Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Molecules ; 25(18)2020 Sep 07.
Article in English | MEDLINE | ID: mdl-32906751

ABSTRACT

In this study, the neural phenotype is explored in rodent models of the spinocerebellar disorder known as the Friedreich Ataxia (FA), which results from mutations within the gene encoding the Frataxin mitochondrial protein. For this, the M12 line, bearing a targeted mutation, which disrupts the Frataxin gene exon 4 was used, together with the M02 line, which, in addition, is hemizygous for the human Frataxin gene mutation (Pook transgene), implying the occurrence of 82-190 GAA repeats within its first intron. The mutant mice phenotype was compared to the one of wild type littermates in regions undergoing differential profiles of neurogenesis, including the cerebellar cortex and the spinal cord by using neuronal (ß-tubulin) and glial (Glial Fibrillary Acidic Protein) markers as well as the Contactin 1 axonal glycoprotein, involved in neurite growth control. Morphological/morphometric analyses revealed that while in Frataxin mutant mice the neuronal phenotype was significantly counteracted, a glial upregulation occurred at the same time. Furthermore, Contactin 1 downregulation suggested that changes in the underlying gene contributed to the disorder pathogenesis. Therefore, the FA phenotype implies an alteration of the developmental profile of neuronal and glial precursors. Finally, epigallocatechin gallate polyphenol administration counteracted the disorder, indicating protective effects of antioxidant administration.


Subject(s)
Contactins/genetics , Disease Susceptibility , Friedreich Ataxia/genetics , Friedreich Ataxia/metabolism , Gene Expression , Animals , Antioxidants/administration & dosage , Cell Communication , Cerebellum/drug effects , Cerebellum/metabolism , Contactins/metabolism , Disease Models, Animal , Humans , Immunohistochemistry , Mice , Mutation , Neuroglia/metabolism , Neurons/drug effects , Neurons/metabolism , Phenotype , Signal Transduction , Spinal Cord/drug effects , Spinal Cord/metabolism
2.
Front Cell Dev Biol ; 7: 119, 2019.
Article in English | MEDLINE | ID: mdl-31380366

ABSTRACT

In this study the mechanisms are explored, which modulate expression and function of cell surface adhesive glycoproteins of the Immunoglobulin Supergene Family (IgSF), and in particular of its Contactin subset, during neuronal precursor developmental events. In this context, a specific topic concerns the significance of the expression profile of such molecules and their ability to modulate signaling pathways activated through nutraceuticals, in particular polyphenols, administration. Both in vitro and in vivo approaches are chosen. As for the former, by using as a model the human SH-SY5Y neuroblastoma line, the effects of grape seed polyphenols are evaluated on proliferation and commitment/differentiation events along the neuronal lineage. In SH-SY5Y cell cultures, polyphenols were found to counteract precursor proliferation while promoting their differentiation, as deduced by studying their developmental parameters through the expression of cell cycle and neuronal commitment/differentiation markers as well as by measuring neurite growth. In such cultures, Cyclin E expression and BrdU incorporation were downregulated, indicating reduced precursor proliferation while increased neuronal differentiation was inferred from upregulation of cell cycle exit (p27-Kip) and neuronal commitment (NeuN) markers as well as by measuring neurite length through morphometric analysis. The polyphenol effects on developmental parameters were also explored in vivo, in cerebellar cortex, by using as a model the TAG/F3 transgenic line, which undergoes delayed neural development as a consequence of Contactin1 adhesive glycoprotein upregulation and premature expression under control of the Contactin2 gene (Cntn-2) promoter. In this transgenic line, a Notch pathway activation is known to occur and polyphenol treatment was found to counteract such an effect, demonstrated through downregulation of the Hes-1 transcription factor. Polyphenols also downregulated the expression of adhesive glycoproteins of the Contactin family themselves, demonstrated for both Contactin1 and Contactin2, indicating the involvement of changes in the expression of the underlying genes in the observed phenotype. These data support the hypothesis that the complex control exerted by polyphenols on neural development involves modulation of expression and function of the genes encoding cell adhesion molecules of the Contactin family and of the associated signaling pathways, indicating potential mechanisms whereby such compounds may control neurogenesis.

3.
Stem Cell Res Ther ; 10(1): 29, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30646960

ABSTRACT

Cognitive deficit has been identified in one third of patients affected by Duchenne Muscular Dystrophy, primarily attributed to loss of the short Dp71 dystrophin, the major brain dystrophin isoform. In this study, we investigated for the first time the Dp71 and Dp71-associated proteins cellular localization and expression in human neurons obtained by differentiation from induced pluripotent stem cell line of a patient affected by cognitive impairment. We found structural and molecular alterations in both pluripotent stem cell and derived neurons, reduced Dp71 expression, and a Ca2+ cytoplasmic overload in neurons coupled with increased expression of the SERCA2 pump in the dystrophic neurons. These results suggest that the reduction of Dp71 protein in the Duchenne muscular dystrophy neurons leads to alterations in SERCA2 and to elevated cytosolic Ca2+ concentration with consequent potential disruption of the dystrophin proteins and Dp71-associated proteins.


Subject(s)
Cognitive Dysfunction/genetics , Dystrophin/genetics , Dystrophin/metabolism , Muscular Dystrophy, Duchenne/genetics , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Adult , Cell Differentiation , Humans , Male , Muscular Dystrophy, Duchenne/metabolism , Neurons , Young Adult
4.
J Pediatr Hematol Oncol ; 40(4): e207-e214, 2018 05.
Article in English | MEDLINE | ID: mdl-29432309

ABSTRACT

Epidemiologic studies have suggested that parental occupations, pesticide use, environmental factors, and genetic polymorphism are involved in the etiology of childhood acute leukemia (CAL). In total, 116 cases of CAL and 162 controls were recruited and submitted to blood drawing to assess the presence of genetic polymorphisms. Parental occupations, pesticides exposure, and other potential determinants were investigated. Increased risk for CAL was associated with prenatal maternal use of insecticides/rodenticides (odds ratio [OR]=1.87; 95% confidence intervals [CI], 1.04-3.33), with subjects living <100 m from pesticide-treated fields (OR=3.21; 95% CI, 1.37-7.53) and with a paternal occupation as traffic warden/policeman (OR=4.02; 95% CI, 1.63-9.87). Associations were found between CAL and genetic polymorphism of CYP2D6*4 for homozygous alleles (mutant type/mutant type: OR=6.39; 95% CI, 1.17-34.66). In conclusion, despite the small sample size, maternal prenatal exposure to pesticides, paternal occupation as a traffic warden/police officer, and CYP2D6*4 polymorphism could play a role in the etiology of CAL.


Subject(s)
Alleles , Cytochrome P-450 CYP2D6/genetics , Leukemia , Maternal Exposure/adverse effects , Occupational Exposure/adverse effects , Pesticides/adverse effects , Polymorphism, Genetic , Acute Disease , Child , Child, Preschool , Female , Humans , Leukemia/enzymology , Leukemia/epidemiology , Leukemia/etiology , Leukemia/genetics , Male , Military Personnel , Police
5.
Ecotoxicol Environ Saf ; 140: 156-161, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28258041

ABSTRACT

BACKGROUND: Particulate matter (PM) is the most efficient vehicle for the inhalation and absorption of toxic substances into the body. METHOD: The present study was aimed at testing the hypothesis that PM10 samples collected on quartz filters exert an angiogenic activity in vivo in the chick embryo chorioallantoic membrane (CAM) assay. RESULTS: When the low, medium, and high PM10 concentrations filters were tested in the CAM assay, an increasing number of microvessels was detectable after 4 days of applications of the filters. Moreover, at histological level, numerous microvessels and a dense inflammatory infiltrate were recognizable in the CAM mesenchyme. CONCLUSION: Our data show a clear dose-response relationship between the dose variable (PM10 and Bap) and the outcome variable. So far, the PM10 target value is determined on the basis of regulatory agreements and is not health-based. In addition, the mere gravimetric measure of PM10 cannot be considered a fully reliable surrogate of the overall toxicity of the mixture.


Subject(s)
Air Pollutants/toxicity , Chorioallantoic Membrane/blood supply , Neovascularization, Pathologic/chemically induced , Particulate Matter/toxicity , Air Pollutants/analysis , Animals , Carcinogens/analysis , Carcinogens/toxicity , Chick Embryo , Chorioallantoic Membrane/chemistry , Metals/analysis , Metals/toxicity , Microvessels/physiology , Nitro Compounds/analysis , Nitro Compounds/toxicity , Particulate Matter/analysis , Polycyclic Aromatic Hydrocarbons/toxicity
6.
Mol Cell Neurosci ; 81: 49-63, 2017 06.
Article in English | MEDLINE | ID: mdl-27871938

ABSTRACT

This review article focuses on the Contactin (CNTN) subset of the Immunoglobulin supergene family (IgC2/FNIII molecules), whose components share structural properties (the association of Immunoglobulin type C2 with Fibronectin type III domains), as well as a general role in cell contact formation and axonal growth control. IgC2/FNIII molecules include 6 highly related components (CNTN 1-6), associated with the cell membrane via a Glycosyl Phosphatidyl Inositol (GPI)-containing lipid tail. Contactin 1 and Contactin 2 share ~50 (49.38)% identity at the aminoacid level. They are components of the cell surface, from which they may be released in soluble forms. They bind heterophilically to multiple partners in cis and in trans, including members of the related L1CAM family and of the Neurexin family Contactin-associated proteins (CNTNAPs or Casprs). Such interactions are important for organising the neuronal membrane, as well as for modulating the growth and pathfinding of axon tracts. In addition, they also mediate the functional maturation of axons by promoting their interactions with myelinating cells at the nodal, paranodal and juxtaparanodal regions. Such interactions also mediate differential ionic channels (both Na+ and K+) distribution, which is of critical relevance in the generation of the peak-shaped action potential. Indeed, thanks to their interactions with Ankyrin G, Na+ channels map within the nodal regions, where they drive axonal depolarization. However, no ionic channels are found in the flanking Contactin1-containing paranodal regions, where CNTN1 interactions with Caspr1 and with the Ig superfamily component Neurofascin 155 in cis and in trans, respectively, build a molecular barrier between the node and the juxtaparanode. In this region K+ channels are clustered, depending upon molecular interactions with Contactin 2 and with Caspr2. In addition to these functions, the Contactins appear to have also a role in degenerative and inflammatory disorders: indeed Contactin 2 is involved in neurodegenerative disorders with a special reference to the Alzheimer disease, given its ability to work as a ligand of the Alzheimer Precursor Protein (APP), which results in increased Alzheimer Intracellular Domain (AICD) release in a γ-secretase-dependent manner. On the other hand Contactin 1 drives Notch signalling activation via the Hes pathway, which could be consistent with its ability to modulate neuroinflammation events, and with the possibility that Contactin 1-dependent interactions may participate to the pathogenesis of the Multiple Sclerosis and of other inflammatory disorders.


Subject(s)
Axons/metabolism , Contactins/metabolism , Neurodevelopmental Disorders/metabolism , Neurogenesis , Animals , Contactins/chemistry , Contactins/genetics , Humans , Neurodevelopmental Disorders/genetics
8.
Exp Cell Res ; 343(2): 190-207, 2016 05 01.
Article in English | MEDLINE | ID: mdl-27015747

ABSTRACT

The blood-brain barrier (BBB) is altered in mdx mouse, an animal model to study Duchenne muscular dystrophy (DMD). Our previous work demonstrated that perivascular glial endfeet control the selective exchanges between blood and neuropil as well as the BBB development and integrity; the alterations of dystrophin and dystrophin-associated protein complex (DAPs) in the glial cells of mdx mouse, parallel damages of the BBB and increase in vascular permeability. The aim of this study was to improve our knowledge about brain cellular components in the mdx mouse through the isolation, for the first time, of the adult neural stem cells (ANSCs). We characterized them by FACS, electron microscopy, confocal immunofluorescence microscopy, Real Time-PCR and western blotting, and we studied the expression of the DAPs aquaporin-4 (AQP4), potassium channel Kir4.1, α- and ß-dystroglycan (αDG, ßDG), α-syntrophin (αSyn), and short dystrophin isoform Dp71 proteins. The results showed that the mdx ANSCs expressed CD133 and Nestin receptor as the control ones, but showed a reduction in Notch receptor and altered cell proliferation with an increment in the apoptotic nuclei. Ultrastructurally, they appeared 50% size reduced compared to control ones, with a few cytoplasmic organelles. Moreover, the mdx ANSCs are devoid in full length dystrophin 427, and they expressed post-transcriptional reduction in the Dp71 in parallel with the ubiquitin proteasome activation, and decrement of DAPs proteins which appeared diffused in the cytoplasm and not polarized on the stem cells plasmamembrane, as prevalently observed in the controls. Overall, these results indicate that structural and molecular alterations affect the neural stem cells in the dystrophic brain, whose increased apoptosis and reduced Dp71 and DAPs proteins expression, together with loss in Dp427 dystrophin, could be responsible of the altered mdx glial maintenance and differentiation and consequent failure in the vessels barrier control occurring in the adult dystrophic brain.


Subject(s)
Cell Separation/methods , Muscular Dystrophy, Animal/pathology , Neural Stem Cells/cytology , AC133 Antigen/metabolism , Adult Stem Cells/cytology , Adult Stem Cells/metabolism , Animals , Aquaporin 4/metabolism , Blotting, Western , Calcium-Binding Proteins , Cell Differentiation , Dystroglycans/metabolism , Dystrophin/metabolism , Flow Cytometry , Fluorescent Antibody Technique , Glial Fibrillary Acidic Protein/metabolism , Membrane Proteins , Mice, Inbred C57BL , Mice, Inbred mdx , Muscle Proteins , Muscular Dystrophy, Animal/genetics , Neural Stem Cells/metabolism , Neural Stem Cells/ultrastructure , Potassium Channels, Inwardly Rectifying/metabolism , Proteasome Endopeptidase Complex/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Spheroids, Cellular/cytology , Spheroids, Cellular/ultrastructure , Ubiquitin/metabolism
9.
Dev Neurobiol ; 75(12): 1420-40, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25820347

ABSTRACT

The Contactin-1 axonal glycoprotein (formerly F3/Contactin) plays a relevant role in cerebellar ontogenesis, as shown in Contactin-1 KO-mice and in transgenic mice misexpressing the corresponding cDNA from a heterologous promoter. Likewise, null mutant mice for the Collier/Olf1/Early B-cell family transcription factor EBF2, in which Purkinje neuron development is primarily affected, exhibit abnormalities in cerebellar corticogenesis. Here, to evaluate the contribution to the Ebf2 null phenotype of changes in the profile of Contactin-1, we study its expression in Ebf2 null mice. In addition, we explore the activation profile of the Cntn1 gene promoter upon transferring the Ebf2 mutation to transgenic mice expressing an enhanced green fluorescent protein reporter under control of Cntn1 gene regulatory sequences. In Ebf2 null mice, Contactin-1 protein expression and Cntn1 gene promoter activity are both downregulated during embryonic and early postnatal cerebellar development, both in the rostral and caudal folia, while in the latter an upregulation is observed at postnatal day 8. In vitro, vectors driving EBF1,2,3 transcription factors from a cytomegalovirus (CMV) promoter transactivate a Cntn1-Choline acetyltransferse (CAT) promoter-reporter construct in cotransfection assays and, accordingly, by chromatin immunoprecipitation, we show that the Cntn1 gene 5' flanking region is bound by the EBF2 transcription factor, consistent with the evidence that this region bears the cognate deoxyribonucleic acid (DNA) consensus sequences. These data indicate that Contactin-1 expression is dependent upon EBF factors, suggesting that the Cntn1 gene belongs to the expanding regulatory cascade driven by these transcriptional regulators so that changes in its activation may contribute to the phenotype of Ebf2 null mutant mice.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cerebellum/growth & development , Cerebellum/metabolism , Contactin 1/metabolism , Trans-Activators/metabolism , Animals , Base Sequence , Basic Helix-Loop-Helix Transcription Factors/genetics , Blotting, Western , Cell Proliferation/physiology , Cerebellum/cytology , Choline O-Acetyltransferase/genetics , Choline O-Acetyltransferase/metabolism , Contactin 1/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Immunohistochemistry , Immunoprecipitation , Mice, Transgenic , Molecular Sequence Data , Promoter Regions, Genetic , Sequence Alignment , Transfection
10.
Mol Cell Neurosci ; 50(3-4): 221-37, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22579730

ABSTRACT

F3/Contactin is a neuronal surface glycoprotein, which plays a general role in neural development and, in particular, in neuronal and oligodendrocyte differentiation. In a previous study using the F3/EGFP transgenic mice, which express an EGFP reporter under control of the regulatory region from the mouse F3/Contactin gene, the activation of the F3/Contactin promoter was found to correlate with granule and Purkinje neuron differentiation in developing cerebellar cortex. Here we report that in developing cerebral cortex and basal ganglia the F3/Contactin gene is mostly activated during early commitment of neuronal precursors, thus indicating a region-specific profile of its developmental activation. We also report that, in the same structures of F3/EGFP mice, a downregulation of the endogenous F3/Contactin gene occurs, which correlates with upregulation of the dopaminergic phenotype and with locomotor pattern abnormalities. Therefore, F3/EGFP transgenic mice exhibit morphological and functional phenotypes recapitulating those arising from imbalance of the striatal dopaminergic pathway. As for the underlying mechanisms, we postulate that in F3/EGFP mice F3/Contactin downregulation results from the ability of transgene promoter sequences to interfere with the activation of the endogenous gene, thus realizing an F3/Contactin knockdown model, while dopaminergic upregulation is consistent with a general F3/Contactin inhibitory effect on the neuronal phenotype.


Subject(s)
Cerebral Cortex/metabolism , Contactin 1/genetics , Dopaminergic Neurons/metabolism , Promoter Regions, Genetic , Substantia Nigra/metabolism , Animals , Cerebral Cortex/growth & development , Contactin 1/metabolism , Gene Expression Regulation, Developmental , Green Fluorescent Proteins/genetics , Mice , Mice, Transgenic , Recombinant Fusion Proteins , Substantia Nigra/growth & development , Transcription, Genetic
11.
Dev Biol ; 365(1): 133-51, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22360968

ABSTRACT

The expression of the cell recognition molecule F3/Contactin (CNTN1) is generally associated with the functions of post-mitotic neurons. In the embryonic cortex, however, we find it expressed by proliferating ventricular zone (VZ) precursors. In contrast to previous findings in the developing cerebellum, F3/Contactin transgenic overexpression in the early cortical VZ promotes proliferation and expands the precursor pool at the expense of neurogenesis. At later stages, when F3/Contactin levels subside, however, neurogenesis resumes, suggesting that F3/Contactin expression in the VZ is inversely related to neurogenesis and plays a role in a feedback control mechanism, regulating the orderly progression of cortical development. The modified F3/Contactin profile therefore results in delayed corticogenesis, as judged by downregulation in upper and lower layer marker expression and by BrdU birth dating, indicating that, in this transgenic model, increased F3/Contactin levels counteract neuronal precursor commitment. These effects also occur in primary cultures and are reproduced by addition of an F3/Fc fusion protein to wild type cultures. Together, these data indicate a completely novel function for F3/Contactin. Parallel changes in the generation of the Notch Intracellular Domain and in the expression of the Hes-1 transcription factor indicate that activation of the Notch pathway plays a role in this phenotype, consistent with previous in vitro reports that F3/Contactin is a Notch1 ligand.


Subject(s)
Cerebral Cortex/embryology , Contactin 1/physiology , Neurogenesis , Animals , Animals, Genetically Modified , Cell Differentiation , Cerebral Cortex/cytology , Cerebral Cortex/physiology , Gene Expression Regulation, Developmental , Humans , Mice , Neurogenesis/genetics , Neurons/cytology , Neurons/physiology , Receptors, Notch/physiology , Signal Transduction
12.
Lab Invest ; 90(11): 1645-60, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20714324

ABSTRACT

In this study, we investigated the involvement of dystrophin-associated proteins (DAPs) and their relationship with the perivascular basement membrane in the brains of mdx mice and controls at the age of 2 months. We analyzed (1) the expression of glial DAPs α-ß-dystroglycan (DG), α-syntrophin, aquaporin-4 (AQP4) water channel, Kir 4.1 and dystrophin isoform (Dp71) by immunocytochemistry, laser confocal microscopy, immunogold electron microscopy, immunoblotting and RT-PCR; (2) the ultrastructure of the basement membrane and expression of laminin and agrin; and (3) the dual immunofluorescence colocalization of AQP4/α-ß-DG, and of Kir 4.1/agrin. The following results were observed in mdx brain as compared with controls: (1) a significant reduction in protein content and mRNA expression of DAPs; (2) ultrastructurally, a thickened and discontinuous appearance of the basement membrane and a significant reduction in laminin and agrin; and (3) a molecular rearrangment of α-ß-DG, coupled with a parallel loss of agrin and Kir 4.1 on basement membrane and glial endfeet. These data indicate that in mdx brain the deficiency in dystrophin and dystrophin isoform (Dp71) is coupled with a reduction of DAP components, coupled with an altered anchoring to the basement membrane.


Subject(s)
Agrin/analysis , Brain/metabolism , Dystrophin-Associated Proteins/analysis , Laminin/analysis , Muscular Dystrophy, Duchenne/metabolism , Animals , Aquaporin 4/analysis , Blotting, Western , Calcium-Binding Proteins/analysis , Disease Models, Animal , Down-Regulation , Dystroglycans/analysis , Fluorescent Antibody Technique , Membrane Proteins/analysis , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Microscopy, Confocal , Microscopy, Electron , Muscle Proteins/analysis , Muscular Dystrophy, Duchenne/pathology , Potassium Channels, Inwardly Rectifying/analysis
13.
Cell Adh Migr ; 3(1): 53-63, 2009.
Article in English | MEDLINE | ID: mdl-19372728

ABSTRACT

F3/Contactin is an immunoglobulin superfamily component expressed in the nervous tissue of several species. Here we focus on the structural and functional properties of its mouse relative, on the mechanisms driving its regulated expression and on its developmental role. F3/Contactin is differentially expressed in distinct populations of central and peripheral neurons and in some non-neuronal cells. Accordingly, the regulatory region of the underlying gene includes promoter elements undergoing differential activation, associated with an intricate splicing profile, indicating that transcriptional and posttranscriptional mechanisms contribute to its expression. Transgenic models allowed to follow F3/Contactin promoter activation in vivo and to modify F3/Contactin gene expression under a heterologous promoter, which resulted in morphological and functional phenotypes. Besides axonal growth and pathfinding, these concerned earlier events, including precursor proliferation and commitment. This wide role in neural ontogenesis is consistent with the recognized interaction of F3/Contactin with developmental control genes belonging to the Notch pathway.


Subject(s)
Cell Adhesion Molecules, Neuronal/chemistry , Cell Adhesion Molecules, Neuronal/metabolism , Gene Expression Regulation, Developmental , Glycoproteins/chemistry , Glycoproteins/metabolism , Animals , Cell Adhesion Molecules, Neuronal/genetics , Contactins , Glycoproteins/genetics , Mice
14.
Brain Pathol ; 17(4): 399-406, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17784876

ABSTRACT

In Duchenne muscular dystrophy (DMD) metabolic and structural alterations of the central nervous system are described. Here, we investigated in the brain of 10 mdx mice and in five control ones, the expression of hypoxia inducible factor-1alpha (HIF-1alpha) and we correlated it with the expression of vascular endothelial growth factor (VEGF) and vascular endothelial growth factor receptor-2 (VEGFR-2) and of the endothelial tight junction proteins zonula occludens-1 (ZO-1) and claudin-1. Results showed an activation of mRNA HIF-1alpha by reverse transcription polymerase chain reaction (RT-PCR) and a strong HIF1-alpha labeling of perivascular glial cells and cortical neurons by immunohistochemistry, in mdx mouse. Moreover, overexpression of VEGF and VEGFR-2, respectively, in neurons and in endothelial cells coupled with changes to endothelial ZO-1 and claudin-1 expression in the latter were detected by immunoblotting and immunohistochemistry, in the mdx brain. Furthermore, by immunoprecipitation, an up-phosphorylation of ZO-1 was demonstrated in mdx endothelial cells in parallel with the reduction in ZO-1 protein content. These data suggest that the activation of HIF-1alpha in the brain of dystrophic mice coupled with VEGF and VEGFR-2 up-regulation and ZO-1 and claudin-1 rearrangement might contribute to both blood-brain barrier opening and increased angiogenesis.


Subject(s)
Brain Diseases, Metabolic, Inborn/metabolism , Brain/metabolism , Hypoxia-Inducible Factor 1/metabolism , Membrane Proteins/metabolism , Muscular Dystrophy, Duchenne/complications , Muscular Dystrophy, Duchenne/metabolism , Phosphoproteins/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/physiopathology , Brain/physiopathology , Brain Diseases, Metabolic, Inborn/genetics , Brain Diseases, Metabolic, Inborn/physiopathology , Claudin-1 , Disease Models, Animal , Endothelial Cells/metabolism , Female , Hypoxia-Inducible Factor 1/genetics , Immunohistochemistry , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Muscular Dystrophy, Duchenne/genetics , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/physiopathology , Neuroglia/metabolism , Neurons/metabolism , Phosphoproteins/genetics , Phosphorylation , RNA, Messenger/metabolism , Up-Regulation/physiology , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism , Zonula Occludens-1 Protein
15.
Mol Cell Neurosci ; 32(4): 403-18, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16857383

ABSTRACT

In this study, we address the activation profile of the gene encoding the mouse axonal glycoprotein F3/Contactin. Promoter sequences previously characterized in vitro are used to drive an Enhanced Green Fluorescent Protein reporter in transgenic mice. In developing cerebellum, differential transgene expression occurs within distinct cell populations. At P0 the transgene is activated in postmitotic granule neurons undergoing radial migration, a sharp upregulation occurring at P6-P8, with a gradual decline from this stage onward. In Purkinje cells, promoter activation, first detected at P3, peaks at around P6 and is fully downregulated by P16. The transgene is also expressed in Ng2- and O4-positive cells, mostly at the end of the first postnatal week, suggesting correlation with early oligodendrocyte differentiation. These data indicate that the complex organization of the regulatory region of the F3/Contactin gene is necessary for directing its articulated expression in different neural cells types and for its developmental function.


Subject(s)
Cell Adhesion Molecules, Neuronal/metabolism , Cerebellum , Gene Expression Regulation, Developmental/physiology , Neurons/metabolism , Age Factors , Animals , Animals, Newborn , Antigens/metabolism , Cell Adhesion Molecules, Neuronal/genetics , Cells, Cultured , Cerebellum/cytology , Cerebellum/growth & development , Cerebellum/metabolism , Contactins , Enzyme Activation/physiology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Immunohistochemistry/methods , Mice , Mice, Transgenic , Neurons/classification , O Antigens/metabolism , Proteoglycans/metabolism , Tubulin/metabolism
16.
Brain Res ; 1013(2): 256-9, 2004 Jul 09.
Article in English | MEDLINE | ID: mdl-15193536

ABSTRACT

In this study, for the first time, we investigated about the localization of VEGF-A, VEGFR-2 and Ang-2 in the choroid plexuses of the adult mouse by Western blot and immunohistochemistry. Results showed that VEGF-A stained epithelial cells, while anti-VEGFR-2 and -Ang-2 antibodies stained endothelial cells. These data suggest that Ang-2, converting blood vessels into a more plastic and immature phenotype, would provide more accessibility of VEGF-A to endothelial cells.


Subject(s)
Angiopoietin-2/metabolism , Choroid Plexus/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Age Factors , Animals , Immunohistochemistry , Mice
17.
Neurochem Res ; 29(3): 547-60, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15038602

ABSTRACT

A study is presented on the expression and activity of complex I, as well as of other complexes of the respiratory chain, in the course of brain development and inherited encephalopathies. Investigations on mouse hippocampal cells show that differentiation of these cells both in vivo and in cell cultures is associated with the expression of a functional complex I, whose activity markedly increases with respect to that of complexes III and IV. Data are presented on genetic defects of complex I in six children with inborn encephalopathy associated with isolated deficiency of the complex. Mutations have been identified in nuclear and mitochondrial genes coding for subunits of the complex. Different mutations were found in the nuclear NDUFS4 gene coding for the 18 kD (IP, AQDQ) subunit of complex I. All the NDUFS4 mutations resulted in impairment of the assembly of a functional complex. The observations presented provide evidence showing a critical role of complex I in differentiation and functional activity of brain cells.


Subject(s)
Chromosome Mapping , Electron Transport Complex I/genetics , Hippocampus/enzymology , Mutation , Animals , Cell Differentiation , DNA, Complementary/genetics , Disease Models, Animal , Hippocampus/cytology , Humans , Mice , Mitochondrial Encephalomyopathies/enzymology , Mitochondrial Encephalomyopathies/genetics , Neurons/cytology , Neurons/enzymology
18.
Brain Res ; 989(1): 9-15, 2003 Oct 31.
Article in English | MEDLINE | ID: mdl-14519506

ABSTRACT

The repair and regeneration of injured tissues and organs depend on the re-establishment of the blood flow needed for cellular infiltration and metabolic support. Among the various materials used in tissue reconstruction, acellular scaffolds have recently been utilized. In this study, we investigated the angiogenic response induced by acellular brain scaffolds implanted in vivo onto the chick embryo chorioallantoic membrane (CAM), a useful model for such investigations. The results show that acellular brain scaffolds are able to induce a strong angiogenic response, comparable to that of fibroblast growth factor-2 (FGF-2), a well known angiogenic cytokine. The response may be considered dependent on a direct angiogenic effect exerted by the scaffold, because no inflammatory infiltrate was detectable in CAM's mesenchyme beneath the implant. Acellular brain scaffolds might induce the release of endogenous angiogenic factors, such as FGF-2 and vascular endothelial growth factor (VEGF) released from the extracellular matrix of the developing CAM. In addition, the angiogenic response may depend, in part, also on the presence in the acellular matrix of transforming growth factor beta 1 (TGFbeta1).


Subject(s)
Allantois/transplantation , Brain Tissue Transplantation , Brain/physiology , Chorion/transplantation , Neovascularization, Physiologic/physiology , Allantois/cytology , Allantois/physiology , Animals , Brain/cytology , Brain Tissue Transplantation/physiology , Chick Embryo , Chorion/cytology , Chorion/physiology , Rats , Rats, Sprague-Dawley , Transplants
19.
Glia ; 42(3): 235-51, 2003 May.
Article in English | MEDLINE | ID: mdl-12673830

ABSTRACT

In this study, we investigated the involvement of the blood-brain barrier (BBB) in the brain of the dystrophin-deficient mdx mouse, an experimental model of Duchenne muscular dystrophy (DMD). To this purpose, we used two tight junction markers, the Zonula occludens (ZO-1) and claudin-1 proteins, and a glial marker, the aquaporin-4 (AQP4) protein, whose expression is correlated with BBB differentiation and integrity. Results showed that most of the brain microvessels in mdx mice were lined by altered endothelial cells that showed open tight junctions and were surrounded by swollen glial processes. Moreover, 18% of the perivascular glial endfeet contained electron-dense cellular debris and were enveloped by degenerating microvessels. Western blot showed a 60% reduction in the ZO-1 protein content in mdx mice and a similar reduction in AQP4 content compared with the control brain. ZO-1 immunocytochemistry and claudin-1 immunofluorescence in mdx mice revealed a diffuse staining of microvessels as compared with the control ones, which displayed a banded staining pattern. ZO-1 immunogold electron microscopy showed unlabeled tight junctions and the presence of gold particles scattered in the endothelial cytoplasm in the mdx mice, whereas ZO-1 gold particles were exclusively located at the endothelial tight junctions in the controls. Dual immunofluorescence staining of alpha-actin and ZO-1 revealed colocalization of these proteins. As in ZO-1 staining, the pattern of immunolabeling with anti-alpha-actin antibody was diffuse in the mdx vessels and pointed or banded in the controls. alpha-actin immunogold electron microscopy showed gold particles in the cytoplasms of endothelial cells and pericytes in the mdx mice, whereas alpha-actin gold particles were revealed on the endothelial tight junctions and the cytoskeletal microfilaments of pericytes in the controls. Perivascular glial processes of the mdx mice appeared faintly stained by anti-AQP4 antibody, while in the controls a strong AQP4 labeling of glial processes was detected at light and electron microscope level. The vascular permeability of the mdx brain microvessels was investigated by means of the horseradish peroxidase (HRP). After HRP injection, extensive perivascular areas of marker escape were observed in mdx mice, whereas HRP was exclusively intravascularly localized in the controls. Inflammatory cells, CD4-, CD8-, CD20-, and CD68-positive cells, were not revealed in the perivascular stroma of the mdx brain. These findings indicate that dystrophin deficiency in the mdx brain leads to severe injury of the endothelial and glial cells with disturbance in alpha-actin cytoskeleton, ZO-1, claudin-1, and AQP4 assembly, as well as BBB breakdown. The BBB alterations suggest that changes in vascular permeability are involved in the pathogenesis of the neurological dysfunction associated with DMD.


Subject(s)
Astrocytes/metabolism , Blood-Brain Barrier/genetics , Brain/blood supply , Brain/metabolism , Endothelium, Vascular/metabolism , Muscular Dystrophy, Duchenne/metabolism , Tight Junctions/metabolism , Actins/metabolism , Animals , Antigens, Surface/metabolism , Aquaporin 4 , Aquaporins/metabolism , Astrocytes/pathology , Astrocytes/ultrastructure , Biomarkers , Brain/physiopathology , Claudin-1 , Disease Models, Animal , Down-Regulation/genetics , Endothelium, Vascular/pathology , Endothelium, Vascular/physiopathology , Female , Horseradish Peroxidase , Immunohistochemistry , Membrane Proteins/metabolism , Mice , Mice, Inbred mdx , Microscopy, Electron , Muscular Dystrophy, Duchenne/pathology , Muscular Dystrophy, Duchenne/physiopathology , Phosphoproteins/metabolism , Tight Junctions/pathology , Tight Junctions/ultrastructure , Zonula Occludens-1 Protein
20.
Brain Res ; 953(1-2): 12-6, 2002 Oct 25.
Article in English | MEDLINE | ID: mdl-12384233

ABSTRACT

Recent data have demonstrated that vascular endothelial growth factor (VEGF) is expressed by subsets of neurons, coincident with angiogenesis within its developing cerebral cortex. In this study, with the aim of elucidating the mechanisms of vascular involvement during brain impairment in Duchenne muscular distrophy (DMD), we have correlated the vascular density with VEGF and VEGF receptor-2 (VEGFR-2) expression in the brain cortex of normal and mdx mouse, an animal model with a genetic defect in a region homologous with the human DMD gene. Results showed that in mdx mouse, tissue area occupied by microvessels positive to factor VIII related antigen and VEGFR-2 increased in parallel to the tissue area occupied by neurons positive to VEGF. Our data suggest that increased vascularity in the brain of mdx mouse may be due, at least in part, to proliferation of endothelial cells in response to VEGF secreted by neuronal cells.


Subject(s)
Cerebral Cortex/chemistry , Endothelial Growth Factors/analysis , Intercellular Signaling Peptides and Proteins/analysis , Lymphokines/analysis , Muscular Dystrophy, Animal/metabolism , Vascular Endothelial Growth Factor Receptor-2/analysis , Animals , Factor VIII/analysis , Female , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Neovascularization, Pathologic , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors , von Willebrand Factor/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...