Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Transl Stroke Res ; 15(2): 411-421, 2024 04.
Article in English | MEDLINE | ID: mdl-36723700

ABSTRACT

Cerebral cavernous malformation (CCM) is a vascular disease that affects the central nervous system, which familial form is due to autosomal dominant mutations in the genes KRIT1(CCM1), MGC4607(CCM2), and PDCD10(CCM3). Patients affected by the PDCD10 mutations usually have the onset of symptoms at an early age and a more aggressive phenotype. The aim of this study is to investigate the molecular mechanism involved with CCM3 disease pathogenesis. Herein, we report two typical cases of CCM3 phenotype and compare the clinical and neuroradiological findings with five patients with a familial form of KRIT1 or CCM2 mutations and six patients with a sporadic form. In addition, we evaluated the PDCD10 gene expression by qPCR and developed a bioinformatic pipeline to understand the structural changes of mutations. The two CCM3 patients had an early onset of symptoms and a high lesion burden. Furthermore, the sequencing showed that Patient 1 had a frameshift mutation in c.222delT; p.(Asn75Thrfs*14) that leads to lacking the last 124 C-terminal amino acids on its primary structure and Patient 2 had a variant on the splicing site region c.475-2A > G. The mRNA expression was fourfold lower in both patients with PDCD10 mutation. Using in silico analysis, we identify that the frameshift mutation transcript lacks the C-terminal FAT-homology domain compared to the wild-type PDCD10 and preserves the N-terminal dimerization domain. The two patients studied here allow estimating the potential impact of mutations in clinical interpretation as well as support to better understand the mechanism and pathogenesis of CCM3.


Subject(s)
Hemangioma, Cavernous, Central Nervous System , Humans , Central Nervous System , Hemangioma, Cavernous, Central Nervous System/diagnostic imaging , Hemangioma, Cavernous, Central Nervous System/genetics , Mutation/genetics , Phenotype , Proto-Oncogene Proteins/genetics
2.
J Neurochem ; 162(6): 501-513, 2022 09.
Article in English | MEDLINE | ID: mdl-35797221

ABSTRACT

Glioblastoma is the most common and malignant type of primary brain tumor. Previous studies have shown that alterations in centrosome amplification and its components are frequently found in treatment-resistant tumors and may be associated with tumor progression. A centrosome protein essential for centrosome biogenesis is the centromere protein J (CENPJ), known to control the proliferation of neural progenitors and hepatocarcinoma cells, and also neuronal migration. However, it remains unknown the role of CENPJ in glioblastoma. Here we show that CENPJ is overexpressed in human glioblastoma cell lines in comparison to human astrocytes. Using bioinformatics analysis, we find that high Cenpj expression is associated with poor prognosis in glioma patients. Examining Cenpj loss of function in glioblastoma by siRNA transfection, we find impairments in cell proliferation and migration. Using a Cenpj mutant version with the deleted PN2-3 or TCP domain, we found that a conserved PN2-3 region is required for glioblastoma migration. Moreover, Cenpj downregulation modulates glioblastoma morphology resulting in microtubules stabilization and actin filaments depolymerization. Altogether, our findings indicate that CENPJ controls relevant aspects of glioblastoma progression and might be a target for therapeutic intervention and a biomarker for glioma malignancy.


Subject(s)
Brain Neoplasms , Glioblastoma , Glioma , Brain Neoplasms/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Centromere/metabolism , Centromere/pathology , Gene Expression Regulation, Neoplastic , Glioblastoma/metabolism , Glioma/metabolism , Humans
3.
Curr Neurovasc Res ; 18(2): 172-180, 2021.
Article in English | MEDLINE | ID: mdl-34082682

ABSTRACT

BACKGROUND: Cerebral Cavernous Malformations (CCM) predispose patients to a lifetime risk of seizures and symptomatic hemorrhage. Only a small percentage of people affected will develop clinical symptoms and the molecular mechanisms underlying lesional activity remain unclear. We analyzed a panel of Single Nucleotide Polymorphisms (SNPs) in CCM patients. We looked for plasmatic inflammatory cytokines, checking for a pattern of plasma expression heterogeneity and any correlation with genetic variations identified with different CCM clinical phenotypes. METHODS: This was a case-control study from a long-term follow-up cohort including 23 CCM patients, of which 16 were symptomatic, and 7 were asymptomatic. A 200-SNP panel was considered through next-generation sequencing and 18 different plasma molecules were assessed through a suspension array system. RESULTS: Fcγ receptor IIa rs1801274 (FCGR2A) and protein tyrosine phosphatase non-receptor type 2 rs72872125 PTPN2 were statistically different between groups. Patients who had a combination of the presence of FCGR2A and the absence of PTPN2 also had symptoms earlier in life. The combination of genetic polymorphisms and serum level of GM-CSF showed the best diagnostic biomarker to distinguish symptomatic patients as formulated: [0.296*(FCGR2A)] + [-0.788*(PTPN2)] + [-0.107*(GM-CSF)]. CONCLUSION: We have shown that SNPs in inflammation genes might be related to a symptomatic phenotype in CCM. We also demonstrated that a formula based on two of these polymorphisms (FCGR2A+ and PTPN2+) is possibly capable of predicting a symptomatic phenotype during a patient's lifetime.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Hemangioma, Cavernous, Central Nervous System/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 2/genetics , Receptors, IgG/genetics , Adult , Aged , Case-Control Studies , Female , Genetic Markers , High-Throughput Nucleotide Sequencing , Humans , Male , Middle Aged , Young Adult
4.
World Neurosurg ; 131: 87-89, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31356970

ABSTRACT

BACKGROUND: Intraorbital and intracerebral cavernous malformation (CM) lesions are considered independent entities. Purely cerebral CMs have variable biology with recent evidence depicting inflammation as an important player and a risk factor for aggressiveness. We describe a case of concomitant left intraaxial and extraaxial CMs, linked by the ipsilateral basal vein, where the extraaxial component has developed an aggressive behavior. CASE DESCRIPTION: A 35-year-old female patient presented with a rapid and progressive exophthalmos and loss of vision on the left eye. Cranial magnetic resonance and angiography examinations demonstrated a left craniofacial CM and large intraorbital component. The lesion was connected through a large basal vein to a cerebral intraventricular CM. Transconjunctival resection showed typical findings of CM. A complete histopathology and immunostaining analysis was performed and revealed a clear acute lymphomononuclear reaction with a predominant immune cellular inflammation. CONCLUSIONS: A case of intraorbital and extracranial cavernomatous mass, connected to a cerebral intraventricular CM through a large basal vein, has presented with an aggressive course. A complete histopathologic and immunohistochemical analysis of the orbital mass has pictured a clear immune-cellular inflammatory reaction adding to the amounting evidence of association between inflammation and site aggressiveness in the setting of CMs.


Subject(s)
Cerebral Veins/diagnostic imaging , Cerebral Ventricle Neoplasms/diagnostic imaging , Hemangioma, Cavernous, Central Nervous System/diagnostic imaging , Neoplasms, Multiple Primary/diagnostic imaging , Orbital Neoplasms/diagnostic imaging , Adult , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Cerebral Angiography , Female , Hemangioma, Cavernous, Central Nervous System/immunology , Hemangioma, Cavernous, Central Nervous System/pathology , Hemangioma, Cavernous, Central Nervous System/surgery , Humans , Inflammation/immunology , Inflammation/pathology , Macrophages/immunology , Macrophages/pathology , Magnetic Resonance Imaging , Neoplasms, Multiple Primary/immunology , Neoplasms, Multiple Primary/pathology , Neoplasms, Multiple Primary/surgery , Orbital Neoplasms/immunology , Orbital Neoplasms/pathology , Orbital Neoplasms/surgery , Plasma Cells/immunology , Plasma Cells/pathology
5.
J Neurochem ; 150(2): 138-157, 2019 07.
Article in English | MEDLINE | ID: mdl-31009074

ABSTRACT

Parkinson's disease (PD) is characterized by selective death of dopaminergic neurons in the substantia nigra, degeneration of the nigrostriatal pathway, increases in glutamatergic synapses in the striatum and aggregation of α-synuclein. Evidence suggests that oligomeric species of α-synuclein (αSO) are the genuine neurotoxins of PD. Although several studies have supported the direct neurotoxic effects of αSO on neurons, their effects on astrocytes have not been directly addressed. Astrocytes are essential to several steps of synapse formation and function, including secretion of synaptogenic factors, control of synaptic elimination and stabilization, secretion of neural/glial modulators, and modulation of extracellular ions, and neurotransmitter levels in the synaptic cleft. Here, we show that αSO induced the astrocyte reactivity and enhanced the synaptogenic capacity of human and murine astrocytes by increasing the levels of the known synaptogenic molecule transforming growth factor beta 1 (TGF-ß1). Moreover, intracerebroventricular injection of αSO in mice increased the number of astrocytes, the density of excitatory synapses, and the levels of TGF-ß1 in the striatum of injected animals. Inhibition of TGF-ß1 signaling impaired the effect of the astrocyte-conditioned medium on glutamatergic synapse formation in vitro and on striatal synapse formation in vivo, whereas addition of TGF-ß1 protected mesencephalic neurons against synapse loss triggered by αSO. Together, our data suggest that αSO have important effects on astrocytic functions and describe TGF-ß1 as a new endogenous astrocyte-derived molecule involved in the increase in striatal glutamatergic synaptic density present in early stages of PD. OPEN SCIENCE BADGES: This article has received a badge for *Open Materials* because it provided all relevant information to reproduce the study in the manuscript. The complete Open Science Disclosure form for this article can be found at the end of the article. More information about the Open Practices badges can be found at https://cos.io/our-services/open-science-badges/. Cover Image for this issue: doi: 10.1111/jnc.14514.


Subject(s)
Astrocytes/metabolism , Parkinsonian Disorders/metabolism , Synapses/metabolism , Transforming Growth Factor beta1/metabolism , alpha-Synuclein/metabolism , Animals , Disease Models, Animal , Humans , Mice , Neurogenesis/physiology , Signal Transduction/physiology
6.
Mol Neurobiol ; 55(8): 6816-6833, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29349577

ABSTRACT

Glioblastoma (GBM) is the most common and aggressive malignant primary brain tumor and still lacks effective therapeutic strategies. It has already been shown that old drugs like sulfasalazine (SAS) and valproic acid (VPA) present antitumoral activities in glioma cell lines. SAS has also been associated with a decrease of intracellular glutathione (GSH) levels through a potent inhibition of xc- glutamate/cystine exchanger leading to an antioxidant deprotection. In the same way, VPA was recently identified as a histone deacetylase (HDAT) inhibitor capable of activating tumor suppression genes. As both drugs are widely used in clinical practice and their profile of adverse effects is well known, the aim of our study was to investigate the effects of the combined treatment with SAS and VPA in GBM cell lines. We observed that both drugs were able to reduce cell viability in a dose-dependent manner and the combined treatment potentiated these effects. Combined treatment also increased cell death and inhibited proliferation of GBM cells, while having no effect on human and rat cultured astrocytes. Also, we observed high protein expression of the catalytic subunit of xc- in all the examined GBM cell lines, and treatment with SAS blocked its activity and decreased intracellular GSH levels. Noteworthy, SAS but not VPA was also able to reduce the [14C]-ascorbate uptake. Together, these data indicate that SAS and VPA exhibit a substantial effect on GBM cell's death related to an intracellular oxidative response imbalance, making this combination of drugs a promising therapeutic strategy.


Subject(s)
Brain Neoplasms/pathology , Glioblastoma/pathology , Intracellular Space/metabolism , Sulfasalazine/pharmacology , Valproic Acid/pharmacology , Amino Acid Transport System y+/metabolism , Animals , Ascorbic Acid/metabolism , Cell Death/drug effects , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Therapy, Combination , Glutathione/metabolism , Humans , Mesoderm/drug effects , Mesoderm/pathology , Neuroglia/drug effects , Neuroglia/metabolism , Neuroglia/pathology , Oxidation-Reduction , Rats , Time Factors
7.
Oncol Rep ; 38(3): 1341-1352, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28714013

ABSTRACT

Glioblastoma (GBM) is a grade IV astrocytoma. GBM patients show resistance to chemotherapy such as temozolomide (TMZ), the gold standard treatment. In order to simulate the molecular mechanisms behind the different chemotherapeutic responses in GBM patients we compared the cellular heterogeneity and chemotherapeutic resistance mechanisms in different GBM cell lines. We isolated and characterized a human GBM cell line obtained from a GBM patient, named GBM11. We studied the GBM11 behaviour when treated with Tamoxifen (TMX) that, among other functions, is a protein kinase C (PKC) inhibitor, alone and in combination with TMZ in comparison with the responses of U87 and U118 human GBM cell lines. We evaluated the cell death, cell cycle arrest and cell proliferation, mainly through PKC expression, by flow cytometry and western blot analysis and, ultimately, cell migration capability and f-actin filament disorganization by fluorescence microscopy. We demonstrated that the constitutive activation of p-PKC seems to be one of the main metabolic implicated on GBM malignancy. Despite of its higher resistance, possibly due to the overexpression of P-glycoprotein and stem-like cell markers, GBM11 cells presented a subtle different chemotherapeutic response compared to U87 and U118 cells. The GBM11, U87, U118 cell lines show subtle molecular differences, which clearly indicate the characterization of GBM heterogeneity, one of the main reasons for tumor resistance. The adding of cellular heterogeneity in molecular behaviour constitutes a step closer in the understanding of resistant molecular mechanisms in GBM, and can circumvents the eventual impaired therapy.


Subject(s)
Astrocytoma/drug therapy , Genetic Heterogeneity , Glioblastoma/drug therapy , Protein Kinase C/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Apoptosis/drug effects , Astrocytoma/genetics , Astrocytoma/pathology , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Dacarbazine/administration & dosage , Dacarbazine/analogs & derivatives , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/drug effects , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Neoplasm Grading , Signal Transduction/drug effects , Tamoxifen/administration & dosage , Temozolomide
8.
Sci Rep ; 7: 45091, 2017 03 27.
Article in English | MEDLINE | ID: mdl-28345587

ABSTRACT

Astrocytes play a critical role in the development and homeostasis of the central nervous system (CNS). Astrocyte dysfunction results in several neurological and degenerative diseases. However, a major challenge to our understanding of astrocyte physiology and pathology is the restriction of studies to animal models, human post-mortem brain tissues, or samples obtained from invasive surgical procedures. Here, we report a protocol to generate human functional astrocytes from cerebral organoids derived from human pluripotent stem cells. The cellular isolation of cerebral organoids yielded cells that were morphologically and functionally like astrocytes. Immunolabelling and proteomic assays revealed that human organoid-derived astrocytes express the main astrocytic molecular markers, including glutamate transporters, specific enzymes and cytoskeletal proteins. We found that organoid-derived astrocytes strongly supported neuronal survival and neurite outgrowth and responded to ATP through transient calcium wave elevations, which are hallmarks of astrocyte physiology. Additionally, these astrocytes presented similar functional pathways to those isolated from adult human cortex by surgical procedures. This is the first study to provide proteomic and functional analyses of astrocytes isolated from human cerebral organoids. The isolation of these astrocytes holds great potential for the investigation of developmental and evolutionary features of the human brain and provides a useful approach to drug screening and neurodegenerative disease modelling.


Subject(s)
Astrocytes/cytology , Cerebral Cortex/cytology , Neuronal Outgrowth , Organoids/cytology , Animals , Astrocytes/metabolism , Calcium Signaling , Cells, Cultured , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Glutamic Acid/metabolism , Humans , Mice , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Proteome/genetics , Proteome/metabolism
10.
World Neurosurg ; 86: 514.e19-22, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26548819

ABSTRACT

BACKGROUND: Cavernous malformation (CM) is a vascular malformation found in the encephalic parenchyma, spinal cord, nerve roots, and extraneural tissue. CM in the trigeminal distribution is exquisitely uncommon and its biological behavior not completely understood. The clinical picture might be diverse, depending on the affected sector of the trigeminal architecture, and literature debating its pathobiology is scarce. CASE DESCRIPTION: We describe a case of 56-year-old woman who presented with left trigeminal neuralgia and a rapidly growing cavernous malformation of the entire distribution of the fifth nerve. The clinical picture evolved to a progressive gait ataxia and follow-up neuroimaging showed a large intracranial mass leading to a brainstem compression. After microsurgical resection, the mass proved to be a typical CM of the trigeminal root. CONCLUSION: We present an uncommonly aggressive progression of a CM of the trigeminal root, Gasserian ganglion, and cavernous sinus evolving to severe brainstem compression. The documentation of this unique case as well as its management is presented is discussed.


Subject(s)
Brain Neoplasms/complications , Brain Neoplasms/diagnosis , Hemangioma, Cavernous, Central Nervous System/complications , Hemangioma, Cavernous, Central Nervous System/diagnosis , Trigeminal Neuralgia/etiology , Brain Neoplasms/surgery , Female , Hemangioma, Cavernous, Central Nervous System/surgery , Humans , Middle Aged , Trigeminal Neuralgia/diagnosis , Trigeminal Neuralgia/therapy
11.
Genet Med ; 17(3): 188-196, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25122144

ABSTRACT

PURPOSE: The phenotypic manifestations of cerebral cavernous malformation disease caused by rare PDCD10 mutations have not been systematically examined, and a mechanistic link to Rho kinase-mediated hyperpermeability, a potential therapeutic target, has not been established. METHODS: We analyzed PDCD10 small interfering RNA-treated endothelial cells for stress fibers, Rho kinase activity, and permeability. Rho kinase activity was assessed in cerebral cavernous malformation lesions. Brain permeability and cerebral cavernous malformation lesion burden were quantified, and clinical manifestations were assessed in prospectively enrolled subjects with PDCD10 mutations. RESULTS: We determined that PDCD10 protein suppresses endothelial stress fibers, Rho kinase activity, and permeability in vitro. Pdcd10 heterozygous mice have greater lesion burden than other Ccm genotypes. We demonstrated robust Rho kinase activity in murine and human cerebral cavernous malformation vasculature and increased brain vascular permeability in humans with PDCD10 mutation. Clinical phenotype is exceptionally aggressive compared with the more common KRIT1 and CCM2 familial and sporadic cerebral cavernous malformation, with greater lesion burden and more frequent hemorrhages earlier in life. We first report other phenotypic features, including scoliosis, cognitive disability, and skin lesions, unrelated to lesion burden or bleeding. CONCLUSION: These findings define a unique cerebral cavernous malformation disease with exceptional aggressiveness, and they inform preclinical therapeutic testing, clinical counseling, and the design of trials.Genet Med 17 3, 188-196.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Central Nervous System Neoplasms/pathology , Hemangioma, Cavernous, Central Nervous System/pathology , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Mutation , Proto-Oncogene Proteins/genetics , rho-Associated Kinases/metabolism , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Adolescent , Adult , Animals , Apoptosis Regulatory Proteins/metabolism , Carrier Proteins/genetics , Cells, Cultured , Central Nervous System Neoplasms/enzymology , Central Nervous System Neoplasms/genetics , Child , Child, Preschool , Disease Models, Animal , Hemangioma, Cavernous, Central Nervous System/enzymology , Hemangioma, Cavernous, Central Nervous System/genetics , Human Umbilical Vein Endothelial Cells , Humans , Infant , Intracellular Signaling Peptides and Proteins/metabolism , Keratin-1/genetics , Membrane Proteins/metabolism , Mice , Middle Aged , Prospective Studies , Proto-Oncogene Proteins/metabolism , Stress Fibers/drug effects , Stress Fibers/metabolism , Young Adult , rho-Associated Kinases/antagonists & inhibitors
12.
BMC Cancer ; 14: 923, 2014 Dec 08.
Article in English | MEDLINE | ID: mdl-25482099

ABSTRACT

BACKGROUND: Glioblastoma (GBM) is the most common primary brain tumor and the most aggressive glial tumor. This tumor is highly heterogeneous, angiogenic, and insensitive to radio- and chemotherapy. Here we have investigated the progression of GBM produced by the injection of human GBM cells into the brain parenchyma of immunocompetent mice. METHODS: Xenotransplanted animals were submitted to magnetic resonance imaging (MRI) and histopathological analyses. RESULTS: Our data show that two weeks after injection, the produced tumor presents histopathological characteristics recommended by World Health Organization for the diagnosis of GBM in humans. The tumor was able to produce reactive gliosis in the adjacent parenchyma, angiogenesis, an intense recruitment of macrophage and microglial cells, and presence of necrosis regions. Besides, MRI showed that tumor mass had enhanced contrast, suggesting a blood-brain barrier disruption. CONCLUSIONS: This study demonstrated that the xenografted tumor in mouse brain parenchyma develops in a very similar manner to those found in patients affected by GBM and can be used to better understand the biology of GBM as well as testing potential therapies.


Subject(s)
Brain Neoplasms/pathology , Brain/pathology , Disease Models, Animal , Glioblastoma/pathology , Tumor Microenvironment , Animals , Brain/blood supply , Brain Neoplasms/complications , Glioblastoma/complications , Glioblastoma/physiopathology , Gliosis/etiology , Humans , Immunocompetence , Macrophage Activation , Magnetic Resonance Imaging , Male , Mice , Microglia/physiology , Necrosis/etiology , Neovascularization, Pathologic/etiology , Transplantation, Heterologous
13.
Brain Res ; 1538: 172-81, 2013 Nov 13.
Article in English | MEDLINE | ID: mdl-24095794

ABSTRACT

Inflammation influences the pathogenesis of seizures by boosting neuronal degeneration of temporal lobe epilepsy with hippocampal sclerosis (TLE-HS). This work aimed to determine the activity of metalloproteases (MMPs) in brain tissue fragments of TLE-HS patients and the effect of lobectomy on circulating inflammatory biomarkers. Surgical fragments (n=4) from epileptogenic focus (EF) e perilesion area (PL), and control hippocampus from autopsy (n=5) were processed for glial protein (GFAP), activated microglia (IB4) immunohistochemistry, and metalloprotease activity (MMP-2, -9). Perilesional area showed GFAP positive cells with morphology of activate astrocyte and reactive gliosis nearby the lesion. In the lesion foci, astrocytes had altered cytoarchitecture with disorganized stroma suggestive of necrosis, and numerous mononuclear cells with few projections and morphological characteristics of activate microglia. Analysis of MMP-9 and MMP-2 in the sera before and after hippocampectomy confirmed the inflammatory pattern of TLE-HS, with high MMP-9 activity; high MMP-9/TIMP-1 and urokinase uPAR plasma levels before lobectomy but low after surgery. Maintenance of MMP-2 activity indicates persistent tissue remodeling in both groups. The present work shows that patients with chronic and medically intractable TLE-HS that undergone amigdalo-hippocampectomy for removal of epileptogenic lesion had a clinical enduring benefit of lack seizure recurrence for up to a year, and consistent reduction of proteases (MMP-9 and uPAR) activation that participate as important inflammatory epileptogenic inducers.


Subject(s)
Epilepsy, Temporal Lobe/metabolism , Epilepsy, Temporal Lobe/pathology , Hippocampus/metabolism , Hippocampus/pathology , Metalloproteases/blood , Receptors, Urokinase Plasminogen Activator/blood , Adolescent , Adult , Anterior Temporal Lobectomy , Cytokines/blood , Encephalitis/metabolism , Epilepsy, Temporal Lobe/blood , Epilepsy, Temporal Lobe/enzymology , Female , Humans , Male , Middle Aged , Tissue Inhibitor of Metalloproteinases/blood , Young Adult
14.
PLoS One ; 8(1): e55605, 2013.
Article in English | MEDLINE | ID: mdl-23383241

ABSTRACT

Connective-tissue growth factor (CTGF/CCN2) is a matricellular-secreted protein involved in complex processes such as wound healing, angiogenesis, fibrosis and metastasis, in the regulation of cell proliferation, migration and extracellular matrix remodeling. Glioblastoma (GBM) is the major malignant primary brain tumor and its adaptation to the central nervous system microenvironment requires the production and remodeling of the extracellular matrix. Previously, we published an in vitro approach to test if neurons can influence the expression of the GBM extracellular matrix. We demonstrated that neurons remodeled glioma cell laminin. The present study shows that neurons are also able to modulate CTGF expression in GBM. CTGF immnoreactivity and mRNA levels in GBM cells are dramatically decreased when these cells are co-cultured with neonatal neurons. As proof of particular neuron effects, neonatal neurons co-cultured onto GBM cells also inhibit the reporter luciferase activity under control of the CTGF promoter, suggesting inhibition at the transcription level. This inhibition seems to be contact-mediated, since conditioned media from embryonic or neonatal neurons do not affect CTGF expression in GBM cells. Furthermore, the inhibition of CTGF expression in GBM/neuronal co-cultures seems to affect the two main signaling pathways related to CTGF. We observed inhibition of TGFß luciferase reporter assay; however phopho-SMAD2 levels did not change in these co-cultures. In addition levels of phospho-p44/42 MAPK were decreased in co-cultured GBM cells. Finally, in transwell migration assay, CTGF siRNA transfected GBM cells or GBM cells co-cultured with neurons showed a decrease in the migration rate compared to controls. Previous data regarding laminin and these results demonstrating that CTGF is down-regulated in GBM cells co-cultured with neonatal neurons points out an interesting view in the understanding of the tumor and cerebral microenvironment interactions and could open up new strategies as well as suggest a new target in GBM control.


Subject(s)
Cell Communication , Connective Tissue Growth Factor/metabolism , Glioblastoma/metabolism , Neurons/metabolism , Animals , Cell Line, Tumor , Cell Movement , Coculture Techniques , Connective Tissue Growth Factor/genetics , Gene Expression , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation , Primary Cell Culture , Promoter Regions, Genetic , Rats , Signal Transduction , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Transcriptional Activation , Transforming Growth Factor beta/metabolism
15.
Curr Top Med Chem ; 12(19): 2082-93, 2012.
Article in English | MEDLINE | ID: mdl-23167797

ABSTRACT

Glioblastoma (GBM) is considered incurable due to its resistance to current cancer treatments. So far, all clinically available alternatives for treating GBM are limited, evoking the development of novel treatment strategies that can more effectively manage these tumors. Extensive effort is being dedicated to characterize the molecular basis of GBM resistance to chemotherapy and to explore novel therapeutic procedures that may improve overall survival. Cytolysins are toxins that form pores in target cell membranes, modifying ion homeostasis and leading to cell death. These pore-forming toxins might be used, therefore, to enhance the efficiency of conventional chemotherapeutic drugs, facilitating their entrance into the cell. In this study, we show that a non-cytotoxic concentration of equinatoxin II (EqTx-II), a pore-forming toxin from the sea anemone Actinia equina, potentiates the cytotoxicity induced by temozolomide (TMZ), a first-line GBM treatment, and by etoposide (VP-16), a second- or third-line GBM treatment. We also suggest that this effect is selective to GBM cells and occurs via PI3K/Akt pathway inhibition. Finally, Magnetic resonance imaging (MRI) revealed that a non-cytotoxic concentration of EqTx-II potentiates the VP-16-induced inhibition of GBM growth in vivo. These combined therapies constitute a new and potentially valuable tool for GBM treatment, leading to the requirement of lower concentrations of chemotherapeutic drugs and possibly reducing, therefore, the adverse effects of chemotherapy.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Brain Neoplasms/pathology , Cnidarian Venoms/pharmacology , Dacarbazine/analogs & derivatives , Etoposide/pharmacology , Glioblastoma/pathology , Animals , Blotting, Western , Cell Line, Tumor , Dacarbazine/pharmacology , Drug Synergism , Humans , Immunohistochemistry , Magnetic Resonance Imaging , Male , Mice , Mice, Inbred C57BL , Temozolomide
16.
J Biol Chem ; 287(49): 41432-45, 2012 Nov 30.
Article in English | MEDLINE | ID: mdl-23055518

ABSTRACT

Assembly of synapses requires proper coordination between pre- and postsynaptic elements. Identification of cellular and molecular events in synapse formation and maintenance is a key step to understand human perception, learning, memory, and cognition. A key role for astrocytes in synapse formation and function has been proposed. Here, we show that transforming growth factor ß (TGF-ß) signaling is a novel synaptogenic pathway for cortical neurons induced by murine and human astrocytes. By combining gain and loss of function approaches, we show that TGF-ß1 induces the formation of functional synapses in mice. Further, TGF-ß1-induced synaptogenesis involves neuronal activity and secretion of the co-agonist of the NMDA receptor, D-serine. Manipulation of D-serine signaling, by either genetic or pharmacological inhibition, prevented the TGF-ß1 synaptogenic effect. Our data show a novel molecular mechanism that might impact synaptic function and emphasize the evolutionary aspect of the synaptogenic property of astrocytes, thus shedding light on new potential therapeutic targets for synaptic deficit diseases.


Subject(s)
Astrocytes/cytology , Cerebral Cortex/metabolism , Neurons/metabolism , Serine/chemistry , Synapses/metabolism , Transforming Growth Factor beta/metabolism , Animals , Cells, Cultured , Cognition , Culture Media, Conditioned/pharmacology , Electrophysiology , Humans , Mice , Models, Biological , Patch-Clamp Techniques , Signal Transduction , Transfection
19.
Surg Neurol ; 72(6): 612-9, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19604557

ABSTRACT

BACKGROUND: Stent-assisted coiling is an accepted endovascular treatment (EVT) for wide-necked intracranial aneurysms. The Neuroform stent (Target Therapeutics, Fremont, Calif) is a flexible nitinol self-expandable stent that was designed to potentially overcome the limitations of balloon expandable coronary stents in the intracranial circulation. The aim of this study was to reenforce the use of this stent for EVT of wide-necked cerebral aneurysms. METHODS: Between March 2005 and March 2008, 24 patients harboring wide-necked cerebral aneurysms were treated with stent reconstruction of the aneurysm neck. Inclusion criteria restricted the group to adult patients with wide-necked intracranial aneurysms (ruptured and unruptured lesions). Immediate postprocedure angiography studies were performed to determine successful coil occlusion of the aneurysm as well as patency of the parent vessel. We assessed the clinical history, aneurysm dimensions, and technical detail of the procedures, including any difficulties with stent placement and deployment, degree of aneurysm occlusion, and complications. Clinical outcome was assessed with the Glasgow Outcome Scale (GOS). RESULTS: The stent was easily navigated and precisely positioned in 24 of 26 cases. However, technical difficulties occurred in 9 patients, including difficulties in crossing the stents interstice in 6 cases, inadvertent stent delivery (n = 1), and incapacity of stent delivery (n = 1) and incapacity of crossing the neck (n = 1). These latter 2 cases were classified as failures of the stent-assisted technique. A single procedural complication occurred, involving transient nonocclusive intrastent thrombus formation, which was treated uneventfully with abciximab. Seventeen patients experienced excellent clinical outcomes (GOS 5), with good outcomes (GOS 4) in 5 patients and a poor outcome (GOS 3) in 2 patients. There were no treatment-related deaths or neurologic complications (mean clinical follow-up, 12 months). Angiographic results consisted of 17 complete occlusions, 4 neck remnants, and 3 incomplete occlusions. CONCLUSIONS: The Neuroform stent is very useful for EVT of wide-necked intracranial aneurysms because it is easy to navigate and to deploy accurately. In most cases, the stent can be deployed precisely, even in very tortuous carotid siphons. Although in some cases delivery and deployment was challenging, clinically significant complications were not observed.


Subject(s)
Alloys , Aneurysm, Ruptured/therapy , Embolization, Therapeutic/instrumentation , Intracranial Aneurysm/therapy , Stents , Adult , Aged , Carotid Artery Diseases/diagnosis , Carotid Artery Diseases/therapy , Cerebral Angiography , Equipment Design , Female , Glasgow Outcome Scale , Humans , Intracranial Aneurysm/diagnosis , Male , Middle Aged , Tomography, X-Ray Computed , Treatment Outcome
20.
Arq Neuropsiquiatr ; 66(4): 795-9, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19099113

ABSTRACT

OBJECTIVE: Multiple cerebral cavernous malformation (CCM) is the hallmark of familial presentation of cavernous malformation in the brain. We describe an ongoing Familial Cerebral Cavernous Malformation Project in the Rio de Janeiro state showing genetic profile and the pattern of emergent neuroimaging findings of this particular population besides a review of the updated recommendations for management of familial CCM versus patients harboring sporadic lesions. METHOD: Four families of our cohort of 9 families were genetically mapped showing mutational profile linked to CCM1. The neuroimaging paradigm was shifted from T2*gradient-echo (GRE) sequence to susceptibility weighting MR phase imaging (SWI). RESULTS: Only two index cases were subjected to surgery. There was no surgical intervention in any of the kindreds of our entire cohort of 9 families of our Neurovascular Program within seven years of follow-up. The genetic sequencing for mutational profile in four of these families has demonstrated only CCM1 gene affected. Our management of the familial CCM is according to the review of the literature recommendations. CONCLUSIONS: The Project of Familial Cerebral Cavernous Malformations of Rio de Janeiro detected mutations of the gene CCM1 in the first four families studied. Familial cavernous malformation are to be settled apart from the more common sporadic lesion. A set of recommendations was searched for in the literature in order to deal with these specific patients and kindreds.


Subject(s)
Germ-Line Mutation/genetics , Intracranial Arteriovenous Malformations/genetics , Microtubule-Associated Proteins/genetics , Proto-Oncogene Proteins/genetics , Adolescent , Adult , Aged , Cohort Studies , Family , Female , Humans , Intracranial Arteriovenous Malformations/therapy , KRIT1 Protein , Magnetic Resonance Imaging , Male , Phenotype
SELECTION OF CITATIONS
SEARCH DETAIL
...