Nf1 loss and Ras hyperactivation in oligodendrocytes induce NOS-driven defects in myelin and vasculature

Debra A Mayes, Tilat A Rizvi, Haley Titus-Mitchell, Rachel Oberst, Georgianne M Ciraolo, Charles V Vorhees, Andrew P Robinson, Stephen D Miller, Jose A Cancelas, Anat O Stemmer-Rachamimov, Nancy Ratner, Debra A Mayes, Tilat A Rizvi, Haley Titus-Mitchell, Rachel Oberst, Georgianne M Ciraolo, Charles V Vorhees, Andrew P Robinson, Stephen D Miller, Jose A Cancelas, Anat O Stemmer-Rachamimov, Nancy Ratner

Abstract

Patients with neurofibromatosis type 1 (NF1) and Costello syndrome Rasopathy have behavioral deficits. In NF1 patients, these may correlate with white matter enlargement and aberrant myelin. To model these features, we induced Nf1 loss or HRas hyperactivation in mouse oligodendrocytes. Enlarged brain white matter tracts correlated with myelin decompaction, downregulation of claudin-11, and mislocalization of connexin-32. Surprisingly, non-cell-autonomous defects in perivascular astrocytes and the blood-brain barrier (BBB) developed, implicating a soluble mediator. Nitric oxide (NO) can disrupt tight junctions and gap junctions, and NO and NO synthases (NOS1-NOS3) were upregulated in mutant white matter. Treating mice with the NOS inhibitor NG-nitro-L-arginine methyl ester or the antioxidant N-acetyl cysteine corrected cellular phenotypes. CNP-HRasG12V mice also displayed locomotor hyperactivity, which could be rescued by antioxidant treatment. We conclude that Nf1/Ras regulates oligodendrocyte NOS and that dysregulated NO signaling in oligodendrocytes can alter the surrounding vasculature. The data suggest that antioxidants may improve some behavioral deficits in Rasopathy patients.

Copyright © 2013 The Authors. Published by Elsevier Inc. All rights reserved.

Figures

Figure 1. Nf1 Loss or HRas Activation…
Figure 1. Nf1 Loss or HRas Activation in Oligodendrocytes Causes Optic-Nerve Enlargement, Myelin Decompaction, Loss of Claudin-11, and Altered Cx32 Localization
(A) Gross micrographs of optic nerves at the level of the chiasm in 12-month-old animals. Ruler shows 1 mm markings. (B) Toluidine-blue-stained semithin optic-nerve cross-sections, 1 mm rostral to the chiasm. (C) Optic-nerve g-ratio scatterplots (~5,000 axons/graph; measurements 1 mm rostral to the chiasm in three to five electron micrographs at 10,000×). Red box: 99% of WT g-ratios superimposed upon graphs of other genotypes. ANOVA, Tukey post hoc, p = 2.2 × 10−16. (D) Diagram of myelin compaction. Brown circle: axon cross-section; gray: major dense lines (MDLs); orange: Cx32 GJs between compact myelin layers; blue lines: claudin-11+ TJs in compact myelin. (E) Electron micrographs (scale bar, 10 nm) of WT, PLPCre;Nf1 fl/fl, and CNP-HRas optic-nerve myelin. White arrows: intraperiod lines (ILs); black arrows: MDLs. (F–I) Western blots of total optic-nerve lysate or whole-brain cytosolic, endosomal, or myelin fractions showing myelin proteins (F), claudin-11 (G), or Cx32 cellular localization (H and I). n = 3–5 animals/genotype/experiment. See also Figures S1, S2, S3, and S4.
Figure 2. GJs Are Lost in Enlarged…
Figure 2. GJs Are Lost in Enlarged Optic-Nerve Astrocyte Endfeet after Nf1 Loss or HRas Activation
(A) Electron micrograph cross-sections of capillaries and perivascular space in optic nerve 1 mm from the chiasm (10,000×; scale bar, 2 μm). (B) Left: Diagram of capillary. Yellow: GJs. As, astrocyte endfoot; BV, blood vessel; Endo, endothelial cell. (B) Electron micrographs of capillaries 1 mm from the chiasm (50,000×; scale bar, 250 nm). Red arrows: GJs on astrocyte endfeet; red arrowheads: astrocyte endfeet lacking GJs. (C) Quantified perivascular area (% total area divided by vascular area). (D) Percentage of astrocyte endfeet with visible GJs. (E and F) Western blots using total optic-nerve lysate (E) or whole-brain endosomal fractions (F) showing Cx43. n = 3–5 animals/genotype. *p

Figure 3. Nf1 Loss or HRas Activation…

Figure 3. Nf1 Loss or HRas Activation in the Optic Nerve Causes Changes in Claudins…

Figure 3. Nf1 Loss or HRas Activation in the Optic Nerve Causes Changes in Claudins and BBB Permeability
(A) Electron micrographs of optic-nerve cross-sections of capillaries 1 mm from the chiasm (50,000×; scale bar, 200nm) show electron-dense TJs between endothelial cells. Blue arrowheads: areas of TJ disruption. (B) Quantification of the total TJs with gaps, with 200–300 TJs counted per genotype. (C) Evans blue stain of longitudinal sections of PLPCre; Nf1fl/fl and CNP-HRas optic nerve. Insets: cross-sections; arrows: blood vessel. (D) Western blots from total optic-nerve lysates showing claudin-1 and claudin-5. All experiments included three to five animals per genotype. *p

Figure 4. Increased ROS after Nf1 Loss…

Figure 4. Increased ROS after Nf1 Loss or HRas Activation

NOS sufficiency for phenotype presentation.…

Figure 4. Increased ROS after Nf1 Loss or HRas Activation
NOS sufficiency for phenotype presentation. (A) Western blot analysis of NOS (NOS1–NOS3) in optic-nerve protein lysates. Blot intensity was measured with ImageJ software and caps at upper intensity levels; “max” indicates intensity beyond the upper intensity limitations. (B) DCF-DA stain in unfixed optic-nerve cross-sections (5×; inset: 40×) from WT and CNP-HRas mice with and without the NOS inhibitor L-NAME. White line shows optic-nerve boundaries. (C) g-Ratio scatterplots show measurements of >1,000 axons/animal. (D) Quantification. (E–K) Electron micrographs of optic-nerve cross-sections 1 mm rostral to the chiasm (E and G: 10,000× and scale bar, 2 μm; I and K: 50,000× and scale bar, 100 nm). (F) Optic area quantified in semithin cross-sections 1 mm rostral to the chiasm. (H, J, and L) Quantification of perivascular area (H), percentage of astrocyte endfeet with visible GJs (J,~300 GJs/animal), and percentage of endothelial cells with TJ gaps (L, ~50–60 TJs/animal) measured using electron microscopy; n = 3–5 animals/genotype. See also Figures S6 and S7.

Figure 5. Heterozygous Nf1 Loss in Oligodendrocytes…

Figure 5. Heterozygous Nf1 Loss in Oligodendrocytes Causes Myelin and Vascular Phenotypes

(A) Gross micrographs…

Figure 5. Heterozygous Nf1 Loss in Oligodendrocytes Causes Myelin and Vascular Phenotypes
(A) Gross micrographs of optic nerves at the level of the chiasm in 12-month-old animals. Ruler shows 1 mm markings. (B–F) Optic nerve 1 mm rostral to the chiasm. (B) Toluidine-blue-stained semithin cross-sections. (C) Scatterplots of g-ratios (~5,000 axons/graph) from three to five electron micrographs (10,000×) of three to five animals/genotype. Red box: 99% of WT g-ratios superimposed upon graphs of other genotypes. ANOVA, Tukey post hoc, p = 2.2 × 10−16. (D) Electron micrographs of capillaries and perivascular space (10,000×; scale bar, 2 μm). (E) Electron micrographs of optic-nerve cross-sections of capillaries (50,000×; scale bar, 200 nm). Red arrows: GJs on astrocyte endfeet; red arrowheads: astrocyte endfeet without GJs. (F) Electron micrographs of TJs between endothelial cells (50,000×, scale bar, 200 nm). Blue arrowheads: areas of TJ disruption. (G) Western blot analysis of NOS (NOS1–NOS3) in total optic-nerve lysates. (H) Western blots in total optic-nerve lysate showing Cx43.

Figure 6. Increased Reactive Oxygen Is Detected…

Figure 6. Increased Reactive Oxygen Is Detected Only in Oligodendrocytes

(A) DCF-DA fluorescence intensity measured…

Figure 6. Increased Reactive Oxygen Is Detected Only in Oligodendrocytes
(A) DCF-DA fluorescence intensity measured by flow cytometry in dissociated forebrain + optic nerves of WT and PLP;Nf1fl+ mice 6 months after tamoxifen injection. Gates for DCF-DA with cell percentages indicated. (B) Gates were used to discriminate cell types. Top row: WT animals are shown with red gates. Middle row: PLP;Nf1fl+ animals are shown with blue gates. Bottom row: DCF-DA fluorescence intensities per cell type with WT (red) and PLP;Nf1fl+ (blue) lines. (C) Cell Rox Orange fluorescence in GFP− and GFP+ cells.

Figure 7. The Antioxidant NAC Rescues Myelin…

Figure 7. The Antioxidant NAC Rescues Myelin and Vascular Phenotypes in PLP;Nf1floxed and CNP-HRas Mice

Figure 7. The Antioxidant NAC Rescues Myelin and Vascular Phenotypes in PLP;Nf1floxed and CNP-HRas Mice
Quantification of phenotypes in WT, PLP;Nf1fl+, PLP;Nf1fl/fl, and CNP-HRas animals after 6 weeks of vehicle or NAC treatment, all in optic nerve 1 mm rostral to the chiasm. (A) Optic-nerve diameter quantified from semithin cross-sections. (B and C) Quantification of g-ratio (B) and axon diameter (C) of >1,000 axons/genotype measured by electron microscopy. (D) Quantification of % perivascular area normalized to blood vessel area. (E) Percentage of astrocyte endfeet with visible GJs (~300 GJs/animal). (F) Percentage of endothelial TJs with gaps (~50–60 TJs/animal); n = 3–5 animals/genotype.

Figure 8. CNP-HRas Animals Have Hyperactive Locomotion…

Figure 8. CNP-HRas Animals Have Hyperactive Locomotion and Hypersensitivity to Startle Behaviors

Quantification of behavior…

Figure 8. CNP-HRas Animals Have Hyperactive Locomotion and Hypersensitivity to Startle Behaviors
Quantification of behavior in CNP-HRas mice after 6 weeks of vehicle or NAC treatment. (A) Peripheral beam interruptions on locomotor behavioral test. *Vehicle p 0.08. (C) Average startle Vavg. † Vehicle p = 0.07; NAC p
All figures (8)
Similar articles
Cited by
References
    1. Acosta MT, Bearden CE, Castellanos FX, Cutting L, Elgersma Y, Gioia G, Gutmann DH, Lee YS, Legius E, Muenke M, et al. The Learning Disabilities Network (LeaDNet): using neurofibromatosis type 1 (NF1) as a paradigm for translational research. Am J Med Genet A. 2012;158A:2225–2232. - PMC - PubMed
    1. Afzal F, Polak J, Buttery L. Endothelial nitric oxide synthase in the control of osteoblastic mineralizing activity and bone integrity. J Pathol. 2004;202:503–510. - PubMed
    1. Ahn M, Lee J, Gustafsson A, Enriquez A, Lancaster E, Sul JY, Haydon PG, Paul DL, Huang Y, Abrams CK, Scherer SS. Cx29 and Cx32, two connexins expressed by myelinating glia, do not interact and are functionally distinct. J Neurosci Res. 2008;86:992–1006. - PMC - PubMed
    1. Angelow S, Ahlstrom R, Yu AS. Biology of claudins. Am J Physiol Renal Physiol. 2008;295:F867–F876. - PMC - PubMed
    1. Bennett MR, Rizvi TA, Karyala S, McKinnon RD, Ratner N. Aberrant growth and differentiation of oligodendrocyte progenitors in neurofibromatosis type 1 mutants. J Neurosci. 2003;23:7207–7217. - PMC - PubMed
Show all 55 references
Publication types
MeSH terms
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM

NCBI Literature Resources

MeSH PMC Bookshelf Disclaimer

The PubMed wordmark and PubMed logo are registered trademarks of the U.S. Department of Health and Human Services (HHS). Unauthorized use of these marks is strictly prohibited.

Follow NCBI
Figure 3. Nf1 Loss or HRas Activation…
Figure 3. Nf1 Loss or HRas Activation in the Optic Nerve Causes Changes in Claudins and BBB Permeability
(A) Electron micrographs of optic-nerve cross-sections of capillaries 1 mm from the chiasm (50,000×; scale bar, 200nm) show electron-dense TJs between endothelial cells. Blue arrowheads: areas of TJ disruption. (B) Quantification of the total TJs with gaps, with 200–300 TJs counted per genotype. (C) Evans blue stain of longitudinal sections of PLPCre; Nf1fl/fl and CNP-HRas optic nerve. Insets: cross-sections; arrows: blood vessel. (D) Western blots from total optic-nerve lysates showing claudin-1 and claudin-5. All experiments included three to five animals per genotype. *p

Figure 4. Increased ROS after Nf1 Loss…

Figure 4. Increased ROS after Nf1 Loss or HRas Activation

NOS sufficiency for phenotype presentation.…

Figure 4. Increased ROS after Nf1 Loss or HRas Activation
NOS sufficiency for phenotype presentation. (A) Western blot analysis of NOS (NOS1–NOS3) in optic-nerve protein lysates. Blot intensity was measured with ImageJ software and caps at upper intensity levels; “max” indicates intensity beyond the upper intensity limitations. (B) DCF-DA stain in unfixed optic-nerve cross-sections (5×; inset: 40×) from WT and CNP-HRas mice with and without the NOS inhibitor L-NAME. White line shows optic-nerve boundaries. (C) g-Ratio scatterplots show measurements of >1,000 axons/animal. (D) Quantification. (E–K) Electron micrographs of optic-nerve cross-sections 1 mm rostral to the chiasm (E and G: 10,000× and scale bar, 2 μm; I and K: 50,000× and scale bar, 100 nm). (F) Optic area quantified in semithin cross-sections 1 mm rostral to the chiasm. (H, J, and L) Quantification of perivascular area (H), percentage of astrocyte endfeet with visible GJs (J,~300 GJs/animal), and percentage of endothelial cells with TJ gaps (L, ~50–60 TJs/animal) measured using electron microscopy; n = 3–5 animals/genotype. See also Figures S6 and S7.

Figure 5. Heterozygous Nf1 Loss in Oligodendrocytes…

Figure 5. Heterozygous Nf1 Loss in Oligodendrocytes Causes Myelin and Vascular Phenotypes

(A) Gross micrographs…

Figure 5. Heterozygous Nf1 Loss in Oligodendrocytes Causes Myelin and Vascular Phenotypes
(A) Gross micrographs of optic nerves at the level of the chiasm in 12-month-old animals. Ruler shows 1 mm markings. (B–F) Optic nerve 1 mm rostral to the chiasm. (B) Toluidine-blue-stained semithin cross-sections. (C) Scatterplots of g-ratios (~5,000 axons/graph) from three to five electron micrographs (10,000×) of three to five animals/genotype. Red box: 99% of WT g-ratios superimposed upon graphs of other genotypes. ANOVA, Tukey post hoc, p = 2.2 × 10−16. (D) Electron micrographs of capillaries and perivascular space (10,000×; scale bar, 2 μm). (E) Electron micrographs of optic-nerve cross-sections of capillaries (50,000×; scale bar, 200 nm). Red arrows: GJs on astrocyte endfeet; red arrowheads: astrocyte endfeet without GJs. (F) Electron micrographs of TJs between endothelial cells (50,000×, scale bar, 200 nm). Blue arrowheads: areas of TJ disruption. (G) Western blot analysis of NOS (NOS1–NOS3) in total optic-nerve lysates. (H) Western blots in total optic-nerve lysate showing Cx43.

Figure 6. Increased Reactive Oxygen Is Detected…

Figure 6. Increased Reactive Oxygen Is Detected Only in Oligodendrocytes

(A) DCF-DA fluorescence intensity measured…

Figure 6. Increased Reactive Oxygen Is Detected Only in Oligodendrocytes
(A) DCF-DA fluorescence intensity measured by flow cytometry in dissociated forebrain + optic nerves of WT and PLP;Nf1fl+ mice 6 months after tamoxifen injection. Gates for DCF-DA with cell percentages indicated. (B) Gates were used to discriminate cell types. Top row: WT animals are shown with red gates. Middle row: PLP;Nf1fl+ animals are shown with blue gates. Bottom row: DCF-DA fluorescence intensities per cell type with WT (red) and PLP;Nf1fl+ (blue) lines. (C) Cell Rox Orange fluorescence in GFP− and GFP+ cells.

Figure 7. The Antioxidant NAC Rescues Myelin…

Figure 7. The Antioxidant NAC Rescues Myelin and Vascular Phenotypes in PLP;Nf1floxed and CNP-HRas Mice

Figure 7. The Antioxidant NAC Rescues Myelin and Vascular Phenotypes in PLP;Nf1floxed and CNP-HRas Mice
Quantification of phenotypes in WT, PLP;Nf1fl+, PLP;Nf1fl/fl, and CNP-HRas animals after 6 weeks of vehicle or NAC treatment, all in optic nerve 1 mm rostral to the chiasm. (A) Optic-nerve diameter quantified from semithin cross-sections. (B and C) Quantification of g-ratio (B) and axon diameter (C) of >1,000 axons/genotype measured by electron microscopy. (D) Quantification of % perivascular area normalized to blood vessel area. (E) Percentage of astrocyte endfeet with visible GJs (~300 GJs/animal). (F) Percentage of endothelial TJs with gaps (~50–60 TJs/animal); n = 3–5 animals/genotype.

Figure 8. CNP-HRas Animals Have Hyperactive Locomotion…

Figure 8. CNP-HRas Animals Have Hyperactive Locomotion and Hypersensitivity to Startle Behaviors

Quantification of behavior…

Figure 8. CNP-HRas Animals Have Hyperactive Locomotion and Hypersensitivity to Startle Behaviors
Quantification of behavior in CNP-HRas mice after 6 weeks of vehicle or NAC treatment. (A) Peripheral beam interruptions on locomotor behavioral test. *Vehicle p 0.08. (C) Average startle Vavg. † Vehicle p = 0.07; NAC p
All figures (8)
Similar articles
Cited by
References
    1. Acosta MT, Bearden CE, Castellanos FX, Cutting L, Elgersma Y, Gioia G, Gutmann DH, Lee YS, Legius E, Muenke M, et al. The Learning Disabilities Network (LeaDNet): using neurofibromatosis type 1 (NF1) as a paradigm for translational research. Am J Med Genet A. 2012;158A:2225–2232. - PMC - PubMed
    1. Afzal F, Polak J, Buttery L. Endothelial nitric oxide synthase in the control of osteoblastic mineralizing activity and bone integrity. J Pathol. 2004;202:503–510. - PubMed
    1. Ahn M, Lee J, Gustafsson A, Enriquez A, Lancaster E, Sul JY, Haydon PG, Paul DL, Huang Y, Abrams CK, Scherer SS. Cx29 and Cx32, two connexins expressed by myelinating glia, do not interact and are functionally distinct. J Neurosci Res. 2008;86:992–1006. - PMC - PubMed
    1. Angelow S, Ahlstrom R, Yu AS. Biology of claudins. Am J Physiol Renal Physiol. 2008;295:F867–F876. - PMC - PubMed
    1. Bennett MR, Rizvi TA, Karyala S, McKinnon RD, Ratner N. Aberrant growth and differentiation of oligodendrocyte progenitors in neurofibromatosis type 1 mutants. J Neurosci. 2003;23:7207–7217. - PMC - PubMed
Show all 55 references
Publication types
MeSH terms
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM
Figure 4. Increased ROS after Nf1 Loss…
Figure 4. Increased ROS after Nf1 Loss or HRas Activation
NOS sufficiency for phenotype presentation. (A) Western blot analysis of NOS (NOS1–NOS3) in optic-nerve protein lysates. Blot intensity was measured with ImageJ software and caps at upper intensity levels; “max” indicates intensity beyond the upper intensity limitations. (B) DCF-DA stain in unfixed optic-nerve cross-sections (5×; inset: 40×) from WT and CNP-HRas mice with and without the NOS inhibitor L-NAME. White line shows optic-nerve boundaries. (C) g-Ratio scatterplots show measurements of >1,000 axons/animal. (D) Quantification. (E–K) Electron micrographs of optic-nerve cross-sections 1 mm rostral to the chiasm (E and G: 10,000× and scale bar, 2 μm; I and K: 50,000× and scale bar, 100 nm). (F) Optic area quantified in semithin cross-sections 1 mm rostral to the chiasm. (H, J, and L) Quantification of perivascular area (H), percentage of astrocyte endfeet with visible GJs (J,~300 GJs/animal), and percentage of endothelial cells with TJ gaps (L, ~50–60 TJs/animal) measured using electron microscopy; n = 3–5 animals/genotype. See also Figures S6 and S7.
Figure 5. Heterozygous Nf1 Loss in Oligodendrocytes…
Figure 5. Heterozygous Nf1 Loss in Oligodendrocytes Causes Myelin and Vascular Phenotypes
(A) Gross micrographs of optic nerves at the level of the chiasm in 12-month-old animals. Ruler shows 1 mm markings. (B–F) Optic nerve 1 mm rostral to the chiasm. (B) Toluidine-blue-stained semithin cross-sections. (C) Scatterplots of g-ratios (~5,000 axons/graph) from three to five electron micrographs (10,000×) of three to five animals/genotype. Red box: 99% of WT g-ratios superimposed upon graphs of other genotypes. ANOVA, Tukey post hoc, p = 2.2 × 10−16. (D) Electron micrographs of capillaries and perivascular space (10,000×; scale bar, 2 μm). (E) Electron micrographs of optic-nerve cross-sections of capillaries (50,000×; scale bar, 200 nm). Red arrows: GJs on astrocyte endfeet; red arrowheads: astrocyte endfeet without GJs. (F) Electron micrographs of TJs between endothelial cells (50,000×, scale bar, 200 nm). Blue arrowheads: areas of TJ disruption. (G) Western blot analysis of NOS (NOS1–NOS3) in total optic-nerve lysates. (H) Western blots in total optic-nerve lysate showing Cx43.
Figure 6. Increased Reactive Oxygen Is Detected…
Figure 6. Increased Reactive Oxygen Is Detected Only in Oligodendrocytes
(A) DCF-DA fluorescence intensity measured by flow cytometry in dissociated forebrain + optic nerves of WT and PLP;Nf1fl+ mice 6 months after tamoxifen injection. Gates for DCF-DA with cell percentages indicated. (B) Gates were used to discriminate cell types. Top row: WT animals are shown with red gates. Middle row: PLP;Nf1fl+ animals are shown with blue gates. Bottom row: DCF-DA fluorescence intensities per cell type with WT (red) and PLP;Nf1fl+ (blue) lines. (C) Cell Rox Orange fluorescence in GFP− and GFP+ cells.
Figure 7. The Antioxidant NAC Rescues Myelin…
Figure 7. The Antioxidant NAC Rescues Myelin and Vascular Phenotypes in PLP;Nf1floxed and CNP-HRas Mice
Quantification of phenotypes in WT, PLP;Nf1fl+, PLP;Nf1fl/fl, and CNP-HRas animals after 6 weeks of vehicle or NAC treatment, all in optic nerve 1 mm rostral to the chiasm. (A) Optic-nerve diameter quantified from semithin cross-sections. (B and C) Quantification of g-ratio (B) and axon diameter (C) of >1,000 axons/genotype measured by electron microscopy. (D) Quantification of % perivascular area normalized to blood vessel area. (E) Percentage of astrocyte endfeet with visible GJs (~300 GJs/animal). (F) Percentage of endothelial TJs with gaps (~50–60 TJs/animal); n = 3–5 animals/genotype.
Figure 8. CNP-HRas Animals Have Hyperactive Locomotion…
Figure 8. CNP-HRas Animals Have Hyperactive Locomotion and Hypersensitivity to Startle Behaviors
Quantification of behavior in CNP-HRas mice after 6 weeks of vehicle or NAC treatment. (A) Peripheral beam interruptions on locomotor behavioral test. *Vehicle p 0.08. (C) Average startle Vavg. † Vehicle p = 0.07; NAC p
All figures (8)

References

    1. Acosta MT, Bearden CE, Castellanos FX, Cutting L, Elgersma Y, Gioia G, Gutmann DH, Lee YS, Legius E, Muenke M, et al. The Learning Disabilities Network (LeaDNet): using neurofibromatosis type 1 (NF1) as a paradigm for translational research. Am J Med Genet A. 2012;158A:2225–2232.
    1. Afzal F, Polak J, Buttery L. Endothelial nitric oxide synthase in the control of osteoblastic mineralizing activity and bone integrity. J Pathol. 2004;202:503–510.
    1. Ahn M, Lee J, Gustafsson A, Enriquez A, Lancaster E, Sul JY, Haydon PG, Paul DL, Huang Y, Abrams CK, Scherer SS. Cx29 and Cx32, two connexins expressed by myelinating glia, do not interact and are functionally distinct. J Neurosci Res. 2008;86:992–1006.
    1. Angelow S, Ahlstrom R, Yu AS. Biology of claudins. Am J Physiol Renal Physiol. 2008;295:F867–F876.
    1. Bennett MR, Rizvi TA, Karyala S, McKinnon RD, Ratner N. Aberrant growth and differentiation of oligodendrocyte progenitors in neurofibromatosis type 1 mutants. J Neurosci. 2003;23:7207–7217.
    1. Beutheu YS, Belmonte L, Galas L, Boukhettala N, Bôle-Feysot C, Déchelotte P, Coëffier M. Methotrexate modulates tight junctions through NF-κB, MEK, and JNK pathways. J Pediatr Gastroenterol Nutr. 2012;54:463–470.
    1. Brannan CI, Perkins AS, Vogel KS, Ratner N, Nordlund ML, Reid SW, Buchberg AM, Jenkins NA, Parada LF, Copeland NG. Targeted disruption of the neurofibromatosis type-1 gene leads to developmental abnormalities in heart and various neural crest-derived tissues. Genes Dev. 1994;8:1019–1029.
    1. Chen Y, Curran CP, Nebert DW, Patel KV, Williams MT, Vorhees CV. Effect of chronic glutathione deficiency on the behavioral phenotype of Gclm−/− knockout mice. Neurotoxicol Teratol. 2012;34:450–457.
    1. Chow E, Mottahedeh J, Prins M, Ridder W, Nusinowitz S, Bronstein JM. Disrupted compaction of CNS myelin in an OSP/Claudin-11 and PLP/DM20 double knockout mouse. Mol Cell Neurosci. 2005;29:405–413.
    1. Daston MM, Scrable H, Nordlund M, Sturbaum AK, Nissen LM, Ratner N. The protein product of the neurofibromatosis type 1 gene is expressed at highest abundance in neurons, Schwann cells, and oligodendrocytes. Neuron. 1992;8:415–428.
    1. DiPaolo DP, Zimmerman RA, Rorke LB, Zackai EH, Bilaniuk LT, Yachnis AT. Neurofibromatosis type 1: pathologic substrate of high-signal-intensity foci in the brain. Radiology. 1995;195:721–724.
    1. Dlugosova K, Okruhlicova L, Mitasikova M, Sotnikova R, Bernatova I, Weismann P, Slezak J, Tribulova N. Modulation of connexin-43 by omega-3 fatty acids in the aorta of old spontaneously hypertensive rats. J Physiol Pharmacol. 2009;60:63–69.
    1. Doerflinger NH, Macklin WB, Popko B. Inducible site-specific recombination in myelinating cells. Genesis. 2003;35:63–72.
    1. Dumont M, Stack C, Elipenahli C, Jainuddin S, Gerges M, Starkova NN, Yang L, Starkov AA, Beal F. Behavioral deficit, oxidative stress, and mitochondrial dysfunction precede tau pathology in P301S transgenic mice. FASEB J. 2011;25:4063–4072.
    1. Ferro E, Goitre L, Retta SF, Trabalzini L. The interplay between ROS and Ras GTPases: physiological and pathological implications. J Signal Transduct. 2012;2012:365769.
    1. Gandhi GK, Ball KK, Cruz NF, Dienel GA. Hyperglycaemia and diabetes impair gap junctional communication among astrocytes. ASN Neuro. 2010;2:e00030.
    1. Gysin S, Salt M, Young A, McCormick F. Therapeutic strategies for targeting ras proteins. Genes Cancer. 2011;2:359–372.
    1. Hegedus B, Dasgupta B, Shin JE, Emnett RJ, Hart-Mahon EK, Elghazi L, Bernal-Mizrachi E, Gutmann DH. Neurofibromatosis-1 regulates neuronal and glial cell differentiation from neuroglial progenitors in vivo by both cAMP- and Ras-dependent mechanisms. Cell Stem Cell. 2007;1:443–457.
    1. Heimfarth L, Loureiro SO, Pierozan P, de Lima BO, Reis KP, Torres EB, Pessoa-Pureur R. Methylglyoxal-induced cytotoxicity in neonatal rat brain: a role for oxidative stress and MAP kinases. Metab Brain Dis. 2013;28:429–438.
    1. Hyman SL, Gill DS, Shores EA, Steinberg A, North KN. T2 hyperintensities in children with neurofibromatosis type 1 and their relationship to cognitive functioning. J Neurol Neurosurg Psychiatry. 2007;78:1088–1091.
    1. Kamens HM, Crabbe JC. The parallel rod floor test: a measure of ataxia in mice. Nat Protoc. 2007;2:277–281.
    1. Karlsgodt KH, Rosser T, Lutkenhoff ES, Cannon TD, Silva A, Bearden CE. Alterations in white matter microstructure in neurofibromatosis-1. PLoS ONE. 2012;7:e47854.
    1. Kim KY, Ju WK, Hegedus B, Gutmann DH, Ellisman MH. Ultrastructural characterization of the optic pathway in a mouse model of neurofibromatosis-1 optic glioma. Neuroscience. 2010;170:178–188.
    1. Komatsu T, Ireland DD, Chung N, Doré A, Yoder M, Reiss CS. Regulation of the BBB during viral encephalitis: roles of IL-12 and NOS. Nitric Oxide. 1999;3:327–339.
    1. Lee KM, Kwon JY, Lee KW, Lee HJ. Ascorbic acid 6-palmitate suppresses gap-junctional intercellular communication through phosphorylation of connexin 43 via activation of the MEK-ERK pathway. Mutat Res. 2009;660:51–56.
    1. Li W, Cui Y, Kushner SA, Brown RA, Jentsch JD, Frankland PW, Cannon TD, Silva AJ. The HMG-CoA reductase inhibitor lovastatin reverses the learning and attention deficits in a mouse model of neurofibromatosis type 1. Curr Biol. 2005;15:1961–1967.
    1. Li X, Newbern JM, Wu Y, Morgan-Smith M, Zhong J, Charron J, Snider WD. MEK is a key regulator of gliogenesis in the developing brain. Neuron. 2012;75:1035–1050.
    1. Magnotti LM, Goodenough DA, Paul DL. Deletion of oligodendrocyte Cx32 and astrocyte Cx43 causes white matter vacuolation, astrocyte loss and early mortality. Glia. 2011;59:1064–1074.
    1. Mayes DA, Rizvi TA, Cancelas JA, Kolasinski NT, Ciraolo GM, Stemmer-Rachamimov AO, Ratner N. Perinatal or adult Nf1 inactivation using tamoxifen-inducible PlpCre each cause neurofibroma formation. Cancer Res. 2011;71:4675–4685.
    1. Patel AV, Eaves D, Jessen WJ, Rizvi TA, Ecsedy JA, Qian MG, Aronow BJ, Perentesis JP, Serra E, Cripe TP, et al. Ras-driven transcriptome analysis identifies aurora kinase A as a potential malignant peripheral nerve sheath tumor therapeutic target. Clin Cancer Res. 2012;18:5020–5030.
    1. Perilongo G, Opocher E, Viscardi E. Optic, hypothalamic, and thalamic tumors. Handb Clin Neurol. 2012;105:607–613.
    1. Pride N, Payne JM, Webster R, Shores EA, Rae C, North KN. Corpus callosum morphology and its relationship to cognitive function in neurofibromatosis type 1. J Child Neurol. 2010;25:834–841.
    1. Pummi KP, Aho HJ, Laato MK, Peltonen JT, Peltonen SA. Tight junction proteins and perineurial cells in neurofibromas. J Histochem Cytochem. 2006;54:53–61.
    1. Radosinska J, Bacova B, Bernatova I, Navarova J, Zhukovska A, Shysh A, Okruhlicova L, Tribulova N. Myocardial NOS activity and connexin-43 expression in untreated and omega-3 fatty acids-treated spontaneously hypertensive and hereditary hypertriglyceridemic rats. Mol Cell Biochem. 2011;347:163–173.
    1. Robertson CA, Green BG, Niedzwiecki L, Harrison RK, Grant SK. Effect of nitric oxide synthase substrate analog inhibitors on rat liver arginase. Biochem Biophys Res Commun. 1993;197:523–528.
    1. Saura M, Tarin C, Zaragoza C. Recent insights into the implication of nitric oxide in osteoblast differentiation and proliferation during bone development. ScientificWorldJournal. 2010;10:624–632.
    1. Segretain D, Falk MM. Regulation of connexin biosynthesis, assembly, gap junction formation, and removal. Biochim Biophys Acta. 2004;1662:3–21.
    1. Selek S, Bulut M, Ocak AR, Kalenderoğlu A, Savaş HA. Evaluation of total oxidative status in adult attention deficit hyperactivity disorder and its diagnostic implications. J Psychiatr Res. 2012;46:451–455.
    1. Skelton MR, Schaefer TL, Graham DL, Degrauw TJ, Clark JF, Williams MT, Vorhees CV. Creatine transporter (CrT; Slc6a8) knockout mice as a model of human CrT deficiency. PLoS ONE. 2011;6:e16187.
    1. Simard M, Arcuino G, Takano T, Liu QS, Nedergaard M. Signaling at the gliovascular interface. J Neurosci. 2003;23:9254–9262.
    1. Solga AC, Gianino SM, Gutmann DH. NG2-cells are not the cell of origin for murine neurofibromatosis-1 (Nf1) optic glioma. Oncogene. 2013 Published online January 14, 2013 .
    1. Taniguchi Ishikawa E, Gonzalez-Nieto D, Ghiaur G, Dunn SK, Ficker AM, Murali B, Madhu M, Gutstein DE, Fishman GI, Barrio LC, Cancelas JA. Connexin-43 prevents hematopoietic stem cell senescence through transfer of reactive oxygen species to bone marrow stromal cells. Proc Natl Acad Sci USA. 2012;109:9071–9076.
    1. Tiboni GM, Giampietro F, Di Giulio C. The nitric oxide synthesis inhibitor Nomega-nitro-L-arginine methyl ester (L-NAME) causes limb defects in mouse fetuses: protective effect of acute hyperoxia. Pediatr Res. 2003;54:69–76.
    1. Tidyman WE, Rauen KA. Noonan, Costello and cardio-faciocutaneous syndromes: dysregulation of the Ras-MAPK pathway. Expert Rev Mol Med. 2008;10:e37.
    1. Tong JJ, Schriner SE, McCleary D, Day BJ, Wallace DC. Life extension through neurofibromin mitochondrial regulation and antioxidant therapy for neurofibromatosis-1 in Drosophila melanogaster. Nat Genet. 2007;39:476–485.
    1. van Engelen SJ, Krab LC, Moll HA, de Goede-Bolder A, Pluijm SM, Catsman-Berrevoets CE, Elgersma Y, Lequin MH. Quantitative differentiation between healthy and disordered brain matter in patients with neurofibromatosis type I using diffusion tensor imaging. AJNR Am J Neuroradiol. 2008;29:816–822.
    1. Vega Rasgado LA, Ceballos Reyes GM, Vega-Diaz MF. Anticonvulsant drugs, oxidative stress and nitric oxide. Proc West Pharmacol Soc. 2011;54:41–48.
    1. Viosca J, Schuhmacher AJ, Guerra C, Barco A. Germline expression of H-Ras(G12V) causes neurological deficits associated to Costello syndrome. Genes Brain Behav. 2009;8:60–71.
    1. Vorhees CV, Williams MT. Morris water maze: procedures for assessing spatial and related forms of learning and memory. Nat Protoc. 2006;1:848–858.
    1. Wang Y, Kim E, Wang X, Novitch BG, Yoshikawa K, Chang LS, Zhu Y. ERK inhibition rescues defects in fate specification of Nf1-deficient neural progenitors and brain abnormalities. Cell. 2012;150:816–830.
    1. Williams VC, Lucas J, Babcock MA, Gutmann DH, Korf B, Maria BL. Neurofibromatosis type 1 revisited. Pediatrics. 2009;123:124–133.
    1. Wolburg H, Lippoldt A. Tight junctions of the blood-brain barrier: development, composition and regulation. Vascul Pharmacol. 2002;38:323–337.
    1. Zatorre RJ, Fields RD, Johansen-Berg H. Plasticity in gray and white: neuroimaging changes in brain structure during learning. Nat Neurosci. 2012;15:528–536.
    1. Zeger M, Popken G, Zhang J, Xuan S, Lu QR, Schwab MH, Nave KA, Rowitch D, D’Ercole AJ, Ye P. Insulin-like growth factor type 1 receptor signaling in the cells of oligodendrocyte lineage is required for normal in vivo oligodendrocyte development and myelination. Glia. 2007;55:400–411.
    1. Zhang JH, Kawashima S, Yokoyama M, Huang P, Hill CE. Increased eNOS accounts for changes in connexin expression in renal arterioles during diabetes. Anat Rec A Discov Mol Cell Evol Biol. 2006;288:1000–1008.

Source: PubMed

3
Se inscrever