Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters








Publication year range
1.
Nat Commun ; 14(1): 602, 2023 02 06.
Article in English | MEDLINE | ID: mdl-36746942

ABSTRACT

Polyglutamine expansion in the androgen receptor (AR) causes spinobulbar muscular atrophy (SBMA). Skeletal muscle is a primary site of toxicity; however, the current understanding of the early pathological processes that occur and how they unfold during disease progression remains limited. Using transgenic and knock-in mice and patient-derived muscle biopsies, we show that SBMA mice in the presymptomatic stage develop a respiratory defect matching defective expression of genes involved in excitation-contraction coupling (ECC), altered contraction dynamics, and increased fatigue. These processes are followed by stimulus-dependent accumulation of calcium into mitochondria and structural disorganization of the muscle triads. Deregulation of expression of ECC genes is concomitant with sexual maturity and androgen raise in the serum. Consistent with the androgen-dependent nature of these alterations, surgical castration and AR silencing alleviate the early and late pathological processes. These observations show that ECC deregulation and defective mitochondrial respiration are early but reversible events followed by altered muscle force, calcium dyshomeostasis, and dismantling of triad structure.


Subject(s)
Androgens , Bulbo-Spinal Atrophy, X-Linked , Mice , Animals , Androgens/metabolism , Bulbo-Spinal Atrophy, X-Linked/genetics , Calcium/metabolism , Muscle, Skeletal/metabolism , Receptors, Androgen/metabolism , Mitochondria/metabolism , Respiration , Disease Models, Animal
2.
Oxid Med Cell Longev ; 2019: 1681254, 2019.
Article in English | MEDLINE | ID: mdl-31737163

ABSTRACT

Neurodegenerative diseases are a large and heterogeneous group of disorders characterized by selective and progressive death of specific neuronal subtypes. In most of the cases, the pathophysiology is still poorly understood, although a number of hypotheses have been proposed. Among these, dysregulation of Ca2+ homeostasis and mitochondrial dysfunction represent two broadly recognized early events associated with neurodegeneration. However, a direct link between these two hypotheses can be drawn. Mitochondria actively participate to global Ca2+ signaling, and increases of [Ca2+] inside organelle matrix are known to sustain energy production to modulate apoptosis and remodel cytosolic Ca2+ waves. Most importantly, while mitochondrial Ca2+ overload has been proposed as the no-return signal, triggering apoptotic or necrotic neuronal death, until now direct evidences supporting this hypothesis, especially in vivo, are limited. Here, we took advantage of the identification of the mitochondrial Ca2+ uniporter (MCU) and tested whether mitochondrial Ca2+ signaling controls neuronal cell fate. We overexpressed MCU both in vitro, in mouse primary cortical neurons, and in vivo, through stereotaxic injection of MCU-coding adenoviral particles in the brain cortex. We first measured mitochondrial Ca2+ uptake using quantitative genetically encoded Ca2+ probes, and we observed that the overexpression of MCU causes a dramatic increase of mitochondrial Ca2+ uptake both at resting and after membrane depolarization. MCU-mediated mitochondrial Ca2+ overload causes alteration of organelle morphology and dysregulation of global Ca2+ homeostasis. Most importantly, MCU overexpression in vivo is sufficient to trigger gliosis and neuronal loss. Overall, we demonstrated that mitochondrial Ca2+ overload is per se sufficient to cause neuronal cell death both in vitro and in vivo, thus highlighting a potential key step in neurodegeneration.


Subject(s)
Calcium Channels/metabolism , Calcium/metabolism , Cerebellar Cortex/physiology , Gliosis/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Neurodegenerative Diseases/metabolism , Neurons/physiology , Adenoviridae/genetics , Animals , Animals, Newborn , Calcium Channels/genetics , Calcium Signaling , Cell Death , Cerebellar Cortex/pathology , Genetic Vectors , Gliosis/genetics , Humans , Membrane Potential, Mitochondrial , Mice , Mice, Inbred C57BL , Mitochondrial Proteins/genetics , Neurodegenerative Diseases/pathology , Up-Regulation
3.
Acta Neuropathol ; 135(4): 529-550, 2018 04.
Article in English | MEDLINE | ID: mdl-29302779

ABSTRACT

Recent evidence indicates synaptic dysfunction as an early mechanism affected in neuroinflammatory diseases, such as multiple sclerosis, which are characterized by chronic microglia activation. However, the mode(s) of action of reactive microglia in causing synaptic defects are not fully understood. In this study, we show that inflammatory microglia produce extracellular vesicles (EVs) which are enriched in a set of miRNAs that regulate the expression of key synaptic proteins. Among them, miR-146a-5p, a microglia-specific miRNA not present in hippocampal neurons, controls the expression of presynaptic synaptotagmin1 (Syt1) and postsynaptic neuroligin1 (Nlg1), an adhesion protein which play a crucial role in dendritic spine formation and synaptic stability. Using a Renilla-based sensor, we provide formal proof that inflammatory EVs transfer their miR-146a-5p cargo to neuron. By western blot and immunofluorescence analysis we show that vesicular miR-146a-5p suppresses Syt1 and Nlg1 expression in receiving neurons. Microglia-to-neuron miR-146a-5p transfer and Syt1 and Nlg1 downregulation do not occur when EV-neuron contact is inhibited by cloaking vesicular phosphatidylserine residues and when neurons are exposed to EVs either depleted of miR-146a-5p, produced by pro-regenerative microglia, or storing inactive miR-146a-5p, produced by cells transfected with an anti-miR-146a-5p. Morphological analysis reveals that prolonged exposure to inflammatory EVs leads to significant decrease in dendritic spine density in hippocampal neurons in vivo and in primary culture, which is rescued in vitro by transfection of a miR-insensitive Nlg1 form. Dendritic spine loss is accompanied by a decrease in the density and strength of excitatory synapses, as indicated by reduced mEPSC frequency and amplitude. These findings link inflammatory microglia and enhanced EV production to loss of excitatory synapses, uncovering a previously unrecognized role for microglia-enriched miRNAs, released in association to EVs, in silencing of key synaptic genes.


Subject(s)
Extracellular Vesicles/immunology , Inflammation/metabolism , MicroRNAs/metabolism , Neuroglia/immunology , Neurons/immunology , Synapses/immunology , Animals , Cells, Cultured , Cerebrospinal Fluid/metabolism , Coculture Techniques , Extracellular Vesicles/pathology , Female , Hippocampus/immunology , Hippocampus/pathology , Humans , Inflammation/pathology , Male , Mice, Inbred C57BL , Neuroglia/pathology , Neuronal Plasticity/physiology , Neurons/pathology , Primary Cell Culture , Rats, Sprague-Dawley , Synapses/pathology
4.
J Biol Chem ; 290(52): 30931-46, 2015 Dec 25.
Article in English | MEDLINE | ID: mdl-26534959

ABSTRACT

The HIV-1 transactivator protein Tat is implicated in the neuronal damage that contributes to neurocognitive impairment affecting people living with HIV/AIDS. Aberrant splicing of TAU exon 10 results in tauopathies characterized by alterations in the proportion of TAU isoforms containing three (3R) or four (4R) microtubule-binding repeats. The splicing factor SC35/SRSF2 binds to nuclear RNA and facilitates the incorporation of exon 10 in the TAU molecule. Here, we utilized clinical samples, an animal model, and neuronal cell cultures and found that Tat promotes TAU 3R up-regulation through increased levels of phosphorylated SC35, which is retained in nuclear speckles. This mechanism involved Tat-mediated increased expression of DYRK1A and was prevented by DYRK1A silencing. In addition, we found that Tat associates with TAU RNA, further demonstrating that Tat interferes with host RNA metabolism in the absence of viral infection. Altogether, our data unravel a novel mechanism of Tat-mediated neuronal toxicity through dysregulation of the SC35-dependent alternative splicing of TAU exon 10. Furthermore, the increased immunostaining of DYRK1A in HIV+ brains without pathology points at dysregulation of DYRK1A as an early event in the neuronal complications of HIV infection.


Subject(s)
Exons , HIV Infections/metabolism , HIV-1/metabolism , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/biosynthesis , Protein-Tyrosine Kinases/biosynthesis , Ribonucleoproteins/metabolism , tat Gene Products, Human Immunodeficiency Virus/metabolism , tau Proteins/metabolism , Animals , Brain/metabolism , Brain/pathology , Brain/virology , Gene Expression Regulation, Enzymologic , HEK293 Cells , HIV Infections/genetics , HIV-1/genetics , Humans , Mice , Neurons/metabolism , Neurons/pathology , Neurons/virology , Nuclear Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/genetics , Ribonucleoproteins/genetics , Serine-Arginine Splicing Factors , Up-Regulation , tat Gene Products, Human Immunodeficiency Virus/genetics , tau Proteins/genetics , Dyrk Kinases
5.
J Biol Chem ; 290(13): 8067-80, 2015 Mar 27.
Article in English | MEDLINE | ID: mdl-25645911

ABSTRACT

Glioblastoma is one of the most aggressive brain tumors. We have previously found up-regulation of growth differentiation factor 15 (GDF15) in glioblastoma cells treated with the anticancer agent fenofibrate. Sequence analysis of GDF15 revealed the presence of a microRNA, miR-3189, in the single intron. We then asked whether miR-3189 was expressed in clinical samples and whether it was functional in glioblastoma cells. We found that expression of miR-3189-3p was down-regulated in astrocytoma and glioblastoma clinical samples compared with control brain tissue. In vitro, the functionality of miR-3189-3p was tested by RNA-binding protein immunoprecipitation, and miR-3189-3p coimmunoprecipitated with Argonaute 2 together with two of its major predicted gene targets, the SF3B2 splicing factor and the guanine nucleotide exchange factor p63RhoGEF. Overexpression of miR-3189-3p resulted in a significant inhibition of cell proliferation and migration through direct targeting of SF3B2 and p63RhoGEF, respectively. Interestingly, miR-3189-3p levels were increased by treatment of glioblastoma cells with fenofibrate, a lipid-lowering drug with multiple anticancer activities. The attenuated expression of miR-3189-3p in clinical samples paralleled the elevated expression of SF3B2, which could contribute to the activation of SF3B2 growth-promoting pathways in these tumors. Finally, miR-3189-3p-mediated inhibition of tumor growth in vivo further supported the function of this microRNA as a tumor suppressor.


Subject(s)
Brain Neoplasms/metabolism , Glioblastoma/metabolism , MicroRNAs/genetics , Animals , Base Sequence , Binding Sites , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Female , Gene Expression , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , Glioblastoma/genetics , Glioblastoma/pathology , Growth Differentiation Factor 15/genetics , Growth Differentiation Factor 15/metabolism , Humans , Mice, Nude , Neoplasm Transplantation , RNA Interference , RNA Splicing Factors , RNA-Binding Proteins/biosynthesis , RNA-Binding Proteins/genetics , Rho Guanine Nucleotide Exchange Factors/biosynthesis , Rho Guanine Nucleotide Exchange Factors/genetics
6.
J Vis Exp ; (83): e51172, 2014 Jan 22.
Article in English | MEDLINE | ID: mdl-24514260

ABSTRACT

MicroRNAs (miRNAs) constitute a potent layer of gene regulation by guiding RISC to target sites located on mRNAs and, consequently, by modulating their translational repression. Changes in miRNA expression have been shown to be involved in the development of all major complex diseases. Furthermore, recent findings showed that miRNAs can be secreted to the extracellular environment and enter the bloodstream and other body fluids where they can circulate with high stability. The function of such circulating miRNAs remains largely elusive, but systematic high throughput approaches, such as miRNA profiling arrays, have lead to the identification of miRNA signatures in several pathological conditions, including neurodegenerative disorders and several types of cancers. In this context, the identification of miRNA expression profile in the cerebrospinal fluid, as reported in our recent study, makes miRNAs attractive candidates for biomarker analysis. There are several tools available for profiling microRNAs, such as microarrays, quantitative real-time PCR (qPCR), and deep sequencing. Here, we describe a sensitive method to profile microRNAs in cerebrospinal fluids by quantitative real-time PCR. We used the Exiqon microRNA ready-to-use PCR human panels I and II V2.R, which allows detection of 742 unique human microRNAs. We performed the arrays in triplicate runs and we processed and analyzed data using the GenEx Professional 5 software. Using this protocol, we have successfully profiled microRNAs in various types of cell lines and primary cells, CSF, plasma, and formalin-fixed paraffin-embedded tissues.


Subject(s)
MicroRNAs/cerebrospinal fluid , Real-Time Polymerase Chain Reaction/methods , Humans
7.
J Cell Physiol ; 228(5): 1070-5, 2013 May.
Article in English | MEDLINE | ID: mdl-23042033

ABSTRACT

MicroRNAs are short non-coding RNAs that modulate gene expression by translational repression. Because of their high stability in intracellular as well as extracellular environments, miRNAs have recently emerged as important biomarkers in several human diseases. However, they have not been tested in the cerebrospinal fluid (CSF) of HIV-1 positive individuals. Here, we present results of a study aimed at determining the feasibility of detecting miRNAs in the CSF of HIV-infected individuals with and without encephalitis (HIVE). We also evaluated similarities and differences between CSF and brain tissue miRNAs in the same clinical setting. We utilized a high throughput approach of miRNA detection arrays and identified differentially expressed miRNAs in the frontal cortex of three cases each of HIV+, HIVE, and HIV- controls, and CSF of 10 HIV-positive and 10 HIV-negative individuals. For the CSF samples, the group of HIV+ individuals contained nine cases of HIV-associated neurological disorders (HAND) and, among those, four had HIVE. All the HIV-negative samples had non-viral acute disseminate encephalomyelitis. A total of 66 miRNAs were found differentially regulated in HIV+ compared to HIV- groups. The greatest difference in miRNA expression was observed when four cases of HIVE were compared to five non-HIVE cases, previously normalized with the HIV-negative group. After statistical analyses, 11 miRNAs were fund significantly up-regulated in HIVE. Although more clinical samples should be examined, this work represents the first report of CSF miRNAs in HIV-infection and offers the basis for future investigation.


Subject(s)
Encephalitis , HIV Infections , MicroRNAs , Cerebral Cortex/metabolism , Cerebral Cortex/virology , Encephalitis/cerebrospinal fluid , Encephalitis/complications , Encephalitis/pathology , Encephalitis/virology , Gene Expression Regulation , HIV Infections/cerebrospinal fluid , HIV Infections/complications , HIV Infections/pathology , HIV Infections/virology , HIV-1/isolation & purification , Humans , MicroRNAs/cerebrospinal fluid , MicroRNAs/genetics , MicroRNAs/metabolism
8.
Genes (Basel) ; 4(1): 46-64, 2013 Mar 04.
Article in English | MEDLINE | ID: mdl-24705102

ABSTRACT

Glioblastoma multiforme is characterized by rapid proliferation, aggressive metastatic potential, and resistance to radio- and chemotherapy. The matricellular protein CYR61 regulates cellular proliferation and migration and is highly expressed in Glioblastomas. MicroRNAs are 22-nucleotides long RNAs that regulate gene expression post-transcriptionally. Here, we utilized the LN229 glioblastoma cell line and found that CYR61 is a target of miR-136, miR-155, and miR-634. Over-expression of miR-136 and miR-634 miRNAs negatively affected proliferation, but not migration, while expression of miR-155 reduced migration but did not affect the proliferation of LN229 cells. Investigation of the molecular mechanisms affected by expression of miR-634 revealed an increased phosphorylation of p70S6 kinase, suggesting an induction of the mammalian target of rapamycin (mTOR) complex 1 pathway. Additionally, in miR-634 overexpressing cells, TSC2, a negative regulator of mTOR signaling, was found to be decreased. Altogether, our study provides insights on the differential roles of miRs-136, -155, and -634 in regulating glioblastoma cell growth and migration, and how microRNAs could be manipulated to decrease the aggressiveness and metastatic potential of tumor cells.

9.
J Vis Exp ; (63): e3965, 2012 May 24.
Article in English | MEDLINE | ID: mdl-22664838

ABSTRACT

We are describing a quick method to dissociate and culture hippocampal or cortical neurons from E15-17 rat embryos. The procedure can be applied successfully to the isolation of mouse and human primary neurons and neural progenitors. Dissociated neurons are maintained in serum-free medium up to several weeks. These cultures can be used for nucleofection, immunocytochemistry, nucleic acids preparation, as well as electrophysiology. Older neuronal cultures can also be transfected with a good efficiency rate by lentiviral transduction and, less efficiently, with calcium phosphate or lipid-based methods such as lipofectamine.


Subject(s)
Cell Culture Techniques/methods , Cerebral Cortex/cytology , Hippocampus/cytology , Neurons/cytology , Animals , Cerebral Cortex/embryology , Embryo, Mammalian/cytology , Glioblastoma/pathology , Hippocampus/embryology , Humans , Rats
10.
Biochim Biophys Acta ; 1813(10): 1836-44, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21745501

ABSTRACT

The Src-homology 3 (SH3) domain is one of the most frequent protein recognition modules (PRMs), being represented in signal transduction pathways and in several pathologies such as cancer and AIDS. Grb2 (growth factor receptor-bound protein 2) is an adaptor protein that contains two SH3 domains and is involved in receptor tyrosine kinase (RTK) signal transduction pathways. The HIV-1 transactivator factor Tat is required for viral replication and it has been shown to bind directly or indirectly to several host proteins, deregulating their functions. In this study, we show interaction between the cellular factor Grb2 and the HIV-1 trans-activating protein Tat. The binding is mediated by the proline-rich sequence of Tat and the SH3 domain of Grb2. As the adaptor protein Grb2 participates in a wide variety of signaling pathways, we characterized at least one of the possible downstream effects of the Tat/Grb2 interaction on the well-known IGF-1R/Raf/MAPK cascade. We show that the binding of Tat to Grb2 impairs activation of the Raf/MAPK pathway, while potentiating the PKA/Raf inhibitory pathway. The Tat/Grb2 interaction affects also viral function by inhibiting the Tat-mediated transactivation of HIV-1 LTR and viral replication in infected primary microglia.


Subject(s)
GRB2 Adaptor Protein/metabolism , HIV Infections/virology , HIV-1/physiology , Microglia/physiology , src Homology Domains , tat Gene Products, Human Immunodeficiency Virus/metabolism , Amino Acid Sequence , Cells, Cultured , Extracellular Signal-Regulated MAP Kinases/metabolism , GRB2 Adaptor Protein/chemistry , GRB2 Adaptor Protein/physiology , HIV Infections/metabolism , HIV Infections/pathology , HIV-1/metabolism , Host-Pathogen Interactions/physiology , Humans , Microglia/metabolism , Microglia/pathology , Microglia/virology , Molecular Sequence Data , Protein Binding , Signal Transduction , Virus Replication/physiology , tat Gene Products, Human Immunodeficiency Virus/physiology
11.
J Cell Physiol ; 226(7): 1763-70, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21506108

ABSTRACT

Inhibitor of differentiation-1 (Id-1) is a member of helix-loop-helix (HLH) family of proteins that regulate gene transcription through their inhibitory binding to basic-HLH transcription factors. Similarly to other members of this family, Id-1 is involved in the repression of cell differentiation and activation of cell growth. The dual function of Id-1, inhibition of differentiation, and stimulation of cell proliferation, might be interdependent, as cell differentiation is generally coupled with the exit from the cell cycle. Fibroblast growth factor-2 (FGF-2) has been reported to play multiple roles in different biological processes during development of the central nervous system (CNS). In addition, FGF-2 has been described to induce "neuronal-like" differentiation and trigger apoptosis in neuroblastoma SK-N-MC cells. Although regulation of Id-1 protein by several mitogenic factors is well-established, little is known about the role of FGF-2 in the regulation of Id-1. Using human neuroblastoma cell line, SK-N-MC, we found that treatment of these cells with FGF-2 resulted in early induction of both Id-1 mRNA and protein. The induction occurs within 1 h from FGF-2 treatment and is mediated by ERK1/2 pathway, which in turn stimulates expression of the early growth response-1 (Egr-1) transcription factor. We also demonstrate direct interaction of Egr-1 with Id-1 promoter in vitro and in cell culture. Finally, inhibition of Id-1 expression results in G(2) /M accumulation of FGF-2-treated cells and delayed cell death.


Subject(s)
Apoptosis , Brain Neoplasms/metabolism , Early Growth Response Protein 1/metabolism , Fibroblast Growth Factor 2/metabolism , Inhibitor of Differentiation Protein 1/metabolism , Neuroblastoma/metabolism , Binding Sites , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Cycle , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Inhibitor of Differentiation Protein 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neuroblastoma/genetics , Neuroblastoma/pathology , Promoter Regions, Genetic , RNA Interference , RNA, Messenger/metabolism , Recombinant Proteins/metabolism , Time Factors , Transfection , Up-Regulation
12.
FASEB J ; 24(7): 2292-300, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20181935

ABSTRACT

MicroRNA-mediated regulation of gene expression appears to be involved in a variety of cellular processes, including development, differentiation, proliferation, and apoptosis. Mir-146a is thought to be involved in the regulation of the innate immune response, and its expression is increased in tissues associated with chronic inflammation. Among the predicted gene targets for mir-146a, the chemokine CCL8/MCP-2 is a ligand for the CCR5 chemokine receptor and a potent inhibitor of CD4/CCR5-mediated HIV-1 entry and replication. In the present study, we have analyzed changes in the expression of mir-146a in primary human fetal microglial cells upon infection with HIV-1 and found increased expression of mir-146a. We further show that CCL8/MCP-2 is a target for mir-146a in HIV-1 infected microglia, as overexpression of mir-146a prevented HIV-induced secretion of MCP-2 chemokine. The clinical relevance of our findings was evaluated in HIV-encephalitis (HIVE) brain samples in which decreased levels of MCP-2 and increased levels of mir-146a were observed, suggesting a role for mir-146a in the maintenance of HIV-mediated chronic inflammation of the brain.


Subject(s)
Chemokine CCL8/antagonists & inhibitors , HIV Infections/etiology , HIV-1/pathogenicity , MicroRNAs/genetics , Microglia/virology , Cells, Cultured , Encephalitis, Viral/pathology , Gene Expression Regulation/immunology , HIV Infections/genetics , HIV Infections/immunology , Humans , Inflammation/virology
SELECTION OF CITATIONS
SEARCH DETAIL