94 results on '"Cohen Salmon M"'
Search Results
2. Same-eye comparison of pupillary dilation with an intraoperative standardized intracameral combination of mydriatics (Mydrane®) versus a preoperative ophthalmic (Mydriasert®) in standard cataract surgery in non-diabetic patients
- Author
-
de Faria, A., Giorgi, R., Cohen Salmon, M., Bonnel, S., Holweck, G., Le Corre, A., and Chazalon, E.
- Published
- 2019
- Full Text
- View/download PDF
3. Reactive astrocyte nomenclature, definitions, and future directions
- Author
-
Escartin, C, Galea, Elena, Lakatos, A., O’Callaghan, J.P., Petzold, G.C., Serrano-Pozo, Alberto, Steinhäuser, C., Volterra, A., Carmignoto, G., Agarwal, A., Allen, N.J, Araque, Alfonso, Barbeito, Luis, Barzilai, Ari, Bergles, D. E., Bonvento, Gilles, Butt, A.M., Chen, W.T., Cohen-Salmon, M., Cunningham, C., Deneen, B., De Strooper, B., Díaz-Castro, B., Farina, C., Freeman, Mark, Gallo, V., Goldman, J.E., Goldman, S.A., Götz, Magdalena, Gutiérrez, A., Haydon, P. G., Heiland, D.H., Hol, E.M., Holt, M.G., Lino, M., Kastanenka, K.V., Kettenmann, H., Khakh, B.S., Koizumi, S., Lee, C.J., Liddelow, S.A., MacVicar, B.A., Magistretti, P., Messing, A., Mishra, A., Molofsky, A.V., Murai, K.K., Norris, C.M., and Okada, S.
- Abstract
Reactive astrocytes are astrocytes undergoing morphological, molecular, and functional remodeling in response to injury, disease, or infection of the CNS. Although this remodeling was first described over a century ago, uncertainties and controversies remain regarding the contribution of reactive astrocytes to CNS diseases, repair, and aging. It is also unclear whether fixed categories of reactive astrocytes exist and, if so, how to identify them. We point out the shortcomings of binary divisions of reactive astrocytes into good-vs-bad, neurotoxic-vs-neuroprotective or A1-vs-A2. We advocate, instead, that research on reactive astrocytes include assessment of multiple molecular and functional parameters—preferably in vivo—plus multivariate statistics and determination of impact on pathological hallmarks in relevant models. These guidelines may spur the discovery of astrocyte-based biomarkers as well as astrocyte-targeting therapies that abrogate detrimental actions of reactive astrocytes, potentiate their neuro- and glioprotective actions, and restore or augment their homeostatic, modulatory, and defensive functions. Funding: CNRS, CEA, ANR, and France Alzheimer to CE.; MCINN (PID2019-107633RB-I00) and Generalitat de Catalunya (2017-SGR547, Grup de demències Sant Pau) to E.G. US Centers for Disease Control and Prevention to J. P.O. Alzheimer’s Association (AACF-17-524184) and NIH-NIA (K08AG064039) to A.S.-P. DFG (SPP1757, STE 552/5, STE 552/4), EU (H2020-MSCA-ITN project 722053 EU-GliaPhD) and BMBF (16GW0182 CONNEXIN) to C.S. Swiss National Science Foundation grant 31003A 173124/1; SNSF NCCR ‘Transcure’ (51NF40-160620); Synapsis Foundation Heidi Seiler-Stiftung 2018-PI01 to A.Volterra. NIH-NINDS (NS084030), Dr. Miriam and Sheldon G. Adelson Medical Foundation and Wings for Life to M.V.S. The authors thank T. Yohannan of Alpha Language Services, Barcelona, for expert copy editing.
- Published
- 2021
4. Nocardia nova sphenoid sinusitis and infratemporal fossa abscess
- Author
-
Giordano, A., Cohen-Salmon, M., Joly, B., and Maffiolo, C.
- Published
- 2016
- Full Text
- View/download PDF
5. Sinusite sphénoïdale et abcès de la fosse infratemporale par Nocardia nova
- Author
-
Giordano, A., Cohen-Salmon, M., Joly, B., and Maffiolo, C.
- Published
- 2016
- Full Text
- View/download PDF
6. Immune Quiescence of the Brain Is Set by Astroglial Connexin 43
- Author
-
Boulay, A.-C., primary, Mazeraud, A., additional, Cisternino, S., additional, Saubamea, B., additional, Mailly, P., additional, Jourdren, L., additional, Blugeon, C., additional, Mignon, V., additional, Smirnova, M., additional, Cavallo, A., additional, Ezan, P., additional, Ave, P., additional, Dingli, F., additional, Loew, D., additional, Vieira, P., additional, Chretien, F., additional, and Cohen-Salmon, M., additional
- Published
- 2015
- Full Text
- View/download PDF
7. Molecular approach to the pathogenesis of renal anomalies in Kallmann's syndrome and in the branchio-oto-renal syndrome
- Author
-
Hardelin, J. P., nadia Soussi-Yanicostas, Levilliers, J., Kalatzis, V., Abdelhak, S., Cohen-Salmon, M., and Petit, C.
- Subjects
Intracellular Signaling Peptides and Proteins ,Trans-Activators ,Humans ,Nuclear Proteins ,Kallmann Syndrome ,Protein Tyrosine Phosphatases ,Kidney ,Branchio-Oto-Renal Syndrome - Published
- 1999
8. Hearing Is Normal without Connexin30
- Author
-
Boulay, A.-C., primary, del Castillo, F. J., additional, Giraudet, F., additional, Hamard, G., additional, Giaume, C., additional, Petit, C., additional, Avan, P., additional, and Cohen-Salmon, M., additional
- Published
- 2013
- Full Text
- View/download PDF
9. Isolation of kidney complementary DNAs down-expressed in Wilms' tumor by a subtractive hybridization approach
- Author
-
Austruy E, Cohen-Salmon M, Corinne ANTIGNAC, Béroud C, Henry I, Vc, Nguyen, Brugières L, Junien C, and Jeanpierre C
- Subjects
Adult ,Base Sequence ,Molecular Sequence Data ,Down-Regulation ,Nucleic Acid Hybridization ,DNA, Neoplasm ,Sequence Analysis, DNA ,Kidney ,Polymerase Chain Reaction ,Wilms Tumor ,Kidney Neoplasms ,Fetus ,Humans ,RNA, Messenger ,RNA, Neoplasm ,Chromosome Deletion - Abstract
We applied a subtractive hybridization approach to isolate genes differentially expressed between mature kidney and Wilms' tumor. We constructed a complementary DNA library from a total mature kidney complementary DNA subtracted by an excess of mRNA from a Wilms' tumor, WAGR4, with a germline deletion of 11p13 and a somatic loss of alleles at 11p15. Six clones presenting a differential pattern of expression, positive with mRNA from the mature kidney and negative with mRNA from the Wilms' tumor WAGR4, were characterized. Among these clones were two as yet unknown expressed sequences (D11S877E and D15S109E) and four sequences from known genes: renal dipeptidase (DPEP1), alpha B-crystallin (CRYA2), uromodulin (UMOD), and plasma glutathione peroxidase (GPX2). The different patterns of expression of these genes in 11 Wilms' tumors, whether or not they are hereditary, reflect the well-documented pathogenetic heterogeneity for Wilms' tumors. We propose that these clones could be helpful for an improved histological characterization of Wilms' tumors.
- Published
- 1993
10. Chromosomal assignment of the uromodulin gene (UMOD) to 16p13.11
- Author
-
Jeanpierre, C., primary, Whitmore, S.A., additional, Austruy, E., additional, Cohen-Salmon, M., additional, Callen, D.F., additional, and Junien, C., additional
- Published
- 1993
- Full Text
- View/download PDF
11. X chromosome-linked Kallmann syndrome: stop mutations validate the candidate gene.
- Author
-
Hardelin, J P, primary, Levilliers, J, additional, del Castillo, I, additional, Cohen-Salmon, M, additional, Legouis, R, additional, Blanchard, S, additional, Compain, S, additional, Bouloux, P, additional, Kirk, J, additional, and Moraine, C, additional
- Published
- 1992
- Full Text
- View/download PDF
12. Spatiotemporal expression of otogelin in the developing and adult mouse inner ear
- Author
-
El-Amraoui, A., Cohen-Salmon, M., Petit, C., and Simmler, M. C.
- Published
- 2001
- Full Text
- View/download PDF
13. Chromosomal assignment of the uromodulin gene (UMOD) to 16p13.11.
- Author
-
Jeanpierre, C., Whitmore, S.A., Austruy, E., Cohen-Salmon, M., Callen, D.F., and Junien, C.
- Published
- 1993
- Full Text
- View/download PDF
14. BRAIN VASCULAR PRESSURE INCREASE OPENS THE BLOOD-BRAIN BARRIER IN ABSENCE OF ASTROGLIAL CONNEXINS
- Author
-
Cohen-Salmon, M., Ezan, P., Andre, P., Cisternino, S., bruno saubamea, and Giaume, C.
15. Characterization of the promoter of the human KAL gene, responsible for the X-chromosome-linked Kallmann syndrome
- Author
-
Cohen-Salmon, M., Tronche, F., Castillo, I. Del, and Petit, C.
- Published
- 1995
- Full Text
- View/download PDF
16. Isolation and characterization of the gene responsible for the X chromosome-linked Kallmann Syndrome
- Author
-
Legouis, R., Cohen-Salmon, M., Castillo, I. Del, and Petit, C.
- Published
- 1994
- Full Text
- View/download PDF
17. Vascular dysfunction is at the onset of oxaliplatin-induced peripheral neuropathy symptoms in mice.
- Author
-
Taïb S, Durand J, Dehais V, Boulay AC, Martin S, Blugeon C, Jourdren L, Freydier R, Cohen-Salmon M, Hazan J, and Brunet I
- Subjects
- Animals, Mice, Male, Disease Models, Animal, Antineoplastic Agents adverse effects, Mice, Inbred C57BL, Brain metabolism, Brain drug effects, Oxaliplatin adverse effects, Peripheral Nervous System Diseases chemically induced, Peripheral Nervous System Diseases metabolism, Peripheral Nervous System Diseases physiopathology, Sciatic Nerve drug effects, Sciatic Nerve physiopathology
- Abstract
Oxaliplatin-induced peripheral neuropathy (OIPN) is an adverse side effect of this chemotherapy used for gastrointestinal cancers. The continuous pain experienced by OIPN patients often results in the reduction or discontinuation of chemotherapy, thereby affecting patient survival. Several pathogenic mechanisms involving sensory neurons were shown to participate in the occurrence of OIPN symptoms. However, the dysfunction of the blood-nerve barrier as a source of nerve alteration had not been thoroughly explored. To characterise the vascular contribution to OIPN symptoms, we undertook two comparative transcriptomic analyses of mouse purified brain and sciatic nerve blood vessels (BVs) and nerve BVs after oxaliplatin or control administration. These analyses reveal distinct molecular landscapes between brain and nerve BVs and the up-regulation of transcripts involved in vascular contraction after oxaliplatin treatment. Anatomical examination of the nerve yet shows the preservation of BV architecture in the acute OIPN mouse model, although treated mice exhibit both neuropathic symptoms and enhanced vasoconstriction reflected by hypoxia. Moreover, vasodilators significantly reduce oxaliplatin-induced neuropathic symptoms and endoneurial hypoxia, establishing the key involvement of nerve blood flow in OIPN., (© 2024 Taïb et al.)
- Published
- 2024
- Full Text
- View/download PDF
18. Dynamic local mRNA localization and translation occurs during the postnatal molecular maturation of perivascular astrocytic processes.
- Author
-
Avila-Gutierrez K, Slaoui L, Alvear-Perez R, Kozlowski E, Oudart M, Augustin E, Claveau C, Mailly P, Monnet H, Mignon V, Saubaméa B, Boulay AC, and Cohen-Salmon M
- Subjects
- Mice, Animals, RNA, Messenger metabolism, Astrocytes metabolism
- Abstract
Astrocytes are highly ramified and send out perivascular processes (PvAPs) that entirely sheathe the brain's blood vessels. PvAPs are equipped with an enriched molecular repertoire that sustains astrocytic regulatory functions at the vascular interface. In the mouse, PvAP development starts after birth and is essentially complete by postnatal day (P) 15. Progressive molecular maturation also occurs over this period, with the acquisition of proteins enriched in PvAPs. The mechanisms controlling the development and molecular maturation of PvAPs have not been extensively characterized. We reported previously that mRNAs are distributed unequally in mature PvAPs and are locally translated. Since dynamic mRNA localization and local translation influence the cell's polarity, we hypothesized that they might sustain the postnatal maturation of PvAPs. Here, we used a combination of molecular biology and imaging approaches to demonstrate that the development of PvAPs is accompanied by the transport of mRNA and polysomal mRNA into PvAPs, the development of a rough endoplasmic reticulum (RER) network and Golgi cisternae, and local translation. By focusing on genes and proteins that are selectively or specifically expressed in astrocytes, we characterized the developmental profile of mRNAs, polysomal mRNAs and proteins in PvAPs from P5 to P60. We found that some polysomal mRNAs polarized progressively towards the PvAPs. Lastly, we found that expression and localization of mRNAs in developing PvAPs is perturbed in a mouse model of megalencephalic leukoencephalopathy with subcortical cysts. Our results indicate that dynamic mRNA localization and local translation influence the postnatal maturation of PvAPs., (© 2024 The Authors. GLIA published by Wiley Periodicals LLC.)
- Published
- 2024
- Full Text
- View/download PDF
19. Evaluation of gliovascular functions of AQP4 readthrough isoforms.
- Author
-
Mueller SM, McFarland White K, Fass SB, Chen S, Shi Z, Ge X, Engelbach JA, Gaines SH, Bice AR, Vasek MJ, Garbow JR, Culver JP, Martinez-Lozada Z, Cohen-Salmon M, Dougherty JD, and Sapkota D
- Abstract
Aquaporin-4 (AQP4) is a water channel protein that links the astrocytic endfeet to the blood-brain barrier (BBB) and regulates water and potassium homeostasis in the brain, as well as the glymphatic clearance of waste products that would otherwise potentiate neurological diseases. Recently, translational readthrough was shown to generate a C-terminally extended variant of AQP4, known as AQP4x, which preferentially localizes around the BBB through interaction with the scaffolding protein α-syntrophin, and loss of AQP4x disrupts waste clearance from the brain. To investigate the function of AQP4x, we generated a novel AQP4 mouse line (AllX) to increase relative levels of the readthrough variant above the ~15% of AQP4 in the brain of wild-type (WT) mice. We validated the line and assessed characteristics that are affected by the presence of AQP4x, including AQP4 and α-syntrophin localization, integrity of the BBB, and neurovascular coupling. We compared AllX
Hom and AllXHet mice to WT and to previously characterized AQP4 NoXHet and NoXHom mice, which cannot produce AQP4x. An increased dose of AQP4x enhanced perivascular localization of α-syntrophin and AQP4, while total protein expression of the two was unchanged. However, at 100% readthrough, AQP4x localization and the formation of higher order complexes were disrupted. Electron microscopy showed that overall blood vessel morphology was unchanged except for an increased proportion of endothelial cells with budding vesicles in NoXHom mice, which may correspond to a leakier BBB or altered efflux that was identified in NoX mice using MRI. These data demonstrate that AQP4x plays a small but measurable role in maintaining BBB integrity as well as recruiting structural and functional support proteins to the blood vessel. This also establishes a new set of genetic tools for quantitatively modulating AQP4x levels., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 Mueller, McFarland White, Fass, Chen, Shi, Ge, Engelbach, Gaines, Bice, Vasek, Garbow, Culver, Martinez-Lozada, Cohen-Salmon, Dougherty and Sapkota.)- Published
- 2023
- Full Text
- View/download PDF
20. The ribosome-associated protein RACK1 represses Kir4.1 translation in astrocytes and influences neuronal activity.
- Author
-
Oudart M, Avila-Gutierrez K, Moch C, Dossi E, Milior G, Boulay AC, Gaudey M, Moulard J, Lombard B, Loew D, Bemelmans AP, Rouach N, Chapat C, and Cohen-Salmon M
- Subjects
- Animals, Mice, Mice, Knockout, Neurons, Receptors for Activated C Kinase metabolism, Ribosomes, Astrocytes metabolism, Neuroglia metabolism
- Abstract
The regulation of translation in astrocytes, the main glial cells in the brain, remains poorly characterized. We developed a high-throughput proteomics screen for polysome-associated proteins in astrocytes and focused on ribosomal protein receptor of activated protein C kinase 1 (RACK1), a critical factor in translational regulation. In astrocyte somata and perisynaptic astrocytic processes (PAPs), RACK1 preferentially binds to a number of mRNAs, including Kcnj10, encoding the inward-rectifying potassium (K
+ ) channel Kir4.1. By developing an astrocyte-specific, conditional RACK1 knockout mouse model, we show that RACK1 represses production of Kir4.1 in hippocampal astrocytes and PAPs. Upregulation of Kir4.1 in the absence of RACK1 increases astrocytic Kir4.1-mediated K+ currents and volume. It also modifies neuronal activity attenuating burst frequency and duration. Reporter-based assays reveal that RACK1 controls Kcnj10 translation through the transcript's 5' untranslated region. Hence, translational regulation by RACK1 in astrocytes represses Kir4.1 expression and influences neuronal activity., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2023 The Author(s). Published by Elsevier Inc. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
21. Physiopathological changes of ferritin mRNA density and distribution in hippocampal astrocytes in the mouse brain.
- Author
-
Tortuyaux R, Avila-Gutierrez K, Oudart M, Mazaré N, Mailly P, Deschemin JC, Vaulont S, Escartin C, and Cohen-Salmon M
- Subjects
- Mice, Animals, Hepcidins, Astrocytes metabolism, Amyloid beta-Peptides, RNA, Messenger, Iron metabolism, Mice, Knockout, Hippocampus metabolism, Ferritins genetics, Ferritins metabolism, Alzheimer Disease pathology
- Abstract
Astrocytes are thought to play a crucial role in brain iron homeostasis. How they accomplish this regulation in vivo is unclear. In a recent transcriptomic analysis, we showed that polysomal Ftl1 and Fth1 mRNAs, encoding the ferritin light (Ftl) and heavy (Fth) chains that assemble into ferritin, a critical complex for iron storage and reduction, are enriched in perisynaptic astrocytic processes as compared to astrocytic soma. These data suggested that ferritin translation plays a specific role at the perisynaptic astrocytic interface and is tighly regulated by local translation. Here, we used our recently described AstroDot 3D in situ methodology to study the density and localization of ferritin mRNAs in astrocytes in the hippocampus in three different contexts in which local or systemic iron overload has been documented: aging, the hepcidin knock-out mouse model of hemochromatosis and the APP/PS1dE9 mouse model of Alzheimer's disease (AD). Our results showed that in wild type mice, Fth1 mRNA density was higher than Ftl1 and that both mRNAs were mostly distributed in astrocyte fine processes. Aging and absence of hepcidin caused an increased Fth1/Ftl1 ratio in astrocytes and in the case of aging, led to a redistribution of Fth1 mRNAs in astrocytic fine processes. In contrast, in AD mice, we observed a lower Fth1/Ftl1 ratio. Fth1 mRNAs became more somatic and Ftl1 mRNAs redistributed in large processes of astrocytes proximal to Amyloid beta (Aß) deposits. Hence, we propose that regulation of ferritin mRNA density and distribution in astrocytes contribute to iron homeostasis in physiology and pathophysiology., (© 2022 The Authors. Journal of Neurochemistry published by John Wiley & Sons Ltd on behalf of International Society for Neurochemistry.)
- Published
- 2023
- Full Text
- View/download PDF
22. In mice and humans, brain microvascular contractility matures postnatally.
- Author
-
Slaoui L, Gilbert A, Rancillac A, Delaunay-Piednoir B, Chagnot A, Gerard Q, Letort G, Mailly P, Robil N, Gelot A, Lefebvre M, Favier M, Dias K, Jourdren L, Federici L, Auvity S, Cisternino S, Vivien D, Cohen-Salmon M, and Boulay AC
- Subjects
- Humans, Mice, Animals, Brain blood supply, Muscle Contraction, Muscle, Smooth, Vascular physiology, Endothelial Cells
- Abstract
Although great efforts to characterize the embryonic phase of brain microvascular system development have been made, its postnatal maturation has barely been described. Here, we compared the molecular and functional properties of brain vascular cells on postnatal day (P)5 vs. P15, via a transcriptomic analysis of purified mouse cortical microvessels (MVs) and the identification of vascular-cell-type-specific or -preferentially expressed transcripts. We found that endothelial cells (EC), vascular smooth muscle cells (VSMC) and fibroblasts (FB) follow specific molecular maturation programs over this time period. Focusing on VSMCs, we showed that the arteriolar VSMC network expands and becomes contractile resulting in a greater cerebral blood flow (CBF), with heterogenous developmental trajectories within cortical regions. Samples of the human brain cortex showed the same postnatal maturation process. Thus, the postnatal phase is a critical period during which arteriolar VSMC contractility required for vessel tone and brain perfusion is acquired and mature., (© 2022. The Author(s), under exclusive licence to Springer-Verlag GmbH Germany, part of Springer Nature.)
- Published
- 2023
- Full Text
- View/download PDF
23. Pannexin 1 activity in astroglia sets hippocampal neuronal network patterns.
- Author
-
Vasile F, Dossi E, Moulard J, Ezan P, Lecoin L, Cohen-Salmon M, Mailly P, Le Bert M, Couillin I, Bemelmans A, and Rouach N
- Subjects
- Animals, Mice, Connexins metabolism, Astrocytes metabolism, Disease Models, Animal
- Abstract
Astroglial release of molecules is thought to actively modulate neuronal activity, but the nature, release pathway, and cellular targets of these neuroactive molecules are still unclear. Pannexin 1, expressed by neurons and astrocytes, form nonselective large pore channels that mediate extracellular exchange of molecules. The functional relevance of these channels has been mostly studied in brain tissues, without considering their specific role in different cell types, or in neurons. Thus, our knowledge of astroglial pannexin 1 regulation and its control of neuronal activity remains very limited, largely due to the lack of tools targeting these channels in a cell-specific way. We here show that astroglial pannexin 1 expression in mice is developmentally regulated and that its activation is activity-dependent. Using astrocyte-specific molecular tools, we found that astroglial-specific pannexin 1 channel activation, in contrast to pannexin 1 activation in all cell types, selectively and negatively regulates hippocampal networks, with their disruption inducing a drastic switch from bursts to paroxysmal activity. This decrease in neuronal excitability occurs via an unconventional astroglial mechanism whereby pannexin 1 channel activity drives purinergic signaling-mediated regulation of hyperpolarisation-activated cyclic nucleotide (HCN)-gated channels. Our findings suggest that astroglial pannexin 1 channel activation serves as a negative feedback mechanism crucial for the inhibition of hippocampal neuronal networks., Competing Interests: The authors have declared that no competing interests exist., (Copyright: © 2022 Vasile et al. This is an open access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.)
- Published
- 2022
- Full Text
- View/download PDF
24. PAI-1 production by reactive astrocytes drives tissue dysfibrinolysis in multiple sclerosis models.
- Author
-
Lebas H, Guérit S, Picot A, Boulay AC, Fournier A, Vivien D, Cohen Salmon M, Docagne F, and Bardou I
- Subjects
- Animals, Astrocytes metabolism, Central Nervous System metabolism, Disease Models, Animal, Fibrin, In Situ Hybridization, Fluorescence, Mice, Mice, Knockout, Serpin E2, Encephalomyelitis, Autoimmune, Experimental genetics, Multiple Sclerosis genetics, Plasminogen Activator Inhibitor 1 genetics
- Abstract
Background: In multiple sclerosis (MS), disturbance of the plasminogen activation system (PAS) and blood brain barrier (BBB) disruption are physiopathological processes that might lead to an abnormal fibrin(ogen) extravasation into the parenchyma. Fibrin(ogen) deposits, usually degraded by the PAS, promote an autoimmune response and subsequent demyelination. However, the PAS disruption is not well understood and not fully characterized in this disorder., Methods: Here, we characterized the expression of PAS actors during different stages of two mouse models of MS (experimental autoimmune encephalomyelitis-EAE), in the central nervous system (CNS) by quantitative RT-PCR, immunohistofluorescence and fluorescent in situ hybridization (FISH). Thanks to constitutive PAI-1 knockout mice (PAI-1 KO) and an immunotherapy using a blocking PAI-1 antibody, we evaluated the role of PAI-1 in EAE models and its impact on physiopathological processes such as fibrin(ogen) deposits, lymphocyte infiltration and demyelination., Results: We report a striking overexpression of PAI-1 in reactive astrocytes during symptomatic phases, in two EAE mouse models of MS. This increase is concomitant with lymphocyte infiltration and fibrin(ogen) deposits in CNS parenchyma. By genetic invalidation of PAI-1 in mice and immunotherapy using a blocking PAI-1 antibody, we demonstrate that abolition of PAI-1 reduces the severity of EAE and occurrence of relapses in two EAE models. These benefits are correlated with a decrease in fibrin(ogen) deposits, infiltration of T4 lymphocytes, reactive astrogliosis, demyelination and axonal damage., Conclusion: These results demonstrate that a deleterious overexpression of PAI-1 by reactive astrocytes leads to intra-parenchymal dysfibrinolysis in MS models and anti-PAI-1 strategies could be a new therapeutic perspective for MS., (© 2022. The Author(s), under exclusive licence to Springer Nature Switzerland AG.)
- Published
- 2022
- Full Text
- View/download PDF
25. A Retro-orbital Sinus Injection Mouse Model to Study Early Events and Reorganization of the Astrocytic Network during Pneumococcal Meningitis.
- Author
-
Bello C, Cohen-Salmon M, and Van Nhieu GT
- Abstract
Pneumococcal (PN) meningitis is a life-threatening disease with high mortality rates that leads to permanent neurological sequelae. Studies of the process of bacterial crossing of the blood brain barrier (BBB) are hampered by the lack of relevant in vitro and in vivo models of meningitis that recapitulate the human disease. PN meningitis involves bacterial access to the bloodstream preceding translocation across the BBB. A large number of PN meningitis models have been developed in mice, with intravenous administration via the lateral tail vein representing the main way to study BBB crossing by PN. While in humans, meningitis is not always associated with bacteremia, PN meningitis after intravenous injection in mice usually develops following sustained and very high bacteremic titers. High grade bacteremia, however, is known to favor inflammation and BBB permeabilization, thereby increasing PN translocation across the BBB and associated damages. Therefore, specific processes associated with early events of PN translocation may be blurred by overall changes in the inflammatory environment and potentially systemic dysfunction in the case of severe sepsis. Here, we report a mouse meningitis model induced by PN injection in the retro-orbital (RO) sinus. We show that, in this model, mice appear to control bacteremic levels during the first 13 h post-infection, while PN crossing of the BBB can be clearly detected by fluorescence confocal microscopy analysis of brain slices as early as 6 h post-infection. Because of the low frequency of events, however, PN translocation across brain parenchymal vessels at early time points requires a rigorous and systematic examination of the brain volume., Competing Interests: Competing interestsThe authors declare no competing interests., (Copyright © 2021 The Authors; exclusive licensee Bio-protocol LLC.)
- Published
- 2021
- Full Text
- View/download PDF
26. Megalencephalic leukoencephalopathy with subcortical cysts is a developmental disorder of the gliovascular unit.
- Author
-
Gilbert A, Elorza-Vidal X, Rancillac A, Chagnot A, Yetim M, Hingot V, Deffieux T, Boulay AC, Alvear-Perez R, Cisternino S, Martin S, Taïb S, Gelot A, Mignon V, Favier M, Brunet I, Declèves X, Tanter M, Estevez R, Vivien D, Saubaméa B, and Cohen-Salmon M
- Subjects
- Animals, Cell Adhesion Molecules, Neuron-Glia metabolism, Disease Models, Animal, Membrane Proteins metabolism, Mice, Mice, Knockout, Nerve Tissue Proteins metabolism, Cell Adhesion Molecules, Neuron-Glia genetics, Cysts genetics, Hereditary Central Nervous System Demyelinating Diseases genetics, Membrane Proteins genetics, Nerve Tissue Proteins genetics
- Abstract
Absence of the astrocyte-specific membrane protein MLC1 is responsible for megalencephalic leukoencephalopathy with subcortical cysts (MLC), a rare type of leukodystrophy characterized by early-onset macrocephaly and progressive white matter vacuolation that lead to ataxia, spasticity, and cognitive decline. During postnatal development (from P5 to P15 in the mouse), MLC1 forms a membrane complex with GlialCAM (another astrocytic transmembrane protein) at the junctions between perivascular astrocytic processes. Perivascular astrocytic processes along with blood vessels form the gliovascular unit. It was not previously known how MLC1 influences the physiology of the gliovascular unit. Here, using the Mlc1 knock-out mouse model of MLC, we demonstrated that MLC1 controls the postnatal development and organization of perivascular astrocytic processes, vascular smooth muscle cell contractility, neurovascular coupling, and intraparenchymal interstitial fluid clearance. Our data suggest that MLC is a developmental disorder of the gliovascular unit, and perivascular astrocytic processes and vascular smooth muscle cell maturation defects are primary events in the pathogenesis of MLC and therapeutic targets for this disease., Competing Interests: AG, XE, AR, AC, MY, VH, TD, AB, RA, SC, SM, ST, AG, VM, MF, IB, XD, MT, RE, DV, BS, MC No competing interests declared, (© 2021, Gilbert et al.)
- Published
- 2021
- Full Text
- View/download PDF
27. Astroglial Cx30 differentially impacts synaptic activity from hippocampal principal cells and interneurons.
- Author
-
Hardy E, Cohen-Salmon M, Rouach N, and Rancillac A
- Subjects
- Animals, Connexin 30 genetics, Connexin 30 metabolism, Interneurons metabolism, Mice, Synapses metabolism, Synaptic Transmission physiology, Astrocytes metabolism, Hippocampus cytology, Hippocampus metabolism
- Abstract
Astrocytes play important roles in brain function via dynamic structural and functional interactions with neurons. Yet the underlying mechanisms remain poorly defined. A typical feature of astrocytes is the high expression of connexins, which mediate their extensive intercellular communication and regulate their structural properties. In particular, connexin 30 (Cx30), one of the two connexins abundantly expressed by astrocytes, was recently shown to be a critical regulator of excitatory synaptic transmission by controlling the astroglial coverage of synapses. However, the role of Cx30 in the regulation of inhibitory synaptic transmission and excitatory/inhibitory balance remains elusive. Here, we investigated the role of astroglial Cx30 on the electrophysiological and morphological properties of five classes of hippocampal CA1 stratum oriens and pyramidale neurons, defined by the unsupervised Ward's clustering. Using Cx30 knockout mice, we found that Cx30 alters specific properties of some subsets of CA1 interneurons, such as resting membrane potential and sag ratio, while other parameters, such as action potential threshold and saturation frequency, were more frequently altered among the different classes of neurons. The excitation-inhibition balance was also differentially and selectively modulated among the different neuron subtypes. Only slight morphological differences were observed on reconstructed neurons. Altogether, these data indicate that Cx30 differentially alters the electrophysiological and morphological properties of hippocampal cell populations, and modulates both their excitatory and inhibitory inputs. Astrocytes, via Cx30, are thus active modulators of both excitatory and inhibitory synapses in the hippocampus., (© 2021 Wiley Periodicals LLC.)
- Published
- 2021
- Full Text
- View/download PDF
28. Identification of the GlialCAM interactome: the G protein-coupled receptors GPRC5B and GPR37L1 modulate megalencephalic leukoencephalopathy proteins.
- Author
-
Alonso-Gardón M, Elorza-Vidal X, Castellanos A, La Sala G, Armand-Ugon M, Gilbert A, Di Pietro C, Pla-Casillanis A, Ciruela F, Gasull X, Nunes V, Martínez A, Schulte U, Cohen-Salmon M, Marazziti D, and Estévez R
- Subjects
- Animals, Astrocytes metabolism, Brain metabolism, Cell Adhesion Molecules, Neuron-Glia genetics, Cell Adhesion Molecules, Neuron-Glia metabolism, Cell Cycle Proteins genetics, Chloride Channels genetics, Cysts metabolism, HEK293 Cells, HeLa Cells, Hereditary Central Nervous System Demyelinating Diseases metabolism, Humans, Leukoencephalopathies genetics, Leukoencephalopathies metabolism, Membrane Proteins genetics, Mice, Mice, Knockout, Mutation, Nerve Tissue Proteins genetics, Nerve Tissue Proteins metabolism, Nervous System Malformations metabolism, Protein Transport, Receptors, G-Protein-Coupled metabolism, Cysts genetics, Hereditary Central Nervous System Demyelinating Diseases genetics, Receptors, G-Protein-Coupled genetics
- Abstract
Megalencephalic Leukoencephalopathy with subcortical Cysts (MLC) is a type of vacuolating leukodystrophy, which is mainly caused by mutations in MLC1 or GLIALCAM. The two MLC-causing genes encode for membrane proteins of yet unknown function that have been linked to the regulation of different chloride channels such as the ClC-2 and VRAC. To gain insight into the role of MLC proteins, we have determined the brain GlialCAM interacting proteome. The proteome includes different transporters and ion channels known to be involved in the regulation of brain homeostasis, proteins related to adhesion or signaling as several G protein-coupled receptors (GPCRs), including the orphan GPRC5B and the proposed prosaposin receptor GPR37L1. Focusing on these two GPCRs, we could validate that they interact directly with MLC proteins. The inactivation of Gpr37l1 in mice upregulated MLC proteins without altering their localization. Conversely, a reduction of GPRC5B levels in primary astrocytes downregulated MLC proteins, leading to an impaired activation of ClC-2 and VRAC. The interaction between the GPCRs and MLC1 was dynamically regulated upon changes in the osmolarity or potassium concentration. We propose that GlialCAM and MLC1 associate with different integral membrane proteins modulating their functions and acting as a recruitment site for various signaling components as the GPCRs identified here. We hypothesized that the GlialCAM/MLC1 complex is working as an adhesion molecule coupled to a tetraspanin-like molecule performing regulatory effects through direct binding or influencing signal transduction events., (© The Author(s) 2021. Published by Oxford University Press.)
- Published
- 2021
- Full Text
- View/download PDF
29. Astrocytes in the regulation of cerebrovascular functions.
- Author
-
Cohen-Salmon M, Slaoui L, Mazaré N, Gilbert A, Oudart M, Alvear-Perez R, Elorza-Vidal X, Chever O, and Boulay AC
- Subjects
- Brain, Neuroglia, Neurons, Astrocytes, Blood-Brain Barrier
- Abstract
Astrocytes are the most numerous type of neuroglia in the brain and have a predominant influence on the cerebrovascular system; they control perivascular homeostasis, the integrity of the blood-brain barrier, the dialogue with the peripheral immune system, the transfer of metabolites from the blood, and blood vessel contractility in response to neuronal activity. These regulatory processes occur in a specialized interface composed of perivascular astrocyte extensions that almost completely cover the cerebral blood vessels. Scientists have only recently started to study how this interface is formed and how it influences cerebrovascular functions. Here, we review the literature on the astrocytes' role in the regulation of the cerebrovascular system. We cover the anatomy and development of the gliovascular interface, the known gliovascular functions, and molecular factors, the latter's implication in certain pathophysiological situations, and recent cutting-edge experimental tools developed to examine the astrocytes' role at the vascular interface. Finally, we highlight some open questions in this field of research., (© 2020 Wiley Periodicals LLC.)
- Published
- 2021
- Full Text
- View/download PDF
30. Dp71 contribution to the molecular scaffold anchoring aquaporine-4 channels in brain macroglial cells.
- Author
-
Belmaati Cherkaoui M, Vacca O, Izabelle C, Boulay AC, Boulogne C, Gillet C, Barnier JV, Rendon A, Cohen-Salmon M, and Vaillend C
- Subjects
- Animals, Aquaporin 4 genetics, Aquaporin 4 metabolism, Astrocytes metabolism, Brain metabolism, Mice, Neuroglia metabolism, Dystroglycans genetics, Dystrophin genetics
- Abstract
Intellectual disability in Duchenne muscular dystrophy has been associated with the loss of dystrophin-protein 71, Dp71, the main dystrophin-gene product in the adult brain. Dp71 shows major expression in perivascular macroglial endfeet, suggesting that dysfunctional glial mechanisms contribute to cognitive impairments. In the present study, we investigated the molecular alterations induced by a selective loss of Dp71 in mice, using semi-quantitative immunogold analyses in electron microscopy and immunofluorescence confocal analyses in brain sections and purified gliovascular units. In macroglial pericapillary endfeet of the cerebellum and hippocampus, we found a drastic reduction (70%) of the polarized distribution of aquaporin-4 (AQP4) channels, a 50% reduction of β-dystroglycan, and a complete loss of α1-syntrophin. Interestingly, in the hippocampus and cortex, these effects were not homogeneous: AQP4 and AQP4ex isoforms were mostly lost around capillaries but preserved in large vessels corresponding to pial arteries, penetrating cortical arterioles, and arterioles of the hippocampal fissure, indicating the presence of Dp71-independent pools of AQP4 in these vascular structures. In conclusion, the depletion of Dp71 strongly alters the distribution of AQP4 selectively in macroglial perivascular endfeet surrounding capillaries. This effect likely affects water homeostasis and blood-brain barrier functions and may thus contribute to the synaptic and cognitive defects associated with Dp71 deficiency., (© 2020 Wiley Periodicals LLC.)
- Published
- 2021
- Full Text
- View/download PDF
31. CC17 group B Streptococcus exploits integrins for neonatal meningitis development.
- Author
-
Deshayes de Cambronne R, Fouet A, Picart A, Bourrel AS, Anjou C, Bouvier G, Candeias C, Bouaboud A, Costa L, Boulay AC, Cohen-Salmon M, Plu I, Rambaud C, Faurobert E, Albigès-Rizo C, Tazi A, Poyart C, and Guignot J
- Subjects
- Adhesins, Bacterial genetics, Animals, Animals, Newborn, Bacterial Adhesion genetics, Blood-Brain Barrier microbiology, Cell Line, Humans, Integrin alphaVbeta3 genetics, Meningitis, Bacterial genetics, Rats, Receptors, Vitronectin genetics, Streptococcal Infections genetics, Streptococcus agalactiae genetics, Adhesins, Bacterial metabolism, Blood-Brain Barrier metabolism, Integrin alphaVbeta3 metabolism, Meningitis, Bacterial metabolism, Receptors, Vitronectin metabolism, Streptococcal Infections metabolism, Streptococcus agalactiae metabolism
- Abstract
Group B Streptococcus (GBS) is the major cause of human neonatal infections. A single clone, designated CC17-GBS, accounts for more than 80% of meningitis cases, the most severe form of the infection. However, the events allowing blood-borne GBS to penetrate the brain remain largely elusive. In this study, we identified the host transmembrane receptors α5β1 and αvβ3 integrins as the ligands of Srr2, a major CC17-GBS-specific adhesin. Two motifs located in the binding region of Srr2 were responsible for the interaction between CC17-GBS and these integrins. We demonstrated in a blood-brain-barrier cellular model that both integrins contributed to the adhesion and internalization of CC17-GBS. Strikingly, both integrins were overexpressed during the postnatal period in the brain vessels of the blood-brain barrier and blood-cerebrospinal fluid barrier and contributed to juvenile susceptibility to CC17 meningitis. Finally, blocking these integrins decreased the ability of CC17-GBS to cross into the CNS of juvenile mice in an in vivo model of meningitis. Our study demonstrated that CC17-GBS exploits integrins in order to cross the brain vessels, leading to meningitis. Importantly, it provides host molecular insights into neonate's susceptibility to CC17-GBS meningitis, thereby opening new perspectives for therapeutic and prevention strategies of GBS-elicited meningitis.
- Published
- 2021
- Full Text
- View/download PDF
32. Reactive astrocyte nomenclature, definitions, and future directions.
- Author
-
Escartin C, Galea E, Lakatos A, O'Callaghan JP, Petzold GC, Serrano-Pozo A, Steinhäuser C, Volterra A, Carmignoto G, Agarwal A, Allen NJ, Araque A, Barbeito L, Barzilai A, Bergles DE, Bonvento G, Butt AM, Chen WT, Cohen-Salmon M, Cunningham C, Deneen B, De Strooper B, Díaz-Castro B, Farina C, Freeman M, Gallo V, Goldman JE, Goldman SA, Götz M, Gutiérrez A, Haydon PG, Heiland DH, Hol EM, Holt MG, Iino M, Kastanenka KV, Kettenmann H, Khakh BS, Koizumi S, Lee CJ, Liddelow SA, MacVicar BA, Magistretti P, Messing A, Mishra A, Molofsky AV, Murai KK, Norris CM, Okada S, Oliet SHR, Oliveira JF, Panatier A, Parpura V, Pekna M, Pekny M, Pellerin L, Perea G, Pérez-Nievas BG, Pfrieger FW, Poskanzer KE, Quintana FJ, Ransohoff RM, Riquelme-Perez M, Robel S, Rose CR, Rothstein JD, Rouach N, Rowitch DH, Semyanov A, Sirko S, Sontheimer H, Swanson RA, Vitorica J, Wanner IB, Wood LB, Wu J, Zheng B, Zimmer ER, Zorec R, Sofroniew MV, and Verkhratsky A
- Subjects
- Animals, Brain Diseases pathology, Brain Injuries pathology, Humans, Spinal Cord Injuries pathology, Aging pathology, Astrocytes pathology, Brain pathology, Spinal Cord pathology
- Abstract
Reactive astrocytes are astrocytes undergoing morphological, molecular, and functional remodeling in response to injury, disease, or infection of the CNS. Although this remodeling was first described over a century ago, uncertainties and controversies remain regarding the contribution of reactive astrocytes to CNS diseases, repair, and aging. It is also unclear whether fixed categories of reactive astrocytes exist and, if so, how to identify them. We point out the shortcomings of binary divisions of reactive astrocytes into good-vs-bad, neurotoxic-vs-neuroprotective or A1-vs-A2. We advocate, instead, that research on reactive astrocytes include assessment of multiple molecular and functional parameters-preferably in vivo-plus multivariate statistics and determination of impact on pathological hallmarks in relevant models. These guidelines may spur the discovery of astrocyte-based biomarkers as well as astrocyte-targeting therapies that abrogate detrimental actions of reactive astrocytes, potentiate their neuro- and glioprotective actions, and restore or augment their homeostatic, modulatory, and defensive functions.
- Published
- 2021
- Full Text
- View/download PDF
33. Local translation in perisynaptic and perivascular astrocytic processes - a means to ensure astrocyte molecular and functional polarity?
- Author
-
Mazaré N, Oudart M, and Cohen-Salmon M
- Subjects
- Neurons, RNA, Messenger genetics, Astrocytes, Synapses
- Abstract
Together with the compartmentalization of mRNAs in distal regions of the cytoplasm, local translation constitutes a prominent and evolutionarily conserved mechanism mediating cellular polarization and the regulation of protein delivery in space and time. The translational regulation of gene expression enables a rapid response to stimuli or to a change in the environment, since the use of pre-existing mRNAs can bypass time-consuming nuclear control mechanisms. In the brain, the translation of distally localized mRNAs has been mainly studied in neurons, whose cytoplasmic protrusions may be more than 1000 times longer than the diameter of the cell body. Importantly, alterations in local translation in neurons have been implicated in several neurological diseases. Astrocytes, the most abundant glial cells in the brain, are voluminous, highly ramified cells that project long processes to neurons and brain vessels, and dynamically regulate distal synaptic and vascular functions. Recent research has demonstrated the presence of local translation at these astrocytic interfaces that might regulate the functional compartmentalization of astrocytes. In this Review, we summarize our current knowledge about the localization and local translation of mRNAs in the distal perisynaptic and perivascular processes of astrocytes, and discuss their possible contribution to the molecular and functional polarity of astrocytes., Competing Interests: Competing interestsThe authors declare no competing or financial interests., (© 2021. Published by The Company of Biologists Ltd.)
- Published
- 2021
- Full Text
- View/download PDF
34. Role of astroglial Connexin 43 in pneumolysin cytotoxicity and during pneumococcal meningitis.
- Author
-
Bello C, Smail Y, Sainte-Rose V, Podglajen I, Gilbert A, Moreira V, Chrétien F, Cohen Salmon M, and Tran Van Nhieu G
- Subjects
- Animals, Bacterial Proteins metabolism, Mice, Mice, Inbred C57BL, Streptococcus pneumoniae metabolism, Streptococcus pneumoniae pathogenicity, Virulence physiology, Virulence Factors metabolism, Astrocytes metabolism, Connexin 43 metabolism, Meningitis, Pneumococcal, Streptolysins metabolism
- Abstract
Streptococcus pneumoniae or pneumococcus (PN) is a major causative agent of bacterial meningitis with high mortality in young infants and elderly people worldwide. The mechanism underlying PN crossing of the blood brain barrier (BBB) and specifically, the role of non-endothelial cells of the neurovascular unit that control the BBB function, remains poorly understood. Here, we show that the astroglial connexin 43 (aCx43), a major gap junctional component expressed in astrocytes, plays a predominant role during PN meningitis. Following intravenous PN challenge, mice deficient for aCx43 developed milder symptoms and showed severely reduced bacterial counts in the brain. Immunofluorescence analysis of brain slices indicated that PN induces the aCx43-dependent destruction of the network of glial fibrillary acid protein (GFAP), an intermediate filament protein specifically expressed in astrocytes and up-regulated in response to brain injury. PN also induced nuclear shrinkage in astrocytes associated with the loss of BBB integrity, bacterial translocation across endothelial vessels and replication in the brain cortex. We found that aCx4-dependent astrocyte damages could be recapitulated using in vitro cultured cells upon challenge with wild-type PN but not with a ply mutant deficient for the pore-forming toxin pneumolysin (Ply). Consistently, we showed that purified Ply requires Cx43 to promote host cell plasma membrane permeabilization in a process involving the Cx43-dependent release of extracellular ATP and prolonged increase of cytosolic Ca2+ in host cells. These results point to a critical role for astrocytes during PN meningitis and suggest that the cytolytic activity of the major virulence factor Ply at concentrations relevant to bacterial infection requires co-opting of connexin plasma membrane channels., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2020
- Full Text
- View/download PDF
35. Immunoprecipitation of Ribosome-Bound mRNAs from Astrocytic Perisynaptic Processes of the Mouse Hippocampus.
- Author
-
Mazaré N, Oudart M, Cheung G, Boulay AC, and Cohen-Salmon M
- Subjects
- Animals, Astrocytes pathology, Astrocytes physiology, Biophysical Phenomena, Cell Communication, Hippocampus physiology, Mice, Phagocytosis, Ribosomes genetics, Ribosomes metabolism, Synapses physiology, Immunoprecipitation methods, RNA, Messenger isolation & purification
- Abstract
Translation of distally localized mRNAs is an evolutionary mechanism occurring in polarized cells. It has been observed in astrocytes, whose processes contact blood vessels and synapses. Here, we describe a protocol for the purification of the entire pool of ribosome-bound mRNAs in perisynaptic astrocytic processes (PAPs). Our procedure combines the preparation of synaptogliosomes with a refined translating ribosome affinity purification technique. This approach can be used in any brain region to probe the physiological relevance of local translation in PAPs. For complete details on the use and execution of this protocol, please refer to Mazaré et al. (2020)., Competing Interests: The authors declare no competing interests., (© 2020 The Author(s).)
- Published
- 2020
- Full Text
- View/download PDF
36. Mitochondrial AIF loss causes metabolic reprogramming, caspase-independent cell death blockade, embryonic lethality, and perinatal hydrocephalus.
- Author
-
Delavallée L, Mathiah N, Cabon L, Mazeraud A, Brunelle-Navas MN, Lerner LK, Tannoury M, Prola A, Moreno-Loshuertos R, Baritaud M, Vela L, Garbin K, Garnier D, Lemaire C, Langa-Vives F, Cohen-Salmon M, Fernández-Silva P, Chrétien F, Migeotte I, and Susin SA
- Subjects
- Animals, Apoptosis physiology, Caspases metabolism, Cell Respiration, Female, Fibroblasts metabolism, Genetic Engineering methods, Glycolysis genetics, Hydrocephalus metabolism, Male, Membrane Potential, Mitochondrial genetics, Membrane Potential, Mitochondrial physiology, Mice, Mice, Inbred C57BL, Mice, Inbred Strains genetics, Mitochondria metabolism, Models, Animal, Oxidative Phosphorylation, Reactive Oxygen Species metabolism, Apoptosis Inducing Factor genetics, Apoptosis Inducing Factor metabolism
- Abstract
Objectives: Apoptosis-Inducing Factor (AIF) is a protein involved in mitochondrial electron transport chain assembly/stability and programmed cell death. The relevant role of this protein is underlined because mutations altering mitochondrial AIF properties result in acute pediatric mitochondriopathies and tumor metastasis. By generating an original AIF-deficient mouse strain, this study attempted to analyze, in a single paradigm, the cellular and developmental metabolic consequences of AIF loss and the subsequent oxidative phosphorylation (OXPHOS) dysfunction., Methods: We developed a novel AIF-deficient mouse strain and assessed, using molecular and cell biology approaches, the cellular, embryonic, and adult mice phenotypic alterations. Additionally, we conducted ex vivo assays with primary and immortalized AIF knockout mouse embryonic fibroblasts (MEFs) to establish the cell death characteristics and the metabolic adaptive responses provoked by the mitochondrial electron transport chain (ETC) breakdown., Results: AIF deficiency destabilized mitochondrial ETC and provoked supercomplex disorganization, mitochondrial transmembrane potential loss, and high generation of mitochondrial reactive oxygen species (ROS). AIF
-/Y MEFs counterbalanced these OXPHOS alterations by mitochondrial network reorganization and a metabolic reprogramming toward anaerobic glycolysis illustrated by the AMPK phosphorylation at Thr172, the overexpression of the glucose transporter GLUT-4, the subsequent enhancement of glucose uptake, and the anaerobic lactate generation. A late phenotype was characterized by the activation of P53/P21-mediated senescence. Notably, approximately 2% of AIF-/Y MEFs diminished both mitochondrial mass and ROS levels and spontaneously proliferated. These cycling AIF-/Y MEFs were resistant to caspase-independent cell death inducers. The AIF-deficient mouse strain was embryonic lethal between E11.5 and E13.5 with energy loss, proliferation arrest, and increased apoptotic levels. Contrary to AIF-/Y MEFs, the AIF KO embryos were unable to reprogram their metabolism toward anaerobic glycolysis. Heterozygous AIF+/- females displayed progressive bone marrow, thymus, and spleen cellular loss. In addition, approximately 10% of AIF+/- females developed perinatal hydrocephaly characterized by brain development impairment, meningeal fibrosis, and medullar hemorrhages; those mice died 5 weeks after birth. AIF+/- with hydrocephaly exhibited loss of ciliated epithelium in the ependymal layer. This phenotype was triggered by the ROS excess. Accordingly, it was possible to diminish the occurrence of hydrocephalus AIF+/- females by supplying dams and newborns with an antioxidant in drinking water., Conclusions: In a single knockout model and at 3 different levels (cell, embryo, and adult mice) we demonstrated that by controlling the mitochondrial OXPHOS/metabolism, AIF is a key factor regulating cell differentiation and fate. Additionally, by providing new insights into the pathological consequences of mitochondrial OXPHOS dysfunction, our new findings pave the way for novel pharmacological strategies., (Copyright © 2020 The Author(s). Published by Elsevier GmbH.. All rights reserved.)- Published
- 2020
- Full Text
- View/download PDF
37. Local Translation in Perisynaptic Astrocytic Processes Is Specific and Changes after Fear Conditioning.
- Author
-
Mazaré N, Oudart M, Moulard J, Cheung G, Tortuyaux R, Mailly P, Mazaud D, Bemelmans AP, Boulay AC, Blugeon C, Jourdren L, Le Crom S, Rouach N, and Cohen-Salmon M
- Subjects
- Animals, Humans, Mice, Astrocytes metabolism, Fear psychology, Neuronal Plasticity physiology
- Abstract
Local translation is a conserved mechanism conferring cells the ability to quickly respond to local stimuli. In the brain, it has been recently reported in astrocytes, whose fine processes contact blood vessels and synapses. Yet the specificity and regulation of astrocyte local translation remain unknown. We study hippocampal perisynaptic astrocytic processes (PAPs) and show that they contain the machinery for translation. Using a refined immunoprecipitation technique, we characterize the entire pool of ribosome-bound mRNAs in PAPs and compare it with the one expressed in the whole astrocyte. We find that a specific pool of mRNAs is highly polarized at the synaptic interface. These transcripts encode an unexpected molecular repertoire, composed of proteins involved in iron homeostasis, translation, cell cycle, and cytoskeleton. Remarkably, we observe alterations in global RNA distribution and ribosome-bound status of some PAP-enriched transcripts after fear conditioning, indicating the role of astrocytic local translation in memory and learning., Competing Interests: Declaration of Interests The authors declare no conflict of interest., (Copyright © 2020 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
38. AstroDot - a new method for studying the spatial distribution of mRNA in astrocytes.
- Author
-
Oudart M, Tortuyaux R, Mailly P, Mazaré N, Boulay AC, and Cohen-Salmon M
- Subjects
- Animals, Glial Fibrillary Acidic Protein genetics, Mice, Microglia, RNA, Messenger genetics, Alzheimer Disease, Astrocytes
- Abstract
Astrocytes are morphologically complex and use local translation to regulate distal functions. To study the distribution of mRNA in astrocytes, we combined mRNA detection via in situ hybridization with immunostaining of the astrocyte-specific intermediate filament glial fibrillary acidic protein (GFAP). mRNAs at the level of GFAP-immunolabelled astrocyte somata, and large and fine processes were analysed using AstroDot, an ImageJ plug-in and the R package AstroStat. Taking the characterization of mRNAs encoding GFAP-α and GFAP-δ isoforms as a proof of concept, we showed that they mainly localized on GFAP processes. In the APPswe/PS1dE9 mouse model of Alzheimer's disease, the density and distribution of both α and δ forms of Gfap mRNA changed as a function of the region of the hippocampus and the astrocyte's proximity to amyloid plaques. To validate our method, we confirmed that the ubiquitous Rpl4 (large subunit ribosomal protein 4) mRNA was present in astrocyte processes as well as in microglia processes immunolabelled for ionized calcium binding adaptor molecule 1 (Iba1; also known as IAF1). In summary, this novel set of tools allows the characterization of mRNA distribution in astrocytes and microglia in physiological or pathological settings., Competing Interests: Competing interestsThe authors declare no competing or financial interests., (© 2020. Published by The Company of Biologists Ltd.)
- Published
- 2020
- Full Text
- View/download PDF
39. Neuronal Activity Drives Astroglial Connexin 30 in Perisynaptic Processes and Shapes Its Functions.
- Author
-
Ghézali G, Vasile F, Curry N, Fantham M, Cheung G, Ezan P, Cohen-Salmon M, Kaminski C, and Rouach N
- Subjects
- Animals, Astrocytes cytology, Female, Hippocampus cytology, Lysosomes physiology, Male, Mice, Inbred C57BL, Mice, Knockout, Proteolysis, Astrocytes physiology, Connexin 30 physiology, Hippocampus physiology, Neurons physiology
- Abstract
Astrocytes play key roles in brain functions through dynamic interactions with neurons. One of their typical features is to express high levels of connexins (Cxs), Cx43 and Cx30, the gap junction (GJ)-forming proteins. Cx30 is involved in basic cognitive processes and shapes synaptic and network activities, as shown by recent studies in transgenic animals. Yet it remains unknown whether astroglial Cx30 expression, localization, and functions are endogenously and dynamically regulated by neuronal activity and could therefore play physiological roles in neurotransmission. We here show that neuronal activity increased hippocampal Cx30 protein levels via a posttranslational mechanism regulating lysosomal degradation. Neuronal activity also increased Cx30 protein levels at membranes and perisynaptic processes, as revealed by superresolution imaging. This translated at the functional level in the activation of Cx30 hemichannels and in Cx30-mediated remodeling of astrocyte morphology independently of GJ biochemical coupling. Altogether, these data show activity-dependent dynamics of Cx30 expression, perisynaptic localization, and functions., (© The Author(s) 2019. Published by Oxford University Press. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2020
- Full Text
- View/download PDF
40. Uncoupling of the Astrocyte Syncytium Differentially Affects AQP4 Isoforms.
- Author
-
Katoozi S, Skauli N, Zahl S, Deshpande T, Ezan P, Palazzo C, Steinhäuser C, Frigeri A, Cohen-Salmon M, Ottersen OP, and Amiry-Moghaddam M
- Subjects
- Animals, Aquaporin 4 genetics, Calcium-Binding Proteins metabolism, Connexins metabolism, Gene Deletion, Hippocampus metabolism, Hippocampus ultrastructure, Membrane Proteins metabolism, Mice, Inbred C57BL, Muscle Proteins metabolism, Protein Isoforms metabolism, RNA, Messenger genetics, RNA, Messenger metabolism, Aquaporin 4 metabolism, Astrocytes metabolism, Giant Cells metabolism
- Abstract
The water channel protein aquaporin-4 (AQP4) and the gap junction forming proteins connexin-43 (Cx43) and connexin-30 (Cx30) are astrocytic proteins critically involved in brain water and ion homeostasis. While AQP4 is mainly involved in water flux across the astrocytic endfeet membranes, astrocytic gap junctions provide syncytial coupling allowing intercellular exchange of water, ions, and other molecules. We have previously shown that mice with targeted deletion of Aqp4 display enhanced gap junctional coupling between astrocytes. Here, we investigate whether uncoupling of the astrocytic syncytium by deletion of the astrocytic connexins Cx43 and Cx30 affects AQP4 membrane localization and expression. By using quantitative immunogold cytochemistry, we show that deletion of astrocytic connexins leads to a substantial reduction of perivascular AQP4, concomitant with a down-regulation of total AQP4 protein and mRNA. Isoform expression analysis shows that while the level of the predominant AQP4 M23 isoform is reduced in Cx43/Cx30 double deficient hippocampal astrocytes, the levels of M1, and the alternative translation AQP4ex isoform protein levels are increased. These findings reveal a complex interdependence between AQP4 and connexins, which are both significantly involved in homeostatic functions and astrogliopathologies., Competing Interests: The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results.
- Published
- 2020
- Full Text
- View/download PDF
41. Astroglial Connexin 43 Deficiency Protects against LPS-Induced Neuroinflammation: A TSPO Brain µPET Study with [ 18 F]FEPPA.
- Author
-
Vignal N, Boulay AC, San C, Cohen-Salmon M, Rizzo-Padoin N, Sarda-Mantel L, Declèves X, Cisternino S, and Hosten B
- Subjects
- Anilides pharmacokinetics, Animals, Area Under Curve, Cerebral Cortex diagnostic imaging, Connexin 43 metabolism, Female, Lipopolysaccharides, Male, Mice, Inbred C57BL, Mice, Knockout, Positron Emission Tomography Computed Tomography, Pyridines pharmacokinetics, Anilides chemistry, Astrocytes metabolism, Brain diagnostic imaging, Brain pathology, Connexin 43 deficiency, Inflammation pathology, Positron-Emission Tomography, Pyridines chemistry, Receptors, GABA metabolism
- Abstract
Astroglial connexin 43 (Cx43) has been recognized as a crucial immunoregulating factor in the brain. Its inactivation leads to a continuous immune recruitment, cytokine expression modification and a specific humoral autoimmune response against the astrocytic extracellular matrix but without brain lesions or cell lysis. To assess the impact of Cx43 deletion on the brain's inflammatory response, TSPO expression was studied by positron emission tomography (PET) imaging with a specific radioligand, [
18 F]FEPPA, in basal conditions or upon Lipopolysaccharides (LPS)-induced inflammatory challenge. Astroglial Cx43-deleted mice underwent [18 F]FEPPA PET/CT dynamic imaging with or without LPS injection (5 mg/kg) 24 h before imaging. Quantification and pharmacokinetic data modelling with a 2TCM-1K compartment model were performed. After collecting the mice brains, TSPO expression was quantified and localized by Western blot and FISH analysis. We found that astroglial Cx43 deficiency does not significantly alter TSPO expression in the basal state as observed with [18 F]FEPPA PET imaging, FISH and Western blot analysis. However, deletion of astrocyte Cx43 abolishes the LPS-induced TSPO increase. Autoimmune encephalopathy observed in astroglial Cx43-deleted mice does not involve TSPO overexpression. Consistent with previous studies showing a unique inflammatory status in the absence of astrocyte Cx43, we show that a deficient expression of astrocytic Cx43 protects the animals from LPS-induced neuroinflammation as addressed by TSPO expression., Competing Interests: The authors declare no conflict of interest.- Published
- 2020
- Full Text
- View/download PDF
42. Postnatal development of the astrocyte perivascular MLC1/GlialCAM complex defines a temporal window for the gliovascular unit maturation.
- Author
-
Gilbert A, Vidal XE, Estevez R, Cohen-Salmon M, and Boulay AC
- Subjects
- ATP Binding Cassette Transporter, Subfamily B, Member 1 metabolism, Age Factors, Animals, Animals, Newborn, Aquaporin 4 metabolism, Blood-Brain Barrier cytology, Brain anatomy & histology, Brain growth & development, Cell Adhesion Molecules, Neuron-Glia genetics, Claudin-5 metabolism, Female, In Vitro Techniques, Lectins metabolism, Male, Membrane Proteins genetics, Mice, Nerve Tissue Proteins genetics, Astrocytes physiology, Blood-Brain Barrier growth & development, Cell Adhesion Molecules, Neuron-Glia metabolism, Gene Expression Regulation, Developmental physiology, Membrane Proteins metabolism, Multiprotein Complexes metabolism, Nerve Tissue Proteins metabolism
- Abstract
Astrocytes, the most abundant glial cells of the central nervous system are morphologically complex. They display numerous processes interacting with synapses and blood vessels. At the vascular interface, astrocyte endfeet-terminated processes almost entirely cover the blood vessel surface and participate to the gliovascular unit where important vascular properties of the brain are set such as the blood-brain barrier (BBB) integrity. How specific morphological and functional interactions between astrocytes and the vascular compartment develop has not been fully investigated. Here, we elaborated an original experimental strategy to study the postnatal development of astrocyte perivascular endfeet. Using purified gliovascular units, we focused on the postnatal expression of MLC1 and GlialCAM, two transmembrane proteins forming a complex enriched at the junction between mature astrocyte perivascular endfeet. We showed that MLC1 and GlialCAM were enriched and assembled into mature complexes in astrocyte perivascular endfeet between postnatal days 10 and 15, after the formation of astrocyte perivascular Aquaporin 4 water channels. These events correlated with the increased expression of Claudin-5 and P-gP, two endothelial-specific BBB components. These results illustrate for the first time that astrocyte perivascular endfeet differentiation is a complex and progressive process which correlates with BBB maturation. Moreover, our results suggest that maturation of the astrocyte endfeet MLC1/GlialCAM complex between postnatal days 10 and 15 might be a key event in the gliovascular unit maturation.
- Published
- 2019
- Full Text
- View/download PDF
43. Preparing the Astrocyte Perivascular Endfeet Transcriptome to Investigate Astrocyte Molecular Regulations at the Brain-Vascular Interface.
- Author
-
Boulay AC, Mazaré N, Saubaméa B, and Cohen-Salmon M
- Subjects
- Protein Biosynthesis, RNA, Messenger genetics, RNA, Messenger isolation & purification, Ribosomes metabolism, Astrocytes metabolism, Blood-Brain Barrier metabolism, Gene Expression Profiling methods, Transcriptome
- Abstract
Astrocytes send out long processes that are terminated by endfeet at the vascular surface and regulate vascular functions in particular through the expression of a specific molecular repertoire in perivascular endfeet. We recently proposed that local translation might sustain this structural and functional polarization. More specifically we showed that a subset of mRNAs is distributed in astrocyte endfeet and characterized this transcriptome. We also identified among these endfeet RNAs, the ones bound to ribosomes, the polysomal astrocyte endfeet mRNAs, which we called the endfeetome. Here, we describe experimental strategies to identify mRNAs and polysomes in astrocyte perivascular endfeet, which are based on the combination of gliovascular unit purification and astrocyte-specific translating ribosome affinity purification.
- Published
- 2019
- Full Text
- View/download PDF
44. Connexin 43 Controls the Astrocyte Immunoregulatory Phenotype.
- Author
-
Boulay AC, Gilbert A, Oliveira Moreira V, Blugeon C, Perrin S, Pouch J, Le Crom S, Ducos B, and Cohen-Salmon M
- Abstract
Astrocytes are the most abundant glial cells of the central nervous system and have recently been recognized as crucial in the regulation of brain immunity. In most neuropathological conditions, astrocytes are prone to a radical phenotypical change called reactivity, which plays a key role in astrocyte contribution to neuroinflammation. However, how astrocytes regulate brain immunity in healthy conditions is an understudied question. One of the astroglial molecule involved in these regulations might be Connexin 43 (Cx43), a gap junction protein highly enriched in astrocyte perivascular endfeet-terminated processes forming the glia limitans. Indeed, Cx43 deletion in astrocytes (Cx43KO) promotes a continuous immune recruitment and an autoimmune response against an astrocyte protein, without inducing any brain lesion. To investigate the molecular basis of this unique immune response, we characterized the polysomal transcriptome of hippocampal astrocytes deleted for Cx43. Our results demonstrate that, in the absence of Cx43, astrocytes adopt an atypical reactive status with no change in most canonical astrogliosis markers, but with an upregulation of molecules promoting immune recruitment, complement activation as well as anti-inflammatory processes. Intriguingly, while several of these upregulated transcriptional events suggested an activation of the γ-interferon pathway, no increase in this cytokine or activation of related signaling pathways were found in Cx43KO. Finally, deletion of astroglial Cx43 was associated with the upregulation of several angiogenic factors, consistent with an increase in microvascular density in Cx43KO brains. Collectively, these results strongly suggest that Cx43 controls immunoregulatory and angiogenic properties of astrocytes., Competing Interests: The authors declare no conflict of interest.
- Published
- 2018
- Full Text
- View/download PDF
45. Connexin 30 is expressed in a subtype of mouse brain pericytes.
- Author
-
Mazaré N, Gilbert A, Boulay AC, Rouach N, and Cohen-Salmon M
- Subjects
- Animals, Astrocytes metabolism, Brain metabolism, Connexin 30 genetics, Glial Fibrillary Acidic Protein metabolism, Lectins metabolism, Mice, Mice, Inbred C57BL, Mice, Transgenic, Receptor, Platelet-Derived Growth Factor beta metabolism, Brain anatomy & histology, Connexin 30 metabolism, Pericytes classification, Pericytes metabolism
- Abstract
Pericytes are mural cells of blood microvessels which play a crucial role at the neurovascular interface of the central nervous system. They are involved in the regulation of blood-brain barrier integrity, angiogenesis, clearance of toxic metabolites, capillary hemodynamic responses, and neuroinflammation, and they demonstrate stem cell activity. Morphological and molecular studies to characterize brain pericytes recently pointed out some heterogeneity in pericyte population. Nevertheless, a clear definition of pericyte subtypes is still lacking. Here, we demonstrate that a fraction of brain pericytes express Connexin 30 (Cx30), a gap junction protein, which, in the brain parenchyma, was thought to be exclusively found in astrocytes. Cx30 could thus be a candidate protein in the composition of the gap junction channels already described between endothelial cells and pericytes. It could also form hemichannels or acts in a channel-independent manner to regulate pericyte morphology, as already observed in astrocytes. Altogether, our results suggest that Cx30 defines a novel brain pericyte subtype.
- Published
- 2018
- Full Text
- View/download PDF
46. Translation in astrocyte distal processes sets molecular heterogeneity at the gliovascular interface.
- Author
-
Boulay AC, Saubaméa B, Adam N, Chasseigneaux S, Mazaré N, Gilbert A, Bahin M, Bastianelli L, Blugeon C, Perrin S, Pouch J, Ducos B, Le Crom S, Genovesio A, Chrétien F, Declèves X, Laplanche JL, and Cohen-Salmon M
- Abstract
Astrocytes send out long processes that are terminated by endfeet at the vascular surface and regulate vascular functions as well as homeostasis at the vascular interface. To date, the astroglial mechanisms underlying these functions have been poorly addressed. Here we demonstrate that a subset of messenger RNAs is distributed in astrocyte endfeet. We identified, among this transcriptome, a pool of messenger RNAs bound to ribosomes, the endfeetome, that primarily encodes for secreted and membrane proteins. We detected nascent protein synthesis in astrocyte endfeet. Finally, we determined the presence of smooth and rough endoplasmic reticulum and the Golgi apparatus in astrocyte perivascular processes and endfeet, suggesting for local maturation of membrane and secreted proteins. These results demonstrate for the first time that protein synthesis occurs in astrocyte perivascular distal processes that may sustain their structural and functional polarization at the vascular interface., Competing Interests: The authors declare no conflict of interest.
- Published
- 2017
- Full Text
- View/download PDF
47. Immunoregulation at the gliovascular unit in the healthy brain: A focus on Connexin 43.
- Author
-
Boulay AC, Cisternino S, and Cohen-Salmon M
- Subjects
- Humans, Astrocytes immunology, Autoimmunity immunology, Blood-Brain Barrier immunology, Brain immunology, Connexin 43 physiology
- Abstract
In the brain, immune cell infiltration is normally kept at a very low level and a unique microenvironment strictly restricts immune reactions and inflammation. Even in such quiescent environment, a constant immune surveillance is at work allowing the brain to rapidly react to threats. To date, knowledge about the factors regulating the brain-immune system interrelationship in healthy conditions remains elusive. Interestingly, astrocytes, the most abundant glial cells in the brain, may participate in many aspects of this unique homeostasis, in particular due to their close interaction with the brain vascular system and expression of a specific molecular repertoire. Indeed, astrocytes maintain the blood-brain barrier (BBB) integrity, interact with immune cells, and participate in the regulation of intracerebral liquid movements. We recently showed that Connexin 43 (Cx43), a gap junction protein highly expressed by astrocytes at the BBB interface, is an immunoregulating factor. The absence of astroglial Cx43 leads to a transient endothelial activation, a continuous immune recruitment as well as the development of a specific humoral autoimmune response against the von Willebrand factor A domain-containing protein 5a, an extracellular matrix protein expressed by astrocytes. In this review, we propose to gather current knowledge on how astrocytes may influence the immune system in the healthy brain, focusing on their roles at the gliovascular interface. We will also consider pathological situations involving astrocyte-specific autoimmunities. Finally, we will discuss the specific role of astroglial Cx43 and the physiological consequences of immune regulations taking place on inflammation, cognition and behavior in the absence of Cx43., (Copyright © 2015 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
48. Purification of Mouse Brain Vessels.
- Author
-
Boulay AC, Saubaméa B, Declèves X, and Cohen-Salmon M
- Subjects
- Animals, Biological Transport, Cytological Techniques methods, Endothelial Cells cytology, Mice, Pericytes cytology, Signal Transduction, Tight Junctions physiology, Blood-Brain Barrier cytology, Brain blood supply
- Abstract
In the brain, most of the vascular system consists of a selective barrier, the blood-brain barrier (BBB) that regulates the exchange of molecules and immune cells between the brain and the blood. Moreover, the huge neuronal metabolic demand requires a moment-to-moment regulation of blood flow. Notably, abnormalities of these regulations are etiological hallmarks of most brain pathologies; including glioblastoma, stroke, edema, epilepsy, degenerative diseases (ex: Parkinson's disease, Alzheimer's disease), brain tumors, as well as inflammatory conditions such as multiple sclerosis, meningitis and sepsis-induced brain dysfunctions. Thus, understanding the signaling events modulating the cerebrovascular physiology is a major challenge. Much insight into the cellular and molecular properties of the various cell types that compose the cerebrovascular system can be gained from primary culture or cell sorting from freshly dissociated brain tissue. However, properties such as cell polarity, morphology and intercellular relationships are not maintained in such preparations. The protocol that we describe here is designed to purify brain vessel fragments, whilst maintaining structural integrity. We show that isolated vessels consist of endothelial cells sealed by tight junctions that are surrounded by a continuous basal lamina. Pericytes, smooth muscle cells as well as the perivascular astrocyte endfeet membranes remain attached to the endothelial layer. Finally, we describe how to perform immunostaining experiments on purified brain vessels.
- Published
- 2015
- Full Text
- View/download PDF
49. The Sarcoglycan complex is expressed in the cerebrovascular system and is specifically regulated by astroglial Cx30 channels.
- Author
-
Boulay AC, Saubaméa B, Cisternino S, Mignon V, Mazeraud A, Jourdren L, Blugeon C, and Cohen-Salmon M
- Abstract
Astrocytes, the most prominent glial cell type in the brain, send specialized processes called endfeet, around blood vessels and express a large molecular repertoire regulating the cerebrovascular system physiology. One of the most striking properties of astrocyte endfeet is their enrichment in gap junction proteins Connexin 43 and 30 (Cx43 and Cx30) allowing in particular for direct intercellular trafficking of ions and small signaling molecules through perivascular astroglial networks. In this study, we addressed the specific role of Cx30 at the gliovascular interface. Using an inactivation mouse model for Cx30 (Cx30(Δ/Δ); Δ means deleted allele) we showed that absence of Cx30 does not affect blood-brain barrier (BBB) organization and permeability. However, it results in the cerebrovascular fraction, in a strong upregulation of Sgcg encoding γ-Sarcoglycan (γ-SG), a member of the Dystrophin-associated protein complex (DAPC) connecting cytoskeleton and the extracellular matrix. The same molecular event occurs in Cx30(T5M/T5M) mutated mice, where Cx30 channels are closed, demonstrating that Sgcg regulation relied on Cx30 channel functions. We further characterized the expression of other Sarcoglycan complex (SGC) molecules in the cerebrovascular system and showed the presence of α-, β-, δ-, γ-, ε- and ζ- SG, as well as Sarcospan. Their expression was however not modified in Cx30(Δ/Δ). These results suggest that a full SGC might be present in the cerebrovascular system, and that expression of one of its member, γ-SG, depends on Cx30 channels. As described in skeletal muscles, the SGC may contribute to membrane stabilization and signal transduction in the cerebrovascular system, which may therefore be regulated by Cx30 channel-mediated functions.
- Published
- 2015
- Full Text
- View/download PDF
50. Connexin 30 sets synaptic strength by controlling astroglial synapse invasion.
- Author
-
Pannasch U, Freche D, Dallérac G, Ghézali G, Escartin C, Ezan P, Cohen-Salmon M, Benchenane K, Abudara V, Dufour A, Lübke JH, Déglon N, Knott G, Holcman D, and Rouach N
- Subjects
- Animals, Astrocytes metabolism, Behavior, Animal, Connexin 30, Female, Hippocampus cytology, Hippocampus metabolism, Hippocampus pathology, Male, Memory physiology, Mice, Inbred C57BL, Mice, Knockout, Mutation genetics, Neuronal Plasticity physiology, Astrocytes pathology, Cell Movement physiology, Connexins metabolism, Glutamic Acid metabolism, Synapses physiology, Synaptic Transmission physiology
- Abstract
Astrocytes play active roles in brain physiology by dynamic interactions with neurons. Connexin 30, one of the two main astroglial gap-junction subunits, is thought to be involved in behavioral and basic cognitive processes. However, the underlying cellular and molecular mechanisms are unknown. We show here in mice that connexin 30 controls hippocampal excitatory synaptic transmission through modulation of astroglial glutamate transport, which directly alters synaptic glutamate levels. Unexpectedly, we found that connexin 30 regulated cell adhesion and migration and that connexin 30 modulation of glutamate transport, occurring independently of its channel function, was mediated by morphological changes controlling insertion of astroglial processes into synaptic clefts. By setting excitatory synaptic strength, connexin 30 plays an important role in long-term synaptic plasticity and in hippocampus-based contextual memory. Taken together, these results establish connexin 30 as a critical regulator of synaptic strength by controlling the synaptic location of astroglial processes.
- Published
- 2014
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.