerves. This strategy regulated both local and systemic inflammation while alleviating depressive-like behaviors in diabetic rats, providing a new clinical perspective for the treatment of diabetes-related and emotional disorders.
Collapse
Affiliation(s)
- Yue Hou
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
| | - Xiaochuan Guo
- Department of Rehabilitation Medicine, Affiliated Hospital of Southwest Jiaotong University, The Third People's Hospital of Chengdu, Chengdu, Sichuan, 610031, China
| | - Jinhui Ran
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
| | - Xiong Lu
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
| | - Chaoming Xie
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
| |
Collapse
|
2
|
Leibovit-Reiben Z, Godfrey H, Jedlowski P, Thiede R. Neurologic adverse events associated with BRAF and MEK inhibitor therapy in patients with malignant melanoma: a disproportionality analysis using the Food and Drug Administration Adverse Event Reporting System. Melanoma Res 2024:00008390-990000000-00181. [PMID: 39656586 DOI: 10.1097/cmr.0000000000001015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
BRAF and MEK inhibitor (BRAFi + MEKi) therapy has improved the treatment of solid tumors with BRAF mutation. However, their neurologic adverse events (nAEs) have been largely unexplored. This study aimed to provide clinicians with more updated knowledge on nAEs associated with BRAFi + MEKi therapy in patients with malignant melanoma compared with nonmelanoma cancers. The United States Food and Drug Administration Adverse Event Reporting System was queried from 2011 to 2022 to capture nAEs reported for the BRAFi + MEKi therapies, vemurafenib plus cobimetinib (V + C), dabrafenib plus trametinib (D + T), and encorafenib plus binimetinib (E + B). A disproportionality analysis was performed to calculate their reporting odds ratios (RORs) and 95% confidence intervals (CIs) using a control group of antineoplastic medications. There were 2881 BRAFi + MEKi therapy-associated nAE cases, the majority of which listed malignant melanoma as the reason for use (87.5, 66.7, and 62.0% for V + C, D + T, and E + B, respectively). Several novel associations were identified; including epidural lipomatosis (ROR: 320.07, 95% CI: 123.76-827.77 for V + C), peripheral nerve lesion (ROR: 185.64, 95% CI: 73.95-466.03 for V + C), Guillain-Barre syndrome (RORs: 8.80, 2.94, and 11.79, 95% CIs: 3.65-21.22, 1.40-6.19, and 5.87-23.66 for V + C, D + T, and E + B), demyelinating polyneuropathy (RORs: 24.72 and 78.98, 95% CI: 8.16-74.86 and 24.84-251.13 for D + T and E + B), and multiple sclerosis (ROR: 5.90, 95% CI: 3.06-11.40 for D + T) in melanoma patients. nAEs in the setting of BRAFi + MEKi therapy should be a safety consideration when utilizing these medications.
Collapse
Affiliation(s)
| | | | - Patrick Jedlowski
- Department of Dermatology, University of Arizona College of Medicine - Tucson, Tucson, Arizona, USA
| | - Rebecca Thiede
- Department of Dermatology, University of Arizona College of Medicine - Tucson, Tucson, Arizona, USA
| |
Collapse
|
3
|
Jo Y, Webster MJ, Kim S, Lee D. Interpretation of SNP combination effects on schizophrenia etiology based on stepwise deep learning with multi-precision data. Brief Funct Genomics 2024; 23:663-671. [PMID: 37738675 PMCID: PMC11428150 DOI: 10.1093/bfgp/elad041] [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: 01/19/2023] [Revised: 07/17/2023] [Accepted: 08/22/2023] [Indexed: 09/24/2023] Open
Abstract
Schizophrenia genome-wide association studies (GWAS) have reported many genomic risk loci, but it is unclear how they affect schizophrenia susceptibility through interactions of multiple SNPs. We propose a stepwise deep learning technique with multi-precision data (SLEM) to explore the SNP combination effects on schizophrenia through intermediate molecular and cellular functions. The SLEM technique utilizes two levels of precision data for learning. It constructs initial backbone networks with more precise but small amount of multilevel assay data. Then, it learns strengths of intermediate interactions with the less precise but massive amount of GWAS data. The learned networks facilitate identifying effective SNP interactions from the intractably large space of all possible SNP combinations. We have shown that the extracted SNP combinations show higher accuracy than any single SNPs and preserve the accuracy in an independent dataset. The learned networks also provide interpretations of molecular and cellular interactions of SNP combinations toward schizophrenia etiology.
Collapse
Affiliation(s)
- Yousang Jo
- Department of Bio and Brain Engineering, KAIST, Daejeon, South Korea
| | - Maree J Webster
- Brain Research Laboratory, Stanley Medical Research Institute, Rockville, MD, USA
| | - Sanghyeon Kim
- Brain Research Laboratory, Stanley Medical Research Institute, Rockville, MD, USA
| | - Doheon Lee
- Department of Bio and Brain Engineering, KAIST, Daejeon, South Korea
| |
Collapse
|
4
|
Nwabo Kamdje AH, Dongmo Fogang HP, Mimche PN. Role of epigenetic in cancer biology, in hematologic malignancies and in anticancer therapy. FRONTIERS IN MOLECULAR MEDICINE 2024; 4:1426454. [PMID: 39308891 PMCID: PMC11412843 DOI: 10.3389/fmmed.2024.1426454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 08/08/2024] [Indexed: 09/25/2024]
Abstract
Major epigenetic changes are associated with carcinogenesis, including aberrant DNA methylations and post-translational modifications of histone. Indeed evidence accumulated in recent years indicates that inactivating DNA hypermethylation preferentially targets the subset of polycomb group (PcG) genes that are regulators of developmental processes. Conversely, activating DNA hypomethylation targets oncogenic signaling pathway genes, but outcomes of both events lead in the overexpression of oncogenic signaling pathways that contribute to the stem-like state of cancer cells. On the basis of recent evidence from population-basedclinical and experimental studies, we hypothesize that factors associated with risk for developing a hematologic malignancy (HM), such as metabolic syndrome and chronic inflammation, may trigger epigenetic mechanisms to increase the transcriptional expression of oncogenes and activate oncogenic signaling pathways. Signaling pathways associated with such risk factors include but are not limited to pro-inflammatory nuclear factor κB (NF-κB) and mitogenic, growth, and survival Janus kinase (JAK) intracellular non-receptor tyrosine kinase-triggered pathways. The latter includes signaling pathways such as transducer and activator of transcription (STAT), Ras GTPases/mitogen-activated protein kinases (MAPKs)/extracellular signal-related kinases (ERKs), phosphatidylinositol 3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR), and β-catenin pathways. Recent findings on epigenetic mechanisms at work in the biology of cancer and in HMs and their importance in the etiology and pathogenesis of these diseases are herein summarized and discussed. Furthermore, the role of epigenetic processes in the determination of biological identity, the consequences for interindividual variability in disease clinical profile, and the potential of epigenetic drugs in HMs are also considered.
Collapse
Affiliation(s)
- Armel Hervé Nwabo Kamdje
- Department of Physiological Sciences and Biochemistry, Faculty of Medicine and Biomedical Sciences, University of Garoua, Garoua, Cameroon
| | - Hervet Paulain Dongmo Fogang
- Department of Physiological Sciences and Biochemistry, Faculty of Medicine and Biomedical Sciences, University of Garoua, Garoua, Cameroon
| | - Patrice N. Mimche
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Medicine, Division of Gastroenterology and Hepatology, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
5
|
Li Z, Yu Y, Bu Y, Liu C, Liu E, Jin J, Chen G, Li C, Wang H, Li H, Han L, Zhang Y, Gong W, Luo J, Xiao H, Yue Z. Targeting macrophagic RasGRP1 with catechin hydrate ameliorates sepsis-induced multiorgan dysfunction. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 130:155733. [PMID: 38759314 DOI: 10.1016/j.phymed.2024.155733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 05/01/2024] [Accepted: 05/11/2024] [Indexed: 05/19/2024]
Abstract
BACKGROUND The proinflammatory response induced by macrophages plays a crucial role in the development of sepsis and the resulting multiorgan dysfunction. Identifying new regulatory targets for macrophage homeostasis and devising effective treatment strategies remains a significant challenge in contemporary research. PURPOSE This study aims to identify new regulatory targets for macrophage homeostasis and develop effective strategies for treating sepsis. STUDY DESIGN AND METHODS Macrophage infiltration in septic patients and in lungs, kidneys, and brains of caecum ligation and puncture (CLP)-induced septic mice was observed using CIBERSORT and immunofluorescence (IF). Upon integrating the MSigDB database and GSE65682 dataset, differently expressed macrophage-associated genes (DEMAGs) were identified. Critical DEMAGs were confirmed through machine learning. The protein level of the critical DEMAG was detected in PBMCs of septic patients, RAW264.7 cells, and mice lungs, kidneys, and brains using ELISA, western blot, immunohistochemistry, and IF. siRNA was applied to investigate the effect of the critical DEMAG in RAW264.7 cells. A natural product library was screened to find a compound targeting the critical DEMAG protein. The binding of compounds and proteins was analyzed through molecular docking, molecular dynamics simulations, CETSA, and MST analysis. The therapeutic efficacy of the compounds against sepsis was then evaluated through in vitro and in vivo experiments. RESULTS Macrophage infiltration was inversely correlated with survival in septic patients. The critical differentially expressed molecule RasGRP1 was frequently observed in the PBMCs of septic patients, LPS-induced RAW264.7 cells, and the lungs, kidneys, and brains of septic mice. Silencing RasGRP1 alleviated proinflammatory response and oxidative stress in LPS-treated RAW264.7 cells. Catechin Hydrate (CH) was identified as an inhibitor of RasGRP1, capable of maintaining macrophage homeostasis and mitigating lung, kidney, and brain damage during sepsis. CONCLUSION This study demonstrates that RasGRP1, a novel activator of macrophage proinflammatory responses, plays a crucial role in the excessive inflammation and oxidative stress associated with sepsis. CH shows potential for treating sepsis by inhibiting RasGRP1.
Collapse
Affiliation(s)
- Zhixi Li
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Harbin 150001, PR China; Heilongjiang Province Key Laboratory of Research on Anesthesiology and Critical Care Medicine, 246 Xuefu Road, Harbin 150001, PR China; The Key Laboratory of Myocardial Ischemia Organization, Chinese Ministry of Education, 246 Xuefu Road, Harbin 150001, PR China
| | - Yongjing Yu
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Harbin 150001, PR China; Heilongjiang Province Key Laboratory of Research on Anesthesiology and Critical Care Medicine, 246 Xuefu Road, Harbin 150001, PR China; The Key Laboratory of Myocardial Ischemia Organization, Chinese Ministry of Education, 246 Xuefu Road, Harbin 150001, PR China
| | - Yue Bu
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Harbin 150001, PR China; Heilongjiang Province Key Laboratory of Research on Anesthesiology and Critical Care Medicine, 246 Xuefu Road, Harbin 150001, PR China; Department of Pain Medicine, Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Harbin 150001, PR China
| | - Chang Liu
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Harbin 150001, PR China; Heilongjiang Province Key Laboratory of Research on Anesthesiology and Critical Care Medicine, 246 Xuefu Road, Harbin 150001, PR China; The Key Laboratory of Myocardial Ischemia Organization, Chinese Ministry of Education, 246 Xuefu Road, Harbin 150001, PR China
| | - Enran Liu
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Harbin 150001, PR China; Heilongjiang Province Key Laboratory of Research on Anesthesiology and Critical Care Medicine, 246 Xuefu Road, Harbin 150001, PR China
| | - Jiaqi Jin
- The Key Laboratory of Myocardial Ischemia Organization, Chinese Ministry of Education, 246 Xuefu Road, Harbin 150001, PR China; Department of Neurology, Xuanwu Hospital, Capital Medical University, 45 Changchun Road, Beijing 100053, PR China
| | - Guangmin Chen
- Department of Anesthesiology, First Affiliated Hospital of Harbin Medical University, 199 Dazhi Road, Harbin 150001, PR China
| | - Chenglong Li
- Department of Anesthesiology, Fourth Affiliated Hospital of Harbin Medical University, 37 Yiyuan Road, Harbin 150001, PR China
| | - Hongyu Wang
- Department of Cardiology, First Affiliated Hospital of Harbin Medical University, 199 Dazhi Road, Harbin 150001, PR China
| | - Hui Li
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Harbin 150001, PR China; Heilongjiang Province Key Laboratory of Research on Anesthesiology and Critical Care Medicine, 246 Xuefu Road, Harbin 150001, PR China
| | - Lei Han
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Harbin 150001, PR China; Heilongjiang Province Key Laboratory of Research on Anesthesiology and Critical Care Medicine, 246 Xuefu Road, Harbin 150001, PR China
| | - Yan Zhang
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Harbin 150001, PR China; Heilongjiang Province Key Laboratory of Research on Anesthesiology and Critical Care Medicine, 246 Xuefu Road, Harbin 150001, PR China
| | - Weidong Gong
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Harbin 150001, PR China; Heilongjiang Province Key Laboratory of Research on Anesthesiology and Critical Care Medicine, 246 Xuefu Road, Harbin 150001, PR China
| | - Juan Luo
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Harbin 150001, PR China; Heilongjiang Province Key Laboratory of Research on Anesthesiology and Critical Care Medicine, 246 Xuefu Road, Harbin 150001, PR China
| | - Haichuan Xiao
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Harbin 150001, PR China; Heilongjiang Province Key Laboratory of Research on Anesthesiology and Critical Care Medicine, 246 Xuefu Road, Harbin 150001, PR China
| | - Ziyong Yue
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Harbin 150001, PR China; Heilongjiang Province Key Laboratory of Research on Anesthesiology and Critical Care Medicine, 246 Xuefu Road, Harbin 150001, PR China.
| |
Collapse
|
6
|
Xu Y, Xu C, Song H, Feng X, Ma L, Zhang X, Li G, Mu C, Tan L, Zhang Z, Liu Z, Luo Z, Yang C. Biomimetic bone-periosteum scaffold for spatiotemporal regulated innervated bone regeneration and therapy of osteosarcoma. J Nanobiotechnology 2024; 22:250. [PMID: 38750519 PMCID: PMC11094931 DOI: 10.1186/s12951-024-02430-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 03/20/2024] [Indexed: 05/19/2024] Open
Abstract
The complexity of repairing large segment defects and eradicating residual tumor cell puts the osteosarcoma clinical management challenging. Current biomaterial design often overlooks the crucial role of precisely regulating innervation in bone regeneration. Here, we develop a Germanium Selenium (GeSe) co-doped polylactic acid (PLA) nanofiber membrane-coated tricalcium phosphate bioceramic scaffold (TCP-PLA/GeSe) that mimics the bone-periosteum structure. This biomimetic scaffold offers a dual functionality, combining piezoelectric and photothermal conversion capabilities while remaining biodegradable. When subjected to ultrasound irradiation, the US-electric stimulation of TCP-PLA/GeSe enables spatiotemporal control of neurogenic differentiation. This feature supports early innervation during bone formation, promoting early neurogenic differentiation of Schwann cells (SCs) by increasing intracellular Ca2+ and subsequently activating the PI3K-Akt and Ras signaling pathways. The biomimetic scaffold also demonstrates exceptional osteogenic differentiation potential under ultrasound irradiation. In rabbit model of large segment bone defects, the TCP-PLA/GeSe demonstrates promoted osteogenesis and nerve fibre ingrowth. The combined attributes of high photothermal conversion capacity and the sustained release of anti-tumor selenium from the TCP-PLA/GeSe enable the synergistic eradication of osteosarcoma both in vitro and in vivo. This strategy provides new insights on designing advanced biomaterials of repairing large segment bone defect and osteosarcoma.
Collapse
Affiliation(s)
- Yan Xu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Chao Xu
- College of Materials Science and Engineering, Wuhan Textile University, Wuhan, 430200, China
| | - Huan Song
- Otorhinolaryngology Head and Neck Surgery, Wuhan Fourth Hospital, Wuhan, Hubei, 430033, China
| | - Xiaobo Feng
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Liang Ma
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Xiaoguang Zhang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Gaocai Li
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Congpu Mu
- Center for High Pressure Science, State Key Laboratory of Metastable Materials Science and Technology, Yanshan University, Qinhuangdao, 066004, China
| | - Lei Tan
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Zhengdong Zhang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430074, China.
- School of Clinical Medicine, Department of Orthopedics, Chengdu Medical College, the First Affiliated Hospital of Chengdu Medical College, Chengdu, 610500, China.
| | - Zhongyuan Liu
- Center for High Pressure Science, State Key Laboratory of Metastable Materials Science and Technology, Yanshan University, Qinhuangdao, 066004, China
| | - Zhiqiang Luo
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Cao Yang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430074, China.
| |
Collapse
|
7
|
Chen Y, Jiang L, Li M, Shen Y, Liu S, Yang D. Huanglian Jiedu decoction alleviates neurobehavioral damage in mice with chronic alcohol exposure through the RAS-RAF-MEK-ERK pathway. Heliyon 2024; 10:e29556. [PMID: 38644875 PMCID: PMC11033144 DOI: 10.1016/j.heliyon.2024.e29556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 04/09/2024] [Accepted: 04/10/2024] [Indexed: 04/23/2024] Open
Abstract
Objective Long-term alcohol consumption can cause organic damage to the brain, resulting in mental and nervous system abnormalities and intellectual impairment. Huanglian Jiedu decoction (HLJDD) is the classic representative of clearing heat and detoxifying. This study aimed to explore the effects and possible mechanisms of HLJDD on brain injury in chronic alcohol-exposed mice. Methods The alcohol-exposed mice were treated with different doses of HLJDD to observe behavioral changes, hippocampal Aβ1-42 deposition, number and ultrastructural changes of neurons in the hippocampus and prefrontal cortex, and expressions of synaptic proteins. On this basis, transcriptome sequencing was used to analyze the differentially expressed genes in different treatment groups, and functional enrichment analysis was performed. Then, WB and RT-PCR were used to verify the expression of the pathway. Results Chronic alcohol exposure reduced body weight in mice, led to motor cognitive impairment, increased Aβ1-42 in the hippocampus, decreased the number of neurons in the hippocampus and prefrontal cortex, and the expression of PSD95 and SYN in the hippocampus. HLJDD significantly improved the cognitive dysfunction of mice and alleviated the damage of the hippocampus and prefrontal cortex. Transcriptome sequencing results showed that the regulatory effects of HLJDD on chronic alcohol-exposed mice may be related to the RAS pathway. Further experiments confirmed that chronic alcohol exposure caused a significant increase in protein and gene expressions of the RAS-RAF-MEK-ERK pathway in mouse, and this activation was reversed by HLJDD. Conclusion HLJDD may ameliorate brain damage caused by chronic alcohol exposure by regulating the RAS-RAF-MEK-ERK pathway.
Collapse
Affiliation(s)
- Yun Chen
- Department of Neurology, Chengdu University of Traditional Chinese Medicine, Sichuan, PR China
- Department of Neurology, The First People's Hospital of Bijie City, Guizhou, PR China
| | - Lianyan Jiang
- Department of Neurology, Chengdu University of Traditional Chinese Medicine, Sichuan, PR China
| | - Mao Li
- Department of Neurology, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, PR China
| | - Yuling Shen
- Department of Neurology, Chengdu University of Traditional Chinese Medicine, Sichuan, PR China
| | - Shanyu Liu
- Department of Neurology, Chengdu University of Traditional Chinese Medicine, Sichuan, PR China
| | - Dongdong Yang
- Department of Neurology, The Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Sichuan, PR China
| |
Collapse
|
8
|
Cherra SJ, Lamb R. Interactions between Ras and Rap signaling pathways during neurodevelopment in health and disease. Front Mol Neurosci 2024; 17:1352731. [PMID: 38463630 PMCID: PMC10920261 DOI: 10.3389/fnmol.2024.1352731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 02/08/2024] [Indexed: 03/12/2024] Open
Abstract
The Ras family of small GTPases coordinates tissue development by modulating cell proliferation, cell-cell adhesion, and cellular morphology. Perturbations of any of these key steps alter nervous system development and are associated with neurological disorders. While the underlying causes are not known, genetic mutations in Ras and Rap GTPase signaling pathways have been identified in numerous neurodevelopmental disorders, including autism spectrum, neurofibromatosis, intellectual disability, epilepsy, and schizophrenia. Despite diverse clinical presentations, intersections between these two signaling pathways may provide a better understanding of how deviations in neurodevelopment give rise to neurological disorders. In this review, we focus on presynaptic and postsynaptic functions of Ras and Rap GTPases. We highlight various roles of these small GTPases during synapse formation and plasticity. Based on genomic analyses, we discuss how disease-related mutations in Ras and Rap signaling proteins may underlie human disorders. Finally, we discuss how recent observations have identified molecular interactions between these pathways and how these findings may provide insights into the mechanisms that underlie neurodevelopmental disorders.
Collapse
Affiliation(s)
- Salvatore J. Cherra
- Department of Neuroscience, University of Kentucky College of Medicine, Lexington, KY, United States
| | | |
Collapse
|
9
|
Zhang FL, Ma HH, Dong PY, Yuan ZN, Zhang SE, Zhao AH, Liu HQ, De Felici M, Shen W, Zhang XF. Aflatoxin B1 disrupts testicular development via the cell cycle-related Ras/PI3K/Akt signaling in mice and pig. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 329:121729. [PMID: 37116564 DOI: 10.1016/j.envpol.2023.121729] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 04/24/2023] [Accepted: 04/25/2023] [Indexed: 05/03/2023]
Abstract
Aflatoxins B1 (AFB1), a type I carcinogen widely present in the environment, not only poses a danger to animal husbandry, but also poses a potential threat to human reproductive health, but its mechanism is still unclear. To address this question, multi-omics were performed on porcine Sertoli cells and mice testis. The data suggest that AFB1 induced testicular damage manifested as decreased expression of GJA1, ZO1 and OCCLUDIN in mice (p < 0.01) and inhibition of porcine Sertoli cell proliferation. Transcriptomic analysis suggested changes in noncoding RNA expression profiles that affect the cell cycle-related Ras/PI3K/Akt signaling pathway after AFB1 exposure both in mice and pigs. Specifically, AFB1 caused abnormal cell cycle of testis with the characterization of decreased expressions of CCNA1, CCNB1 and CDK1 (p < 0.01). Flow cytometry revealed that the G2/M phase was significantly increased after AFB1 exposure. Meanwhile, AFB1 downregulated the expressions of Ras, PI3K and AKT both in porcine Sertoli cell (p < 0.01) and mice testis (p < 0.01). Metabolome analysis verified the alterations in the PI3K/Akt signaling pathway (p < 0.05). Moreover, the joint analysis of metabolome and microbiome found that the changes of metabolites were correlated with the expression of flora. In conclusion, we have demonstrated that AFB1 impairs testicular development via the cell cycle-related Ras/PI3K/Akt signaling.
Collapse
Affiliation(s)
- Fa-Li Zhang
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, 266100, China; College of Life Sciences, Key Laboratory of Animal Reproduction and Biotechnology in Universities of Shandong, Qingdao Agricultural University, Qingdao, 266109, China; College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an, 271018, China
| | - Hao-Hai Ma
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, 266100, China
| | - Pei-Yu Dong
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, 266100, China
| | - Zheng-Ning Yuan
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, 266100, China
| | - Shu-Er Zhang
- Animal Husbandry General Station of Shandong Province, Jinan, 250010, China
| | - Ai-Hong Zhao
- Qingdao Academy of Agricultural Sciences, Qingdao, 266100, China
| | - Huan-Qi Liu
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, 266100, China
| | - Massimo De Felici
- Department of Biomedicine and Prevention, University of Rome 'Tor Vergata', Rome, 00133, Italy
| | - Wei Shen
- College of Life Sciences, Key Laboratory of Animal Reproduction and Biotechnology in Universities of Shandong, Qingdao Agricultural University, Qingdao, 266109, China
| | - Xi-Feng Zhang
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, 266100, China.
| |
Collapse
|
10
|
Ye Q, Srivastava P, Al-Kuwari N, Chen X. Oncogenic BRAFV600E induces microglial proliferation through extracellular signal-regulated kinase and neuronal death through c-Jun N-terminal kinase. Neural Regen Res 2023; 18:1613-1622. [PMID: 36571370 PMCID: PMC10075110 DOI: 10.4103/1673-5374.361516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 06/13/2022] [Accepted: 10/18/2022] [Indexed: 11/19/2022] Open
Abstract
Activating V600E in v-Raf murine sarcoma viral oncogene homolog B (BRAF) is a common driver mutation in cancers of multiple tissue origins, including melanoma and glioma. BRAFV600E has also been implicated in neurodegeneration. The present study aims to characterize BRAFV600E during cell death and proliferation of three major cell types of the central nervous system: neurons, astrocytes, and microglia. Multiple primary cultures (primary cortical mixed culture) and cell lines of glial cells (BV2) and neurons (SH-SY5Y) were employed. BRAFV600E and BRAFWT expression was mediated by lentivirus or retrovirus. Blockage of downstream effectors (extracellular signal-regulated kinase 1/2 and JNK1/2) were achieved by siRNA. In astrocytes and microglia, BRAFV600E induces cell proliferation, and the proliferative effect in microglia is mediated by activated extracellular signal-regulated kinase, but not c-Jun N-terminal kinase. Conditioned medium from BRAFV600E-expressing microglia induced neuronal death. In neuronal cells, BRAFV600E directly induces neuronal death, through c-Jun N-terminal kinase but not extracellular signal-regulated kinase. We further show that BRAF-related genes are enriched in pathways in patients with Parkinson's disease. Our study identifies distinct consequences mediated by distinct downstream effectors in dividing glial cells and in neurons following the same BRAF mutational activation and a causal link between BRAF-activated microglia and neuronal cell death that does not require physical proximity. It provides insight into a possibly important role of BRAF in neurodegeneration as a result of either dysregulated BRAF in neurons or its impact on glial cells.
Collapse
Affiliation(s)
- Qing Ye
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
- Department of Neurology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Pranay Srivastava
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Nasser Al-Kuwari
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Xiqun Chen
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| |
Collapse
|
11
|
Yang Y, Song R, Gao Y, Yu H, Wang S. Regulatory mechanisms and therapeutic potential of JAB1 in neurological development and disorders. Mol Med 2023; 29:80. [PMID: 37365502 DOI: 10.1186/s10020-023-00675-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 05/30/2023] [Indexed: 06/28/2023] Open
Abstract
c-Jun activation domain binding protein-1 (JAB1) is a multifunctional regulator that plays vital roles in diverse cellular processes. It regulates AP-1 transcriptional activity and also acts as the fifth component of the COP9 signalosome complex. While JAB1 is considered an oncoprotein that triggers tumor development, recent studies have shown that it also functions in neurological development and disorders. In this review, we summarize the general features of the JAB1 gene and protein, and present recent updates on the regulation of JAB1 expression. Moreover, we also highlight the functional roles and regulatory mechanisms of JAB1 in neurodevelopmental processes such as neuronal differentiation, synaptic morphogenesis, myelination, and hair cell development and in the pathogenesis of some neurological disorders such as Alzheimer's disease, multiple sclerosis, neuropathic pain, and peripheral nerve injury. Furthermore, current challenges and prospects are discussed, including updates on drug development targeting JAB1.
Collapse
Affiliation(s)
- Yu Yang
- Department of Psychiatry, Jining Medical University, Jianshe South Road No. 45, Jining, Shandong, China
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Jining Medical University, Jining, Shandong, China
| | - Ruying Song
- Department of Psychiatry, Jining Medical University, Jianshe South Road No. 45, Jining, Shandong, China
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Jining Medical University, Jining, Shandong, China
| | - Yiming Gao
- Department of Psychiatry, Jining Medical University, Jianshe South Road No. 45, Jining, Shandong, China
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Jining Medical University, Jining, Shandong, China
| | - Hao Yu
- Department of Psychiatry, Jining Medical University, Jianshe South Road No. 45, Jining, Shandong, China.
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Jining Medical University, Jining, Shandong, China.
| | - Shuai Wang
- Department of Psychiatry, Jining Medical University, Jianshe South Road No. 45, Jining, Shandong, China.
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Jining Medical University, Jining, Shandong, China.
| |
Collapse
|
12
|
Kim HW, Wang S, Davies AJ, Oh SB. The therapeutic potential of natural killer cells in neuropathic pain. Trends Neurosci 2023:S0166-2236(23)00133-9. [PMID: 37385878 DOI: 10.1016/j.tins.2023.05.008] [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: 03/15/2023] [Revised: 05/04/2023] [Accepted: 05/17/2023] [Indexed: 07/01/2023]
Abstract
Novel disease-modifying treatments for neuropathic pain are urgently required. The cellular immune response to nerve injury represents a promising target for therapeutic development. Recently, the role of natural killer (NK) cells in both CNS and PNS disease has been the subject of growing interest. In this opinion article, we set out the case for NK cell-based intervention as a promising avenue for development in the management of neuropathic pain. We explore the potential cellular and molecular targets of NK cells in the PNS by contrasting with their reported functional roles in CNS diseases, and we suggest strategies for using the beneficial functions of NK cells and immune-based therapeutics in the context of neuropathic pain.
Collapse
Affiliation(s)
- Hyoung Woo Kim
- Department of Neurobiology and Physiology, School of Dentistry, and Dental Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Shuaiwei Wang
- Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Alexander J Davies
- Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford, UK.
| | - Seog Bae Oh
- Department of Neurobiology and Physiology, School of Dentistry, and Dental Research Institute, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
13
|
Kiyota N, Namekata K, Nishijima E, Guo X, Kimura A, Harada C, Nakazawa T, Harada T. Effects of constitutively active K-Ras on axon regeneration after optic nerve injury. Neurosci Lett 2023; 799:137124. [PMID: 36780941 DOI: 10.1016/j.neulet.2023.137124] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/24/2023] [Accepted: 02/09/2023] [Indexed: 02/13/2023]
Abstract
Visual disturbance after optic nerve injury is a serious problem. Attempts have been made to enhance the intrinsic ability of retinal ganglion cells (RGCs) to regenerate their axons, and the importance of PI3K/Akt and RAF/MEK/ERK signal activation has been suggested. Since these signals are shared with oncogenic signaling cascades, in this study, we focused on a constitutively active form of K-Ras, K-RasV12, to determine if overexpression of this molecule could stimulate axon regeneration. We confirmed that K-RasV12 phosphorylated Akt and ERK in vitro. Intravitreal delivery of AAV2-K-RasV12 increased the number of surviving RGCs and promoted 1.0 mm of axon regeneration one week after optic nerve injury without inducing abnormal proliferative effects in the RGCs. In addition, AAV2-K-RasV12 induced robust RGC axon regeneration, reaching as far as approximately 2.5 mm from the injury site, in eight weeks. Our findings suggest that AAV2-K-RasV12 could provide a good model for speedy and efficient analysis of the mechanism underlying axon regeneration in vivo.
Collapse
Affiliation(s)
- Naoki Kiyota
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan; Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kazuhiko Namekata
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan.
| | - Euido Nishijima
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Xiaoli Guo
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Atsuko Kimura
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Chikako Harada
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Toru Nakazawa
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takayuki Harada
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| |
Collapse
|
14
|
Díaz A, Flores I, Treviño S. Neurotrophic fragments as therapeutic alternatives to ameliorate brain aging. Neural Regen Res 2023; 18:51-56. [PMID: 35799508 PMCID: PMC9241392 DOI: 10.4103/1673-5374.331867] [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] [Indexed: 12/02/2022] Open
Abstract
Aging is a global phenomenon and a complex biological process of all living beings that introduces various changes. During this physiological process, the brain is the most affected organ due to changes in its structural and chemical functions, such as changes in plasticity and decrease in the number, diameter, length, and branching of dendrites and dendritic spines. Likewise, it presents a great reduction in volume resulting from the contraction of the gray matter. Consequently, aging can affect not only cognitive functions, including learning and memory, but also the quality of life of older people. As a result of the phenomena, various molecules with notable neuroprotective capacity have been proposed, which provide a therapeutic alternative for people under conditions of aging or some neurodegenerative diseases. It is important to indicate that in recent years the use of molecules with neurotrophic activity has shown interesting results when evaluated in in vivo models. This review aims to describe the neurotrophic potential of molecules such as resveratrol (3,5,4′-trihydroxystilbene), neurotrophins (brain-derived neurotrophic factor), and neurotrophic-type compounds such as the terminal carboxyl domain of the heavy chain of tetanus toxin, cerebrolysin, neuropeptide-12, and rapamycin. Most of these molecules have been evaluated by our research group. Studies suggest that these molecules exert an important therapeutic potential, restoring brain function in aging conditions or models of neurodegenerative diseases. Hence, our interest is in describing the current scientific evidence that supports the therapeutic potential of these molecules with active neurotrophic.
Collapse
|
15
|
Ni Q, Zhu B, Ji Y, Zheng Q, Liang X, Ma N, Jiang H, Zhang F, Shang Y, Wang Y, Xu S, Zhang E, Yuan Y, Chen T, Yin F, Cao H, Huang J, Xia J, Ding X, Qiu X, Ding K, Song C, Zhou W, Wu M, Wang K, Lui R, Lin Q, Chen W, Li Z, Cheng S, Wang X, Xie D, Li J. PPDPF Promotes the Development of Mutant KRAS-Driven Pancreatic Ductal Adenocarcinoma by Regulating the GEF Activity of SOS1. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2202448. [PMID: 36453576 PMCID: PMC9839844 DOI: 10.1002/advs.202202448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 10/11/2022] [Indexed: 06/17/2023]
Abstract
The guanine nucleotide exchange factor (GEF) SOS1 catalyzes the exchange of GDP for GTP on RAS. However, regulation of the GEF activity remains elusive. Here, the authors report that PPDPF functions as an important regulator of SOS1. The expression of PPDPF is significantly increased in pancreatic ductal adenocarcinoma (PDAC), associated with poor prognosis and recurrence of PDAC patients. Overexpression of PPDPF promotes PDAC cell growth in vitro and in vivo, while PPDPF knockout exerts opposite effects. Pancreatic-specific deletion of PPDPF profoundly inhibits tumor development in KRASG12D -driven genetic mouse models of PDAC. PPDPF can bind GTP and transfer GTP to SOS1. Mutations of the GTP-binding sites severely impair the tumor-promoting effect of PPDPF. Consistently, mutations of the critical amino acids mediating SOS1-PPDPF interaction significantly impair the GEF activity of SOS1. Therefore, this study demonstrates a novel model of KRAS activation via PPDPF-SOS1 axis, and provides a promising therapeutic target for PDAC.
Collapse
|
16
|
Kenney-Jung DL, Rogers DJ, Kroening SJ, Zatkalik AL, Whitmarsh AE, Roberts AE, Zenker M, Gambardella ML, Contaldo I, Leoni C, Onesimo R, Zampino G, Tartaglia M, Battaglia DI, Pierpont EI. Infantile epileptic spasms syndrome in children with cardiofaciocutanous syndrome: Clinical presentation and associations with genotype. AMERICAN JOURNAL OF MEDICAL GENETICS. PART C, SEMINARS IN MEDICAL GENETICS 2022; 190:501-509. [PMID: 36448195 PMCID: PMC9825647 DOI: 10.1002/ajmg.c.32022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 11/11/2022] [Accepted: 11/21/2022] [Indexed: 12/05/2022]
Abstract
Gene variants that dysregulate signaling through the RAS-MAPK pathway cause cardiofaciocutaneous syndrome (CFCS), a rare multi-system disorder. Infantile epileptic spasms syndrome (IESS) and other forms of epilepsy are among the most serious complications. To investigate clinical presentation, treatment outcomes, and genotype-phenotype associations in CFCS patients with IESS, molecular genetics and clinical neurological history were reviewed across two large clinical research cohorts (n = 180). IESS presented in 18/180 (10%) cases, including 16 patients with BRAF variants and 2 with MAP2K1 variants. Among IESS patients with BRAF variants, 16/16 (100%) had sequence changes affecting the protein kinase domain (exons 11-16), although only 57% of total BRAF variants occurred in this domain. Clinical onset of spasms occurred at a median age of 5.4 months (range: 1-24 months). Among 13/18 patients whose IESS resolved with anti-seizure medications, 10 were treated with ACTH and/or vigabatrin. A substantial majority of CFCS patients with IESS subsequently developed other epilepsy types (16/18; 89%). In terms of neurodevelopmental outcomes, gross motor function and verbal communication were more limited in patients with a history of IESS compared to those without IESS. These findings can inform clinical neurological care guidelines for CFCS and development of relevant pre-clinical models for severe epilepsy phenotypes.
Collapse
Affiliation(s)
| | - Dante J. Rogers
- Department of Pediatrics, University of Minnesota Medical School
| | | | | | | | - Amy E. Roberts
- Department of Cardiology and Department of Pediatrics, Division of Genetics and Genomics, Boston Children’s Hospital
| | - Martin Zenker
- Institute of Human Genetics, University Hospital, Otto-von-Guericke University Magdeburg, Germany Magdeburg
| | - Maria Luigia Gambardella
- Pediatric Neuropsychiatry, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Ilaria Contaldo
- Pediatric Neuropsychiatry, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Chiara Leoni
- Center for Rare Disease and Congenital Defects, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, 00168 Rome, Italy
| | - Roberta Onesimo
- Center for Rare Disease and Congenital Defects, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, 00168 Rome, Italy
| | - Giuseppe Zampino
- Center for Rare Disease and Congenital Defects, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, 00168 Rome, Italy
| | - Marco Tartaglia
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | - Domenica I. Battaglia
- Pediatric Neuropsychiatry, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy,Dipartimento Scienze della Vita, Università Cattolica del Sacro Cuore, Rome, Italy
| | | |
Collapse
|
17
|
Pyrido[2,3-d]pyrimidine-2,4(1H,3H)-dione derivatives as RAF-MEK-ERK pathway signaling pathway blockers: Synthesis, cytotoxic activity, mechanistic investigation and structure-activity relationships. Eur J Med Chem 2022; 240:114579. [DOI: 10.1016/j.ejmech.2022.114579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 06/26/2022] [Accepted: 06/27/2022] [Indexed: 11/21/2022]
|
18
|
Neurologic and neurodevelopmental complications in cardiofaciocutaneous syndrome are associated with genotype: A multinational cohort study. Genet Med 2022; 24:1556-1566. [PMID: 35524774 DOI: 10.1016/j.gim.2022.04.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 03/30/2022] [Accepted: 04/01/2022] [Indexed: 01/07/2023] Open
Abstract
PURPOSE Dysregulation of RAS or its major effector pathway is the molecular mechanism of RASopathies, a group of multisystemic congenital disorders. Neurologic complications are especially challenging in the management of the rare RASopathy cardiofaciocutaneous (CFC) syndrome. This study evaluated clinical neurologic and neurodevelopmental features and their associations with CFC syndrome gene variants. METHODS A multinational cohort of 138 individuals with CFC syndrome (BRAF = 90, MAP2K1 = 36, MAP2K2 = 10, KRAS = 2) was recruited. Neurologic presentation was captured via clinician review of medical records and caregiver-completed electronic surveys. Validated measures of seizure severity, adaptive function, and gross motor function were obtained. RESULTS The overall frequency of intellectual disability and seizures was 82% and 55%, respectively. The frequency and severity of seizures was higher among individuals with BRAF or MAP2K1 variants than in those with MAP2K2 variants. A disproportionate incidence of severe, treatment-resistant seizures was observed in patients with variants in the catalytic protein kinase domain of BRAF and at the common p.Y130 site of MAP2K1. Neurodevelopmental outcomes were associated with genotype as well as seizure severity. CONCLUSION Molecular genetic testing can aid in prediction of epilepsy and neurodevelopmental phenotypes in CFC syndrome. Study results identified potential CFC syndrome-associated variants in the development of relevant animal models for neurologic, neurocognitive, and motor function impairment.
Collapse
|
19
|
Tan MH, Xu XH, Yuan TJ, Hou X, Wang J, Jiang ZH, Peng LH. Self-powered smart patch promotes skin nerve regeneration and sensation restoration by delivering biological-electrical signals in program. Biomaterials 2022; 283:121413. [DOI: 10.1016/j.biomaterials.2022.121413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 02/09/2022] [Accepted: 02/13/2022] [Indexed: 11/02/2022]
|
20
|
Adachi H, Morizane A, Torikoshi S, Raudzus F, Taniguchi Y, Miyamoto S, Sekiguchi K, Takahashi J. OUP accepted manuscript. Stem Cells Transl Med 2022; 11:767-777. [PMID: 35605097 PMCID: PMC9299512 DOI: 10.1093/stcltm/szac033] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 04/18/2022] [Indexed: 11/12/2022] Open
Affiliation(s)
- Hiromasa Adachi
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Asuka Morizane
- Corresponding authors: Asuka Morizane, MD, PhD, Kobe City Medical Center General Hospital, Center for Clinical Research and Innovation, 2-1-1, Minatojima-Minamimachi, Chuo-ku, Kobe, Hyogo 650 0046, Japan, Tel: +81 78 302 4321; Fax: +81 78 302 7537;
| | - Sadaharu Torikoshi
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Fabian Raudzus
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
- Neuronal Signaling and Regeneration Unit, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Medical Education Center/International Education Section, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | | | - Susumu Miyamoto
- Kobe City Medical Center General Hospital, Center for Clinical Research and Innovation, Hyogo, Japan
| | - Kiyotoshi Sekiguchi
- Kiyotoshi Sekiguchi, PhD (for chimeric laminin fragments), Division of Matrixome Research and Application, Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka 565-0871, Japan. Tel: +81 6 6105 5935; Fax: +81 6 6105 5935; Email;
| | - Jun Takahashi
- Jun Takahashi, MD, PhD, Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan. Tel: +81 75 366 7052; Fax: +81 75 366 7071;
| |
Collapse
|
21
|
Liu X, Song L, Ma X, Liu Y, Huang H, Xu Y, Yan W. Overexpression of RAPGEF3 enhances the therapeutic effect of dezocine in treatment of neuropathic pain. Genet Mol Biol 2021; 44:e20200463. [PMID: 34807222 PMCID: PMC8607529 DOI: 10.1590/1678-4685-gmb-2020-0463] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 08/05/2021] [Indexed: 12/20/2022] Open
Abstract
Pain is a significant problem worldwide that affects the quality of life of patients. Dezocine is a non-addictive analgesic drug with kappa-opioid antagonist activity and has been successfully used to alleviate of postoperative pain. In addition, dezocine has an analgesic effect similar to that of morphine, alleviating moderate to severe pain. Rap guanine nucleotide exchange factor 3 (RAPGEF3) is a guanine nucleotide exchange factor for GTPases Rap1 and Rap2, which could enhance the activity of Rap1 to promote cell adhesion and axon regeneration, as well as promote neurite extension by interacting with nerve growth factors. Here, we first observed that overexpression of RAPGEF3 increased cell viability, as shown by a CCK-8 assay, and recovered brain function in rats. The expression of inflammation-related factors at the mRNA level was detected using qPCR, and the concentration of these factors in a cultured cell medium and rat serum samples were decreased as shown by ELISA after RAPGEF3 overexpression. Through western blotting, we further found that pro-inflammatory proteins were decreased, and these effects might be mediated by inhibition of the Ras/p-38 MAPK signaling pathway. Taken together, we speculated that RAPGEF3overexpression enhances the therapeutic effect of dezocine on neuropathic pain by inhibiting the inflammatory response through inhibition of the Ras/p-38 MAPK signaling pathway.
Collapse
Affiliation(s)
- Xue Liu
- The Affiliated Hospital of Qingdao University, Department of Anesthesiology, Qingdao, Shandong, China
| | - Li Song
- The Affiliated Hospital of Qingdao University, Department of Anesthesiology, Qingdao, Shandong, China
| | - Xiaojun Ma
- The Affiliated Hospital of Qingdao University, Department of Anesthesiology, Qingdao, Shandong, China
| | - Yong Liu
- The Affiliated Hospital of Qingdao University, Department of Anesthesiology, Qingdao, Shandong, China
| | - Hui Huang
- The Affiliated Hospital of Qingdao University, Department of Anesthesiology, Qingdao, Shandong, China
| | - Yongsheng Xu
- The Affiliated Hospital of Qingdao University, Department of Anesthesiology, Qingdao, Shandong, China
| | - Wei Yan
- The Affiliated Hospital of Qingdao University, Department of Anesthesiology, Qingdao, Shandong, China
| |
Collapse
|
22
|
Niu F, Sharma A, Wang Z, Feng L, Muresanu DF, Sahib S, Tian ZR, Lafuente JV, Buzoianu AD, Castellani RJ, Nozari A, Menon PK, Patnaik R, Wiklund L, Sharma HS. Nanodelivery of oxiracetam enhances memory, functional recovery and induces neuroprotection following concussive head injury. PROGRESS IN BRAIN RESEARCH 2021; 265:139-230. [PMID: 34560921 DOI: 10.1016/bs.pbr.2021.06.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Military personnel are the most susceptible to concussive head injury (CHI) caused by explosion, blast or missile or blunt head trauma. Mild to moderate CHI could induce lifetime functional and cognitive disturbances causing significant decrease in quality of life. Severe CHI leads to instant death and lifetime paralysis. Thus, further exploration of novel therapeutic agents or new features of known pharmacological agents are needed to enhance quality of life of CHI victims. Previous reports from our laboratory showed that mild CHI induced by weight drop technique causing an impact of 0.224N results in profound progressive functional deficit, memory impairment and brain pathology from 5h after trauma that continued over several weeks of injury. In this investigation we report that TiO2 nanowired delivery of oxiracetam (50mg/kg, i.p.) daily for 5 days after CHI resulted in significant improvement of functional deficit on the 8th day. This was observed using Rota Rod treadmill, memory improvement assessed by the time spent in finding hidden platform under water. The motor function improvement is seen in oxiracetam treated CHI group by placing forepaw on an inclined mesh walking and foot print analysis for stride length and distance between hind feet. TiO2-nanowired oxiracetam also induced marked improvements in the cerebral blood flow, reduction in the BBB breakdown and edema formation as well as neuroprotection of neuronal, glial and myelin damages caused by CHI at light and electron microscopy on the 7th day after 5 days TiO2 oxiracetam treatment. Adverse biochemical events such as upregulation of CSF nitrite and nitrate, IL-6, TNF-a and p-Tau are also reduced significantly in oxiracetam treated CHI group. On the other hand post treatment of 100mg/kg dose of normal oxiracetam in identical conditions after CHI is needed to show slight but significant neuroprotection together with mild recovery of memory function and functional deficits on the 8th day. These observations are the first to point out that nanowired delivery of oxiracetam has superior neuroprotective ability in CHI. These results indicate a promising clinical future of TiO2 oxiracetam in treating CHI patients for better quality of life and neurorehabilitation, not reported earlier.
Collapse
Affiliation(s)
- Feng Niu
- CSPC NBP Pharmaceutical Medicine, Shijiazhuang, China
| | - Aruna Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| | - Zhenguo Wang
- CSPC NBP Pharmaceutical Medicine, Shijiazhuang, China
| | - Lianyuan Feng
- Department of Neurology, Bethune International Peace Hospital, Shijiazhuang, China
| | - Dafin F Muresanu
- Department of Clinical Neurosciences, University of Medicine & Pharmacy, Cluj-Napoca, Romania; "RoNeuro" Institute for Neurological Research and Diagnostic, Cluj-Napoca, Romania
| | - Seaab Sahib
- Department of Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, United States
| | - Z Ryan Tian
- Department of Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, United States
| | - José Vicente Lafuente
- LaNCE, Department of Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| | - Anca D Buzoianu
- Department of Clinical Pharmacology and Toxicology, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Rudy J Castellani
- Department of Pathology, University of Maryland, Baltimore, MD, United States
| | - Ala Nozari
- Anesthesiology & Intensive Care, Massachusetts General Hospital, Boston, MA, United States
| | - Preeti K Menon
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Ranjana Patnaik
- Department of Biomaterials, School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University, Varanasi, India
| | - Lars Wiklund
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - Hari Shanker Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
23
|
Liu KX, Sethi RV, Pulsifer MB, D'Gama AM, LaVally B, Ebb DH, Tarbell NJ, Yock TI, MacDonald SM. Clinical outcomes of pediatric patients with autism spectrum disorder and other neurodevelopmental disorders and intracranial germ cell tumors. Pediatr Blood Cancer 2021; 68:e28935. [PMID: 33694260 DOI: 10.1002/pbc.28935] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 01/11/2021] [Accepted: 01/18/2021] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Intracranial germ cell tumors (IGCTs) are rare tumors of the central nervous system with peak incidence around puberty. Given the developmental origins of IGCTs, we investigated the prevalence of neurodevelopmental disorders (NDDs) in patients with IGCTs and characterized outcomes for patients with NDD and IGCTs. METHODS A retrospective review of medical records was conducted for 111 patients diagnosed with IGCTs between 1998 and 2018 and evaluated at the Massachusetts General Hospital. Kaplan-Meier method and log-rank test was used for survival analyses. Cox regression analyses were performed for parameters associated with progression-free survival (PFS). RESULTS Median age at IGCT diagnosis was 12.8 years (range: 4.3-21.7) and median follow-up was 6.5 years (range: 0.2-20.5). Eighteen patients were diagnosed with NDDs prior to IGCT diagnosis, including five patients with autism spectrum disorder (ASD). Of the 67 patients with pure germinomas, four (6.0 %) had prior ASD diagnoses. Patients with NDD had significantly inferior PFS in the nongerminomatous germ cell tumor (NGGCT) cohort. On univariate and multivariable analyses, craniospinal irradiation (CSI) was significantly associated with improved PFS in the NGGCT cohort. CONCLUSIONS Our study found an ASD prevalence in the pure germinoma cohort more than threefold greater than the national prevalence, suggesting an association between ASD and pure germinomas. Furthermore, patients with NDD and NGGCT had worse PFS, possibly due to fewer patients with NDD receiving CSI. Future prospective studies with larger cohorts are needed to examine associations between NDDs and IGCTs, and further characterize outcomes for patients with NDDs and IGCTs.
Collapse
Affiliation(s)
- Kevin X Liu
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Roshan V Sethi
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Margaret B Pulsifer
- Department of Psychiatry, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Alissa M D'Gama
- Division of Newborn Medicine, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Beverly LaVally
- Department of Pediatric Oncology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - David H Ebb
- Department of Pediatric Oncology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Nancy J Tarbell
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Torunn I Yock
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Shannon M MacDonald
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
24
|
MAPK and Notch-Mediated Effects of Meso-Xanthin F199 Compounds on Proliferative Activity and Apoptosis of Human Melanocytes in Three-Dimensional Culture. BIOMED RESEARCH INTERNATIONAL 2021; 2021:8463161. [PMID: 34337053 PMCID: PMC8315846 DOI: 10.1155/2021/8463161] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 06/08/2021] [Accepted: 07/06/2021] [Indexed: 11/18/2022]
Abstract
Meso-Xanthin (Meso-Xanthin F199™) is a highly active antiaging injection drug of the latest generation. The main acting compound is fucoxanthin, supplemented with several growth factors, vitamins, and hyaluronic acid. Previous examination of fucoxanthin on melanocytes showed its ability to inhibit skin pigmentation through different signaling pathways focused on suppression of melanogenic-stimulating receptors. In turn, the anticancer property of fucoxanthin is realized through MAPK and PI3K pathways. We aimed to evaluate the effect of fucoxanthin and supplemented growth factors on melanocyte growth and transformation at a proteomic level. The effect of fucoxanthin on melanocytes cultivated in three-dimensional (3D) condition was examined using high-throughput proteomic and system biology approaches to disclose key molecular events of the targeted action. Our results demonstrated significant inhibition of cell differentiation and ubiquitination processes. We found that the negative regulation of PSME1 and PTGIS largely determines the inhibition of NF-κB and MAPK2. Besides, fucoxanthin selectively inhibits cell differentiation via negative regulation of Raf signaling and the upstream activation of IL-1 signaling. It is assumed that inhibition of Raf influences the Notch-4 signaling and switches off the MAPK/MAPK2 cascade. Blockage of MAPK/MAPK2 is feasible due to suppression of Ras and NF-κB by the addressed action of IKKB, IKK2, and TRAF6. Suggestively, Meso-Xanthin F199™ can manage processes of proliferative activity and inhibition of apoptosis due to composition of fucoxanthin and growth-stimulating factors, which may increase the risk of skin cancer development under certain condition.
Collapse
|
25
|
Binamé F, Pham-Van LD, Bagnard D. Manipulating oligodendrocyte intrinsic regeneration mechanism to promote remyelination. Cell Mol Life Sci 2021; 78:5257-5273. [PMID: 34019104 PMCID: PMC11073109 DOI: 10.1007/s00018-021-03852-4] [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: 02/03/2021] [Revised: 04/14/2021] [Accepted: 05/08/2021] [Indexed: 02/06/2023]
Abstract
In demyelinated lesions, astrocytes, activated microglia and infiltrating macrophages secrete several factors regulating oligodendrocyte precursor cells' behaviour. What appears to be the initiation of an intrinsic mechanism of myelin repair is only leading to partial recovery and inefficient remyelination, a process worsening over the course of the disease. This failure is largely due to the concomitant accumulation of inhibitory cues in and around the lesion sites opposing to growth promoting factors. Here starts a complex game of interactions between the signalling pathways controlling oligodendrocytes migration or differentiation. Receptors of positive or negative cues are modulating Ras, PI3K or RhoGTPases pathways acting on oligodendrocyte cytoskeleton remodelling. From the description of this intricate signalling network, this review addresses the extent to which the modulation of the global response to inhibitory cues may pave the route towards novel therapeutic approaches for myelin repair.
Collapse
Affiliation(s)
- Fabien Binamé
- INSERM U1119, Biopathology of Myelin, Neuroprotection and Therapeutic Strategy (BMNST Lab), Labex Medalis, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Pôle API, Ecole Supérieure de Biotechnologie, 300 Boulevard Sébastien Brant, 67412, Illkirch, France
| | - Lucas D Pham-Van
- INSERM U1119, Biopathology of Myelin, Neuroprotection and Therapeutic Strategy (BMNST Lab), Labex Medalis, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Pôle API, Ecole Supérieure de Biotechnologie, 300 Boulevard Sébastien Brant, 67412, Illkirch, France
| | - Dominique Bagnard
- INSERM U1119, Biopathology of Myelin, Neuroprotection and Therapeutic Strategy (BMNST Lab), Labex Medalis, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Pôle API, Ecole Supérieure de Biotechnologie, 300 Boulevard Sébastien Brant, 67412, Illkirch, France.
| |
Collapse
|
26
|
Lindborg JA, Tran NM, Chenette DM, DeLuca K, Foli Y, Kannan R, Sekine Y, Wang X, Wollan M, Kim IJ, Sanes JR, Strittmatter SM. Optic nerve regeneration screen identifies multiple genes restricting adult neural repair. Cell Rep 2021; 34:108777. [PMID: 33657370 PMCID: PMC8009559 DOI: 10.1016/j.celrep.2021.108777] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 12/21/2020] [Accepted: 01/29/2021] [Indexed: 12/22/2022] Open
Abstract
Adult mammalian central nervous system (CNS) trauma interrupts neural networks and, because axonal regeneration is minimal, neurological deficits persist. Repair via axonal growth is limited by extracellular inhibitors and cell-autonomous factors. Based on results from a screen in vitro, we evaluate nearly 400 genes through a large-scale in vivo regeneration screen. Suppression of 40 genes using viral-driven short hairpin RNAs (shRNAs) promotes retinal ganglion cell (RGC) axon regeneration after optic nerve crush (ONC), and most are validated by separate CRISPR-Cas9 editing experiments. Expression of these axon-regeneration-suppressing genes is not significantly altered by axotomy. Among regeneration-limiting genes, loss of the interleukin 22 (IL-22) cytokine allows an early, yet transient, inflammatory response in the retina after injury. Reduced IL-22 drives concurrent activation of signal transducer and activator of transcription 3 (Stat3) and dual leucine zipper kinase (DLK) pathways and upregulation of multiple neuron-intrinsic regeneration-associated genes (RAGs). Including IL-22, our screen identifies dozens of genes that limit CNS regeneration. Suppression of these genes in the context of axonal damage could support improved neural repair.
Collapse
Affiliation(s)
- Jane A Lindborg
- Cellular Neuroscience, Neurodegeneration, Repair, Departments of Neurology and of Neuroscience, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Nicholas M Tran
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA
| | - Devon M Chenette
- Cellular Neuroscience, Neurodegeneration, Repair, Departments of Neurology and of Neuroscience, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Kristin DeLuca
- Cellular Neuroscience, Neurodegeneration, Repair, Departments of Neurology and of Neuroscience, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Yram Foli
- Cellular Neuroscience, Neurodegeneration, Repair, Departments of Neurology and of Neuroscience, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Ramakrishnan Kannan
- Cellular Neuroscience, Neurodegeneration, Repair, Departments of Neurology and of Neuroscience, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Yuichi Sekine
- Cellular Neuroscience, Neurodegeneration, Repair, Departments of Neurology and of Neuroscience, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Xingxing Wang
- Cellular Neuroscience, Neurodegeneration, Repair, Departments of Neurology and of Neuroscience, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Marius Wollan
- Cellular Neuroscience, Neurodegeneration, Repair, Departments of Neurology and of Neuroscience, Yale University School of Medicine, New Haven, CT 06536, USA
| | - In-Jung Kim
- Department of Ophthalmology and Visual Science, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Joshua R Sanes
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA
| | - Stephen M Strittmatter
- Cellular Neuroscience, Neurodegeneration, Repair, Departments of Neurology and of Neuroscience, Yale University School of Medicine, New Haven, CT 06536, USA.
| |
Collapse
|
27
|
Fadeev FO, Bashirov FV, Markosyan VA, Izmailov AA, Povysheva TV, Sokolov ME, Kuznetsov MS, Eremeev AA, Salafutdinov II, Rizvanov AA, Lee HJ, Islamov RR. Combination of epidural electrical stimulation with ex vivo triple gene therapy for spinal cord injury: a proof of principle study. Neural Regen Res 2021; 16:550-560. [PMID: 32985487 PMCID: PMC7996027 DOI: 10.4103/1673-5374.293150] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 11/03/2019] [Accepted: 04/29/2020] [Indexed: 02/07/2023] Open
Abstract
Despite emerging contemporary biotechnological methods such as gene- and stem cell-based therapy, there are no clinically established therapeutic strategies for neural regeneration after spinal cord injury. Our previous studies have demonstrated that transplantation of genetically engineered human umbilical cord blood mononuclear cells producing three recombinant therapeutic molecules, including vascular endothelial growth factor (VEGF), glial cell-line derived neurotrophic factor (GDNF), and neural cell adhesion molecule (NCAM) can improve morpho-functional recovery of injured spinal cord in rats and mini-pigs. To investigate the efficacy of human umbilical cord blood mononuclear cells-mediated triple-gene therapy combined with epidural electrical stimulation in the treatment of spinal cord injury, in this study, rats with moderate spinal cord contusion injury were intrathecally infused with human umbilical cord blood mononuclear cells expressing recombinant genes VEGF165, GDNF, NCAM1 at 4 hours after spinal cord injury. Three days after injury, epidural stimulations were given simultaneously above the lesion site at C5 (to stimulate the cervical network related to forelimb functions) and below the lesion site at L2 (to activate the central pattern generators) every other day for 4 weeks. Rats subjected to the combined treatment showed a limited functional improvement of the knee joint, high preservation of muscle fiber area in tibialis anterior muscle and increased H/M ratio in gastrocnemius muscle 30 days after spinal cord injury. However, beneficial cellular outcomes such as reduced apoptosis and increased sparing of the gray and white matters, and enhanced expression of heat shock and synaptic proteins were found in rats with spinal cord injury subjected to the combined epidural electrical stimulation with gene therapy. This study presents the first proof of principle study of combination of the multisite epidural electrical stimulation with ex vivo triple gene therapy (VEGF, GDNF and NCAM) for treatment of spinal cord injury in rat models. The animal protocols were approved by the Kazan State Medical University Animal Care and Use Committee (approval No. 2.20.02.18) on February 20, 2018.
Collapse
Affiliation(s)
- Filip Olegovich Fadeev
- Department of Medical Biology and Genetics, Kazan State Medical University, Kazan, Russia
| | | | | | | | | | | | | | | | | | | | - Hyun Joon Lee
- Department of Neurology, University of Mississippi Medical Center, Jackson, MS, USA
- Research Service, G.V. (Sonny) Montgomery VA Medical Center, Jackson, MS, USA
| | | |
Collapse
|
28
|
Using antisense oligonucleotides for the physiological modulation of the alternative splicing of NF1 exon 23a during PC12 neuronal differentiation. Sci Rep 2021; 11:3661. [PMID: 33574490 PMCID: PMC7878752 DOI: 10.1038/s41598-021-83152-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 01/29/2021] [Indexed: 01/11/2023] Open
Abstract
Neurofibromatosis Type 1 (NF1) is a genetic condition affecting approximately 1:3500 persons worldwide. The NF1 gene codes for neurofibromin protein, a GTPase activating protein (GAP) and a negative regulator of RAS. The NF1 gene undergoes alternative splicing of exon 23a (E23a) that codes for 21 amino acids placed at the center of the GAP related domain (GRD). E23a-containing type II neurofibromin exhibits a weaker Ras-GAP activity compared to E23a-less type I isoform. Exon E23a has been related with the cognitive impairment present in NF1 individuals. We designed antisense Phosphorodiamidate Morpholino Oligomers (PMOs) to modulate E23a alternative splicing at physiological conditions of gene expression and tested their impact during PC12 cell line neuronal differentiation. Results show that any dynamic modification of the natural ratio between type I and type II isoforms disturbed neuronal differentiation, altering the proper formation of neurites and deregulating both the MAPK/ERK and cAMP/PKA signaling pathways. Our results suggest an opposite regulation of these pathways by neurofibromin and the possible existence of a feedback loop sensing neurofibromin-related signaling. The present work illustrates the utility of PMOs to study alternative splicing that could be applied to other alternatively spliced genes in vitro and in vivo.
Collapse
|
29
|
Delhaye S, Bardoni B. Role of phosphodiesterases in the pathophysiology of neurodevelopmental disorders. Mol Psychiatry 2021; 26:4570-4582. [PMID: 33414502 PMCID: PMC8589663 DOI: 10.1038/s41380-020-00997-9] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 12/03/2020] [Accepted: 12/09/2020] [Indexed: 12/12/2022]
Abstract
Phosphodiesterases (PDEs) are enzymes involved in the homeostasis of both cAMP and cGMP. They are members of a family of proteins that includes 11 subfamilies with different substrate specificities. Their main function is to catalyze the hydrolysis of cAMP, cGMP, or both. cAMP and cGMP are two key second messengers that modulate a wide array of intracellular processes and neurobehavioral functions, including memory and cognition. Even if these enzymes are present in all tissues, we focused on those PDEs that are expressed in the brain. We took into consideration genetic variants in patients affected by neurodevelopmental disorders, phenotypes of animal models, and pharmacological effects of PDE inhibitors, a class of drugs in rapid evolution and increasing application to brain disorders. Collectively, these data indicate the potential of PDE modulators to treat neurodevelopmental diseases characterized by learning and memory impairment, alteration of behaviors associated with depression, and deficits in social interaction. Indeed, clinical trials are in progress to treat patients with Alzheimer's disease, schizophrenia, depression, and autism spectrum disorders. Among the most recent results, the application of some PDE inhibitors (PDE2A, PDE3, PDE4/4D, and PDE10A) to treat neurodevelopmental diseases, including autism spectrum disorders and intellectual disability, is a significant advance, since no specific therapies are available for these disorders that have a large prevalence. In addition, to highlight the role of several PDEs in normal and pathological neurodevelopment, we focused here on the deregulation of cAMP and/or cGMP in Down Syndrome, Fragile X Syndrome, Rett Syndrome, and intellectual disability associated with the CC2D1A gene.
Collapse
Affiliation(s)
- Sébastien Delhaye
- grid.429194.30000 0004 0638 0649Université Côte d’Azur, CNRS UMR7275, Institute of Molecular and Cellular Pharmacology, 06560 Valbonne, France
| | - Barbara Bardoni
- Université Côte d'Azur, Inserm, CNRS UMR7275, Institute of Molecular and Cellular Pharmacology, 06560, Valbonne, France.
| |
Collapse
|
30
|
Toricelli M, Pereira AAR, Souza Abrao G, Malerba HN, Maia J, Buck HS, Viel TA. Mechanisms of neuroplasticity and brain degeneration: strategies for protection during the aging process. Neural Regen Res 2021; 16:58-67. [PMID: 32788448 PMCID: PMC7818866 DOI: 10.4103/1673-5374.286952] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Aging is a dynamic and progressive process that begins at conception and continues until death. This process leads to a decrease in homeostasis and morphological, biochemical and psychological changes, increasing the individual’s vulnerability to various diseases. The growth in the number of aging populations has increased the prevalence of chronic degenerative diseases, impairment of the central nervous system and dementias, such as Alzheimer’s disease, whose main risk factor is age, leading to an increase of the number of individuals who need daily support for life activities. Some theories about aging suggest it is caused by an increase of cellular senescence and reactive oxygen species, which leads to inflammation, oxidation, cell membrane damage and consequently neuronal death. Also, mitochondrial mutations, which are generated throughout the aging process, can lead to changes in energy production, deficiencies in electron transport and apoptosis induction that can result in decreased function. Additionally, increasing cellular senescence and the release of proinflammatory cytokines can cause irreversible damage to neuronal cells. Recent reports point to the importance of changing lifestyle by increasing physical exercise, improving nutrition and environmental enrichment to activate neuroprotective defense mechanisms. Therefore, this review aims to address the latest information about the different mechanisms related to neuroplasticity and neuronal death and to provide strategies that can improve neuroprotection and decrease the neurodegeneration caused by aging and environmental stressors.
Collapse
Affiliation(s)
- Mariana Toricelli
- Department of Physiological Sciences, Santa Casa de Sao Paulo School of Medical Sciences, Sao Paulo, Brazil
| | - Arthur Antonio Ruiz Pereira
- Laboratory of Neurobiology of Aging, School of Arts, Sciences and Humanities, Universidade de São Paulo, Sao Paulo, Brazil
| | - Guilherme Souza Abrao
- Laboratory of Neurobiology of Aging, School of Arts, Sciences and Humanities, Universidade de São Paulo, Sao Paulo, Brazil
| | - Helena Nascimento Malerba
- Laboratory of Neurobiology of Aging, School of Arts, Sciences and Humanities, Universidade de São Paulo, Sao Paulo, Brazil
| | - Julia Maia
- Laboratory of Neurobiology of Aging, School of Arts, Sciences and Humanities, Universidade de São Paulo, Sao Paulo, Brazil
| | - Hudson Sousa Buck
- Department of Physiological Sciences, Santa Casa de Sao Paulo School of Medical Sciences, Sao Paulo, Brazil
| | - Tania Araujo Viel
- Laboratory of Neurobiology of Aging, School of Arts, Sciences and Humanities, Universidade de São Paulo, Sao Paulo, Brazil
| |
Collapse
|
31
|
King LB, Boto T, Botero V, Aviles AM, Jomsky BM, Joseph C, Walker JA, Tomchik SM. Developmental loss of neurofibromin across distributed neuronal circuits drives excessive grooming in Drosophila. PLoS Genet 2020; 16:e1008920. [PMID: 32697780 PMCID: PMC7398555 DOI: 10.1371/journal.pgen.1008920] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 08/03/2020] [Accepted: 06/08/2020] [Indexed: 02/06/2023] Open
Abstract
Neurofibromatosis type 1 is a monogenetic disorder that predisposes individuals to tumor formation and cognitive and behavioral symptoms. The neuronal circuitry and developmental events underlying these neurological symptoms are unknown. To better understand how mutations of the underlying gene (NF1) drive behavioral alterations, we have examined grooming in the Drosophila neurofibromatosis 1 model. Mutations of the fly NF1 ortholog drive excessive grooming, and increased grooming was observed in adults when Nf1 was knocked down during development. Furthermore, intact Nf1 Ras GAP-related domain signaling was required to maintain normal grooming. The requirement for Nf1 was distributed across neuronal circuits, which were additive when targeted in parallel, rather than mapping to discrete microcircuits. Overall, these data suggest that broadly-distributed alterations in neuronal function during development, requiring intact Ras signaling, drive key Nf1-mediated behavioral alterations. Thus, global developmental alterations in brain circuits/systems function may contribute to behavioral phenotypes in neurofibromatosis type 1.
Collapse
Affiliation(s)
- Lanikea B. King
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida, United States of America
| | - Tamara Boto
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida, United States of America
| | - Valentina Botero
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida, United States of America
| | - Ari M. Aviles
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida, United States of America
- Honors College, Florida Atlantic University, Jupiter, Florida, United States of America
| | - Breanna M. Jomsky
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida, United States of America
- Honors College, Florida Atlantic University, Jupiter, Florida, United States of America
| | - Chevara Joseph
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida, United States of America
- Honors College, Florida Atlantic University, Jupiter, Florida, United States of America
| | - James A. Walker
- Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Cambridge, Massachusetts, United States of America
| | - Seth M. Tomchik
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida, United States of America
| |
Collapse
|
32
|
Oh M, Kim SY, Gil JE, Byun JS, Cha DW, Ku B, Lee W, Kim WK, Oh KJ, Lee EW, Bae KH, Lee SC, Han BS. Nurr1 performs its anti-inflammatory function by regulating RasGRP1 expression in neuro-inflammation. Sci Rep 2020; 10:10755. [PMID: 32612143 PMCID: PMC7329810 DOI: 10.1038/s41598-020-67549-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 06/10/2020] [Indexed: 12/21/2022] Open
Abstract
Nurr1, a transcription factor belonging to the orphan nuclear receptor, has an essential role in the generation and maintenance of dopaminergic neurons and is important in the pathogenesis of Parkinson’ disease (PD). In addition, Nurr1 has a non-neuronal function, and it is especially well known that Nurr1 has an anti-inflammatory function in the Parkinson’s disease model. However, the molecular mechanisms of Nurr1 have not been elucidated. In this study, we describe a novel mechanism of Nurr1 function. To provide new insights into the molecular mechanisms of Nurr1 in the inflammatory response, we performed Chromatin immunoprecipitation sequencing (ChIP-Seq) on LPS-induced inflammation in BV2 cells and finally identified the RasGRP1 gene as a novel target of Nurr1. Here, we show that Nurr1 directly binds to the RasGRP1 intron to regulate its expression. Moreover, we also identified that RasGRP1 regulates the Ras-Raf-MEK-ERK signaling cascade in LPS-induced inflammation signaling. Finally, we conclude that RasGRP1 is a novel regulator of Nurr1’s mediated inflammation signaling.
Collapse
Affiliation(s)
- Mihee Oh
- Biodefense Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea
| | - Sun Young Kim
- Biodefense Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea
| | - Jung-Eun Gil
- Biodefense Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea
| | - Jeong-Su Byun
- Biodefense Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea
| | - Dong-Wook Cha
- Biodefense Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea.,Department of Functional Genomics, University of Science and Technology (UST) of Korea, Daejeon, 34113, Republic of Korea
| | - Bonsu Ku
- Disease Target Structure Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea
| | | | - Won-Kon Kim
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea.,Department of Functional Genomics, University of Science and Technology (UST) of Korea, Daejeon, 34113, Republic of Korea
| | - Kyoung-Jin Oh
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea.,Department of Functional Genomics, University of Science and Technology (UST) of Korea, Daejeon, 34113, Republic of Korea
| | - Eun-Woo Lee
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea
| | - Kwang-Hee Bae
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea.,Department of Functional Genomics, University of Science and Technology (UST) of Korea, Daejeon, 34113, Republic of Korea
| | - Sang Chul Lee
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea. .,Department of Functional Genomics, University of Science and Technology (UST) of Korea, Daejeon, 34113, Republic of Korea.
| | - Baek-Soo Han
- Biodefense Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea. .,Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea. .,Department of Functional Genomics, University of Science and Technology (UST) of Korea, Daejeon, 34113, Republic of Korea.
| |
Collapse
|
33
|
Accogli A, Scala M, Pavanello M, Severino M, Gandolfo C, De Marco P, Musacchia F, Torella A, Pinelli M, Nigro V, Capra V. Sinus pericranii, skull defects, and structural brain anomalies in TRAF7-related disorder. Birth Defects Res 2020; 112:1085-1092. [PMID: 32459067 DOI: 10.1002/bdr2.1711] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 04/25/2020] [Indexed: 12/23/2022]
Abstract
BACKGROUND Several somatic mutations in TRAF7 have been reported in cancers, whereas a few germline heterozygous mutations have been recently linked to a neurodevelopmental disorder, characterized by craniofacial dysmorphisms, congenital heart defects, and digital anomalies. CASES We report two subjects harboring de novo heterozygous missense variants in TRAF7, namely the recurrent 1964G>A(p.Arg655Gln) and the novel missense c.1204C>G(p.Leu402Val) variants. In addition to the typical hallmarks of the TRAF7-related disorder, both subjects presented with a recognizable "pear-shaped" skull due to multiple craniosynostosis, sinus pericranii, skull base/cranio-cervical junction anomalies, dysgyria, and inferior cerebellar vermis hypoplasia. CONCLUSIONS Hence, we expand the genotypic and phenotypic spectrum of this neurodevelopmental disorder, discussing possible implications for clinical management of subjects with germline TRAF7 mutations.
Collapse
Affiliation(s)
- Andrea Accogli
- Unit of Medical Genetics, IRCCS Istituto Giannina Gaslini, Genoa, Italy.,Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
| | - Marcello Scala
- Unit of Medical Genetics, IRCCS Istituto Giannina Gaslini, Genoa, Italy.,Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
| | - Marco Pavanello
- UOC Neurosurgery, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | | | - Carlo Gandolfo
- Neuroradiology Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Patrizia De Marco
- Unit of Medical Genetics, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | | | - Annalaura Torella
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy.,Medical Genetics, Department of Precision Medicine, Università degli Studi della Campania 'Luigi Vanvitelli', Naples, Italy
| | - Michele Pinelli
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy
| | - Vincenzo Nigro
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy.,Medical Genetics, Department of Precision Medicine, Università degli Studi della Campania 'Luigi Vanvitelli', Naples, Italy
| | - Valeria Capra
- UOC Neurosurgery, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| |
Collapse
|
34
|
de Castro LF, Ovejero D, Boyce AM. DIAGNOSIS OF ENDOCRINE DISEASE: Mosaic disorders of FGF23 excess: Fibrous dysplasia/McCune-Albright syndrome and cutaneous skeletal hypophosphatemia syndrome. Eur J Endocrinol 2020; 182:R83-R99. [PMID: 32069220 PMCID: PMC7104564 DOI: 10.1530/eje-19-0969] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 02/17/2020] [Indexed: 12/11/2022]
Abstract
Fibrous dysplasia/McCune-Albright Syndrome (FD/MAS), arising from gain-of-function mutations in Gαs, and cutaneous skeletal hypophosphatemia syndrome (CSHS), arising from gain-of-function mutations in the Ras/MAPK pathway, are strikingly complex, mosaic diseases with overlapping phenotypes. Both disorders are defined by mosaic skin and bone involvement, and both are complicated by increased FGF23 production. These similarities have frequently led to mis-diagnoses, primarily in patients with CSHS who are often assumed to have FD/MAS. The intriguing similarities in skeletal involvement in these genetically distinct disorders have led to novel insights into FGF23 physiology, making an understanding of FD/MAS and CSHS relevant to both clinicians and researchers interested in bone and endocrine disorders. This review will give an overview of FD/MAS and CSHS, focusing on the roles of mosaicism and FGF23 in the pathogenesis and clinical presentation of these disorders.
Collapse
Affiliation(s)
- Luis F de Castro
- Skeletal Disorders and Mineral Homeostasis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, USA
| | - Diana Ovejero
- Skeletal Disorders and Mineral Homeostasis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, USA
- Musculoskeletal Research Unit, Hospital del Mar Institute of Medical Investigation (IMIM), Barcelona, Spain
- National Research Council, Institute of Clinical Physiology, Lecce, Italy
| | - Alison M Boyce
- Skeletal Disorders and Mineral Homeostasis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
35
|
Santra T, Herrero A, Rodriguez J, von Kriegsheim A, Iglesias-Martinez LF, Schwarzl T, Higgins D, Aye TT, Heck AJR, Calvo F, Agudo-Ibáñez L, Crespo P, Matallanas D, Kolch W. An Integrated Global Analysis of Compartmentalized HRAS Signaling. Cell Rep 2020; 26:3100-3115.e7. [PMID: 30865897 DOI: 10.1016/j.celrep.2019.02.038] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 12/16/2018] [Accepted: 02/11/2019] [Indexed: 12/27/2022] Open
Abstract
Modern omics technologies allow us to obtain global information on different types of biological networks. However, integrating these different types of analyses into a coherent framework for a comprehensive biological interpretation remains challenging. Here, we present a conceptual framework that integrates protein interaction, phosphoproteomics, and transcriptomics data. Applying this method to analyze HRAS signaling from different subcellular compartments shows that spatially defined networks contribute specific functions to HRAS signaling. Changes in HRAS protein interactions at different sites lead to different kinase activation patterns that differentially regulate gene transcription. HRAS-mediated signaling is the strongest from the cell membrane, but it regulates the largest number of genes from the endoplasmic reticulum. The integrated networks provide a topologically and functionally resolved view of HRAS signaling. They reveal distinct HRAS functions including the control of cell migration from the endoplasmic reticulum and TP53-dependent cell survival when signaling from the Golgi apparatus.
Collapse
Affiliation(s)
- Tapesh Santra
- Systems Biology Ireland, University College Dublin, Belfield, Dublin, Ireland
| | - Ana Herrero
- Systems Biology Ireland, University College Dublin, Belfield, Dublin, Ireland
| | - Javier Rodriguez
- Systems Biology Ireland, University College Dublin, Belfield, Dublin, Ireland
| | - Alex von Kriegsheim
- Systems Biology Ireland, University College Dublin, Belfield, Dublin, Ireland
| | | | - Thomas Schwarzl
- Systems Biology Ireland, University College Dublin, Belfield, Dublin, Ireland
| | - Des Higgins
- Systems Biology Ireland, University College Dublin, Belfield, Dublin, Ireland; Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Belfield, Ireland; School of Medicine and Medical Science, University College Dublin, Belfield, Ireland
| | - Thin-Thin Aye
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Centre for Biomolecular Research and Utrecht Institute for Pharmaceutical Science, Utrecht University, Padualaan 8, 3584 Utrecht, the Netherlands
| | - Albert J R Heck
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Centre for Biomolecular Research and Utrecht Institute for Pharmaceutical Science, Utrecht University, Padualaan 8, 3584 Utrecht, the Netherlands
| | - Fernando Calvo
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), Consejo Superior de Investigaciones Científicas (CSIC) - Universidad de Cantabria, Santander 39011, Spain
| | - Lorena Agudo-Ibáñez
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), Consejo Superior de Investigaciones Científicas (CSIC) - Universidad de Cantabria, Santander 39011, Spain
| | - Piero Crespo
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), Consejo Superior de Investigaciones Científicas (CSIC) - Universidad de Cantabria, Santander 39011, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| | - David Matallanas
- Systems Biology Ireland, University College Dublin, Belfield, Dublin, Ireland.
| | - Walter Kolch
- Systems Biology Ireland, University College Dublin, Belfield, Dublin, Ireland; Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Belfield, Ireland; School of Medicine and Medical Science, University College Dublin, Belfield, Ireland.
| |
Collapse
|
36
|
Kopra K, Vuorinen E, Abreu-Blanco M, Wang Q, Eskonen V, Gillette W, Pulliainen AT, Holderfield M, Härmä H. Homogeneous Dual-Parametric-Coupled Assay for Simultaneous Nucleotide Exchange and KRAS/RAF-RBD Interaction Monitoring. Anal Chem 2020; 92:4971-4979. [PMID: 32106676 PMCID: PMC7143314 DOI: 10.1021/acs.analchem.9b05126] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 02/28/2020] [Indexed: 02/07/2023]
Abstract
We have developed a rapid and sensitive single-well dual-parametric method introduced in linked RAS nucleotide exchange and RAS/RAF-RBD interaction assays. RAS mutations are frequent drivers of multiple different human cancers, but the development of therapeutic strategies has been challenging. Traditionally, efforts to disrupt the RAS function have focused on nucleotide exchange inhibitors, GTP-RAS interaction inhibitors, and activators increasing GTPase activity of mutant RAS proteins. As the amount of biological knowledge grows, targeted biochemical assays enabling high-throughput screening have become increasingly interesting. We have previously introduced a homogeneous quenching resonance energy transfer (QRET) assay for nucleotide binding studies with RAS and heterotrimeric G proteins. Here, we introduce a novel homogeneous signaling technique called QTR-FRET, which combine QRET technology and time-resolved Förster resonance energy transfer (TR-FRET). The dual-parametric QTR-FRET technique enables the linking of guanine nucleotide exchange factor-induced Eu3+-GTP association to RAS, monitored at 615 nm, and subsequent Eu3+-GTP-loaded RAS interaction with RAF-RBD-Alexa680 monitored at 730 nm. Both reactions were monitored in a single-well assay applicable for inhibitor screening and real-time reaction monitoring. This homogeneous assay enables separable detection of both nucleotide exchange and RAS/RAF interaction inhibitors using low nanomolar protein concentrations. To demonstrate a wider applicability as a screening and real-time reaction monitoring method, the QTR-FRET technique was also applied for G(i)α GTP-loading and pertussis toxin-catalyzed ADP-ribosylation of G(i)α, for which we synthesized a novel γ-GTP-Eu3+ molecule. The study indicates that the QTR-FRET detection technique presented here can be readily applied to dual-parametric assays for various targets.
Collapse
Affiliation(s)
- Kari Kopra
- Materials
Chemistry and Chemical Analysis, University
of Turku, Vatselankatu 2, 20500 Turku, Finland
| | - Emmiliisa Vuorinen
- Materials
Chemistry and Chemical Analysis, University
of Turku, Vatselankatu 2, 20500 Turku, Finland
| | - Maria Abreu-Blanco
- Leidos
Biomedical Research, Inc., Frederick National
Laboratory for Cancer Research, 8560 Progress Dr., Frederick, Maryland 21702, United States
| | - Qi Wang
- Institute
of Biomedicine, University of Turku, Kiinamyllynkatu 10, 20520 Turku, Finland
| | - Ville Eskonen
- Materials
Chemistry and Chemical Analysis, University
of Turku, Vatselankatu 2, 20500 Turku, Finland
| | - William Gillette
- Leidos
Biomedical Research, Inc., Frederick National
Laboratory for Cancer Research, 8560 Progress Dr., Frederick, Maryland 21702, United States
| | - Arto T. Pulliainen
- Institute
of Biomedicine, University of Turku, Kiinamyllynkatu 10, 20520 Turku, Finland
| | - Matthew Holderfield
- Leidos
Biomedical Research, Inc., Frederick National
Laboratory for Cancer Research, 8560 Progress Dr., Frederick, Maryland 21702, United States
| | - Harri Härmä
- Materials
Chemistry and Chemical Analysis, University
of Turku, Vatselankatu 2, 20500 Turku, Finland
| |
Collapse
|
37
|
Zhang D, Zhang Y, Gao Y, Chai X, Pi R, Chan G, Hu Y. Translating traditional herbal formulas into modern drugs: a network-based analysis of Xiaoyao decoction. Chin Med 2020; 15:25. [PMID: 32190105 PMCID: PMC7075005 DOI: 10.1186/s13020-020-00302-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 02/12/2020] [Indexed: 12/22/2022] Open
Abstract
Background Traditional Chinese medicine (TCM) encompasses numerous herbal formulas which play critical therapeutic roles through "multi-components, multi-targets and multi-pathways" mechanisms. Exploring the interaction among these mechanisms can certainly help to depict the core therapeutic function of herbal formulas. Xiaoyao decoction (XYD) is one of the most well-known traditional Chinese medicine formulas which has been widely applied to treat various diseases. In this study, taking XYD as an example, we proposed a network pharmacology-based method to identify the main therapeutic targets of this herbal concoctions. Methods Chemical data of XYD were retrieved from the Traditional Chinese Medicine Systems Pharmacology Database (TCMSP), Traditional Chinese Medicines Integrated Database (TCMID) and Compound Reference Database (CRD) and screened oral bioavailability attributes from SwissADME using Veber's filter. Targets of sample chemicals were identified using the online tool similarity ensemble approach (SEA), and pathways were enriched using STRING database. On the basis of targets-pathways interactions from the enrichment, a "targets-pathways-targets" (TPT) network was constructed. In the TPT network, the importance of each target was calculated by the declining value of network efficiency, which represents the influential strength of a specific set-off target on the whole network. Network-based predictive results were statistically validated with existing experimental evidence. Results The TPT network was comprised of 279 nodes and 6549 edges. The declining value of network efficiency of the sample targets was significantly correlated with their involvement frequency in existing studies of XYD using Spearman's test (p < 0.001). The top 10% of candidate targets, such as AKT1, PIK3R1, NFKB1 and RELA, etc., were chosen as XYD's main therapeutic targets, which further show pharmacological functions synergistically through 11 main pathways. These pathways are responsible for endocrine, nutritional or metabolic diseases, neoplasms and diseases of the nervous system, etc. Conclusions The network pharmacology-based approach in the present study shows promising potential for identifying the main therapeutic targets from TCM formulas. This study provides valuable information for TCM researchers and clinicians for better understanding the main therapeutic targets and therapeutic roles of herbal decoctions in clinical settings.
Collapse
Affiliation(s)
- Daiyan Zhang
- 1State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Yun Zhang
- 1State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Yan Gao
- 1State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Xingyun Chai
- 2Modern Research Center for Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029 China
| | - Rongbiao Pi
- 3School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006 China
| | - Ging Chan
- 1State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Yuanjia Hu
- 1State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| |
Collapse
|
38
|
Chottekalapanda RU, Kalik S, Gresack J, Ayala A, Gao M, Wang W, Meller S, Aly A, Schaefer A, Greengard P. AP-1 controls the p11-dependent antidepressant response. Mol Psychiatry 2020; 25:1364-1381. [PMID: 32439846 PMCID: PMC7303013 DOI: 10.1038/s41380-020-0767-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 04/10/2020] [Accepted: 04/28/2020] [Indexed: 01/10/2023]
Abstract
Selective serotonin reuptake inhibitors (SSRIs) are the most widely prescribed drugs for mood disorders. While the mechanism of SSRI action is still unknown, SSRIs are thought to exert therapeutic effects by elevating extracellular serotonin levels in the brain, and remodel the structural and functional alterations dysregulated during depression. To determine their precise mode of action, we tested whether such neuroadaptive processes are modulated by regulation of specific gene expression programs. Here we identify a transcriptional program regulated by activator protein-1 (AP-1) complex, formed by c-Fos and c-Jun that is selectively activated prior to the onset of the chronic SSRI response. The AP-1 transcriptional program modulates the expression of key neuronal remodeling genes, including S100a10 (p11), linking neuronal plasticity to the antidepressant response. We find that AP-1 function is required for the antidepressant effect in vivo. Furthermore, we demonstrate how neurochemical pathways of BDNF and FGF2, through the MAPK, PI3K, and JNK cascades, regulate AP-1 function to mediate the beneficial effects of the antidepressant response. Here we put forth a sequential molecular network to track the antidepressant response and provide a new avenue that could be used to accelerate or potentiate antidepressant responses by triggering neuroplasticity.
Collapse
Affiliation(s)
- Revathy U. Chottekalapanda
- 0000 0001 2166 1519grid.134907.8Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, 1230 York Avenue, New York, NY 10065 USA
| | - Salina Kalik
- 0000 0001 2166 1519grid.134907.8Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, 1230 York Avenue, New York, NY 10065 USA
| | - Jodi Gresack
- 0000 0001 2166 1519grid.134907.8Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, 1230 York Avenue, New York, NY 10065 USA
| | - Alyssa Ayala
- 0000 0001 2166 1519grid.134907.8Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, 1230 York Avenue, New York, NY 10065 USA
| | - Melanie Gao
- 0000 0001 2166 1519grid.134907.8Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, 1230 York Avenue, New York, NY 10065 USA
| | - Wei Wang
- 0000 0001 2166 1519grid.134907.8Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, 1230 York Avenue, New York, NY 10065 USA
| | - Sarah Meller
- 0000 0001 2166 1519grid.134907.8Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, 1230 York Avenue, New York, NY 10065 USA
| | - Ammar Aly
- 0000 0001 2166 1519grid.134907.8Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, 1230 York Avenue, New York, NY 10065 USA
| | - Anne Schaefer
- 0000 0001 0670 2351grid.59734.3cFriedman Brain Institute, Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Paul Greengard
- 0000 0001 2166 1519grid.134907.8Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, 1230 York Avenue, New York, NY 10065 USA
| |
Collapse
|
39
|
de Carvalho JB, de Morais GL, Vieira TCDS, Rabelo NC, Llerena JC, Gonzalez SMDC, de Vasconcelos ATR. miRNA Genetic Variants Alter Their Secondary Structure and Expression in Patients With RASopathies Syndromes. Front Genet 2019; 10:1144. [PMID: 31798637 PMCID: PMC6863982 DOI: 10.3389/fgene.2019.01144] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 10/21/2019] [Indexed: 12/22/2022] Open
Abstract
RASopathies are a group of rare genetic diseases caused by germline mutations in genes involved in the RAS–mitogen-activated protein kinase (RAS-MAPK) pathway. Whole-exome sequencing (WES) is a powerful approach for identifying new variants in coding and noncoding DNA sequences, including miRNAs. miRNAs are fine-tuning negative regulators of gene expression. The presence of variants in miRNAs could lead to malfunctions of regulation, resulting in diseases. Here, we identified 41 variants in mature miRNAs through WES analysis in five patients with previous clinical diagnosis of RASopathies syndromes. The pathways, biological processes, and diseases that were over-represented among the target genes of the mature miRNAs harboring variants included the RAS, MAPK, RAP1, and PIK3-Akt signaling pathways, neuronal differentiation, neurogenesis and nervous system development, congenital cardiac defects (hypertrophic cardiomyopathy, dilated cardiomyopathy, and arrhythmogenic right ventricular cardiomyopathy), and the phenotypes and syndromes of RASopathies (Noonan syndrome, Legius syndrome, Costello syndrome, Cafe au lait spots multiple, subaortic stenosis, pulmonary valve stenosis, and LEOPARD syndrome). Furthermore, eight selected variants in nine mature miRNAs (hsa-miR-1304, hsa-miR-146a, hsa-miR-196a2, hsa-miR-499a/hsa-miR-499b, hsa-miR-449b, hsa-miR-548l, hsa-miR-575, and hsa-miR-593) may have caused alterations in the secondary structures of miRNA precursor. Selected miRNAs containing variants such as hsa-miR-146a-3p, hsa-miR-196a-3p, hsa-miR-548l, hsa-miR-449b-5p, hsa-miR-575, and hsa-miR499a-3p could regulate classical genes associated with Rasopathies and RAS-MAPK pathways, contributing to modify the expression pattern of miRNAs in patients. RT-qPCR expression analysis revealed four differentially expressed miRNAs that were downregulated: miRNA-146a-3p in P1, P2, P3, P4, and P5, miR-1304-3p in P2, P3, P4, and P5, miR-196a2-3p in P3, and miR-499b-5p in P1. miR-499a-3p was upregulated in P1, P3, and P5. These results indicate that miRNAs show different expression patterns when these variants are present in patients. Therefore, this study characterized the role of miRNAs harboring variants related to RASopathies for the first time and indicated the possible implications of these variants for phenotypes of RASopathies such as congenital cardiac defects and cardio-cerebrovascular diseases. The expression and existence of miRNA variants may be used in the study of biomarkers of the RASopathies.
Collapse
Affiliation(s)
- Joseane Biso de Carvalho
- Bioinformatics Laboratory (LABINFO), National Laboratory for Scientific Computing (LNCC), Petrópolis, Brazil
| | - Guilherme Loss de Morais
- Bioinformatics Laboratory (LABINFO), National Laboratory for Scientific Computing (LNCC), Petrópolis, Brazil
| | - Thays Cristine Dos Santos Vieira
- Laboratory of Genomic Medicine, National Institute of Women, Children and Adolescents Health Fernandes Figueira (IFF/FIOCRUZ), Rio de Janeiro, Brazil
| | - Natana Chaves Rabelo
- Laboratory of Genomic Medicine, National Institute of Women, Children and Adolescents Health Fernandes Figueira (IFF/FIOCRUZ), Rio de Janeiro, Brazil
| | - Juan Clinton Llerena
- Department of Medical Genetics, National Institute of Women, Children and Adolescents Health Fernades Figueira, Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro, Brazil
| | - Sayonara Maria de Carvalho Gonzalez
- Laboratory of Genomic Medicine, National Institute of Women, Children and Adolescents Health Fernandes Figueira (IFF/FIOCRUZ), Rio de Janeiro, Brazil
| | | |
Collapse
|
40
|
Sadanandan N, Di Santo S, Widmer HR. Another win for endothelial progenitor cells: Endothelial progenitor cell-derived conditioned medium promotes proliferation and exerts neuroprotection in cultured neuronal progenitor cells. Brain Circ 2019; 5:106-111. [PMID: 31620656 PMCID: PMC6785943 DOI: 10.4103/bc.bc_41_19] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Revised: 09/10/2019] [Accepted: 09/11/2019] [Indexed: 01/08/2023] Open
Abstract
Progress in stem cell research demonstrates stem cells' potential for treating neurodegenerative diseases. Stem cells have proliferative/differentiative properties and produce a variety of paracrine factors that can potentially be used to regenerate nervous tissue. Previous studies have shown the positive regenerative effects of endothelial progenitor cells (EPCs), and thus, they may be used as a tool for regeneration. A study by Di Santo et al. explored whether EPC-derived conditioned medium (EPC-CM) promotes the survival of cultured striatal progenitor cells and attempted to find the paracrine factors and signaling pathways involved with EPC-CM's effects. The neuronal progenitor cells that were cultured with EPC-CM had much higher densities of GABA-immunoreactive (GABA-ir) neurons. It was shown that phosphatidylinositol-3-kinase/AKT and mitogen-activated protein kinase/ERK signaling pathways are involved in the proliferation of GABAergic neurons, as inhibition of these pathways decreased GABAergic densities. In addition, the results suggest that paracrine factors from EPC, both proteinaceous and lipidic, significantly elevated the viability and/or differentiation in the cultures. Importantly, it was found that EPC-CM provided neuroprotection against toxins from 3-nitropropionic acid. In sum, EPC-CM engendered proliferation and regeneration of the cultured striatal cells through paracrine factors and imparted neuroprotection. Furthermore, the effects of EPC-CM may generate a cell-free therapeutic strategy to address neurodegeneration.
Collapse
Affiliation(s)
- Nadia Sadanandan
- Department of Neurosurgery and Brain Repair, College of Medicine, University of South Florida Morsani, Tampa, FL, USA
| | - Stefano Di Santo
- Department of Neurosurgery, Neurocenter and Regenerative Neuroscience Cluster, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Hans Rudolf Widmer
- Department of Neurosurgery, Neurocenter and Regenerative Neuroscience Cluster, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
41
|
Lang C, Shu X, Peng L, Yu X. The ERK signaling pathway is involved in cardiotrophin-1-induced neural differentiation of human umbilical cord blood mesenchymal stem cells in vitro. Cytotechnology 2019; 71:977-988. [PMID: 31489528 DOI: 10.1007/s10616-019-00339-w] [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: 10/16/2018] [Accepted: 08/20/2019] [Indexed: 11/29/2022] Open
Abstract
Central nervous system diseases remain the most challenging pathologies, with limited or even no therapeutic possibilities and a poor prognosis. This study aimed to investigate the differentiation properties of human umbilical cord blood mesenchymal stem cells (hUCB-MSCs) transfected with recombinant adenovirus expressing enhanced green fluorescence protein cardiotrophin-1 (Adv-EGFP-CT-1) and the possible mechanisms involved. Cells were isolated, and MSC immunophenotypes were confirmed. The resulting differentiated cells treated with Adv-EGFP-CT-1 and cultured in neural induction medium (NIM) expressed higher levels of Nestin, neuronal nuclei (NeuN) and glial fibrillary acidic protein (GFAP) markers than cells in other treatments. Expression of glycoprotein 130/leukemia inhibitory factor receptor β (gp130/LiFRβ), Raf-1, phosphorylated Raf-1 (p-Raf-1), extracellular signal-regulated kinase 1/2 (ERK1/2) and phospho-ERK1/2 (p-ERK1/2) increased gradually within 72 h after transfection with Adv-EGFP-CT-1 and NIM culture. Additionally, inhibition of extracellular signal-regulated kinase kinase (MEK) abrogated expression of p-ERK1/2, Nestin, GFAP and NeuN. Thus, the ERK1/2 pathway may contribute to CT1-stimulated neural differentiation of hUCB-MSCs.
Collapse
Affiliation(s)
- Changhui Lang
- Department of Pediatrics, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, Guizhou, China
| | - Xiaomei Shu
- Department of Pediatrics, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, Guizhou, China.
| | - Longying Peng
- Department of Pediatrics, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, Guizhou, China
| | - Xiaohua Yu
- Department of Pediatrics, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, Guizhou, China
| |
Collapse
|
42
|
Zhang ZW, Pang B, Chen YC, Peng AQ. TMPRSS3 regulates cell viability and apoptosis processes of HEI-OC1 cells via regulation of the circ-Slc4a2, miR-182 and Akt cascade. J Gene Med 2019; 21:e3118. [PMID: 31408246 DOI: 10.1002/jgm.3118] [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: 05/30/2019] [Revised: 08/08/2019] [Accepted: 08/09/2019] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND The present study aimed to investigate the functions and regulation mechanism of the transmembrane protease, serine 3 (TMPRSS3), which plays an important role in sensorineural hearing loss. METHODS House Ear Institute-Organ of Corti 1 (HEI-OC1) cells, comprising auditory-related cells, were used in the present study. An overexpression vector and small hairpin RNA target on TMPRSS3 were designed and transfected into HEI-OC1 cells. Circular RNA (circRNA) sequencing was conducted and expression profiles were obtained. The circular structure of circRNAs was validated with a polymerase chain reaction and Sanger sequencing using convergent and divergent primers. RESULTS Overexpression of TMPRSS3 increased cell viability, whereas suppression of TMPRSS3 increased the percentage of apoptotic cells and decreased cell viability, compared to the control group. circRNA sequencing provided expression profiles indicating that the overexpression of TMPRSS3 increased the expression level of 195 circRNAs. Results of GO (Gene Ontology) and KEGG (Kyoto Encyclopedia of Genes and Genomes) studies indicated that the circRNAs are focused on the RAS signaling pathway. The pathway, circ-Slc41a2 (chr10: 82744115|82767120), miR-182 and Akt, might comprise one of the key cascades of TMPRSS3. CONCLUSIONS TMPRSS3 is an important molecule in the regulation of cell viability and cell apoptosis of HEI-OC1 cells. Its functions are dependent on the circ-Slc41a2, miR-182 and Akt cascade.
Collapse
Affiliation(s)
- Zhi-Wen Zhang
- Department of Otolaryngology, Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Bo Pang
- Department of Otolaryngology, Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yi-Chao Chen
- Department of Otolaryngology, Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - An-Quan Peng
- Department of Otolaryngology, Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
43
|
Novel Tools towards Magnetic Guidance of Neurite Growth: (I) Guidance of Magnetic Nanoparticles into Neurite Extensions of Induced Human Neurons and In Vitro Functionalization with RAS Regulating Proteins. J Funct Biomater 2019; 10:jfb10030032. [PMID: 31315182 PMCID: PMC6787644 DOI: 10.3390/jfb10030032] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 07/08/2019] [Accepted: 07/11/2019] [Indexed: 12/18/2022] Open
Abstract
Parkinson’s disease (PD) is a neurodegenerative disease associated with loss or dysfunction of dopaminergic neurons located in the substantia nigra (SN), and there is no cure available. An emerging new approach for treatment is to transplant human induced dopaminergic neurons directly into the denervated striatal brain target region. Unfortunately, neurons grafted into the substantia nigra are unable to grow axons into the striatum and thus do not allow recovery of the original connectivity. Towards overcoming this general limitation in guided neuronal regeneration, we develop here magnetic nanoparticles functionalized with proteins involved in the regulation of axonal growth. We show covalent binding of constitutive active human rat sarcoma (RAS) proteins or RAS guanine nucleotide exchange factor catalytic domain of son of sevenless (SOS) by fluorescence correlation spectroscopy and multiangle light scattering as well as the characterization of exchange factor activity. Human dopaminergic neurons were differentiated from neural precursor cells and characterized by electrophysiological and immune histochemical methods. Furthermore, we demonstrate magnetic translocation of cytoplasmic γ-Fe2O3@SiO2 core-shell nanoparticles into the neurite extensions of induced human neurons. Altogether, we developed tools towards remote control of directed neurite growth in human dopaminergic neurons. These results may have relevance for future therapeutic approaches of cell replacement therapy in Parkinson’s disease.
Collapse
|
44
|
Huang N, Zhang Y, Chen M, Jin H, Nie J, Luo Y, Zhou S, Shi J, Jin F. Resveratrol delays 6-hydroxydopamine-induced apoptosis by activating the PI3K/Akt signaling pathway. Exp Gerontol 2019; 124:110653. [PMID: 31295526 DOI: 10.1016/j.exger.2019.110653] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 05/30/2019] [Accepted: 07/05/2019] [Indexed: 01/10/2023]
Abstract
This study aimed to determine whether resveratrol (Res) delays the progression of 6-hydroxydopamine (6-OHDA)-induced apoptosis via activating the phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) signaling pathway. Sprague-Dawley (SD) rats were unilaterally injected with 6-OHDA (8 μg/4 μL) into the substantia nigra of the midbrain. Res (15 and 30 mg/kg) was given orally to the rats for a total of 36 days to examine its protective effects. We first tested whether Res can delay the progression of 6-OHDA-induced damage by measuring weight and performance on behavioral tests (rotarod, open field test and grid test) and further explored whether this effect is related to the activation of the PI3K/Akt signaling pathway using immunohistochemistry (IHC) and Western blotting (WB). Our results showed that the damage induced by 6-OHDA gradually worsened, while Res 30 mg/kg treatment significantly improved motor function and increased body weight. Compared with those in the model group, the number of dopaminergic neurons cells and the expression of PI3K-110α, p-Akt Ser473, and pro-caspase-3 in the Res 30 mg/kg group were significantly increased, and the Bax/Bcl-2 ratio and the level of activated caspase-3 was decreased. The results indicate that Res ameliorates 6-OHDA-induced apoptosis and motor dysfunction via activating the PI3K/Akt signaling pathway, delaying the progression of Parkinson's disease (PD) symptoms in this model.
Collapse
Affiliation(s)
- Nanqu Huang
- Key Laboratory of Basic Pharmacology and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Guizhou, China; Drug Clinical Trial Institution, The First People's Hospital of Zunyi & The Third Affiliated Hospital of Zunyi Medical University, Guizhou, China
| | - Ying Zhang
- Key Laboratory of Basic Pharmacology and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Guizhou, China
| | - Mingji Chen
- Key Laboratory of Basic Pharmacology and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Guizhou, China
| | - Hai Jin
- Institute of Digestive Diseases of Affiliated Hospital, Zunyi Medical University, Guizhou, China
| | - Jing Nie
- Key Laboratory of Basic Pharmacology and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Guizhou, China
| | - Yong Luo
- Department of Neurology, The First People's Hospital of Zunyi & The Third Affiliated Hospital of Zunyi Medical University, Guizhou, China
| | - Shaoyu Zhou
- Key Laboratory of Basic Pharmacology and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Guizhou, China; Department of Environmental Health, Indiana University Bloomington, IN, United States
| | - Jingshan Shi
- Key Laboratory of Basic Pharmacology and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Guizhou, China
| | - Feng Jin
- Key Laboratory of Basic Pharmacology and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Guizhou, China.
| |
Collapse
|
45
|
Transcriptomic Analysis of MAPK Signaling in NSC-34 Motor Neurons Treated with Vitamin E. Nutrients 2019; 11:nu11051081. [PMID: 31096690 PMCID: PMC6566669 DOI: 10.3390/nu11051081] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 05/08/2019] [Accepted: 05/10/2019] [Indexed: 12/14/2022] Open
Abstract
Vitamin E family is composed of different tocopherols and tocotrienols that are well-known as antioxidants but that exert also non-antioxidant effects. Oxidative stress may be involved in the progression of neurodegenerative disorders including amyotrophic lateral sclerosis (ALS), characterized by motor neuron death. The aim of the study was the evaluation of the changes induced in the transcriptional profile of NSC-34 motor neurons treated with α-tocopherol. In particular, cells were treated for 24 h with 10 µM α-tocopherol, RNA was extracted and transcriptomic analysis was performed using Next Generation Sequencing. Vitamin E treatment modulated MAPK signaling pathway. The evaluation revealed that 34 and 12 genes, respectively belonging to “Classical MAP kinase pathway” and “JNK and p38 MAP kinase pathway”, were involved. In particular, a downregulation of the genes encoding for p38 (Log2 fold change −0.87 and −0.67) and JNK (Log2 fold change −0.16) was found. On the contrary, the gene encoding for ERK showed a higher expression in cells treated with vitamin E (Log2 fold change 0.30). Since p38 and JNK seem more involved in cell death, while ERK in cell survival, the data suggested that vitamin E treatment may exert a protective role in NSC-34 motor neurons. Moreover, Vitamin E treatment reduced the expression of the genes which encode proteins involved in mitophagy. These results indicate that vitamin E may be an efficacious therapy in preventing motor neuron death, opening new strategies for those diseases that involve motor neurons, including ALS.
Collapse
|
46
|
Subaraja M, Kulandaisamy A, Shanmugam NRS, Vanisree AJ. Homology modeling identified for purported drug targets to the neuroprotective effects of levodopa and asiaticoside-D in degenerated cerebral ganglions of Lumbricus terrestris. Indian J Pharmacol 2019; 51:31-39. [PMID: 31031465 PMCID: PMC6444839 DOI: 10.4103/ijp.ijp_600_18] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
CONTEXT: Homology modeling plays role in determining the therapeutic targets dreadful for condition such as neurodegenerative diseases (NDD), which pose challenge in achieving the effective managements. The structures of the serotonin transporter (SERT), aquaporin (AQP), and tropomyosin receptor kinase (TrkA) which are implicated in NDD pathology are still unknown for Lumbricus terrestris, but the three-dimensional (3D) structure of the human counterpart for modeling. AIM: This study aims to generate and evaluate the 3D structure of TrkA, SERT, and AQP proteins and their interaction with the ligands, namely Asiaticoside-D (AD) and levodopa (L-DOPA) the anti-NDD agents. SUBJECTS AND METHODS: Homology modeling for SERT, AQP, and TrkA proteins of Lumbricus terrestris using SWISS-MODEL Server and the modeled structure was validated using Rampage Server. Wet-lab analysis of their correspondent m-RNA levels was also done to validate the in silico data. RESULTS: It was found that TrkA had moderately high homology (67%) to human while SERT and AQP could exhibit 58% and 42%, respectively. The reliability of the model was assessed by Ramachandran plot analysis. Interactions of AD with the SERT, AQP-4, and TrkA showed the binding energies as −9.93, 8.88, and −7.58 of Kcal/mol, respectively, while for L-DOPA did show −3.93, −5.13, and −6.0 Kcal/mol, respectively. The levels of SERT, TrkA, and AQP-4 were significantly reduced (P < 0.001) on ROT induced when compared to those of control worms. On ROT + AD supplementation group (III), m-RNA levels were significantly increased (P < 0.05) when compared to those of ROT induced worms (group II). CONCLUSION: Our pioneering docking data propose the possible of target which is proved useful for therapeutic investigations against the unconquered better of NDD.
Collapse
Affiliation(s)
- Mamangam Subaraja
- Department of Biochemistry, Guindy Campus, University of Madras, Chennai, Tamil Nadu, India
| | - A Kulandaisamy
- Department of Biotechnology, Indian Institute of Technology, Chennai, Tamil Nadu, India
| | - N R Siva Shanmugam
- Department of Biotechnology, Indian Institute of Technology, Chennai, Tamil Nadu, India
| | | |
Collapse
|
47
|
Santo SD, Seiler S, Andres R, Widmer HR. Endothelial Progenitor Cells Conditioned Medium Supports Number of GABAergic Neurons and Exerts Neuroprotection in Cultured Striatal Neuronal Progenitor Cells. Cell Transplant 2019; 28:367-378. [PMID: 31017468 PMCID: PMC6628568 DOI: 10.1177/0963689719835192] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
There is growing evidence that stem and progenitor cells exert regenerative actions by means of paracrine factors. In line with these notions, we recently demonstrated that endothelial progenitor cell (EPC)-derived conditioned medium (EPC-CM) substantially increased viability of brain microvascular cells. In the present study, we aimed at investigating whether EPC-CM supports cell survival of cultured striatal progenitor cells. For that purpose, primary cultures from fetal rat embryonic (E14) ganglionic eminence were prepared and grown for 7 days in vitro (DIV). EPC-CM was administered from DIV5–7. Treatment of the striatal cultures with EPC-CM resulted in significantly increased densities of GABA-immunoreactive (-ir) neurons. Inhibition of mitogen-activated protein kinase and phosphatidylinositol-3-kinase, but not of the ROCK pathway, significantly attenuated the EPC-CM induced increase in GABA-ir cell densities. Similar results were observed when EPC-CM was subjected to proteolytic digestion and lipid extraction. Furthermore, inhibition of translation abolished the EPC-CM induced effects. Importantly, EPC-CM displayed neuroprotection against 3-nitropropionic acid induced toxicity. These findings demonstrate that EPC-derived paracrine factors substantially promote survival and/or differentiation of cultured striatal progenitor cells involving both proteinaceous factors and lipidic factors. In sum, EPC-CM constituents might lead to a novel cell-free therapeutic strategy to challenge neuronal degeneration.
Collapse
Affiliation(s)
- Stefano Di Santo
- 1 Department of Neurosurgery, Bern University Hospital, Switzerland
| | - Stefanie Seiler
- 1 Department of Neurosurgery, Bern University Hospital, Switzerland
| | - Robert Andres
- 1 Department of Neurosurgery, Bern University Hospital, Switzerland
| | | |
Collapse
|
48
|
Baquedano Lobera I, Izquierdo Álvarez S, Oliván Del Cacho MJ. Rasopathies case report: concurrence of two pathogenic variations de novo in NF1 and KRAS genes in a patient. BMC Pediatr 2019; 19:92. [PMID: 30953504 PMCID: PMC6449997 DOI: 10.1186/s12887-019-1463-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 03/19/2019] [Indexed: 01/11/2023] Open
Abstract
Background Rasopathies are a group of genetic malformative syndromes including neurofibromatosis 1, Noonan, LEOPARD, Costello, cardio-facio-cutaneous, Legius, and capillary malformation-arteriovenous malformation syndromes. Case presentation We present a female newborn that consulted at the emergency department with refusal to eat and sleepiness. A shortened femur, thickened nucal fold and suspect for agenesis of the corpus callosum were observed in prenatal ultrasound. Her phenotype included hypertelorism, antimongoloid obliquity of the palpebral fissure, prominent forehead, long filtrum, thickened nucal fold, separated nipples, widespread thickened skinfolds and café-au-lait spots. She had a systolic murmur due to pulmonary valve stenosis. The NF1 gene testing found the pathogenic variant p.E2586X (c.7756G > T) in exon 53, not described in any international database or scientific publications yet. Also, a mutation in the Kras gene was detected (p.Val14lle), which is associated with mild Noonan phenotype. Both variations were de novo. Conclusions Not all genes and mutations have already been discovered, so it’s important to document new findings, like our patient’s, to enrich and update the international database and broaden all possible knowledge about rasopathies. This is the first case to be described presenting simultaneously two mutations in Kras and NF1 genes, whose possible synergic effect regarding its pathogenicity is unknown, but could be interesting towards therapeutic alternatives.
Collapse
Affiliation(s)
- Irene Baquedano Lobera
- Pediatrics Department, Miguel Servet Children's Hospital, Isabel la Católica Avenue 1-3, 50009, Zaragoza, Spain.
| | - Silvia Izquierdo Álvarez
- Clinical Genetics and Assisted Reproduction, Clinical Biochemistry Department, Miguel Servet Hospital, Padre Arrupe Street, 50009, Zaragoza, Spain
| | - María Jesús Oliván Del Cacho
- Neonatology Department, Miguel Servet Children's Hospital, Isabel la Católica Avenue 1-3, 50009, Zaragoza, Spain
| |
Collapse
|
49
|
Zhang Y, Huang N, Chen M, Jin H, Nie J, Shi J, Jin F. Procyanidin protects against 6-hydroxydopamine-induced dopaminergic neuron damage via the regulation of the PI3K/Akt signalling pathway. Biomed Pharmacother 2019; 114:108789. [PMID: 30925459 DOI: 10.1016/j.biopha.2019.108789] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 02/21/2019] [Accepted: 03/13/2019] [Indexed: 12/15/2022] Open
Abstract
Over the past 200 years, Parkinson's disease (PD) has remained an insurmountable challenge. Despite the existence of numerous therapeutic drugs, there are still not enough treatments in the face of the complex pathogenesis of PD. Therefore, the search for more effective therapeutic drugs for PD has extremely important practical significance. Procyanidin (PC), widely found in plants such as grapes, is a bioflavonoid antioxidant with a special molecular structure that can effectively remove free radicals in the human body. To explore its possible mechanism in PD, we used 6-hydroxydopamine (6-OHDA, 8 μg) to mimic dopaminergic (DA) neuronal damage and validated this model in vivo and in vitro. in vivo, we detected an effect of PC (60 mg/kg) on the behavioural changes exhibited in 6-OHDA model rats, the number of DA neurons and the phosphorylation of protein kinase B (Akt). in vitro, we detected changes in cell viability, mitochondrial membrane potential (MMP) and total superoxide dismutase (SOD) and explored the role of PC (50 μM) by inhibiting the phosphoinositide 3-kinase (PI3K)/Akt signalling pathway with LY294002 (20 μM). The present study demonstrates that PC plays a protective role against 6-OHDA-induced neurotoxicity, which may be mediated through the activation of the PI3K/Akt signalling pathway. This study indicates a potential use for PC in the treatment and prevention of PD.
Collapse
Affiliation(s)
- Ying Zhang
- Key Laboratory of Basic Pharmacology and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Guizhou, China
| | - Nanqu Huang
- Drug Clinical Trial Institution, The First People's Hospital of Zunyi, Guizhou, China
| | - Mingji Chen
- Key Laboratory of Basic Pharmacology and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Guizhou, China
| | - Hai Jin
- Institute of Digestive Diseases of Affiliated Hospital, Zunyi Medical University, Guizhou, China
| | - Jing Nie
- Key Laboratory of Basic Pharmacology and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Guizhou, China
| | - Jingshan Shi
- Key Laboratory of Basic Pharmacology and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Guizhou, China
| | - Feng Jin
- Key Laboratory of Basic Pharmacology and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Guizhou, China.
| |
Collapse
|
50
|
Perspectives of RAS and RHEB GTPase Signaling Pathways in Regenerating Brain Neurons. Int J Mol Sci 2018; 19:ijms19124052. [PMID: 30558189 PMCID: PMC6321366 DOI: 10.3390/ijms19124052] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 12/05/2018] [Accepted: 12/13/2018] [Indexed: 12/29/2022] Open
Abstract
Cellular activation of RAS GTPases into the GTP-binding “ON” state is a key switch for regulating brain functions. Molecular protein structural elements of rat sarcoma (RAS) and RAS homolog protein enriched in brain (RHEB) GTPases involved in this switch are discussed including their subcellular membrane localization for triggering specific signaling pathways resulting in regulation of synaptic connectivity, axonal growth, differentiation, migration, cytoskeletal dynamics, neural protection, and apoptosis. A beneficial role of neuronal H-RAS activity is suggested from cellular and animal models of neurodegenerative diseases. Recent experiments on optogenetic regulation offer insights into the spatiotemporal aspects controlling RAS/mitogen activated protein kinase (MAPK) or phosphoinositide-3 kinase (PI3K) pathways. As optogenetic manipulation of cellular signaling in deep brain regions critically requires penetration of light through large distances of absorbing tissue, we discuss magnetic guidance of re-growing axons as a complementary approach. In Parkinson’s disease, dopaminergic neuronal cell bodies degenerate in the substantia nigra. Current human trials of stem cell-derived dopaminergic neurons must take into account the inability of neuronal axons navigating over a large distance from the grafted site into striatal target regions. Grafting dopaminergic precursor neurons directly into the degenerating substantia nigra is discussed as a novel concept aiming to guide axonal growth by activating GTPase signaling through protein-functionalized intracellular magnetic nanoparticles responding to external magnets.
Collapse
|