Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Biomedicines ; 9(4)2021 Apr 08.
Article in English | MEDLINE | ID: mdl-33917746

ABSTRACT

Currently, engineered skin substitutes (ESS) are unable to regenerate cutaneous appendages. Recent studies have shown that skin-derived precursors (SKPs), which are extensively available, have the potential to induce hair follicle neogenesis. Here, we demonstrate that ESS consisting of culture-expanded SKPs and epidermal stem cells (Epi-SCs) reconstitute the skin with hair follicle regeneration after grafting into nude mice. SKPs seeded in a C-GAG matrix proliferated and expressed higher levels of hair induction signature genes-such as Akp2, Sox2, CD133 and Bmp6-compared to dermal fibroblasts. Moreover, when ESS prepared by seeding a mixture of culture-expanded murine SKPs and human adult Epi-SCs into a C-GAG matrix was grafted into full-thickness skin wounds in nude mice, black hairs were generated within 3 weeks. Immunofluorescence analysis showed that the SKPs were localized to the dermal papillae of the newly-formed hair follicle. Our results indicate that SKPs can serve as the hair-inductive cells in ESS to furnish it with hair genesis potential.

2.
Gene Ther ; 28(3-4): 155-161, 2021 04.
Article in English | MEDLINE | ID: mdl-32994568

ABSTRACT

Follicle-stimulating hormone (FSH) is a pituitary glycoprotein that regulates follicle maturation through its binding to follicle-stimulating hormone receptor (FSHR). Endothelial cells express FSHR, but its exact role in endothelial cells remains unclear. Here we show that FSHR expression was detectable in human umbilical vein endothelial cells (HUVECs). FSH stimulation promoted HUVECs migration but not proliferation. Because FSHR is a GPCR, FSH treatment triggers the activation of cAMP-PKA signaling pathways, and the JAK-STAT, PI3K-AKT, and JNK-MAPK pathways. RNAi of FSHR dramatically attenuated the activation effect of FSH on HUVECs migration, as well as the related signaling pathways. Treatment of FSH in HUVECs also transcriptionally upregulated the expression of VAV3 and LAMA2, suppression either of VAV3 or LAMA2 by RNAi attenuated the FSH's effect on HUVECs migration. All of these results indicated a functional role of FSH in the regulation of endothelial cells.


Subject(s)
Cell Movement , Follicle Stimulating Hormone, Human/metabolism , Follicle Stimulating Hormone , Receptors, FSH/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Phosphatidylinositol 3-Kinases/metabolism , Receptors, FSH/genetics , Signal Transduction
3.
ACS Med Chem Lett ; 11(7): 1392-1401, 2020 Jul 09.
Article in English | MEDLINE | ID: mdl-32676145

ABSTRACT

Dysregulation of the inflammatory response is a key driver of many debilitating and costly diseases including immune disorders, cancer, and infection. Pyroptosis is a highly inflammatory form of programmed cell death, triggered by various stimuli and meditated by the activation of inflammatory caspases. Pharmacologic agents that provide strategies to modulate pyroptosis for research and clinical practice are still very limited. In current study, we identify 3-difluoroalkyl quaternary oxindoles as chemical inhibitors of caspase-1, the pyroptosis driving caspase. Our results demonstrated compound 6 could directly bind to the CARD domain of pro-caspase-1 to inhibit its infammasome recruitment and pharmacologic inhibition of pyroptotic cell death by compound 6 is partially efficacious in sepsis models. Compound 6 is thus a potential therapeutic for inflammatory disorders and a tool for further study of the inflammation in human health and disease.

4.
Proc Natl Acad Sci U S A ; 117(9): 4770-4780, 2020 03 03.
Article in English | MEDLINE | ID: mdl-32071245

ABSTRACT

Recurrence and metastasis remain the major obstacles to successful treatment of hepatocellular carcinoma (HCC). Chromatin remodeling factor ARID2 is commonly mutated in HCC, indicating its important role in cancer development. However, its role in HCC metastasis is largely elusive. In this study, we find that ARID2 expression is significantly decreased in metastatic HCC tissues, showing negative correlation with pathological grade, organ metastasis and positive association with survival of HCC patients. ARID2 inhibits migration and invasion of HCC cells in vitro and metastasis in vivo. Moreover, ARID2 knockout promotes pulmonary metastasis in different HCC mouse models. Mechanistic study reveals that ARID2 represses epithelial-mesenchymal transition (EMT) of HCC cells by recruiting DNMT1 to Snail promoter, which increases promoter methylation and inhibits Snail transcription. In addition, we discover that ARID2 mutants with disrupted C2H2 domain lose the metastasis suppressor function, exhibiting a positive association with HCC metastasis and poor prognosis. In conclusion, our study reveals the metastasis suppressor role as well as the underlying mechanism of ARID2 in HCC and provides a potential therapeutic target for ARID2-deficient HCC.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , Chromatin Assembly and Disassembly/physiology , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , Liver Neoplasms/drug therapy , Neoplasm Metastasis/drug therapy , Transcription Factors/metabolism , Animals , CYS2-HIS2 Zinc Fingers , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Movement/drug effects , Disease Models, Animal , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Mice , Mice, Knockout , Mutation , Neoplasm Metastasis/pathology , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics
5.
J Clin Endocrinol Metab ; 105(3)2020 Mar 01.
Article in English | MEDLINE | ID: mdl-31652310

ABSTRACT

CONTEXT: Obesity-related insulin resistance (OIR) is one of the main contributors to type 2 diabetes and other metabolic diseases. Protein kinases are implicated in insulin signaling and glucose metabolism. Molecular mechanisms underlying OIR involving global kinase activities remain incompletely understood. OBJECTIVE: To investigate abnormal kinase activity associated with OIR in human skeletal muscle. DESIGN: Utilization of stable isotopic labeling-based quantitative proteomics combined with affinity-based active enzyme probes to profile in vivo kinase activity in skeletal muscle from lean control (Lean) and OIR participants. PARTICIPANTS: A total of 16 nondiabetic adults, 8 Lean and 8 with OIR, underwent hyperinsulinemic-euglycemic clamp with muscle biopsy. RESULTS: We identified the first active kinome, comprising 54 active protein kinases, in human skeletal muscle. The activities of 23 kinases were different in OIR muscle compared with Lean muscle (11 hyper- and 12 hypo-active), while their protein abundance was the same between the 2 groups. The activities of multiple kinases involved in adenosine monophosphate-activated protein kinase (AMPK) and p38 signaling were lower in OIR compared with Lean. On the contrary, multiple kinases in the c-Jun N-terminal kinase (JNK) signaling pathway exhibited higher activity in OIR vs Lean. The kinase-substrate-prediction based on experimental data further confirmed a potential downregulation of insulin signaling (eg, inhibited phosphorylation of insulin receptor substrate-1 and AKT1/2). CONCLUSIONS: These findings provide a global view of the kinome activity in OIR and Lean muscle, pinpoint novel specific impairment in kinase activities in signaling pathways important for skeletal muscle insulin resistance, and may provide potential drug targets (ie, abnormal kinase activities) to prevent and/or reverse skeletal muscle insulin resistance in humans.


Subject(s)
Insulin Resistance , Muscle, Skeletal/enzymology , Obesity/metabolism , Protein Kinases/physiology , Proteome , AMP-Activated Protein Kinases/physiology , Adult , Female , Humans , Male , Signal Transduction/physiology , p38 Mitogen-Activated Protein Kinases/physiology
6.
World J Stem Cells ; 11(8): 535-547, 2019 Aug 26.
Article in English | MEDLINE | ID: mdl-31523372

ABSTRACT

Human hepatocyte-like cells (HLCs) derived from human pluripotent stem cells (hPSCs) promise a valuable source of cells with human genetic background, physiologically relevant liver functions, and unlimited supply. With over 10 years' efforts in this field, great achievements have been made. HLCs have been successfully derived and applied in disease modeling, toxicity testing and drug discovery. Large cohorts of induced pluripotent stem cells-derived HLCs have been recently applied in studying population genetics and functional outputs of common genetic variants in vitro. This has offered a new paradigm for genome-wide association studies and possibly in vitro pharmacogenomics in the nearly future. However, HLCs have not yet been successfully applied in bioartificial liver devices and have only displayed limited success in cell transplantation. HLCs still have an immature hepatocyte phenotype and exist as a population with great heterogeneity, and HLCs derived from different hPSC lines display variable differentiation efficiency. Therefore, continuous improvement to the quality of HLCs, deeper investigation of relevant biological processes, and proper adaptation of recent advances in cell culture platforms, genome editing technology, and bioengineering systems are required before HLCs can fulfill the needs in basic and translational research. In this review, we summarize the discoveries, achievements, and challenges in the derivation and applications of HLCs.

7.
Cell Rep ; 26(4): 884-892.e4, 2019 01 22.
Article in English | MEDLINE | ID: mdl-30673611

ABSTRACT

DNA variants in the SLC16A11 coding region were identified to be strongly associated with type 2 diabetes (T2DM) in a Mexican population. Previous studies suggested that these variants disrupt SLC16A11 function and therefore proposed to revive SLC16A11 levels or activity to achieve therapeutic benefit. However, with knockout mouse models, here we show that Slc16a11 depletion has no significant metabolic defects. Further studies demonstrate that reconstitution of the mutant, but not the wild-type Slc16a11, in the liver of knockout mice causes more triglyceride accumulation and induction of insulin resistance via upregulation of lipin 1, suggesting gaining of aberrant functions of the mutant protein that affects lipid metabolism. Our findings offer a different explanation to the function of these diabetic variants, challenging the concept of enhancing SLC16A11 function to treat T2DM. The contradictory results by our and previous studies suggest that how the SLC16A11 locus contributes to human metabolism warrants further investigation.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2 , Gain of Function Mutation , Insulin Resistance/genetics , Monocarboxylic Acid Transporters , Triglycerides , Animals , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , HEK293 Cells , Humans , Mice , Mice, Knockout , Monocarboxylic Acid Transporters/genetics , Monocarboxylic Acid Transporters/metabolism , Phosphatidate Phosphatase/genetics , Phosphatidate Phosphatase/metabolism , Triglycerides/genetics , Triglycerides/metabolism
8.
J Proteome Res ; 17(9): 2963-2977, 2018 09 07.
Article in English | MEDLINE | ID: mdl-30014700

ABSTRACT

Charcoal-stripped fetal bovine serum (CS-FBS) is commonly used to study androgen responsiveness and androgen metabolism in cultured prostate cancer (CaP) cells. Switching CaP cells from FBS to CS-FBS may reduce the activity of androgen receptor (AR), inhibit cell proliferation, or modulate intracellular androgen metabolism. The removal of proteins by charcoal stripping may cause changes in biological functions and has not yet been investigated. Here we profiled proteins in FBS and CS-FBS using an ion-current-based quantitative platform consisting of reproducible surfactant-aided precipitation/on-pellet digestion, long-column nanoliquid chromatography separation, and ion-current-based analysis. A total of 143 proteins were identified in FBS, among which 14 proteins including insulin-like growth factor 2 (IGF-2) and IGF binding protein (IGFBP)-2 and -6 were reduced in CS-FBS. IGF-1 receptor (IGF1R) and insulin receptor were sensitized to IGFs in CS-FBS. IGF-1 and IGF-2 stimulation fully compensated for the loss of AR activity to maintain cell growth in CS-FBS. Endogenous production of IGF and IGFBPs was verified in CaP cells and clinical CaP specimens. This study provided the most comprehensive protein profiles of FBS and CS-FBS and offered an opportunity to identify new protein regulators and signaling pathways that regulate AR activity, androgen metabolism, and proliferation of CaP cells.


Subject(s)
Blood Proteins/isolation & purification , Epithelial Cells/drug effects , Prostatic Neoplasms/metabolism , Proteomics/methods , Testosterone/pharmacology , Adsorption , Animals , Blood Proteins/chemistry , Cattle , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Charcoal/chemistry , Culture Media/chemistry , Culture Media/pharmacology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Fetus , Gene Expression , Humans , Insulin-Like Growth Factor Binding Protein 2/isolation & purification , Insulin-Like Growth Factor Binding Protein 6/isolation & purification , Insulin-Like Growth Factor I/isolation & purification , Insulin-Like Growth Factor I/pharmacology , Insulin-Like Growth Factor II/isolation & purification , Insulin-Like Growth Factor II/pharmacology , Male , Prostate/drug effects , Prostate/metabolism , Prostate/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Receptor, IGF Type 1/isolation & purification , Receptor, Insulin/isolation & purification , Receptors, Androgen/biosynthesis , Receptors, Androgen/genetics , Testosterone/isolation & purification
9.
Stem Cell Reports ; 11(1): 22-31, 2018 07 10.
Article in English | MEDLINE | ID: mdl-29861165

ABSTRACT

Hepatocyte-like cells (HLCs) derived from human pluripotent stem cells (hPSCs) offer a promising cell resource for disease modeling and transplantation. However, differentiated HLCs exhibit an immature phenotype and comprise a heterogeneous population. Thus, a better understanding of HLC differentiation will improve the likelihood of future application. Here, by taking advantage of CRISPR-Cas9-based genome-wide screening technology and a high-throughput hPSC screening platform with a reporter readout, we identified several potential genetic regulators of HLC differentiation. By using a chemical screening approach within our platform, we also identified compounds that can further promote HLC differentiation and preserve the characteristics of in vitro cultured primary hepatocytes. Remarkably, both screenings identified histone deacetylase 3 (HDAC3) as a key regulator in hepatic differentiation. Mechanistically, HDAC3 formed a complex with liver transcriptional factors, e.g., HNF4, and co-regulated the transcriptional program during hepatic differentiation. This study highlights a broadly useful approach for studying and optimizing hPSC differentiation.


Subject(s)
Cell Differentiation , Hepatocytes/cytology , Hepatocytes/metabolism , Histone Deacetylases/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Benzamides , CRISPR-Cas Systems , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line , Cells, Cultured , Flow Cytometry , Gene Editing , Gene Expression Regulation, Developmental , Gene Targeting , Genes, Reporter , Genes, abl , Hepatocyte Nuclear Factor 4/metabolism , Histone Deacetylases/genetics , Humans , Models, Biological , Phenylenediamines/pharmacology
10.
Anal Chem ; 90(4): 2434-2439, 2018 02 20.
Article in English | MEDLINE | ID: mdl-29376338

ABSTRACT

Membrane proteins may act as transporters, receptors, enzymes, and adhesion-anchors, accounting for nearly 70% of pharmaceutical drug targets. Difficulties in efficient enrichment, extraction, and solubilization still exist because of their relatively low abundance and poor solubility. A simplified membrane protein extraction approach with advantages of user-friendly sample processing procedures, good repeatability and significant effectiveness was developed in the current research for enhancing enrichment and identification of membrane proteins. This approach combining centrifugation and detergent along with LC-MS/MS successfully identified higher proportion of membrane proteins, integral proteins and transmembrane proteins in membrane fraction (76.6%, 48.1%, and 40.6%) than in total cell lysate (41.6%, 16.4%, and 13.5%), respectively. Moreover, our method tended to capture membrane proteins with high degree of hydrophobicity and number of transmembrane domains as 486 out of 2106 (23.0%) had GRAVY > 0 in membrane fraction, 488 out of 2106 (23.1%) had TMs ≥ 2. It also provided for improved identification of membrane proteins as more than 60.6% of the commonly identified membrane proteins in two cell samples were better identified in membrane fraction with higher sequence coverage. Data are available via ProteomeXchange with identifier PXD008456.

11.
J Proteome Res ; 17(3): 1300-1308, 2018 03 02.
Article in English | MEDLINE | ID: mdl-29369637

ABSTRACT

Failure to properly repair damaged due to myocardial infarction is a major cause of heart failure. In contrast with adult mammals, zebrafish hearts show remarkable regenerative capabilities after substantial damage. To characterize protein dynamics during heart regeneration, we employed an HPLC-ESI-MS/MS (mass spectrometry) approach. Myocardium tissues were taken from sham-operated fish and ventricle-resected sample at three different time points (2, 7, and 14 days); dynamics of protein expression were analyzed by an ion-current-based quantitative platform. More than 2000 protein groups were quantified in all 16 experiments. Two hundred and nine heart-regeneration-related protein groups were quantified and clustered into six time-course patterns. Functional analysis indicated that multiple molecular function and metabolic pathways were involved in heart regeneration. Interestingly, Ingenuity Pathway Analysis revealed that P53 signaling was inhibited during the heart regeneration, which was further verified by real-time quantitative polymerase chain reaction (Q-PCR). In summary, we applied systematic proteomics analysis on regenerating zebrafish heart, uncovered the dynamics of regenerative genes expression and regulatory pathways, and provided invaluable insight into design regenerative-based strategies in human hearts.


Subject(s)
Fish Proteins/genetics , Heart Injuries/genetics , Heart Ventricles/metabolism , Myocardium/metabolism , Proteomics/methods , Regeneration/genetics , Animals , Chromatography, High Pressure Liquid , Fish Proteins/metabolism , Gene Ontology , Heart Injuries/metabolism , Heart Injuries/rehabilitation , Heart Ventricles/injuries , Metabolic Networks and Pathways/genetics , Molecular Sequence Annotation , Proteomics/instrumentation , Real-Time Polymerase Chain Reaction , Spectrometry, Mass, Electrospray Ionization , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Zebrafish
12.
Obesity (Silver Spring) ; 24(7): 1506-14, 2016 07.
Article in English | MEDLINE | ID: mdl-27345962

ABSTRACT

OBJECTIVE: To provide a more global view of adipocyte changes in human insulin resistance by proteomics analyses. METHODS: Baseline biopsies of abdominal subcutaneous adipose tissue were obtained from 23 subjects without diabetes. Euglycemic clamps were used to divide subjects into an insulin-resistant group (IR, N = 10) and an insulin-sensitive (IS, N = 13) group, which were of similar age and gender but unequal adiposity (greater in IR). Proteins of isolated adipocytes were quantified by mass spectrometry using normalized spectral abundance factors. RESULTS: Of 1,245 proteins assigned, 30 were detected in at least 12 of the 23 subjects that differed significantly in abundance ≥1.5-fold between IR and IS. IR displayed a pattern of increased cytoskeletal proteins and decreased mitochondrial proteins and FABP4 and FABP5. In subgroup analyses of adiposity-matched subjects, several of these changes were less pronounced in IR, but the abundance of proteins related to lipid metabolism and the unfolded/misfolded protein response were significantly and unfavorably altered. CONCLUSIONS: These results confirm lower abundance of mitochondrial proteins and suggest increased cytoskeletal proteins and decreased FABP4 and FABP5 in subcutaneous adipocytes of typical IR individuals. Changes in proteins related to lipid metabolism and the unfolded/misfolded protein may discriminate IR and IS individuals of equal adiposity.


Subject(s)
Adipocytes/chemistry , Insulin Resistance , Proteomics , Subcutaneous Fat, Abdominal/cytology , Adiposity , Adult , Cytoskeletal Proteins/analysis , Fatty Acid-Binding Proteins/analysis , Female , Healthy Volunteers , Humans , Male , Middle Aged , Mitochondrial Proteins/analysis
13.
Mol Cell Endocrinol ; 424: 1-11, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26780722

ABSTRACT

Protein phosphatase 2A (PP2A) is one of the major serine/threonine phosphatases. We hypothesize that PP2A regulates signaling cascades in pancreatic ß-cells in the context of glucose-stimulated insulin secretion (GSIS). Using co-immunoprecipitation (co-IP) and tandem mass spectrometry, we globally identified the protein interaction partners of the PP2A catalytic subunit (PP2Ac) in insulin-secreting pancreatic ß-cells. Among the 514 identified PP2Ac interaction partners, 476 were novel. This represents the first global view of PP2Ac protein-protein interactions caused by hyperglycemic conditions. Additionally, numerous PP2Ac partners were found involved in a variety of signaling pathways in the ß-cell function, such as insulin secretion. Our data suggest that PP2A interacts with various signaling proteins necessary for physiological insulin secretion as well as signaling proteins known to regulate cell dysfunction and apoptosis in the pancreatic ß-cells.


Subject(s)
Gene Regulatory Networks , Insulin-Secreting Cells/physiology , Protein Phosphatase 2/metabolism , Proteomics/methods , Cells, Cultured , Glucose/pharmacology , Humans , Protein Interaction Mapping/methods , Tandem Mass Spectrometry
14.
PLoS One ; 10(10): e0140255, 2015.
Article in English | MEDLINE | ID: mdl-26465754

ABSTRACT

Insulin resistance and Type 2 diabetes are marked by an aberrant response in the insulin signaling network. The phosphoinositide-dependent serine/threonine kinase, Akt2, plays a key role in insulin signaling and glucose uptake, most notably within skeletal muscle. Protein-protein interaction regulates the functional consequence of Akt2 and in turn, Akt2's role in glucose uptake. However, only few insulin-responsive Akt2 interaction partners have been identified in skeletal muscle cells. In the present work, rat L6 myoblasts, a widely used insulin sensitive skeletal muscle cell line, were used to examine endogenous, insulin-stimulated Akt2 protein interaction partners. Akt2 co-immunoprecipitation was coupled with 1D-SDS-PAGE and fractions were analyzed by HPLC-ESI-MS/MS to reveal Akt2 protein-protein interactions. The pull-down assay displayed specificity for the Akt2 isoform; Akt1 and Akt3 unique peptides were not detected. A total of 49 were detected with a significantly increased (47) or decreased (2) association with Akt2 following insulin administration (n = 4; p<0.05). Multiple pathways were identified for the novel Akt2 interaction partners, such as the EIF2 and ubiquitination pathways. These data suggest that multiple new endogenous proteins may associate with Akt2 under basal as well as insulin-stimulated conditions, providing further insight into the insulin signaling network. Data are available via ProteomeXchange with identifier PXD002557.


Subject(s)
Carrier Proteins/metabolism , Myoblasts/metabolism , Protein Interaction Mapping , Proto-Oncogene Proteins c-akt/metabolism , Animals , Cells, Cultured , Protein Binding , Rats , Signal Transduction , rho-Associated Kinases/metabolism
15.
J Proteome Res ; 14(11): 4662-73, 2015 Nov 06.
Article in English | MEDLINE | ID: mdl-26390080

ABSTRACT

The two key steps for analyzing proteomic data generated by high-resolution MS are database searching and postprocessing. While the two steps are interrelated, studies on their combinatory effects and the optimization of these procedures have not been adequately conducted. Here, we investigated the performance of three popular search engines (SEQUEST, Mascot, and MS Amanda) in conjunction with five filtering approaches, including respective score-based filtering, a group-based approach, local false discovery rate (LFDR), PeptideProphet, and Percolator. A total of eight data sets from various proteomes (e.g., E. coli, yeast, and human) produced by various instruments with high-accuracy survey scan (MS1) and high- or low-accuracy fragment ion scan (MS2) (LTQ-Orbitrap, Orbitrap-Velos, Orbitrap-Elite, Q-Exactive, Orbitrap-Fusion, and Q-TOF) were analyzed. It was found combinations involving Percolator achieved markedly more peptide and protein identifications at the same FDR level than the other 12 combinations for all data sets. Among these, combinations of SEQUEST-Percolator and MS Amanda-Percolator provided slightly better performances for data sets with low-accuracy MS2 (ion trap or IT) and high accuracy MS2 (Orbitrap or TOF), respectively, than did other methods. For approaches without Percolator, SEQUEST-group performs the best for data sets with MS2 produced by collision-induced dissociation (CID) and IT analysis; Mascot-LFDR gives more identifications for data sets generated by higher-energy collisional dissociation (HCD) and analyzed in Orbitrap (HCD-OT) and in Orbitrap Fusion (HCD-IT); MS Amanda-Group excels for the Q-TOF data set and the Orbitrap Velos HCD-OT data set. Therefore, if Percolator was not used, a specific combination should be applied for each type of data set. Moreover, a higher percentage of multiple-peptide proteins and lower variation of protein spectral counts were observed when analyzing technical replicates using Percolator-associated combinations; therefore, Percolator enhanced the reliability for both identification and quantification. The analyses were performed using the specific programs embedded in Proteome Discoverer, Scaffold, and an in-house algorithm (BuildSummary). These results provide valuable guidelines for the optimal interpretation of proteomic results and the development of fit-for-purpose protocols under different situations.


Subject(s)
Algorithms , Peptides/analysis , Proteome/analysis , Proteomics/methods , Search Engine/methods , Software , Cell Line, Tumor , Databases, Protein , Escherichia coli/genetics , Escherichia coli/metabolism , Humans , Proteome/genetics , Proteome/metabolism , Proteomics/instrumentation , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Tandem Mass Spectrometry
16.
J Proteomics ; 109: 63-75, 2014 Sep 23.
Article in English | MEDLINE | ID: mdl-24972320

ABSTRACT

Serine/threonine protein phosphatase 1 regulatory subunit 12A (PPP1R12A) modulates the activity and specificity of the catalytic subunit of protein phosphatase 1, regulating various cellular processes via dephosphorylation. Nonetheless, little is known about phosphorylation events controlled by PPP1R12A in skeletal muscle insulin signaling. Here, we used quantitative phosphoproteomics to generate a global picture of phosphorylation events regulated by PPP1R12A in a L6 skeletal muscle cell line, which were engineered for inducible PPP1R12A knockdown. Phosphoproteomics revealed 3876 phosphorylation sites (620 were novel) in these cells. Furthermore, PPP1R12A knockdown resulted in increased overall phosphorylation in L6 cells at the basal condition, and changed phosphorylation levels for 698 sites (assigned to 295 phosphoproteins) at the basal and/or insulin-stimulated conditions. Pathway analysis on the 295 phosphoproteins revealed multiple significantly enriched pathways related to insulin signaling, such as mTOR signaling and RhoA signaling. Moreover, phosphorylation levels for numerous regulatory sites in these pathways were significantly changed due to PPP1R12A knockdown. These results indicate that PPP1R12A indeed plays a role in skeletal muscle insulin signaling, providing novel insights into the biology of insulin action. This new information may facilitate the design of experiments to better understand mechanisms underlying skeletal muscle insulin resistance and type 2 diabetes. BIOLOGICAL SIGNIFICANCE: These results identify a large number of potential new substrates of serine/threonine protein phosphatase 1 and suggest that serine/threonine protein phosphatase 1 regulatory subunit 12A indeed plays a regulatory role in multiple pathways related to insulin action, providing novel insights into the biology of skeletal muscle insulin signaling. This information may facilitate the design of experiments to better understand the molecular mechanism responsible for skeletal muscle insulin resistance and associated diseases, such as type 2 diabetes and cardiovascular diseases.


Subject(s)
Insulin/metabolism , Muscle Proteins/metabolism , Myosin-Light-Chain Phosphatase/metabolism , Protein Phosphatase 1/metabolism , Proteomics , Signal Transduction/physiology , Animals , Cell Line , Diabetes Mellitus, Type 2/metabolism , Humans , Insulin Resistance , Phosphorylation/physiology , Rats , TOR Serine-Threonine Kinases/metabolism , rhoA GTP-Binding Protein/metabolism
17.
Diabetes ; 63(6): 1933-47, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24584551

ABSTRACT

Insulin receptor substrate 1 (IRS1) is a key mediator of insulin signal transduction. Perturbations involving IRS1 complexes may lead to the development of insulin resistance and type 2 diabetes (T2D). Surprisingly little is known about the proteins that interact with IRS1 in humans under health and disease conditions. We used a proteomic approach to assess IRS1 interaction partners in skeletal muscle from lean healthy control subjects (LCs), obese insulin-resistant nondiabetic control subjects (OCs), and participants with T2D before and after insulin infusion. We identified 113 novel endogenous IRS1 interaction partners, which represents the largest IRS1 interactome in humans and provides new targets for studies of IRS1 complexes in various diseases. Furthermore, we generated the first global picture of IRS1 interaction partners in LCs, and how they differ in OCs and T2D patients. Interestingly, dozens of proteins in OCs and/or T2D patients exhibited increased associations with IRS1 compared with LCs under the basal and/or insulin-stimulated conditions, revealing multiple new dysfunctional IRS1 pathways in OCs and T2D patients. This novel abnormality, increased interaction of multiple proteins with IRS1 in obesity and T2D in humans, provides new insights into the molecular mechanism of insulin resistance and identifies new targets for T2D drug development.


Subject(s)
Blood Glucose/metabolism , Diabetes Mellitus, Type 2/metabolism , Insulin Receptor Substrate Proteins/metabolism , Insulin Resistance/genetics , Muscle, Skeletal/metabolism , Obesity/metabolism , Thinness/metabolism , Body Mass Index , Diabetes Mellitus, Type 2/genetics , Female , Genetic Predisposition to Disease , Genetic Variation , Glucose Clamp Technique , Humans , Insulin/administration & dosage , Insulin Receptor Substrate Proteins/genetics , Linkage Disequilibrium , Male , Middle Aged , Obesity/genetics , Promoter Regions, Genetic , RNA, Messenger , Risk Factors , Signal Transduction , Thinness/genetics
18.
Oncotarget ; 4(9): 1427-37, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23965803

ABSTRACT

Mineral dust-induced gene (mdig) had been linked to the development of human lung cancers associated with environmental exposure to mineral dust, tobacco smoke or other carcinogens. In the present studies, we demonstrated that the overexpression of mdig in A549 adenocarcinomic human alveolar type II epithelial cells decreases the heterochromatin conformation of the cells and de-represses the transcription of genes in the tandemly repeated DNA regions. Although mdig can only cause a marginal decrease of the total histone H3 lysine 9 trimethylation (H3K9me3), a significant reduction of H3K9me3 in the promoter region of H19, the paternally imprinted but maternally expressed gene transcribing a large intergenic non-coding RNA (lincRNA), was observed in the cells with mdig overexpression. Silencing mdig by either shRNA or siRNA not only increased the level of H3K9me3 in the promoter region of H19 but also attenuated the transcription of H19 long non-coding RNA. Demethylation assays using immunoprecipitated mdig and histone H3 peptide substrate suggested that mdig is able to remove the methyl groups from H3K9me3. Clinically, we found that higher levels of mdig and H19 expression correlate with poorer survival of the lung cancer patients. Taken together, our results imply that mdig is involved in the regulation of H3K9me3 to influence the heterochromatin structure of the genome and the expression of genes important for cell growth or transformation.


Subject(s)
Heterochromatin/genetics , Histones/genetics , Nuclear Proteins/genetics , RNA, Long Noncoding/genetics , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma of Lung , Cell Line, Tumor , Dioxygenases , Down-Regulation , Heterochromatin/metabolism , Histone Demethylases , Histones/metabolism , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Methylation , Nuclear Proteins/metabolism , RNA, Long Noncoding/metabolism , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Survival Analysis , Transcription, Genetic , Transfection
19.
Exp Hematol Oncol ; 2: 18, 2013.
Article in English | MEDLINE | ID: mdl-23890079

ABSTRACT

Oxythiamine (OT), an analogue of anti-metabolite, can suppress the nonoxidative synthesis of ribose and induce cell apoptosis by causing a G1 phase arrest in vitro and in vivo. However, the molecular mechanism remains unclear yet. In the present study, a quantitative proteomic analysis using the modified SILAC method (mSILAC) was performed to determine the effect of metabolic inhibition on dynamic changes of protein expression in MIA PaCa-2 cancer cells treated with OT at various doses (0 µM, 5 µM, 50 µM and 500 µM) and time points (0 h, 12 h and 48 h). A total of 52 differential proteins in MIA PaCa-2 cells treated with OT were identified, including 14 phosphorylated proteins. Based on the dynamic expression pattern, these proteins were categorized in three clusters, straight down-regulation (cluster 1, 37% of total proteins), upright "V" shape expression pattern (cluster 2, 47.8% total), and downright "V" shape pattern (cluster 3, 15.2% total). Among them, Annexin A1 expression was significantly down-regulated by OT treatment in time-dependent manner, while no change of this protein was observed in OT dose-dependent fashion. Pathway analysis suggested that inhibition of transketolase resulted in changes of multiple cellular signaling pathways associated with cell apoptosis. The temporal expression patterns of proteins revealed that OT altered dynamics of protein expression in time-dependent fashion by suppressing phosphor kinase expression, resulting in cancer cell apoptosis. Results from this study suggest that interference of single metabolic enzyme activity altered multiple cellular signaling pathways.

20.
Elife ; 2: e00444, 2013 Apr 09.
Article in English | MEDLINE | ID: mdl-23580231

ABSTRACT

The secretory pathway of eukaryotic cells packages cargo proteins into COPII-coated vesicles for transport from the endoplasmic reticulum (ER) to the Golgi. We now report that complete genetic deficiency for the COPII component SEC24A is compatible with normal survival and development in the mouse, despite the fundamental role of SEC24 in COPII vesicle formation and cargo recruitment. However, these animals exhibit markedly reduced plasma cholesterol, with mutations in Apoe and Ldlr epistatic to Sec24a, suggesting a receptor-mediated lipoprotein clearance mechanism. Consistent with these data, hepatic LDLR levels are up-regulated in SEC24A-deficient cells as a consequence of specific dependence of PCSK9, a negative regulator of LDLR, on SEC24A for efficient exit from the ER. Our findings also identify partial overlap in cargo selectivity between SEC24A and SEC24B, suggesting a previously unappreciated heterogeneity in the recruitment of secretory proteins to the COPII vesicles that extends to soluble as well as trans-membrane cargoes. DOI:http://dx.doi.org/10.7554/eLife.00444.001.


Subject(s)
COP-Coated Vesicles/enzymology , Cholesterol/blood , Dyslipidemias/enzymology , Endoplasmic Reticulum/enzymology , Liver/enzymology , Proprotein Convertases/blood , Serine Endopeptidases/blood , Vesicular Transport Proteins/deficiency , Animals , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , COP-Coated Vesicles/metabolism , Cell Line, Tumor , Down-Regulation , Dyslipidemias/blood , Dyslipidemias/genetics , Endoplasmic Reticulum/metabolism , Epistasis, Genetic , Genotype , HEK293 Cells , Humans , Liver/metabolism , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mutation , Phenotype , Proprotein Convertase 9 , Proprotein Convertases/metabolism , Protein Transport , Receptors, LDL/genetics , Receptors, LDL/metabolism , Serine Endopeptidases/metabolism , Transfection , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...