51
|
Zheng M, Liu Z, Mana L, Qin G, Huang S, Gong Z, Tian M, He Y, Wang P. Shenzhiling oral liquid protects the myelin sheath against Alzheimer's disease through the PI3K/Akt-mTOR pathway. JOURNAL OF ETHNOPHARMACOLOGY 2021; 278:114264. [PMID: 34082015 DOI: 10.1016/j.jep.2021.114264] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 05/22/2021] [Accepted: 05/27/2021] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Shenzhiling oral liquid (SZL), a traditional Chinese medicine (TCM) compound, is firstly approved by the Chinese Food and Drug Administration (CFDA) for the treatment of mild to moderate Alzheimer's disease (AD). SZL is composed of ten Chinese herbs, and the precise therapy mechanism of its action to AD is far from fully understood. AIM OF THE STUDY The purpose of this study was to observe whether SZL is an effective therapy for amyloid-beta (Aβ)-induced myelin sheath and oligodendrocytes impairments. Notably, the primary aim was to elucidate whether and through what underlying mechanism SZL protects the myelin sheath through the PI3K/Akt-mTOR signaling pathway in Aβ42-induced OLN-93 oligodendrocytes in vitro. MATERIALS AND METHODS APP/PS1 mice were treated with SZL or donepezil continuously for three months, and Aβ42-induced oligodendrocyte OLN-93 cells mimicking AD pathogenesis of myelin sheath impairments were incubated with SZL-containing serum or with donepezil. LC-MS/MS was used to analysis the active components of SZL and SZL-containing serum. The Y maze test was administered after 3 months of treatment, and the hippocampal tissues of the APP/PS1 mice were then harvested for observation of myelin sheath and oligodendrocyte morphology. Cell viability and toxicity were assessed using CCK-8 and lactate dehydrogenase (LDH) release assays, and flow cytometry was used to measure cell apoptosis. The expression of the myelin proteins MBP, PLP, and MAG and that of Aβ42 and Aβ40 in the hippocampi of APP/PS1 mice were examined after SZL treatment. Simultaneously, the expression of p-PI3K, PI3K, p-Akt, Akt, p-mTOR, and mTOR were also examined. The expression of proteins, including CNPase, Olig2, NKX2.2, MBP, PLP, MAG, MOG, p-PI3K, PI3K, p-Akt, Akt, p-mTOR, and mTOR, was determined by immunofluorescence and Western blot, and the corresponding gene expression was evaluated by qPCR in Aβ42-induced OLN-93 oligodendrocytes. RESULTS LC-MS/MS detected a total of 126 active compounds in SZL-containing serum, including terpenoids, flavones, phenols, phenylpropanoids and phenolic acids. SZL treatment significantly improved memory and cognition in APP/PS1 mice and decreased the G-ratio of myelin sheath, alleviated myelin sheath and oligodendrocyte impairments by decreasing Aβ42 and Aβ40 accumulation and increasing the expression of myelin proteins MBP, PLP, MAG, and PI3K/Akt-mTOR signaling pathway associated protein in the hippocampi of APP/PS1 mice. SZL-containing serum also significantly reversed the OLN-93 cell injury induced by Aβ42 by increasing cell viability and enhanced the expression of MBP, PLP, MAG, and MOG. Meanwhile, SZL-containing serum facilitated the maturation and differentiation of oligodendrocytes in Aβ42-induced OLN-93 cells by heightening the expression of CNPase, Olig2 and NKX2.2. SZL-containing serum treatment also fostered the expression of p-PI3K, PI3K, p-Akt, Akt, p-mTOR, and mTOR, indicating an activating PI3K/Akt-mTOR signaling pathway in OLN-93 cells. Furthermore, the effects of SZL on myelin proteins, p-Akt, and p-mTOR were clearly inhibited by LY294002 and/or rapamycin, antagonists of PI3K and m-TOR, respectively. CONCLUSIONS Our findings indicate that SZL exhibits a neuroprotective effect on the myelin sheath by promoting the expression of myelin proteins during AD, and its mechanism of action is closely related to the activation of the PI3K/Akt-mTOR signaling pathway.
Collapse
Affiliation(s)
- Mingcui Zheng
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine (BUCM), Beijing, 100700, China; Key Laboratory of Pharmacology Dongzhimen Hospital (BUCM), State Administration of Traditional Chinese Medicine, Beijing, 100700, China.
| | - Zhenhong Liu
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine (BUCM), Beijing, 100700, China; Key Laboratory of Pharmacology Dongzhimen Hospital (BUCM), State Administration of Traditional Chinese Medicine, Beijing, 100700, China; Institute for Brain Disorders, Beijing University of Chinese Medicine (BUCM), Beijing, 100029, China.
| | - Lulu Mana
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine (BUCM), Beijing, 100700, China; Key Laboratory of Pharmacology Dongzhimen Hospital (BUCM), State Administration of Traditional Chinese Medicine, Beijing, 100700, China; Xinjiang Medical University, Urumqi, 830011, China.
| | - Gaofeng Qin
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine (BUCM), Beijing, 100700, China; Key Laboratory of Pharmacology Dongzhimen Hospital (BUCM), State Administration of Traditional Chinese Medicine, Beijing, 100700, China.
| | - Shuaiyang Huang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine (BUCM), Beijing, 100700, China; Key Laboratory of Pharmacology Dongzhimen Hospital (BUCM), State Administration of Traditional Chinese Medicine, Beijing, 100700, China.
| | - Zhuoyan Gong
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine (BUCM), Beijing, 100700, China; Key Laboratory of Pharmacology Dongzhimen Hospital (BUCM), State Administration of Traditional Chinese Medicine, Beijing, 100700, China.
| | - Meijing Tian
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine (BUCM), Beijing, 100700, China; Key Laboratory of Pharmacology Dongzhimen Hospital (BUCM), State Administration of Traditional Chinese Medicine, Beijing, 100700, China.
| | - Yannan He
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine (BUCM), Beijing, 100700, China; Key Laboratory of Pharmacology Dongzhimen Hospital (BUCM), State Administration of Traditional Chinese Medicine, Beijing, 100700, China.
| | - Pengwen Wang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine (BUCM), Beijing, 100700, China; Key Laboratory of Pharmacology Dongzhimen Hospital (BUCM), State Administration of Traditional Chinese Medicine, Beijing, 100700, China.
| |
Collapse
|
52
|
Abstract
Myelin is a key evolutionary specialization and adaptation of vertebrates formed by the plasma membrane of glial cells, which insulate axons in the nervous system. Myelination not only allows rapid and efficient transmission of electric impulses in the axon by decreasing capacitance and increasing resistance but also influences axonal metabolism and the plasticity of neural circuits. In this review, we will focus on Schwann cells, the glial cells which form myelin in the peripheral nervous system. Here, we will describe the main extrinsic and intrinsic signals inducing Schwann cell differentiation and myelination and how myelin biogenesis is achieved. Finally, we will also discuss how the study of human disorders in which molecules and pathways relevant for myelination are altered has enormously contributed to the current knowledge on myelin biology.
Collapse
Affiliation(s)
- Alessandra Bolino
- Human Inherited Neuropathies Unit, Institute of Experimental Neurology INSPE, Division of Neuroscience, IRCCS Ospedale San Raffaele, Via Olgettina 60, 20132, Milan, Italy.
| |
Collapse
|
53
|
Xu R, He Q, Wang Y, Yang Y, Guo ZN. Therapeutic Potential of Remote Ischemic Conditioning in Vascular Cognitive Impairment. Front Cell Neurosci 2021; 15:706759. [PMID: 34413726 PMCID: PMC8370253 DOI: 10.3389/fncel.2021.706759] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Accepted: 06/29/2021] [Indexed: 12/21/2022] Open
Abstract
Vascular cognitive impairment (VCI) is a heterogeneous disease caused by a variety of cerebrovascular diseases. Patients with VCI often present with slower cognitive processing speed and poor executive function, which affects their independence in daily life, thus increasing social burden. Remote ischemic conditioning (RIC) is a non-invasive and efficient intervention that triggers endogenous protective mechanisms to generate neuroprotection. Over the past decades, evidence from basic and clinical research has shown that RIC is promising for the treatment of VCI. To further our understanding of RIC and improve the management of VCI, we summarize the evidence on the therapeutic potential of RIC in relation to the risk factors and pathobiologies of VCI, including reducing the risk of recurrent stroke, decreasing high blood pressure, improving cerebral blood flow, restoring white matter integrity, protecting the neurovascular unit, attenuating oxidative stress, and inhibiting the inflammatory response.
Collapse
Affiliation(s)
- Rui Xu
- Department of Neurology, Stroke Center & Clinical Trial and Research Center for Stroke, The First Hospital of Jilin University, Changchun, China.,China National Comprehensive Stroke Center, Changchun, China.,Jilin Provincial Key Laboratory of Cerebrovascular Disease, Changchun, China
| | - Qianyan He
- Department of Neurology, Stroke Center & Clinical Trial and Research Center for Stroke, The First Hospital of Jilin University, Changchun, China.,China National Comprehensive Stroke Center, Changchun, China.,Jilin Provincial Key Laboratory of Cerebrovascular Disease, Changchun, China
| | - Yan Wang
- Department of Neurology, Stroke Center & Clinical Trial and Research Center for Stroke, The First Hospital of Jilin University, Changchun, China.,China National Comprehensive Stroke Center, Changchun, China.,Jilin Provincial Key Laboratory of Cerebrovascular Disease, Changchun, China
| | - Yi Yang
- Department of Neurology, Stroke Center & Clinical Trial and Research Center for Stroke, The First Hospital of Jilin University, Changchun, China.,China National Comprehensive Stroke Center, Changchun, China.,Jilin Provincial Key Laboratory of Cerebrovascular Disease, Changchun, China
| | - Zhen-Ni Guo
- Department of Neurology, Stroke Center & Clinical Trial and Research Center for Stroke, The First Hospital of Jilin University, Changchun, China.,China National Comprehensive Stroke Center, Changchun, China.,Jilin Provincial Key Laboratory of Cerebrovascular Disease, Changchun, China
| |
Collapse
|
54
|
Bernardo A, De Nuccio C, Visentin S, Martire A, Minghetti L, Popoli P, Ferrante A. Myelin Defects in Niemann-Pick Type C Disease: Mechanisms and Possible Therapeutic Perspectives. Int J Mol Sci 2021; 22:ijms22168858. [PMID: 34445564 PMCID: PMC8396228 DOI: 10.3390/ijms22168858] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/11/2021] [Accepted: 08/16/2021] [Indexed: 12/25/2022] Open
Abstract
Niemann–Pick type C (NPC) disease is a wide-spectrum clinical condition classified as a neurovisceral disorder affecting mainly the liver and the brain. It is caused by mutations in one of two genes, NPC1 and NPC2, coding for proteins located in the lysosomes. NPC proteins are deputed to transport cholesterol within lysosomes or between late endosome/lysosome systems and other cellular compartments, such as the endoplasmic reticulum and plasma membrane. The first trait of NPC is the accumulation of unesterified cholesterol and other lipids, like sphingosine and glycosphingolipids, in the late endosomal and lysosomal compartments, which causes the blockade of autophagic flux and the impairment of mitochondrial functions. In the brain, the main consequences of NPC are cerebellar neurodegeneration, neuroinflammation, and myelin defects. This review will focus on myelin defects and the pivotal importance of cholesterol for myelination and will offer an overview of the molecular targets and the pharmacological strategies so far proposed, or an object of clinical trials for NPC. Finally, it will summarize recent data on a new and promising pharmacological perspective involving A2A adenosine receptor stimulation in genetic and pharmacological NPC dysmyelination models.
Collapse
Affiliation(s)
- Antonietta Bernardo
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (A.B.); (S.V.); (A.M.); (P.P.)
| | - Chiara De Nuccio
- Research Coordination and Support Service, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (C.D.N.); (L.M.)
| | - Sergio Visentin
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (A.B.); (S.V.); (A.M.); (P.P.)
| | - Alberto Martire
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (A.B.); (S.V.); (A.M.); (P.P.)
| | - Luisa Minghetti
- Research Coordination and Support Service, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (C.D.N.); (L.M.)
| | - Patrizia Popoli
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (A.B.); (S.V.); (A.M.); (P.P.)
| | - Antonella Ferrante
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (A.B.); (S.V.); (A.M.); (P.P.)
- Correspondence: ; Tel.: +39-06-49902050
| |
Collapse
|
55
|
Dysregulation of myelin synthesis and actomyosin function underlies aberrant myelin in CMT4B1 neuropathy. Proc Natl Acad Sci U S A 2021; 118:2009469118. [PMID: 33653949 DOI: 10.1073/pnas.2009469118] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Charcot-Marie-Tooth type 4B1 (CMT4B1) is a severe autosomal recessive demyelinating neuropathy with childhood onset, caused by loss-of-function mutations in the myotubularin-related 2 (MTMR2) gene. MTMR2 is a ubiquitously expressed catalytically active 3-phosphatase, which in vitro dephosphorylates the 3-phosphoinositides PtdIns3P and PtdIns(3,5)P 2, with a preference for PtdIns(3,5)P 2 A hallmark of CMT4B1 neuropathy are redundant loops of myelin in the nerve termed myelin outfoldings, which can be considered the consequence of altered growth of myelinated fibers during postnatal development. How MTMR2 loss and the resulting imbalance of 3'-phosphoinositides cause CMT4B1 is unknown. Here we show that MTMR2 by regulating PtdIns(3,5)P 2 levels coordinates mTORC1-dependent myelin synthesis and RhoA/myosin II-dependent cytoskeletal dynamics to promote myelin membrane expansion and longitudinal myelin growth. Consistent with this, pharmacological inhibition of PtdIns(3,5)P 2 synthesis or mTORC1/RhoA signaling ameliorates CMT4B1 phenotypes. Our data reveal a crucial role for MTMR2-regulated lipid turnover to titrate mTORC1 and RhoA signaling thereby controlling myelin growth.
Collapse
|
56
|
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
|
57
|
Ishii A, Furusho M, Bansal R. Mek/ERK1/2-MAPK and PI3K/Akt/mTOR signaling plays both independent and cooperative roles in Schwann cell differentiation, myelination and dysmyelination. Glia 2021; 69:2429-2446. [PMID: 34157170 DOI: 10.1002/glia.24049] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 04/29/2021] [Accepted: 06/04/2021] [Indexed: 01/15/2023]
Abstract
Multiple signals are involved in the regulation of developmental myelination by Schwann cells and in the maintenance of a normal myelin homeostasis throughout adult life, preserving the integrity of the axons in the PNS. Recent studies suggest that Mek/ERK1/2-MAPK and PI3K/Akt/mTOR intracellular signaling pathways play important, often overlapping roles in the regulation of myelination in the PNS. In addition, hyperactivation of these signaling pathways in Schwann cells leads to a late onset of various pathological changes in the sciatic nerves. However, it remains poorly understood whether these pathways function independently or sequentially or converge using a common mechanism to facilitate Schwann cell differentiation and myelin growth during development and in causing pathological changes in the adult animals. To address these questions, we analyzed multiple genetically modified mice using simultaneous loss- and constitutive gain-of-function approaches. We found that during development, the Mek/ERK1/2-MAPK pathway plays a primary role in Schwann cell differentiation, distinct from mTOR. However, during active myelination, ERK1/2 is dependent on mTOR signaling to drive the growth of the myelin sheath and regulate its thickness. Finally, our data suggest that peripheral nerve pathology during adulthood caused by hyperactivation of Mek/ERK1/2-MAPK or PI3K is likely to be independent or dependent on mTOR-signaling in different contexts. Thus, this study highlights the complexities in the roles played by two major intracellular signaling pathways in Schwann cells that affect their differentiation, myelination, and later PNS pathology and predicts that potential therapeutic modulation of these pathways in PNS neuropathies could be a complex process.
Collapse
Affiliation(s)
- Akihiro Ishii
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| | - Miki Furusho
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| | - Rashmi Bansal
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| |
Collapse
|
58
|
Hou J, Bi H, Ye Z, Huang W, Zou G, Zou X, Shi YS, Shen Y, Ma Q, Kirchhoff F, Hu Y, Chen G. Pen-2 Negatively Regulates the Differentiation of Oligodendrocyte Precursor Cells into Astrocytes in the Central Nervous System. J Neurosci 2021; 41:4976-4990. [PMID: 33972402 PMCID: PMC8197633 DOI: 10.1523/jneurosci.2455-19.2021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 03/30/2021] [Accepted: 04/28/2021] [Indexed: 11/21/2022] Open
Abstract
Mutations on γ-secretase subunits are associated with neurologic diseases. Whereas the role of γ-secretase in neurogenesis has been intensively studied, little is known about its role in astrogliogenesis. Recent evidence has demonstrated that astrocytes can be generated from oligodendrocyte precursor cells (OPCs). However, it is not well understood what mechanism may control OPCs to differentiate into astrocytes. To address the above questions, we generated two independent lines of oligodendrocyte lineage-specific presenilin enhancer 2 (Pen-2) conditional KO mice. Both male and female mice were used. Here we demonstrate that conditional inactivation of Pen-2 mediated by Olig1-Cre or NG2-CreERT2 causes enhanced generation of astrocytes. Lineage-tracing experiments indicate that abnormally generated astrocytes are derived from Cre-expressing OPCs in the CNS in Pen-2 conditional KO mice. Mechanistic analysis reveals that deletion of Pen-2 inhibits the Notch signaling to upregulate signal transducer and activator of transcription 3, which triggers activation of GFAP to promote astrocyte differentiation. Together, these novel findings indicate that Pen-2 regulates the specification of astrocytes from OPCs through the signal transducer and activator of transcription 3 signaling.SIGNIFICANCE STATEMENT Astrocytes and oligodendrocyte (OLs) play critical roles in the brain. Recent evidence has demonstrated that astrocytes can be generated from OL precursor cells (OPCs). However, it remains poorly understood what mechanism governs the differentiation of OPCs into astrocytes. In this study, we took advantage of OL lineage cells specific presenilin enhancer 2 (Pen-2) conditional KO mice. We show that deletion of Pen-2 leads to dramatically enhanced astrocyte differentiation from OPCs in the CNS. Mechanistic analysis reveals that deletion of Pen-2 inhibits Hes1 and activates signal transducer and activator of transcription 3 to trigger GFAP activation which promotes astrocyte differentiation. Overall, this study identifies a novel function of Pen-2 in astrogliogenesis from OPCs.
Collapse
Affiliation(s)
- Jinxing Hou
- State Key Laboratory of Pharmaceutical Biotechnology, MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Nanjing, 210061, China
| | - Huiru Bi
- State Key Laboratory of Pharmaceutical Biotechnology, MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Nanjing, 210061, China
| | - Zhuoyang Ye
- State Key Laboratory of Pharmaceutical Biotechnology, MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Nanjing, 210061, China
| | - Wenhui Huang
- Department of Molecular Physiology, Center for Integrative Physiology and Molecular Medicine, University of Saarland, Homburg, D-66421, Germany
| | - Gang Zou
- Department of General Surgery, Second Clinical Medical College, Shenzhen People's Hospital, Jinan University, Shenzhen, 518000, China
| | - Xiaochuan Zou
- State Key Laboratory of Pharmaceutical Biotechnology, MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Nanjing, 210061, China
| | - Yun Stone Shi
- State Key Laboratory of Pharmaceutical Biotechnology, MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Nanjing, 210061, China
| | - Ying Shen
- Department of Neurobiology, Key Laboratory of Medical Neurobiology of the Ministry of Health, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Quanhong Ma
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Institute of Neuroscience, Second Affiliated Hospital, Soochow University, Suzhou, 215123, China
| | - Frank Kirchhoff
- Department of Molecular Physiology, Center for Integrative Physiology and Molecular Medicine, University of Saarland, Homburg, D-66421, Germany
| | - Yimin Hu
- Department of Anesthesiology, Second Affiliated Changzhou People's Hospital of Nanjing Medical University, Changzhou, Jiangsu 213000, China
| | - Guiquan Chen
- State Key Laboratory of Pharmaceutical Biotechnology, MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Nanjing, 210061, China
| |
Collapse
|
59
|
Chua JP, De Calbiac H, Kabashi E, Barmada SJ. Autophagy and ALS: mechanistic insights and therapeutic implications. Autophagy 2021; 18:254-282. [PMID: 34057020 PMCID: PMC8942428 DOI: 10.1080/15548627.2021.1926656] [Citation(s) in RCA: 72] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Mechanisms of protein homeostasis are crucial for overseeing the clearance of misfolded and toxic proteins over the lifetime of an organism, thereby ensuring the health of neurons and other cells of the central nervous system. The highly conserved pathway of autophagy is particularly necessary for preventing and counteracting pathogenic insults that may lead to neurodegeneration. In line with this, mutations in genes that encode essential autophagy factors result in impaired autophagy and lead to neurodegenerative conditions such as amyotrophic lateral sclerosis (ALS). However, the mechanistic details underlying the neuroprotective role of autophagy, neuronal resistance to autophagy induction, and the neuron-specific effects of autophagy-impairing mutations remain incompletely defined. Further, the manner and extent to which non-cell autonomous effects of autophagy dysfunction contribute to ALS pathogenesis are not fully understood. Here, we review the current understanding of the interplay between autophagy and ALS pathogenesis by providing an overview of critical steps in the autophagy pathway, with special focus on pivotal factors impaired by ALS-causing mutations, their physiologic effects on autophagy in disease models, and the cell type-specific mechanisms regulating autophagy in non-neuronal cells which, when impaired, can contribute to neurodegeneration. This review thereby provides a framework not only to guide further investigations of neuronal autophagy but also to refine therapeutic strategies for ALS and related neurodegenerative diseases.Abbreviations: ALS: amyotrophic lateral sclerosis; Atg: autophagy-related; CHMP2B: charged multivesicular body protein 2B; DPR: dipeptide repeat; FTD: frontotemporal dementia; iPSC: induced pluripotent stem cell; LIR: LC3-interacting region; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MTOR: mechanistic target of rapamycin kinase; PINK1: PTEN induced kinase 1; RNP: ribonuclear protein; sALS: sporadic ALS; SPHK1: sphingosine kinase 1; TARDBP/TDP-43: TAR DNA binding protein; TBK1: TANK-binding kinase 1; TFEB: transcription factor EB; ULK: unc-51 like autophagy activating kinase; UPR: unfolded protein response; UPS: ubiquitin-proteasome system; VCP: valosin containing protein.
Collapse
Affiliation(s)
- Jason P Chua
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
| | - Hortense De Calbiac
- Recherche translationnelle sur les maladies neurologiques, Institut Imagine, UMR-1163 INSERM et Université Paris Descartes, Hôpital Universitaire Necker-Enfants Malades, Paris, France
| | - Edor Kabashi
- Recherche translationnelle sur les maladies neurologiques, Institut Imagine, UMR-1163 INSERM et Université Paris Descartes, Hôpital Universitaire Necker-Enfants Malades, Paris, France
| | - Sami J Barmada
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
60
|
Saiki T, Nakamura N, Miyabe M, Ito M, Minato T, Sango K, Matsubara T, Naruse K. The Effects of Insulin on Immortalized Rat Schwann Cells, IFRS1. Int J Mol Sci 2021; 22:ijms22115505. [PMID: 34071138 PMCID: PMC8197103 DOI: 10.3390/ijms22115505] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 05/18/2021] [Accepted: 05/19/2021] [Indexed: 01/02/2023] Open
Abstract
Schwann cells play an important role in peripheral nerve function, and their dysfunction has been implicated in the pathogenesis of diabetic neuropathy and other demyelinating diseases. The physiological functions of insulin in Schwann cells remain unclear and therefore define the aim of this study. By using immortalized adult Fischer rat Schwann cells (IFRS1), we investigated the mechanism of the stimulating effects of insulin on the cell proliferation and expression of myelin proteins (myelin protein zero (MPZ) and myelin basic protein (MBP). The application of insulin to IFRS1 cells increased the proliferative activity and induced phosphorylation of Akt and ERK, but not P38-MAPK. The proliferative potential of insulin-stimulated IFRS1 was significantly suppressed by the addition of LY294002, a PI3 kinase inhibitor. The insulin-stimulated increase in MPZ expression was significantly suppressed by the addition of PD98059, a MEK inhibitor. Furthermore, insulin-increased MBP expression was significantly suppressed by the addition of LY294002. These findings suggest that both PI3-K/Akt and ERK/MEK pathways are involved in insulin-induced cell growth and upregulation of MPZ and MBP in IFRS1 Schwann cells.
Collapse
Affiliation(s)
- Tomokazu Saiki
- Department of Pharmacy, Aichi Gakuin University Dental Hospital, Nagoya 464-8651, Japan;
| | - Nobuhisa Nakamura
- Department of Internal Medicine, School of Dentistry, Aichi Gakuin University, Nagoya 464-8651, Japan; (M.M.); (M.I.); (T.M.); (K.N.)
- Correspondence: ; Tel.: +81-52-759-2111; Fax: +81-52-759-2168
| | - Megumi Miyabe
- Department of Internal Medicine, School of Dentistry, Aichi Gakuin University, Nagoya 464-8651, Japan; (M.M.); (M.I.); (T.M.); (K.N.)
| | - Mizuho Ito
- Department of Internal Medicine, School of Dentistry, Aichi Gakuin University, Nagoya 464-8651, Japan; (M.M.); (M.I.); (T.M.); (K.N.)
| | - Tomomi Minato
- Department of Clinical Laboratory, Aichi Gakuin University Dental Hospital, Nagoya 464-8651, Japan;
| | - Kazunori Sango
- Diabetic Neuropathy Project, Department of Diseases and Infection, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan;
| | - Tatsuaki Matsubara
- Department of Internal Medicine, School of Dentistry, Aichi Gakuin University, Nagoya 464-8651, Japan; (M.M.); (M.I.); (T.M.); (K.N.)
| | - Keiko Naruse
- Department of Internal Medicine, School of Dentistry, Aichi Gakuin University, Nagoya 464-8651, Japan; (M.M.); (M.I.); (T.M.); (K.N.)
| |
Collapse
|
61
|
Fletcher JL, Makowiecki K, Cullen CL, Young KM. Oligodendrogenesis and myelination regulate cortical development, plasticity and circuit function. Semin Cell Dev Biol 2021; 118:14-23. [PMID: 33863642 DOI: 10.1016/j.semcdb.2021.03.017] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 03/23/2021] [Accepted: 03/25/2021] [Indexed: 12/17/2022]
Abstract
During cortical development and throughout adulthood, oligodendrocytes add myelin internodes to glutamatergic projection neurons and GABAergic inhibitory neurons. In addition to directing node of Ranvier formation, to enable saltatory conduction and influence action potential transit time, oligodendrocytes support axon health by communicating with axons via the periaxonal space and providing metabolic support that is particularly critical for healthy ageing. In this review we outline the timing of oligodendrogenesis in the developing mouse and human cortex and describe the important role that oligodendrocytes play in sustaining and modulating neuronal function. We also provide insight into the known and speculative impact that myelination has on cortical axons and their associated circuits during the developmental critical periods and throughout life, particularly highlighting their life-long role in learning and remembering.
Collapse
Affiliation(s)
- Jessica L Fletcher
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia
| | - Kalina Makowiecki
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia
| | - Carlie L Cullen
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia
| | - Kaylene M Young
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia.
| |
Collapse
|
62
|
Rivera AD, Pieropan F, Chacon‐De‐La‐Rocha I, Lecca D, Abbracchio MP, Azim K, Butt AM. Functional genomic analyses highlight a shift in Gpr17-regulated cellular processes in oligodendrocyte progenitor cells and underlying myelin dysregulation in the aged mouse cerebrum. Aging Cell 2021; 20:e13335. [PMID: 33675110 PMCID: PMC8045941 DOI: 10.1111/acel.13335] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 01/18/2021] [Accepted: 02/09/2021] [Indexed: 12/14/2022] Open
Abstract
Brain ageing is characterised by a decline in neuronal function and associated cognitive deficits. There is increasing evidence that myelin disruption is an important factor that contributes to the age-related loss of brain plasticity and repair responses. In the brain, myelin is produced by oligodendrocytes, which are generated throughout life by oligodendrocyte progenitor cells (OPCs). Currently, a leading hypothesis points to ageing as a major reason for the ultimate breakdown of remyelination in Multiple Sclerosis (MS). However, an incomplete understanding of the cellular and molecular processes underlying brain ageing hinders the development of regenerative strategies. Here, our combined systems biology and neurobiological approach demonstrate that oligodendroglial and myelin genes are amongst the most altered in the ageing mouse cerebrum. This was underscored by the identification of causal links between signalling pathways and their downstream transcriptional networks that define oligodendroglial disruption in ageing. The results highlighted that the G-protein coupled receptor Gpr17 is central to the disruption of OPCs in ageing and this was confirmed by genetic fate-mapping and cellular analyses. Finally, we used systems biology strategies to identify therapeutic agents that rejuvenate OPCs and restore myelination in age-related neuropathological contexts.
Collapse
Affiliation(s)
- Andrea D. Rivera
- School of Pharmacy and Biomedical ScienceUniversity of PortsmouthPortsmouthUK
- Department of NeuroscienceInstitute of Human AnatomyUniversity of PaduaPaduaItaly
| | - Francesca Pieropan
- School of Pharmacy and Biomedical ScienceUniversity of PortsmouthPortsmouthUK
| | | | - Davide Lecca
- Department of Pharmaceutical SciencesUniversity of MilanMilanItaly
| | | | - Kasum Azim
- Department of NeurologyNeuroregenerationMedical FacultyHeinrich‐Heine‐UniversityDüsseldorfGermany
| | - Arthur M. Butt
- School of Pharmacy and Biomedical ScienceUniversity of PortsmouthPortsmouthUK
| |
Collapse
|
63
|
Ratti S, Rusciano I, Mongiorgi S, Owusu Obeng E, Cappellini A, Teti G, Falconi M, Talozzi L, Capellari S, Bartoletti-Stella A, Guaraldi P, Cortelli P, Suh PG, Cocco L, Manzoli L, Ramazzotti G. Cell signaling pathways in autosomal-dominant leukodystrophy (ADLD): the intriguing role of the astrocytes. Cell Mol Life Sci 2021; 78:2781-2795. [PMID: 33034697 PMCID: PMC8004488 DOI: 10.1007/s00018-020-03661-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 09/02/2020] [Accepted: 09/28/2020] [Indexed: 12/17/2022]
Abstract
Autosomal-dominant leukodystrophy (ADLD) is a rare fatal neurodegenerative disorder with overexpression of the nuclear lamina component, Lamin B1 due to LMNB1 gene duplication or deletions upstream of the gene. The molecular mechanisms responsible for driving the onset and development of this pathology are not clear yet. Vacuolar demyelination seems to be one of the most significant histopathological observations of ADLD. Considering the role of oligodendrocytes, astrocytes, and leukemia inhibitory factor (LIF)-activated signaling pathways in the myelination processes, this work aims to analyze the specific alterations in different cell populations from patients with LMNB1 duplications and engineered cellular models overexpressing Lamin B1 protein. Our results point out, for the first time, that astrocytes may be pivotal in the evolution of the disease. Indeed, cells from ADLD patients and astrocytes overexpressing LMNB1 show severe ultrastructural nuclear alterations, not present in oligodendrocytes overexpressing LMNB1. Moreover, the accumulation of Lamin B1 in astrocytes induces a reduction in LIF and in LIF-Receptor (LIF-R) levels with a consequential decrease in LIF secretion. Therefore, in both our cellular models, Jak/Stat3 and PI3K/Akt axes, downstream of LIF/LIF-R, are downregulated. Significantly, the administration of exogenous LIF can partially reverse the toxic effects induced by Lamin B1 accumulation with differences between astrocytes and oligodendrocytes, highlighting that LMNB1 overexpression drastically affects astrocytic function reducing their fundamental support to oligodendrocytes in the myelination process. In addition, inflammation has also been investigated, showing an increased activation in ADLD patients' cells.
Collapse
Affiliation(s)
- Stefano Ratti
- Cellular Signalling Laboratory, Department of Biomedical and NeuroMotor Sciences (DIBINEM), University of Bologna, Bologna, Italy
| | - Isabella Rusciano
- Cellular Signalling Laboratory, Department of Biomedical and NeuroMotor Sciences (DIBINEM), University of Bologna, Bologna, Italy
| | - Sara Mongiorgi
- Cellular Signalling Laboratory, Department of Biomedical and NeuroMotor Sciences (DIBINEM), University of Bologna, Bologna, Italy
| | - Eric Owusu Obeng
- Cellular Signalling Laboratory, Department of Biomedical and NeuroMotor Sciences (DIBINEM), University of Bologna, Bologna, Italy
| | - Alessandra Cappellini
- Cellular Signalling Laboratory, Department of Biomedical and NeuroMotor Sciences (DIBINEM), University of Bologna, Bologna, Italy
| | - Gabriella Teti
- Department of Biomedical and NeuroMotor Sciences (DIBINEM), University of Bologna, Via Irnerio 48, Bologna, Italy
| | - Mirella Falconi
- Department of Biomedical and NeuroMotor Sciences (DIBINEM), University of Bologna, Via Irnerio 48, Bologna, Italy
| | - Lia Talozzi
- Functional MR Unit, Department of Biomedical and NeuroMotor Sciences (DIBINEM), University of Bologna, Bologna, Italy
| | - Sabina Capellari
- Department of Biomedical and NeuroMotor Sciences (DIBINEM), University of Bologna, Bologna, Italy
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC NeuroMet, Bologna, Italy
| | | | - Pietro Guaraldi
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC NeuroMet, Bologna, Italy
| | - Pietro Cortelli
- Department of Biomedical and NeuroMotor Sciences (DIBINEM), University of Bologna, Bologna, Italy
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC NeuroMet, Bologna, Italy
| | - Pann-Ghill Suh
- Korea Brain Research Institute, Daegu, Republic of Korea
- School of Life Sciences, UNIST, Ulsan, Republic of Korea
| | - Lucio Cocco
- Cellular Signalling Laboratory, Department of Biomedical and NeuroMotor Sciences (DIBINEM), University of Bologna, Bologna, Italy.
| | - Lucia Manzoli
- Cellular Signalling Laboratory, Department of Biomedical and NeuroMotor Sciences (DIBINEM), University of Bologna, Bologna, Italy.
| | - Giulia Ramazzotti
- Cellular Signalling Laboratory, Department of Biomedical and NeuroMotor Sciences (DIBINEM), University of Bologna, Bologna, Italy
| |
Collapse
|
64
|
Mühlebner A, van Scheppingen J, de Neef A, Bongaarts A, Zimmer TS, Mills JD, Jansen FE, Spliet WGM, Krsek P, Zamecnik J, Coras R, Blumcke I, Feucht M, Scholl T, Gruber VE, Hainfellner JA, Söylemezoğlu F, Kotulska K, Lagae L, Jansen AC, Kwiatkowski DJ, Jozwiak S, Curatolo P, Aronica E. Myelin Pathology Beyond White Matter in Tuberous Sclerosis Complex (TSC) Cortical Tubers. J Neuropathol Exp Neurol 2021; 79:1054-1064. [PMID: 32954437 PMCID: PMC7559237 DOI: 10.1093/jnen/nlaa090] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Tuberous sclerosis complex (TSC) is a monogenetic disease that arises due to mutations in either the TSC1 or TSC2 gene and affects multiple organ systems. One of the hallmark manifestations of TSC are cortical malformations referred to as cortical tubers. These tubers are frequently associated with treatment-resistant epilepsy. Some of these patients are candidates for epilepsy surgery. White matter abnormalities, such as loss of myelin and oligodendroglia, have been described in a small subset of resected tubers but mechanisms underlying this phenomenon are unclear. Herein, we analyzed a variety of neuropathologic and immunohistochemical features in gray and white matter areas of resected cortical tubers from 46 TSC patients using semi-automated quantitative image analysis. We observed divergent amounts of myelin basic protein as well as numbers of oligodendroglia in both gray and white matter when compared with matched controls. Analyses of clinical data indicated that reduced numbers of oligodendroglia were associated with lower numbers on the intelligence quotient scale and that lower amounts of myelin-associated oligodendrocyte basic protein were associated with the presence of autism-spectrum disorder. In conclusion, myelin pathology in cortical tubers extends beyond the white matter and may be linked to cognitive dysfunction in TSC patients.
Collapse
Affiliation(s)
- Angelika Mühlebner
- Department of (Neuro)Pathology, Amsterdam Neuroscience, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Jackelien van Scheppingen
- Department of (Neuro)Pathology, Amsterdam Neuroscience, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Andrew de Neef
- Department of (Neuro)Pathology, Amsterdam Neuroscience, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Anika Bongaarts
- Department of (Neuro)Pathology, Amsterdam Neuroscience, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Till S Zimmer
- Department of (Neuro)Pathology, Amsterdam Neuroscience, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - James D Mills
- Department of (Neuro)Pathology, Amsterdam Neuroscience, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Floor E Jansen
- Department of Pediatric Neurology, Brain Center University Medical Center
| | - Wim G M Spliet
- Department of Pathology, University Medical Center Utrecht (WGMS) Utrecht, The Netherlands
| | | | | | - Roland Coras
- Second Faculty of Medicine, Charles University, Motol University Hospital, Prague, Czech Republic; Department of Neuropathology, University Hospital Erlangen, Erlangen, Germany
| | - Ingmar Blumcke
- Second Faculty of Medicine, Charles University, Motol University Hospital, Prague, Czech Republic; Department of Neuropathology, University Hospital Erlangen, Erlangen, Germany
| | | | | | | | | | - Figen Söylemezoğlu
- Medical University of Vienna, Vienna, Austria; Department of Pathology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Katarzyna Kotulska
- Department of Neurology and Epileptology, The Children's Memorial Health Institute, Warsaw, Poland
| | - Lieven Lagae
- Department of Development and Regeneration-Section Pediatric Neurology, University Hospitals KU Leuven, Leuven
| | - Anna C Jansen
- Pediatric Neurology Unit-UZ Brussel, Brussels Belgium
| | | | - Sergiusz Jozwiak
- Department of Neurology and Epileptology, The Children's Memorial Health Institute.,Department of Child Neurology, Medical University of Warsaw Warsaw, Poland
| | - Paolo Curatolo
- Child Neurology and Psychiatry Unit, Systems Medicine Department, Tor Vergata University, Rome, Italy
| | - Eleonora Aronica
- Department of (Neuro)Pathology, Amsterdam Neuroscience, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Stichting Epilepsie Instellingen Nederland (SEIN), Heemstede, The Netherlands
| |
Collapse
|
65
|
Comprehensive Analysis of Age-related Changes in Lipid Metabolism and Myelin Sheath Formation in Sciatic Nerves. J Mol Neurosci 2021; 71:2310-2323. [PMID: 33492614 DOI: 10.1007/s12031-020-01768-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 11/30/2020] [Indexed: 02/06/2023]
Abstract
To investigate the molecular changes related to myelin formation and lipid metabolism in the sciatic nerve in Sprague Dawley (SD) rats during aging. Thirty-six healthy male SD rats were divided into five groups according to age: 1 week, 1 month, 6 months, 12 months, and 24 months. Sciatic nerves were collected from 1-month-old and 24-month-old SD rats (n = 3) to perform next-generation sequencing (NGS) and bioinformatics analysis. Specimens from each group were harvested and analyzed by qPCR, Western blotting, and transmission electron microscopy (TEM). Protein-protein interaction (PPI) networks of differentially expressed mRNAs (DEmRNAs) related to myelin and lipid metabolism were constructed. DEmRNAs in subnetworks were verified using qPCR. A total of 4580 DEmRNAs were found during aging. The top enriched GO biological processes were primarily clustered in cholesterol and lipid metabolism, including the cholesterol biosynthetic process (RF = 3.16), sterol biosynthetic process (RF = 3.03), cholesterol metabolic process (RF = 2.15), sterol metabolic process (RF = 2.11), fatty acid biosynthetic process (RF = 2.09), and lipid biosynthetic process (RF = 1.79). The mRNA levels of MBP, PMP22, and MPZ were downregulated during aging, while the protein expression of MBP showed an increasing trend. The TEM results showed thin myelin sheaths and an increased number of unmyelinated axons in the 1-week-old rats, and the sheaths became thickened with degenerated axons appearing in older animals. Forty PPI subnetworks related to lipid metabolism were constructed, including one primary subnetwork and two smaller subnetworks. The hub genes were mTOR in sub-network 1, Akt1 in sub-network 2, and SIRT1 in sub-network 3. No gene expression was found consistent with the sequencing results, while in the downregulated genes, AKT1, CEBPA, LIPE, LRP5, PHB, and Rara were significantly downregulated in 24-month-old rats. Lipid metabolism might play an important role in maintaining the structure and physiological function in sciatic nerves during aging and could be candidates for nerve aging research.
Collapse
|
66
|
de Curtis M, Garbelli R, Uva L. A hypothesis for the role of axon demyelination in seizure generation. Epilepsia 2021; 62:583-595. [PMID: 33493363 DOI: 10.1111/epi.16824] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 01/04/2021] [Accepted: 01/05/2021] [Indexed: 01/06/2023]
Abstract
Loss of myelin and altered oligodendrocyte distribution in the cerebral cortex are commonly observed both in postsurgical tissue derived from different focal epilepsies (such as focal cortical dysplasias and tuberous sclerosis) and in animal models of focal epilepsy. Moreover, seizures are a frequent symptom in demyelinating diseases, such as multiple sclerosis, and in animal models of demyelination and oligodendrocyte dysfunction. Finally, the excessive activity reported in demyelinated axons may promote hyperexcitability. We hypothesize that the extracellular potassium rise generated during epileptiform activity may be amplified by the presence of axons without appropriate myelin coating and by alterations in oligodendrocyte function. This process could facilitate the triggering of recurrent spontaneous seizures in areas of altered myelination and could result in further demyelination, thus promoting epileptogenesis.
Collapse
Affiliation(s)
- Marco de Curtis
- Epilepsy Unit, IRCCS Foundation Carlo Besta Neurological Institute, Milan, Italy
| | - Rita Garbelli
- Epilepsy Unit, IRCCS Foundation Carlo Besta Neurological Institute, Milan, Italy
| | - Laura Uva
- Epilepsy Unit, IRCCS Foundation Carlo Besta Neurological Institute, Milan, Italy
| |
Collapse
|
67
|
Krzyżanowska M, Rębała K, Steiner J, Kaliszan M, Pieśniak D, Karnecki K, Wiergowski M, Brisch R, Braun K, Jankowski Z, Kosmowska M, Chociej J, Gos T. Reduced ribosomal DNA transcription in the prefrontal cortex of suicide victims: consistence of new molecular RT-qPCR findings with previous morphometric data from AgNOR-stained pyramidal neurons. Eur Arch Psychiatry Clin Neurosci 2021; 271:567-576. [PMID: 33501518 PMCID: PMC7981327 DOI: 10.1007/s00406-021-01232-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 01/11/2021] [Indexed: 12/12/2022]
Abstract
Prefrontal cortical regions play a key role in behavioural regulation, which is profoundly disturbed in suicide. The study was carried out on frozen cortical samples from the anterior cingulate cortex (dorsal and ventral parts, ACd and ACv), the orbitofrontal cortex (OFC), and the dorsolateral cortex (DLC) obtained from 20 suicide completers (predominantly violent) with unknown psychiatric diagnosis and 21 non-suicidal controls. The relative level of ribosomal RNA (rRNA) as a marker of the transcriptional activity of ribosomal DNA (rDNA) was evaluated bilaterally in prefrontal regions mentioned above (i.e. in eight regions of interest, ROIs) by reverse transcription and quantitative polymerase chain reaction (RT-qPCR). The overall statistical analysis revealed a decrease in rDNA activity in suicide victims versus controls, particularly in male subjects. Further ROI-specific post hoc analyses revealed a significant decrease in this activity in suicides compared to non-suicides in five ROIs. This effect was accentuated in the ACv, where it was observed bilaterally. Our findings suggest that decreased rDNA transcription in the prefrontal cortex plays an important role in suicide pathogenesis and corresponds with our previous morphometric analyses of AgNOR-stained neurons.
Collapse
Affiliation(s)
- Marta Krzyżanowska
- grid.11451.300000 0001 0531 3426Department of Forensic Medicine, Medical University of Gdańsk, ul. Dębowa 23, 80-204 Gdańsk, Poland
| | - Krzysztof Rębała
- grid.11451.300000 0001 0531 3426Department of Forensic Medicine, Medical University of Gdańsk, ul. Dębowa 23, 80-204 Gdańsk, Poland
| | - Johann Steiner
- grid.5807.a0000 0001 1018 4307Department of Psychiatry, Otto von Guericke University, Magdeburg, Germany
| | - Michał Kaliszan
- grid.11451.300000 0001 0531 3426Department of Forensic Medicine, Medical University of Gdańsk, ul. Dębowa 23, 80-204 Gdańsk, Poland
| | - Dorota Pieśniak
- grid.11451.300000 0001 0531 3426Department of Forensic Medicine, Medical University of Gdańsk, ul. Dębowa 23, 80-204 Gdańsk, Poland
| | - Karol Karnecki
- grid.11451.300000 0001 0531 3426Department of Forensic Medicine, Medical University of Gdańsk, ul. Dębowa 23, 80-204 Gdańsk, Poland
| | - Marek Wiergowski
- grid.11451.300000 0001 0531 3426Department of Forensic Medicine, Medical University of Gdańsk, ul. Dębowa 23, 80-204 Gdańsk, Poland
| | - Ralf Brisch
- grid.11451.300000 0001 0531 3426Department of Forensic Medicine, Medical University of Gdańsk, ul. Dębowa 23, 80-204 Gdańsk, Poland
| | - Katharina Braun
- grid.5807.a0000 0001 1018 4307Department of Zoology/Developmental Neurobiology, Institute of Biology, Otto von Guericke University, Magdeburg, Germany
| | - Zbigniew Jankowski
- grid.11451.300000 0001 0531 3426Department of Forensic Medicine, Medical University of Gdańsk, ul. Dębowa 23, 80-204 Gdańsk, Poland
| | - Monika Kosmowska
- grid.11451.300000 0001 0531 3426Department of Forensic Medicine, Medical University of Gdańsk, ul. Dębowa 23, 80-204 Gdańsk, Poland
| | - Joanna Chociej
- grid.11451.300000 0001 0531 3426Department of Forensic Medicine, Medical University of Gdańsk, ul. Dębowa 23, 80-204 Gdańsk, Poland
| | - Tomasz Gos
- Department of Forensic Medicine, Medical University of Gdańsk, ul. Dębowa 23, 80-204, Gdańsk, Poland. .,Department of Psychiatry, Otto von Guericke University, Magdeburg, Germany. .,Department of Zoology/Developmental Neurobiology, Institute of Biology, Otto von Guericke University, Magdeburg, Germany.
| |
Collapse
|
68
|
A Golgi-associated lipid kinase controls peripheral nerve myelination. Proc Natl Acad Sci U S A 2020; 117:30873-30875. [PMID: 33188090 DOI: 10.1073/pnas.2021130117] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
69
|
Affiliation(s)
- Amelia Trimarco
- INSPE, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| | - Carla Taveggia
- INSPE, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
70
|
Wang H, Liu M, Zou G, Wang L, Duan W, He X, Ji M, Zou X, Hu Y, Yang J, Chen G. Deletion of PDK1 in oligodendrocyte lineage cells causes white matter abnormality and myelination defect in the central nervous system. Neurobiol Dis 2020; 148:105212. [PMID: 33276084 DOI: 10.1016/j.nbd.2020.105212] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 11/17/2020] [Accepted: 11/29/2020] [Indexed: 12/01/2022] Open
Abstract
PDK1 (3-Phosphoinositide dependent protein kinase-1) is a member in the PI3K (phosphatidylinositol 3 kinase) pathway and is implicated in neurodevelopmental disease with microcephaly. Although the role of PDK1 in neurogenesis has been broadly studied, it remains unknown how PDK1 may regulate oligogenesis in the central nervous system (CNS). To address this question, we generated oligodendrocyte (OL) lineage cells specific PDK1 conditional knockout (cKO) mice. We find that PDK1 cKOs display abnormal white matter (WM), massive loss of mature OLs and severe defect in myelination in the CNS. In contrast, these mutants exhibit normal neuronal development and unchanged apoptosis in the CNS. We demonstrate that deletion of PDK1 severely impairs OL differentiation. We show that genetic or pharmacological inhibition of PDK1 causes deficit in the mammalian target of rapamycin (mTor) signaling and down-regulation of Sox10. Together, these results highlight a critical role of PDK1 in OL differentiation during postnatal CNS development.
Collapse
Affiliation(s)
- He Wang
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, No. 1 East Jianshe Road, Zhengzhou 450052, China
| | - Mengjia Liu
- State Key Laboratory of Pharmaceutical Biotechnology, MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, 12 Xuefu Avenue, Nanjing, Jiangsu Province 210061, China
| | - Gang Zou
- Department of Anesthesiology, The Second Affiliated Hospital, Nanjing Medical University, 121 Jiangjiayuan Avenue, Nanjing, Jiangsu Province 210003, China
| | - Long Wang
- State Key Laboratory of Pharmaceutical Biotechnology, MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, 12 Xuefu Avenue, Nanjing, Jiangsu Province 210061, China
| | - Wenbin Duan
- Department of General Surgery, The Second Clinical Medical College, Shenzhen People's Hospital, Jinan University, Shenzhen 518000, China
| | - Xue He
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, No. 1 East Jianshe Road, Zhengzhou 450052, China
| | - Muhuo Ji
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, No. 1 East Jianshe Road, Zhengzhou 450052, China
| | - Xiaochuan Zou
- State Key Laboratory of Pharmaceutical Biotechnology, MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, 12 Xuefu Avenue, Nanjing, Jiangsu Province 210061, China
| | - Yimin Hu
- Department of General Surgery, The Second Clinical Medical College, Shenzhen People's Hospital, Jinan University, Shenzhen 518000, China.
| | - Jianjun Yang
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, No. 1 East Jianshe Road, Zhengzhou 450052, China.
| | - Guiquan Chen
- State Key Laboratory of Pharmaceutical Biotechnology, MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, 12 Xuefu Avenue, Nanjing, Jiangsu Province 210061, China.
| |
Collapse
|
71
|
Environmental and Nutritional "Stressors" and Oligodendrocyte Dysfunction: Role of Mitochondrial and Endoplasmatic Reticulum Impairment. Biomedicines 2020; 8:biomedicines8120553. [PMID: 33265917 PMCID: PMC7760976 DOI: 10.3390/biomedicines8120553] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 11/25/2020] [Accepted: 11/27/2020] [Indexed: 12/13/2022] Open
Abstract
Oligodendrocytes are myelinating cells of the central nervous system which are generated by progenitor oligodendrocytes as a result of maturation processes. The main function of mature oligodendrocytes is to produce myelin, a lipid-rich multi-lamellar membrane that wraps tightly around neuronal axons, insulating them and facilitating nerve conduction through saltatory propagation. The myelination process requires the consumption a large amount of energy and a high metabolic turnover. Mitochondria are essential organelles which regulate many cellular functions, including energy production through oxidative phosphorylation. Any mitochondrial dysfunction impacts cellular metabolism and negatively affects the health of the organism. If the functioning of the mitochondria is unbalanced, the myelination process is impaired. When myelination has finished, oligodendrocyte will have synthesized about 40% of the total lipids present in the brain. Since lipid synthesis occurs in the cellular endoplasmic reticulum, the dysfunction of this organelle can lead to partial or deficient myelination, triggering numerous neurodegenerative diseases. In this review, the induced malfunction of oligodendrocytes by harmful exogenous stimuli has been outlined. In particular, the effects of alcohol consumption and heavy metal intake are discussed. Furthermore, the response of the oligodendrocyte to excessive mitochondrial oxidative stress and to the altered regulation of the functioning of the endoplasmic reticulum will be explored.
Collapse
|
72
|
Insights into Potential Targets for Therapeutic Intervention in Epilepsy. Int J Mol Sci 2020; 21:ijms21228573. [PMID: 33202963 PMCID: PMC7697405 DOI: 10.3390/ijms21228573] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/04/2020] [Accepted: 11/11/2020] [Indexed: 02/06/2023] Open
Abstract
Epilepsy is a chronic brain disease that affects approximately 65 million people worldwide. However, despite the continuous development of antiepileptic drugs, over 30% patients with epilepsy progress to drug-resistant epilepsy. For this reason, it is a high priority objective in preclinical research to find novel therapeutic targets and to develop effective drugs that prevent or reverse the molecular mechanisms underlying epilepsy progression. Among these potential therapeutic targets, we highlight currently available information involving signaling pathways (Wnt/β-catenin, Mammalian Target of Rapamycin (mTOR) signaling and zinc signaling), enzymes (carbonic anhydrase), proteins (erythropoietin, copine 6 and complement system), channels (Transient Receptor Potential Vanilloid Type 1 (TRPV1) channel) and receptors (galanin and melatonin receptors). All of them have demonstrated a certain degree of efficacy not only in controlling seizures but also in displaying neuroprotective activity and in modifying the progression of epilepsy. Although some research with these specific targets has been done in relation with epilepsy, they have not been fully explored as potential therapeutic targets that could help address the unsolved issue of drug-resistant epilepsy and develop new antiseizure therapies for the treatment of epilepsy.
Collapse
|
73
|
Alcover-Sanchez B, Garcia-Martin G, Escudero-Ramirez J, Gonzalez-Riano C, Lorenzo P, Gimenez-Cassina A, Formentini L, de la Villa-Polo P, Pereira MP, Wandosell F, Cubelos B. Absence of R-Ras1 and R-Ras2 causes mitochondrial alterations that trigger axonal degeneration in a hypomyelinating disease model. Glia 2020; 69:619-637. [PMID: 33010069 DOI: 10.1002/glia.23917] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 08/18/2020] [Accepted: 09/21/2020] [Indexed: 12/11/2022]
Abstract
Fast synaptic transmission in vertebrates is critically dependent on myelin for insulation and metabolic support. Myelin is produced by oligodendrocytes (OLs) that maintain multilayered membrane compartments that wrap around axonal fibers. Alterations in myelination can therefore lead to severe pathologies such as multiple sclerosis. Given that hypomyelination disorders have complex etiologies, reproducing clinical symptoms of myelin diseases from a neurological perspective in animal models has been difficult. We recently reported that R-Ras1-/- and/or R-Ras2-/- mice, which lack GTPases essential for OL survival and differentiation processes, present different degrees of hypomyelination in the central nervous system with a compounded hypomyelination in double knockout (DKO) mice. Here, we discovered that the loss of R-Ras1 and/or R-Ras2 function is associated with aberrant myelinated axons with increased numbers of mitochondria, and a disrupted mitochondrial respiration that leads to increased reactive oxygen species levels. Consequently, aberrant myelinated axons are thinner with cytoskeletal phosphorylation patterns typical of axonal degeneration processes, characteristic of myelin diseases. Although we observed different levels of hypomyelination in a single mutant mouse, the combined loss of function in DKO mice lead to a compromised axonal integrity, triggering the loss of visual function. Our findings demonstrate that the loss of R-Ras function reproduces several characteristics of hypomyelinating diseases, and we therefore propose that R-Ras1-/- and R-Ras2-/- neurological models are valuable approaches for the study of these myelin pathologies.
Collapse
Affiliation(s)
- Berta Alcover-Sanchez
- Departamento de Biología Molecular and Centro Biología Molecular "Severo Ochoa", Universidad Autónoma de Madrid - Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Gonzalo Garcia-Martin
- Departamento de Biología Molecular and Centro Biología Molecular "Severo Ochoa", Universidad Autónoma de Madrid - Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Juan Escudero-Ramirez
- Departamento de Biología Molecular and Centro Biología Molecular "Severo Ochoa", Universidad Autónoma de Madrid - Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Carolina Gonzalez-Riano
- CEMBIO (Centre for Metabolomics and Bioanalysis), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - Paz Lorenzo
- CEMBIO (Centre for Metabolomics and Bioanalysis), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - Alfredo Gimenez-Cassina
- Departamento de Biología Molecular and Centro Biología Molecular "Severo Ochoa", Universidad Autónoma de Madrid - Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Laura Formentini
- Departamento de Biología Molecular and Centro Biología Molecular "Severo Ochoa", Universidad Autónoma de Madrid - Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Pedro de la Villa-Polo
- Departamento de Biología de Sistemas, Universidad de Alcalá, Madrid, Spain.,Grupo de Neurofisiología Visual, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Marta P Pereira
- Departamento de Biología Molecular and Centro Biología Molecular "Severo Ochoa", Universidad Autónoma de Madrid - Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Francisco Wandosell
- Departamento de Biología Molecular and Centro Biología Molecular "Severo Ochoa", Universidad Autónoma de Madrid - Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Beatriz Cubelos
- Departamento de Biología Molecular and Centro Biología Molecular "Severo Ochoa", Universidad Autónoma de Madrid - Consejo Superior de Investigaciones Científicas, Madrid, Spain
| |
Collapse
|
74
|
Huerga-Gómez A, Aguado T, Sánchez-de la Torre A, Bernal-Chico A, Matute C, Mato S, Guzmán M, Galve-Roperh I, Palazuelos J. Δ 9 -Tetrahydrocannabinol promotes oligodendrocyte development and CNS myelination in vivo. Glia 2020; 69:532-545. [PMID: 32956517 PMCID: PMC7821226 DOI: 10.1002/glia.23911] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 09/03/2020] [Accepted: 09/04/2020] [Indexed: 12/20/2022]
Abstract
Δ9‐Tetrahydrocannabinol (THC), the main bioactive compound found in the plant Cannabis sativa, exerts its effects by activating cannabinoid receptors present in many neural cells. Cannabinoid receptors are also physiologically engaged by endogenous cannabinoid compounds, the so‐called endocannabinoids. Specifically, the endocannabinoid 2‐arachidonoylglycerol has been highlighted as an important modulator of oligodendrocyte (OL) development at embryonic stages and in animal models of demyelination. However, the potential impact of THC exposure on OL lineage progression during the critical periods of postnatal myelination has never been explored. Here, we show that acute THC administration at early postnatal ages in mice enhanced OL development and CNS myelination in the subcortical white matter by promoting oligodendrocyte precursor cell cycle exit and differentiation. Mechanistically, THC‐induced‐myelination was mediated by CB1 and CB2 cannabinoid receptors, as demonstrated by the blockade of THC actions by selective receptor antagonists. Moreover, the THC‐mediated modulation of oligodendroglial differentiation relied on the activation of the mammalian target of rapamycin complex 1 (mTORC1) signaling pathway, as mTORC1 pharmacological inhibition prevented the THC effects. Our study identifies THC as an effective pharmacological strategy to enhance oligodendrogenesis and CNS myelination in vivo.
Collapse
Affiliation(s)
- Alba Huerga-Gómez
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Department of Biochemistry and Molecular Biology and Instituto Universitario de Investigación en Neuroquímica (IUIN), Complutense University, Madrid, Spain
| | - Tania Aguado
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Department of Biochemistry and Molecular Biology and Instituto Universitario de Investigación en Neuroquímica (IUIN), Complutense University, Madrid, Spain
| | - Aníbal Sánchez-de la Torre
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Department of Biochemistry and Molecular Biology and Instituto Universitario de Investigación en Neuroquímica (IUIN), Complutense University, Madrid, Spain
| | - Ana Bernal-Chico
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Department of Neurosciences, University of the Basque Country UPV/EHU, Leioa, Spain.,Achucarro Basque Center for Neuroscience, Leioa, Spain
| | - Carlos Matute
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Department of Neurosciences, University of the Basque Country UPV/EHU, Leioa, Spain.,Achucarro Basque Center for Neuroscience, Leioa, Spain
| | - Susana Mato
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Department of Neurosciences, University of the Basque Country UPV/EHU, Leioa, Spain.,Achucarro Basque Center for Neuroscience, Leioa, Spain.,Biocruces Bizkaia, Multiple Sclerosis and Other Demyelinating Diseases Unit, Barakaldo, Spain
| | - Manuel Guzmán
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Department of Biochemistry and Molecular Biology and Instituto Universitario de Investigación en Neuroquímica (IUIN), Complutense University, Madrid, Spain
| | - Ismael Galve-Roperh
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Department of Biochemistry and Molecular Biology and Instituto Universitario de Investigación en Neuroquímica (IUIN), Complutense University, Madrid, Spain
| | - Javier Palazuelos
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Department of Biochemistry and Molecular Biology and Instituto Universitario de Investigación en Neuroquímica (IUIN), Complutense University, Madrid, Spain
| |
Collapse
|
75
|
Zimmer TS, Broekaart DWM, Gruber VE, van Vliet EA, Mühlebner A, Aronica E. Tuberous Sclerosis Complex as Disease Model for Investigating mTOR-Related Gliopathy During Epileptogenesis. Front Neurol 2020; 11:1028. [PMID: 33041976 PMCID: PMC7527496 DOI: 10.3389/fneur.2020.01028] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Accepted: 08/06/2020] [Indexed: 12/13/2022] Open
Abstract
Tuberous sclerosis complex (TSC) represents the prototypic monogenic disorder of the mammalian target of rapamycin (mTOR) pathway dysregulation. It provides the rational mechanistic basis of a direct link between gene mutation and brain pathology (structural and functional abnormalities) associated with a complex clinical phenotype including epilepsy, autism, and intellectual disability. So far, research conducted in TSC has been largely neuron-oriented. However, the neuropathological hallmarks of TSC and other malformations of cortical development also include major morphological and functional changes in glial cells involving astrocytes, oligodendrocytes, NG2 glia, and microglia. These cells and their interglial crosstalk may offer new insights into the common neurobiological mechanisms underlying epilepsy and the complex cognitive and behavioral comorbidities that are characteristic of the spectrum of mTOR-associated neurodevelopmental disorders. This review will focus on the role of glial dysfunction, the interaction between glia related to mTOR hyperactivity, and its contribution to epileptogenesis in TSC. Moreover, we will discuss how understanding glial abnormalities in TSC might give valuable insight into the pathophysiological mechanisms that could help to develop novel therapeutic approaches for TSC or other pathologies characterized by glial dysfunction and acquired mTOR hyperactivation.
Collapse
Affiliation(s)
- Till S Zimmer
- Department of (Neuro)Pathology, Amsterdam Neuroscience, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | - Diede W M Broekaart
- Department of (Neuro)Pathology, Amsterdam Neuroscience, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | | | - Erwin A van Vliet
- Department of (Neuro)Pathology, Amsterdam Neuroscience, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands.,Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam, Netherlands
| | - Angelika Mühlebner
- Department of (Neuro)Pathology, Amsterdam Neuroscience, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | - Eleonora Aronica
- Department of (Neuro)Pathology, Amsterdam Neuroscience, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands.,Stichting Epilepsie Instellingen Nederland (SEIN), Heemstede, Netherlands
| |
Collapse
|
76
|
The neuromuscular junction is a focal point of mTORC1 signaling in sarcopenia. Nat Commun 2020; 11:4510. [PMID: 32908143 PMCID: PMC7481251 DOI: 10.1038/s41467-020-18140-1] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 08/05/2020] [Indexed: 12/21/2022] Open
Abstract
With human median lifespan extending into the 80s in many developed countries, the societal burden of age-related muscle loss (sarcopenia) is increasing. mTORC1 promotes skeletal muscle hypertrophy, but also drives organismal aging. Here, we address the question of whether mTORC1 activation or suppression is beneficial for skeletal muscle aging. We demonstrate that chronic mTORC1 inhibition with rapamycin is overwhelmingly, but not entirely, positive for aging mouse skeletal muscle, while genetic, muscle fiber-specific activation of mTORC1 is sufficient to induce molecular signatures of sarcopenia. Through integration of comprehensive physiological and extensive gene expression profiling in young and old mice, and following genetic activation or pharmacological inhibition of mTORC1, we establish the phenotypically-backed, mTORC1-focused, multi-muscle gene expression atlas, SarcoAtlas (https://sarcoatlas.scicore.unibas.ch/), as a user-friendly gene discovery tool. We uncover inter-muscle divergence in the primary drivers of sarcopenia and identify the neuromuscular junction as a focal point of mTORC1-driven muscle aging.
Collapse
|
77
|
Montani L. Lipids in regulating oligodendrocyte structure and function. Semin Cell Dev Biol 2020; 112:114-122. [PMID: 32912639 DOI: 10.1016/j.semcdb.2020.07.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 07/17/2020] [Accepted: 07/29/2020] [Indexed: 12/15/2022]
Abstract
Oligodendrocytes enwrap central nervous system axons with myelin, a lipid enriched highly organized multi-layer membrane structure that allows for fast long-distance saltatory conduction of neuronal impulses. Myelin has an extremely high lipid content (∼80 % of its dry weight) and a peculiar lipid composition, with a 2:2:1 cholesterol:phospholipid:glycolipid ratio. Inherited neurodegenerative diseases of the lipids (caused by mutations in lipogenic enzymes) often present oligodendrocyte and/or myelin defects which contribute to the overall disease pathophysiology. These phenomena triggered an increasing number of studies over the functions lipid exert to shape and maintain myelin, and brought to the finding that lipids are more than only structural building blocks. They act as signaling molecules to drive proliferation and differentiation of oligodendrocyte progenitor cells, as well as proliferation of premyelinating oligodendrocytes, and their maturation into myelinating ones. Here, we summarize key findings in these areas, while presenting the main related human diseases. Despite many advances in the field, various questions remain open which we briefly discuss. This article is part of a special issue entitled "Role of Lipids in CNS Cell Physiology and Pathology".
Collapse
Affiliation(s)
- Laura Montani
- Institute of Molecular Health Sciences, Department of Biology, Swiss Federal Institute of Technology, ETH Zürich, Zürich, CH-8093, Switzerland.
| |
Collapse
|
78
|
Castets P, Ham DJ, Rüegg MA. The TOR Pathway at the Neuromuscular Junction: More Than a Metabolic Player? Front Mol Neurosci 2020; 13:162. [PMID: 32982690 PMCID: PMC7485269 DOI: 10.3389/fnmol.2020.00162] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 08/05/2020] [Indexed: 12/18/2022] Open
Abstract
The neuromuscular junction (NMJ) is the chemical synapse connecting motor neurons and skeletal muscle fibers. NMJs allow all voluntary movements, and ensure vital functions like breathing. Changes in the structure and function of NMJs are hallmarks of numerous pathological conditions that affect muscle function including sarcopenia, the age-related loss of muscle mass and function. However, the molecular mechanisms leading to the morphological and functional perturbations in the pre- and post-synaptic compartments of the NMJ remain poorly understood. Here, we discuss the role of the metabolic pathway associated to the kinase TOR (Target of Rapamycin) in the development, maintenance and alterations of the NMJ. This is of particular interest as the TOR pathway has been implicated in aging, but its role at the NMJ is still ill-defined. We highlight the respective functions of the two TOR-associated complexes, TORC1 and TORC2, and discuss the role of localized protein synthesis and autophagy regulation in motor neuron terminals and sub-synaptic regions of muscle fibers and their possible effects on NMJ maintenance.
Collapse
Affiliation(s)
- Perrine Castets
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | | | | |
Collapse
|
79
|
Xia W, Zhu J, Wang X, Tang Y, Zhou P, Hou M, Li S. ANXA1 directs Schwann cells proliferation and migration to accelerate nerve regeneration through the FPR2/AMPK pathway. FASEB J 2020; 34:13993-14005. [PMID: 32856352 DOI: 10.1096/fj.202000726rrr] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 08/06/2020] [Accepted: 08/10/2020] [Indexed: 12/17/2022]
Abstract
Many factors are involved in the process of nerve regeneration. Understanding the mechanisms regarding how these factors promote an efficient remyelination is crucial to deciphering the molecular and cellular processes required to promote nerve repair. Schwann cells (SCs) play a central role in the process of peripheral nerve repair/regeneration. Using a model of facial nerve crush injury and repair, we identified Annexin A1 (ANXA1) as the extracellular trigger of SC proliferation and migration. ANXA1 activated formyl peptide receptor 2 (FPR2) receptors and the downstream adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK) signaling cascade, leading to SC proliferation and migration in vitro. SCs lacking FPR2 or AMPK displayed a defect in proliferation and migration. After facial nerve injury (FNI), ANXA1 promoted the proliferation of SCs and nerve regeneration in vivo. Collectively, these data identified the ANXA1/FPR2/AMPK axis as an important pathway in SC proliferation and migration. ANXA1-induced remyelination and SC proliferation promotes FNI regeneration.
Collapse
Affiliation(s)
- Wenzheng Xia
- Department of Neurosurgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Center for Diagnosis and Treatment of Cranial Nerve Diseases, Shanghai Jiao Tong University, Shanghai, China
| | - Jin Zhu
- Department of Neurosurgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Center for Diagnosis and Treatment of Cranial Nerve Diseases, Shanghai Jiao Tong University, Shanghai, China
| | - Xueyi Wang
- Department of Neurosurgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Center for Diagnosis and Treatment of Cranial Nerve Diseases, Shanghai Jiao Tong University, Shanghai, China
| | - Yinda Tang
- Department of Neurosurgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Center for Diagnosis and Treatment of Cranial Nerve Diseases, Shanghai Jiao Tong University, Shanghai, China
| | - Ping Zhou
- Department of Neurosurgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Center for Diagnosis and Treatment of Cranial Nerve Diseases, Shanghai Jiao Tong University, Shanghai, China
| | - Meng Hou
- Department of Radiation Oncology, First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Shiting Li
- Department of Neurosurgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Center for Diagnosis and Treatment of Cranial Nerve Diseases, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
80
|
R-Ras GTPases Signaling Role in Myelin Neurodegenerative Diseases. Int J Mol Sci 2020; 21:ijms21165911. [PMID: 32824627 PMCID: PMC7460555 DOI: 10.3390/ijms21165911] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 08/13/2020] [Accepted: 08/14/2020] [Indexed: 12/18/2022] Open
Abstract
Myelination is required for fast and efficient synaptic transmission in vertebrates. In the central nervous system, oligodendrocytes are responsible for creating myelin sheaths that isolate and protect axons, even throughout adulthood. However, when myelin is lost, the failure of remyelination mechanisms can cause neurodegenerative myelin-associated pathologies. From oligodendrocyte progenitor cells to mature myelinating oligodendrocytes, myelination is a highly complex process that involves many elements of cellular signaling, yet many of the mechanisms that coordinate it, remain unknown. In this review, we will focus on the three major pathways involved in myelination (PI3K/Akt/mTOR, ERK1/2-MAPK, and Wnt/β-catenin) and recent advances describing the crosstalk elements which help to regulate them. In addition, we will review the tight relation between Ras GTPases and myelination processes and discuss its potential as novel elements of crosstalk between the pathways. A better understanding of the crosstalk elements orchestrating myelination mechanisms is essential to identify new potential targets to mitigate neurodegeneration.
Collapse
|
81
|
Wang J, Yang L, Dong C, Wang J, Xu L, Qiu Y, Weng Q, Zhao C, Xin M, Lu QR. EED-mediated histone methylation is critical for CNS myelination and remyelination by inhibiting WNT, BMP, and senescence pathways. SCIENCE ADVANCES 2020; 6:eaaz6477. [PMID: 32851157 PMCID: PMC7423366 DOI: 10.1126/sciadv.aaz6477] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Accepted: 06/30/2020] [Indexed: 05/07/2023]
Abstract
Mutations in the polycomb repressive complex 2 (PRC2) can cause Weaver-like syndrome, wherein a patient cohort exhibits abnormal white matter; however, PRC2 functions in CNS myelination and regeneration remain elusive. We show here that H3K27me3, the PRC2 catalytic product, increases during oligodendrocyte maturation. Depletion of embryonic ectoderm development (EED), a core PRC2 subunit, reduces differentiation of oligodendrocyte progenitors (OPCs), and causes an OPC-to-astrocyte fate switch in a region-specific manner. Although dispensable for myelin maintenance, EED is critical for oligodendrocyte remyelination. Genomic occupancy and transcriptomic analyses indicate that EED establishes a chromatin landscape that selectively represses inhibitory WNT and bone morphogenetic protein (BMP) signaling, and senescence-associated programs. Blocking WNT or BMP pathways partially restores differentiation defects in EED-deficient OPCs. Thus, our findings reveal that EED/PRC2 is a crucial epigenetic programmer of CNS myelination and repair, while demonstrating a spatiotemporal-specific role of PRC2-mediated chromatin silencing in shaping oligodendrocyte identity and lineage plasticity.
Collapse
Affiliation(s)
- Jiajia Wang
- Department of Pediatrics, Brain Tumor Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Lijun Yang
- Department of Pediatrics, Brain Tumor Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Chen Dong
- Department of Pediatrics, Brain Tumor Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Jincheng Wang
- Department of Pediatrics, Brain Tumor Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Lingli Xu
- Department of Pediatrics, Brain Tumor Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Yueping Qiu
- Center for Drug Safety Evaluation and Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Qinjie Weng
- Center for Drug Safety Evaluation and Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Chuntao Zhao
- Department of Pediatrics, Brain Tumor Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Mei Xin
- Department of Pediatrics, Brain Tumor Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Q. Richard Lu
- Department of Pediatrics, Brain Tumor Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Corresponding author.
| |
Collapse
|
82
|
Li M, Meng N, Guo X, Niu X, Zhao Z, Wang W, Xie X, Lv P. Dl-3-n-Butylphthalide Promotes Remyelination and Suppresses Inflammation by Regulating AMPK/SIRT1 and STAT3/NF-κB Signaling in Chronic Cerebral Hypoperfusion. Front Aging Neurosci 2020; 12:137. [PMID: 32581761 PMCID: PMC7296049 DOI: 10.3389/fnagi.2020.00137] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 04/22/2020] [Indexed: 12/12/2022] Open
Abstract
Demyelination in vascular dementia (VD) is partly attributable to inflammation induced by chronic cerebral hypoperfusion (CCH). Remyelination contributes to the recovery of cognitive impairment by inducing the proliferation and differentiation of oligodendrocyte progenitor cells. It was previously reported that Dl-3-n-butylphthalide (NBP) promotes cognitive improvement. However, whether NBP can stimulate remyelination and suppress inflammation after CCH remains unclear. To answer this question, the present study investigated the effects of NBP on remyelination in a rat model of CCH established by bilateral carotid artery occlusion. Functional recovery was evaluated with the Morris water maze (MWM) test, and myelin integrity, regeneration of mature oligodendrocytes, and inhibition of astrocyte proliferation were assessed by immunohistochemistry and histologic analysis. Additionally, activation of 5′ AMP-activated protein kinase (AMPK)/Sirtuin (SIRT)1 and Signal transducer and activator of transcription (STAT)3/nuclear factor (NF)-κB signaling pathways was evaluated by western blotting. The results showed that NBP treatment improved memory and learning performance in CCH rats, which was accompanied by increased myelin integrity and oligodendrocyte regeneration, and reduced astrocyte proliferation and inflammation. Additionally, NBP induced the activation of AMPK/SIRT1 signaling while inhibiting the STAT3/NF-κB pathway. These results indicate that NBP alleviates cognitive impairment following CCH by promoting remyelination and suppressing inflammation via modulation of AMPK/SIRT1 and STAT3/NF-κB signaling.
Collapse
Affiliation(s)
- Meixi Li
- Department of Neurology, Hebei Medical University, Shijiazhuang, China.,Department of Neurology, Hebei General Hospital, Shijiazhuang, China
| | - Nan Meng
- Department of Neurology, Hebei Medical University, Shijiazhuang, China.,Department of Neurology, Hebei General Hospital, Shijiazhuang, China
| | - Xin Guo
- Department of Neurology, Hebei Medical University, Shijiazhuang, China
| | - Xiaoli Niu
- Department of Neurology, Hebei General Hospital, Shijiazhuang, China
| | - Zhongmin Zhao
- Department of Neurology, Hebei Medical University, Shijiazhuang, China
| | - Wei Wang
- Department of Neurology, Hebei Medical University, Shijiazhuang, China
| | - Xiaohua Xie
- Department of Neurology, Hebei General Hospital, Shijiazhuang, China
| | - Peiyuan Lv
- Department of Neurology, Hebei Medical University, Shijiazhuang, China.,Department of Neurology, Hebei General Hospital, Shijiazhuang, China
| |
Collapse
|
83
|
Sawade L, Grandi F, Mignanelli M, Patiño-López G, Klinkert K, Langa-Vives F, Di Guardo R, Echard A, Bolino A, Haucke V. Rab35-regulated lipid turnover by myotubularins represses mTORC1 activity and controls myelin growth. Nat Commun 2020; 11:2835. [PMID: 32503983 PMCID: PMC7275063 DOI: 10.1038/s41467-020-16696-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 05/18/2020] [Indexed: 01/21/2023] Open
Abstract
Inherited peripheral neuropathies (IPNs) represent a broad group of disorders including Charcot-Marie-Tooth (CMT) neuropathies characterized by defects primarily arising in myelin, axons, or both. The molecular mechanisms by which mutations in nearly 100 identified IPN/CMT genes lead to neuropathies are poorly understood. Here we show that the Ras-related GTPase Rab35 controls myelin growth via complex formation with the myotubularin-related phosphatidylinositol (PI) 3-phosphatases MTMR13 and MTMR2, encoded by genes responsible for CMT-types 4B2 and B1 in humans, and found that it downregulates lipid-mediated mTORC1 activation, a pathway known to crucially regulate myelin biogenesis. Targeted disruption of Rab35 leads to hyperactivation of mTORC1 signaling caused by elevated levels of PI 3-phosphates and to focal hypermyelination in vivo. Pharmacological inhibition of phosphatidylinositol 3,5-bisphosphate synthesis or mTORC1 signaling ameliorates this phenotype. These findings reveal a crucial role for Rab35-regulated lipid turnover by myotubularins to repress mTORC1 activity and to control myelin growth. Charcot-Marie-Tooth (CMT) is an inherited peripheral neuropathy. Here, the authors show that Rab35 forms a complex with genes implicated in CMT, MTMR13 and MTMR2, which regulates myelin growth by controlling mTORC1 signaling through lipid turnover.
Collapse
Affiliation(s)
- Linda Sawade
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Robert-Rössle-Strasse 10, 13125, Berlin, Germany
| | - Federica Grandi
- Institute of Experimental Neurology (InSpe), Division of Neuroscience, IRCCS Ospedale San Raffaele, Via Olgettina 60, 20132, Milan, Italy
| | - Marianna Mignanelli
- Institute of Experimental Neurology (InSpe), Division of Neuroscience, IRCCS Ospedale San Raffaele, Via Olgettina 60, 20132, Milan, Italy.,San Raffaele Vita-Salute University, Via Olgettina 60, 20132, Milan, Italy
| | - Genaro Patiño-López
- Laboratorio de Investigación en Inmunología y Proteómica, Hospital Infantil de México, Federico Gómez. C.P, 06720, Ciudad de México, México
| | - Kerstin Klinkert
- Membrane Traffic and Cell Division Lab, Institut Pasteur, UMR3691, CNRS, 25-28 rue du Dr Roux, F-75015, Paris, France.,Sorbonne Université, Collège doctoral, F-75005, Paris, France
| | - Francina Langa-Vives
- Centre d'Ingénierie Génétique Murine, Institut Pasteur, 25-28 rue du Dr Roux, F-75015, Paris, France
| | - Roberta Di Guardo
- Institute of Experimental Neurology (InSpe), Division of Neuroscience, IRCCS Ospedale San Raffaele, Via Olgettina 60, 20132, Milan, Italy
| | - Arnaud Echard
- Membrane Traffic and Cell Division Lab, Institut Pasteur, UMR3691, CNRS, 25-28 rue du Dr Roux, F-75015, Paris, France
| | - Alessandra Bolino
- Institute of Experimental Neurology (InSpe), Division of Neuroscience, IRCCS Ospedale San Raffaele, Via Olgettina 60, 20132, Milan, Italy.
| | - Volker Haucke
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Robert-Rössle-Strasse 10, 13125, Berlin, Germany. .,Freie Universität Berlin, Faculty of Biology, Chemistry and Pharmacy, 14195, Berlin, Germany. .,Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt- Universität zu Berlin, NeuroCure Cluster of Excellence, 10117, Berlin, Germany.
| |
Collapse
|
84
|
Warnock A, Toomey LM, Wright AJ, Fisher K, Won Y, Anyaegbu C, Fitzgerald M. Damage Mechanisms to Oligodendrocytes and White Matter in Central Nervous System Injury: The Australian Context. J Neurotrauma 2020; 37:739-769. [DOI: 10.1089/neu.2019.6890] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Affiliation(s)
- Andrew Warnock
- Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia
| | - Lillian M. Toomey
- Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia
- Perron Institute for Neurological and Translational Science, Nedlands, Western Australia, Australia
| | - Alexander J. Wright
- Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia
| | - Katherine Fisher
- School of Human Sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Yerim Won
- School of Human Sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Chidozie Anyaegbu
- Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia
| | - Melinda Fitzgerald
- Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia
- Perron Institute for Neurological and Translational Science, Nedlands, Western Australia, Australia
| |
Collapse
|
85
|
Nanocatalytic activity of clean-surfaced, faceted nanocrystalline gold enhances remyelination in animal models of multiple sclerosis. Sci Rep 2020; 10:1936. [PMID: 32041968 PMCID: PMC7010780 DOI: 10.1038/s41598-020-58709-w] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 01/17/2020] [Indexed: 12/26/2022] Open
Abstract
Development of pharmacotherapies that promote remyelination is a high priority for multiple sclerosis (MS), due to their potential for neuroprotection and restoration of function through repair of demyelinated lesions. A novel preparation of clean-surfaced, faceted gold nanocrystals demonstrated robust remyelinating activity in response to demyelinating agents in both chronic cuprizone and acute lysolecithin rodent animal models. Furthermore, oral delivery of gold nanocrystals improved motor functions of cuprizone-treated mice in both open field and kinematic gait studies. Gold nanocrystal treatment of oligodendrocyte precursor cells in culture resulted in oligodendrocyte maturation and expression of myelin differentiation markers. Additional in vitro data demonstrated that these gold nanocrystals act via a novel energy metabolism pathway involving the enhancement of key indicators of aerobic glycolysis. In response to gold nanocrystals, co-cultured central nervous system cells exhibited elevated levels of the redox coenzyme nicotine adenine dinucleotide (NAD+), elevated total intracellular ATP levels, and elevated extracellular lactate levels, along with upregulation of myelin-synthesis related genes, collectively resulting in functional myelin generation. Based on these preclinical studies, clean-surfaced, faceted gold nanocrystals represent a novel remyelinating therapeutic for multiple sclerosis.
Collapse
|
86
|
Belgrad J, De Pace R, Fields RD. Autophagy in Myelinating Glia. J Neurosci 2020; 40:256-266. [PMID: 31744863 PMCID: PMC6948934 DOI: 10.1523/jneurosci.1066-19.2019] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 10/17/2019] [Accepted: 11/08/2019] [Indexed: 12/14/2022] Open
Abstract
Autophagy is the cellular process involved in transportation and degradation of membrane, proteins, pathogens, and organelles. This fundamental cellular process is vital in development, plasticity, and response to disease and injury. Compared with neurons, little information is available on autophagy in glia, but it is paramount for glia to perform their critical responses to nervous system disease and injury, including active tissue remodeling and phagocytosis. In myelinating glia, autophagy has expanded roles, particularly in phagocytosis of mature myelin and in generating the vast amounts of membrane proteins and lipids that must be transported to form new myelin. Notably, autophagy plays important roles in removing excess cytoplasm to promote myelin compaction and development of oligodendrocytes, as well as in remyelination by Schwann cells after nerve trauma. This review summarizes the cell biology of autophagy, detailing the major pathways and proteins involved, as well as the roles of autophagy in Schwann cells and oligodendrocytes in development, plasticity, and diseases in which myelin is affected. This includes traumatic brain injury, Alexander's disease, Alzheimer's disease, hypoxia, multiple sclerosis, hereditary spastic paraplegia, and others. Promising areas for future research are highlighted.
Collapse
Affiliation(s)
| | - Raffaella De Pace
- Section on Intracellular Protein Trafficking, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892
| | | |
Collapse
|
87
|
Ornelas IM, Khandker L, Wahl SE, Hashimoto H, Macklin WB, Wood TL. The mechanistic target of rapamycin pathway downregulates bone morphogenetic protein signaling to promote oligodendrocyte differentiation. Glia 2020; 68:1274-1290. [PMID: 31904150 DOI: 10.1002/glia.23776] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 11/26/2019] [Accepted: 11/29/2019] [Indexed: 12/13/2022]
Abstract
Oligodendrocyte precursor cells (OPCs) differentiate and mature into oligodendrocytes, which produce myelin in the central nervous system. Prior studies have shown that the mechanistic target of rapamycin (mTOR) is necessary for proper myelination of the mouse spinal cord and that bone morphogenetic protein (BMP) signaling inhibits oligodendrocyte differentiation, in part by promoting expression of inhibitor of DNA binding 2 (Id2). Here we provide evidence that mTOR functions specifically in the transition from early stage OPC to immature oligodendrocyte by downregulating BMP signaling during postnatal spinal cord development. When mTOR is deleted from the oligodendrocyte lineage, expression of the FK506 binding protein 1A (FKBP12), a suppressor of BMP receptor activity, is reduced, downstream Smad activity is increased and Id2 expression is elevated. Additionally, mTOR inhibition with rapamycin in differentiating OPCs alters the transcriptional complex present at the Id2 promoter. Deletion of mTOR in oligodendroglia in vivo resulted in fewer late stage OPCs and fewer newly formed oligodendrocytes in the spinal cord with no effect on OPC proliferation or cell cycle exit. Finally, we demonstrate that inhibiting BMP signaling rescues the rapamycin-induced deficit in myelin protein expression. We conclude that mTOR promotes early oligodendrocyte differentiation by suppressing BMP signaling in OPCs.
Collapse
Affiliation(s)
- Isis M Ornelas
- Department of Pharmacology, Physiology & Neuroscience, New Jersey Medical School, Rutgers University, Newark, New Jersey
| | - Luipa Khandker
- Department of Pharmacology, Physiology & Neuroscience, New Jersey Medical School, Rutgers University, Newark, New Jersey
| | - Stacey E Wahl
- Department of Pharmacology, Physiology & Neuroscience, New Jersey Medical School, Rutgers University, Newark, New Jersey
| | - Hirokazu Hashimoto
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, Colorado
| | - Wendy B Macklin
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, Colorado
| | - Teresa L Wood
- Department of Pharmacology, Physiology & Neuroscience, New Jersey Medical School, Rutgers University, Newark, New Jersey
| |
Collapse
|
88
|
Hackett AR, Strickland A, Milbrandt J. Disrupting insulin signaling in Schwann cells impairs myelination and induces a sensory neuropathy. Glia 2019; 68:963-978. [PMID: 31758725 DOI: 10.1002/glia.23755] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 10/28/2019] [Accepted: 11/05/2019] [Indexed: 12/14/2022]
Abstract
Although diabetic mice have been studied for decades, little is known about the cell type specific contributions to diabetic neuropathy (DN). Schwann cells (SCs) myelinate and provide trophic support to peripheral nervous system axons. Altered SC metabolism leads to myelin defects, which can be seen both in inherited and DNs. How SC metabolism is altered in DN is not fully understood, but it is clear that insulin resistance underlies impaired lipid metabolism in many cell types throughout the body via the phosphoinositide 3-kinase/protein kinase b (PKB)/mammalian target of rapamycin (PI3K/AKT/mTOR) pathway. Here, we created an insulin resistant SC by deleting both insulin receptor (INSR) and insulin-like growth factor receptor 1 (IGF1R), to determine the role of this signaling pathway in development and response to injury in order to understand SC defects in DN. We found that myelin is thinner throughout development and adulthood in INSR/IGF1R Schwann cell specific knock out mice. The nerves of these mutant mice had reduced expression of key genes that mediate fatty acid and cholesterol synthesis due to reduced mTOR-sterol regulatory element-binding protein signaling. In adulthood, these mice show sensory neuropathy phenotypes reminiscent of diabetic mice. Altogether, these data suggest that SCs may play an important role in DN and targeting their metabolism could lead to new therapies for DN.
Collapse
Affiliation(s)
- Amber R Hackett
- Department of Genetics, Washington University School of Medicine, Saint Louis, Missouri
| | - Amy Strickland
- Department of Genetics, Washington University School of Medicine, Saint Louis, Missouri
| | - Jeffrey Milbrandt
- Department of Genetics, Washington University School of Medicine, Saint Louis, Missouri
| |
Collapse
|
89
|
Sen NE, Arsovic A, Meierhofer D, Brodesser S, Oberschmidt C, Canet-Pons J, Kaya ZE, Halbach MV, Gispert S, Sandhoff K, Auburger G. In Human and Mouse Spino-Cerebellar Tissue, Ataxin-2 Expansion Affects Ceramide-Sphingomyelin Metabolism. Int J Mol Sci 2019; 20:E5854. [PMID: 31766565 PMCID: PMC6928749 DOI: 10.3390/ijms20235854] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 11/19/2019] [Accepted: 11/20/2019] [Indexed: 02/08/2023] Open
Abstract
Ataxin-2 (human gene symbol ATXN2) acts during stress responses, modulating mRNA translation and nutrient metabolism. Ataxin-2 knockout mice exhibit progressive obesity, dyslipidemia, and insulin resistance. Conversely, the progressive ATXN2 gain of function due to the fact of polyglutamine (polyQ) expansions leads to a dominantly inherited neurodegenerative process named spinocerebellar ataxia type 2 (SCA2) with early adipose tissue loss and late muscle atrophy. We tried to understand lipid dysregulation in a SCA2 patient brain and in an authentic mouse model. Thin layer chromatography of a patient cerebellum was compared to the lipid metabolome of Atxn2-CAG100-Knockin (KIN) mouse spinocerebellar tissue. The human pathology caused deficits of sulfatide, galactosylceramide, cholesterol, C22/24-sphingomyelin, and gangliosides GM1a/GD1b despite quite normal levels of C18-sphingomyelin. Cerebellum and spinal cord from the KIN mouse showed a consistent decrease of various ceramides with a significant elevation of sphingosine in the more severely affected spinal cord. Deficiency of C24/26-sphingomyelins contrasted with excess C18/20-sphingomyelin. Spinocerebellar expression profiling revealed consistent reductions of CERS protein isoforms, Sptlc2 and Smpd3, but upregulation of Cers2 mRNA, as prominent anomalies in the ceramide-sphingosine metabolism. Reduction of Asah2 mRNA correlated to deficient S1P levels. In addition, downregulations for the elongase Elovl1, Elovl4, Elovl5 mRNAs and ELOVL4 protein explain the deficit of very long-chain sphingomyelin. Reduced ASMase protein levels correlated to the accumulation of long-chain sphingomyelin. Overall, a deficit of myelin lipids was prominent in SCA2 nervous tissue at prefinal stage and not compensated by transcriptional adaptation of several metabolic enzymes. Myelination is controlled by mTORC1 signals; thus, our human and murine observations are in agreement with the known role of ATXN2 yeast, nematode, and mouse orthologs as mTORC1 inhibitors and autophagy promoters.
Collapse
Affiliation(s)
- Nesli-Ece Sen
- Experimental Neurology, Building 89, Goethe University Medical Faculty, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (N.-E.S.); (A.A.); (C.O.); (J.C.-P.); (Z.-E.K.); (M.-V.H.); (S.G.)
- Faculty of Biosciences, Goethe-University, 60438 Frankfurt am Main, Germany
| | - Aleksandar Arsovic
- Experimental Neurology, Building 89, Goethe University Medical Faculty, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (N.-E.S.); (A.A.); (C.O.); (J.C.-P.); (Z.-E.K.); (M.-V.H.); (S.G.)
| | - David Meierhofer
- Max Planck Institute for Molecular Genetics, Ihnestrasse 63-73, 14195 Berlin, Germany;
| | - Susanne Brodesser
- Membrane Biology and Lipid Biochemistry Unit, Life and Medical Sciences Institute, University of Bonn, 53121 Bonn, Germany;
| | - Carola Oberschmidt
- Experimental Neurology, Building 89, Goethe University Medical Faculty, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (N.-E.S.); (A.A.); (C.O.); (J.C.-P.); (Z.-E.K.); (M.-V.H.); (S.G.)
| | - Júlia Canet-Pons
- Experimental Neurology, Building 89, Goethe University Medical Faculty, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (N.-E.S.); (A.A.); (C.O.); (J.C.-P.); (Z.-E.K.); (M.-V.H.); (S.G.)
| | - Zeynep-Ece Kaya
- Experimental Neurology, Building 89, Goethe University Medical Faculty, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (N.-E.S.); (A.A.); (C.O.); (J.C.-P.); (Z.-E.K.); (M.-V.H.); (S.G.)
- Cerrahpasa School of Medicine, Istanbul University, 34098 Istanbul, Turkey
| | - Melanie-Vanessa Halbach
- Experimental Neurology, Building 89, Goethe University Medical Faculty, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (N.-E.S.); (A.A.); (C.O.); (J.C.-P.); (Z.-E.K.); (M.-V.H.); (S.G.)
| | - Suzana Gispert
- Experimental Neurology, Building 89, Goethe University Medical Faculty, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (N.-E.S.); (A.A.); (C.O.); (J.C.-P.); (Z.-E.K.); (M.-V.H.); (S.G.)
| | - Konrad Sandhoff
- Membrane Biology and Lipid Biochemistry Unit, Life and Medical Sciences Institute, University of Bonn, 53121 Bonn, Germany;
| | - Georg Auburger
- Experimental Neurology, Building 89, Goethe University Medical Faculty, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (N.-E.S.); (A.A.); (C.O.); (J.C.-P.); (Z.-E.K.); (M.-V.H.); (S.G.)
| |
Collapse
|
90
|
Fledrich R, Kungl T, Nave KA, Stassart RM. Axo-glial interdependence in peripheral nerve development. Development 2019; 146:146/21/dev151704. [PMID: 31719044 DOI: 10.1242/dev.151704] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
During the development of the peripheral nervous system, axons and myelinating Schwann cells form a unique symbiotic unit, which is realized by a finely tuned network of molecular signals and reciprocal interactions. The importance of this complex interplay becomes evident after injury or in diseases in which aspects of axo-glial interaction are perturbed. This Review focuses on the specific interdependence of axons and Schwann cells in peripheral nerve development that enables axonal outgrowth, Schwann cell lineage progression, radial sorting and, finally, formation and maintenance of the myelin sheath.
Collapse
Affiliation(s)
- Robert Fledrich
- Institute of Anatomy, Leipzig University, 04103 Leipzig, Germany .,Department of Neurogenetics, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany
| | - Theresa Kungl
- Institute of Anatomy, Leipzig University, 04103 Leipzig, Germany.,Department of Neurogenetics, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany
| | - Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany
| | - Ruth M Stassart
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany .,Department of Neuropathology, University Clinic Leipzig, 04103 Leipzig, Germany
| |
Collapse
|
91
|
Thomas L, Pasquini LA. Galectin-3 Exerts a Pro-differentiating and Pro-myelinating Effect Within a Temporal Window Spanning Precursors and Pre-oligodendrocytes: Insights into the Mechanisms of Action. Mol Neurobiol 2019; 57:976-987. [PMID: 31654317 DOI: 10.1007/s12035-019-01787-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 09/12/2019] [Indexed: 11/29/2022]
Abstract
Oligodendrocytes (OLG) are the cells resident in the CNS responsible for myelination. OLG undergo a succession of morphological and molecular changes along several maturational stages. Galectin-3 (Gal-3) is a 25- to 35-KDa protein belonging to the family of carbohydrate-binding galectins, which bind to glycoconjugates containing β-galactosides. Gal-3 lacks a specific receptor and its binding is thus rather unspecific, as it depends on the cellular environment and the repertoire of glycomolecules at the time when Gal-3 is present. Our previous work revealed that recombinant Gal-3 (rGal-3)-treated OLG showed accelerated differentiation, evidenced by an increase in the number of mature cells to the detriment of immature ones and accelerated actin cytoskeleton dynamics. These changes were a consequence of rGal-3 influence on Akt, Erk 1/2, and β-catenin signaling pathways. Considering this previous evidence, the aim of this study was to identify the temporal window of rGal-3 action on the OLG lineage to induce OLG maturation by using specific single pulses of rGal-3 over the different maturational stages of OLG, and to unravel its main direct targets promoting OLG differentiation by mass spectrometry analysis. Our results reveal a key temporal window spanning between OPC and pre-OLG states in which rGal-3 action promotes OLG differentiation, and identify several targets for rGal-3 binding including proteins related to the cytoskeleton, signaling pathways, metabolism and intracellular trafficking, among others. These results highlight the relevance of Gal-3 in signaling pathways regulating oligodendroglial differentiation and support a potential therapeutic role for rGal-3 in demyelinating diseases such as multiple sclerosis.
Collapse
Affiliation(s)
- Laura Thomas
- Department of Biological Chemistry, School of Pharmacy and Biochemistry, Institute of Chemistry and Biological Physicochemistry (IQUIFIB), University of Buenos Aires and National Research Council (CONICET), Buenos Aires, Argentina
| | - Laura Andrea Pasquini
- Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 956, C1113, Buenos Aires, Argentina.
| |
Collapse
|
92
|
Abstract
Autism spectrum disorder (ASD) has been hypothesized to be a result of altered connectivity in the brain. Recent imaging studies suggest accelerated maturation of the white matter in young children with ASD, with underlying mechanisms unknown. Myelin is an integral part of the white matter and critical for connectivity; however, its role in ASD remains largely unclear. Here, we investigated myelin development in a model of idiopathic ASD, the BTBR mice. Magnetic resonance imaging revealed that fiber tracts in the frontal brain of the BTBR mice had increased volume at postnatal day 6, but the difference reduced over time, reminiscent of the findings in young patients. We further identified that myelination in the frontal brain of both male and female neonatal BTBR mice was increased, associated with elevated levels of myelin basic protein. However, myelin pattern was unaltered in adult BTBR mice, revealing accelerated developmental trajectory of myelination. Consistently, we found that signaling of platelet-derived growth factor receptor alpha (PDGFRα) was reduced in the frontal brain of neonatal BTBR mice. However, levels of microRNA species known to regulate PDGFRα signaling and myelination were unaltered. Together, these results suggest that precocious myelination could potentially contribute to increased volume and connectivity of the white matter observed in young children with ASD.
Collapse
|
93
|
Machado J, Silveira WA, Gonçalves DA, Schavinski AZ, Khan MM, Zanon NM, Diaz MB, Rudolf R, Kettelhut IC, Navegantes LC. α-Calcitonin gene-related peptide inhibits autophagy and calpain systems and maintains the stability of neuromuscular junction in denervated muscles. Mol Metab 2019; 28:91-106. [PMID: 31331823 PMCID: PMC6822259 DOI: 10.1016/j.molmet.2019.06.024] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 06/23/2019] [Accepted: 06/28/2019] [Indexed: 11/25/2022] Open
Abstract
OBJECTIVE Although it is well established that a-calcitonin gene-related peptide (CGRP) stabilizes muscle-type cholinergic receptors nicotinic subunits (AChR), the underlying mechanism by which this neuropeptide regulates muscle protein metabolism and neuromuscular junction (NMJ) morphology is unclear. METHODS To elucidate the mechanisms how CGRP controls NMJ stability in denervated mice skeletal muscles, we carried out physiological, pharmacological, and molecular analyses of atrophic muscles induced by sciatic nerve transection. RESULTS Here, we report that CGRP treatment in vivo abrogated the deleterious effects on NMJ upon denervation (DEN), an effect that was associated with suppression of skeletal muscle proteolysis, but not stimulation of protein synthesis. CGRP also blocked the DEN-induced increase in endocytic AChR vesicles and the elevation of autophagosomes per NMJ area. The treatment of denervated animals with rapamycin blocked the stimulatory effects of CGRP on mTORC1 and its inhibitory actions on autophagic flux and NMJ degeneration. Furthermore, CGRP inhibited the DEN-induced hyperactivation of Ca2+-dependent proteolysis, a degradative system that has been shown to destabilize NMJ. Consistently, calpain was found to be activated by cholinergic stimulation in myotubes leading to the dispersal of AChR clusters, an effect that was abolished by CGRP. CONCLUSION Taken together, these data suggest that the inhibitory effect of CGRP on autophagy and calpain may represent an important mechanism for the preservation of synapse morphology when degradative machinery is exacerbated upon denervation conditions.
Collapse
Affiliation(s)
- Juliano Machado
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil; Department of Biochemistry/Immunology, Ribeirão Preto Medical School/University of São Paulo, Ribeirão Preto, São Paulo, Brazil; Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, 85764, Neuherberg, Germany; Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine I, Heidelberg University Hospital, 69120, Heidelberg, Germany.
| | - Wilian A Silveira
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil.
| | - Dawit A Gonçalves
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil; Department of Biochemistry/Immunology, Ribeirão Preto Medical School/University of São Paulo, Ribeirão Preto, São Paulo, Brazil.
| | - Aline Zanatta Schavinski
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil.
| | - Muzamil M Khan
- Institute of Molecular and Cell Biology, University of Applied Sciences Mannheim, Mannheim, Germany; Institute of Medical Technology, University of Heidelberg and University of Applied Sciences Mannheim, Mannheim, Germany.
| | - Neusa M Zanon
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil.
| | - Mauricio Berriel Diaz
- Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, 85764, Neuherberg, Germany; Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine I, Heidelberg University Hospital, 69120, Heidelberg, Germany.
| | - Rüdiger Rudolf
- Institute of Molecular and Cell Biology, University of Applied Sciences Mannheim, Mannheim, Germany; Institute of Medical Technology, University of Heidelberg and University of Applied Sciences Mannheim, Mannheim, Germany.
| | - Isis C Kettelhut
- Department of Biochemistry/Immunology, Ribeirão Preto Medical School/University of São Paulo, Ribeirão Preto, São Paulo, Brazil.
| | - Luiz C Navegantes
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil.
| |
Collapse
|
94
|
Zhang SJ, Li XX, Yu Y, Chiu AP, Lo LH, To JC, Rowlands DK, Keng VW. Schwann cell-specific PTEN and EGFR dysfunctions affect neuromuscular junction development by impairing Agrin signaling and autophagy. Biochem Biophys Res Commun 2019; 515:50-56. [PMID: 31122699 DOI: 10.1016/j.bbrc.2019.05.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 05/02/2019] [Indexed: 10/26/2022]
Abstract
The neuromuscular junction (NMJ) is formed by motor nerve terminals, post-junctional muscle membranes, and terminal Schwann cells (SCs). The formation of NMJ requires complex and dynamic molecular interactions. Nerve- and muscle-derived molecules have been well characterized but the mechanistic involvement of SC in NMJ development remains poorly understood. SC-specific phosphatase and tensin homolog (Pten) inactivation and epidermal growth factor receptor (EGFR) overexpression (Dhh-Cre; Cnp-EGFR; Ptenflox/flox or DET) mice were used and NMJ malformation was observed in these mice. Acetylcholine receptors (AChRs) were distorted and varicose presynaptic nerve terminals appeared in the tibialis anterior (TA) muscle of DET mice. Agrin signaling related to NMJ development, was downregulated in TA muscle. Both RAS/MEK/ERK and PI3K/AKT/mTOR signaling pathways were activated in the sciatic nerves of DET mice. In addition, autophagy was downregulated in these sciatic nerves. Interestingly, the use of Torin 2, an mTOR inhibitor, rescued the phenotype. The downregulated-autophagy might account for Agrin signaling abnormity, which induced NMJ malformation. Taken together, our results indicate that SCs-specific Pten and EGFR cooperation are essential for NMJ development.
Collapse
Affiliation(s)
- Shi-Jie Zhang
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China; Department of Neurology, Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiao-Xiao Li
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| | - Yuyu Yu
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| | - Amy P Chiu
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| | - Lilian H Lo
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| | - Jeffrey C To
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| | - Dewi K Rowlands
- Laboratory Animal Services Centre, The Chinese University of Hong Kong, Sha Tin, New Territories, Hong Kong SAR, China
| | - Vincent W Keng
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China.
| |
Collapse
|
95
|
Wu F, Xu K, Liu L, Zhang K, Xia L, Zhang M, Teng C, Tong H, He Y, Xue Y, Zhang H, Chen D, Hu A. Vitamin B 12 Enhances Nerve Repair and Improves Functional Recovery After Traumatic Brain Injury by Inhibiting ER Stress-Induced Neuron Injury. Front Pharmacol 2019; 10:406. [PMID: 31105562 PMCID: PMC6491933 DOI: 10.3389/fphar.2019.00406] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 04/01/2019] [Indexed: 12/31/2022] Open
Abstract
Traumatic brain injury (TBI) is one of the most common causes of neurological damage in young human populations. Vitamin B12 has been reported to promote axon growth of neuronal cells after peripheral nerve injury, which is currently used for the treatment of peripheral nerve damage in the clinical trial. Thus, we hypothesized that TBI can be attenuated by vitaminB12 treatment through its beneficial role on axon regeneration after nerve injury. To confirm it, the biological function of vitaminB12 was characterized using hematoxylin and eosin (H&E) staining, Luxol fast blue (LFB) staining, western blot analysis, and immunohistochemistry staining. The results showed that the neurological functional recovery was improved in the VitaminB12-treated group after TBI, which may be due to downregulation of the endoplasmic reticulum stress-related apoptosis signaling pathway. Moreover, the microtubule stabilization, remyelination and myelin reparation were rescued by vitamin B12, which was consistent with the treatment of 4-phenylbutyric acid (4-PBA), an endoplasmic reticulum stress inhibitor. The study suggests that vitamin B12 may be useful as a novel neuroprotective drug for TBI.
Collapse
Affiliation(s)
- Fangfang Wu
- Department of Emergency, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China.,School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Ke Xu
- Institute of Life Sciences, Wenzhou University, Wenzhou, China
| | - Lei Liu
- Department of Emergency, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Kairui Zhang
- Department of Emergency, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Leilei Xia
- Department of Emergency, Wenzhou People's Hospital, The Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou, China
| | - Man Zhang
- Department of Emergency, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Chenhuai Teng
- Department of Emergency, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Heyan Tong
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Yifang He
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Yujie Xue
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Hongyu Zhang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Daqing Chen
- Department of Emergency, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Aiping Hu
- Department of Emergency, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China.,School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
96
|
Sokol DK, Maloney B, Westmark CJ, Lahiri DK. Novel Contribution of Secreted Amyloid-β Precursor Protein to White Matter Brain Enlargement in Autism Spectrum Disorder. Front Psychiatry 2019; 10:165. [PMID: 31024350 PMCID: PMC6469489 DOI: 10.3389/fpsyt.2019.00165] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 03/06/2019] [Indexed: 12/27/2022] Open
Abstract
The most replicated neuroanatomical finding in autism is the tendency toward brain overgrowth, especially in younger children. Research shows that both gray and white matter are enlarged. Proposed mechanisms underlying brain enlargement include abnormal inflammatory and neurotrophic signals that lead to excessive, aberrant dendritic connectivity via disrupted pruning and cell adhesion, and enlargement of white matter due to excessive gliogenesis and increased myelination. Amyloid-β protein precursor (βAPP) and its metabolites, more commonly associated with Alzheimer's disease (AD), are also dysregulated in autism plasma and brain tissue samples. This review highlights findings that demonstrate how one βAPP metabolite, secreted APPα, and the ADAM family α-secretases, may lead to increased brain matter, with emphasis on increased white matter as seen in autism. sAPPα and the ADAM family α-secretases contribute to the anabolic, non-amyloidogenic pathway, which is in contrast to the amyloid (catabolic) pathway known to contribute to Alzheimer disease. The non-amyloidogenic pathway could produce brain enlargement via genetic mechanisms affecting mRNA translation and polygenic factors that converge on molecular pathways (mitogen-activated protein kinase/MAPK and mechanistic target of rapamycin/mTOR), promoting neuroinflammation. A novel mechanism linking the non-amyloidogenic pathway to white matter enlargement is proposed: α-secretase and/or sAPPα, activated by ERK receptor signaling activates P13K/AKt/mTOR and then Rho GTPases favoring myelination via oligodendrocyte progenitor cell (OPC) activation of cofilin. Applying known pathways in AD to autism should allow further understanding and provide options for new drug targets.
Collapse
Affiliation(s)
- Deborah K. Sokol
- Pediatrics Section, Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Bryan Maloney
- Indiana Alzheimers Disease Center, Department of Psychiatry, Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Cara J. Westmark
- Department of Neurology, University of Wisconsin, Madison, WI, United States
| | - Debomoy K. Lahiri
- Indiana Alzheimers Disease Center, Department of Psychiatry, Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
97
|
Jessen KR, Arthur-Farraj P. Repair Schwann cell update: Adaptive reprogramming, EMT, and stemness in regenerating nerves. Glia 2019; 67:421-437. [PMID: 30632639 DOI: 10.1002/glia.23532] [Citation(s) in RCA: 212] [Impact Index Per Article: 42.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 08/20/2018] [Accepted: 09/05/2018] [Indexed: 12/16/2022]
Abstract
Schwann cells respond to nerve injury by cellular reprogramming that generates cells specialized for promoting regeneration and repair. These repair cells clear redundant myelin, attract macrophages, support survival of damaged neurons, encourage axonal growth, and guide axons back to their targets. There are interesting parallels between this response and that found in other tissues. At the cellular level, many other tissues also react to injury by cellular reprogramming, generating cells specialized to promote tissue homeostasis and repair. And at the molecular level, a common feature possessed by Schwann cells and many other cells is the injury-induced activation of genes associated with epithelial-mesenchymal transitions and stemness, differentiation states that are linked to cellular plasticity and that help injury-induced tissue remodeling. The number of signaling systems regulating Schwann cell plasticity is rapidly increasing. Importantly, this includes mechanisms that are crucial for the generation of functional repair Schwann cells and nerve regeneration, although they have no or a minor role elsewhere in the Schwann cell lineage. This encourages the view that selective tools can be developed to control these particular cells, amplify their repair supportive functions and prevent their deterioration. In this review, we discuss the emerging similarities between the injury response seen in nerves and in other tissues and survey the transcription factors, epigenetic mechanisms, and signaling cascades that control repair Schwann cells, with emphasis on systems that selectively regulate the Schwann cell injury response.
Collapse
Affiliation(s)
- Kristjan R Jessen
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - Peter Arthur-Farraj
- John Van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
98
|
Ishii A, Furusho M, Macklin W, Bansal R. Independent and cooperative roles of the Mek/ERK1/2-MAPK and PI3K/Akt/mTOR pathways during developmental myelination and in adulthood. Glia 2019; 67:1277-1295. [PMID: 30761608 DOI: 10.1002/glia.23602] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 01/25/2019] [Accepted: 01/29/2019] [Indexed: 12/20/2022]
Abstract
Multiple extracellular and intracellular signals regulate the functions of oligodendrocytes as they progress through the complex process of developmental myelination and then maintain a functionally intact myelin sheath throughout adult life, preserving the integrity of the axons. Recent studies suggest that Mek/ERK1/2-MAPK and PI3K/Akt/mTOR intracellular signaling pathways play important, often overlapping roles in the regulation of myelination. However, it remains poorly understood whether they function independently, sequentially, or converge using a common mechanism to facilitate oligodendrocyte differentiation, myelin growth, and maintenance. To address these questions, we analyzed multiple genetically modified mice and asked whether the deficits due to the conditional loss-of-function of ERK1/2 or mTOR could be abrogated by simultaneous constitutive activation of PI3K/Akt or Mek, respectively. From these studies, we concluded that while PI3K/Akt, not Mek/ERK1/2, plays a key role in promoting oligodendrocyte differentiation and timely initiation of myelination through mTORC1 signaling, Mek/ERK1/2-MAPK functions largely independently of mTORC1 to preserve the integrity of the myelinated axons during adulthood. However, to promote the efficient growth of the myelin sheath, these two pathways cooperate with each other converging at the level of mTORC1, both in the context of normal developmental myelination or following forced reactivation of the myelination program during adulthood. Thus, Mek/ERK1/2-MAPK and the PI3K/Akt/mTOR signaling pathways work both independently and cooperatively to maintain a finely tuned, temporally regulated balance as oligodendrocytes progress through different phases of developmental myelination into adulthood. Therapeutic strategies aimed at targeting remyelination in demyelinating diseases are expected to benefit from these findings.
Collapse
Affiliation(s)
- Akihiro Ishii
- Department of Neuroscience, University of Connecticut Medical School, Farmington, Connecticut, USA
| | - Miki Furusho
- Department of Neuroscience, University of Connecticut Medical School, Farmington, Connecticut, USA
| | - Wendy Macklin
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Rashmi Bansal
- Department of Neuroscience, University of Connecticut Medical School, Farmington, Connecticut, USA
| |
Collapse
|
99
|
Elbaz B, Popko B. Molecular Control of Oligodendrocyte Development. Trends Neurosci 2019; 42:263-277. [PMID: 30770136 DOI: 10.1016/j.tins.2019.01.002] [Citation(s) in RCA: 168] [Impact Index Per Article: 33.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 01/07/2019] [Accepted: 01/15/2019] [Indexed: 12/27/2022]
Abstract
Myelin is a multilayer lipid membrane structure that wraps and insulates axons, allowing for the efficient propagation of action potentials. During developmental myelination of the central nervous system (CNS), oligodendrocyte progenitor cells (OPCs) proliferate and migrate to their final destination, where they terminally differentiate into mature oligodendrocytes and myelinate axons. Lineage progression and terminal differentiation of oligodendrocyte lineage cells are under tight transcriptional and post-transcriptional control. The characterization of several recently identified regulatory factors that govern these processes, which are the focus of this review, has greatly increased our understanding of oligodendrocyte development and function. These insights are critical to facilitate efforts to enhance OPC differentiation in neurological disorders that disrupt CNS myelin.
Collapse
Affiliation(s)
- Benayahu Elbaz
- The Center for Peripheral Neuropathy, The Department of Neurology, University of Chicago, Chicago, IL, USA
| | - Brian Popko
- The Center for Peripheral Neuropathy, The Department of Neurology, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
100
|
Zhang Y, Lu XY, Ye ZQ, Ciric B, Ma CG, Rostami A, Li X, Zhang GX. Combination Therapy With Fingolimod and Neural Stem Cells Promotes Functional Myelination in vivo Through a Non-immunomodulatory Mechanism. Front Cell Neurosci 2019; 13:14. [PMID: 30804753 PMCID: PMC6371042 DOI: 10.3389/fncel.2019.00014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 01/15/2019] [Indexed: 01/03/2023] Open
Abstract
Myelination, which occurs predominantly postnatally and continues throughout life, is important for proper neurologic function of the mammalian central nervous system (CNS). We have previously demonstrated that the combination therapy of fingolimod (FTY720) and transplanted neural stem cells (NSCs) had a significantly enhanced therapeutic effect on the chronic stage of experimental autoimmune encephalomyelitis, an animal model of CNS autoimmunity, compared to using either one of them alone. However, reduced disease severity may be secondary to the immunomodulatory effects of FTY720 and NSCs, while whether this therapy directly affects myelinogenesis remains unknown. To investigate this important question, we used three myelination models under minimal or non-inflammatory microenvironments. Our results showed that FTY720 drives NSCs to differentiate into oligodendrocytes and promotes myelination in an ex vivo brain slice culture model, and in the developing CNS of healthy postnatal mice in vivo. Elevated levels of neurotrophic factors, e.g., brain-derived neurotrophic factor and glial cell line-derived neurotrophic factor, were observed in the CNS of the treated infant mice. Further, FTY720 and NSCs efficiently prolonged the survival and improved sensorimotor function of shiverer mice. Together, these data demonstrate a direct effect of FTY720, beyond its known immunomodulatory capacity, in NSC differentiation and myelin development as a novel mechanism underlying its therapeutic effect in demyelinating diseases.
Collapse
Affiliation(s)
- Yuan Zhang
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, United States,National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, Shaanxi Normal University, Xi'an, China
| | - Xin-Yu Lu
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, Shaanxi Normal University, Xi'an, China
| | - Ze-Qin Ye
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, Shaanxi Normal University, Xi'an, China
| | - Bogoljub Ciric
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Cun-Gen Ma
- Department of Neurology, Institute of Brain Science, Shanxi Datong University Medical School, Datong, China
| | - Abdolmohamad Rostami
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Xing Li
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, United States,National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, Shaanxi Normal University, Xi'an, China,*Correspondence: Xing Li
| | - Guang-Xian Zhang
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, United States,Guang-Xian Zhang
| |
Collapse
|