1
|
Dickstein DL, Zhang R, Ru N, Vozenin MC, Perry BC, Wang J, Baulch JE, Acharya MM, Limoli CL. Structural plasticity of pyramidal cell neurons measured after FLASH and conventional dose-rate irradiation. Brain Struct Funct 2025; 230:41. [PMID: 40024988 PMCID: PMC11872753 DOI: 10.1007/s00429-025-02902-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 02/17/2025] [Indexed: 03/04/2025]
Abstract
Evidence shows that ultra-high dose-rate FLASH-radiotherapy (FLASH-RT) provides relative protection against normal tissue complications and functional decrements in the irradiated brain. Past work has shown that radiation-induced cognitive impairment, neuroinflammation and reduced structural complexity ofgranule cell neurons were not observed to the same extent after FLASH-RT (> MGy/s) compared to conventional dose-rate (CONV, 0.1 Gy/s) delivery. In this study, we explored the sensitivity of hippocampal CA1 and medial prefrontal cortex (mPFC) pyramidal neurons to cranial irradiation and dose-rate modulation using electron and confocal microscopy. Neuron ultrastructural analyses by electron microscopy after 10 Gy FLASH- or CONV-RT exposures indicated that irradiation had little impact on dendritic complexity and synapse density in the CA1, but did increase the length and head diameter of smaller non-perforated synapses. Similarly, irradiation caused no change in mPFC prelimbic/infralimbic axospinous synapse density, but reductions in non-perforated synapse diameters. While irradiation resulted in thinner myelin sheaths compared to controls, none of these metrics were dose-rate sensitive. Analysis of fluorescently labeled CA1 neurons revealed no radiation-induced or dose-rate-dependent changes in overall dendritic complexity or spine density, in contrast to our past analysis of granule cell neurons. Super-resolution confocal microscopy following a clinical dosing paradigm (3 × 10 Gy) showed significant reductions in excitatory vesicular glutamate transporter 1 and inhibitory vesicular GABA transporter puncta density within the CA1 that were largely dose-rate independent. Collectively, these data reveal that, compared to granule cell neurons, CA1 and mPFC neurons are relatively more radioresistant irrespective of radiation dose-rate.
Collapse
Affiliation(s)
- Dara L Dickstein
- Department of Pathology, Uniformed Services University of Health Sciences, Bethesda, MD, 20814, USA.
- The Henry M. Jackson Foundation for the Advancement of Military Health Inc, Bethesda, MD, USA.
| | - Richard Zhang
- Department of Radiation Oncology, University of California, Irvine School of Medicine, Irvine, CA, USA
| | - Ning Ru
- Department of Radiation Oncology, University of California, Irvine School of Medicine, Irvine, CA, USA
| | - Marie-Catherine Vozenin
- Secteur Radio-Oncologie et Radiobiologie, Hôpitaux Universitaires de Genève, Geneva, Switzerland
| | - Bayley C Perry
- Department of Pathology, Uniformed Services University of Health Sciences, Bethesda, MD, 20814, USA
- The Henry M. Jackson Foundation for the Advancement of Military Health Inc, Bethesda, MD, USA
| | - Juan Wang
- Department of Pathology, Uniformed Services University of Health Sciences, Bethesda, MD, 20814, USA
- The Henry M. Jackson Foundation for the Advancement of Military Health Inc, Bethesda, MD, USA
| | - Janet E Baulch
- Department of Radiation Oncology, University of California, Irvine School of Medicine, Irvine, CA, USA
| | - Munjal M Acharya
- Department of Radiation Oncology, University of California, Irvine School of Medicine, Irvine, CA, USA
- Department of Anatomy and Neurobiology, University of California, Irvine School of Medicine, Irvine, CA, USA
| | - Charles L Limoli
- Department of Radiation Oncology, University of California, Irvine School of Medicine, Irvine, CA, USA.
| |
Collapse
|
2
|
Dai R, Sun Y. Altered GnRH neuron-glia networks close to interface of polycystic ovary syndrome: Molecular mechanism and clinical perspectives. Life Sci 2025; 361:123318. [PMID: 39719166 DOI: 10.1016/j.lfs.2024.123318] [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: 08/27/2024] [Revised: 11/24/2024] [Accepted: 12/16/2024] [Indexed: 12/26/2024]
Abstract
Polycystic ovary syndrome (PCOS) has been noticed as a neuroendocrine syndrome manifested by reproductive hormone dysregulation involving increased luteinizing hormone (LH) pulse frequency and an increased LH to follicle-stimulating hormone ratio, yet theory is just beginning to be established. Neuroglia located in the arcuate nucleus and median eminence (ARC-ME) that are close to gonadotropin-releasing hormone (GnRH) axon terminals, comprise the blood-brain barrier and fenestrated vessels implying their putative roles in the modulation of the abnormal GnRH pulse in PCOS. This review outlines the disturbances of neuron-glia networks that underlie hypothetically the deregulation of GnRH-LH release and impaired sex hormone negative feedback in PCOS. We then discuss chronic and low-grade inflammatory status together with gut dysbiosis and how the detriments may intrude the hypothalamus by virtue of violating interfaces between the brain and periphery, which might contribute to the etiology of the impaired neural circuits in the ARC-ME to induce PCOS.
Collapse
Affiliation(s)
- Ruoxi Dai
- Hospital & Institute of Obstetrics and Gynecology, Fudan University, Shanghai 200081, China
| | - Yan Sun
- Hospital & Institute of Obstetrics and Gynecology, Fudan University, Shanghai 200081, China; The Academy of Integrative Medicine, Fudan University, Shanghai 200081, China; Shanghai Key Laboratory of Female Reproductive Endocrine-related Disease, Shanghai 200081, China.
| |
Collapse
|
3
|
Schuurmans IME, Mordelt A, de Witte LD. Orchestrating the neuroglial compartment: Ontogeny and developmental interaction of astrocytes, oligodendrocytes, and microglia. HANDBOOK OF CLINICAL NEUROLOGY 2025; 209:27-47. [PMID: 40122629 DOI: 10.1016/b978-0-443-19104-6.00011-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
Neuroglial cells serve as the master regulators of the central nervous system, making it imperative for glial development to be tightly regulated both spatially and temporally to ensure optimal brain function. In this chapter, we will discuss the origin and development of the three major glia cells such as astrocytes, oligodendrocytes, and microglia in the central nervous system. While much of our understanding of neuroglia development stems from studies using animal models, we will also explore recent insights into human glial development and potential differences from rodent models. Finally, the extensive crosstalk between glia cells will be highlighted, discussing how interactions among astrocyte, oligodendrocyte, and microglial influence their respective developmental pathways.
Collapse
Affiliation(s)
- Imke M E Schuurmans
- Department of Pediatrics, Radboud University Medical Center, Amalia Children's Hospital, Nijmegen, The Netherlands; Emma Center for Personalized Medicine, Departments of Pediatrics and Human Genetics, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Annika Mordelt
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands; Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
| | - Lot D de Witte
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands; Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands; Department of Psychiatry, Radboud University Medical Center, Nijmegen, The Netherlands.
| |
Collapse
|
4
|
Nocera S, Marchena MA, Fernández-Gómez B, Gómez-Martín P, Sánchez-Jiménez E, Macías-Castellano A, Laó Y, Cordano C, Gómez-Torres Ó, Luján R, de Castro F. Activation of Shh/Smo is sufficient to maintain oligodendrocyte precursor cells in an undifferentiated state and is not necessary for myelin formation and (re)myelination. Glia 2024; 72:1469-1483. [PMID: 38771121 DOI: 10.1002/glia.24540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 05/22/2024]
Abstract
Myelination is the terminal step in a complex and precisely timed program that orchestrates the proliferation, migration and differentiation of oligodendroglial cells. It is thought that Sonic Hedgehog (Shh) acting on Smoothened (Smo) participates in regulating this process, but that these effects are highly context dependent. Here, we investigate oligodendroglial development and remyelination from three specific transgenic lines: NG2-CreERT2 (control), Smofl/fl/NG2-CreERT2 (loss of function), and SmoM2/NG2-CreERT2 (gain of function), as well as pharmacological manipulation that enhance or inhibit the Smo pathway (Smoothened Agonist (SAG) or cyclopamine treatment, respectively). To explore the effects of Shh/Smo on differentiation and myelination in vivo, we developed a highly quantifiable model by transplanting oligodendrocyte precursor cells (OPCs) in the retina. We find that myelination is greatly enhanced upon cyclopamine treatment and hypothesize that Shh/Smo could promote OPC proliferation to subsequently inhibit differentiation. Consistent with this hypothesis, we find that the genetic activation of Smo significantly increased numbers of OPCs and decreased oligodendrocyte differentiation when we examined the corpus callosum during development and after cuprizone demyelination and remyelination. However, upon loss of function with the conditional ablation of Smo, myelination in the same scenarios are unchanged. Taken together, our present findings suggest that the Shh pathway is sufficient to maintain OPCs in an undifferentiated state, but is not necessary for myelination and remyelination.
Collapse
Affiliation(s)
- Sonia Nocera
- Grupo de Neurobiología del Desarrollo-GNDe, Instituto Cajal-CSIC, Madrid, Spain
| | - Miguel A Marchena
- Grupo de Neurobiología del Desarrollo-GNDe, Instituto Cajal-CSIC, Madrid, Spain
- Facultad HM de Ciencias de la Salud de la UCJC, Universidad Camilo José Cela, Madrid, Spain
- NeuroLab, Instituto de Investigación Sanitaria HM Hospitales, Madrid, Spain
| | | | - Paula Gómez-Martín
- Grupo de Neurobiología del Desarrollo-GNDe, Instituto Cajal-CSIC, Madrid, Spain
| | | | | | - Yolanda Laó
- Grupo de Neurobiología del Desarrollo-GNDe, Instituto Cajal-CSIC, Madrid, Spain
| | - Christian Cordano
- Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, California, USA
- Department of Neurology, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health. University of Genoa, Italy
- Department of Neuroscience, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Óscar Gómez-Torres
- Facultad de Ciencias Ambientales, Universidad de Castilla-La Mancha, Toledo, Spain
| | - Rafael Luján
- Facultad de Medicina, Universidad de Castilla-La Mancha, Albacete, Spain
| | - Fernando de Castro
- Grupo de Neurobiología del Desarrollo-GNDe, Instituto Cajal-CSIC, Madrid, Spain
| |
Collapse
|
5
|
Dickstein DL, Zhang R, Ru N, Vozenin MC, Perry BC, Wang J, Baulch J, Acharya MM, Limoli CL. Structural plasticity of pyramidal cell neurons measured after FLASH and conventional dose-rate irradiation. RESEARCH SQUARE 2024:rs.3.rs-4656938. [PMID: 39108471 PMCID: PMC11302692 DOI: 10.21203/rs.3.rs-4656938/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
Evidence shows that ultra-high dose-rate FLASH-radiotherapy (FLASH-RT) protects against normal tissue complications and functional decrements in the irradiated brain. Past work has shown that radiation-induced cognitive impairment, neuroinflammation and reduced structural complexity of granule cell neurons were not observed to the same extent after FLASH-RT (> MGy/s) compared to conventional dose-rate (CONV, 0.1 Gy/s) delivery. To explore the sensitivity of different neuronal populations to cranial irradiation and dose-rate modulation, hippocampal CA1 and medial prefrontal cortex (PFC) pyramidal neurons were analyzed by electron and confocal microscopy. Neuron ultrastructural analyses by electron microscopy after 10 Gy FLASH- or CONV-RT exposures indicated that irradiation had little impact on dendritic complexity and synapse density in the CA1, but did increase length and head diameter of smaller non-perforated synapses. Similarly, irradiation caused no change in PFC prelimbic/infralimbic axospinous synapse density, but reductions in non-perforated synapse diameters. While irradiation resulted in thinner myelin sheaths compared to controls, none of these metrics were dose-rate sensitive. Analysis of fluorescently labeled CA1 neurons revealed no radiation-induced or dose-rate-dependent changes in overall dendritic complexity or spine density, in contrast to our past analysis of granule cell neurons. Super-resolution confocal microscopy following a clinical dosing paradigm (3×10Gy) showed significant reductions in excitatory vesicular glutamate transporter 1 and inhibitory vesicular GABA transporter puncta density within the CA1 that were largely dose-rate independent. Collectively, these data reveal that, compared to granule cell neurons, CA1 and mPFC neurons are more radioresistant irrespective of radiation dose-rate.
Collapse
Affiliation(s)
| | | | - Ning Ru
- University of California, Irvine School of Medicine
| | | | | | - Juan Wang
- Uniformed Services University of Health Sciences
| | - Janet Baulch
- University of California, Irvine School of Medicine
| | | | | |
Collapse
|
6
|
Chen M, Guo P, Ru X, Chen Y, Zuo S, Feng H. Myelin sheath injury and repairment after subarachnoid hemorrhage. Front Pharmacol 2023; 14:1145605. [PMID: 37077816 PMCID: PMC10106687 DOI: 10.3389/fphar.2023.1145605] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 03/24/2023] [Indexed: 04/05/2023] Open
Abstract
Subarachnoid hemorrhage (SAH) can lead to damage to the myelin sheath in white matter. Through classification and analysis of relevant research results, the discussion in this paper provides a deeper understanding of the spatiotemporal change characteristics, pathophysiological mechanisms and treatment strategies of myelin sheath injury after SAH. The research progress for this condition was also systematically reviewed and compared related to myelin sheath in other fields. Serious deficiencies were identified in the research on myelin sheath injury and treatment after SAH. It is necessary to focus on the overall situation and actively explore different treatment methods based on the spatiotemporal changes in the characteristics of the myelin sheath, as well as the initiation, intersection and common action point of the pathophysiological mechanism, to finally achieve accurate treatment. We hope that this article can help researchers in this field to further clarify the challenges and opportunities in the current research on myelin sheath injury and treatment after SAH.
Collapse
Affiliation(s)
- Mao Chen
- Department of Neurology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Peiwen Guo
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xufang Ru
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yujie Chen
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- *Correspondence: Yujie Chen, ; Shilun Zuo,
| | - Shilun Zuo
- Department of Neurology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- *Correspondence: Yujie Chen, ; Shilun Zuo,
| | - Hua Feng
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
7
|
Villadiego J, García-Swinburn R, García-González D, Lebrón-Galán R, Murcia-Belmonte V, García-Roldán E, Suárez-Luna N, Nombela C, Marchena M, de Castro F, Toledo-Aral JJ. Extracellular matrix protein anosmin-1 overexpression alters dopaminergic phenotype in the CNS and the PNS with no pathogenic consequences in a MPTP model of Parkinson's disease. Brain Struct Funct 2023; 228:907-920. [PMID: 36995433 PMCID: PMC10147818 DOI: 10.1007/s00429-023-02631-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 03/15/2023] [Indexed: 03/31/2023]
Abstract
The development and survival of dopaminergic neurons are influenced by the fibroblast growth factor (FGF) pathway. Anosmin-1 (A1) is an extracellular matrix protein that acts as a major regulator of this signaling pathway, controlling FGF diffusion, and receptor interaction and shuttling. In particular, previous work showed that A1 overexpression results in more dopaminergic neurons in the olfactory bulb. Prompted by those intriguing results, in this study, we investigated the effects of A1 overexpression on different populations of catecholaminergic neurons in the central (CNS) and the peripheral nervous systems (PNS). We found that A1 overexpression increases the number of dopaminergic substantia nigra pars compacta (SNpc) neurons and alters the striosome/matrix organization of the striatum. Interestingly, these numerical and morphological changes in the nigrostriatal pathway of A1-mice did not confer an altered susceptibility to experimental MPTP-parkinsonism with respect to wild-type controls. Moreover, the study of the effects of A1 overexpression was extended to different dopaminergic tissues associated with the PNS, detecting a significant reduction in the number of dopaminergic chemosensitive carotid body glomus cells in A1-mice. Overall, our work shows that A1 regulates the development and survival of dopaminergic neurons in different nuclei of the mammalian nervous system.
Collapse
Affiliation(s)
- Javier Villadiego
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío-CSIC-Universidad de Sevilla, Avda. Manuel Siurot s/n, 41013, Seville, Spain
- Departamento de Fisiología Médica y Biofísica, Universidad de Sevilla, 41009, Sevilla, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Roberto García-Swinburn
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío-CSIC-Universidad de Sevilla, Avda. Manuel Siurot s/n, 41013, Seville, Spain
- Departamento de Fisiología Médica y Biofísica, Universidad de Sevilla, 41009, Sevilla, Spain
| | - Diego García-González
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío-CSIC-Universidad de Sevilla, Avda. Manuel Siurot s/n, 41013, Seville, Spain
- Grupo de Neurobiología del Desarrollo-GNDe, Hospital Nacional de Parapléjicos, 45071, Toledo, Spain
| | - Rafael Lebrón-Galán
- Grupo de Neurobiología del Desarrollo-GNDe, Hospital Nacional de Parapléjicos, 45071, Toledo, Spain
| | - Verónica Murcia-Belmonte
- Grupo de Neurobiología del Desarrollo-GNDe, Hospital Nacional de Parapléjicos, 45071, Toledo, Spain
- Instituto de Neurociencias, UMH-CSIC, Sant Joan d´Alacant, 03550, Alicante, Spain
| | - Ernesto García-Roldán
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío-CSIC-Universidad de Sevilla, Avda. Manuel Siurot s/n, 41013, Seville, Spain
- Departamento de Fisiología Médica y Biofísica, Universidad de Sevilla, 41009, Sevilla, Spain
- Servicio de Neurología y Neurofisiología Clínica, Hospital Universitario Virgen del Rocío, 41013, Sevilla, Spain
| | - Nela Suárez-Luna
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío-CSIC-Universidad de Sevilla, Avda. Manuel Siurot s/n, 41013, Seville, Spain
- Departamento de Fisiología Médica y Biofísica, Universidad de Sevilla, 41009, Sevilla, Spain
| | - Cristina Nombela
- Departamento de Psicología Biológica y de la Salud, Universidad Autónoma de Madrid, 28049, Madrid, Spain
| | - Miguel Marchena
- Grupo de Neurobiología del Desarrollo-GNDe, Instituto Cajal-CSIC, Avenida Doctor Arce 37, 28002, Madrid, Spain
- Departamento de Medicina, Universidad Europea de Madrid-UEM, Villaviciosa de Odón, 28670, Madrid, Spain
| | - Fernando de Castro
- Grupo de Neurobiología del Desarrollo-GNDe, Hospital Nacional de Parapléjicos, 45071, Toledo, Spain.
- Grupo de Neurobiología del Desarrollo-GNDe, Instituto Cajal-CSIC, Avenida Doctor Arce 37, 28002, Madrid, Spain.
| | - Juan José Toledo-Aral
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío-CSIC-Universidad de Sevilla, Avda. Manuel Siurot s/n, 41013, Seville, Spain.
- Departamento de Fisiología Médica y Biofísica, Universidad de Sevilla, 41009, Sevilla, Spain.
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
| |
Collapse
|
8
|
Zhang X, Han J, Fan D, Wang J, Lin X, Zhang H, Zhang C, Bai J, Huang H, Gu Y. Lysine-40 succinylation of TAGLN2 induces glioma angiogenesis and tumor growth through regulating TMSB4X. Cancer Gene Ther 2023; 30:172-181. [PMID: 36131066 DOI: 10.1038/s41417-022-00534-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 08/12/2022] [Accepted: 09/06/2022] [Indexed: 01/19/2023]
Abstract
Protein lysine succinylation (Ksucc) represents an important regulatory mechanism of tumor development. In this work, the difference of protein Ksucc between HCMEC/D3 co-cultured with U87 (glioma endothelia cells, GEC) and without U87 (normal endothelia cells, NEC) was investigated using TMT labeling and affinity enrichment followed by high-resolution LC-MS/MS analysis. Interestingly, TAGLN2 was highly succinylated at K40 in GEC (15.36 folds vs. NEC). Compared to the Vector group, TAGLN2WT and a succinylation-mimetic TAGLN2K40E greatly promoted the angiogenesis of glioma in vitro and in vivo. Furthermore, the adhesion and metastasis of U87 co-cultured with GEC in the TAGLN2WT or TAGLN2K40E group were also significantly promoted. This was consistent with the increased expression of VE-cadherin and actin cytoskeleton remodeling induced by TAGLN2 K40succ in GEC. In addition, high K40succ of TAGLN2 was associated with poor prognosis in patients with glioma. Overexpression of TAGLN2K40E also markedly promoted the proliferation and migration of glioma cells, further analysis of in vivo xenograft tumors showed that there was a significant decrease in tumor size and angiogenesis in the TAGLN2K40R group. Notably, the co-localization of TMSB4X and TAGLN2 mainly in the nucleus and cytoplasm of glioma cells was detected by immunofluorescence staining. We identified TMSB4X as a potential target of TAGLN2, which was proved to interact with TAGLN2WT rather than TAGLN2K40A. And the inhibition of TMSB4X could markedly attenuate the proliferation and migration of glioma cells induced by TAGLN2 K40succ. The results revealed K40succ of TAGLN2 could be a novelty diagnosis and therapeutic target for gliomas.
Collapse
Affiliation(s)
- Xiaoyi Zhang
- Department of Pharmacology, Life Science and Biopharmaceutical Institution, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning Province, P. R. China
| | - Jin Han
- Department of Pharmacology, Life Science and Biopharmaceutical Institution, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning Province, P. R. China
| | - Di Fan
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, P. R. China
| | - Jiahong Wang
- Department of Pharmacology, Life Science and Biopharmaceutical Institution, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning Province, P. R. China
| | - Xiangdan Lin
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, P. R. China
| | - Hong Zhang
- Department of Pharmacology, Life Science and Biopharmaceutical Institution, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning Province, P. R. China
| | - Cai Zhang
- Department of Pharmacology, Life Science and Biopharmaceutical Institution, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning Province, P. R. China
| | - Jialing Bai
- Department of Pharmacology, Life Science and Biopharmaceutical Institution, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning Province, P. R. China
| | - Hailan Huang
- Department of Pharmacology, Life Science and Biopharmaceutical Institution, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning Province, P. R. China
| | - Yanting Gu
- Department of Pharmacology, Life Science and Biopharmaceutical Institution, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning Province, P. R. China.
| |
Collapse
|
9
|
Anosmin 1 N-terminal domains modulate prokineticin receptor 2 activation by prokineticin 2. Cell Signal 2022; 98:110417. [PMID: 35878754 DOI: 10.1016/j.cellsig.2022.110417] [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: 05/20/2022] [Revised: 07/12/2022] [Accepted: 07/19/2022] [Indexed: 11/23/2022]
Abstract
The X-linked form of Kallmann syndrome (KS), characterized by hypogonadotropic hypogonadism and anosmia, is due to mutations in the ANOS1 gene that encodes for the extracellular matrix (ECM) protein anosmin 1. Prokineticins (PKs) exert their biological functions through the activation of the G protein-coupled receptors (GPCRs) prokineticin receptor 1 and 2 (PKR1, 2), and mutations in the PK2 and PKR2 genes are involved in the pathogenesis of KS. We have previously shown interaction between PKR2 and anosmin 1 in vitro. In the current report we present evidence of the modulation of PK2/PKR2 activity by anosmin 1, since this protein is able to enhance the activation of the ERK1/2 (extracellular signal-regulated kinase 1/2) pathway elicited by PK2 through PKR2. We also show that the N-terminal region of anosmin 1, capable of binding to the PK2-binding domain of PKR2, seems to be responsible for this effect. The whey acidic protein domain (WAP) is necessary for this modulatory activity, although data from GST pull-down (glutathione-S-transferase) and analysis of the N267K mutation in the fibronectin type III domain 1 (FnIII.1) suggest the cysteine-rich (CR) and the FnIII.1 domains could assist the WAP domain both in the binding to PKR2 and in the modulation of the activation of the receptor by PK2. Our data support the idea of a modulatory role of anosmin 1 in the biological effects controlled by the PK2/PKR2 system.
Collapse
|
10
|
Ornitz DM, Itoh N. New developments in the biology of fibroblast growth factors. WIREs Mech Dis 2022; 14:e1549. [PMID: 35142107 PMCID: PMC10115509 DOI: 10.1002/wsbm.1549] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 11/08/2021] [Accepted: 11/09/2021] [Indexed: 01/28/2023]
Abstract
The fibroblast growth factor (FGF) family is composed of 18 secreted signaling proteins consisting of canonical FGFs and endocrine FGFs that activate four receptor tyrosine kinases (FGFRs 1-4) and four intracellular proteins (intracellular FGFs or iFGFs) that primarily function to regulate the activity of voltage-gated sodium channels and other molecules. The canonical FGFs, endocrine FGFs, and iFGFs have been reviewed extensively by us and others. In this review, we briefly summarize past reviews and then focus on new developments in the FGF field since our last review in 2015. Some of the highlights in the past 6 years include the use of optogenetic tools, viral vectors, and inducible transgenes to experimentally modulate FGF signaling, the clinical use of small molecule FGFR inhibitors, an expanded understanding of endocrine FGF signaling, functions for FGF signaling in stem cell pluripotency and differentiation, roles for FGF signaling in tissue homeostasis and regeneration, a continuing elaboration of mechanisms of FGF signaling in development, and an expanding appreciation of roles for FGF signaling in neuropsychiatric diseases. This article is categorized under: Cardiovascular Diseases > Molecular and Cellular Physiology Neurological Diseases > Molecular and Cellular Physiology Congenital Diseases > Stem Cells and Development Cancer > Stem Cells and Development.
Collapse
Affiliation(s)
- David M Ornitz
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Nobuyuki Itoh
- Kyoto University Graduate School of Pharmaceutical Sciences, Sakyo, Kyoto, Japan
| |
Collapse
|
11
|
Mitroi DN, Tian M, Kawaguchi R, Lowry WE, Carmichael ST. Single-nucleus transcriptome analysis reveals disease- and regeneration-associated endothelial cells in white matter vascular dementia. J Cell Mol Med 2022; 26:3183-3195. [PMID: 35543222 PMCID: PMC9170821 DOI: 10.1111/jcmm.17315] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 02/01/2022] [Accepted: 03/12/2022] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Vascular dementia (VaD) is the accumulation of vascular lesions in the subcortical white matter of the brain. These lesions progress and there is no direct medical therapy. AIMS To determine the specific cellular responses in VaD so as to provide molecular targets for therapeutic development. MATERIALS AND METHODS Single-nucleus transcriptome analysis was performed in human periventricular white matter (PVWM) samples of VaD and normal control (NC) subjects. RESULTS Differential analysis shows that cell type-specific transcriptomic changes in VaD are associated with the disruption of specific biological processes, including angiogenesis, immune activation, axonal injury and myelination. Each cell type in the neurovascular unit within white matter has a specific alteration in gene expression in VaD. In a central cell type for this disease, subcluster analysis of endothelial cells (EC) indicates that VaD contains a disease-associated EC subcluster that expresses genes associated with programmed cell death and a response to protein folding. Two other subpopulations of EC in VaD express molecular systems associated with regenerative processes in angiogenesis, and in axonal sprouting and oligodendrocyte progenitor cell maturation. CONCLUSION This comprehensive molecular profiling of brain samples from patients with VaD reveals previously unknown molecular changes in cells of the neurovascular niche, and an attempt at regeneration in injured white matter.
Collapse
Affiliation(s)
- Daniel N. Mitroi
- Department of Psychiatry and Biobehavioral SciencesDavid Geffen School of Medicine at UCLALos AngelesCaliforniaUSA
| | - Min Tian
- Department of Psychiatry and Biobehavioral SciencesDavid Geffen School of Medicine at UCLALos AngelesCaliforniaUSA
| | - Riki Kawaguchi
- Department of Psychiatry and Biobehavioral SciencesDavid Geffen School of Medicine at UCLALos AngelesCaliforniaUSA
| | - William E. Lowry
- Department of Molecular, Cell and Developmental BiologyUCLALos AngelesCaliforniaUSA
| | - S. Thomas Carmichael
- Department of Psychiatry and Biobehavioral SciencesDavid Geffen School of Medicine at UCLALos AngelesCaliforniaUSA
| |
Collapse
|
12
|
Di Schiavi E, Vistoli G, Moretti RM, Corrado I, Zuccarini G, Gervasoni S, Casati L, Bottai D, Merlo GR, Maggi R. Anosmin-1-Like Effect of UMODL1/Olfactorin on the Chemomigration of Mouse GnRH Neurons and Zebrafish Olfactory Axons Development. Front Cell Dev Biol 2022; 10:836179. [PMID: 35223856 PMCID: PMC8874799 DOI: 10.3389/fcell.2022.836179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 01/17/2022] [Indexed: 11/13/2022] Open
Abstract
The impairment of development/migration of hypothalamic gonadotropin-releasing hormone (GnRH) neurons is the main cause of Kallmann's syndrome (KS), an inherited disorder characterized by hypogonadism, anosmia, and other developmental defects. Olfactorin is an extracellular matrix protein encoded by the UMODL1 (uromodulin-like 1) gene expressed in the mouse olfactory region along the migratory route of GnRH neurons. It shares a combination of WAP and FNIII repeats, expressed in complementary domains, with anosmin-1, the product of the ANOS1 gene, identified as the causative of KS. In the present study, we have investigated the effects of olfactorin in vitro and in vivo models. The results show that olfactorin exerts an anosmin-1-like strong chemoattractant effect on mouse-immortalized GnRH neurons (GN11 cells) through the activation of the FGFR and MAPK pathways. In silico analysis of olfactorin and anosmin-1 reveals a satisfactory similarity at the N-terminal region for the overall arrangement of corresponding WAP and FNIII domains and marked similarities between WAP domains’ binding modes of interaction with the resolved FGFR1–FGF2 complex. Finally, in vivo experiments show that the down-modulation of the zebrafish z-umodl1 gene (orthologous of UMODL1) in both GnRH3:GFP and omp2k:gap-CFPrw034 transgenic zebrafish strains leads to a clear disorganization and altered fasciculation of the neurites of GnRH3:GFP neurons crossing at the anterior commissure and a significant increase in olfactory CFP + fibers with altered trajectory. Thus, our study shows olfactorin as an additional factor involved in the development of olfactory and GnRH systems and proposes UMODL1 as a gene worthy of diagnostic investigation in KS.
Collapse
Affiliation(s)
- Elia Di Schiavi
- Institute of Biosciences and Bioresources, National Research Council of Italy, Naples, Italy
| | - Giulio Vistoli
- Department of Pharmaceutical Sciences DISFARM, Università degli Studi di Milano, Milano, Italy
| | - Roberta Manuela Moretti
- Department of Pharmacological and Biomolecular Sciences DISFEB, Università degli Studi di Milano, Milano, Italy
| | - Ilaria Corrado
- Department Molecular Biotechnology and Health Science, University of Torino, Torino, Italy
| | - Giulia Zuccarini
- Department Molecular Biotechnology and Health Science, University of Torino, Torino, Italy
| | - Silvia Gervasoni
- Department of Pharmaceutical Sciences DISFARM, Università degli Studi di Milano, Milano, Italy
| | - Lavinia Casati
- Department of Pharmaceutical Sciences DISFARM, Università degli Studi di Milano, Milano, Italy
| | - Daniele Bottai
- Department of Pharmaceutical Sciences DISFARM, Università degli Studi di Milano, Milano, Italy
| | - Giorgio Roberto Merlo
- Department Molecular Biotechnology and Health Science, University of Torino, Torino, Italy
| | - Roberto Maggi
- Department of Pharmaceutical Sciences DISFARM, Università degli Studi di Milano, Milano, Italy
- *Correspondence: Roberto Maggi,
| |
Collapse
|
13
|
David-Bercholz J, Kuo CT, Deneen B. Astrocyte and Oligodendrocyte Responses From the Subventricular Zone After Injury. Front Cell Neurosci 2022; 15:797553. [PMID: 35002630 PMCID: PMC8740317 DOI: 10.3389/fncel.2021.797553] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 11/15/2021] [Indexed: 11/14/2022] Open
Abstract
Under normal conditions, neural stem cells (NSCs or B cells) in the adult subventricular zone (SVZ) give rise to amplifying neural progenitor cells (NPCs or C cells), which can produce neuroblasts (or A cells) that migrate to the olfactory bulb and differentiate into new neurons. However, following brain injury, these cells migrate toward the injury site where they differentiate into astrocytes and oligodendrocytes. In this review, we will focus on recent findings that chronicle how astrocytes and oligodendrocytes derived from SVZ-NSCs respond to different types of injury. We will also discuss molecular regulators of SVZ-NSC proliferation and their differentiation into astrocytes and oligodendrocytes. Overall, the goal of this review is to highlight how SVZ-NSCs respond to injury and to summarize the regulatory mechanisms that oversee their glial response. These molecular and cellular processes will provide critical insights needed to develop strategies to promote brain repair following injury using SVZ-NSCs.
Collapse
Affiliation(s)
- Jennifer David-Bercholz
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, United States.,Department of Anesthesiology, Duke University School of Medicine, Durham, NC, United States
| | - Chay T Kuo
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, United States
| | - Benjamin Deneen
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, United States.,Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States.,Department of Neurosurgery, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
14
|
Ghelman J, Grewing L, Windener F, Albrecht S, Zarbock A, Kuhlmann T. SKAP2 as a new regulator of oligodendroglial migration and myelin sheath formation. Glia 2021; 69:2699-2716. [PMID: 34324225 DOI: 10.1002/glia.24066] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 07/13/2021] [Accepted: 07/15/2021] [Indexed: 02/06/2023]
Abstract
Oligodendroglial progenitor cells (OPCs) are highly proliferative and migratory cells, which differentiate into complex myelin forming and axon ensheathing mature oligodendrocytes during myelination. Recent studies indicate that the oligodendroglial cell population is heterogeneous on transcriptional and functional level depending on the location in the central nervous system. Here, we compared intrinsic properties of OPC from spinal cord and brain on functional and transcriptional level. Spinal cord OPC demonstrated increased migration as well as differentiation capacity. Moreover, transcriptome analysis revealed differential expression of several genes between both OPC populations. In spinal cord OPC, we confirmed upregulation of SKAP2, a cytoplasmatic adaptor protein known for its implication in cytoskeletal remodeling and migration in other cell types. Recent findings suggest that actin dynamics determine not only oligodendroglial migration, but also differentiation: Whereas actin polymerization is important for process extension, actin destabilization and depolymerization is required for myelin sheath formation. Downregulation or complete lack of SKAP2 in OPC resulted in reduced migration and impaired morphological maturation in oligodendrocytes. In contrast, overexpression of SKAP2 as well as constitutively active SKAP2 increased OPC migration suggesting that SKAP2 function is dependent on activation by phosphorylation. Furthermore, lack of SKAP2 enhanced the positive effect on OPC migration after integrin activation suggesting that SKAP2 acts as modulator of integrin dependent migration. In summary, we demonstrate the presence of intrinsic differences between spinal cord and brain OPC and identified SKAP2 as a new regulator of oligodendroglial migration and sheath formation.
Collapse
Affiliation(s)
- Julia Ghelman
- Institute of Neuropathology, University Hospital Muenster, Muenster, Germany
| | - Laureen Grewing
- Institute of Neuropathology, University Hospital Muenster, Muenster, Germany
| | - Farina Windener
- Institute of Neuropathology, University Hospital Muenster, Muenster, Germany
| | - Stefanie Albrecht
- Institute of Neuropathology, University Hospital Muenster, Muenster, Germany
| | - Alexander Zarbock
- Department of Anesthesiology, Intensive Care, and Pain Medicine, University Hospital Muenster, University of Muenster, Muenster, Germany
| | - Tanja Kuhlmann
- Institute of Neuropathology, University Hospital Muenster, Muenster, Germany
| |
Collapse
|
15
|
Chronic Low Dose Neutron Exposure Results in Altered Neurotransmission Properties of the Hippocampus-Prefrontal Cortex Axis in Both Mice and Rats. Int J Mol Sci 2021; 22:ijms22073668. [PMID: 33915974 PMCID: PMC8036585 DOI: 10.3390/ijms22073668] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 03/29/2021] [Accepted: 03/31/2021] [Indexed: 12/18/2022] Open
Abstract
The proposed deep space exploration to the moon and later to Mars will result in astronauts receiving significant chronic exposures to space radiation (SR). SR exposure results in multiple neurocognitive impairments. Recently, our cross-species (mouse/rat) studies reported impaired associative memory formation in both species following a chronic 6-month low dose exposure to a mixed field of neutrons (1 mGy/day for a total dose pf 18 cGy). In the present study, we report neutron exposure induced synaptic plasticity in the medial prefrontal cortex, accompanied by microglial activation and significant synaptic loss in the hippocampus. In a parallel study, neutron exposure was also found to alter fluorescence assisted single synaptosome LTP (FASS-LTP) in the hippocampus of rats, that may be related to a reduced ability to insert AMPAR into the post-synaptic membrane, which may arise from increased phosphorylation of the serine 845 residue of the GluA1 subunit. Thus, we demonstrate for the first time, that low dose chronic neutron irradiation impacts homeostatic synaptic plasticity in the hippocampal-cortical circuit in two rodent species, and that the ability to successfully encode associative recognition memory is a dynamic, multicircuit process, possibly involving compensatory changes in AMPAR density on the synaptic surface.
Collapse
|
16
|
Alcover-Sanchez B, Garcia-Martin G, Escudero-Ramirez J, Gonzalez-Riano C, Lorenzo P, Gimenez-Cassina A, Formentini L, de la Villa-Polo P, Pereira MP, Wandosell F, Cubelos B. Absence of R-Ras1 and R-Ras2 causes mitochondrial alterations that trigger axonal degeneration in a hypomyelinating disease model. Glia 2020; 69:619-637. [PMID: 33010069 DOI: 10.1002/glia.23917] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 08/18/2020] [Accepted: 09/21/2020] [Indexed: 12/11/2022]
Abstract
Fast synaptic transmission in vertebrates is critically dependent on myelin for insulation and metabolic support. Myelin is produced by oligodendrocytes (OLs) that maintain multilayered membrane compartments that wrap around axonal fibers. Alterations in myelination can therefore lead to severe pathologies such as multiple sclerosis. Given that hypomyelination disorders have complex etiologies, reproducing clinical symptoms of myelin diseases from a neurological perspective in animal models has been difficult. We recently reported that R-Ras1-/- and/or R-Ras2-/- mice, which lack GTPases essential for OL survival and differentiation processes, present different degrees of hypomyelination in the central nervous system with a compounded hypomyelination in double knockout (DKO) mice. Here, we discovered that the loss of R-Ras1 and/or R-Ras2 function is associated with aberrant myelinated axons with increased numbers of mitochondria, and a disrupted mitochondrial respiration that leads to increased reactive oxygen species levels. Consequently, aberrant myelinated axons are thinner with cytoskeletal phosphorylation patterns typical of axonal degeneration processes, characteristic of myelin diseases. Although we observed different levels of hypomyelination in a single mutant mouse, the combined loss of function in DKO mice lead to a compromised axonal integrity, triggering the loss of visual function. Our findings demonstrate that the loss of R-Ras function reproduces several characteristics of hypomyelinating diseases, and we therefore propose that R-Ras1-/- and R-Ras2-/- neurological models are valuable approaches for the study of these myelin pathologies.
Collapse
Affiliation(s)
- Berta Alcover-Sanchez
- Departamento de Biología Molecular and Centro Biología Molecular "Severo Ochoa", Universidad Autónoma de Madrid - Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Gonzalo Garcia-Martin
- Departamento de Biología Molecular and Centro Biología Molecular "Severo Ochoa", Universidad Autónoma de Madrid - Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Juan Escudero-Ramirez
- Departamento de Biología Molecular and Centro Biología Molecular "Severo Ochoa", Universidad Autónoma de Madrid - Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Carolina Gonzalez-Riano
- CEMBIO (Centre for Metabolomics and Bioanalysis), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - Paz Lorenzo
- CEMBIO (Centre for Metabolomics and Bioanalysis), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - Alfredo Gimenez-Cassina
- Departamento de Biología Molecular and Centro Biología Molecular "Severo Ochoa", Universidad Autónoma de Madrid - Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Laura Formentini
- Departamento de Biología Molecular and Centro Biología Molecular "Severo Ochoa", Universidad Autónoma de Madrid - Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Pedro de la Villa-Polo
- Departamento de Biología de Sistemas, Universidad de Alcalá, Madrid, Spain.,Grupo de Neurofisiología Visual, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Marta P Pereira
- Departamento de Biología Molecular and Centro Biología Molecular "Severo Ochoa", Universidad Autónoma de Madrid - Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Francisco Wandosell
- Departamento de Biología Molecular and Centro Biología Molecular "Severo Ochoa", Universidad Autónoma de Madrid - Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Beatriz Cubelos
- Departamento de Biología Molecular and Centro Biología Molecular "Severo Ochoa", Universidad Autónoma de Madrid - Consejo Superior de Investigaciones Científicas, Madrid, Spain
| |
Collapse
|
17
|
Amini H, Shroff N, Stamova B, Ferino E, Carmona‐Mora P, Zhan X, Sitorus PP, Hull H, Jickling GC, Sharp FR, Ander BP. Genetic variation contributes to gene expression response in ischemic stroke: an eQTL study. Ann Clin Transl Neurol 2020; 7:1648-1660. [PMID: 32785988 PMCID: PMC7480928 DOI: 10.1002/acn3.51154] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 06/27/2020] [Accepted: 07/13/2020] [Indexed: 01/06/2023] Open
Abstract
OBJECTIVE Single nucleotide polymorphisms (SNPs) contribute to complex disorders such as ischemic stroke (IS). Since SNPs could affect IS by altering gene expression, we studied the association of common SNPs with changes in mRNA expression (i.e. expression quantitative trait loci; eQTL) in blood after IS. METHODS RNA and DNA were isolated from 137 patients with acute IS and 138 vascular risk factor controls (VRFC). Gene expression was measured using Affymetrix HTA 2.0 microarrays and SNP variants were assessed with Axiom Biobank Genotyping microarrays. A linear model with a genotype (SNP) × diagnosis (IS and VRFC) interaction term was fit for each SNP-gene pair. RESULTS The eQTL interaction analysis revealed significant genotype × diagnosis interaction for four SNP-gene pairs as cis-eQTL and 70 SNP-gene pairs as trans-eQTL. Cis-eQTL involved in the inflammatory response to IS included rs56348411 which correlated with neurogranin expression (NRGN), rs78046578 which correlated with CXCL10 expression, rs975903 which correlated with SMAD4 expression, and rs62299879 which correlated with CD38 expression. These four genes are important in regulating inflammatory response and BBB stabilization. SNP rs148791848 was a strong trans-eQTL for anosmin-1 (ANOS1) which is involved in neural cell adhesion and axonal migration and may be important after stroke. INTERPRETATION This study highlights the contribution of genetic variation to regulating gene expression following IS. Specific inflammatory response to stroke is at least partially influenced by genetic variation. This has implications for progressing toward personalized treatment strategies. Additional research is required to investigate these genes as therapeutic targets.
Collapse
Affiliation(s)
- Hajar Amini
- Department of NeurologyUniversity of California at DavisSacramentoCalifornia95817
| | - Natasha Shroff
- Department of NeurologyUniversity of California at DavisSacramentoCalifornia95817
| | - Boryana Stamova
- Department of NeurologyUniversity of California at DavisSacramentoCalifornia95817
| | - Eva Ferino
- Department of NeurologyUniversity of California at DavisSacramentoCalifornia95817
| | - Paulina Carmona‐Mora
- Department of NeurologyUniversity of California at DavisSacramentoCalifornia95817
| | - Xinhua Zhan
- Department of NeurologyUniversity of California at DavisSacramentoCalifornia95817
| | - Preston P. Sitorus
- Department of NeurologyUniversity of California at DavisSacramentoCalifornia95817
| | - Heather Hull
- Department of NeurologyUniversity of California at DavisSacramentoCalifornia95817
| | - Glen C. Jickling
- Department of NeurologyUniversity of California at DavisSacramentoCalifornia95817
| | - Frank R. Sharp
- Department of NeurologyUniversity of California at DavisSacramentoCalifornia95817
| | - Bradley P. Ander
- Department of NeurologyUniversity of California at DavisSacramentoCalifornia95817
| |
Collapse
|
18
|
Deboux C, Spigoni G, Caillava C, Garcia-Diaz B, Ypsilanti A, Sarrazin N, Bachelin C, Chédotal A, Baron-Van Evercooren A. Slit1 Protein Regulates SVZ-Derived Precursor Mobilization in the Adult Demyelinated CNS. Front Cell Neurosci 2020; 14:168. [PMID: 32670024 PMCID: PMC7332780 DOI: 10.3389/fncel.2020.00168] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 05/19/2020] [Indexed: 01/03/2023] Open
Abstract
Slit1 is a secreted axon guidance molecule, also involved in adult neurogenesis. In physiological conditions, Slit1 loss promotes ectopic dispersal of SVZ-derived neural precursors (SVZ-NPCs) into periventricular structures such as the corpus callosum. Demyelination of the corpus callosum triggers SVZ-NPC migration to ectopic locations and their recruitment by the lesion, suggesting a possible role for Slit1 in SVZ-NPCs ectopic dispersal regulation in pathological conditions. Here, we have investigated the function of Slit1 protein in the recruitment of SVZ-NPCs after CNS demyelination. We find that the dynamics of oligodendrogenesis and temporal profile of developmental myelination in Slit1–/– mice are similar to Slit1+/− controls. SVZ micro-dissection and RT-PCR from wild-type mice, show that Slits and Robos are physiologically regulated at the transcriptional level in response to corpus callosum demyelination suggesting their role in the process of SVZ-NPC ectopic migration in demyelinating conditions. Moreover, we find that the number of SVZ-NPCs recruited by the lesion increases in Sli1–/– mice compared to Slit1+/− mice, leading to higher numbers of Olig2+ cells within the lesion. Time-lapse video-microscopy of immuno-purified NPCs shows that Slit1-deficient cells migrate faster and make more frequent directional changes than control NPCs, supporting a cell-autonomous mechanism of action of Slit1 in NPC migration. In conclusion, while Slit1 does not affect the normal developmental process of oligodendrogenesis and myelination, it regulates adult SVZ-NPC ectopic migration in response to demyelination, and consequently oligodendrocyte renewal within the lesion.
Collapse
Affiliation(s)
- C Deboux
- Institut du Cerveau et de la Moelle épinière-Groupe Hospitalier Pitié-Salpêtrière, INSERM U1127, CNRS, UMR 7225, Sorbonne Université, UM75, Paris, France
| | - G Spigoni
- Institut du Cerveau et de la Moelle épinière-Groupe Hospitalier Pitié-Salpêtrière, INSERM U1127, CNRS, UMR 7225, Sorbonne Université, UM75, Paris, France
| | - C Caillava
- Institut du Cerveau et de la Moelle épinière-Groupe Hospitalier Pitié-Salpêtrière, INSERM U1127, CNRS, UMR 7225, Sorbonne Université, UM75, Paris, France
| | - B Garcia-Diaz
- Institut du Cerveau et de la Moelle épinière-Groupe Hospitalier Pitié-Salpêtrière, INSERM U1127, CNRS, UMR 7225, Sorbonne Université, UM75, Paris, France
| | - A Ypsilanti
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, Paris, France
| | - N Sarrazin
- Institut du Cerveau et de la Moelle épinière-Groupe Hospitalier Pitié-Salpêtrière, INSERM U1127, CNRS, UMR 7225, Sorbonne Université, UM75, Paris, France
| | - C Bachelin
- Institut du Cerveau et de la Moelle épinière-Groupe Hospitalier Pitié-Salpêtrière, INSERM U1127, CNRS, UMR 7225, Sorbonne Université, UM75, Paris, France
| | - A Chédotal
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, Paris, France
| | - A Baron-Van Evercooren
- Institut du Cerveau et de la Moelle épinière-Groupe Hospitalier Pitié-Salpêtrière, INSERM U1127, CNRS, UMR 7225, Sorbonne Université, UM75, Paris, France
| |
Collapse
|
19
|
Ma W, Mao J, Wang X, Duan L, Song Y, Lian X, Zheng J, Liu Z, Nie M, Wu X. Novel Microdeletion in the X Chromosome Leads to Kallmann Syndrome, Ichthyosis, Obesity, and Strabismus. Front Genet 2020; 11:596. [PMID: 32670353 PMCID: PMC7327112 DOI: 10.3389/fgene.2020.00596] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 05/15/2020] [Indexed: 11/23/2022] Open
Abstract
Background A large deletion in Xp22.3 can result in contiguous gene syndromes, including X-linked ichthyosis (XLI) and Kallmann syndrome (KS), presenting with short stature, chondrodysplasia punctata, intellectual disability, and strabismus. XLI and KS are caused by the deletion of STS and ANOS1, respectively. Method Two KS patients with XLI were screened to identify possible pathogenic mutations using whole exome sequencing. The clinical characteristics, molecular genetics, treatment outcomes, and genotype–phenotype association for each patient were analyzed. Results We identified a novel 3,923 kb deletion within the Xp22.31 region (chrX: 5810838–9733877) containing STS, ANOS1, GPR143, NLGN4X, VCX-A, PUDP, and PNPLA4 in patient 1, who presented with KS, XLI, obesity, hyperlipidemia, and strabismus. We identified a novel 5,807 kb deletion within the Xp22.31-p22.33 regions (chrX: 2700083–8507807) containing STS, ANOS1, and other 24 genes in patient 2, who presented with KS, XLI, obesity, and strabismus. No developmental delay, abnormal speech development, or autistic behavior were noticed in either patient. Conclusion We identified two novel microdeletions in the X chromosome leading to KS and XLI. These findings contribute to the understanding of the molecular mechanisms that drive contiguous gene syndromes. Our research confirmed that the Kallmann-Ichthyosis phenotype is caused by microdeletions at the chromosome level.
Collapse
Affiliation(s)
- Wanlu Ma
- Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Jiangfeng Mao
- Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Xi Wang
- Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Lian Duan
- Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Yuwen Song
- Department of Endocrinology, The Second Hospital of Shandong University, Jinan, China
| | - Xiaolan Lian
- Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Junjie Zheng
- Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Zhaoxiang Liu
- Department of Endocrinology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Min Nie
- Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Xueyan Wu
- Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
20
|
Functional Heterogeneity of Mouse and Human Brain OPCs: Relevance for Preclinical Studies in Multiple Sclerosis. J Clin Med 2020; 9:jcm9061681. [PMID: 32498223 PMCID: PMC7355819 DOI: 10.3390/jcm9061681] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 05/19/2020] [Indexed: 02/08/2023] Open
Abstract
Besides giving rise to oligodendrocytes (the only myelin-forming cell in the Central Nervous System (CNS) in physiological conditions), Oligodendrocyte Precursor Cells (OPCs) are responsible for spontaneous remyelination after a demyelinating lesion. They are present along the mouse and human CNS, both during development and in adulthood, yet how OPC physiological behavior is modified throughout life is not fully understood. The activity of adult human OPCs is still particularly unexplored. Significantly, most of the molecules involved in OPC-mediated remyelination are also involved in their development, a phenomenon that may be clinically relevant. In the present article, we have compared the intrinsic properties of OPCs isolated from the cerebral cortex of neonatal, postnatal and adult mice, as well as those recovered from neurosurgical adult human cerebral cortex tissue. By analyzing intact OPCs for the first time with 1H High Resolution Magic Angle Spinning Nuclear Magnetic Resonance (1H HR-MAS NMR) spectroscopy, we show that these cells behave distinctly and that they have different metabolic patterns in function for their stage of maturity. Moreover, their response to Fibroblast Growth Gactor-2 (FGF-2) and anosmin-1 (two molecules that have known effects on OPC biology during development and that are overexpressed in individuals with Multiple Sclerosis (MS)) differs in relation to their developmental stage and in the function of the species. Our data reveal that the behavior of adult human and mouse OPCs differs in a very dynamic way that should be very relevant when testing drugs and for the proper design of effective pharmacological and/or cell therapies for MS.
Collapse
|
21
|
Traiffort E, Kassoussi A, Zahaf A, Laouarem Y. Astrocytes and Microglia as Major Players of Myelin Production in Normal and Pathological Conditions. Front Cell Neurosci 2020; 14:79. [PMID: 32317939 PMCID: PMC7155218 DOI: 10.3389/fncel.2020.00079] [Citation(s) in RCA: 126] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 03/19/2020] [Indexed: 12/13/2022] Open
Abstract
Myelination is an essential process that consists of the ensheathment of axons by myelin. In the central nervous system (CNS), myelin is synthesized by oligodendrocytes. The proliferation, migration, and differentiation of oligodendrocyte precursor cells constitute a prerequisite before mature oligodendrocytes extend their processes around the axons and progressively generate a multilamellar lipidic sheath. Although myelination is predominately driven by oligodendrocytes, the other glial cells including astrocytes and microglia, also contribute to this process. The present review is an update of the most recent emerging mechanisms involving astrocyte and microglia in myelin production. The contribution of these cells will be first described during developmental myelination that occurs in the early postnatal period and is critical for the proper development of cognition and behavior. Then, we will report the novel findings regarding the beneficial or deleterious effects of astroglia and microglia, which respectively promote or impair the endogenous capacity of oligodendrocyte progenitor cells (OPCs) to induce spontaneous remyelination after myelin loss. Acute delineation of astrocyte and microglia activities and cross-talk should uncover the way towards novel therapeutic perspectives aimed at recovering proper myelination during development or at breaking down the barriers impeding the regeneration of the damaged myelin that occurs in CNS demyelinating diseases.
Collapse
Affiliation(s)
| | | | - Amina Zahaf
- U1195 Inserm, University Paris-Saclay, Kremlin-Bicêtre, France
| | - Yousra Laouarem
- U1195 Inserm, University Paris-Saclay, Kremlin-Bicêtre, France
| |
Collapse
|
22
|
Kanda M, Suh YS, Park DJ, Tanaka C, Ahn SH, Kong SH, Lee HJ, Kobayashi D, Fujiwara M, Shimada H, Cho B, Murotani K, Kim HH, Yang HK, Kodera Y. Serum levels of ANOS1 serve as a diagnostic biomarker of gastric cancer: a prospective multicenter observational study. Gastric Cancer 2020; 23:203-211. [PMID: 31377880 DOI: 10.1007/s10120-019-00995-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 07/26/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND Development of high-performance serum biomarkers will likely improve treatment outcomes of patients with gastric cancer (GC). We previously identified the candidate serum markers, anosmin 1 (ANOS1), dihydropyrimidinase-like 3 (DPYSL3), and melanoma-associated antigen D2 (MAGE-D2) and evaluated their clinical significance through a single-center retrospective analysis. Here we conducted a prospective multicenter observational study aimed at validating the diagnostic performance of these potential markers. METHODS We analyzed serum levels before and after surgery of the three potential biomarkers in patients with GC and healthy volunteers. Quantification of serum and GC tissue levels was performed using an ELISA. RESULTS Area under the curve (AUC) values that discriminated patients with GC from healthy controls were - 0.7058, 0.6188, and 0.5031 for ANOS1, DPYSL3, and MAGED2, respectively. The sensitivity and specificity of the ANOS1 assay were 0.36 and 0.85, respectively. The AUC value of ANOS1 that discriminated patients with stage I GC from healthy controls was 0.7131. Serum ANOS1 levels were significantly elevated in patients with stage I GC compared with those of healthy controls (median 1179 ng/ml and 461 ng/ml, respectively, P < 0.0001) and decreased after resection of primary GC lesions (P < 0.0001). The combination of serum ANOS1 and DPYSL3 levels increased the AUC value that discriminated patients with GC from healthy controls. Serum levels of ANOS1 did not significantly correlate with those of carcinoembryonic antigen, carbohydrate antigen 19-9, or other markers of inflammation. CONCLUSIONS Serum levels of ANOS1 may serve as a useful diagnostic tool for managing GC.
Collapse
Affiliation(s)
- Mitsuro Kanda
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Yun-Suhk Suh
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Do Joong Park
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Chie Tanaka
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Sang-Hoon Ahn
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Seong-Ho Kong
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Hyuk-Joon Lee
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
- Cancer Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Daisuke Kobayashi
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Michitaka Fujiwara
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Hideaki Shimada
- Department of Gastroenterological Surgery, Graduate School of Medicine, Toho University, Ota City, Japan
| | - BeLong Cho
- Department of Family Medicine, Center for Health Promotion and Optimal Aging, Seoul National University College of Medicine and Hospital, Seoul, Korea
- Institute On Aging, Seoul National University College of Medicine, Seoul, Korea
| | - Kenta Murotani
- Biostatistics Center, Graduate School of Medicine, Kurume University, Kurume, Japan
| | - Hyung-Ho Kim
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Han-Kwang Yang
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
- Cancer Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Yasuhiro Kodera
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan.
| |
Collapse
|
23
|
Scheuer T, Klein LS, Bührer C, Endesfelder S, Schmitz T. Transient Improvement of Cerebellar Oligodendroglial Development in a Neonatal Hyperoxia Model by PDGFA Treatment. Dev Neurobiol 2019; 79:222-235. [PMID: 30674088 DOI: 10.1002/dneu.22667] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 01/10/2019] [Accepted: 01/12/2019] [Indexed: 12/13/2022]
Abstract
In preterm infants, the changes from fetal life to ex-utero conditions often coincide with reduced growth and white matter damage of the cerebellum. The premature increase in arterial oxygen tension caused by preterm birth may dysregulate cerebellar development. In a hyperoxia rat model of white matter damage to mimic a steep increase in oxygen levels by 24 h exposure to 80% O2 from postnatal day 6 (P6) to day 7, we analyzed growth factor (GF) synthesis of cerebellar astrocytes. Determination of GF production was performed in astrocytes after Magnetic-activated cell sorting (MACS) isolation from cerebelli after hyperoxia exposure ex vivo, and also in astroglial cultures. Oligodendrocyte progenitor cell (OPC) function was analyzed in cerebellar OPCs isolated by MACS after hyperoxia. Administration of PDGFA from P6 to P11, during hyperoxia and during 4 days recovery, was finally tested for protection of oligodendroglia and myelination. As a result, expression of the GFs Pdgfa, Fgf2, and Bdnf was diminished in cerebellar astrocytes in vitro and in vivo. Gene expression of Olig1, Olig2, Sox9, Sox10, and Cnp was reduced in OPCs in vivo. Nasal PDGFA application improved oligodendroglial proliferation after hyperoxia at P7. However, this treatment effect vanished until P9. Impaired MBP expression after hyperoxia was attenuated by PDGFA treatment until P11, but not beyond when PDGFA supply was stopped. In this study on neonatal cerebellar injury, it is documented for the first time that improvement of oligodendroglial proliferation and of myelination can be achieved by PDGFA treatment. However, the treatment benefit is not maintained long term.
Collapse
Affiliation(s)
- Till Scheuer
- Department for Neonatology, Charité University Medical Center, Berlin, Germany
| | - Luisa Sophie Klein
- Department for Neonatology, Charité University Medical Center, Berlin, Germany
| | - Christoph Bührer
- Department for Neonatology, Charité University Medical Center, Berlin, Germany
| | | | - Thomas Schmitz
- Department for Neonatology, Charité University Medical Center, Berlin, Germany
| |
Collapse
|
24
|
Pepper RE, Pitman KA, Cullen CL, Young KM. How Do Cells of the Oligodendrocyte Lineage Affect Neuronal Circuits to Influence Motor Function, Memory and Mood? Front Cell Neurosci 2018; 12:399. [PMID: 30524235 PMCID: PMC6262292 DOI: 10.3389/fncel.2018.00399] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 10/17/2018] [Indexed: 12/11/2022] Open
Abstract
Oligodendrocyte progenitor cells (OPCs) are immature cells in the central nervous system (CNS) that can rapidly respond to changes within their environment by modulating their proliferation, motility and differentiation. OPCs differentiate into myelinating oligodendrocytes throughout life, and both cell types have been implicated in maintaining and modulating neuronal function to affect motor performance, cognition and emotional state. However, questions remain about the mechanisms employed by OPCs and oligodendrocytes to regulate circuit function, including whether OPCs can only influence circuits through their generation of new oligodendrocytes, or can play other regulatory roles within the CNS. In this review, we detail the molecular and cellular mechanisms that allow OPCs, newborn oligodendrocytes and pre-existing oligodendrocytes to regulate circuit function and ultimately influence behavioral outcomes.
Collapse
Affiliation(s)
- Renee E Pepper
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Kimberley A Pitman
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Carlie L Cullen
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Kaylene M Young
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| |
Collapse
|
25
|
Dickstein DL, Talty R, Bresnahan E, Varghese M, Perry B, Janssen WGM, Sowa A, Giedzinski E, Apodaca L, Baulch J, Acharya M, Parihar V, Limoli CL. Alterations in synaptic density and myelination in response to exposure to high-energy charged particles. J Comp Neurol 2018; 526:2845-2855. [PMID: 30198564 DOI: 10.1002/cne.24530] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 07/06/2018] [Accepted: 08/21/2018] [Indexed: 12/21/2022]
Abstract
High-energy charged particles are considered particularly hazardous components of the space radiation environment. Such particles include fully ionized energetic nuclei of helium, silicon, and oxygen, among others. Exposure to charged particles causes reactive oxygen species production, which has been shown to result in neuronal dysfunction and myelin degeneration. Here we demonstrate that mice exposed to high-energy charged particles exhibited alterations in dendritic spine density in the hippocampus, with a significant decrease of thin spines in mice exposed to helium, oxygen, and silicon, compared to sham-irradiated controls. Electron microscopy confirmed these findings and revealed a significant decrease in overall synapse density and in nonperforated synapse density, with helium and silicon exhibiting more detrimental effects than oxygen. Degeneration of myelin was also evident in exposed mice with significant changes in the percentage of myelinated axons and g-ratios. Our data demonstrate that exposure to all types of high-energy charged particles have a detrimental effect, with helium and silicon having more synaptotoxic effects than oxygen. These results have important implications for the integrity of the central nervous system and the cognitive health of astronauts after prolonged periods of space exploration.
Collapse
Affiliation(s)
- Dara L Dickstein
- Uniformed Services University of Health Sciences, Bethesda, Maryland.,Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Ronan Talty
- Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Erin Bresnahan
- Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Merina Varghese
- Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Bayley Perry
- Uniformed Services University of Health Sciences, Bethesda, Maryland
| | - William G M Janssen
- Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Allison Sowa
- Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Erich Giedzinski
- Department of Radiation Oncology, University of California, Irvine, California
| | - Lauren Apodaca
- Department of Radiation Oncology, University of California, Irvine, California
| | - Janet Baulch
- Department of Radiation Oncology, University of California, Irvine, California
| | - Munjal Acharya
- Department of Radiation Oncology, University of California, Irvine, California
| | - Vipan Parihar
- Department of Radiation Oncology, University of California, Irvine, California
| | - Charles L Limoli
- Department of Radiation Oncology, University of California, Irvine, California
| |
Collapse
|
26
|
de Castro F, Seal R, Maggi R. ANOS1: a unified nomenclature for Kallmann syndrome 1 gene (KAL1) and anosmin-1. Brief Funct Genomics 2018; 16:205-210. [PMID: 27899353 PMCID: PMC5860151 DOI: 10.1093/bfgp/elw037] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
It is accepted that confusion regarding the description of genetic variants occurs when researchers do not use standard nomenclature. The Human Genome Organization Gene Nomenclature Committee contacted a panel of consultants, all working on the KAL1 gene, to propose an update of the nomenclature of the gene, as there was a convention in the literature of using the ‘KAL1’ symbol, when referring to the gene, but using the name ‘anosmin-1’ when referring to the protein. The new name, ANOS1, reflects protein name and is more transferrable across species.
Collapse
|
27
|
R-Ras1 and R-Ras2 Are Essential for Oligodendrocyte Differentiation and Survival for Correct Myelination in the Central Nervous System. J Neurosci 2018; 38:5096-5110. [PMID: 29720552 DOI: 10.1523/jneurosci.3364-17.2018] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 03/14/2018] [Accepted: 04/10/2018] [Indexed: 12/21/2022] Open
Abstract
Rapid and effective neural transmission of information requires correct axonal myelination. Modifications in myelination alter axonal capacity to transmit electric impulses and enable pathological conditions. In the CNS, oligodendrocytes (OLs) myelinate axons, a complex process involving various cellular interactions. However, we know little about the mechanisms that orchestrate correct myelination. Here, we demonstrate that OLs express R-Ras1 and R-Ras2. Using female and male mutant mice to delete these proteins, we found that activation of the PI3K/Akt and Erk1/2-MAPK pathways was weaker in mice lacking one or both of these GTPases, suggesting that both proteins coordinate the activity of these two pathways. Loss of R-Ras1 and/or R-Ras2 diminishes the number of OLs in major myelinated CNS tracts and increases the proportion of immature OLs. In R-Ras1-/- and R-Ras2-/--null mice, OLs show aberrant morphologies and fail to differentiate correctly into myelin-forming phenotypes. The smaller OL population and abnormal OL maturation induce severe hypomyelination, with shorter nodes of Ranvier in R-Ras1-/- and/or R-Ras2-/- mice. These defects explain the slower conduction velocity of myelinated axons that we observed in the absence of R-Ras1 and R-Ras2. Together, these results suggest that R-Ras1 and R-Ras2 are upstream elements that regulate the survival and differentiation of progenitors into OLs through the PI3K/Akt and Erk1/2-MAPK pathways for proper myelination.SIGNIFICANCE STATEMENT In this study, we show that R-Ras1 and R-Ras2 play essential roles in regulating myelination in vivo and control fundamental aspects of oligodendrocyte (OL) survival and differentiation through synergistic activation of PI3K/Akt and Erk1/2-MAPK signaling. Mice lacking R-Ras1 and/or R-Ras2 show a diminished OL population with a higher proportion of immature OLs, explaining the observed hypomyelination in main CNS tracts. In vivo electrophysiology recordings demonstrate a slower conduction velocity of nerve impulses in the absence of R-Ras1 and R-Ras2. Therefore, R-Ras1 and R-Ras2 are essential for proper axonal myelination and accurate neural transmission.
Collapse
|
28
|
van Tilborg E, de Theije CGM, van Hal M, Wagenaar N, de Vries LS, Benders MJ, Rowitch DH, Nijboer CH. Origin and dynamics of oligodendrocytes in the developing brain: Implications for perinatal white matter injury. Glia 2017; 66:221-238. [PMID: 29134703 PMCID: PMC5765410 DOI: 10.1002/glia.23256] [Citation(s) in RCA: 183] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 10/17/2017] [Accepted: 10/20/2017] [Indexed: 12/11/2022]
Abstract
Infants born prematurely are at high risk to develop white matter injury (WMI), due to exposure to hypoxic and/or inflammatory insults. Such perinatal insults negatively impact the maturation of oligodendrocytes (OLs), thereby causing deficits in myelination. To elucidate the precise pathophysiology underlying perinatal WMI, it is essential to fully understand the cellular mechanisms contributing to healthy/normal white matter development. OLs are responsible for myelination of axons. During brain development, OLs are generally derived from neuroepithelial zones, where neural stem cells committed to the OL lineage differentiate into OL precursor cells (OPCs). OPCs, in turn, develop into premyelinating OLs and finally mature into myelinating OLs. Recent studies revealed that OPCs develop in multiple waves and form potentially heterogeneous populations. Furthermore, it has been shown that myelination is a dynamic and plastic process with an excess of OPCs being generated and then abolished if not integrated into neural circuits. Myelination patterns between rodents and humans show high spatial and temporal similarity. Therefore, experimental studies on OL biology may provide novel insights into the pathophysiology of WMI in the preterm infant and offers new perspectives on potential treatments for these patients.
Collapse
Affiliation(s)
- Erik van Tilborg
- Laboratory of Neuroimmunology and Developmental Origins of Disease, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Caroline G M de Theije
- Laboratory of Neuroimmunology and Developmental Origins of Disease, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Maurik van Hal
- Laboratory of Neuroimmunology and Developmental Origins of Disease, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Nienke Wagenaar
- Department of Neonatology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Linda S de Vries
- Department of Neonatology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Manon J Benders
- Department of Neonatology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - David H Rowitch
- Department of Pediatrics, Eli and Edythe Broad Center for Stem Cell Research and Regeneration Medicine, University of California, San Francisco, San Francisco, California.,Department of Paediatrics, Wellcome Trust-MRC Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Cora H Nijboer
- Laboratory of Neuroimmunology and Developmental Origins of Disease, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
29
|
Neben CL, Lo M, Jura N, Klein OD. Feedback regulation of RTK signaling in development. Dev Biol 2017; 447:71-89. [PMID: 29079424 DOI: 10.1016/j.ydbio.2017.10.017] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 10/17/2017] [Accepted: 10/23/2017] [Indexed: 02/07/2023]
Abstract
Precise regulation of the amplitude and duration of receptor tyrosine kinase (RTK) signaling is critical for the execution of cellular programs and behaviors. Understanding these control mechanisms has important implications for the field of developmental biology, and in recent years, the question of how augmentation or attenuation of RTK signaling via feedback loops modulates development has become of increasing interest. RTK feedback regulation is also important for human disease research; for example, germline mutations in genes that encode RTK signaling pathway components cause numerous human congenital syndromes, and somatic alterations contribute to the pathogenesis of diseases such as cancers. In this review, we survey regulators of RTK signaling that tune receptor activity and intracellular transduction cascades, with a focus on the roles of these genes in the developing embryo. We detail the diverse inhibitory mechanisms utilized by negative feedback regulators that, when lost or perturbed, lead to aberrant increases in RTK signaling. We also discuss recent biochemical and genetic insights into positive regulators of RTK signaling and how these proteins function in tandem with negative regulators to guide embryonic development.
Collapse
Affiliation(s)
- Cynthia L Neben
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, San Francisco 94143, USA
| | - Megan Lo
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, San Francisco 94143, USA; Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Natalia Jura
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA; Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA.
| | - Ophir D Klein
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, San Francisco 94143, USA; Department of Pediatrics and Institute for Human Genetics, University of California, San Francisco, San Francisco 94143, USA.
| |
Collapse
|
30
|
Manso Y, Holland PR, Kitamura A, Szymkowiak S, Duncombe J, Hennessy E, Searcy JL, Marangoni M, Randall AD, Brown JT, McColl BW, Horsburgh K. Minocycline reduces microgliosis and improves subcortical white matter function in a model of cerebral vascular disease. Glia 2017; 66:34-46. [PMID: 28722234 DOI: 10.1002/glia.23190] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 06/26/2017] [Accepted: 06/27/2017] [Indexed: 12/19/2022]
Abstract
Chronic cerebral hypoperfusion is a key mechanism associated with white matter disruption in cerebral vascular disease and dementia. In a mouse model relevant to studying cerebral vascular disease, we have previously shown that cerebral hypoperfusion disrupts axon-glial integrity and the distribution of key paranodal and internodal proteins in subcortical myelinated axons. This disruption of myelinated axons is accompanied by increased microglia and cognitive decline. The aim of the present study was to investigate whether hypoperfusion impairs the functional integrity of white matter, its relation with axon-glial integrity and microglial number, and whether by targeting microglia these effects can be improved. We show that in response to increasing durations of hypoperfusion, the conduction velocity of myelinated fibres in the corpus callosum is progressively reduced and that paranodal and internodal axon-glial integrity is disrupted. The number of microglial cells increases in response to hypoperfusion and correlates with disrupted paranodal and internodal integrity and reduced conduction velocities. Further minocycline, a proposed anti-inflammatory and microglia inhibitor, restores white matter function related to a reduction in the number of microglia. The study suggests that microglial activation contributes to the structural and functional alterations of myelinated axons induced by cerebral hypoperfusion and that dampening microglia numbers/proliferation should be further investigated as potential therapeutic benefit in cerebral vascular disease.
Collapse
Affiliation(s)
- Yasmina Manso
- Centre for Neuroregeneration, University of Edinburgh, Chancellor's Building, 49 Little France Crescent, Edinburgh, EH16 4SB, United Kingdom
| | - Philip R Holland
- Centre for Neuroregeneration, University of Edinburgh, Chancellor's Building, 49 Little France Crescent, Edinburgh, EH16 4SB, United Kingdom
| | - Akihiro Kitamura
- Centre for Neuroregeneration, University of Edinburgh, Chancellor's Building, 49 Little France Crescent, Edinburgh, EH16 4SB, United Kingdom
| | - Stefan Szymkowiak
- University of Edinburgh, The Roslin Institute, Easter Bush, Edinburgh, EH25 9RG
| | - Jessica Duncombe
- Centre for Neuroregeneration, University of Edinburgh, Chancellor's Building, 49 Little France Crescent, Edinburgh, EH16 4SB, United Kingdom
| | - Edel Hennessy
- Centre for Neuroregeneration, University of Edinburgh, Chancellor's Building, 49 Little France Crescent, Edinburgh, EH16 4SB, United Kingdom
| | - James L Searcy
- Centre for Neuroregeneration, University of Edinburgh, Chancellor's Building, 49 Little France Crescent, Edinburgh, EH16 4SB, United Kingdom
| | - Martina Marangoni
- Centre for Neuroregeneration, University of Edinburgh, Chancellor's Building, 49 Little France Crescent, Edinburgh, EH16 4SB, United Kingdom
| | - Andrew D Randall
- University of Exeter Medical School, University of Exeter, Exeter, UK
| | - Jon T Brown
- University of Exeter Medical School, University of Exeter, Exeter, UK
| | - Barry W McColl
- University of Edinburgh, The Roslin Institute, Easter Bush, Edinburgh, EH25 9RG.,UK Dementia Research Institute, University of Edinburgh, Edinburgh Medical School, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
| | - Karen Horsburgh
- Centre for Neuroregeneration, University of Edinburgh, Chancellor's Building, 49 Little France Crescent, Edinburgh, EH16 4SB, United Kingdom
| |
Collapse
|
31
|
Ono K, Yoshii K, Tominaga H, Gotoh H, Nomura T, Takebayashi H, Ikenaka K. Oligodendrocyte precursor cells in the mouse optic nerve originate in the preoptic area. Brain Struct Funct 2017; 222:2441-2448. [PMID: 28293728 DOI: 10.1007/s00429-017-1394-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 02/24/2017] [Indexed: 11/28/2022]
Abstract
The present study aims to examine the origin of oligodendrocyte progenitor cells (OPCs) in the mouse optic nerve (ON) by labeling OPCs in the fetal forebrain. The labeling of OPCs in the ON was performed by injection of a retrovirus vector carrying the lacZ gene into the lateral ventricle, or by inducible Cre/loxP of Olig2-positive cells. The retrovirus labeling revealed that ventricular zone-derived cells of the fetal forebrain relocated to the ON and differentiated into oligodendrocytes. In addition, lineage tracing of Olig2-positive cells and whole-mount staining of PDGFRα-positive cells demonstrated that OPCs appeared by E12.5 in the preoptic area, and spread caudally to enter the ON. Our results also suggest that OPCs generated during the early stage are depleted from the ON after maturation.
Collapse
Affiliation(s)
- Katsuhiko Ono
- Department of Biology, Kyoto Prefectural University of Medicine, Kyoto, 606-0823, Japan.
| | - Kengo Yoshii
- Departments of Mathematics, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hiroyuki Tominaga
- Department of Biology, Kyoto Prefectural University of Medicine, Kyoto, 606-0823, Japan
| | - Hitoshi Gotoh
- Department of Biology, Kyoto Prefectural University of Medicine, Kyoto, 606-0823, Japan
| | - Tadashi Nomura
- Department of Biology, Kyoto Prefectural University of Medicine, Kyoto, 606-0823, Japan
| | | | - Kazuhiro Ikenaka
- Division of Neurobiology and Bioinformatics, National Institute for Physiological Sciences, Okazaki, Japan
| |
Collapse
|
32
|
Medina-Rodríguez EM, Bribián A, Boyd A, Palomo V, Pastor J, Lagares A, Gil C, Martínez A, Williams A, de Castro F. Promoting in vivo remyelination with small molecules: a neuroreparative pharmacological treatment for Multiple Sclerosis. Sci Rep 2017; 7:43545. [PMID: 28256546 PMCID: PMC5335257 DOI: 10.1038/srep43545] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 01/25/2017] [Indexed: 12/22/2022] Open
Abstract
Multiple Sclerosis (MS) is a neurodegenerative disease where immune-driven demyelination occurs with inefficient remyelination, but therapies are limited, especially those to enhance repair. Here, we show that the dual phosphodiesterase (PDE)7- glycogen synthase kinase (GSK)3 inhibitor, VP3.15, a heterocyclic small molecule with good pharmacokinetic properties and safety profile, improves in vivo remyelination in mouse and increases both adult mouse and adult human oligodendrocyte progenitor cell (OPC) differentiation, in addition to its immune regulatory action. The dual inhibition is synergistic, as increasing intracellular levels of cAMP by cyclic nucleotide PDE inhibition both suppresses the immune response and increases remyelination, and in addition, inhibition of GSK3 limits experimental autoimmune encephalomyelitis in mice. This combination of an advantageous effect on the immune response and an enhancement of repair, plus demonstration of its activity on adult human OPCs, leads us to propose dual PDE7-GSK3 inhibition, and specifically VP3.15, as a neuroprotective and neuroreparative disease-modifying treatment for MS.
Collapse
Affiliation(s)
- Eva María Medina-Rodríguez
- Grupo de Neurobiología del Desarrollo-GNDe, Hospital Nacional de Parapléjicos, Finca la Peraleda s/n, E- 45071, Toledo, Spain
| | - Ana Bribián
- Grupo de Neurobiología del Desarrollo-GNDe, Hospital Nacional de Parapléjicos, Finca la Peraleda s/n, E- 45071, Toledo, Spain
- Instituto Cajal-CSIC, Avda. Dr. Arce 37, E-28002, Madrid, Spain
| | - Amanda Boyd
- MRC-Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive, EH164UU, Edinburgh, UK
| | - Valle Palomo
- Centro de Investigaciones Biológicas, CIB-CSIC, Calle Ramiro de Maeztu 9, E-28040, Madrid, Spain
| | - Jesús Pastor
- Servicio de Neurofisiología Clínica, Hospital La Princesa, Calle Diego de León 62, E-28006,Madrid, Spain
| | - Alfonso Lagares
- Servicio de Neurocirugía, Hospital 12 de Octubre, Avda. de Córdoba s/n, E-28041,Madrid, Spain
| | - Carmen Gil
- Centro de Investigaciones Biológicas, CIB-CSIC, Calle Ramiro de Maeztu 9, E-28040, Madrid, Spain
| | - Ana Martínez
- Centro de Investigaciones Biológicas, CIB-CSIC, Calle Ramiro de Maeztu 9, E-28040, Madrid, Spain
| | - Anna Williams
- MRC-Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive, EH164UU, Edinburgh, UK
| | - Fernando de Castro
- Grupo de Neurobiología del Desarrollo-GNDe, Hospital Nacional de Parapléjicos, Finca la Peraleda s/n, E- 45071, Toledo, Spain
- Instituto Cajal-CSIC, Avda. Dr. Arce 37, E-28002, Madrid, Spain
| |
Collapse
|
33
|
Schneider S, Gruart A, Grade S, Zhang Y, Kröger S, Kirchhoff F, Eichele G, Delgado García JM, Dimou L. Decrease in newly generated oligodendrocytes leads to motor dysfunctions and changed myelin structures that can be rescued by transplanted cells. Glia 2016; 64:2201-2218. [PMID: 27615452 DOI: 10.1002/glia.23055] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 08/11/2016] [Accepted: 08/22/2016] [Indexed: 11/09/2022]
Abstract
NG2-glia in the adult brain are known to proliferate and differentiate into mature and myelinating oligodendrocytes throughout lifetime. However, the role of these newly generated oligodendrocytes in the adult brain still remains little understood. Here we took advantage of the Sox10-iCreERT2 x CAG-eGFP x Esco2fl/fl mouse line in which we can specifically ablate proliferating NG2-glia in adult animals. Surprisingly, we observed that the generation of new oligodendrocytes in the adult brain was severely affected, although the number of NG2-glia remained stable due to the enhanced proliferation of non-recombined cells. This lack of oligodendrogenesis led to the elongation of the nodes of Ranvier as well as the associated paranodes, which could be locally rescued by myelinating oligodendrocytes differentiated from transplanted NG2-glia deriving from wildtype mice. Repetitive measurements of conduction velocity in the corpus callosum of awake animals revealed a progressive deceleration specifically in the mice lacking adult oligodendrogenesis that resulted in progressive motor deficits. In summary, here we demonstrated for the first time that axon function is not only controlled by the reliable organization of myelin, but also requires a dynamic and continuous generation of new oligodendrocytes in the adult brain. GLIA 2016;64:2201-2218.
Collapse
Affiliation(s)
- Sarah Schneider
- Physiological Genomics, Biomedical Center, Ludwig-Maximilians University, Munich, Germany.,Institute of Stem Cell Research, Helmholtz Zentrum, Neuherberg, Germany.,Graduate School of Systemic Neurosciences, Ludwig-Maximilians University, Munich, Germany
| | - Agnès Gruart
- División de Neurosciencias, Universidad Pablo de Olavide, Seville, Spain
| | - Sofia Grade
- Physiological Genomics, Biomedical Center, Ludwig-Maximilians University, Munich, Germany.,Institute of Stem Cell Research, Helmholtz Zentrum, Neuherberg, Germany
| | - Yina Zhang
- Physiological Genomics, Biomedical Center, Ludwig-Maximilians University, Munich, Germany
| | - Stephan Kröger
- Physiological Genomics, Biomedical Center, Ludwig-Maximilians University, Munich, Germany
| | - Frank Kirchhoff
- Molecular Physiology, Center of Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Homburg, Germany
| | - Gregor Eichele
- Department of Genes and Behavior, MPI for Biophysical Chemistry, Göttingen, Germany
| | | | - Leda Dimou
- Physiological Genomics, Biomedical Center, Ludwig-Maximilians University, Munich, Germany. .,Institute of Stem Cell Research, Helmholtz Zentrum, Neuherberg, Germany. .,Graduate School of Systemic Neurosciences, Ludwig-Maximilians University, Munich, Germany. .,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| |
Collapse
|
34
|
Azim K, Berninger B, Raineteau O. Mosaic Subventricular Origins of Forebrain Oligodendrogenesis. Front Neurosci 2016; 10:107. [PMID: 27047329 PMCID: PMC4805584 DOI: 10.3389/fnins.2016.00107] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 03/05/2016] [Indexed: 12/30/2022] Open
Abstract
In the perinatal as well as the adult CNS, the subventricular zone (SVZ) of the forebrain is the largest and most active source of neural stem cells (NSCs) that generates neurons and oligodendrocytes (OLs), the myelin forming cells of the CNS. Recent advances in the field are beginning to shed light regarding SVZ heterogeneity, with the existence of spatially segregated microdomains that are intrinsically biased to generate phenotypically distinct neuronal populations. Although most research has focused on this regionalization in the context of neurogenesis, newer findings underline that this also applies for the genesis of OLs under the control of specific patterning molecules. In this mini review, we discuss the origins as well as the mechanisms that induce and maintain SVZ regionalization. These come in the flavor of specific signaling ligands and subsequent initiation of transcriptional networks that provide a basis for subdividing the SVZ into distinct lineage-specific microdomains. We further emphasize canonical Wnts and FGF2 as essential signaling pathways for the regional genesis of OL progenitors from NSCs of the dorsal SVZ. This aspect of NSC biology, which has so far received little attention, may unveil new avenues for appropriately recruiting NSCs in demyelinating diseases.
Collapse
Affiliation(s)
- Kasum Azim
- Focus Translational Neuroscience, Institute of Physiological Chemistry, University of Mainz Mainz, Germany
| | - Benedikt Berninger
- Focus Translational Neuroscience, Institute of Physiological Chemistry, University of Mainz Mainz, Germany
| | - Olivier Raineteau
- Inserm U1208, Stem Cell and Brain Research Institute, Université Lyon 1 Bron, France
| |
Collapse
|
35
|
Díaz-Balzac CA, Lázaro-Peña MI, Ramos-Ortiz GA, Bülow HE. The Adhesion Molecule KAL-1/anosmin-1 Regulates Neurite Branching through a SAX-7/L1CAM-EGL-15/FGFR Receptor Complex. Cell Rep 2015; 11:1377-84. [PMID: 26004184 PMCID: PMC4464948 DOI: 10.1016/j.celrep.2015.04.057] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Revised: 03/20/2015] [Accepted: 04/27/2015] [Indexed: 01/10/2023] Open
Abstract
Neurite branching is essential for correct assembly of neural circuits, yet it remains a poorly understood process. For example, the neural cell adhesion molecule KAL-1/anosmin-1, which is mutated in Kallmann syndrome, regulates neurite branching through mechanisms largely unknown. Here, we show that KAL-1/anosmin-1 mediates neurite branching as an autocrine co-factor with EGL-17/FGF through a receptor complex consisting of the conserved cell adhesion molecule SAX-7/L1CAM and the fibroblast growth factor receptor EGL-15/FGFR. This protein complex, which appears conserved in humans, requires the immunoglobulin (Ig) domains of SAX-7/L1CAM and the FN(III) domains of KAL-1/anosmin-1 for formation in vitro as well as function in vivo. The kinase domain of the EGL-15/FGFR is required for branching, and genetic evidence suggests that ras-mediated signaling downstream of EGL-15/FGFR is necessary to effect branching. Our studies establish a molecular pathway that regulates neurite branching during development of the nervous system.
Collapse
Affiliation(s)
- Carlos A Díaz-Balzac
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - María I Lázaro-Peña
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Gibram A Ramos-Ortiz
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Biology, University of Puerto Rico-Río Piedras, San Juan 00931, Puerto Rico
| | - Hannes E Bülow
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| |
Collapse
|