1
|
Giri SS, Tripathi AS, Erkekoğlu P, Zaki MEA. Molecular pathway of pancreatic cancer-associated neuropathic pain. J Biochem Mol Toxicol 2024; 38:e23638. [PMID: 38613466 DOI: 10.1002/jbt.23638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 11/29/2023] [Accepted: 12/21/2023] [Indexed: 04/15/2024]
Abstract
The pancreas is a heterocrine gland that has both exocrine and endocrine parts. Most pancreatic cancer begins in the cells that line the ducts of the pancreas and is called pancreatic ductal adenocarcinoma (PDAC). PDAC is the most encountered pancreatic cancer type. One of the most important characteristic features of PDAC is neuropathy which is primarily due to perineural invasion (PNI). PNI develops tumor microenvironment which includes overexpression of fibroblasts cells, macrophages, as well as angiogenesis which can be responsible for neuropathy pain. In tumor microenvironment inactive fibroblasts are converted into an active form that is cancer-associated fibroblasts (CAFs). Neurotrophins they also increase the level of Substance P, calcitonin gene-related peptide which is also involved in pain. Matrix metalloproteases are the zinc-associated proteases enzymes which activates proinflammatory interleukin-1β into its activated form and are responsible for release and activation of Substance P which is responsible for neuropathic pain by transmitting pain signal via dorsal root ganglion. All the molecules and their role in being responsible for neuropathic pain are described below.
Collapse
Affiliation(s)
| | - Alok Shiomurti Tripathi
- Department of Pharmacology, Era College of Pharmacy, Era University, Lucknow, Uttar Pradesh, India
| | - Pınar Erkekoğlu
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Magdi E A Zaki
- Department of Chemistry, Faculty of Science, Imam Mohammad lbn Saud Islamic University, Riyadh, Saudi Arabia
| |
Collapse
|
2
|
Louie AY, Kim JS, Drnevich J, Dibaeinia P, Koito H, Sinha S, McKim DB, Soto-Diaz K, Nowak RA, Das A, Steelman AJ. Influenza A virus infection disrupts oligodendrocyte homeostasis and alters the myelin lipidome in the adult mouse. J Neuroinflammation 2023; 20:190. [PMID: 37596606 PMCID: PMC10439573 DOI: 10.1186/s12974-023-02862-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Accepted: 07/25/2023] [Indexed: 08/20/2023] Open
Abstract
BACKGROUND Recent data suggest that myelin may be altered by physiological events occurring outside of the central nervous system, which may cause changes to cognition and behavior. Similarly, peripheral infection by non-neurotropic viruses is also known to evoke changes to cognition and behavior. METHODS Mice were inoculated with saline or influenza A virus. Bulk RNA-seq, lipidomics, RT-qPCR, flow cytometry, immunostaining, and western blots were used to determine the effect of infection on OL viability, protein expression and changes to the lipidome. To determine if microglia mediated infection-induced changes to OL homeostasis, mice were treated with GW2580, an inhibitor of microglia activation. Additionally, conditioned medium experiments using primary glial cell cultures were also used to test whether secreted factors from microglia could suppress OL gene expression. RESULTS Transcriptomic and RT-qPCR analyses revealed temporal downregulation of OL-specific transcripts with concurrent upregulation of markers characteristic of cellular stress. OLs isolated from infected mice had reduced cellular expression of myelin proteins compared with those from saline-inoculated controls. In contrast, the expression of these proteins within myelin was not different between groups. Similarly, histological and immunoblotting analysis performed on various brain regions indicated that infection did not alter OL viability, but increased expression of a cellular stress marker. Shot-gun lipidomic analysis revealed that infection altered the lipid profile within the prefrontal cortex as well as in purified brain myelin and that these changes persisted after recovery from infection. Treatment with GW2580 during infection suppressed the expression of genes associated with glial activation and partially restored OL-specific transcripts to baseline levels. Finally, conditioned medium from activated microglia reduced OL-gene expression in primary OLs without altering their viability. CONCLUSIONS These findings show that peripheral respiratory viral infection with IAV is capable of altering OL homeostasis and indicate that microglia activation is likely involved in the process.
Collapse
Affiliation(s)
- Allison Y Louie
- Neuroscience Program, 2325/21 Beckman Institute, 405 North Mathews Ave., Urbana, IL, 61801, USA
| | - Justin S Kim
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, 1201 W. Gregory Dr., Urbana, IL, 61801, USA
- School of Chemistry and Biochemistry, Georgia Institute of Technology, 3306, IBB, Parker H. Petit Institute for Bioengineering and Biosciences, 315 Fernst Dr. NW, Atlanta, GA, 30332, USA
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, 3516 Veterinary Medicine Basic Sciences Bldg., 2001 South Lincoln Avenue, Urbana, IL, 61802, USA
| | - Jenny Drnevich
- Roy J. Carver Biotechnology Center, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Payam Dibaeinia
- Department of Computer Science, University of Illinois at Urbana-Champaign, 201 North Goodwin Avenue, Urbana, IL, 61801, USA
| | - Hisami Koito
- Department of Pharmaceutical Sciences, Josai University, 1-1 Keyakidai, Sakado-shi, Saitama, 350-0295, Japan
| | - Saurabh Sinha
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, 1201 W. Gregory Dr., Urbana, IL, 61801, USA
- Department of Computer Science, University of Illinois at Urbana-Champaign, 201 North Goodwin Avenue, Urbana, IL, 61801, USA
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, 1206 West Gregory Dr., Urbana, IL, 61801, USA
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, USA
| | - Daniel B McKim
- Neuroscience Program, 2325/21 Beckman Institute, 405 North Mathews Ave., Urbana, IL, 61801, USA
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, 1201 W. Gregory Dr., Urbana, IL, 61801, USA
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, 1201 W. Gregory Dr., Urbana, IL, 61801, USA
| | - Katiria Soto-Diaz
- Neuroscience Program, 2325/21 Beckman Institute, 405 North Mathews Ave., Urbana, IL, 61801, USA
| | - Romana A Nowak
- Department of Computer Science, University of Illinois at Urbana-Champaign, 201 North Goodwin Avenue, Urbana, IL, 61801, USA
- Department of Bioengineering, Cancer Center at Illinois, Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, 405 N. Mathews Ave., Urbana, IL, 61801, USA
| | - Aditi Das
- Neuroscience Program, 2325/21 Beckman Institute, 405 North Mathews Ave., Urbana, IL, 61801, USA.
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, 1201 W. Gregory Dr., Urbana, IL, 61801, USA.
- School of Chemistry and Biochemistry, Georgia Institute of Technology, 3306, IBB, Parker H. Petit Institute for Bioengineering and Biosciences, 315 Fernst Dr. NW, Atlanta, GA, 30332, USA.
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, 3516 Veterinary Medicine Basic Sciences Bldg., 2001 South Lincoln Avenue, Urbana, IL, 61802, USA.
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, 1206 West Gregory Dr., Urbana, IL, 61801, USA.
| | - Andrew J Steelman
- Neuroscience Program, 2325/21 Beckman Institute, 405 North Mathews Ave., Urbana, IL, 61801, USA.
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, 1201 W. Gregory Dr., Urbana, IL, 61801, USA.
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, 1206 West Gregory Dr., Urbana, IL, 61801, USA.
- Department of Bioengineering, Cancer Center at Illinois, Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, 405 N. Mathews Ave., Urbana, IL, 61801, USA.
| |
Collapse
|
3
|
Basu K, Appukuttan S, Manchanda R, Sik A. Difference in axon diameter and myelin thickness between excitatory and inhibitory callosally projecting axons in mice. Cereb Cortex 2022; 33:4101-4115. [PMID: 36205478 PMCID: PMC10068302 DOI: 10.1093/cercor/bhac329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 07/18/2022] [Accepted: 08/02/2022] [Indexed: 11/13/2022] Open
Abstract
Synchronization of network oscillation in spatially distant cortical areas is essential for normal brain activity. Precision in synchronization between hemispheres depends on the axonal conduction velocity, which is determined by physical parameters of the axons involved, including diameter, and extent of myelination. To compare these parameters in long-projecting excitatory and inhibitory axons in the corpus callosum, we used genetically modified mice and virus tracing to separately label CaMKIIα expressing excitatory and GABAergic inhibitory axons. Using electron microscopy analysis, we revealed that (i) the axon diameters of excitatory fibers (myelinated axons) are significantly larger than those of nonmyelinated excitatory axons; (ii) the diameters of bare axons of excitatory myelinated fibers are significantly larger than those of their inhibitory counterparts; and (iii) myelinated excitatory fibers are significantly larger than myelinated inhibitory fibers. Also, the thickness of myelin ensheathing inhibitory axons is significantly greater than for excitatory axons, with the ultrastructure of the myelin around excitatory and inhibitory fibers also differing. We generated a computational model to investigate the functional consequences of these parameter divergences. Our simulations indicate that impulses through inhibitory and excitatory myelinated fibers reach the target almost simultaneously, whereas action potentials conducted by nonmyelinated axons reach target cells with considerable delay.
Collapse
Affiliation(s)
- Kaustuv Basu
- Facility for Electron Microscopy Research, McGill University, Montreal, QC H3A 0C72, Canada.,Department of Anatomy & Cell Biology, McGill University, Montreal, Canada
| | - Shailesh Appukuttan
- Biomedical Engineering Group, Department of Biosciences & Bioengineering IIT Bombay, Powai, Mumbay, 4000764, India
| | - Rohit Manchanda
- Biomedical Engineering Group, Department of Biosciences & Bioengineering IIT Bombay, Powai, Mumbay, 4000764, India
| | - Attila Sik
- College of Medical and Dental Sciences, University of Birmingham, Vincent Drive, Birmingham B15 2TT, United Kingdom.,Institute of Physiology, Medical School, University of Pecs, Pecs H-7624, Hungary.,Institute of Transdisciplinary Discoveries, Medical School, University of Pecs, Pecs H-7624, Hungary
| |
Collapse
|
4
|
Lam M, Takeo K, Almeida RG, Cooper MH, Wu K, Iyer M, Kantarci H, Zuchero JB. CNS myelination requires VAMP2/3-mediated membrane expansion in oligodendrocytes. Nat Commun 2022; 13:5583. [PMID: 36151203 PMCID: PMC9508103 DOI: 10.1038/s41467-022-33200-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 09/07/2022] [Indexed: 11/08/2022] Open
Abstract
Myelin is required for rapid nerve signaling and is emerging as a key driver of CNS plasticity and disease. How myelin is built and remodeled remains a fundamental question of neurobiology. Central to myelination is the ability of oligodendrocytes to add vast amounts of new cell membrane, expanding their surface areas by many thousand-fold. However, how oligodendrocytes add new membrane to build or remodel myelin is not fully understood. Here, we show that CNS myelin membrane addition requires exocytosis mediated by the vesicular SNARE proteins VAMP2/3. Genetic inactivation of VAMP2/3 in myelinating oligodendrocytes caused severe hypomyelination and premature death without overt loss of oligodendrocytes. Through live imaging, we discovered that VAMP2/3-mediated exocytosis drives membrane expansion within myelin sheaths to initiate wrapping and power sheath elongation. In conjunction with membrane expansion, mass spectrometry of oligodendrocyte surface proteins revealed that VAMP2/3 incorporates axon-myelin adhesion proteins that are collectively required to form nodes of Ranvier. Together, our results demonstrate that VAMP2/3-mediated membrane expansion in oligodendrocytes is indispensable for myelin formation, uncovering a cellular pathway that could sculpt myelination patterns in response to activity-dependent signals or be therapeutically targeted to promote regeneration in disease.
Collapse
Affiliation(s)
- Mable Lam
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Koji Takeo
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
- Pharmaceutical Research Laboratories, Toray Industries, Inc., Kamakura, Kanagawa, Japan
| | - Rafael G Almeida
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Madeline H Cooper
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Kathryn Wu
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Manasi Iyer
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Husniye Kantarci
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - J Bradley Zuchero
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
5
|
Mukherjee A, Ha P, Wai KC, Naara S. The Role of ECM Remodeling, EMT, and Adhesion Molecules in Cancerous Neural Invasion: Changing Perspectives. Adv Biol (Weinh) 2022; 6:e2200039. [PMID: 35798312 DOI: 10.1002/adbi.202200039] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 05/05/2022] [Indexed: 01/28/2023]
Abstract
Perineural invasion (PNI) refers to the cancerous invasion of nerves. It provides an alternative route for metastatic invasion and can exist independently in the absence of lymphatic or vascular invasion. It is a prominent characteristic of specific aggressive malignancies where it correlates with poor prognosis. The clinical significance of PNI is widely recognized despite a lack of understanding of the molecular mechanisms underlying its pathogenesis. The interaction between the nerve and the cancer cells is the most pivotal PNI step which is mediated by the activation or inhibition of multiple signaling pathways that include chemokines, interleukins, nerve growth factors, and matrix metalloproteinases, to name a few. The nerve-cancer cell interaction brings about specific changes in the perineural niche, which not only affects the regular nerve functions, but also enhances the migratory, invasive, and adherent properties of the tumor cells. This review aims to elucidate the vital role of adhesion molecules, extracellular matrix, and epithelial-mesenchymal proteins that promote PNI, which may serve as therapeutic targets in the future.
Collapse
Affiliation(s)
- Abhishek Mukherjee
- Department of Genetics and Developmental BiologyRappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, 3525422, Israel
| | - Patrick Ha
- Department of Otolaryngology-Head and Neck Surgery, University of California-San Francisco, Helen Diller Family Comprehensive Cancer Center, San Francisco, CA, 94158, USA
| | - Katherine C Wai
- Department of Otolaryngology-Head and Neck Surgery, University of California-San Francisco, Helen Diller Family Comprehensive Cancer Center, San Francisco, CA, 94158, USA
| | - Shorook Naara
- Department of Genetics and Developmental BiologyRappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, 3525422, Israel.,Department of Otolaryngology-Head and Neck Surgery, University of California-San Francisco, Helen Diller Family Comprehensive Cancer Center, San Francisco, CA, 94158, USA
| |
Collapse
|
6
|
Yu W, Li Y, Hu J, Wu J, Huang Y. A Study on the Pathogenesis of Vascular Cognitive Impairment and Dementia: The Chronic Cerebral Hypoperfusion Hypothesis. J Clin Med 2022; 11:jcm11164742. [PMID: 36012981 PMCID: PMC9409771 DOI: 10.3390/jcm11164742] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 07/27/2022] [Accepted: 08/02/2022] [Indexed: 11/16/2022] Open
Abstract
The pathogenic mechanisms underlying vascular cognitive impairment and dementia (VCID) remain controversial due to the heterogeneity of vascular causes and complexity of disease neuropathology. However, one common feature shared among all these vascular causes is cerebral blood flow (CBF) dysregulation, and chronic cerebral hypoperfusion (CCH) is the universal consequence of CBF dysregulation, which subsequently results in an insufficient blood supply to the brain, ultimately contributing to VCID. The purpose of this comprehensive review is to emphasize the important contributions of CCH to VCID and illustrate the current findings about the mechanisms involved in CCH-induced VCID pathological changes. Specifically, evidence is mainly provided to support the molecular mechanisms, including Aβ accumulation, inflammation, oxidative stress, blood-brain barrier (BBB) disruption, trophic uncoupling and white matter lesions (WMLs). Notably, there are close interactions among these multiple mechanisms, and further research is necessary to elucidate the hitherto unsolved questions regarding these interactions. An enhanced understanding of the pathological features in preclinical models could provide a theoretical basis, ultimately achieving the shift from treatment to prevention.
Collapse
Affiliation(s)
- Weiwei Yu
- Department of Neurology, Peking University Shenzhen Hospital, 1120 Lianhua Road, Futian District, Shenzhen 518036, China
| | - Yao Li
- Department of Neurology, Peking University Shenzhen Hospital, 1120 Lianhua Road, Futian District, Shenzhen 518036, China
| | - Jun Hu
- Department of Neurology, Peking University Shenzhen Hospital, 1120 Lianhua Road, Futian District, Shenzhen 518036, China
| | - Jun Wu
- Department of Neurology, Peking University Shenzhen Hospital, 1120 Lianhua Road, Futian District, Shenzhen 518036, China
- Correspondence: (J.W.); (Y.H.); Tel.: +86-0755-8392-2833 (J.W.); +86-010-83572857 (Y.H.)
| | - Yining Huang
- Department of Neurology, Peking University First Hospital, 8 Xishiku Street Xicheng District, Beijing 100034, China
- Correspondence: (J.W.); (Y.H.); Tel.: +86-0755-8392-2833 (J.W.); +86-010-83572857 (Y.H.)
| |
Collapse
|
7
|
Zhang Q, Li Y, Zhuo Y. Synaptic or Non-synaptic? Different Intercellular Interactions with Retinal Ganglion Cells in Optic Nerve Regeneration. Mol Neurobiol 2022; 59:3052-3072. [PMID: 35266115 PMCID: PMC9016027 DOI: 10.1007/s12035-022-02781-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 02/24/2022] [Indexed: 12/31/2022]
Abstract
Axons of adult neurons in the mammalian central nervous system generally fail to regenerate by themselves, and few if any therapeutic options exist to reverse this situation. Due to a weak intrinsic potential for axon growth and the presence of strong extrinsic inhibitors, retinal ganglion cells (RGCs) cannot regenerate their axons spontaneously after optic nerve injury and eventually undergo apoptosis, resulting in permanent visual dysfunction. Regarding the extracellular environment, research to date has generally focused on glial cells and inflammatory cells, while few studies have discussed the potentially significant role of interneurons that make direct connections with RGCs as part of the complex retinal circuitry. In this study, we provide a novel angle to summarize these extracellular influences following optic nerve injury as "intercellular interactions" with RGCs and classify these interactions as synaptic and non-synaptic. By discussing current knowledge of non-synaptic (glial cells and inflammatory cells) and synaptic (mostly amacrine cells and bipolar cells) interactions, we hope to accentuate the previously neglected but significant effects of pre-synaptic interneurons and bring unique insights into future pursuit of optic nerve regeneration and visual function recovery.
Collapse
Affiliation(s)
- Qi Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Yiqing Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, Guangzhou, 510060, China.
| | - Yehong Zhuo
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, Guangzhou, 510060, China.
| |
Collapse
|
8
|
The oligodendrocyte-enriched orphan G protein-coupled receptor Gpr62 is dispensable for central nervous system myelination. Neural Dev 2021; 16:6. [PMID: 34844642 PMCID: PMC8630896 DOI: 10.1186/s13064-021-00156-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 11/15/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Myelination is a highly regulated process in the vertebrate central nervous system (CNS) whereby oligodendrocytes wrap axons with multiple layers of insulating myelin in order to allow rapid electrical conduction. Establishing the proper pattern of myelin in neural circuits requires communicative axo-glial interactions, however, the molecular interactions that occur between oligodendrocytes and axons during developmental myelination and myelin maintenance remain to be fully elucidated. Our previous work identified G protein-coupled receptor 62 (Gpr62), an uncharacterized orphan g-protein coupled receptor, as being selectively expressed by mature oligodendrocytes within the CNS, suggesting a potential role in myelination or axoglial interactions. However, no studies to date have assessed the functional requirement for Gpr62 in oligodendrocyte development or CNS myelination. METHODS To address this, we generated a knockout mouse strain lacking the Gpr62 gene. We assessed CNS myelination during both postnatal development and adulthood using immunohistochemistry, electron microscopy and western blot. In addition, we utilized AAV-mediated expression of a tagged Gpr62 in oligodendrocytes to determine the subcellular localization of the protein in vivo. RESULTS We find that virally expressed Gpr62 protein is selectively expressed on the adaxonal myelin layer, suggestive of a potential role for Gpr62 in axo-myelinic signaling. Nevertheless, Gpr62 knockout mice display normal oligodendrocyte numbers and apparently normal myelination within the CNS during both postnatal development and adulthood. CONCLUSIONS We conclude that in spite of being well-placed to mediate neuronal-oligodendrocyte communications, Gpr62 is overall dispensable for CNS myelination.
Collapse
|
9
|
Schäfer D, Henze J, Pfeifer R, Schleicher A, Brauner J, Mockel-Tenbrinck N, Barth C, Gudert D, Al Rawashdeh W, Johnston ICD, Hardt O. A Novel Siglec-4 Derived Spacer Improves the Functionality of CAR T Cells Against Membrane-Proximal Epitopes. Front Immunol 2020; 11:1704. [PMID: 32849600 PMCID: PMC7426717 DOI: 10.3389/fimmu.2020.01704] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 06/25/2020] [Indexed: 11/13/2022] Open
Abstract
A domain that is often neglected in the assessment of chimeric antigen receptor (CAR) functionality is the extracellular spacer module. However, several studies have elucidated that membrane proximal epitopes are best targeted through CARs comprising long spacers, while short spacer CARs exhibit highest activity on distal epitopes. This finding can be explained by the requirement to have an optimal distance between the effector T cell and target cell. Commonly used long spacer domains are the CH2-CH3 domains of IgG molecules. However, CARs containing these spacers generally show inferior in vivo efficacy in mouse models compared to their observed in vitro activity, which is linked to unspecific Fcγ-Receptor binding and can be abolished by mutating the respective regions. Here, we first assessed a CAR therapy targeting membrane proximal CD20 using such a modified long IgG1 spacer. However, despite these mutations, this construct failed to unfold its observed in vitro cytotoxic potential in an in vivo model, while a shorter but less structured CD8α spacer CAR showed complete tumor clearance. Given the shortage of well-described long spacer domains with a favorable functionality profile, we designed a novel class of CAR spacers with similar attributes to IgG spacers but without unspecific off-target binding, derived from the Sialic acid-binding immunoglobulin-type lectins (Siglecs). Of five constructs tested, a Siglec-4 derived spacer showed highest cytotoxic potential and similar performance to a CD8α spacer in a CD20 specific CAR setting. In a pancreatic ductal adenocarcinoma model, a Siglec-4 spacer CAR targeting a membrane proximal (TSPAN8) epitope was efficiently engaged in vitro, while a membrane distal (CD66c) epitope did not activate the T cell. Transfer of the TSPAN8 specific Siglec-4 spacer CAR to an in vivo setting maintained the excellent tumor killing characteristics being indistinguishable from a TSPAN8 CD8α spacer CAR while outperforming an IgG4 long spacer CAR and, at the same time, showing an advantageous central memory CAR T cell phenotype with lower release of inflammatory cytokines. In summary, we developed a novel spacer that combines cytotoxic potential with an advantageous T cell and cytokine release phenotype, which make this an interesting candidate for future clinical applications.
Collapse
Affiliation(s)
- Daniel Schäfer
- Translational Molecular Imaging, Institute for Diagnostic and Interventional Radiology & Clinic for Hematology and Medical Oncology, University Medical Center Göttingen, Göttingen, Germany.,R&D Reagents, Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany
| | - Janina Henze
- Translational Molecular Imaging, Institute for Diagnostic and Interventional Radiology & Clinic for Hematology and Medical Oncology, University Medical Center Göttingen, Göttingen, Germany.,R&D Reagents, Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany
| | - Rita Pfeifer
- R&D Reagents, Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany
| | - Anna Schleicher
- Faculty of Chemistry and Biosciences, Karlsruher Institute of Technology, Karlsruhe, Germany
| | - Janina Brauner
- R&D Reagents, Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany
| | | | - Carola Barth
- R&D Reagents, Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany
| | - Daniela Gudert
- R&D Reagents, Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany
| | | | - Ian C D Johnston
- R&D Reagents, Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany
| | - Olaf Hardt
- R&D Reagents, Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany
| |
Collapse
|
10
|
Scapin C, Ferri C, Pettinato E, Zambroni D, Bianchi F, Del Carro U, Belin S, Caruso D, Mitro N, Pellegatta M, Taveggia C, Schwab MH, Nave KA, Feltri ML, Wrabetz L, D'Antonio M. Enhanced axonal neuregulin-1 type-III signaling ameliorates neurophysiology and hypomyelination in a Charcot-Marie-Tooth type 1B mouse model. Hum Mol Genet 2020; 28:992-1006. [PMID: 30481294 DOI: 10.1093/hmg/ddy411] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 10/30/2018] [Accepted: 11/22/2018] [Indexed: 12/11/2022] Open
Abstract
Charcot-Marie-Tooth (CMT) neuropathies are a group of genetic disorders that affect the peripheral nervous system with heterogeneous pathogenesis and no available treatment. Axonal neuregulin 1 type III (Nrg1TIII) drives peripheral nerve myelination by activating downstream signaling pathways such as PI3K/Akt and MAPK/Erk that converge on master transcriptional regulators of myelin genes, such as Krox20. We reasoned that modulating Nrg1TIII activity may constitute a general therapeutic strategy to treat CMTs that are characterized by reduced levels of myelination. Here we show that genetic overexpression of Nrg1TIII ameliorates neurophysiological and morphological parameters in a mouse model of demyelinating CMT1B, without exacerbating the toxic gain-of-function that underlies the neuropathy. Intriguingly, the mechanism appears not to be related to Krox20 or myelin gene upregulation, but rather to a beneficial rebalancing in the stoichiometry of myelin lipids and proteins. Finally, we provide proof of principle that stimulating Nrg1TIII signaling, by pharmacological suppression of the Nrg1TIII inhibitor tumor necrosis factor-alpha-converting enzyme (TACE/ADAM17), also ameliorates the neuropathy. Thus, modulation of Nrg1TIII by TACE/ADAM17 inhibition may represent a general treatment for hypomyelinating neuropathies.
Collapse
Affiliation(s)
| | | | | | | | - Francesca Bianchi
- INSPE, Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Ubaldo Del Carro
- INSPE, Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | | | - Donatella Caruso
- DiSFeB-Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Nico Mitro
- DiSFeB-Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Marta Pellegatta
- INSPE, Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Carla Taveggia
- INSPE, Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Markus H Schwab
- Max Planck Institute for Experimental Medicine, 37075 Göttingen, Germany.,Cellular Neurophysiology, Hannover Medical School, Hannover, Germany
| | - Klaus-Armin Nave
- Max Planck Institute for Experimental Medicine, 37075 Göttingen, Germany
| | - M Laura Feltri
- DIBIT, Divisions of Genetics and Cell Biology.,Hunter James Kelly Research Institute.,Department of Neurology.,Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - Lawrence Wrabetz
- DIBIT, Divisions of Genetics and Cell Biology.,Hunter James Kelly Research Institute.,Department of Neurology.,Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | | |
Collapse
|
11
|
Torii T, Miyamoto Y, Yamauchi J. Cellular Signal-Regulated Schwann Cell Myelination and Remyelination. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1190:3-22. [PMID: 31760634 DOI: 10.1007/978-981-32-9636-7_1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Increasing studies have demonstrated multiple signaling molecules responsible for oligodendrocytes and Schwann cells development such as migration, differentiation, myelination, and axo-glial interaction. However, complicated roles in these events are still poorly understood. This chapter focuses on well established intracellular signaling transduction and recent topics that control myelination and are elucidated from accumulating evidences. The underlying molecular mechanisms, which involved in membrane trafficking through small GTPase Arf6 and its activator cytohesins, demonstrate the crosstalk between well established intracellular signaling transduction and a new finding signaling pathway in glial cells links to physiological phenotype and essential role in peripheral nerve system (PNS). Since Arf family proteins affect the expression levels of myelin protein zero (MPZ) and Krox20, which is a transcription factor regulatory factor in early developmental stages of Schwann cells, Arf proteins likely to be key regulator for Schwann cells development. Herein, we discuss how intracellular signaling transductions in Schwann cells associate with myelination in CNS and PNS.
Collapse
Affiliation(s)
- Tomohiro Torii
- Graduate School of Brain Science, Doshisha University, Kyotanabe-shi, Kyoto, Japan
| | - Yuki Miyamoto
- Department of Pharmacology, National Research Institute for Child Health and Development, Setagaya, Tokyo, Japan
| | - Junji Yamauchi
- Laboratory of Molecular Neuroscience and Neurology, Tokyo University of Pharmacy and Life Science, Hachioji, Tokyo, Japan.
| |
Collapse
|
12
|
Urban MW, Ghosh B, Block CG, Charsar BA, Smith GM, Wright MC, Li S, Lepore AC. Protein Tyrosine Phosphatase σ Inhibitory Peptide Promotes Recovery of Diaphragm Function and Sprouting of Bulbospinal Respiratory Axons after Cervical Spinal Cord Injury. J Neurotrauma 2019; 37:572-579. [PMID: 31392919 DOI: 10.1089/neu.2019.6586] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Damage to respiratory neural circuitry and consequent loss of diaphragm function is a major cause of morbidity and mortality after cervical spinal cord injury (SCI). Upon SCI, inspiratory signals originating in the medullary rostral ventral respiratory group (rVRG) become disrupted from their phrenic motor neuron (PhMN) targets, resulting in diaphragm paralysis. Limited growth of both damaged and spared axon populations occurs after central nervous system trauma attributed, in part, to expression of various growth inhibitory molecules, some that act through direct interaction with the protein tyrosine phosphatase sigma (PTPσ) receptor located on axons. In the rat model of C2 hemisection SCI, we aimed to block PTPσ signaling to investigate potential mechanisms of axon plasticity and respiratory recovery using a small molecule peptide mimetic that inhibits PTPσ. The peptide was soaked into a biocompatible gelfoam and placed directly over the injury site immediately after hemisection and replaced with a freshly soaked piece 1 week post-SCI. At 8 weeks post-hemisection, PTPσ peptide significantly improved ipsilateral hemidiaphragm function, as assessed in vivo with electromyography recordings. PTPσ peptide did not promote regeneration of axotomized rVRG fibers originating in ipsilateral medulla, as assessed by tracing after adeno-associated virus serotype 2/mCherry injection into the rVRG. Conversely, PTPσ peptide stimulated robust sprouting of contralateral-originating rVRG fibers and serotonergic axons within the PhMN pool ipsilateral to hemisection. Further, relesion through the hemisection did not compromise diaphragm recovery, suggesting that PTPσ peptide-induced restoration of function was attributed to plasticity of spared axon pathways descending in contralateral spinal cord. These data demonstrate that inhibition of PTPσ signaling can promote significant recovery of diaphragm function after SCI by stimulating plasticity of critical axon populations spared by the injury and consequently enhancing descending excitatory input to PhMNs.
Collapse
Affiliation(s)
- Mark W Urban
- Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Biswarup Ghosh
- Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Cole G Block
- Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Brittany A Charsar
- Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania
| | - George M Smith
- Department of Neuroscience, Shriners Hospitals Pediatric Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Megan C Wright
- Department of Biology, Arcadia University, Glenside, Pennsylvania
| | - Shuxin Li
- Department of Neuroscience, Shriners Hospitals Pediatric Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Angelo C Lepore
- Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania
| |
Collapse
|
13
|
Fratta P, Ornaghi F, Dati G, Zambroni D, Saveri P, Belin S, D'Adamo P, Shy M, Quattrini A, Laura Feltri M, Wrabetz L. A nonsense mutation in myelin protein zero causes congenital hypomyelination neuropathy through altered P0 membrane targeting and gain of abnormal function. Hum Mol Genet 2019; 28:124-132. [PMID: 30239779 PMCID: PMC6298235 DOI: 10.1093/hmg/ddy336] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 09/18/2018] [Indexed: 12/13/2022] Open
Abstract
Protein zero (P0) is the major structural protein in peripheral myelin, and mutations in the Myelin Protein Zero (Mpz) gene produce wide-ranging hereditary neuropathy phenotypes. To gain insight in the mechanisms underlying a particularly severe form, congenital hypomyelination (CH), we targeted mouse Mpz to encode P0Q215X, a nonsense mutation associated with the disease, that we show escapes nonsense mediated decay and is expressed in CH patient nerves. The knock-in mice express low levels of the resulting truncated protein, producing a milder phenotype when compared to patients, allowing to dissect the subtle pathogenic mechanisms occurring in otherwise very compromised peripheral myelin. We find that P0Q215X does not elicit an unfolded protein response, which is a key mechanism for other pathogenic MPZ mutations, but is instead in part aberrantly trafficked to non-myelin plasma membranes and induces defects in radial sorting of axons by Schwann cells. We show that the loss of the C-terminal Tyr-Ala-Met-Leu motif is responsible for P0 mislocalization, as its addition is able to restore correct P0Q215X trafficking in vitro. Lastly, we show that P0Q215X acts through dose-dependent gain of abnormal function, as wild-type P0 is unable to rescue the hypomyelination phenotype. Collectively, these data indicate that alterations at the premyelinating stage, linked to altered targeting of P0, may be responsible for CH, and that different types of gain of abnormal function produce the diverse neuropathy phenotypes associated with MPZ, supporting future allele-specific therapeutic silencing strategies.
Collapse
Affiliation(s)
- Pietro Fratta
- UCL Institute of Neurology, Queen Square, London WC1N, UK.,IRCCS San Raffaele Scientific Institute, DIBIT, Milan, Italy
| | - Francesca Ornaghi
- IRCCS San Raffaele Scientific Institute, DIBIT, Milan, Italy.,SR-TIGET, Milan, Italy.,Departments of Neurology and Biochemistry, Jacobs School of Medicine and Biomedical Sciences, Hunter James Kelly Research Institute, State University of New York at Buffalo, Buffalo, NY, USA
| | - Gabriele Dati
- IRCCS San Raffaele Scientific Institute, DIBIT, Milan, Italy
| | | | - Paola Saveri
- IRCCS San Raffaele Scientific Institute, DIBIT, Milan, Italy.,Unit of Rare Neurodegenerative and Neurometabolic Diseases, Department of Clinical Neurosciences, C. Besta Neurological Institute IRCCS Foundation, Milan, Italy
| | - Sophie Belin
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College Department of Neuroscience and Experimental Therapeutics, Albany, NY, USA.,Departments of Neurology and Biochemistry, Jacobs School of Medicine and Biomedical Sciences, Hunter James Kelly Research Institute, State University of New York at Buffalo, Buffalo, NY, USA
| | | | - Michael Shy
- Department of Neurology, University of Iowa, Iowa City, IA, USA
| | | | - M Laura Feltri
- IRCCS San Raffaele Scientific Institute, DIBIT, Milan, Italy.,Departments of Neurology and Biochemistry, Jacobs School of Medicine and Biomedical Sciences, Hunter James Kelly Research Institute, State University of New York at Buffalo, Buffalo, NY, USA
| | - Lawrence Wrabetz
- IRCCS San Raffaele Scientific Institute, DIBIT, Milan, Italy.,Departments of Neurology and Biochemistry, Jacobs School of Medicine and Biomedical Sciences, Hunter James Kelly Research Institute, State University of New York at Buffalo, Buffalo, NY, USA
| |
Collapse
|
14
|
Malheiro AR, Correia B, Ferreira da Silva T, Bessa-Neto D, Van Veldhoven PP, Brites P. Leukodystrophy caused by plasmalogen deficiency rescued by glyceryl 1-myristyl ether treatment. Brain Pathol 2019; 29:622-639. [PMID: 30667116 DOI: 10.1111/bpa.12710] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 01/16/2019] [Indexed: 12/29/2022] Open
Abstract
Plasmalogens are the most abundant form of ether phospholipids in myelin and their deficiency causes Rhizomelic Chondrodysplasia Punctata (RCDP), a severe developmental disorder. Using the Gnpat-knockout (KO) mouse as a model of RCDP, we determined the consequences of a plasmalogen deficiency during myelination and myelin homeostasis in the central nervous system (CNS). We unraveled that the lack of plasmalogens causes a generalized hypomyelination in several CNS regions including the optic nerve, corpus callosum and spinal cord. The defect in myelin content evolved to a progressive demyelination concomitant with generalized astrocytosis and white matter-selective microgliosis. Oligodendrocyte precursor cells (OPC) and mature oligodendrocytes were abundant in the CNS of Gnpat KO mice during the active period of demyelination. Axonal loss was minimal in plasmalogen-deficient mice, although axonal damage was observed in spinal cords from aged Gnpat KO mice. Characterization of the plasmalogen-deficient myelin identified myelin basic protein and septin 7 as early markers of dysmyelination, whereas myelin-associated glycoprotein was associated with the active demyelination phase. Using in vitro myelination assays, we unraveled that the intrinsic capacity of oligodendrocytes to ensheath and initiate membrane wrapping requires plasmalogens. The defect in plasmalogens was rescued with glyceryl 1-myristyl ether [1-O-tetradecyl glycerol (1-O-TDG)], a novel alternative precursor in the plasmalogen biosynthesis pathway. 1-O-TDG treatment rescued myelination in plasmalogen-deficient oligodendrocytes and in mutant mice. Our results demonstrate the importance of plasmalogens for oligodendrocyte function and myelin assembly, and identified a novel strategy to promote myelination in nervous tissue.
Collapse
Affiliation(s)
- Ana R Malheiro
- Neurolipid Biology, Instituto de Investigação e Inovação em Saúde - i3S, Instituto de Biologia Molecular e Celular - IBMC e Universidade do Porto, Porto, Portugal.,ICBAS, Instituto Ciências Biomédicas Abel Salazar, Porto, Portugal
| | - Barbara Correia
- Neurolipid Biology, Instituto de Investigação e Inovação em Saúde - i3S, Instituto de Biologia Molecular e Celular - IBMC e Universidade do Porto, Porto, Portugal
| | - Tiago Ferreira da Silva
- Neurolipid Biology, Instituto de Investigação e Inovação em Saúde - i3S, Instituto de Biologia Molecular e Celular - IBMC e Universidade do Porto, Porto, Portugal
| | - Diogo Bessa-Neto
- Neurolipid Biology, Instituto de Investigação e Inovação em Saúde - i3S, Instituto de Biologia Molecular e Celular - IBMC e Universidade do Porto, Porto, Portugal
| | - Paul P Van Veldhoven
- Laboratory of Lipid Biochemistry and Protein Interactions (LIPIT), KU Leuven, Leuven, Belgium
| | - Pedro Brites
- Neurolipid Biology, Instituto de Investigação e Inovação em Saúde - i3S, Instituto de Biologia Molecular e Celular - IBMC e Universidade do Porto, Porto, Portugal
| |
Collapse
|
15
|
The Extracellular Environment of the CNS: Influence on Plasticity, Sprouting, and Axonal Regeneration after Spinal Cord Injury. Neural Plast 2018; 2018:2952386. [PMID: 29849554 PMCID: PMC5932463 DOI: 10.1155/2018/2952386] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 01/22/2018] [Accepted: 02/06/2018] [Indexed: 11/17/2022] Open
Abstract
The extracellular environment of the central nervous system (CNS) becomes highly structured and organized as the nervous system matures. The extracellular space of the CNS along with its subdomains plays a crucial role in the function and stability of the CNS. In this review, we have focused on two components of the neuronal extracellular environment, which are important in regulating CNS plasticity including the extracellular matrix (ECM) and myelin. The ECM consists of chondroitin sulfate proteoglycans (CSPGs) and tenascins, which are organized into unique structures called perineuronal nets (PNNs). PNNs associate with the neuronal cell body and proximal dendrites of predominantly parvalbumin-positive interneurons, forming a robust lattice-like structure. These developmentally regulated structures are maintained in the adult CNS and enhance synaptic stability. After injury, however, CSPGs and tenascins contribute to the structure of the inhibitory glial scar, which actively prevents axonal regeneration. Myelin sheaths and mature adult oligodendrocytes, despite their important role in signal conduction in mature CNS axons, contribute to the inhibitory environment existing after injury. As such, unlike the peripheral nervous system, the CNS is unable to revert to a “developmental state” to aid neuronal repair. Modulation of these external factors, however, has been shown to promote growth, regeneration, and functional plasticity after injury. This review will highlight some of the factors that contribute to or prevent plasticity, sprouting, and axonal regeneration after spinal cord injury.
Collapse
|
16
|
Díez-Revuelta N, Higuero AM, Velasco S, Peñas-de-la-Iglesia M, Gabius HJ, Abad-Rodríguez J. Neurons define non-myelinated axon segments by the regulation of galectin-4-containing axon membrane domains. Sci Rep 2017; 7:12246. [PMID: 28947766 PMCID: PMC5612983 DOI: 10.1038/s41598-017-12295-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 09/01/2017] [Indexed: 01/01/2023] Open
Abstract
The mechanism underlying selective myelination of axons versus dendrites or neuronal somata relies on the expression of somatodendritic membrane myelination inhibitors (i.e. JAM2). However, axons still present long unmyelinated segments proposed to contribute to axonal plasticity and higher order brain functions. Why these segments remain unmyelinated is still an unresolved issue. The bifunctional lectin galectin-4 (Gal-4) organizes the transport of axon glycoproteins by binding to N-acetyllactosamine (LacNac) termini of N-glycans. We have shown that Gal-4 is sorted to segmental domains (G4Ds) along the axon surface, reminiscent of these long unmyelinated axon segments in cortical neurons. We report here that oligodendrocytes (OLGs) do not deposit myelin on Gal-4 covered surfaces or myelinate axonal G4Ds. In addition, Gal-4 interacts and co-localizes in G4Ds with contactin-1, a marker of another type of non-myelinated segments, the nodes of Ranvier. Neither Gal-4 expression nor G4D dimensions are affected by myelin extracts or myelinating OLGs, but are reduced with neuron maturation. As in vitro, Gal-4 is consistently segregated from myelinated structures in the brain. Our data shape the novel concept that neurons establish axon membrane domains expressing Gal-4, the first inhibitor of myelination identified in axons, whose regulated boundaries delineate myelination-incompetent axon segments along development.
Collapse
Affiliation(s)
- Natalia Díez-Revuelta
- Membrane Biology and Axonal Repair Laboratory, Hospital Nacional de Parapléjicos (SESCAM), Finca La Peraleda s/n, E-45071, Toledo, Spain
| | - Alonso M Higuero
- Membrane Biology and Axonal Repair Laboratory, Hospital Nacional de Parapléjicos (SESCAM), Finca La Peraleda s/n, E-45071, Toledo, Spain
| | - Silvia Velasco
- Membrane Biology and Axonal Repair Laboratory, Hospital Nacional de Parapléjicos (SESCAM), Finca La Peraleda s/n, E-45071, Toledo, Spain
| | - María Peñas-de-la-Iglesia
- Membrane Biology and Axonal Repair Laboratory, Hospital Nacional de Parapléjicos (SESCAM), Finca La Peraleda s/n, E-45071, Toledo, Spain
| | - Hans-Joachim Gabius
- Institut für Physiologische Chemie, Tierärztliche Fakultät, Ludwig-Maximilians-Universität, Veterinärstr. 13, D-80539, München, Germany
| | - José Abad-Rodríguez
- Membrane Biology and Axonal Repair Laboratory, Hospital Nacional de Parapléjicos (SESCAM), Finca La Peraleda s/n, E-45071, Toledo, Spain.
| |
Collapse
|
17
|
Granados-Durán P, López-Ávalos MD, Cifuentes M, Pérez-Martín M, Fernández-Arjona MDM, Hughes TR, Johnson K, Morgan BP, Fernández-Llebrez P, Grondona JM. Microbial Neuraminidase Induces a Moderate and Transient Myelin Vacuolation Independent of Complement System Activation. Front Neurol 2017; 8:78. [PMID: 28326060 PMCID: PMC5339270 DOI: 10.3389/fneur.2017.00078] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 02/20/2017] [Indexed: 02/05/2023] Open
Abstract
AIMS Some central nervous system pathogens express neuraminidase (NA) on their surfaces. In the rat brain, a single intracerebroventricular (ICV) injection of NA induces myelin vacuolation in axonal tracts. Here, we explore the nature, the time course, and the role of the complement system in this damage. METHODS The spatiotemporal analysis of myelin vacuolation was performed by optical and electron microscopy. Myelin basic protein-positive area and oligodendrocyte transcription factor (Olig2)-positive cells were quantified in the damaged bundles. Neuronal death in the affected axonal tracts was assessed by Fluoro-Jade B and anti-caspase-3 staining. To evaluate the role of the complement, membrane attack complex (MAC) deposition on damaged bundles was analyzed using anti-C5b9. Rats ICV injected with the anaphylatoxin C5a were studied for myelin damage. In addition, NA-induced vacuolation was studied in rats with different degrees of complement inhibition: normal rats treated with anti-C5-blocking antibody and C6-deficient rats. RESULTS The stria medullaris, the optic chiasm, and the fimbria were the most consistently damaged axonal tracts. Vacuolation peaked 7 days after NA injection and reverted by day 15. Olig2+ cell number in the damaged tracts was unaltered, and neurodegeneration associated with myelin alterations was not detected. MAC was absent on damaged axonal tracts, as revealed by C5b9 immunostaining. Rats ICV injected with the anaphylatoxin C5a displayed no myelin injury. When the complement system was experimentally or constitutively inhibited, NA-induced myelin vacuolation was similar to that observed in normal rats. CONCLUSION Microbial NA induces a moderate and transient myelin vacuolation that is not caused either by neuroinflammation or complement system activation.
Collapse
Affiliation(s)
- Pablo Granados-Durán
- Laboratorio de Fisiología Animal, Facultad de Ciencias, Departamento de Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga , Málaga , Spain
| | - María Dolores López-Ávalos
- Laboratorio de Fisiología Animal, Facultad de Ciencias, Departamento de Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga , Málaga , Spain
| | - Manuel Cifuentes
- Laboratorio de Fisiología Animal, Facultad de Ciencias, Departamento de Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga, Málaga, Spain; Centro de Investigaciones Biomédicas en Red de Bioingeniería, Biomateriales y Nanomedicina, CIBER BBN, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain
| | - Margarita Pérez-Martín
- Laboratorio de Fisiología Animal, Facultad de Ciencias, Departamento de Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga , Málaga , Spain
| | - María Del Mar Fernández-Arjona
- Laboratorio de Fisiología Animal, Facultad de Ciencias, Departamento de Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga , Málaga , Spain
| | - Timothy R Hughes
- Division of Infection and Immunity, School of Medicine, Cardiff University , Cardiff , UK
| | | | - B Paul Morgan
- Division of Infection and Immunity, School of Medicine, Cardiff University , Cardiff , UK
| | - Pedro Fernández-Llebrez
- Laboratorio de Fisiología Animal, Facultad de Ciencias, Departamento de Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga , Málaga , Spain
| | - Jesús M Grondona
- Laboratorio de Fisiología Animal, Facultad de Ciencias, Departamento de Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga , Málaga , Spain
| |
Collapse
|
18
|
Dell'Anno MT, Strittmatter SM. Rewiring the spinal cord: Direct and indirect strategies. Neurosci Lett 2016; 652:25-34. [PMID: 28007647 DOI: 10.1016/j.neulet.2016.12.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 11/15/2016] [Accepted: 12/02/2016] [Indexed: 12/23/2022]
Abstract
Spinal cord injury is currently incurable. Treatment is limited to minimizing secondary complications and maximizing residual function by rehabilitation. Neurologic recovery is prevented by the poor intrinsic regenerative capacity of neurons in the adult central nervous system and by the presence of growth inhibitors in the adult brain and spinal cord. Here we identify three approaches to rewire the spinal cord after injury: axonal regeneration (direct endogenous reconnection), axonal sprouting (indirect endogenous reconnection) and neural stem cell transplantation (indirect exogenous reconnection). Regeneration and sprouting of axonal fibers can be both enhanced through the neutralization of myelin- and extracellular matrix-associated inhibitors described in the first part of this review. Alternatively, in the second part we focus on the formation of a novel circuit through the grafting of neural stem cells in the lesion site. Transplanted neural stem cells differentiate in vivo into neurons and glial cells which form an intermediate station between the rostral and caudal segment of the recipient spinal cord. In particular, here we describe how neural stem cells-derived neurons are endowed with the ability to extend long-distance axons to regain the transmission of motor and sensory information.
Collapse
Affiliation(s)
- Maria Teresa Dell'Anno
- Program in Cellular Neuroscience, Neurodegeneration & Repair, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Stephen M Strittmatter
- Program in Cellular Neuroscience, Neurodegeneration & Repair, Yale University School of Medicine, New Haven, CT 06536, USA.
| |
Collapse
|
19
|
Palavicini JP, Wang C, Chen L, Ahmar S, Higuera JD, Dupree JL, Han X. Novel molecular insights into the critical role of sulfatide in myelin maintenance/function. J Neurochem 2016; 139:40-54. [PMID: 27417284 DOI: 10.1111/jnc.13738] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Revised: 06/17/2016] [Accepted: 06/22/2016] [Indexed: 01/19/2023]
Abstract
Cerebroside sulfotransferase (CST) catalyzes the production of sulfatide, a major class of myelin-specific lipids. CST knockout (CST(-/-) ) mice in which sulfatide is completely depleted are born healthy, but display myelin abnormalities and progressive tremors starting at 4-6 weeks of age. Although these phenotypes suggest that sulfatide plays a critical role in myelin maintenance/function, the underlying mechanisms remain largely unknown. We analyzed the major CNS myelin proteins and the major lipids enriched in the myelin in a spatiotemporal manner. We found a one-third reduction of the major compact myelin proteins (myelin basic protein, myelin basic protein, and proteolipid protein, PLP) and an equivalent post-developmental loss of myelin lipids, providing the molecular basis behind the thinner myelin sheaths. Our lipidomics data demonstrated that the observed global reduction of myelin lipid content was not because of an increase of lipid degradation but rather to the reduction of their synthesis by oligodendrocytes. We also showed that sulfatide depletion leads to region-specific effects on non-compact myelin, dramatically affecting the paranode (neurofascin 155) and the major inner tongue myelin protein (myelin-associated glycoprotein). Moreover, we demonstrated that sulfatide promotes the interaction between adjacent PLP extracellular domains, evidenced by a progressive decline of high molecular weight PLP complexes in CST(-/-) mice, providing an explanation at a molecular level regarding the uncompacted myelin sheaths. Finally, we proposed that the dramatic losses of neurofascin 155 and PLP interactions are responsible for the progressive tremors and eventual ataxia. In summary, we unraveled novel molecular insights into the critical role of sulfatide in myelin maintenance/function. Cerebroside sulfotransferase (CST) catalyzes the production of sulfatide, a major class of myelin-specific lipids. CST knockout (CST(-/-) ) mice in which sulfatide is completely depleted are born healthy, but display myelin abnormalities We show in our study that sulfatide depletion leads to losses of myelin proteins and lipids, and impairment of myelin functions, unraveling novel molecular insights into the critical role of sulfatide in myelin maintenance/function.
Collapse
Affiliation(s)
- Juan Pablo Palavicini
- Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida, USA
| | - Chunyan Wang
- Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida, USA
| | - Linyuan Chen
- Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida, USA
| | - Sareen Ahmar
- Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida, USA
| | - Juan Diego Higuera
- Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida, USA
| | - Jeffrey L Dupree
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, Virginia, USA.,Research Division, McGuire Veterans Affairs Medical Center, Richmond, Virginia, USA
| | - Xianlin Han
- Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida, USA.
| |
Collapse
|
20
|
Papuć E, Kurys-Denis E, Krupski W, Tatara M, Rejdak K. Can Antibodies Against Glial Derived Antigens be Early Biomarkers of Hippocampal Demyelination and Memory Loss in Alzheimer's Disease? J Alzheimers Dis 2016; 48:115-21. [PMID: 26401933 DOI: 10.3233/jad-150309] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) is known to exhibit well characterized pathologies including the extracellular accumulation of amyloid plaques, intra-axonal presence of neurofibrillary tangles, and glial hypertrophy. Nevertheless, the nature of myelin pathology in AD has not been well studied. Recent studies on animal models of AD, however, revealed focal demyelination within amyloid-β plaques in hippocampus. OBJECTIVES In a view of this finding, we decided to assess humoral response against proteins of myelin sheath in AD, in the hope of identifying early biomarkers of memory loss and neuropathological process characteristic of AD. METHODS We assessed antibodies levels against proteins of the myelin sheath: myelin oligodendrocyte glycoprotein (MOG), myelin basic protein (MBP), myelin-associated glycoprotein (MAG), and proteolipoprotein (PLP) in sera of 26 AD patients and 26 healthy controls, using commercially available ELISA system (Mediagnost, Germany). RESULTS In the AD patient subgroup, significantly higher titers were observed for all types of assessed IgG autoantibodies compared to healthy control subjects (anti-MOG, anti-MAG, anti-MBP, anti-PLP). The titers of most of the investigated IgM antibodies were also higher in AD patients (p < 0.05), with the exception of anti-MAG IgM antibodies (p > 0.05). CONCLUSION The study provides the evidence for the significantly increased production of autoantibodies against proteins of myelin sheath in AD. These results can be of importance in the light of emerging data from animal models of AD, indicating early demyelination of hippocampal region. Further studies on larger population are necessary to confirm whether these autoantibodies could serve as early biomarkers of AD in humans.
Collapse
Affiliation(s)
- Ewa Papuć
- Chair and Department, Neurology of Medical University of Lublin, Poland
| | - Ewa Kurys-Denis
- 2nd Department of Radiology, Medical University of Lublin, Poland
| | - Witold Krupski
- 2nd Department of Radiology, Medical University of Lublin, Poland
| | - Marcin Tatara
- 2nd Department of Radiology, Medical University of Lublin, Poland.,Department of Animal Physiology, University of Life Sciences, Lublin, Poland
| | - Konrad Rejdak
- Chair and Department, Neurology of Medical University of Lublin, Poland
| |
Collapse
|
21
|
Trapp BD, Ransohoff RM, Fisher E, Rudick RA. Neurodegeneration in Multiple Sclerosis: Relationship to Neurological Disability. Neuroscientist 2016. [DOI: 10.1177/107385849900500107] [Citation(s) in RCA: 128] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating disease of the CNS. Most MS patients follow a relapsing-remitting course (RR-MS) for 8 to 15 years that transforms into a secondary progressive disease course (SP-MS). In this review, we discuss current data that describe MS as a neurodegenerative disease in which axonal loss is the major cause of irreversible neurological disability in MS patients. Neurological deficits in MS patients have two pathogeneses: acute inflammatory demyelination and axonal degeneration. Disability caused by inflammatory demyelination clinically dominates the early stages of RR-MS and is reversible. Axonal transection occurs at sites of inflammation and begins at disease onset but is clinically silent in RR-MS because the CNS compensates for neuronal loss. Once a threshold of axon loss is ex ceeded, MS patients enter an irreversible secondary progressive stage. In SP-MS, axonal degeneration is caused by chronic demyelination and may be irreversibly progressive. This view of MS provides a concep tional framework that explains conversion of RR-MS to SP-MS and provides a rationale for early aggressive anti-inflammatory and neuroprotective therapies. NEUROSCIENTIST 5:48-57, 1999
Collapse
Affiliation(s)
- Bruce D. Trapp
- Departments of Neurosciences, The Cleveland Clinic Foundation
Cleveland, Ohio
| | - Richard M. Ransohoff
- Departments of Neurosciences, Lerner Research Institute
and the Mellen Center for Multiple Sclerosis Treatment and Research (RMR,
RAR) The Cleveland Clinic Foundation Cleveland, Ohio
| | - Elizabeth Fisher
- Biomedical Engineering, The Cleveland Clinic Foundation
Cleveland, Ohio
| | - Richard A. Rudick
- Departments of Neurosciences, Lerner Research Institute
and the Mellen Center for Multiple Sclerosis Treatment and Research (RMR,
RAR) The Cleveland Clinic Foundation Cleveland, Ohio
| |
Collapse
|
22
|
Gangliosides of the Vertebrate Nervous System. J Mol Biol 2016; 428:3325-3336. [PMID: 27261254 DOI: 10.1016/j.jmb.2016.05.020] [Citation(s) in RCA: 132] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 05/11/2016] [Accepted: 05/20/2016] [Indexed: 12/14/2022]
Abstract
Gangliosides, sialylated glycosphingolipids, found on all vertebrate cells and tissues, are major molecular determinants on the surfaces of vertebrate nerve cells. Composed of a sialylated glycan attached to a ceramide lipid, the same four structures-GM1, GD1a, GD1b, and GT1b-represent the vast majority (>90%) of gangliosides in the brains of all mammals and birds. Primarily found on the outer surface of the plasma membrane with their glycans facing outward, gangliosides associate laterally with each other, sphingomyelin, cholesterol, and select proteins in lipid rafts-the dynamic functional subdomains of the plasma membrane. The functions of gangliosides in the human nervous system are revealed by congenital mutations in ganglioside biosynthetic genes. Mutations in ST3GAL5, which codes for an enzyme early in brain ganglioside biosynthesis, result in an early-onset seizure disorder with profound motor and cognitive decay, whereas mutations in B4GALNT1, a gene encoding a later step, result in hereditary spastic paraplegia accompanied by intellectual deficits. The molecular functions of brain gangliosides include regulation of receptors in the same membrane via lateral (cis) associations and regulation of cell-cell recognition by trans interaction with ganglioside binding proteins on apposing cells. Gangliosides also affect the aggregation of Aβ (Alzheimer's disease) and α-synuclein (Parkinson's Disease). As analytical, biochemical, and genetic tools advance, research on gangliosides promises to reveal mechanisms of molecular control related to nerve and glial cell differentiation, neuronal excitability, axon outgrowth after nervous system injury, and protein folding in neurodegenerative diseases.
Collapse
|
23
|
Al-Bashir N, Mellado W, Filbin MT. Sialic Acid Is Required for Neuronal Inhibition by Soluble MAG but not for Membrane Bound MAG. Front Mol Neurosci 2016; 9:21. [PMID: 27065798 PMCID: PMC4817280 DOI: 10.3389/fnmol.2016.00021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2015] [Accepted: 03/14/2016] [Indexed: 11/13/2022] Open
Abstract
Myelin-Associated Glycoprotein (MAG), a major inhibitor of axonal growth, is a member of the immunoglobulin (Ig) super-family. Importantly, MAG (also known as Siglec-4) is a member of the Siglec family of proteins (sialic acid-binding, immunoglobulin-like lectins), MAG binds to complex gangliosides, specifically GD1a and/or GT1b. Therefore, it has been proposed as neuronal receptors for MAG inhibitory effect of axonal growth. Previously, we showed that MAG binds sialic acid through domain 1 at Arg118 and is able to inhibit axonal growth through domain 5. We developed a neurite outgrowth (NOG) assay, in which both wild type MAG and mutated MAG (MAG Arg118) are expressed on cells. In addition we also developed a soluble form NOG in which we utilized soluble MAG-Fc and mutated MAG (Arg118-Fc). Only MAG-Fc is able to inhibit NOG, but not mutated MAG (Arg118)-Fc that has been mutated at its sialic acid binding site. However, both forms of membrane bound MAG- and MAG (Arg118)- expressing cells still inhibit NOG. Here, we review various results from different groups regarding MAG’s inhibition of axonal growth. Also, we propose a model in which the sialic acid binding is not necessary for the inhibition induced by the membrane form of MAG, but it is necessary for the soluble form of MAG. This finding highlights the importance of understanding the different mechanisms by which MAG inhibits NOG in both the soluble fragmented form and the membrane-bound form in myelin debris following CNS damage.
Collapse
Affiliation(s)
- Najat Al-Bashir
- Biology Department, Hunter College, City University of New York New York, NY, USA
| | - Wilfredo Mellado
- Biology Department, Hunter College, City University of New YorkNew York, NY, USA; Burke-Cornell Medical Research Institute White Plains, NY, USA
| | - Marie T Filbin
- Biology Department, Hunter College, City University of New York New York, NY, USA
| |
Collapse
|
24
|
Liang D, Shi S, Xu J, Zhang B, Qin Y, Ji S, Xu W, Liu J, Liu L, Liu C, Long J, Ni Q, Yu X. New insights into perineural invasion of pancreatic cancer: More than pain. Biochim Biophys Acta Rev Cancer 2016; 1865:111-22. [PMID: 26794395 DOI: 10.1016/j.bbcan.2016.01.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Revised: 12/26/2015] [Accepted: 01/11/2016] [Indexed: 01/01/2023]
Abstract
Pancreatic cancer is one of the most malignant human tumors. Perineural invasion, whereby a cancer cell invades the perineural spaces surrounding nerves, is acknowledged as a gradual contributor to cancer aggressiveness. Furthermore, perineural invasion is considered one of the root causes of the recurrence and metastasis observed after pancreatic resection, and it is also an independent predictor of prognosis. Advanced research has demonstrated that the neural microenvironment is closely associated with perineural invasion in pancreatic cancer. Therapy targeting the molecular mechanism of perineural invasion may enable the durable clinical treatment of this formidable disease. This review provides an overview of the present status of perineural invasion, the relevant molecular mechanisms of perineural invasion, pain and hyperglycemia associated with perineural invasion in pancreatic cancer, and the targeted therapeutics based on these studies.
Collapse
Affiliation(s)
- Dingkong Liang
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Si Shi
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Jin Xu
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Bo Zhang
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Yi Qin
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Shunrong Ji
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Wenyan Xu
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Jiang Liu
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Liang Liu
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Chen Liu
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Jiang Long
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Quanxing Ni
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Xianjun Yu
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Pancreatic Cancer Institute, Fudan University, Shanghai, China.
| |
Collapse
|
25
|
von Büdingen HC, Mei F, Greenfield A, Jahn S, Shen YAA, Reid HH, McKemy DD, Chan JR. The myelin oligodendrocyte glycoprotein directly binds nerve growth factor to modulate central axon circuitry. J Cell Biol 2015; 210:891-8. [PMID: 26347141 PMCID: PMC4576870 DOI: 10.1083/jcb.201504106] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Myelin oligodendrocyte glycoprotein, expressed on the outermost lamellae of the
myelin sheath, is a novel and specific binding partner for NGF that may modulate
local concentrations of the neurotrophin in the spinal cord microenvironment. Myelin oligodendrocyte glycoprotein (MOG) is a central nervous system myelin-specific
molecule expressed on the outer lamellae of myelin. To date, the exact function of
MOG has remained unknown, with MOG knockout mice displaying normal myelin
ultrastructure and no apparent specific phenotype. In this paper, we identify nerve
growth factor (NGF) as a binding partner for MOG and demonstrate that this
interaction is capable of sequestering NGF from TrkA-expressing neurons to modulate
axon growth and survival. Deletion of MOG results in aberrant sprouting of
nociceptive neurons in the spinal cord. Binding of NGF to MOG may offer widespread
implications into mechanisms that underlie pain pathways.
Collapse
|
26
|
Baldwin KT, Giger RJ. Insights into the physiological role of CNS regeneration inhibitors. Front Mol Neurosci 2015; 8:23. [PMID: 26113809 PMCID: PMC4462676 DOI: 10.3389/fnmol.2015.00023] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 05/26/2015] [Indexed: 12/14/2022] Open
Abstract
The growth inhibitory nature of injured adult mammalian central nervous system (CNS) tissue constitutes a major barrier to robust axonal outgrowth and functional recovery following trauma or disease. Prototypic CNS regeneration inhibitors are broadly expressed in the healthy and injured brain and spinal cord and include myelin-associated glycoprotein (MAG), the reticulon family member NogoA, oligodendrocyte myelin glycoprotein (OMgp), and chondroitin sulfate proteoglycans (CSPGs). These structurally diverse molecules strongly inhibit neurite outgrowth in vitro, and have been most extensively studied in the context of nervous system injury in vivo. The physiological role of CNS regeneration inhibitors in the naïve, or uninjured, CNS remains less well understood, but has received growing attention in recent years and is the focus of this review. CNS regeneration inhibitors regulate myelin development and axon stability, consolidate neuronal structure shaped by experience, and limit activity-dependent modification of synaptic strength. Altered function of CNS regeneration inhibitors is associated with neuropsychiatric disorders, suggesting crucial roles in brain development and health.
Collapse
Affiliation(s)
- Katherine T Baldwin
- Department of Cell and Developmental Biology, University of Michigan School of Medicine Ann Arbor, MI, USA ; Cellular and Molecular Biology Graduate Program, University of Michigan School of Medicine Ann Arbor, MI, USA
| | - Roman J Giger
- Department of Cell and Developmental Biology, University of Michigan School of Medicine Ann Arbor, MI, USA ; Department of Neurology, University of Michigan School of Medicine Ann Arbor, MI, USA
| |
Collapse
|
27
|
All AH, Gharibani P, Gupta S, Bazley FA, Pashai N, Chou BK, Shah S, Resar LM, Cheng L, Gearhart JD, Kerr CL. Early intervention for spinal cord injury with human induced pluripotent stem cells oligodendrocyte progenitors. PLoS One 2015; 10:e0116933. [PMID: 25635918 PMCID: PMC4311989 DOI: 10.1371/journal.pone.0116933] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Accepted: 12/16/2014] [Indexed: 12/16/2022] Open
Abstract
Induced pluripotent stem (iPS) cells are at the forefront of research in regenerative medicine and are envisaged as a source for personalized tissue repair and cell replacement therapy. Here, we demonstrate for the first time that oligodendrocyte progenitors (OPs) can be derived from iPS cells generated using either an episomal, non-integrating plasmid approach or standard integrating retroviruses that survive and differentiate into mature oligodendrocytes after early transplantation into the injured spinal cord. The efficiency of OP differentiation in all 3 lines tested ranged from 40% to 60% of total cells, comparable to those derived from human embryonic stem cells. iPS cell lines derived using episomal vectors or retroviruses generated a similar number of early neural progenitors and glial progenitors while the episomal plasmid-derived iPS line generated more OPs expressing late markers O1 and RIP. Moreover, we discovered that iPS-derived OPs (iPS-OPs) engrafted 24 hours following a moderate contusive spinal cord injury (SCI) in rats survived for approximately two months and that more than 70% of the transplanted cells differentiated into mature oligodendrocytes that expressed myelin associated proteins. Transplanted OPs resulted in a significant increase in the number of myelinated axons in animals that received a transplantation 24 h after injury. In addition, nearly a 5-fold reduction in cavity size and reduced glial scarring was seen in iPS-treated groups compared to the control group, which was injected with heat-killed iPS-OPs. Although further investigation is needed to understand the mechanisms involved, these results provide evidence that patient-specific, iPS-derived OPs can survive for three months and improve behavioral assessment (BBB) after acute transplantation into SCI. This is significant as determining the time in which stem cells are injected after SCI may influence their survival and differentiation capacity.
Collapse
Affiliation(s)
- Angelo H. All
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Singapore Institute for Neurotechnology, National University of Singapore, Singapore, Singapore
- Departments of Orthopedic Surgery, Biomedical Engineering and Medicine, Division of Neurology, National University of Singapore, Singapore, Singapore
| | - Payam Gharibani
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Siddharth Gupta
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Faith A. Bazley
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Singapore Institute for Neurotechnology, National University of Singapore, Singapore, Singapore
| | - Nikta Pashai
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Bin-Kuan Chou
- Graduate Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Sandeep Shah
- Division of Hematology in Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Linda M. Resar
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Division of Hematology in Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Linzhao Cheng
- Graduate Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Division of Hematology in Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - John D. Gearhart
- Department of Cell and Developmental Biology in the School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Department of Animal Biology in the School of Veterinary Medicine; University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Candace L. Kerr
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Obstetrics and Gynecology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Biochemistry and Molecular Biology, Unversity of Maryland School of Medicine, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
28
|
Perineural growth in head and neck squamous cell carcinoma: a review. Oral Oncol 2014; 51:16-23. [PMID: 25456006 DOI: 10.1016/j.oraloncology.2014.10.004] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Revised: 09/30/2014] [Accepted: 10/06/2014] [Indexed: 02/07/2023]
Abstract
Perineural growth is a unique route of tumor metastasis that is associated with poor prognosis in several solid malignancies. It is diagnosed by the presence of tumor cells inside the neural space seen on histological or imaging evaluations. Little is known about molecular mechanisms involved in the growth and spread of tumor cells in neural spaces. The poor prognosis associated with perineural growth and lack of targeted approaches necessitates the study of molecular factors involved in communication between tumor and neural cells. Perineural growth rates, shown to be as high as 63% in head and neck squamous cell carcinoma (HNSCC), correlate with increased local recurrence and decreased disease-free survival. Here we describe the literature on perineural growth in HNSCC. In addition, we discuss factors implicated in perineural growth of cancer. These factors include brain-derived neurotrophic factor (BDNF), nerve growth factor (NGF), neurotrophin-3 and -4, glial cell-line derived neurotrophic factor (GDNF), the neural cell adhesion molecule (NCAM), substance P (SP), and chemokines. We also explore the literature on membrane receptors, including the Trk family and the low-affinity nerve growth factor receptor. This review highlights areas for further study of the mechanisms of perineural invasion which may facilitate the identification of therapeutic targets in HNSCC.
Collapse
|
29
|
Narayanan M, Huynh JL, Wang K, Yang X, Yoo S, McElwee J, Zhang B, Zhang C, Lamb JR, Xie T, Suver C, Molony C, Melquist S, Johnson AD, Fan G, Stone DJ, Schadt EE, Casaccia P, Emilsson V, Zhu J. Common dysregulation network in the human prefrontal cortex underlies two neurodegenerative diseases. Mol Syst Biol 2014; 10:743. [PMID: 25080494 PMCID: PMC4299500 DOI: 10.15252/msb.20145304] [Citation(s) in RCA: 123] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Using expression profiles from postmortem prefrontal cortex samples of 624 dementia patients and non-demented controls, we investigated global disruptions in the co-regulation of genes in two neurodegenerative diseases, late-onset Alzheimer's disease (AD) and Huntington's disease (HD). We identified networks of differentially co-expressed (DC) gene pairs that either gained or lost correlation in disease cases relative to the control group, with the former dominant for both AD and HD and both patterns replicating in independent human cohorts of AD and aging. When aligning networks of DC patterns and physical interactions, we identified a 242-gene subnetwork enriched for independent AD/HD signatures. This subnetwork revealed a surprising dichotomy of gained/lost correlations among two inter-connected processes, chromatin organization and neural differentiation, and included DNA methyltransferases, DNMT1 and DNMT3A, of which we predicted the former but not latter as a key regulator. To validate the inter-connection of these two processes and our key regulator prediction, we generated two brain-specific knockout (KO) mice and show that Dnmt1 KO signature significantly overlaps with the subnetwork (P = 3.1 × 10−12), while Dnmt3a KO signature does not (P = 0.017).
Collapse
Affiliation(s)
| | - Jimmy L Huynh
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kai Wang
- Merck Research Laboratories Merck & Co., Inc., Whitehouse Station, NJ, USA
| | - Xia Yang
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA, USA
| | - Seungyeul Yoo
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Joshua McElwee
- Merck Research Laboratories Merck & Co., Inc., Whitehouse Station, NJ, USA
| | - Bin Zhang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Chunsheng Zhang
- Merck Research Laboratories Merck & Co., Inc., Whitehouse Station, NJ, USA
| | - John R Lamb
- Merck Research Laboratories Merck & Co., Inc., Whitehouse Station, NJ, USA
| | - Tao Xie
- Merck Research Laboratories Merck & Co., Inc., Whitehouse Station, NJ, USA
| | | | - Cliona Molony
- Merck Research Laboratories Merck & Co., Inc., Whitehouse Station, NJ, USA
| | - Stacey Melquist
- Merck Research Laboratories Merck & Co., Inc., Whitehouse Station, NJ, USA
| | | | - Guoping Fan
- Department of Human Genetics, University of California, Los Angeles, CA, USA
| | - David J Stone
- Merck Research Laboratories Merck & Co., Inc., Whitehouse Station, NJ, USA
| | - Eric E Schadt
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Patrizia Casaccia
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Valur Emilsson
- Icelandic Heart Association, Kopavogur, Iceland Faculty of Pharmaceutical Sciences, University of Iceland, Reykjavik, Iceland
| | - Jun Zhu
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
30
|
Yin X, Kiryu-Seo S, Kidd GJ, Feltri ML, Wrabetz L, Trapp BD. Proteolipid protein cannot replace P0 protein as the major structural protein of peripheral nervous system myelin. Glia 2014; 63:66-77. [PMID: 25066805 DOI: 10.1002/glia.22733] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Accepted: 07/14/2014] [Indexed: 11/10/2022]
Abstract
The central nervous system (CNS) of terrestrial vertebrates underwent a prominent molecular change when proteolipid protein (PLP) replaced P0 protein as the most abundant protein of CNS myelin. However, PLP did not replace P0 in peripheral nervous system (PNS) myelin. To investigate the possible consequences of a PLP to P0 shift in PNS myelin, we engineered mice to express PLP instead of P0 in PNS myelin (PLP-PNS mice). PLP-PNS mice had severe neurological disabilities and died between 3 and 6 months of age. Schwann cells in sciatic nerves from PLP-PNS mice sorted axons into one-to-one relationships but failed to form myelin internodes. Mice with equal amounts of P0 and PLP had normal PNS myelination and lifespans similar to wild-type (WT) mice. When PLP was overexpressed with one copy of the P0 gene, sciatic nerves were hypomyelinated; mice displayed motor deficits, but had normal lifespans. These data support the hypothesis that while PLP can co-exist with P0 in PNS myelin, PLP cannot replace P0 as the major structural protein of PNS myelin.
Collapse
Affiliation(s)
- Xinghua Yin
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | | | | | | | | | | |
Collapse
|
31
|
Schnaar RL, Gerardy-Schahn R, Hildebrandt H. Sialic acids in the brain: gangliosides and polysialic acid in nervous system development, stability, disease, and regeneration. Physiol Rev 2014; 94:461-518. [PMID: 24692354 DOI: 10.1152/physrev.00033.2013] [Citation(s) in RCA: 510] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Every cell in nature carries a rich surface coat of glycans, its glycocalyx, which constitutes the cell's interface with its environment. In eukaryotes, the glycocalyx is composed of glycolipids, glycoproteins, and proteoglycans, the compositions of which vary among different tissues and cell types. Many of the linear and branched glycans on cell surface glycoproteins and glycolipids of vertebrates are terminated with sialic acids, nine-carbon sugars with a carboxylic acid, a glycerol side-chain, and an N-acyl group that, along with their display at the outmost end of cell surface glycans, provide for varied molecular interactions. Among their functions, sialic acids regulate cell-cell interactions, modulate the activities of their glycoprotein and glycolipid scaffolds as well as other cell surface molecules, and are receptors for pathogens and toxins. In the brain, two families of sialoglycans are of particular interest: gangliosides and polysialic acid. Gangliosides, sialylated glycosphingolipids, are the most abundant sialoglycans of nerve cells. Mouse genetic studies and human disorders of ganglioside metabolism implicate gangliosides in axon-myelin interactions, axon stability, axon regeneration, and the modulation of nerve cell excitability. Polysialic acid is a unique homopolymer that reaches >90 sialic acid residues attached to select glycoproteins, especially the neural cell adhesion molecule in the brain. Molecular, cellular, and genetic studies implicate polysialic acid in the control of cell-cell and cell-matrix interactions, intermolecular interactions at cell surfaces, and interactions with other molecules in the cellular environment. Polysialic acid is essential for appropriate brain development, and polymorphisms in the human genes responsible for polysialic acid biosynthesis are associated with psychiatric disorders including schizophrenia, autism, and bipolar disorder. Polysialic acid also appears to play a role in adult brain plasticity, including regeneration. Together, vertebrate brain sialoglycans are key regulatory components that contribute to proper development, maintenance, and health of the nervous system.
Collapse
|
32
|
Tanaka M, Izawa T, Yamate J, Franklin RJM, Kuramoto T, Serikawa T, Kuwamura M. The VF rat with abnormal myelinogenesis has a mutation in Dopey1. Glia 2014; 62:1530-42. [PMID: 24863653 DOI: 10.1002/glia.22698] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 04/30/2014] [Accepted: 05/07/2014] [Indexed: 11/09/2022]
Abstract
The vacuole formation (VF) rat is an autosomal recessive myelin mutant characterized by generalized tremor, hypomyelination, and periaxonal vacuole formation of the central nervous system (CNS). Here, we report the most likely causative gene for neurological disease in the VF rat and pursue its roles in the development and maintenance of the CNS myelin. We identified a nonsense mutation in the dopey family member 1 (Dopey1) located on rat chromosome 8. Expression level of Dopey1 mRNA was decreased and DOPEY1 protein was undetectable both in the white and gray matter of the spinal cords in the VF rats. Double immunohistochemistry demonstrated that DOPEY1 was mainly expressed in neurons and oligodendrocytes in the wild-type rats, whereas no positive cells were detected in the VF rats. We also demonstrated a marked reduction in myelin components both at mRNA and protein levels during myelinogenesis in the VF rats. In addition, proteolipid protein and myelin-associated glycoprotein accumulated in oligodendrocyte cell body, suggesting that Dopey1 is likely to be involved in the traffic of myelin components. Our results highlighted the importance of Dopey1 for the development and maintenance of the CNS myelin.
Collapse
Affiliation(s)
- Miyuu Tanaka
- Laboratory of Veterinary Pathology, Osaka Prefecture University, Izumisano, Osaka, 598-8531, Japan
| | | | | | | | | | | | | |
Collapse
|
33
|
Espinosa-Jeffrey A, Paez PM, Cheli VT, Spreuer V, Wanner I, de Vellis J. Impact of simulated microgravity on oligodendrocyte development: implications for central nervous system repair. PLoS One 2013; 8:e76963. [PMID: 24324574 PMCID: PMC3850904 DOI: 10.1371/journal.pone.0076963] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Accepted: 08/28/2013] [Indexed: 01/11/2023] Open
Abstract
We have recently established a culture system to study the impact of simulated microgravity on oligodendrocyte progenitor cells (OPCs) development. We subjected mouse and human OPCs to a short exposure of simulated microgravity produced by a 3D-Clinostat robot. Our results demonstrate that rodent and human OPCs display enhanced and sustained proliferation when exposed to simulated microgravity as assessed by several parameters, including a decrease in the cell cycle time. Additionally, OPC migration was examined in vitro using time-lapse imaging of cultured OPCs. Our results indicated that OPCs migrate to a greater extent after stimulated microgravity than in normal conditions, and this enhanced motility was associated with OPC morphological changes. The lack of normal gravity resulted in a significant increase in the migration speed of mouse and human OPCs and we found that the average leading process in migrating bipolar OPCs was significantly longer in microgravity treated cells than in controls, demonstrating that during OPC migration the lack of gravity promotes leading process extension, an essential step in the process of OPC migration. Finally, we tested the effect of simulated microgravity on OPC differentiation. Our data showed that the expression of mature oligodendrocyte markers was significantly delayed in microgravity treated OPCs. Under conditions where OPCs were allowed to progress in the lineage, simulated microgravity decreased the proportion of cells that expressed mature markers, such as CC1 and MBP, with a concomitant increased number of cells that retained immature oligodendrocyte markers such as Sox2 and NG2. Development of methodologies aimed at enhancing the number of OPCs and their ability to progress on the oligodendrocyte lineage is of great value for treatment of demyelinating disorders. To our knowledge, this is the first report on the gravitational modulation of oligodendrocyte intrinsic plasticity to increase their progenies.
Collapse
Affiliation(s)
- Araceli Espinosa-Jeffrey
- Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine at UCLA, Intellectual and Developmental Disabilities Research Center, Los Angeles, California, United States of America
- * E-mail:
| | - Pablo M. Paez
- Hunter James Kelly Research Institute, Department of Pharmacology and Toxicology, School of Medicine and Biomedical Sciences, SUNY at Buffalo, NYS Center of Excellence, Buffalo, New York, United States of America
| | - Veronica T. Cheli
- Hunter James Kelly Research Institute, Department of Pharmacology and Toxicology, School of Medicine and Biomedical Sciences, SUNY at Buffalo, NYS Center of Excellence, Buffalo, New York, United States of America
| | - Vilma Spreuer
- Hunter James Kelly Research Institute, Department of Pharmacology and Toxicology, School of Medicine and Biomedical Sciences, SUNY at Buffalo, NYS Center of Excellence, Buffalo, New York, United States of America
| | - Ina Wanner
- Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine at UCLA, Intellectual and Developmental Disabilities Research Center, Los Angeles, California, United States of America
| | - Jean de Vellis
- Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine at UCLA, Intellectual and Developmental Disabilities Research Center, Los Angeles, California, United States of America
| |
Collapse
|
34
|
Jones MV, Nguyen TT, Ewaleifoh O, Lebson L, Whartenby KA, Griffin JW, Calabresi PA. Accelerated axon loss in MOG35-55 experimental autoimmune encephalomyelitis (EAE) in myelin-associated glycoprotein-deficient (MAGKO) mice. J Neuroimmunol 2013; 262:53-61. [PMID: 23899666 DOI: 10.1016/j.jneuroim.2013.06.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Revised: 06/10/2013] [Accepted: 06/20/2013] [Indexed: 01/16/2023]
Abstract
Myelin-associated glycoprotein (MAG) expressed by oligodendrocytes promotes the stability of axons but also impedes neural repair by inhibiting axon extension through lesioned white matter. We previously reported exacerbated axon losses in MAGKO as compared to wild type mice, 30days into experimental autoimmune encephalitis (EAE). Here, we report the time course of axon losses in EAE and show this occurs as early as 7days post-immunization, confirming MAG is protective against immune-mediated axon transection events. MAGKO mice also exhibit increased microglial activation prior to EAE, which is not seen in B4galnt1KO mice that also have axon loss, suggesting that the microglial activation may be a consequence of the loss of MAG inhibitory influence, and not a simple result of axonal degeneration.
Collapse
MESH Headings
- Animals
- Axons/pathology
- Disease Models, Animal
- Encephalomyelitis, Autoimmune, Experimental/chemically induced
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Female
- Male
- Mice
- Mice, Congenic
- Mice, Inbred C57BL
- Mice, Knockout
- Microglia/metabolism
- Microglia/pathology
- Myelin-Associated Glycoprotein/deficiency
- Time Factors
Collapse
Affiliation(s)
- Melina V Jones
- Johns Hopkins University, Department of Neurology, Room 625, 600N. Wolfe Street, Baltimore 21287, MD, USA
| | | | | | | | | | | | | |
Collapse
|
35
|
|
36
|
Barker R, Wellington D, Esiri MM, Love S. Assessing white matter ischemic damage in dementia patients by measurement of myelin proteins. J Cereb Blood Flow Metab 2013; 33:1050-7. [PMID: 23532085 PMCID: PMC3705431 DOI: 10.1038/jcbfm.2013.46] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2012] [Revised: 02/11/2013] [Accepted: 03/04/2013] [Indexed: 11/09/2022]
Abstract
White matter ischemia is difficult to quantify histologically. Myelin-associated glycoprotein (MAG) is highly susceptible to ischemia, being expressed only adaxonally, far from the oligodendrocyte cell body. Myelin-basic protein (MBP) and proteolipid protein (PLP) are expressed throughout the myelin sheath. We compared MAG, MBP, and PLP levels in parietal white matter homogenates from 17 vascular dementia (VaD), 49 Alzheimer's disease (AD), and 33 control brains, after assessing the post-mortem stability of these proteins. Small vessel disease (SVD) and cerebral amyloid angiopathy (CAA) severity had been assessed in paraffin sections. The concentration of MAG remained stable post-mortem, declined with increasing SVD, and was significantly lower in VaD than controls. The concentration of MBP fell progressively post-mortem, limiting its diagnostic utility in this context. Proteolipid protein was stable post-mortem and increased significantly with SVD severity. The MAG/PLP ratio declined significantly with SVD and CAA severity. The MAG and PLP levels and MAG/PLP did not differ significantly between AD and control brains. We validated the utility of MAG and MAG/PLP measurements on analysis of 74 frontal white matter samples from an Oxford cohort in which SVD had previously been scored. MAG concentration and the MAG/PLP ratio are useful post-mortem measures of ante-mortem white matter ischemia.
Collapse
Affiliation(s)
- Rachel Barker
- Dementia Research Group, Institute of Clinical Neurosciences, School of Clinical Sciences, University of Bristol, Bristol, UK
| | | | | | | |
Collapse
|
37
|
|
38
|
Masaki T. Polarization and myelination in myelinating glia. ISRN NEUROLOGY 2012; 2012:769412. [PMID: 23326681 PMCID: PMC3544266 DOI: 10.5402/2012/769412] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2012] [Accepted: 11/13/2012] [Indexed: 01/13/2023]
Abstract
Myelinating glia, oligodendrocytes in central nervous system and Schwann cells in peripheral nervous system, form myelin sheath, a multilayered membrane system around axons enabling salutatory nerve impulse conduction and maintaining axonal integrity. Myelin sheath is a polarized structure localized in the axonal side and therefore is supposed to be formed based on the preceding polarization of myelinating glia. Thus, myelination process is closely associated with polarization of myelinating glia. However, cell polarization has been less extensively studied in myelinating glia than other cell types such as epithelial cells. The ultimate goal of this paper is to provide insights for the field of myelination research by applying the information obtained in polarity study in other cell types, especially epithelial cells, to cell polarization of myelinating glia. Thus, in this paper, the main aspects of cell polarization study in general are summarized. Then, they will be compared with polarization in oligodendrocytes. Finally, the achievements obtained in polarization study for epithelial cells, oligodendrocytes, and other types of cells will be translated into polarization/myelination process by Schwann cells. Then, based on this model, the perspectives in the study of Schwann cell polarization/myelination will be discussed.
Collapse
Affiliation(s)
- Toshihiro Masaki
- Department of Medical Science, Teikyo University of Science, 2-2-1 Senju-Sakuragi, Adachi-ku, Tokyo 120-0045, Japan
| |
Collapse
|
39
|
Abstract
Myelination organizes axons into distinct domains that allow nerve impulses to propagate in a saltatory manner. The edges of the myelin sheath are sealed at the paranodes by axon-glial junctions that have a crucial role in organizing the axonal cytoskeleton. Here we propose a model in which the myelinated axons depend on the axon-glial junctions to stabilize the cytoskeletal transition at the paranodes. Thus paranodal regions are likely to be particularly susceptible to damage induced by demyelinating diseases such as multiple sclerosis.
Collapse
|
40
|
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating disease that is considered by many people to have an autoimmune aetiology. In recent years, new data emerging from histopathology, imaging and other studies have expanded our understanding of the disease and may change the way in which it is treated. Conceptual shifts have included: first, an appreciation of the extent to which the neuron and its axon are affected in MS, and second, elucidation of how the neurobiology of axon-glial and, particularly, axon-myelin interaction may influence disease progression. In this article, we review advances in both areas, focusing on the molecular mechanisms underlying axonal loss in acute inflammation and in chronic demyelination, and discussing how the restoration of myelin sheaths via the regenerative process of remyelination might prevent axon degeneration. An understanding of these processes could lead to better strategies for the prevention and treatment of axonal loss, which will ultimately benefit patients with MS.
Collapse
|
41
|
Abstract
Perineural invasion (PNI) is a prominent characteristic of pancreatic cancer. PNI is a process whereby cancer cells invade the surrounding nerves, thus providing an alternative route for metastatic spread and pain generation. PNI is thought to be an indicator of aggressive tumour behaviour and has been shown to correlate with poor prognosis of patients with pancreatic cancer. Recent studies demonstrated that some signalling molecules and pathways that are involved in PNI are also involved in pain generation. Targeting these signalling pathways has shown some promise in alleviating pain and reducing PNI, which could potentially improve treatment outcomes for patients with pancreatic cancer.
Collapse
Affiliation(s)
- Aditi A Bapat
- Clinical Translational Research Division, Translational Genomics Research Institute, 13208 East Shea Boulevard, Scottsdale, Arizona 85259, USA
| | | | | | | |
Collapse
|
42
|
Von Bartheld CS, Altick AL. Multivesicular bodies in neurons: distribution, protein content, and trafficking functions. Prog Neurobiol 2011; 93:313-40. [PMID: 21216273 DOI: 10.1016/j.pneurobio.2011.01.003] [Citation(s) in RCA: 141] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2010] [Revised: 12/22/2010] [Accepted: 01/03/2011] [Indexed: 11/27/2022]
Abstract
Multivesicular bodies (MVBs) are intracellular endosomal organelles characterized by multiple internal vesicles that are enclosed within a single outer membrane. MVBs were initially regarded as purely prelysosomal structures along the degradative endosomal pathway of internalized proteins. MVBs are now known to be involved in numerous endocytic and trafficking functions, including protein sorting, recycling, transport, storage, and release. This review of neuronal MVBs summarizes their research history, morphology, distribution, accumulation of cargo and constitutive proteins, transport, and theories of functions of MVBs in neurons and glia. Due to their complex morphologies, neurons have expanded trafficking and signaling needs, beyond those of "geometrically simpler" cells, but it is not known whether neuronal MVBs perform additional transport and signaling functions. This review examines the concept of compartment-specific MVB functions in endosomal protein trafficking and signaling within synapses, axons, dendrites and cell bodies. We critically evaluate reports of the accumulation of neuronal MVBs based on evidence of stress-induced MVB formation. Furthermore, we discuss potential functions of neuronal and glial MVBs in development, in dystrophic neuritic syndromes, injury, disease, and aging. MVBs may play a role in Alzheimer's, Huntington's, and Niemann-Pick diseases, some types of frontotemporal dementia, prion and virus trafficking, as well as in adaptive responses of neurons to trauma and toxin or drug exposure. Functions of MVBs in neurons have been much neglected, and major gaps in knowledge currently exist. Developing truly MVB-specific markers would help to elucidate the roles of neuronal MVBs in intra- and intercellular signaling of normal and diseased neurons.
Collapse
Affiliation(s)
- Christopher S Von Bartheld
- Department of Physiology and Cell Biology, Mailstop 352, University of Nevada School of Medicine, Reno, NV 89557, USA.
| | | |
Collapse
|
43
|
Lopez PHH, Ahmad AS, Mehta NR, Toner M, Rowland EA, Zhang J, Doré S, Schnaar RL. Myelin-associated glycoprotein protects neurons from excitotoxicity. J Neurochem 2011; 116:900-8. [PMID: 21214567 DOI: 10.1111/j.1471-4159.2010.07069.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
In addition to supporting rapid nerve conduction, myelination nurtures and stabilizes axons and protects them from acute toxic insults. One myelin molecule that protects and sustains axons is myelin-associated glycoprotein (MAG). MAG is expressed on the innermost wrap of myelin, apposed to the axon surface, where it interacts with axonal receptors that reside in lateral membrane domains including gangliosides, the glycosylphosphatidylinositol-anchored Nogo receptors, and β1-integrin. We report here that MAG protection extends beyond the axon to the neurons from which those axons emanate, protecting them from excitotoxicity. Compared to wild type mice, Mag-null mice displayed markedly increased seizure activity in response to intraperitoneal injection of kainic acid, an excitotoxic glutamate receptor agonist. Mag-null mice also had larger lesion volumes in response to intrastriatal injection of the excitotoxin NMDA. Prior injection of a soluble form of MAG partially protected Mag-null mice from NMDA-induced lesions. Hippocampal neurons plated on proteins extracted from wild-type rat or mouse myelin were resistant to kainic acid-induced excitotoxicity, whereas neurons plated on proteins from Mag-null myelin were not. Protection was reversed by anti-MAG antibody and replicated by addition of soluble MAG. MAG-mediated protection from excitotoxicity was dependent on Nogo receptors and β1-integrin. We conclude that MAG engages membrane-domain resident neuronal receptors to protect neurons from excitotoxicity, and that soluble MAG mitigates excitotoxic damage in vivo.
Collapse
Affiliation(s)
- Pablo H H Lopez
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Pupil-involving third nerve palsy as a manifestation of anti-myelin-associated glycoprotein neuropathy. J Neuroophthalmol 2010; 31:29-33. [PMID: 21164358 DOI: 10.1097/wno.0b013e3181f2e27a] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
A 56-year-old man developed a pupil-involving left third nerve palsy. Imaging studies of the brain and intracranial vessels were normal. Neurological examination demonstrated a sensory polyneuropathy and mild distal weakness. Nerve conduction studies showed prolonged distal motor latencies. An enzyme-linked immunosorbent assay test detected high titers of anti-myelin-associated glycoprotein (MAG) antibodies. The patient improved with prednisone and rituximab treatment. Anti-MAG neuropathy should be considered when evaluating a patient with an undiagnosed cranial neuropathy, especially in the setting of a sensory neuropathy.
Collapse
|
45
|
Barrie JA, Montague P, Karim S, Kirkham D, Nave KA, Anderson TJ, Griffiths IR, McLaughlin M. Modulation of rumpshaker phenotype with wild-type PLP/DM20 suggests several pathogenic mechanisms. J Neurosci Res 2010; 88:2135-45. [PMID: 20175203 DOI: 10.1002/jnr.22379] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The rumpshaker mutation of the murine myelin proteolipid protein 1 (Plp1) gene generates misfolded PLP/DM20 protein, resulting in dysmyelination, increased oligodendrocyte apoptosis, and death prior to P40 when expressed on the C57 BL/6 background. In this study, we used transgenic complementation to normalize the levels of PLP/DM20 in myelin with wild-type protein to determine whether loss of normal PLP function or gain of toxic function is responsible for dysmyelination in the rumpshaker. Restoring myelin PLP/DM20 levels extended the survival time to at least P60, significantly reduced the density of apoptotic cells, increased myelin volume, and restored normal periodicity of myelin. Biochemical analysis found that several myelin proteins that are reduced in rumpshaker, including MAG, CNP, and SirT2, are markedly elevated at peak myelination (P20) in the rumpshaker transgenic mouse. Myelin basic protein, however, remained low at peak myelination but was restored at P60 when myelin had matured and entered into a maintenance phase. Markers of the unfolded protein response (UPR), BiP and XBP1, remained activated with the introduction of wild-type PLP. These data demonstrate that restoring wild-type PLP/DM20 levels in rumpshaker improves the phenotype and the integrity of myelin, but hypomyelination persists and stress pathways remain activated. This suggests that both gain- and loss-of-function mechanisms are involved in the pathogenesis of the rumpshaker.
Collapse
Affiliation(s)
- Jennifer A Barrie
- Applied Neurobiology Group, Institute of Comparative Medicine, University of Glasgow, Bearsden, Glasgow, Scotland
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Hsu C, Morohashi Y, Yoshimura SI, Manrique-Hoyos N, Jung S, Lauterbach MA, Bakhti M, Grønborg M, Möbius W, Rhee J, Barr FA, Simons M. Regulation of exosome secretion by Rab35 and its GTPase-activating proteins TBC1D10A-C. ACTA ACUST UNITED AC 2010; 189:223-32. [PMID: 20404108 PMCID: PMC2856897 DOI: 10.1083/jcb.200911018] [Citation(s) in RCA: 609] [Impact Index Per Article: 43.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
A screen in oligodendrocytes establishes a Rab family member and its GAPs as regulators of exosome secretion by controlling endocytic vesicle docking with the plasma membrane. Oligodendrocytes secrete vesicles into the extracellular space, where they might play a role in neuron–glia communication. These exosomes are small vesicles with a diameter of 50–100 nm that are formed within multivesicular bodies and are released after fusion with the plasma membrane. The intracellular pathways that generate exosomes are poorly defined. Because Rab family guanosine triphosphatases (GTPases) together with their regulators are important membrane trafficking organizers, we investigated which Rab GTPase-activating proteins interfere with exosome release. We find that TBC1D10A–C regulate exosome secretion in a catalytic activity–dependent manner. We show that Rab35 is the target of TBC1D10A–C and that the inhibition of Rab35 function leads to intracellular accumulation of endosomal vesicles and impairs exosome secretion. Rab35 localizes to the surface of oligodendroglia in a GTP-dependent manner, where it increases the density of vesicles, suggesting a function in docking or tethering. These findings provide a basis for understanding the biogenesis and function of exosomes in the central nervous system.
Collapse
Affiliation(s)
- Chieh Hsu
- Max Planck Institute for Experimental Medicine, 37075 Göttingen, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Schnaar RL, Lopez PHH. Myelin-associated glycoprotein and its axonal receptors. J Neurosci Res 2010; 87:3267-76. [PMID: 19156870 DOI: 10.1002/jnr.21992] [Citation(s) in RCA: 108] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Myelin-associated glycoprotein (MAG) is expressed on the innermost myelin membrane wrap, directly apposed to the axon surface. Although it is not required for myelination, MAG enhances long-term axon-myelin stability, helps to structure nodes of Ranvier, and regulates the axon cytoskeleton. In addition to its role in axon-myelin stabilization, MAG inhibits axon regeneration after injury; MAG and a discrete set of other molecules on residual myelin membranes at injury sites actively signal axons to halt elongation. Both the stabilizing and the axon outgrowth inhibitory effects of MAG are mediated by complementary MAG receptors on the axon surface. Two MAG receptor families have been described, sialoglycans (specifically gangliosides GD1a and GT1b) and Nogo receptors (NgRs). Controversies remain about which receptor(s) mediates which of MAG's biological effects. Here we review the findings and challenges in associating MAG's biological effects with specific receptors.
Collapse
Affiliation(s)
- Ronald L Schnaar
- Department of Pharmacology, The Johns Hopkins School of Medicine, 725 N. Wolfe Street, Baltimore, MD 21205, USA.
| | | |
Collapse
|
48
|
Bauer NG, Richter-Landsberg C, Ffrench-Constant C. Role of the oligodendroglial cytoskeleton in differentiation and myelination. Glia 2010; 57:1691-705. [PMID: 19455583 DOI: 10.1002/glia.20885] [Citation(s) in RCA: 141] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Oligodendrocytes, the myelin-forming cells of the central nervous system, are in culture characterized by an elaborate process network, terminating in flat membranous sheets that are rich in myelin-specific proteins and lipids, and spirally wrap axons forming a compact insulating layer in vivo. By analogy with other cell types, maintenance and stability of these processes, as well as the formation of the myelin sheath, likely rely on a pronounced cytoskeleton consisting of microtubules and microfilaments. While the specialized process of wrapping and compaction forming the myelin sheath is not well understood, considerably more is known about how cytoskeletal organization is mediated by extracellular and intracellular signals and other interaction partners during oligodendrocyte differentiation and myelination. Here, we review the current state of knowledge on the role of the oligodendrocyte cytoskeleton in differentiation with an emphasis on signal transduction mechanisms and will attempt to draw out implications for its significance in myelination.
Collapse
Affiliation(s)
- Nina G Bauer
- MRC Centre for Regenerative Medicine, Centre for Multiple Sclerosis Research, The University of Edinburgh, Queen's Medical Research Institute, Edinburgh EH16 4TJ, United Kingdom.
| | | | | |
Collapse
|
49
|
Jackman N, Ishii A, Bansal R. Oligodendrocyte development and myelin biogenesis: parsing out the roles of glycosphingolipids. Physiology (Bethesda) 2009; 24:290-7. [PMID: 19815855 DOI: 10.1152/physiol.00016.2009] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The myelin sheath is an extension of the oligoddendrocyte (OL) plasma membrane enriched in lipids that ensheaths the axons of the central and peripheral nervous system. Here, we review the involvement of glycosphingolipids in myelin/OL functions, including the regulation of OL differentiation, lipid raft-mediated trafficking and signaling, and neuron-glia interactions.
Collapse
Affiliation(s)
- Nicole Jackman
- Department of Neuroscience, University of Connecticut Medical School, Farmington, Connecticut, USA
| | | | | |
Collapse
|
50
|
Sanchez ES, Bigbee JW, Fobbs W, Robinson SE, Sato-Bigbee C. Opioid addiction and pregnancy: perinatal exposure to buprenorphine affects myelination in the developing brain. Glia 2008; 56:1017-27. [PMID: 18381654 DOI: 10.1002/glia.20675] [Citation(s) in RCA: 103] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Buprenorphine is a mu-opioid receptor partial agonist and kappa-opioid receptor antagonist currently on trials for the management of pregnant opioid-dependent addicts. However, little is known about the effects of buprenorphine on brain development. Oligodendrocytes express opioid receptors in a developmentally regulated manner and thus, it is logical to hypothesize that perinatal exposure to buprenorphine could affect myelination. To investigate this possibility, pregnant rats were implanted with minipumps to deliver buprenorphine at 0.3 or 1 mg/kg/day. Analysis of their pups at different postnatal ages indicated that exposure to 0.3 mg/kg/day buprenorphine caused an accelerated and significant increase in the brain expression of all myelin basic protein (MBP) splicing isoforms. In contrast, treatment with the higher dose caused a developmental delay in MBP expression. Examination of corpus callosum at 26-days of age indicated that both buprenorphine doses cause a significant increase in the caliber of the myelinated axons. Surprisingly, these axons have a disproportionately thinner myelin sheath, suggesting alterations at the level of axon-glial interactions. Analysis of myelin associated glycoprotein (MAG) expression and glycosylation indicated that this molecule may play a crucial role in mediating these effects. Co-immunoprecipitation studies also suggested a mechanism involving a MAG-dependent activation of the Src-family tyrosine kinase Fyn. These results support the idea that opioid signaling plays an important role in regulating myelination in vivo and stress the need for further studies investigating potential effects of perinatal buprenorphine exposure on brain development.
Collapse
Affiliation(s)
- Emilse S Sanchez
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, Virginia 23298-0614, USA
| | | | | | | | | |
Collapse
|