Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Int J Mol Sci ; 22(15)2021 Jul 25.
Article in English | MEDLINE | ID: mdl-34360699

ABSTRACT

Reactive astrocytes are a hallmark of neurodegenerative disease including multiple sclerosis. It is widely accepted that astrocytes may adopt alternative phenotypes depending on a combination of environmental cues and intrinsic features in a highly plastic and heterogeneous manner. However, we still lack a full understanding of signals and associated signaling pathways driving astrocyte reaction and of the mechanisms by which they drive disease. We have previously shown in the experimental autoimmune encephalomyelitis mouse model that deficiency of the molecular adaptor Rai reduces disease severity and demyelination. Moreover, using primary mouse astrocytes, we showed that Rai contributes to the generation of a pro-inflammatory central nervous system (CNS) microenvironment through the production of nitric oxide and IL-6 and by impairing CD39 activity in response to soluble factors released by encephalitogenic T cells. Here, we investigated the impact of Rai expression on astrocyte function both under basal conditions and in response to IL-17 treatment using a proteomic approach. We found that astrocytes and astrocyte-derived extracellular vesicles contain a set of proteins, to which Rai contributes, that are involved in the regulation of oligodendrocyte differentiation and myelination, nitrogen metabolism, and oxidative stress. The HIF-1α pathway and cellular energetic metabolism were the most statistically relevant molecular pathways and were related to ENOA and HSP70 dysregulation.


Subject(s)
Astrocytes/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Extracellular Vesicles/metabolism , Interleukin-17/pharmacology , Neuroprotection , Oligodendroglia/physiology , Src Homology 2 Domain-Containing, Transforming Protein 3/genetics , Animals , Cell Differentiation , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Gene Expression Regulation , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myelin Sheath , Proteomics , Src Homology 2 Domain-Containing, Transforming Protein 3/metabolism
2.
Cell Death Dis ; 12(3): 278, 2021 03 15.
Article in English | MEDLINE | ID: mdl-33723262

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the most common cancers with an insidious onset, strong invasiveness, insensitivity to chemotherapy, and poor prognosis, thus makes clinical treatment challenging. The mechanisms require further elucidation for developing novel therapies and targeting drug resistance. Here, we observed high Shc3 expression in patients with chemoresistant and recurrent HCCs. Shc3 overexpression induced a significant increase in MDR1/P-glycoprotein expression, whereas Shc3 knockdown impaired this expression. Further, Shc3 inhibition significantly restored HCC cell sensitivity to doxorubicin and sorafenib. Mechanistically, Shc3 interacted with ß-catenin, inhibited destruction complex stability, promoted ß-catenin release, and dampened ß-catenin ubiquitination. Shc3 bound ß-catenin and facilitated its nuclear translocation, prompting the ß-catenin/TCF pathway to elevate MDR1 transcription. ß-catenin blockage abolished the discrepancy in drug resistance between Shc3-depleted HCC cells and control cells, which further validating that ß-catenin is required for Shc3-mediated liver chemotherapy. We also determined the effect of Shc3 on the sensitivity of HCC to chemotherapy in vivo. Collectively, this study provides a potential strategy to target these pathways concurrently with systemic chemotherapy that can improve the clinical treatment of HCC.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/drug therapy , Doxorubicin/pharmacology , Drug Resistance, Neoplasm , Liver Neoplasms/drug therapy , Neoplastic Stem Cells/drug effects , Src Homology 2 Domain-Containing, Transforming Protein 3/metabolism , beta Catenin/metabolism , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily B/metabolism , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Female , Gene Expression Regulation, Neoplastic , Hep G2 Cells , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Proteolysis , Signal Transduction , Src Homology 2 Domain-Containing, Transforming Protein 3/genetics , TCF Transcription Factors/genetics , TCF Transcription Factors/metabolism , Ubiquitination , Xenograft Model Antitumor Assays
3.
Front Immunol ; 10: 1041, 2019.
Article in English | MEDLINE | ID: mdl-31134091

ABSTRACT

Multiple sclerosis is an autoimmune disease caused by autoreactive immune cell infiltration into the central nervous system leading to inflammation, demyelination, and neuronal loss. While myelin-reactive Th1 and Th17 are centrally implicated in multiple sclerosis pathogenesis, the local CNS microenvironment, which is shaped by both infiltrated immune cells and central nervous system resident cells, has emerged a key player in disease onset and progression. We have recently demonstrated that ShcC/Rai is as a novel astrocytic adaptor whose loss in mice protects from experimental autoimmune encephalomyelitis. Here, we have explored the mechanisms that underlie the ability of Rai-/- astrocytes to antagonize T cell-dependent neuroinflammation. We show that Rai deficiency enhances the ability of astrocytes to upregulate the expression and activity of the ectonucleotidase CD39, which catalyzes the conversion of extracellular ATP to the immunosuppressive metabolite adenosine, through both contact-dependent and-independent mechanisms. As a result, Rai-deficient astrocytes acquire an enhanced ability to suppress T-cell proliferation, which involves suppression of T cell receptor signaling and upregulation of the inhibitory receptor CTLA-4. Additionally, Rai-deficient astrocytes preferentially polarize to the neuroprotective A2 phenotype. These results identify a new mechanism, to which Rai contributes to a major extent, by which astrocytes modulate the pathogenic potential of autoreactive T cells.


Subject(s)
Antigens, CD/metabolism , Apyrase/metabolism , Astrocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Src Homology 2 Domain-Containing, Transforming Protein 3/genetics , Animals , CD8-Positive T-Lymphocytes/immunology , CTLA-4 Antigen/metabolism , Cell Proliferation/genetics , Cells, Cultured , Central Nervous System/immunology , Central Nervous System/pathology , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Src Homology 2 Domain-Containing, Transforming Protein 3/metabolism
4.
Cell Physiol Biochem ; 49(3): 971-984, 2018.
Article in English | MEDLINE | ID: mdl-30184529

ABSTRACT

BACKGROUND/AIMS: Parkinson's disease (PD) is a prevalent disease that leads to motor and cognitive disabilities, and oxidative stress (OS) injury was found to be related to the etiology of PD. Increasing evidence has shown that SHC3 is aberrantly expressed in neurons. The current study examines the involvement of SHC3 silencing in OS injury in the nigral dopamine neurons in rats with PD via the PI3K-AKT-FoxO signaling pathway. METHODS: To study the mechanisms and functions of SHC3 silencing in PD at the tissue level, 170 rats were selected, and a lentivirus-based packaging system was designed to silence SHC3 expression in rats. Furthermore, PC12 cells were selected for in vitro experimentation. To evaluate the effect of SHC3 silencing in nigral dopamine neuronal growth, an MTT assay, propidium iodide (PI) single staining and Annexin V-PI double staining were performed to detect cell viability, cell cycle progression and cell apoptosis, respectively. RESULTS: SHC3 shRNA led to decreased SOD and MDA levels and enhanced GSH activity, indicating that SHC3 silencing leads to motor retardation. SHC3 silencing repressed the extent of Akt and FoxO phosphorylation, thereby inhibiting the PI3K-AKT-FoxO signaling pathway. Furthermore, in cell experiments, SHC3 silencing suppressed PC12 cell proliferation and cell cycle progression, whereas it enhanced cell apoptosis. CONCLUSION: The current study provides evidence suggesting that SHC3 silencing may aggravate OS injury in nigral dopamine neurons via downregulation of the PI3K-AKT-FoxO signaling pathway in PD rats.


Subject(s)
Dopaminergic Neurons/metabolism , Oxidative Stress , Signal Transduction , Src Homology 2 Domain-Containing, Transforming Protein 3/metabolism , Animals , Behavior, Animal , Disease Models, Animal , Forkhead Transcription Factors/metabolism , Lentivirus/genetics , Male , Malondialdehyde/metabolism , PC12 Cells , Parkinson Disease/metabolism , Parkinson Disease/pathology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Src Homology 2 Domain-Containing, Transforming Protein 3/antagonists & inhibitors , Src Homology 2 Domain-Containing, Transforming Protein 3/genetics , Superoxide Dismutase/metabolism
5.
Cancer Res ; 78(9): 2219-2232, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29330146

ABSTRACT

Invasion and intrahepatic metastasis are major factors of poor prognosis in patients with hepatocellular carcinoma (HCC). In this study, we show that increased Src homolog and collagen homolog 3 (Shc3) expression in malignant HCC cell lines associate with HCC invasion and metastasis. Shc3 (N-Shc) was significantly upregulated in tumors of 33 HCC patient samples as compared with adjacent normal tissues. Further analysis of 52 HCC patient samples showed that Shc3 expression correlated with microvascular invasion, cancer staging, and poor prognosis. Shc3 interacted with major vault protein, resulting in activation of MEK1/2 and ERK1/2 independently of Shc1 and c-Raf; this interaction consequently induced epithelial-mesenchymal transition and promoted HCC cell proliferation and metastasis. The observed increase in Shc3 levels was due to demethylation of its upstream promoter, which allowed c-Jun binding. In turn, Shc3 expression promoted c-Jun phosphorylation in a positive feedback loop. Analysis of metastasis using a tumor xenograft mouse model further confirmed the role of Shc3 in vivo Taken together, our results indicate the importance of Shc3 in HCC progression and identify Shc3 as a novel biomarker and potential therapeutic target in HCC.Significance: Ectopic expression of Shc3 forms a complex with MVP/MEK/ERK to potentiate ERK activation and plays an important role in sorafinib resistance in HCC. Cancer Res; 78(9); 2219-32. ©2018 AACR.


Subject(s)
Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Gene Expression Regulation, Neoplastic , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , MAP Kinase Signaling System , Proto-Oncogene Proteins c-raf/metabolism , Src Homology 2 Domain-Containing, Transforming Protein 3/genetics , Animals , Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Movement/genetics , DNA Methylation , Gene Expression Profiling , Humans , Liver Neoplasms/pathology , Male , Mice , Models, Biological , Promoter Regions, Genetic , Protein Binding , Protein Kinase Inhibitors/pharmacology , Sorafenib/pharmacology , Transcriptome
SELECTION OF CITATIONS
SEARCH DETAIL
...