Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Front Endocrinol (Lausanne) ; 11: 613879, 2020.
Article in English | MEDLINE | ID: mdl-33716952

ABSTRACT

Papillary thyroid carcinoma (PTC) is the most common thyroid cancer with a rapidly increasing incidence globally. Bioinformatics analyses suggested that SHCBP1 (SHC SH2 Domain-Binding Protein 1) was significantly up-regulated in PTC tumor tissues, which was further confirmed by immunohistochemical staining and qPCR analyses in Xuzhou cohort. Moreover, the results indicated that the mRNA level of SHCBP1 was negatively associated with patients' disease-free survival rate, and further analysis reveals that patients with high SHCBP1 expression tend to have more lymph node metastasis. Afterward, MTT, colony formation, cell-cycle assay, FACS apoptosis assay, invasion, migration, as well as scratch assay were performed to study the phenotypes change of PTC cells after knocking down SHCBP1. The in vivo subcutaneous tumor model was developed to study the proliferation ability of PTC cells after SHCBP1 knockdown. We show that knock down of SHCBP1 significantly inhibits PTC cell proliferation, cell cycle, invasion and migration in vivo and in vitro. Western blot and qRT-PCR showed that knockdown of SHCBP1 could significantly reduce MYC, KLF4, CD44, ITGA6, ITGB1, ITGB5, and COL4A2 expression at both RNA and protein levels, which indicated that SHCBP1 might be involved in PTC carcinogenesis and progression through targeting formation of integrin and collagen and cell stemness pathways, and can be a potential diagnosis biomarker and therapeutic target for PTC.


Subject(s)
Collagen/metabolism , Integrins/metabolism , Neoplastic Stem Cells/metabolism , Shc Signaling Adaptor Proteins/biosynthesis , Thyroid Cancer, Papillary/metabolism , Thyroid Neoplasms/metabolism , Animals , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/deficiency , Cell Line, Tumor , Cohort Studies , Disease Progression , Female , Humans , Kruppel-Like Factor 4 , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness/pathology , Neoplastic Stem Cells/pathology , Shc Signaling Adaptor Proteins/deficiency , Thyroid Cancer, Papillary/pathology , Thyroid Neoplasms/pathology
2.
Cell Biochem Funct ; 35(8): 527-537, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29243276

ABSTRACT

The purpose of this study was to determine if Shc proteins influence the metabolic response to acute (7 days) feeding of a high-fat diet (HFD). To this end, whole animal energy expenditure (EE) and substrate oxidation were measured in the Shc knockout (ShcKO) and wild-type (WT) mice fed a control or HFD. The activities of enzymes of glycolysis, the citric acid cycle, electron transport chain (ETC), and ß-oxidation were also investigated in liver and skeletal muscle of ShcKO and WT animals. The study showed that ShcKO increases (P < .05) EE adjusted for either total body weight or lean mass. This change in EE could contribute to decreases in weight gain in ShcKO versus WT mice fed an HFD. Thus, our results indicate that Shc proteins should be considered as potential targets for developing interventions to mitigate weight gain on HFD by stimulating EE. Although decreased levels of Shc proteins influenced the activity of some enzymes in response to high-fat feeding (eg, increasing the activity of acyl-CoA dehydrogenase), it did not produce concerted changes in enzymes of glycolysis, citric acid cycle, or the ETC. The physiological significance of observed changes in select enzyme activities remains to be determined. SIGNIFICANCE OF THE STUDY: We report higher EE in ShcKO versus WT mice when consuming the HFD. Although decreased levels of Shc proteins influenced the activity of a central enzyme of ß-oxidation in response to high-fat feeding, it did not produce concerted changes in enzymes of glycolysis, citric acid cycle, or the ETC. Thus, an increase in EE in response to consumption of an HFD may be a mechanism that leads to decreased weight gain previously reported in ShcKO mice with long-term consumption of an HFD.


Subject(s)
Diet, High-Fat/adverse effects , Energy Metabolism , Shc Signaling Adaptor Proteins/metabolism , Animals , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Shc Signaling Adaptor Proteins/deficiency , Weight Gain
3.
PLoS One ; 10(3): e0119797, 2015.
Article in English | MEDLINE | ID: mdl-25790295

ABSTRACT

The adaptor protein p66Shc regulates intracellular oxidant levels through the modulation of a forkhead-related transcription factor (FOXO3a). The genetic ablation of p66Shc (p66Shc-/-) renders mice resistant to oxidative stress and p53-dependent apoptosis. We investigated whether p66Shc ablation in mice modifies lung cellular and molecular responses to cigarette smoke (CS) exposure. No differences between wild type (WT) and p66Shc-/- mice were observed in terms of inflammation and oxidant burden after acute CS exposure; however,p66Shc ablation modifies specific features of chronic inflammation induced by repeated exposure to CS. Unlike WT mice, p66Shc-/- mice did not develop emphysema, showing protection toward oxidative damage to DNA and apoptosis as revealed by a trivial 8-hydroxyguanosine staining and faint TUNEL and caspase-3 positivity on alveolar epithelial cells. Unexpectedly, CS exposure in p66Shc-/- mice resulted in respiratory bronchiolitis with fibrosis in surrounded alveoli. Respiratory bronchiolitis was characterized by peribronchiolar infiltrates of lymphocytes and histiocytes, accumulation of ageing pigmented macrophages within and around bronchioles, and peribronchiolar fibrosis. The blockage of apoptosis interferes with the macrophage "clearance" from alveolar spaces, favouring the accumulation of aging macrophages into alveoli and the progressive accumulation of iron pigment in long-lived senescent cells. The presence of areas of interstitial and alveolar fibrosis in peripheral parenchyma often accompanied the bronchiolar changes. Macrophages from smoking p66Shc-/- mice elaborate M2 cytokines (i.e., IL-4 and IL-13) and enzymes (i.e., chitinase and arginase I), which can promote TGF-beta expression, collagen deposition, and fibrosis in the surrounding areas. We demonstrate here that resistance to oxidative stress and p53-dependent apoptosis can modify tissue responses to CS caused by chronic inflammation without influencing early inflammatory response to CS exposure.


Subject(s)
Bronchiolitis/etiology , Bronchiolitis/genetics , Fibrosis/genetics , Shc Signaling Adaptor Proteins/genetics , Smoking/adverse effects , Animals , Apoptosis , Arginase/metabolism , Bronchiolitis/pathology , Chitinases/metabolism , Desmosine/metabolism , Hydroxyproline/metabolism , Interleukin-13/metabolism , Interleukin-4/metabolism , Lung/metabolism , Lung/pathology , Macrophages/enzymology , Macrophages/metabolism , Mice , Mice, Knockout , Oxidative Stress , Oxidoreductases/metabolism , Pulmonary Emphysema/pathology , Shc Signaling Adaptor Proteins/deficiency , Shc Signaling Adaptor Proteins/metabolism , Src Homology 2 Domain-Containing, Transforming Protein 1 , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Tumor Suppressor Protein p53/metabolism
4.
Eur Heart J ; 36(8): 516-26a, 2015 Feb 21.
Article in English | MEDLINE | ID: mdl-25336219

ABSTRACT

AIMS: Several intracellular mediators have been implicated as new therapeutic targets against myocardial ischaemia and reperfusion injury. However, clinically effective salvage pathways remain undiscovered. Here, we focused on the potential role of the adaptor protein p66(Shc) as a regulator of myocardial injury in a mouse model of cardiac ischaemia and reperfusion. METHODS AND RESULTS: Adult male p66(Shc) deficient (p66(Shc) (-/-)) and C57Bl/6 wild-type (WT) mice were exposed to 30, 45, or 60 min of ischaemia and reperfusion (5, 15 min, or 24 h). Infarct size, systemic and intracardiac inflammation and oxidants, as well as cytosolic and mitochondrial apoptotic pathways were investigated. Following 30, but not 45 or 60 min of ischaemia, genetic p66(Shc) deficiency was associated with larger infarcts. In WT mice, in vivo p66(Shc) knock down by siRNA with transient protein deficiency confirmed these findings. P66(Shc) inhibition was not associated with any modification in post-infarction inflammation, oxidative burst nor cardiac vessel density or structure. However, in p66(Shc) (-/-) mice activation of the protective and anti-apoptotic Reperfusion Injury Salvage Kinases and Survivor Activating Factor Enhancement pathways were blunted and mitochondrial swelling and cellular apoptosis via the caspase-3 pathway increased compared with WT. CONCLUSIONS: Genetic deletion of p66(Shc) increased susceptibility to myocardial injury in response to short-term ischaemia and reperfusion in mice. Still, additional studies are needed for assessing the role of this pathway in acute coronary syndrome patients.


Subject(s)
Gene Deletion , Myocardial Reperfusion Injury/genetics , Shc Signaling Adaptor Proteins/genetics , Animals , Apoptosis/genetics , Biomarkers/metabolism , Gene Knockdown Techniques , MAP Kinase Signaling System/genetics , Male , Mice, Inbred C57BL , Mitochondrial Swelling/genetics , Myocardial Reperfusion/methods , Myocardial Reperfusion Injury/pathology , Myocardium/metabolism , STAT3 Transcription Factor/genetics , Shc Signaling Adaptor Proteins/deficiency , Shc Signaling Adaptor Proteins/metabolism , Src Homology 2 Domain-Containing, Transforming Protein 1 , Troponin I/metabolism
5.
Aging Cell ; 13(6): 1049-58, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25257068

ABSTRACT

Adipose tissue is an important metabolic organ that integrates a wide array of homeostatic processes and is crucial for whole-body insulin sensitivity and energy metabolism. Brown adipose tissue (BAT) is a key thermogenic tissue with a well-established role in energy expenditure. BAT dissipates energy and protects against both hypothermia and obesity. Thus, BAT stimulation therapy is a rational strategy for the looming pandemic of obesity, whose consequences and comorbidities have a huge impact on the aged. Shc-deficient mice (ShcKO) were previously shown to be lean, insulin sensitive, and resistant to high-fat diet and obesity. We investigated the contribution of BAT to this phenotype. Insulin-dependent BAT glucose uptake was higher in ShcKO mice. Primary ShcKO BAT cells exhibited increased mitochondrial respiration; increased expression of several mitochondrial and lipid-oxidative enzymes was observed in ShcKO BAT. Levels of brown fat-specific markers of differentiation, UCP1, PRDM16, ELOVL3, and Cox8b, were higher in ShcKO BAT. In vitro, Shc knockdown in BAT cell line increased insulin sensitivity and metabolic activity. In vivo, pharmacological stimulation of ShcKO BAT resulted in higher energy expenditure. Conversely, pharmacological inhibition of BAT abolished the improved metabolic parameters, that is the increased insulin sensitivity and glucose tolerance of ShcKO mice. Similarly, in vitro Shc knockdown in BAT cell lines increased their expression of UCP1 and metabolic activity. These data suggest increased BAT activity significantly contributes to the improved metabolic phenotype of ShcKO mice.


Subject(s)
Adipose Tissue, Brown/metabolism , Shc Signaling Adaptor Proteins/deficiency , Shc Signaling Adaptor Proteins/metabolism , Animals , Energy Metabolism , Mice , Mice, Knockout , Thermogenesis/physiology
6.
Atherosclerosis ; 236(2): 426-9, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25150941

ABSTRACT

OBJECTIVE: Obesity-induced insulin resistance (IR) precipitates cardiovascular disease (CVD). Impairment of insulin signalling in the endothelium is emerging as a trigger of IR but the underlying mechanisms remain elusive. The mitochondrial adaptor p66(Shc) drives endothelial dysfunction via reactive oxygen species (ROS) generation. This study investigates p66(Shc) role in obesity-induced impairment of endothelial insulin signalling. METHODS: All experiments were performed in leptin-deficient (Lep(Ob/Ob)) and wild-type (WT) mice. RESULTS: Endothelium-dependent relaxations to insulin were blunted in Lep(Ob/Ob) as compared to WT. Interestingly, in vivo gene silencing of p66(Shc) restored insulin response via IRS-1/Akt/eNOS pathway. Furthermore, p66(Shc) knockdown in endothelial cells isolated from Lep(Ob/Ob) mice attenuated ROS production, free fatty acids (FFA) oxidation and prevented dysregulation of redox-sensitive pathways such as nuclear factor-kappa-B (NF-kB), AGE precursor methylglyoxal and PGI2 synthase. CONCLUSIONS: Targeting endothelial p66(Shc) may represent a promising strategy to prevent IR and CVD in obese individuals.


Subject(s)
Endothelium, Vascular/physiopathology , Insulin Resistance/physiology , Shc Signaling Adaptor Proteins/physiology , Animals , Cytochrome P-450 Enzyme System/metabolism , Fatty Acids, Nonesterified/metabolism , Insulin/pharmacology , Intramolecular Oxidoreductases/metabolism , Leptin/deficiency , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Obese , Molecular Targeted Therapy , NF-kappa B/metabolism , Nitric Oxide Synthase Type III/antagonists & inhibitors , Obesity/genetics , Obesity/metabolism , Oxidation-Reduction , Phenylephrine/pharmacology , Pyruvaldehyde/metabolism , RNA Interference , RNA, Small Interfering/pharmacology , Reactive Oxygen Species , Shc Signaling Adaptor Proteins/antagonists & inhibitors , Shc Signaling Adaptor Proteins/deficiency , Src Homology 2 Domain-Containing, Transforming Protein 1 , Superoxides/metabolism , Vasodilation/drug effects
7.
Cell Death Dis ; 5: e1068, 2014 Feb 20.
Article in English | MEDLINE | ID: mdl-24556683

ABSTRACT

Shc (Src homology 2 domain containing) adaptors are ubiquitous components of the signaling pathways triggered by tyrosine kinase-coupled receptors. In lymphocytes, similar to other cell types, the p52 and p66 isoforms of ShcA/Shc participate in a self-limiting loop where p52Shc acts as a positive regulator of antigen receptor signaling by promoting Ras activation, whereas p66Shc limits this activity by competitively inhibiting p52Shc. Based on the fact that many signaling mediators are shared by antigen and chemokine receptors, including p52Shc, we have assessed the potential implication of p66Shc in the regulation of B-cell responses to chemokines, focusing on the homing receptors CXCR4 (C-X-C chemokine receptor type 4) and CXCR5 (C-X-C chemokine receptor type 5). The results identify p66Shc as a negative regulator of the chemotactic responses triggered by these receptors, including adhesion, polarization and migration. We also provide evidence that this function is dependent on the ability of p66Shc to interact with the chemokine receptors and promote the assembly of an inhibitory complex, which includes the phosphatases SHP-1 (Src homology phosphatase-1) and SHIP-1 (SH2 domain-containing inositol 5'-phosphatase-1), that results in impaired Vav-dependent reorganization of the actin cytoskeleton. This function maps to the phosphorylatable tyrosine residues in the collagen homology 1 (CH1) domain. The results identify p66Shc as a negative regulator of B-cell chemotaxis and suggest a role for this adaptor in the control of B-cell homing.


Subject(s)
B-Lymphocytes/metabolism , Chemotaxis, Leukocyte , Receptors, CXCR4/metabolism , Receptors, CXCR5/metabolism , Shc Signaling Adaptor Proteins/metabolism , Animals , Cell Adhesion , Cell Line , Cytoskeleton/metabolism , Humans , Inositol Polyphosphate 5-Phosphatases , Mice , Mice, 129 Strain , Mice, Knockout , Phosphatidylinositol 3-Kinase/metabolism , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases , Phosphoric Monoester Hydrolases/metabolism , Phosphorylation , Protein Structure, Tertiary , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Proto-Oncogene Proteins c-vav/metabolism , Shc Signaling Adaptor Proteins/deficiency , Signal Transduction , Src Homology 2 Domain-Containing, Transforming Protein 1 , Time Factors , Transfection , Tyrosine , src-Family Kinases/metabolism
8.
J Leukoc Biol ; 95(2): 285-92, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24078705

ABSTRACT

The extracellular vesicular compartment has emerged as a novel system of intercellular communication; however, the mechanisms involved in membrane vesicle biogenesis and secretion are as yet unclear. Among immune cells releasing membrane vesicles-mast cells that reside near tissues exposed to the environment-are master modulators of immune responses. Here, we have addressed the role of p66Shc, a novel regulator of mast cell activation and homeostasis, in the dynamic reorganization of the actin cytoskeleton that is associated with morphological changes during secretion. We show that p66Shc is recruited as a complex with the lipid phosphatase SHIP1 to the F-actin skeleton and impairs antigen-dependent cortical F-actin disassembly and membrane ruffling through the inhibition of Vav and paxillin phosphorylation. We also show that in addition to acting as a negative regulator of antigen-dependent mast cell degranulation, p66Shc limits the basal release of granule contents by inhibiting microvesicle budding from the plasma membrane and piecemeal degranulation. These findings identify p66Shc as a critical regulator of actin dynamics in mast cells, providing a basis for understanding the molecular mechanisms involved in vesicle-mediated secretion in these cells.


Subject(s)
Actins/metabolism , Mast Cells/metabolism , Secretory Vesicles/metabolism , Shc Signaling Adaptor Proteins/metabolism , Animals , Bone Marrow Cells/cytology , Cell Degranulation , Cell Surface Extensions/metabolism , Cell Surface Extensions/ultrastructure , Cytoplasmic Granules/metabolism , Cytoplasmic Granules/ultrastructure , Cytoskeleton/metabolism , Mast Cells/cytology , Mast Cells/ultrastructure , Mice , Paxillin/metabolism , Phosphorylation , Proto-Oncogene Proteins c-vav/metabolism , Rats , Receptors, IgE/metabolism , Secretory Vesicles/ultrastructure , Shc Signaling Adaptor Proteins/deficiency , Src Homology 2 Domain-Containing, Transforming Protein 1 , Tetraspanin 30/metabolism
9.
Nature ; 499(7457): 166-71, 2013 Jul 11.
Article in English | MEDLINE | ID: mdl-23846654

ABSTRACT

Cell-surface receptors frequently use scaffold proteins to recruit cytoplasmic targets, but the rationale for this is uncertain. Activated receptor tyrosine kinases, for example, engage scaffolds such as Shc1 that contain phosphotyrosine (pTyr)-binding (PTB) domains. Using quantitative mass spectrometry, here we show that mammalian Shc1 responds to epidermal growth factor (EGF) stimulation through multiple waves of distinct phosphorylation events and protein interactions. After stimulation, Shc1 rapidly binds a group of proteins that activate pro-mitogenic or survival pathways dependent on recruitment of the Grb2 adaptor to Shc1 pTyr sites. Akt-mediated feedback phosphorylation of Shc1 Ser 29 then recruits the Ptpn12 tyrosine phosphatase. This is followed by a sub-network of proteins involved in cytoskeletal reorganization, trafficking and signal termination that binds Shc1 with delayed kinetics, largely through the SgK269 pseudokinase/adaptor protein. Ptpn12 acts as a switch to convert Shc1 from pTyr/Grb2-based signalling to SgK269-mediated pathways that regulate cell invasion and morphogenesis. The Shc1 scaffold therefore directs the temporal flow of signalling information after EGF stimulation.


Subject(s)
Epidermal Growth Factor/metabolism , Shc Signaling Adaptor Proteins/metabolism , Signal Transduction , Animals , Breast/cytology , Cell Line , Epithelial Cells/cytology , ErbB Receptors/agonists , ErbB Receptors/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Feedback, Physiological , GRB2 Adaptor Protein/deficiency , GRB2 Adaptor Protein/genetics , GRB2 Adaptor Protein/metabolism , Humans , Mice , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Phosphorylation , Protein Binding , Protein-Tyrosine Kinases , Proto-Oncogene Proteins c-akt/metabolism , Rats , Shc Signaling Adaptor Proteins/deficiency , Shc Signaling Adaptor Proteins/genetics , Src Homology 2 Domain-Containing, Transforming Protein 1 , Time Factors
10.
Oxid Med Cell Longev ; 2013: 719407, 2013.
Article in English | MEDLINE | ID: mdl-23766859

ABSTRACT

Mitochondrial-mediated oxidative stress and apoptosis play a crucial role in neurodegenerative disease and aging. Both mitochondrial permeability transition (PT) and swelling of mitochondria have been involved in neurodegeneration. Indeed, knockout mice for cyclophilin-D (Cyc-D), a key regulatory component of the PT pore (PTP) that triggers mitochondrial swelling, resulted to be protected in preclinical models of multiple sclerosis (MS), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS). However, how neuronal stress is transduced into mitochondrial oxidative stress and swelling is unclear. Recently, the aging determinant p66Shc that generates H2O2 reacting with cytochrome c and induces oxidation of PTP and mitochondrial swelling was found to be involved in MS and ALS. To investigate the role of p66Shc/PTP pathway in neurodegeneration, we performed experimental autoimmune encephalomyelitis (EAE) experiments in p66Shc knockout mice (p66Shc-/-), knock out mice for cyclophilin-D (Cyc-D-/-), and p66Shc Cyc-D double knock out (p66Shc/Cyc-D-/-) mice. Results confirm that deletion of p66Shc protects from EAE without affecting immune response, whereas it is not epistatic to the Cyc-D mutation. These findings demonstrate that p66Shc contributes to EAE induced neuronal damage most likely through the opening of PTP suggesting that p66Shc/PTP pathway transduces neurodegenerative stresses.


Subject(s)
Mitochondrial Membrane Transport Proteins/metabolism , Nerve Degeneration/metabolism , Shc Signaling Adaptor Proteins/metabolism , Animals , Apoptosis , Cell Line , Peptidyl-Prolyl Isomerase F , Cyclophilins/deficiency , Cyclophilins/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Enzyme-Linked Immunosorbent Assay , Gene Deletion , Humans , Mice , Mice, Knockout , Mitochondrial Permeability Transition Pore , Nerve Degeneration/pathology , Neurons/metabolism , Neurons/pathology , Rats , Shc Signaling Adaptor Proteins/deficiency , Spinal Cord/metabolism , Spinal Cord/pathology , Src Homology 2 Domain-Containing, Transforming Protein 1
11.
Circ Res ; 113(1): 32-39, 2013 Jun 21.
Article in English | MEDLINE | ID: mdl-23661718

ABSTRACT

RATIONALE: Arteriogenesis, the shear stress-driven remodeling of collateral arteries, is critical in restoring blood flow to ischemic tissue after a vascular occlusion. Our previous work has shown that the adaptor protein Shc mediates endothelial responses to shear stress in vitro. OBJECTIVE: To examine the role of the adaptor protein Shc in arteriogenesis and endothelial-dependent responses to shear stress in vivo. METHODS AND RESULTS: Conditional knockout mice in which Shc is deleted from endothelial cells were subjected to femoral artery ligation. Hindlimb perfusion recovery was attenuated in Shc conditional knockout mice compared with littermate controls. Reduced perfusion was associated with blunted collateral remodeling and reduced capillary density. Bone marrow transplantation experiments revealed that endothelial Shc is required for perfusion recovery because loss of Shc in bone marrow-derived hematopoietic cells had no effect on recovery. Mechanistically, Shc deficiency resulted in impaired activation of the nuclear factor κ-light-chain-enhancer of activated B-cell-dependent inflammatory pathway and reduced CD45⁺ cell infiltration. Unexpectedly, Shc was required for arterial specification of the remodeling arteriole by mediating upregulation of the arterial endothelial cell marker ephrinB2 and activation of the Notch pathway. In vitro experiments confirmed that Shc was required for shear stress-induced activation of the Notch pathway and downstream arterial specification through a mechanism that involves upregulation of Notch ligands delta-like 1 and delta-like 4. CONCLUSIONS: Shc mediates activation of 2 key signaling pathways that are critical for inflammation and arterial specification; collectively, these pathways contribute to arteriogenesis and the recovery of blood perfusion.


Subject(s)
Arteritis/etiology , Ischemia/physiopathology , NF-kappa B/physiology , Neovascularization, Physiologic/genetics , Receptors, Notch/physiology , Shc Signaling Adaptor Proteins/physiology , Adaptor Proteins, Signal Transducing , Animals , Arteritis/genetics , Bone Marrow Transplantation , Calcium-Binding Proteins , Cell Adhesion , Collateral Circulation , Endothelial Cells/metabolism , Ephrin-B2/physiology , Femoral Artery/surgery , Genes, Synthetic , Hematopoietic Stem Cells/metabolism , Hemorheology , Hindlimb/blood supply , Intercellular Signaling Peptides and Proteins/physiology , Intracellular Signaling Peptides and Proteins/physiology , Leukocytes/physiology , Ligation , Male , Mechanoreceptors/physiology , Membrane Proteins/physiology , Mice , Mice, Knockout , Shc Signaling Adaptor Proteins/deficiency , Shc Signaling Adaptor Proteins/genetics , Signal Transduction , Stress, Mechanical
12.
Exp Mol Pathol ; 94(3): 445-52, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23506954

ABSTRACT

Overwhelming oxidative stress and compromised tubular cell antioxidant response have been incriminated for cisplatin (Cis)-induced acute kidney injury (AKI). We hypothesized that Cis-induced AKI was the outcome of the deactivated redox-sensitive stress response program (RSSRP). Wild type (WT) and heterozygous p66ShcA(p66(+/-)) mice in groups of six were administered either normal saline (WT) or Cis (12.5 mg/kg, intraperitoneal, Cis/WT). Renal biomarkers were collected and kidneys were harvested for renal histology. Cis/WT showed elevated blood urea nitrogen levels and enhanced tubular cell apoptosis, necrosis, and dilated tubules filled with casts when compared to Cis/p66(+/-). Cis/p66(+/-) developed only a clinically occult AKI (normal blood urea levels and only microscopic alterations). Immunoblots from the lysates of renal tissues of Cis/WT displayed enhanced expression of phospho-p66ShcA, and phospho-Foxo3A but attenuated expression of MnSOD and catalase; conversely, p66 deficit prevented these alterations in Cis milieu. In in vitro studies, Cis treated mouse proximal tubular cells (MPTCs) displayed enhanced phosphorylation of p66ShcA and no increase in tubular cell expression of MnSOD. In addition, renal tissues of Cis/WT and Cis-treated MPTCs displayed enhanced phosphorylation of p53 and Bax expression. However, MPTC partially silenced for p66ShcA displayed partial inhibition of Cis-induced tubular cell apoptosis as well as necrosis. These findings indicate that Cis-induced AKI is the outcome of the deactivated RSSRP (attenuated anti-oxidant response) and activation of pro-apoptotic (p53-induced Bax expression) pathway.


Subject(s)
Acute Kidney Injury/metabolism , Antineoplastic Agents/toxicity , Cisplatin/toxicity , Oxidoreductases/metabolism , Shc Signaling Adaptor Proteins/deficiency , Shc Signaling Adaptor Proteins/metabolism , Acute Kidney Injury/chemically induced , Acute Kidney Injury/pathology , Animals , Apoptosis/drug effects , Blood Urea Nitrogen , Female , Gene Silencing , Heterozygote , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Male , Mice , Mice, Knockout , Necrosis/chemically induced , Oxidation-Reduction , Oxidative Stress/drug effects , Phosphorylation , Shc Signaling Adaptor Proteins/genetics , Tumor Suppressor Protein p53/metabolism , bcl-2-Associated X Protein/metabolism
13.
Methods Mol Biol ; 965: 341-53, 2013.
Article in English | MEDLINE | ID: mdl-23296670

ABSTRACT

Nutrient availability and nutrient-dependent biochemical signals represent major determinants of cellular senescence and organismal aging. The present chapter describes simple procedures to reliably evaluate the response of cultured cell to nutrients through the longevity protein p66(SHC1) and the mTOR/S6K cascade, which might be used to study cellular senescence and its chemical modulation by pharmaceutical agents in vitro.


Subject(s)
Longevity , Shc Signaling Adaptor Proteins/metabolism , Signal Transduction , Aging , Electrophoresis , Gene Knockdown Techniques , HEK293 Cells , Humans , Phosphorylation , Shc Signaling Adaptor Proteins/deficiency , Shc Signaling Adaptor Proteins/genetics , Shc Signaling Adaptor Proteins/isolation & purification , Src Homology 2 Domain-Containing, Transforming Protein 1 , Staining and Labeling , Transfection
14.
Aging Cell ; 12(2): 177-83, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23237310

ABSTRACT

Upon oxidative challenge the genome accumulates adducts and breaks that activate the DNA damage response to repair, arrest, or eliminate the damaged cell. Thus, reactive oxygen species (ROS) generated by endogenous oxygen metabolism are thought to affect mutation frequency. However, few studies determined the mutation frequency when oxidative stress is reduced. To test whether in vivo spontaneous mutation frequency is altered in mice with reduced oxidative stress and cell death rate, we crossed p66Shc knockout (p66KO) mice, characterized by reduced intracellular concentration of ROS and by impaired apoptosis, with a transgenic line harboring multiple copies of the lacZ mutation reporter gene as part of a plasmid that can be recovered from organs into Escherichia coli to measure mutation rate. Liver and small intestine from 2- to 24-month-old, lacZ (p66Shc+/+) and lacZp66KO mice, were investigated revealing no difference in overall mutation frequency but a significant increase in the frequency of size-change mutations in the intestine of lacZp66KO mice. This difference was further increased upon irradiation of mice with X-ray. In addition, we found that knocking down cyclophilin D, a gene that facilitates mitochondrial apoptosis acting downstream of p66Shc, increased the size-change mutation frequency in small intestine. Size-change mutations also accumulated in death-resistant embryonic fibroblasts from lacZp66KO mice treated with H2 O2 . These results indicate that p66Shc plays a role in the accumulation of DNA rearrangements and suggest that p66Shc functions to clear damaged cells rather than affect DNA metabolism.


Subject(s)
Intestine, Small/metabolism , Lac Operon , Liver/metabolism , Mutation Rate , Shc Signaling Adaptor Proteins/genetics , Animals , Peptidyl-Prolyl Isomerase F , Cyclophilins/genetics , Cyclophilins/metabolism , Escherichia coli/genetics , Gamma Rays , Gene Expression Regulation/drug effects , Gene Expression Regulation/radiation effects , Hydrogen Peroxide/pharmacology , Intestine, Small/drug effects , Intestine, Small/radiation effects , Liver/drug effects , Liver/radiation effects , Mice , Mice, Knockout , Mutation , Oxidative Stress , Reactive Oxygen Species/metabolism , Shc Signaling Adaptor Proteins/deficiency , Signal Transduction/drug effects , Signal Transduction/radiation effects , Src Homology 2 Domain-Containing, Transforming Protein 1 , Transgenes
15.
Am J Physiol Renal Physiol ; 303(12): F1629-40, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23019230

ABSTRACT

Candidate genes have been identified that confer increased risk for diabetic glomerulosclerosis (DG). Mice heterozygous for the Akita (Ins2(+/C96Y)) diabetogenic mutation with a second mutation introduced at the bradykinin 2 receptor (B2R(-/-)) locus express a disease phenotype that approximates human DG. Src homology 2 domain transforming protein 1 (p66) controls mitochondrial metabolism and cellular responses to oxidative stress, aging, and apoptosis. We generated p66-null Akita mice to test whether inactivating mutations at the p66 locus will rescue kidneys of Akita mice from disease-causing mutations at the Ins2 and B2R loci. Here we show null mutations at the p66 and B2R loci interact with the Akita (Ins2(+/C96Y)) mutation, independently and in combination, inducing divergent phenotypes in the kidney. The B2R(-/-) mutation induces detrimental phenotypes, as judged by increased systemic and renal levels of oxidative stress, histology, and urine albumin excretion, whereas the p66-null mutation confers a powerful protection phenotype. To elucidate the mechanism(s) of the protection phenotype, we turned to our in vitro system. Experiments with cultured podocytes revealed previously unrecognized cross talk between p66 and the redox-sensitive transcription factor p53 that controls hyperglycemia-induced ROS metabolism, transcription of p53 target genes (angiotensinogen, angiotensin II type-1 receptor, and bax), angiotensin II generation, and apoptosis. RNA-interference targeting p66 inhibits all of the above. Finally, protein levels of p53 target genes were upregulated in kidneys of Akita mice but unchanged in p66-null Akita mice. Taken together, p66 is a potential molecular target for therapeutic intervention in DG.


Subject(s)
Diabetes Mellitus/genetics , Insulin/genetics , Mutation/genetics , Phenotype , Receptor, Bradykinin B2/genetics , Shc Signaling Adaptor Proteins/genetics , Animals , Apoptosis , Cells, Cultured , Diabetes Mellitus/metabolism , Diabetes Mellitus/pathology , Disease Models, Animal , Hyperglycemia/metabolism , Hyperglycemia/pathology , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Podocytes/metabolism , Podocytes/pathology , Receptor, Bradykinin B2/deficiency , Receptor, Bradykinin B2/metabolism , Shc Signaling Adaptor Proteins/deficiency , Shc Signaling Adaptor Proteins/metabolism , Src Homology 2 Domain-Containing, Transforming Protein 1 , Tumor Suppressor Protein p53/metabolism
17.
Circ Res ; 111(3): 278-89, 2012 Jul 20.
Article in English | MEDLINE | ID: mdl-22693349

ABSTRACT

RATIONALE: Hyperglycemic memory may explain why intensive glucose control has failed to improve cardiovascular outcomes in patients with diabetes. Indeed, hyperglycemia promotes vascular dysfunction even after glucose normalization. However, the molecular mechanisms of this phenomenon remain to be elucidated. OBJECTIVE: The present study investigated the role of mitochondrial adaptor p66(Shc) in this setting. METHODS AND RESULTS: In human aortic endothelial cells (HAECs) exposed to high glucose and aortas of diabetic mice, activation of p66(Shc) by protein kinase C ßII (PKCßII) persisted after returning to normoglycemia. Persistent p66(Shc) upregulation and mitochondrial translocation were associated with continued reactive oxygen species (ROS) production, reduced nitric oxide bioavailability, and apoptosis. We show that p66(Shc) gene overexpression was epigenetically regulated by promoter CpG hypomethylation and general control nonderepressible 5-induced histone 3 acetylation. Furthermore, p66(Shc)-derived ROS production maintained PKCßII upregulation and PKCßII-dependent inhibitory phosphorylation of endothelial nitric oxide synthase at Thr-495, leading to a detrimental vicious cycle despite restoration of normoglycemia. Moreover, p66(Shc) activation accounted for the persistent elevation of the advanced glycated end product precursor methylglyoxal. In vitro and in vivo gene silencing of p66(Shc), performed at the time of glucose normalization, blunted ROS production, restored endothelium-dependent vasorelaxation, and attenuated apoptosis by limiting cytochrome c release, caspase 3 activity, and cleavage of poly (ADP-ribose) polymerase. CONCLUSIONS: p66(Shc) is the key effector driving vascular hyperglycemic memory in diabetes. Our study provides molecular insights for the progression of diabetic vascular complications despite glycemic control and may help to define novel therapeutic targets.


Subject(s)
Diabetes Mellitus, Experimental/genetics , Gene Silencing , Hyperglycemia/genetics , Hyperglycemia/physiopathology , Mitochondria/pathology , Shc Signaling Adaptor Proteins/deficiency , Shc Signaling Adaptor Proteins/genetics , Animals , Cells, Cultured , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Gene Knockdown Techniques/methods , Gene Silencing/physiology , Glucose/pharmacology , Humans , Hyperglycemia/pathology , Male , Mice , Mice, 129 Strain , Mitochondria/drug effects , Organ Culture Techniques , Shc Signaling Adaptor Proteins/physiology , Src Homology 2 Domain-Containing, Transforming Protein 1
18.
Eur J Neurosci ; 35(4): 562-71, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22277070

ABSTRACT

Although multiple sclerosis (MS) has traditionally been considered to be an inflammatory disease, recent evidence has brought neurodegeneration into the spotlight, suggesting that accumulated damage and loss of axons is critical to disease progression and the associated irreversible disability. Proposed mechanisms of axonal degeneration in MS posit cytosolic and subsequent mitochondrial Ca(2+) overload, accumulation of pathologic reactive oxygen species (ROS), and mitochondrial dysfunction leading to cell death. In this context, the role of the p66 isoform of ShcA protein (p66) may be significant. The ShcA isoform is uniquely targeted to the mitochondrial intermembrane space in response to elevated oxidative stress, and serves as a redox enzyme amplifying ROS generation in a positive feedforward loop that eventually mediates cell death by activation of the mitochondrial permeability transition pore. Consequently, we tested the hypothesis that genetic inactivation of p66 would reduce axonal injury in a murine model of MS, experimental autoimmune encephalomyelitis (EAE). As predicted, the p66-knockout (p66-KO) mice developed typical signs of EAE, but had less severe clinical impairment and paralysis than wild-type (WT) mice. Histologic examination of spinal cords and optic nerves showed significant axonal protection in the p66-KO tissue, despite similar levels of inflammation. Furthermore, cultured p66-KO neurons treated with agents implicated in MS neurodegenerative pathways showed greater viability than WT neurons. These results confirm the critical role of ROS-mediated mitochondrial dysfunction in the axonal loss that accompanies EAE, and identify p66 as a new pharmacologic target for MS neuroprotective therapeutics.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Shc Signaling Adaptor Proteins/deficiency , Animals , Axons/pathology , Axons/ultrastructure , Cell Proliferation , Cells, Cultured , Cerebral Cortex/cytology , Peptidyl-Prolyl Isomerase F , Cyclophilins/deficiency , Cytokines/metabolism , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/pathology , Freund's Adjuvant/adverse effects , Glycoproteins/adverse effects , Hydrogen Peroxide/pharmacology , Leukemic Infiltration/drug therapy , Leukemic Infiltration/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Transmission , Myelin-Oligodendrocyte Glycoprotein , Nerve Fibers, Myelinated/pathology , Neurons/metabolism , Neurons/ultrastructure , Optic Nerve/immunology , Optic Nerve/metabolism , Optic Nerve/pathology , Optic Nerve/ultrastructure , Peptide Fragments/adverse effects , Shc Signaling Adaptor Proteins/metabolism , Spinal Cord/immunology , Spinal Cord/metabolism , Spinal Cord/pathology , Spinal Cord/ultrastructure , Src Homology 2 Domain-Containing, Transforming Protein 1 , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
19.
J Cell Biochem ; 113(2): 599-610, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21956685

ABSTRACT

Cancer chemopreventive response to D,L-sulforaphane (SFN), a synthetic racemic analogue of broccoli constituent L-sulforaphane, is partly attributable to apoptosis induction, but the mechanism of cell death is not fully understood. The present study demonstrates a critical role for adapter protein p66(Shc) in SFN-induced apoptosis. Immortalized mouse embryonic fibroblasts (MEF) derived from p66(shc) knockout mice were significantly more resistant to SFN-induced apoptosis, collapse of mitochondrial membrane potential, and reactive oxygen species (ROS) production compared with MEF obtained from the wild-type mice. Notably, a spontaneously immortalized and non-tumorigenic human mammary epithelial cell line (MCF-10A) was resistant to SFN-induced ROS production and apoptosis. Stable overexpression of manganese superoxide dismutase in MCF-7 and MDA-MB-231 human breast cancer cells conferred near complete protection against SFN-induced apoptosis and mitochondrial membrane potential collapse. SFN treatment resulted in increased S36 phosphorylation and mitochondrial translocation of p66(shc) in MDA-MB-231 and MCF-7 cells, and SFN-induced apoptosis was significantly attenuated by RNA interference of p66(shc) in both cells. SFN-treated MDA-MB-231 and MCF-7 cells also exhibited a marked decrease in protein level of peptidyl prolyl isomerase (Pin1), which is implicated in mitochondrial translocation of p66(shc) . However, stable overexpression of Pin1 failed to alter proapoptotic response to SFN at least in MCF-7 cells. Finally, SFN-induced S36 phosphorylation of p66(Shc) was mediated by protein kinase Cß (PKCß), and pharmacological inhibition of PKCß significantly inhibited apoptotic cell death resulting from SFN exposure. In conclusion, the present study provides new insight into the mechanism of SFN-induced apoptosis involving PKCß -mediated S36 phosphorylation of p66(shc).


Subject(s)
Anticarcinogenic Agents/pharmacology , Apoptosis/drug effects , Shc Signaling Adaptor Proteins/metabolism , Thiocyanates/pharmacology , Animals , Breast Neoplasms , Cell Line, Tumor , Cell Survival/drug effects , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Gene Knockout Techniques , Humans , Isothiocyanates , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Knockout , NIMA-Interacting Peptidylprolyl Isomerase , Peptidylprolyl Isomerase/genetics , Peptidylprolyl Isomerase/metabolism , Phosphorylation , Protein Kinase C/metabolism , Protein Kinase C beta , Reactive Oxygen Species/metabolism , Shc Signaling Adaptor Proteins/deficiency , Shc Signaling Adaptor Proteins/genetics , Src Homology 2 Domain-Containing, Transforming Protein 1 , Sulfoxides , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism
20.
Aging Cell ; 11(1): 162-8, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22081964

ABSTRACT

Deletion of the p66(Shc) gene results in lean and healthy mice, retards aging, and protects from aging-associated diseases, raising the question of why p66(Shc) has been selected, and what is its physiological role. We have investigated survival and reproduction of p66(Shc)-/- mice in a population living in a large outdoor enclosure for a year, subjected to food competition and exposed to winter temperatures. Under these conditions, deletion of p66(Shc) was strongly counterselected. Laboratory studies revealed that p66(Shc)-/- mice have defects in fat accumulation, thermoregulation, and reproduction, suggesting that p66(Shc) has been evolutionarily selected because of its role in energy metabolism. These findings imply that the health impact of targeting aging genes might depend on the specific energetic niche and caution should be exercised against premature conclusions regarding gene functions that have only been observed in protected laboratory conditions.


Subject(s)
Aging/genetics , Longevity/genetics , Shc Signaling Adaptor Proteins/genetics , Aging/metabolism , Animals , Biological Evolution , Body Temperature Regulation/genetics , Energy Metabolism/genetics , Female , Genetic Fitness/genetics , Heterozygote , Homozygote , Lipid Metabolism/genetics , Male , Mice , Mice, Knockout , Seasons , Shc Signaling Adaptor Proteins/deficiency , Src Homology 2 Domain-Containing, Transforming Protein 1
SELECTION OF CITATIONS
SEARCH DETAIL
...