Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 384
Filtrer
1.
AIDS Res Ther ; 20(1): 82, 2023 11 19.
Article de Anglais | MEDLINE | ID: mdl-37981694

RÉSUMÉ

BACKGROUND: Human immunodeficiency virus (HIV) infection is associated with an elevated incidence of cervical cancer, and accelerated disease progression, but the underlying mechanisms are not well understood. This study aimed to investigate the relationship between HIV infection and epithelial-mesenchymal transition (EMT) in cervical cancer. METHODS: Tissue samples from HIV-positive and negative patients with cervical intraepithelial neoplasia (CIN) and cervical cancer were analyzed for EMT-related proteins. Human cervical cancer SiHa cells were treated with HIV Tat and gp120 proteins to test their effects on EMT, migration, and invasion. RESULTS: HIV-positive patients had lower E-cadherin and cytokeratin, and higher N-cadherin and vimentin levels than HIV-negative patients. HIV Tat and gp120 proteins induced EMT, migration, and invasion in SiHa cells. Transcriptome sequencing analysis revealed that, compared to the control group, the protein-treated group showed upregulation of 22 genes and downregulation of 77 genes. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses revealed the involvement of the Wnt signaling pathway in EMT. Further analysis of gene expression related to this pathway revealed upregulation of DVL1, TCF7, KRT17, and VMAC, while GSK3ß, SFRP2, and CDH1 were downregulated. Immunofluorescence assay demonstrated that HIVgp120 and Tat proteins treatment induced elevated ß-catenin expression with nuclear accumulation in SiHa cells. CONCLUSIONS: The treatment of SiHa cells with HIV Tat and gp120 proteins induces EMT and activates the Wnt/ß-catenin pathway, suggesting that the Wnt/ß-catenin pathway may play a crucial role in promoting EMT progression in cervical lesion tissues of HIV-infected patients.


Sujet(s)
Infections à VIH , Tumeurs du col de l'utérus , Femelle , Humains , bêta-Caténine/génétique , bêta-Caténine/métabolisme , bêta-Caténine/pharmacologie , Tumeurs du col de l'utérus/génétique , Tumeurs du col de l'utérus/métabolisme , Tumeurs du col de l'utérus/anatomopathologie , Lignée cellulaire tumorale , Protéines du gène tat/pharmacologie , Transition épithélio-mésenchymateuse/physiologie
2.
Neurochem Int ; 167: 105538, 2023 Jul.
Article de Anglais | MEDLINE | ID: mdl-37207854

RÉSUMÉ

Oxidative stress plays a key role in the pathogenesis of neuronal injury, including ischemia. Ras-related nuclear protein (RAN), a member of the Ras superfamily, involves in a variety of biological roles, such as cell division, proliferation, and signal transduction. Although RAN reveals antioxidant effect, its precise neuroprotective mechanisms are still unclear. Therefore, we investigated the effects of RAN on HT-22 cell which were exposed to H2O2-induced oxidative stress and ischemia animal model by using the cell permeable Tat-RAN fusion protein. We showed that Tat-RAN transduced into HT-22 cells, and markedly inhibited cell death, DNA fragmentation, and reactive oxygen species (ROS) generation under oxidative stress. This fusion protein also controlled cellular signaling pathways, including mitogen-activated protein kinases (MAPKs), NF-κB, and apoptosis (Caspase-3, p53, Bax and Bcl-2). In the cerebral forebrain ischemia animal model, Tat-RAN significantly inhibited both neuronal cell death, and astrocyte and microglia activation. These results indicate that RAN significantly protects against hippocampal neuronal cell death, suggesting Tat-RAN will help to develop the therapies for neuronal brain diseases including ischemic injury.


Sujet(s)
Lésions encéphaliques , Encéphalopathie ischémique , Neuroprotecteurs , Animaux , Peroxyde d'hydrogène/pharmacologie , Protéine G ran/métabolisme , Protéine G ran/pharmacologie , Hippocampe/métabolisme , Ischémie/métabolisme , Stress oxydatif , Encéphalopathie ischémique/métabolisme , Apoptose , Protéines du gène tat/génétique , Protéines du gène tat/métabolisme , Protéines du gène tat/pharmacologie , Modèles animaux de maladie humaine , Lésions encéphaliques/métabolisme , Neuroprotecteurs/pharmacologie
3.
Sci Rep ; 13(1): 5653, 2023 04 06.
Article de Anglais | MEDLINE | ID: mdl-37024665

RÉSUMÉ

Malate dehydrogenase (MDH) plays an important role in the conversion of malate to oxaloacetate during the tricarboxylic acid cycle. In this study, we examined the role of cytoplasmic MDH (MDH1) in hydrogen peroxide (H2O2)-induced oxidative stress in HT22 cells and ischemia-induced neuronal damage in the gerbil hippocampus. The Tat-MDH1 fusion protein was constructed to enable the delivery of MDH1 into the intracellular space and penetration of the blood-brain barrier. Tat-MDH1, but not MDH1 control protein, showed significant cellular delivery in HT22 cells in a concentration- and time-dependent manner and gradual intracellular degradation in HT22 cells. Treatment with 4 µM Tat-MDH1 significantly ameliorated 200 µM H2O2-induced cell death, DNA fragmentation, and reactive oxygen species formation in HT22 cells. Transient increases in MDH1 immunoreactivity were detected in the hippocampal CA1 region 6-12 h after ischemia, but MDH1 activity significantly decreased 2 days after ischemia. Supplementation of Tat-MDH1 immediately after ischemia alleviated ischemia-induced hyperlocomotion and neuronal damage 1 and 4 days after ischemia. In addition, treatment with Tat-MDH1 significantly ameliorated the increases in hydroperoxides, lipid peroxidation, and reactive oxygen species 2 days after ischemia. Tat-MDH1 treatment maintained the redox status of the glutathione system in the hippocampus 2 days after ischemia. These results suggest that Tat-MDH1 exerts neuroprotective effects by reducing oxidative stress and maintaining glutathione redox system in the hippocampus.


Sujet(s)
Protéines du gène tat , Ischémie , Malate dehydrogenase , Neuroprotecteurs , Stress oxydatif , Animaux , Protéines du gène tat/pharmacologie , Gerbillinae , Hippocampe/métabolisme , Peroxyde d'hydrogène/métabolisme , Ischémie/traitement médicamenteux , Malate dehydrogenase/pharmacologie , Neurones/métabolisme , Neuroprotecteurs/pharmacologie , Oxydoréduction , Stress oxydatif/effets des médicaments et des substances chimiques , Espèces réactives de l'oxygène/métabolisme , Protéines de fusion recombinantes/pharmacologie , Souris
4.
CNS Neurosci Ther ; 29(1): 365-377, 2023 01.
Article de Anglais | MEDLINE | ID: mdl-36419337

RÉSUMÉ

INTRODUCTION: Mitochondrial-associated ER membranes (MAMs) control many cellular functions, including calcium and lipid exchange, intracellular trafficking, and mitochondrial biogenesis. The disruption of these functions contributes to neurocognitive disorders, such as spatial memory impairment and premature brain aging. Using neuronal cells, we demonstrated that HIV-1 Tat protein deregulates the mitochondria. METHODS& RESULTS: To determine the mechanisms, we used a neuronal cell line and showed that Tat-induced changes in expression and interactions of both MAM-associated proteins and MAM tethering proteins. The addition of HIV-1 Tat protein alters expression levels of PTPIP51 and VAPB proteins in the MAM fraction but not the whole cell. Phosphorylation of PTPIP51 protein regulates its subcellular localization and function. We demonstrated that the Tat protein promotes PTPIP51 phosphorylation on tyrosine residues and prevents its binding to VAPB. Treatment of the cells with a kinase inhibitor restores the PTPIP51-VAPB interaction and overcomes the effect of Tat. CONCLUSION: These results suggest that Tat disrupts the MAM, through the induction of PTPIP51 phosphorylation, leading to ROS accumulation, mitochondrial stress, and altered movement. Hence, we concluded that interfering in the MAM-associated cellular pathways contributes to spatial memory impairment and premature brain aging often observed in HIV-1-infected patients.


Sujet(s)
VIH-1 (Virus de l'Immunodéficience Humaine de type 1) , Humains , Encéphale/métabolisme , Protéines du gène tat/métabolisme , Protéines du gène tat/pharmacologie , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/métabolisme , Mitochondries/métabolisme , Protein Tyrosine Phosphatases/métabolisme , Protein Tyrosine Phosphatases/pharmacologie , Réticulum endoplasmique/métabolisme
5.
Neuropharmacology ; 220: 109256, 2022 12 01.
Article de Anglais | MEDLINE | ID: mdl-36162528

RÉSUMÉ

Methamphetamine (METH) is a psychostimulant that is abused throughout the world. METH is a highly addictive drug commonly used by persons living with HIV, and its use can result in cognitive impairment and memory deficits. METH and human immunodeficiency virus-1 transactivator of transcription (HIV-1Tat) have toxic and synergistic effects on the nervous system; however, the mechanism of their synergistic effects has not been clarified. We used BV2 cells, primary microglia, Nrf2-KO C57BL/6J mice, and autopsied brain tissues of METH-abusing, HIV infection, and METH-abusing individuals comorbid with HIV to explore the regulatory role of Nrf2/NQO1/HO-1 signal pathway on microglia autophagy. Our results showed that microglia were significantly activated by METH and HIV-1Tat protein. METH and HIV-1Tat protein combination significantly increase the autophagy-related proteins (LC3-II, Beclin-1, ATG5, and ATG7) expression in microglia and striatum of C57BL/6J mice. After silencing or knocking out the Nrf2 gene, the expression levels of autophagy-related proteins were significantly increased. In human brain tissue, microglia were activated, Nrf2, LC3-II, and Beclin-1 expression levels were raised, and the p62 expression level was decreased. Our results suggested that METH and HIV or HIV-1Tat synergistically affect autophagy. And the Nrf2 pathway plays a vital role in regulating the synergistic induction of microglial autophagy by METH and HIV-1Tat protein. This study may provide a theoretical basis and new ideas for effective targets for pharmacological intervention in HIV-infected patients with drug abuse.


Sujet(s)
Stimulants du système nerveux central , Infections à VIH , VIH-1 (Virus de l'Immunodéficience Humaine de type 1) , Métamfétamine , Animaux , Autophagie , Bécline-1/métabolisme , Stimulants du système nerveux central/pharmacologie , Protéines du gène tat/pharmacologie , Humains , Métamfétamine/effets indésirables , Souris , Souris de lignée C57BL , Microglie , NADPH dehydrogenase (quinone) , Facteur-2 apparenté à NF-E2/métabolisme , Transduction du signal
6.
Cell Rep ; 36(3): 109396, 2021 07 20.
Article de Anglais | MEDLINE | ID: mdl-34289359

RÉSUMÉ

Many neurological disorders show an increased prevalence of GluA2-lacking, Ca2+-permeable AMPA receptors (CP-AMPARs), which dramatically alters synaptic function. However, the molecular mechanism underlying this distinct synaptic plasticity remains enigmatic. Here, we show that nerve injury potentiates postsynaptic, but not presynaptic, CP-AMPARs in the spinal dorsal horn via α2δ-1. Overexpressing α2δ-1, previously regarded as a Ca2+ channel subunit, augments CP-AMPAR levels at the cell surface and synapse. Mechanistically, α2δ-1 physically interacts with both GluA1 and GluA2 via its C terminus, inhibits the GluA1/GluA2 heteromeric assembly, and increases GluA2 retention in the endoplasmic reticulum. Consequently, α2δ-1 diminishes the availability and synaptic expression of GluA1/GluA2 heterotetramers in the spinal cord in neuropathic pain. Inhibiting α2δ-1 with gabapentin or disrupting the α2δ-1-AMPAR complex fully restores the intracellular assembly and synaptic dominance of heteromeric GluA1/GluA2 receptors. Thus, α2δ-1 is a pivotal AMPAR-interacting protein that controls the subunit composition and Ca2+ permeability of postsynaptic AMPARs.


Sujet(s)
Sous-unités de protéines/métabolisme , Récepteur de l'AMPA/métabolisme , Synapses/métabolisme , Adolescent , Adulte , Animaux , Calcium/métabolisme , Perméabilité des membranes cellulaires/effets des médicaments et des substances chimiques , Réticulum endoplasmique/métabolisme , Femelle , Gabapentine/pharmacologie , Protéines du gène tat/pharmacologie , Cellules HEK293 , Humains , Mâle , Névralgie/métabolisme , Névralgie/anatomopathologie , Peptides/métabolisme , Peptides/pharmacologie , Phénotype , Liaison aux protéines/effets des médicaments et des substances chimiques , Rat Sprague-Dawley , Moelle spinale/anatomopathologie , Synapses/effets des médicaments et des substances chimiques , Jeune adulte
7.
Neuropharmacology ; 181: 108326, 2020 12 15.
Article de Anglais | MEDLINE | ID: mdl-32966847

RÉSUMÉ

We have previously shown that sphingosine kinase 2 (SPK2) interacts with Bcl-2 via its BH3 domain, activating autophagy by inducing the dissociation of Beclin-1/Bcl-2 complexes, and that a TAT-SPK2 peptide containing the BH3 domain of SPK2 protects neurons against ischemic injury. The goals of the present study were to establish the functional significance of these findings, by testing whether TAT-SPK2 was effective in a mouse model of ischemic stroke, and to explore potential underlying mechanisms. Mice were administered with TAT-SPK2 by intraperitoneal injection before or after transient middle cerebral artery occlusion (tMCAO). Infarct volume, neurological deficit and brain water content were assessed 24 h after reperfusion. Mitophagy inhibitor Mdivi-1 and BNIP3 siRNAs were used to examine the involvement of BNIP3-dependent mitophagy in the neuroprotection of TAT-SPK2. Mitophagy was quantified by immunoblotting, immunofluorescence and electron microscopy. The interaction between TAT-SPK2 and Bcl-2, Bcl-2 and BNIP3 was detected by co-immunoprecipitation. In the tMCAO model, pre-treatment with TAT-SPK2 significantly reduced infarct volume, improved neurological function and decreased brain edema. Neuroprotection by TAT-SPK2 was still seen when the peptide was administered 3 h after reperfusion. TAT-SPK2 also significantly improved functional recovery and reduced long-term brain atrophy of the ischemic hemisphere 30 days after administration. Our studies further showed that TAT-SPK2 directly binds to Bcl-2 and disrupts Bcl-2/Beclin-1 or Bcl-2/BNIP3 complexes to induce mitophagy. These results suggest that TAT-SPK2 protects neurons against ischemia reperfusion injury by activating BNIP3-mediated mitophagy. Agents exploiting this molecular mechanism are potential candidates for the treatment of ischemic stroke.


Sujet(s)
Protéines du gène tat/pharmacologie , Protéines membranaires/agonistes , Protéines mitochondriales/agonistes , Mitophagie/effets des médicaments et des substances chimiques , Neurones/effets des médicaments et des substances chimiques , Neuroprotecteurs/pharmacologie , Peptides/pharmacologie , Phosphotransferases (Alcohol Group Acceptor)/pharmacologie , Lésion d'ischémie-reperfusion/prévention et contrôle , Animaux , Autophagie , Bécline-1 , Oedème cérébral/prévention et contrôle , Infarctus du territoire de l'artère cérébrale moyenne/traitement médicamenteux , Accident vasculaire cérébral ischémique/traitement médicamenteux , Mâle , Souris , Souris de lignée ICR , Mitochondries/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , Protéines proto-oncogènes c-bcl-2/effets des médicaments et des substances chimiques , Petit ARN interférent/pharmacologie
8.
Exp Eye Res ; 199: 108180, 2020 10.
Article de Anglais | MEDLINE | ID: mdl-32777209

RÉSUMÉ

PURPOSE: Previously we developed TAT-N24 as a synthetic cell-permeable peptide inhibitor of p55PIK signaling and demonstrated its anti-inflammatory effects. This study aimed to evaluate the potential of TAT-N24 as a new agent for the treatment of ocular inflammatory diseases. METHODS: The endotoxin-induced uveitis (EIU) model was established by intravitreal injection of lipopolysaccharide (LPS) in BALB/c mice and experimental autoimmune uveitis (EAU) model was established by subcutaneous injection of a peptide spanning amino acid residues 161-180 of interphotoreceptor retinoid binding protein (IRBP161-180) with complete Freund's adjuvant (CFA) in B10.RIII mice. TAT-N24 was topically administered in EIU model and intraperitoneally administered in EAU model. The severity levels of uveitis were assessed by clinical and histopathological scores. The mRNA levels of inflammatory cytokines in iris-ciliary body (ICB) and retina were analyzed by reverse transcription quantitative polymerase chain reaction (RT-qPCR). The protein levels of inflammatory factors were determined by ELISA or Western blotting. RESULTS: The results showed that TAT-N24 alleviated clinical signs, decreased inflammatory cell infiltration and the expression of inflammatory cytokines in both EIU and EAU models. Furthermore, protein levels of tumor necrosis factor-alpha (TNF-α), interleukin-1ß (IL-1ß) and interleukin-6 (IL-6) in aqueous humor and mRNA and protein levels of NF-κB p65 in the ICB significantly decreased in EIU model. In EAU model, TAT-N24 application induced a significant decrease of IFN-gamma (IFN-γ) and interleukin-17 (IL-17) in the retina, which were secreted by Th1 and Th17 cells, respectively. CONCLUSION: In conclusion, TAT-N24 suppressed intraocular inflammation in both EIU and EAU models, and the anti-inflammatory effects were mediated by suppressing the expression of inflammatory cytokines by PI3K/NF-κB signaling pathway. TAT-N24 could be potential candidate for the treatment of ocular inflammatory diseases.


Sujet(s)
Cytokines/métabolisme , Protéines du gène tat/pharmacologie , Phosphatidylinositol 3-kinases/effets des médicaments et des substances chimiques , Uvéite/métabolisme , Animaux , Technique de Western , Modèles animaux de maladie humaine , Femelle , Souris , Souris de lignée BALB C , Phosphatidylinositol 3-kinases/métabolisme , Transduction du signal , Uvéite/traitement médicamenteux , Uvéite/anatomopathologie
9.
Osteoarthritis Cartilage ; 28(10): 1394-1400, 2020 10.
Article de Anglais | MEDLINE | ID: mdl-32683043

RÉSUMÉ

OBJECT: Autophagy maintains cartilage homeostasis and is compromised during osteoarthritis (OA), contributing to cartilage degeneration. We sought to determine if D-isomer TAT-Beclin-1, a potent inducer of autophagy, could attenuate post-traumatic OA in mice. METHODS: 10-week-old mice underwent destabilization of the medial meniscus (DMM) surgery to induce post-traumatic OA, or sham surgery (control), and injected intra-articularly with D-isomer TAT-Beclin-1 (0.5-2 mg/kg) or PBS 1 week post-surgery for up to 9 weeks. Mice were sacrificed at 2 or 10 weeks post-surgery. Knee joint sections were evaluated by histopathology for cartilage degeneration and synovitis, and immunostaining for key markers of autophagy (LC3B), cell proliferation (nuclear Ki67), activated fibroblasts (αSMA), and cells of hematopoietic origin (CD45). RESULTS: All D-isomer TAT-Beclin-1-treated DMM mice had no difference in the degree of cartilage degeneration compared to PBS-injected DMM mice. Surprisingly, all D-isomer TAT-Beclin-1-treated mice exhibited substantial synovial hyperplasia, with increased cellularity and ECM deposition (fibrosis-like phenotype), as compared to PBS-injected mice. Synovial effects of D-isomer TAT-Beclin-1 were dose- and injection frequency-dependent. An increased percentage of cells positive for LC3B and nuclear Ki67 were found in the synovial intima early after injection, which persisted after frequent injections. CONCLUSIONS: D-isomer TAT-Beclin-1 did not attenuate cartilage degeneration, but rather induced synovial hyperplasia associated with increased expression of key markers of autophagy and cell proliferation and a fibrosis-like phenotype, independent of markers of fibroblast activation or persistent hematopoietic-origin cell infiltration. These data suggest that, if not tissue-targeted, caution should be taken using autophagy activators due to diverse cellular responses in the joint.


Sujet(s)
Autophagie/effets des médicaments et des substances chimiques , Bécline-1/pharmacologie , Cartilage articulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Gonarthrose/anatomopathologie , Membrane synoviale/effets des médicaments et des substances chimiques , Animaux , Cartilage articulaire/anatomopathologie , Modèles animaux de maladie humaine , Fibroblastes/effets des médicaments et des substances chimiques , Protéines du gène tat/pharmacologie , Hyperplasie , Injections articulaires , Ménisques de l'articulation du genou/chirurgie , Souris , Membrane synoviale/anatomopathologie , Synovite/anatomopathologie , Lésions du ménisque externe
10.
Sci Rep ; 10(1): 6591, 2020 04 20.
Article de Anglais | MEDLINE | ID: mdl-32313258

RÉSUMÉ

The paper aims to investigate the cytotoxic effect on tumor cells of irradiated AuNPs in green light and subsequently functionalized with HS-PEG-NH2. The toxicity level of gold conjugates after their functionalization with DOX and TAT peptide was also evaluated. The AuNPs were prepared using the modified Turkevich method and exposed to visible light at a wavelength of 520 nm prior their PEGylation. The optical properties were analyzed by UV-vis spectroscopy, the surface modification was investigated using FTIR and XPS spectroscopies and their sizes and morphologies were evaluated by TEM and DLS techniques. DOX and TAT peptide were linked to the surface of PEGylated AuNPs by reacting their amino groups with glycidyloxypropyl of PEGylated DOX or TAT conjugates under mild conditions at room temperature and in the presence of ethanol as catalyst. The conjugates containing DOX or DOX and TAT have been characterized by fluorescence and FTIR techniques. The changes of electrochemical features were observed using cyclic voltammetry, suggesting a better stability of irradiated nanoparticles. By mass spectrometry it was confirmed that the compounds of interest were obtained. The cell viability test showed that irradiated and non-irradiated nanoparticles coated with PEG are not toxic in normal cells. Tumor cell viability analysis showed that the PEGylated nanoparticles modified with DOX and TAT peptide were more effective than pristine DOX, indicating cytotoxicity up to 10% higher than non-irradiated ones.


Sujet(s)
Doxorubicine/pharmacologie , Protéines du gène tat/pharmacologie , Nanoparticules métalliques/composition chimique , Ostéosarcome/traitement médicamenteux , Antinéoplasiques/composition chimique , Antinéoplasiques/pharmacologie , Lignée cellulaire tumorale , Survie cellulaire/effets des médicaments et des substances chimiques , Doxorubicine/composition chimique , Protéines du gène tat/composition chimique , Or/composition chimique , Humains , Ostéosarcome/génétique , Ostéosarcome/anatomopathologie , Peptides/composition chimique , Peptides/pharmacologie , Polyéthylène glycols/composition chimique , Polyéthylène glycols/pharmacologie , Spectroscopie infrarouge à transformée de Fourier
11.
Proc Natl Acad Sci U S A ; 116(47): 23437-23443, 2019 11 19.
Article de Anglais | MEDLINE | ID: mdl-31685638

RÉSUMÉ

Antibiotic resistance has become one of the major threats to global health. Photodynamic inactivation (PDI) develops little antibiotic resistance; thus, it becomes a promising strategy in the control of bacterial infection. During a PDI process, light-induced reactive oxygen species (ROS) damage the membrane components, leading to the membrane rupture and bacteria death. Due to the short half-life and reaction radius of ROS, achieving the cell-membrane intercalation of photosensitizers is a key challenge for PDI of bacteria. In this work, a tetraphenylethylene-based discrete organoplatinum(II) metallacycle (1) acts as a photosensitizer with aggregation-induced emission. It self-assembles with a transacting activator of transduction (TAT) peptide-decorated virus coat protein (2) through electrostatic interactions. This assembly (3) exhibits both ROS generation and strong membrane-intercalating ability, resulting in significantly enhanced PDI efficiency against bacteria. By intercalating in the bacterial cell membrane or entering the bacteria, assembly 3 decreases the survival rate of gram-negative Escherichia coli to nearly zero and that of gram-positive Staphylococcus aureus to ∼30% upon light irradiation. This study has wide implications from the generation of multifunctional nanomaterials to the control of bacterial infection, especially for gram-negative bacteria.


Sujet(s)
Acides acycliques/pharmacologie , Antibactériens/pharmacologie , Protéines de capside/pharmacologie , Membrane cellulaire/effets des médicaments et des substances chimiques , Escherichia coli/effets des médicaments et des substances chimiques , Protéines du gène tat/pharmacologie , Composés organiques du platine/pharmacologie , Photosensibilisants/pharmacologie , Staphylococcus aureus/effets des médicaments et des substances chimiques , Stilbènes/pharmacologie , Acides acycliques/composition chimique , Spectroscopie de résonance de spin électronique , Escherichia coli/effets des radiations , Escherichia coli/ultrastructure , Microscopie électronique , Photothérapie dynamique/méthodes , Espèces réactives de l'oxygène , Staphylococcus aureus/effets des radiations , Staphylococcus aureus/ultrastructure , Électricité statique , Virus de la mosaïque du tabac
12.
ACS Nano ; 13(8): 8577-8588, 2019 08 27.
Article de Anglais | MEDLINE | ID: mdl-31339295

RÉSUMÉ

Ischemic stroke caused by a thrombus clog and ischemia is one of the most lethal and disabling cerebrovascular diseases. A sequentially targeted delivery system is highly desired to deliver thrombolytics and neuroprotectant to the site of the thrombus and ischemic penumbra, respectively, to pursue a maximized combinational effect. Inspired by the vital roles that platelets play in thrombus formation, herein, we develop a bioengineered "nanoplatelet" (tP-NP-rtPA/ZL006e) for sequentially site-specific delivery of recombinant tissue plasminogen activator (rtPA) and neuroprotectant (ZL006e) for ischemic stroke treatment. The tP-NP-rtPA/ZL006e consists of a ZL006e-loaded dextran derivative polymeric nanoparticle core and platelet membrane shell conjugated with thrombin-cleavable Tat-peptide-coupled rtPA. Mediated by the cloak of the platelet membrane, tP-NP-rtPA/ZL006e targets the thrombus site and rtPA is triggered to release by the upregulated thrombin. Subsequently, the in situ exposed Tat peptide enhanced penetration of the "nanoplatelet" across the blood-brain barrier into ischemic brain for ZL006e site-specific delivery. From the in vitro and in vivo evaluation, tP-NP-rtPA/ZL006e is demonstrated to significantly enhance the anti-ischemic stroke efficacy in the rat model  with middle cerebral artery occlusion, showing a 63 and 72% decrease in ischemic area and reactive oxygen species level compared to that with free drug combination, respectively.


Sujet(s)
Plaquettes/composition chimique , Encéphalopathie ischémique/traitement médicamenteux , Nanoparticules/composition chimique , Accident vasculaire cérébral/traitement médicamenteux , Animaux , Barrière hémato-encéphalique/effets des médicaments et des substances chimiques , Encéphalopathie ischémique/anatomopathologie , Dextrane/composition chimique , Dextrane/pharmacologie , Fibrinolytiques/composition chimique , Fibrinolytiques/pharmacologie , Protéines du gène tat/composition chimique , Protéines du gène tat/pharmacologie , Humains , Mâle , Nanoparticules/usage thérapeutique , Neuroprotecteurs/composition chimique , Neuroprotecteurs/pharmacologie , Rats , Protéines recombinantes/composition chimique , Protéines recombinantes/pharmacologie , Accident vasculaire cérébral/anatomopathologie , Thrombine/composition chimique , Thrombine/pharmacologie , Activateur tissulaire du plasminogène/composition chimique , Activateur tissulaire du plasminogène/pharmacologie
13.
ACS Chem Neurosci ; 10(1): 412-423, 2019 01 16.
Article de Anglais | MEDLINE | ID: mdl-30117729

RÉSUMÉ

Early brain injury (EBI) after aneurysmal subarachnoid hemorrhage (SAH) contributes to high morbidity and mortality. Although it is well recognized that acute neuroinflammation reaction is one of the most important triggers of EBI, pharmacotherapy proved to be clinically effective against the initiating of neuroinflammation after SAH is lacking. The resident microglia and infiltrated peripheral monocyte are two main types of immune cells in central nervous system (CNS) and control the inflammation process in brain after SAH. But the time course and relative contributions of these two immune cell activations after SAH are unknown. The p75 neurotrophin receptor (p75NTR), member of TNF receptor superfamily, expresses on infiltrated peripheral monocytes and suppresses their proinflammatory action after brain insults. But the p75NTR expression on resident microglia in vivo is rarely explored and their function keeps elusive. Therefore, we designed this study to investigate the time course of resident microglia activation and peripheral monocyte infiltration, as well as the microglial expression of p75NTR by using CX3C-chemokine receptor 1 (Cx3cr1) and chemokine receptor 2 (Ccr2) double transgenic mice (Cx3cr1GFP/+Ccr2RFP/+) after SAH. The results showed activated microglia was observed in cortex as early as 24 h and further increased at 48 and 72 h post SAH, while the infiltrated monocyte was not found until 72h. In addition, activated microglia expressed p75NTR acutely and p75NTR specific antagonist TAT-Pep5 significantly reduced microglia activation, neuroinflammation and EBI from 24 to 72 h. Together, these data suggest that the early neuroinflammation reaction might be initiated and intensified mainly by resident microglia rather than infiltrated monocyte at least in the first 48 h after SAH and p75NTR blockading by TAT-Pep5P might alleviate EBI through mediating microglial activation.


Sujet(s)
Lésions encéphaliques/métabolisme , Microglie/métabolisme , Monocytes/métabolisme , Neuroprotecteurs/pharmacologie , Récepteurs facteur croissance nerf/métabolisme , Hémorragie meningée/métabolisme , Animaux , Lésions encéphaliques/étiologie , Lésions encéphaliques/prévention et contrôle , Femelle , Protéines du gène tat/pharmacologie , Protéines du gène tat/usage thérapeutique , Mâle , Souris , Souris de lignée C57BL , Souris transgéniques , Microglie/effets des médicaments et des substances chimiques , Monocytes/effets des médicaments et des substances chimiques , Neuroprotecteurs/usage thérapeutique , Répartition aléatoire , Récepteurs facteur croissance nerf/antagonistes et inhibiteurs , Hémorragie meningée/complications , Hémorragie meningée/traitement médicamenteux
14.
Metab Brain Dis ; 33(5): 1743-1753, 2018 10.
Article de Anglais | MEDLINE | ID: mdl-29987524

RÉSUMÉ

Human immunodeficiency virus type 1 (HIV-1) affects the central nervous system (CNS) that may lead to the development of HIV-associated neuropathologies. Tat protein is one of the viral proteins that have been linked to the neurotoxic effects of HIV. Since many individuals living with HIV often experience significant adverse circumstances, the present study investigated whether exposure to stressful conditions would exacerbate harmful effects of tat protein on brain function. Tat protein (10 µg/10 µl) was injected bilaterally into the dorsal hippocampus of the animal using stereotaxic techniques. The control group received an injection of saline (10 µl). Some control and tat protein-treated animals were subjected to restrain stress for 6 h per day for 28 days and compared to a non-stress group. All animals underwent two behavioural tests, the open field test (OFT) and the novel object recognition test (NORT) to assess their mood state and cognitive function respectively. The release of pro-inflammatory cytokines (TNF-α and IL-1ß) and the expression of mineralocorticoid (MR) and glucocorticoid (GR) receptors were also measured to see whether the impact of the repetitive stress on Tat protein-induced behavioural effects was mediated by elements of the immune system and the HPA axis. Rats treated with tat protein showed the following behavioural changes when compared to control animals: there was a significant decrease in time spent in the center of the open field during the OFT, a significant reduction in time spent with the novel object during the NORT, but no change in locomotor activity. Real-time PCR data showed that the expression levels of GR and MR mRNA were significantly reduced, while Western blot analysis showed that the protein expression levels of TNF-α and IL-1ß were significantly increased. The present findings indicated that injection of tat protein into the hippocampus of rats not subjected to stress may lead to anxiety-like behaviour and deficits in learning and memory. Tat-treated animals subjected to stress evoked only a modest effect on their behaviour and neurochemistry, while stress alone led to behavioural and neurochemical changes similar to tat protein.


Sujet(s)
Protéines du gène tat/pharmacologie , VIH (Virus de l'Immunodéficience Humaine) , Hippocampe/effets des médicaments et des substances chimiques , Hippocampe/métabolisme , Interleukine-1 bêta/métabolisme , Récepteurs aux glucocorticoïdes/métabolisme , Récepteurs des minéralocorticoïdes/métabolisme , Facteur de nécrose tumorale alpha/métabolisme , Protéines virales/pharmacologie , Animaux , Comportement animal/effets des médicaments et des substances chimiques , Protéines du gène tat/administration et posologie , Mâle , Mémoire/effets des médicaments et des substances chimiques , ARN messager/métabolisme , Rats , Contention physique/physiologie , Contention physique/psychologie , Techniques stéréotaxiques , Stress psychologique/métabolisme , Protéines virales/administration et posologie
15.
Autophagy ; 14(9): 1596-1619, 2018.
Article de Anglais | MEDLINE | ID: mdl-29966509

RÉSUMÉ

While the advent of combination antiretroviral therapy (cART) has dramatically increased the life expectancy of HIV-1 infected individuals, paradoxically, however, the prevalence of HIV-1-associated neurocognitive disorders is on the rise. Based on the premise that the cytotoxic HIV-1 protein, transactivator of transcription (TAT), a known activator of glial cells that is found to persist in the central nervous system (CNS) despite cART, we sought to explore the role of defective mitophagy in HIV-1 TAT-mediated microglial activation. Our results demonstrated that exposure of mouse primary microglia to HIV-1 TAT resulted in cellular activation involving altered mitochondrial membrane potential that was accompanied by accumulation of damaged mitochondria. Exposure of microglia to HIV-1 TAT resulted in increased expression of mitophagy signaling proteins, such as PINK1, PRKN, and DNM1L, with a concomitant increase in the formation of autophagosomes, as evidenced by increased expression of BECN1 and MAP1LC3B-II. Intriguingly, exposure of cells to HIV-1 TAT also resulted in increased expression of SQSTM1, signifying thereby a possible blockade of the mitophagy flux, leading, in turn, to the accumulation of mitophagosomes. Interestingly, HIV-1 TAT-mediated activation of microglia was associated with decreased rate of extracellular acidification and mitochondrial oxygen consumption and increased expression of proinflammatory cytokines, such as Tnf, Il1b, and Il6. HIV-1 TAT-mediated defective mitophagy leading to microglial activation was further validated in vivo in the brains of HIV-1 transgenic rats. In conclusion, HIV-1 TAT activates microglia by increasing mitochondrial damage via defective mitophagy. ABBREVIATIONS: 3-MA: 3-methyladenine; Δψm: mitochondrial membrane potential; ACTB: actin, beta; AIF1: allograft inflammatory factor 1; ATP: adenosine triphosphate; BAF: bafilomycin A1; BECN1: beclin 1, autophagy related; cART: combined antiretroviral therapy; CNS: central nervous system; DNM1L: dynamin 1 like; DMEM: Dulbecco modified Eagle medium; DAPI: 4,6-diamidino-2-phenylindole ; ECAR: extracellular acidification rate; FBS: fetal bovine serum; FCCP: trifluoromethoxy carbonylcyanide phenylhydrazone; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; HAND: HIV-1-associated neurocognitive disorders; HIV-1 TAT: human immunodeficiency virus-1 transactivator of transcription; IL1B: interleukin 1, beta; IL6: interleukin 6; ITGAM: integrin subunit alpha M; MAP1LC3B: microtubule-associated protein 1 light chain 3 beta; mPMs: mouse primary microglial cells; MRC: maximal respiratory capacity; mt-CO1: mitochondrially encoded cytochrome c oxidase; mt-ND6: mitochondrially encoded NADH:ubiquinone oxidoreductase core subunit 6; NFKB1: nuclear factor kappa B subunit 1; NLRP3: NLR family pyrin domain containing 3; OCR: oxygen consumption rate; PBS: phosphate-buffered saline; PINK1: PTEN induced putative kinase 1; PRKN: parkin RBR E3 ubiquitin protein ligase; ROS: reactive oxygen species; siRNA: small interfering RNA; SQSTM1: sequestosome 1; TNF: tumor necrosis factor.


Sujet(s)
Protéines du gène tat/pharmacologie , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/composition chimique , Microglie/métabolisme , Mitochondries/anatomopathologie , Mitophagie/effets des médicaments et des substances chimiques , Animaux , Autophagie/effets des médicaments et des substances chimiques , Bécline-1/métabolisme , Marqueurs biologiques/métabolisme , Cellules cultivées , Cytokines/métabolisme , Métabolisme énergétique/effets des médicaments et des substances chimiques , Extinction de l'expression des gènes/effets des médicaments et des substances chimiques , Médiateurs de l'inflammation/métabolisme , Potentiel de membrane mitochondriale/effets des médicaments et des substances chimiques , Souris de lignée C57BL , Microglie/effets des médicaments et des substances chimiques , Microglie/ultrastructure , Mitochondries/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , Mitochondries/ultrastructure , Modèles biologiques , Phagosomes/effets des médicaments et des substances chimiques , Phagosomes/métabolisme , Phagosomes/ultrastructure , Protein kinases/métabolisme , Rats transgéniques , Transduction du signal/effets des médicaments et des substances chimiques , Régulation positive/effets des médicaments et des substances chimiques
16.
Am J Physiol Renal Physiol ; 315(3): F595-F606, 2018 09 01.
Article de Anglais | MEDLINE | ID: mdl-29790391

RÉSUMÉ

Acute glomerulonephritis is characterized by rapid glomerular neutrophil recruitment, proteinuria, and glomerular hypercellularity. The current study tested the hypothesis that the release of neutrophil granule contents plays a role in both the loss of filtration barrier leading to proteinuria and the increase in glomerular cells. Inhibition of neutrophil exocytosis with a peptide inhibitor prevented proteinuria and attenuated podocyte and endothelial cell injury but had no effect on glomerular hypercellularity in an experimental acute glomerulonephritis model in mice. Cultivation of podocytes with neutrophil granule contents disrupted cytoskeletal organization, an in vitro model for podocyte effacement and loss of filtration barrier. Activated, cultured podocytes released cytokines that stimulated neutrophil chemotaxis, primed respiratory burst activity, and stimulated neutrophil exocytosis. We conclude that crosstalk between podocytes and neutrophils contributes to disruption of the glomerular filtration barrier in acute glomerulonephritis. Neutrophil granule products induce podocyte injury but do not participate in the proliferative response of intrinsic glomerular cells.


Sujet(s)
Cytosquelette d'actine/métabolisme , Maladie des anticorps antimembrane basale glomérulaire/métabolisme , Communication cellulaire , Exocytose , Débit de filtration glomérulaire , Granulocytes neutrophiles/métabolisme , Podocytes/métabolisme , Protéinurie/métabolisme , Cytosquelette d'actine/anatomopathologie , Animaux , Maladie des anticorps antimembrane basale glomérulaire/anatomopathologie , Maladie des anticorps antimembrane basale glomérulaire/physiopathologie , Maladie des anticorps antimembrane basale glomérulaire/prévention et contrôle , Lignée cellulaire , Cytokines/métabolisme , Modèles animaux de maladie humaine , Exocytose/effets des médicaments et des substances chimiques , Femelle , Protéines du gène tat/pharmacologie , Humains , Mâle , Souris de lignée C57BL , Activation des neutrophiles , Infiltration par les neutrophiles , Granulocytes neutrophiles/effets des médicaments et des substances chimiques , Podocytes/anatomopathologie , Protéinurie/anatomopathologie , Protéinurie/physiopathologie , Protéinurie/prévention et contrôle , Espèces réactives de l'oxygène/métabolisme , Protéines de fusion recombinantes/pharmacologie , Stimulation du métabolisme oxydatif , Protéines SNARE/pharmacologie
17.
J Nanobiotechnology ; 16(1): 29, 2018 Mar 26.
Article de Anglais | MEDLINE | ID: mdl-29580233

RÉSUMÉ

BACKGROUND: Gene therapy has been developed and used in medical treatment for many years, especially for the enhancement of endothelialization and angiogenesis. But slow endosomal escape rate is still one of the major barriers to successful gene delivery. In order to evaluate whether introducing oligohistidine (Hn) sequence into gene carriers can promote endosomal escape and gene transfection or not, we designed and synthesized Arg-Glu-Asp-Val (REDV) peptide functionalized TAT-NLS-Hn (TAT: typical cell-penetrating peptide, NLS: nuclear localization signals, Hn: oligohistidine sequence, n: 4, 8 and 12) peptides with different Hn sequence lengths. pEGFP-ZNF580 (pZNF580) was condensed by these peptides to form gene complexes, which were used to transfect human umbilical vein endothelial cells (HUVECs). RESULTS: MTT assay showed that the gene complexes exhibited low cytotoxicity for HUVECs. The results of cellular uptake and co-localization ratio demonstrated that the gene complexes prepared from TAT-NLS-Hn with long Hn sequence (n = 12) benefited for high internalization efficiency of pZNF580. In addition, the results of western blot analysis and PCR assay of REDV-TAT-NLS-H12/pZNF580 complexes showed significantly enhanced gene expression at protein and mRNA level. Wound healing assay and transwell migration assay also confirmed the improved proliferation and migration ability of the transfected HUVECs by these complexes. Furthermore, the in vitro and in vivo angiogenesis assay illustrated that these complexes could promote the tube formation ability of HUVECs. CONCLUSION: The above results indicated that the delivery efficiency of pZNF580 and its expression could be enhanced by introducing Hn sequence into gene carriers. The Hn sequence in REDV-TAT-NLS-Hn is beneficial for high gene transfection. These REDV and Hn functionalized TAT-NLS peptides are promising gene carriers in gene therapy.


Sujet(s)
Peptides de pénétration cellulaire/composition chimique , Protéines du gène tat/composition chimique , Histidine/composition chimique , Signaux de localisation nucléaire/composition chimique , Facteurs de transcription/génétique , Transfection/méthodes , Transport biologique , Mouvement cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques , Peptides de pénétration cellulaire/pharmacologie , Endosomes/métabolisme , Expression des gènes , Protéines du gène tat/pharmacologie , Histidine/pharmacologie , Cellules endothéliales de la veine ombilicale humaine , Humains , Signaux de localisation nucléaire/pharmacologie , Oligopeptides/composition chimique , Oligopeptides/pharmacologie , Plasmides/composition chimique , Plasmides/métabolisme , Relation structure-activité , Facteurs de transcription/métabolisme
18.
Int J Gynecol Cancer ; 27(6): 1082-1087, 2017 07.
Article de Anglais | MEDLINE | ID: mdl-28604448

RÉSUMÉ

BACKGROUND: Paclitaxel is recommended as a first-line chemotherapeutic agent against ovarian cancer, but drug resistance becomes a major limitation. The key molecule or mechanism associated with paclitaxel resistance in ovarian cancer still remains unclear. Recent studies have revealed an association between autophagy and drug resistance. METHODS: We previously synthesized a MAPK kinase-recombinant fusion protein, MAP2K6-FP, that contains 3 domains: a protein transduction domain TAT, a human ovarian cancer HO8910 cell-specific binding peptide, and a potential antitumor effector domain MKK6(E). In this study, we investigated the effect of MAP2K6-FP on HO8910 cells treated with paclitaxel. RESULTS: The IC50 (concentration by which 50% cell growth was inhibited) was 20 µM for paclitaxel alone, 1.5 µg/mL for MAP2K6-FP alone, and 0.3 µg/mL for MAP2K6-FP and 15 µM for paclitaxel if combined, respectively. In addition, immunohistochemistry assay demonstrated that tumor tissues from ovarian cancer patients showed higher expression of LC-3, the autophagy-related protein, compared with normal ovarian tissues. MAP2K6-FP (0, 2.5, 5, 10, 20, and 40 µg/mL) dose-dependently increased the LC-3 expression in HO8910 cells. Immunofluorescence assay showed that paclitaxel alone increased the expression of LC-3 in HO8910 cells, which was further enhanced by the combination with MAP2K6-FP. Downregulation of LC-3 expression using LC-3 small interfering RNA inhibited the cytotoxicity effect of MAP2K6-FP. Furthermore, either MAP2K6-FP alone or in combination with paclitaxel increased the ratio of expressions of Beclin-1/Bcl-2, another autophagy-related markers, compared with paclitaxel alone. CONCLUSIONS: MAP2K6-FP enhanced the sensitiveness of paclitaxel for ovarian cancer via inducing autophagy.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique/pharmacologie , MAP Kinase Kinase 6/pharmacologie , Tumeurs de l'ovaire/traitement médicamenteux , Paclitaxel/pharmacologie , Protéines de fusion recombinantes/pharmacologie , Autophagie/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Synergie des médicaments , Femelle , Protéines du gène tat/génétique , Protéines du gène tat/pharmacologie , Humains , MAP Kinase Kinase 6/génétique , Tumeurs de l'ovaire/anatomopathologie , Paclitaxel/administration et posologie , Domaines protéiques , Protéines de fusion recombinantes/administration et posologie , Protéines de fusion recombinantes/génétique
19.
Vet Res Commun ; 41(3): 211-217, 2017 Sep.
Article de Anglais | MEDLINE | ID: mdl-28589421

RÉSUMÉ

The Tat protein is able to translocate through the plasma membrane and when it is fused with other peptides may acts as a protein transduction system. This ability appears particularly interesting to induce tissue-specific differentiation when the Tat protein is associated to transcription factors. In the present work, the potential of the complex Tat-MyoD in inducing equine peripheral blood mesenchymal stem cells (PB-MSCs) towards the myogenic fate, was evaluated. Results showed that the internalization process of Tat-MyoD happens only in serum free conditions and that the nuclear localization of the fused complex is observed after 15 hours of incubation. However, the supplement of Tat-MyoD only was not sufficient to induce myogenesis and, therefore, in order to achieve the myogenic differentiation of PB-MSCs, conditioned medium from C2C12 cells was added without direct contact. Real Time PCR and immunofluorescence methods evaluated the establishment of a myogenic program. Our results suggest that TAT- transduction of Tat-MyoD, when supported by conditioned medium, represents a useful methodology to induce myogenic differentiation.


Sujet(s)
Différenciation cellulaire/effets des médicaments et des substances chimiques , Protéines du gène tat/pharmacologie , Cellules souches mésenchymateuses/effets des médicaments et des substances chimiques , Protéine MyoD/pharmacologie , Animaux , Milieux de culture conditionnés/pharmacologie , Equus caballus , Cellules souches mésenchymateuses/physiologie , Réaction de polymérisation en chaine en temps réel/médecine vétérinaire , Transduction du signal
20.
Mol Med Rep ; 15(6): 4312-4318, 2017 Jun.
Article de Anglais | MEDLINE | ID: mdl-28487986

RÉSUMÉ

The present study investigated the protective effects of small ubiquitin-like modifier 1 (SUMO-1) on spinal cord ischemic damage in rabbits. A trans­activator of transcription (Tat)­SUMO­1 fusion protein was prepared, and transient spinal cord ischemia was induced by occlusion of the abdominal aorta for 15 min. Vehicle (glycerol) or 1 mg/kg Tat-1-SUMO­1 was administered intraperitoneally to the rabbits immediately following ischemia/reperfusion. Administration of Tat-SUMO-1 did not lead to significant alterations in arterial blood gases [partial pressure (Pa)CO2 and PaO2], pH, or blood glucose levels prior to ischemia, 10 min after occlusion or 10 min after reperfusion. Mean arterial pressure was significantly decreased only during occlusion. Motor behaviors were assessed at 24, 48 and 72 h after ischemia/reperfusion using Tarlov's criteria. Administration of Tat­SUMO­1 significantly improved Tarlov scores 24 h after ischemia/reperfusion and the number of cresyl violet positive neurons was significantly increased in the ventral horn of the spinal cord compared with the vehicle­treated group. However, Tarlov scores were consistently decreased at 48 and 72 h after ischemia/reperfusion in the Tat­SUMO­1­treated group, and Tarlov scores and the number of cresyl violet positive neurons were not significantly different between the vehicle­ and Tat­SUMO­1­treated groups after 72 h. Tat-SUMO­1 administration significantly ameliorated a reduction in Cu, Zn­superoxide dismutase activity and an increase in lipid peroxidation 24 h after ischemia/reperfusion; however, these effects were not present at 72 h. These results suggested that Tat­SUMO­1 may delay, although not protect against, neuronal death by regulating oxidative stress in the ventral horn of the spinal cord and that combination therapy using Tat­SUMO­1 with other compounds may provide a therapeutic approach to decrease neuronal damage.


Sujet(s)
Neurones/effets des médicaments et des substances chimiques , Neuroprotecteurs/pharmacologie , Lésion d'ischémie-reperfusion/traitement médicamenteux , Protéine SUMO-1/pharmacologie , Ischémie de la moelle épinière/traitement médicamenteux , Moelle spinale/effets des médicaments et des substances chimiques , Animaux , Protéines du gène tat/métabolisme , Protéines du gène tat/pharmacologie , Peroxydation lipidique/effets des médicaments et des substances chimiques , Mâle , Neurones/métabolisme , Lapins , Lésion d'ischémie-reperfusion/métabolisme , Protéine SUMO-1/métabolisme , Moelle spinale/métabolisme , Ischémie de la moelle épinière/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE