Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Int J Mol Sci ; 21(8)2020 Apr 19.
Article in English | MEDLINE | ID: mdl-32325885

ABSTRACT

This study reports a relationship between Akt3 expression and tissue-specific regulation of the pI3K/Akt/mTOR signaling pathway by copaiba essential oil. Akt3, a protein kinase B isoform important for the regulation of neuronal development, exhibited differential expression levels in cells of various origins. In neuronal and microglial cells, where Akt3 is present, copaiba essential oil positively regulated the pI3K/Akt/mTOR signaling pathway. In contrast, in liver cells and T lymphocytes, where Akt3 is absent, copaiba essential oil negatively regulated the pI3K/Akt/mTOR signaling pathway. The expression of Akt3 via plasmid DNA in liver cells led to positive regulatory effects by copaiba essential oil on the pI3K/Akt/mTOR signaling pathway. In contrast, inhibition of Akt3 expression in neuronal cells via small interfering RNA molecules targeting Akt3 transcripts abrogated the regulatory effects of copaiba essential oil on the pI3K/Akt/mTOR signaling pathway. Interestingly, Akt3 expression did not impact the regulatory effects of copaiba essential oil on other signaling pathways. For example, copaiba essential oil consistently upregulated the MAPK and JAK/STAT signaling pathways in all evaluated cell types, independent of the Akt3 expression level. Collectively, the data indicated that Akt3 expression was required for the positive regulatory effects of copaiba essential oil, specifically on the pI3K/Akt/mTOR signaling pathway.


Subject(s)
Fabaceae/chemistry , Gene Expression Regulation/drug effects , Oils, Volatile/pharmacology , Proto-Oncogene Proteins c-akt/genetics , Cell Line, Tumor , Humans , Janus Kinases/metabolism , Oils, Volatile/chemistry , Organ Specificity/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , STAT Transcription Factors/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism
2.
Int J Mol Sci ; 21(7)2020 Mar 25.
Article in English | MEDLINE | ID: mdl-32218156

ABSTRACT

This study examined the biological activities of copaiba essential oil via measurement of its effects on signaling pathways in the SH-SY5Y neuronal cell line. Nanofluidic proteomic technologies were deployed to measure the phosphorylation of biomarker proteins within the signaling cascades. Interestingly, copaiba essential oil upregulated the pI3K/Akt/mTOR, MAPK, and JAK/STAT signaling pathways in neuronal cells. The effects of copaiba essential oil peaked at 30 min post-treatment, with a half-maximal effective concentration (EC50) of approximately 80 ng/mL. Treatment with cannabinoid receptor 2 (CB2) agonist AM1241 or the inverse agonist BML190 abrogated the regulatory effects of copaiba essential oil on the pI3K/Akt/mTOR signaling pathway. Surprisingly, copaiba essential oil also activated the apoptosis signaling pathway and reduced the viability of SH-SY5Y cells with an EC50 of approximately 400 ng/mL. Furthermore, ß-caryophyllene, a principal constituent of copaiba essential oil, downregulated the pI3K/Akt/mTOR signaling pathway. Taken together, the findings indicated that copaiba essential oil upregulated signaling pathways associated with cell metabolism, growth, immunity, and apoptosis. The biological activities of copaiba essential oil were determined to be fast acting, CB2 mediated, and dependent on multiple chemical constituents of the oil. Nanofluidic proteomics provided a powerful means to assess the biological activities of copaiba essential oil.


Subject(s)
Fabaceae/chemistry , Neuroblastoma/metabolism , Oils, Volatile/pharmacology , Plant Oils/pharmacology , Signal Transduction/drug effects , Cell Line, Tumor , Cyclic AMP-Dependent Protein Kinases/metabolism , Humans , Neuroblastoma/pathology , Phosphatidylinositol 3-Kinases/metabolism , TOR Serine-Threonine Kinases/metabolism
3.
Molecules ; 24(13)2019 Jun 27.
Article in English | MEDLINE | ID: mdl-31252611

ABSTRACT

Current methods for the authentication of essential oils focus on analyzing their chemical composition. This study describes the use of nanofluidic protein post-translational modification (PTM) profiling to differentiate essential oils by analyzing their biochemical effects. Protein PTM profiling was used to measure the effects of four essential oils, copaiba, mandarin, Melissa, and turmeric, on the phosphorylation of MEK1, MEK2, and ERK1/2 in the MAPK signaling pathway; Akt and 4EBP1 in the pI3K/Akt/mTOR signaling pathway; and STAT3 in the JAK/STAT signaling pathway in cultured HepG2 cells. The gain or loss of the phosphorylation of these proteins served as direct read-outs for the positive or negative regulatory effects of essential oils on their respective signaling pathways. Furthermore, protein PTM profiling and GC-MS were employed side-by-side to assess the quality of the essential oils. In general, protein PTM profiling data concurred with GC-MS data on the identification of adulterated mandarin, Melissa, and turmeric essential oils. Most interestingly, protein PTM profiling data identified the differences in biochemical effects between copaiba essential oils, which were indistinguishable with GC-MS data on their chemical composition. Taken together, nanofluidic protein PTM profiling represents a robust method for the assessment of the quality and therapeutic potential of essential oils.


Subject(s)
Citrus/chemistry , Curcuma/chemistry , MAP Kinase Signaling System/drug effects , Melissa/chemistry , Oils, Volatile/pharmacology , Gas Chromatography-Mass Spectrometry , Hep G2 Cells , Humans , Microfluidic Analytical Techniques , Oils, Volatile/chemistry , Phosphorylation/drug effects , Plant Oils/chemistry , Plant Oils/pharmacology , Protein Processing, Post-Translational/drug effects
4.
J Pathol ; 238(5): 641-50, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26725750

ABSTRACT

We describe an alternative approach to classifying fatty liver by profiling protein post-translational modifications (PTMs) with high-throughput capillary isoelectric focusing (cIEF) immunoassays. Four strains of mice were studied, with fatty livers induced by different causes, such as ageing, genetic mutation, acute drug usage, and high-fat diet. Nutrient-sensitive PTMs of a panel of 12 liver metabolic and signalling proteins were simultaneously evaluated with cIEF immunoassays, using nanograms of total cellular protein per assay. Changes to liver protein acetylation, phosphorylation, and O-N-acetylglucosamine glycosylation were quantified and compared between normal and diseased states. Fatty liver tissues could be distinguished from one another by distinctive protein PTM profiles. Fatty liver is currently classified by morphological assessment of lipid droplets, without identifying the underlying molecular causes. In contrast, high-throughput profiling of protein PTMs has the potential to provide molecular classification of fatty liver.


Subject(s)
Fatty Liver/metabolism , High-Throughput Screening Assays , Liver/metabolism , Protein Processing, Post-Translational , Proteins/metabolism , Proteomics/methods , Acetylation , Age Factors , Animals , Biomarkers/metabolism , Disease Models, Animal , Fatty Liver/classification , Fatty Liver/diagnosis , Fatty Liver/etiology , Fatty Liver/genetics , Genetic Predisposition to Disease , Glycosylation , Immunoassay , Isoelectric Focusing , Liver/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phenotype , Phosphorylation
5.
J Lipid Res ; 54(4): 1044-57, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23355744

ABSTRACT

We report in this study an intrinsic link between pyrimidine metabolism and liver lipid accumulation utilizing a uridine phosphorylase 1 transgenic mouse model UPase1-TG. Hepatic microvesicular steatosis is induced by disruption of uridine homeostasis through transgenic overexpression of UPase1, an enzyme of the pyrimidine catabolism and salvage pathway. Microvesicular steatosis is also induced by the inhibition of dihydroorotate dehydrogenase (DHODH), an enzyme of the de novo pyrimidine biosynthesis pathway. Interestingly, uridine supplementation completely suppresses microvesicular steatosis in both scenarios. The effective concentration (EC(50)) for uridine to suppress microvesicular steatosis is approximately 20 µM in primary hepatocytes of UPase1-TG mice. We find that uridine does not have any effect on in vitro DHODH enzymatic activity. On the other hand, uridine supplementation alters the liver NAD(+)/NADH and NADP(+)/NADPH ratios and the acetylation profile of metabolic, oxidation-reduction, and antioxidation enzymes. Protein acetylation is emerging as a key regulatory mechanism for cellular metabolism. Therefore, we propose that uridine suppresses fatty liver by modulating the liver protein acetylation profile. Our findings reveal a novel link between uridine homeostasis, pyrimidine metabolism, and liver lipid metabolism.


Subject(s)
Liver/metabolism , Pyrimidines/metabolism , Uridine/metabolism , Animals , Cells, Cultured , Fatty Acids/metabolism , Lipid Metabolism/drug effects , Liver/drug effects , Male , Mice , Mice, Transgenic , Triglycerides/metabolism , Uridine Phosphorylase/genetics , Uridine Phosphorylase/metabolism
6.
BMC Cancer ; 12: 540, 2012 Nov 21.
Article in English | MEDLINE | ID: mdl-23171028

ABSTRACT

BACKGROUND: Circulating tumour cells (CTC) are an important indicator of metastasis and associated with a poor prognosis. Detection sensitivity and specificity of CTC in the peripheral blood of metastatic cancer patient remain a technical challenge. METHODS: Coherent anti-Stokes Raman scattering (CARS) microscopy was employed to examine the lipid content of CTC isolated from the peripheral blood of metastatic prostate cancer patients. CARS microscopy was also employed to evaluate lipid uptake and mobilization kinetics of a metastatic human prostate cancer cell line. RESULTS: One hundred CTC from eight metastatic prostate cancer patients exhibited strong CARS signal which arose from intracellular lipid. In contrast, leukocytes exhibited weak CARS signal which arose mostly from cellular membrane. On average, CARS signal intensity of prostate CTC was 7-fold higher than that of leukocytes (P<0.0000001). When incubated with human plasma, C4-2 metastatic human prostate cancer cells exhibited rapid lipid uptake kinetics and slow lipid mobilization kinetics. Higher expression of lipid transport proteins in C4-2 cells compared to non-transformed RWPE-1 and non-malignant BPH-1 prostate epithelial cells further indicated strong affinity for lipid of metastatic prostate cancer cells. CONCLUSIONS: Intracellular lipid could serve as a biomarker for prostate CTC which could be sensitively detected with CARS microscopy in a label-free manner. Strong affinity for lipid by metastatic prostate cancer cells could be used to improve detection sensitivity and therapeutic targeting of prostate CTC.


Subject(s)
Lipid Metabolism/physiology , Neoplastic Cells, Circulating/metabolism , Neoplastic Cells, Circulating/pathology , Prostatic Neoplasms/blood , Prostatic Neoplasms/pathology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line , Cell Line, Tumor , Cell Membrane/genetics , Cell Membrane/metabolism , Cell Membrane/pathology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Humans , Leukocytes/metabolism , Leukocytes/pathology , Male , Microscopy/methods , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/genetics , Sensitivity and Specificity , Spectrum Analysis, Raman/methods
7.
Nutrients ; 14(16)2022 Aug 12.
Article in English | MEDLINE | ID: mdl-36014807

ABSTRACT

In this study, the effects of cinnamaldehyde and curcumin on Akt2, a serine/threonine protein kinase central to the insulin signaling pathway, were examined in preadipocytes. Cinnamaldehyde or curcumin treatment increased Akt2 phosphorylation at multiple sites including T450 and Y475, but had no effect on Akt2 phosphorylation at S474, which is critical for Akt2 activation. Surprisingly, insulin treatment with cinnamaldehyde or curcumin increased p-Akt2 (S474) by 3.5-fold versus insulin treatment alone. Furthermore, combined cinnamaldehyde, curcumin, and insulin treatment increased p-Akt2 (S474) by 7-fold versus insulin treatment alone. Interestingly, cinnamaldehyde and curcumin inhibited both serine/threonine phosphatase 2A (PP2A) and protein tyrosine phosphatase 1B (PTP1B). Akt2 activation is a multistep process that requires phosphorylation at T450 for proper folding and maturation, and phosphorylation of both Y475 and S474 for stabilization of the catalytic domain. It is plausible that by inhibiting PP2A and PTP1B, cinnamaldehyde and curcumin increase phosphorylation at T450 and Y475, and prime Akt2 for insulin-stimulated phosphorylation at S474. Notably, the combination of a PP2A inhibitor, okadaic acid, and a PTP1B inhibitor increased p-Akt2 (S474), even in the absence of insulin. Future combinations of PP2A and PTP1B inhibitors provide a rational platform to engineer new therapeutics for insulin resistance syndrome.


Subject(s)
Curcumin , Insulin , Acrolein/analogs & derivatives , Curcumin/pharmacology , Enzyme Inhibitors , Insulin/metabolism , Insulin/pharmacology , Phosphorylation , Serine/metabolism
8.
Nutrients ; 14(20)2022 Oct 16.
Article in English | MEDLINE | ID: mdl-36297009

ABSTRACT

Obesity is an established risk factor for metabolic disease. This study explores the functional complementation of anti-adipogenic phytonutrients for obesity prevention and management. Nine phytonutrients were selected based on their ability to affect the expression of one or more selected adipogenic biomarker proteins. The phytonutrients include berberine, luteolin, resveratrol, fisetin, quercetin, fucoidan, epigallocatechin gallate, hesperidin, and curcumin. The selected adipogenic biomarker proteins include PPARÉ£, SREBP1c, FASN, PLIN1, FABP4, and ß-catenin. Individually, phytonutrients had variable effects on the expression level of selected adipogenic biomarker proteins. Collectively, the functional complementation of nine phytonutrients suppressed de novo fatty acid biosynthesis via the negative regulation of PPARÉ£, FASN, PLIN1, and FABP4 expression; activated glycolysis via the positive regulation of SREBP1c expression; and preserved cell-cell adhesion via the inhibition of ß-catenin degradation. In primary human subcutaneous preadipocytes, the composition of nine phytonutrients had more potent and longer lasting anti-adipogenic effects compared to individual phytonutrients. In a diet-induced obesity murine model, the composition of nine phytonutrients improved glucose tolerance and reduced weight gain, liver steatosis, visceral adiposity, circulating triglycerides, low-density lipoprotein cholesterol, and inflammatory cytokines and chemokines. The functional complementation of anti-adipogenic phytonutrients provides an effective approach toward engineering novel therapeutics for the prevention and management of obesity and metabolic syndrome.


Subject(s)
Obesity , Phytochemicals , Animals , Humans , Mice , Adipocytes , Adipogenesis , Berberine/pharmacology , beta Catenin/metabolism , Cholesterol/metabolism , Curcumin/pharmacology , Cytokines/metabolism , Fatty Acids/metabolism , Glucose/metabolism , Hesperidin/pharmacology , Lipoproteins, LDL/metabolism , Luteolin/pharmacology , Obesity/prevention & control , Obesity/metabolism , Phytochemicals/pharmacology , PPAR gamma/metabolism , Quercetin/pharmacology , Resveratrol/pharmacology , Sterol Regulatory Element Binding Protein 1/metabolism , Triglycerides/metabolism
9.
Nutrients ; 14(18)2022 Sep 14.
Article in English | MEDLINE | ID: mdl-36145160

ABSTRACT

Maintaining healthy body weight is an important component of any effective diabetes management plan. However, glycemic management using insulin generally leads to weight gain. In addition, weight loss medications prescribed for diabetes management are often associated with adverse side effects, which limit their long-term usage. Alternatively, nutrition intervention provides a safe, readily accessible, and inexpensive option for diabetes management. This study describes a composition of phytonutrients comprising berberine, cinnamaldehyde, and curcumin for glycemic and weight management. Functional complementarity between berberine, cinnamaldehyde, and curcumin provides an effective means to improve insulin sensitivity without increasing adiposity. In primary human omental preadipocytes, cinnamaldehyde and curcumin additively enhance insulin-stimulated activation of Akt2 and glucose uptake, whereas berberine inhibits de novo fatty acid biosynthesis and fat cell differentiation. In a diet-induced obesity murine model, a dietary supplement with berberine, cinnamaldehyde, and curcumin prevents weight gain, improves glucose tolerance, and reduces HbA1c, blood lipids, visceral adiposity, and liver steatosis. Collectively, the composition of phytonutrients comprising berberine, cinnamaldehyde, and curcumin protects against obesity and pre-diabetic conditions in a diet-induced obesity murine model. Safety and efficacy assessment of nutrition intervention using combined berberine, cinnamaldehyde, and curcumin for glycemic and weight management in future clinical trials are warranted.


Subject(s)
Berberine , Curcumin , Diabetes Mellitus , Insulin Resistance , Acrolein/analogs & derivatives , Animals , Berberine/pharmacology , Berberine/therapeutic use , Blood Glucose , Curcumin/pharmacology , Curcumin/therapeutic use , Disease Models, Animal , Fatty Acids/therapeutic use , Glycated Hemoglobin , Humans , Insulin , Lipids , Mice , Obesity/complications , Obesity/drug therapy , Phytochemicals/pharmacology , Phytochemicals/therapeutic use , Weight Gain
10.
BMC Cancer ; 11: 51, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21284881

ABSTRACT

BACKGROUND: CD26 (dipeptidyl peptidase IV, DPPIV) is a 110 kDa surface glycoprotein expressed in most normal tissues, and is a potential novel therapeutic target for selected cancers. Our work evaluates the mechanism involved in confluence-dependent CD26 expression in colon cancer. METHODS: Colon adenocarcinoma cells were grown to confluence, and expression of CD26 and transcription factors implicated in its regulation was confirmed by immunofluorescence and Western blotting. Real-time PCR was also performed to evaluate CD26 upregulation at the transcriptional level. The influence of c-Myc on CD26 expression during different growth conditions was further evaluated following transient transfection of a c-Myc-expressing plasmid and a c-Myc specific siRNA. RESULTS: We found that the colon cancer cell lines HCT-116 and HCT-15 exhibited a confluence-dependent increase in CD26 mRNA and protein, associated with decreased expression of c-Myc, increased USF-1 and Cdx 2 levels, and unchanged HNF-1α expression. Meanwhile, ectopic expression of c-Myc in both cell lines led to decreased CD26 expression. In contrast, transfection of a siRNA targeted to Cdx2 resulted in decreased CD26 level. Importantly, culturing of cells in serum-depleted media, but not acidic conditions, upregulated CD26. While HIF-1α level also increased when cells were cultured in serum-depleted media, its expression was required but not sufficient for CD26 upregulation. CONCLUSIONS: CD26 mRNA and protein levels increase in a confluence-dependent manner in colon carcinoma cell lines, with c-Myc acting as a repressor and Cdx2 acting as an enhancer of CD26 expression. The enhanced expression of CD26 in serum-depleted media and a requirement for HIF-1α suggest a role for nutrients or growth factors in the regulation of CD26 protein expression.


Subject(s)
Cell Movement , Cell Proliferation , Dipeptidyl Peptidase 4/genetics , Gene Expression Regulation, Neoplastic/genetics , Blotting, Western , CDX2 Transcription Factor , Cell Adhesion , Cell Line, Tumor , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Dipeptidyl Peptidase 4/metabolism , HCT116 Cells , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Microscopy, Fluorescence , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Trans-Activators/genetics , Trans-Activators/metabolism , Transfection
11.
Nutrients ; 12(2)2020 Jan 30.
Article in English | MEDLINE | ID: mdl-32019055

ABSTRACT

This study used nanofluidic protein posttranslational modification (PTM) profiling to measure the effects of six cannabidiol (CBD) oils and isolated CBD on the signaling pathways of a cultured SH-SY5Y neuronal cell line. Chemical composition analysis revealed that all CBD oils met the label claims and legal regulatory limit regarding the CBD and tetrahydrocannabinol (THC) contents, respectively. Isolated CBD was cytotoxic, with an effective concentration (EC50) of 40 µM. In contrast, the CBD oils had no effect on cell viability at CBD concentrations exceeding 1.2 mM. Interestingly, only an unadulterated CBD oil had strong and statistically significant suppressive effects on the pI3K/Akt/mTOR signaling pathway with an EC50 value of 143 µM and a slow-acting timescale requiring hours. Systematic profiling of twenty-six proteins, which served as biomarkers for nine signaling pathways, revealed that the unadulterated CBD oil downregulated seven signaling pathways but had no measurable effect on the other two signaling pathways. The remaining CBD oils, which were adulterated, and isolated CBD had weak, variable, or undetectable effects on neuronal signaling pathways. Our data clearly showed that adulteration diminished the biological activities of CBD oils. In addition, nanofluidic protein PTM profiling provided a robust means for potency assessment of CBD oils.


Subject(s)
Cannabidiol/pharmacology , Plant Oils/pharmacology , Protein Processing, Post-Translational/drug effects , Signal Transduction/drug effects , Cell Survival/drug effects , Cells, Cultured , Humans
12.
Front Biosci ; 13: 1634-45, 2008 Jan 01.
Article in English | MEDLINE | ID: mdl-17981655

ABSTRACT

CD26/DPPIV is a multifunctional cell surface protein that is widely expressed in most cell types including T lymphocytes, on which it is a marker of activation. It is also present in serum and other body fluids in a truncated form (sCD26/DPPIV). It preferentially cleaves N-terminal dipeptides from polypeptides with proline or alanine in the penultimate position, and in doing so, regulates the activities of a number of cytokines and chemokines. Due in part to this ability to regulate the activity of biopeptides, it can act as a tumor suppressor or activator. It can associate with several proteins, among them fibroblast activating protein-alpha (FAP-alpha), plasminogen, adenosine deaminase (ADA), the tyrosine phosphatase CD45, and the chemokine receptor CXCR4. It can also bind to the extracellular matrix (ECM) and depending on the presence of other ligands, this process can either lead to increased or decreased invasive activity of the cells on which it is expressed. As a result of these characteristics, CD26/DPPIV plays an important role in tumor biology, and is useful as a marker for various cancers, with its levels either on the cell surface or in the serum being increased in some neoplasms and decreased in others. Our group has shown that CD26/DPPIV can be manipulated by such agents as CD26 cDNA-carrying plasmids, siRNA and monoclonal antibodies, resulting in both in vitro and in vivo inhibition of cell growth, enhanced sensitivity to selected chemotherapeutic agents, and enhanced survival of mouse xenograft models. These studies have demonstrated the utility of these tools as potential targeted therapies for specific cancers expressing CD26/DPPIV.


Subject(s)
Dipeptidyl Peptidase 4/physiology , Gene Expression Regulation, Neoplastic , Neoplasms/metabolism , Animals , Cell Line , Cell Line, Tumor , Dipeptidyl Peptidase 4/metabolism , Extracellular Matrix/metabolism , Gene Expression Profiling , Hematologic Neoplasms/metabolism , Humans , Mice , Models, Biological , RNA, Small Interfering/metabolism
13.
Sci Rep ; 8(1): 9991, 2018 07 03.
Article in English | MEDLINE | ID: mdl-29968772

ABSTRACT

Mitogen-activated protein kinases (MAPKs) play an important role in the regulation of cell proliferation, oncogenic transformation, and drug resistance. This study examined the capability of nanofluidic proteomics to identify aberrations in the MAPK signaling cascade, monitor its drug response, and guide the rational design of intervention strategies. Specifically, the protein post-translational modification (PTM) profiles of MEK1, MEK2, and ERK1/2 were measured in breast carcinoma and breast cancer cell lines. Nanofluidic proteomics revealed hyper-phosphorylation of MAPKs in breast carcinoma and breast cancer cells treated with kinase inhibitors that interfere with cell cycle regulation, such as dinaciclib, an inhibitor of cyclin-dependent kinases, and rigosertib, an inhibitor of polo-like kinase 1. A pMEK1 (Thr286) phosphor-isoform, which serves as a biomarker of cell cycle-regulated negative feedback phosphorylation in breast cancer cells, was detected in breast carcinoma. Inhibition of the MAPK pathway with dabrafenib, a B-Raf inhibitor, or trametinib, a MEK1/2 inhibitor, suppressed both the positively regulated phosphorylation of MAPKs and the negatively regulated phosphorylation of MEK1. Interestingly, the combinations of dabrafenib and rigosertib or trametinib and rigosertib permitted the suppression of positively regulated MAPK phosphorylation together with the promotion of negatively regulated MEK1 phosphorylation. The effectiveness of protein PTM-guided drug combinations for inhibition of the MAPK pathway remains to be experimentally tested. Via protein PTM profiling, nanofluidic proteomics provides a robust means to detect anomalies in the MAPK signaling cascade, monitor its drug response, and guide the possible design of drug combinations for MAPK pathway-focused targeting.


Subject(s)
Breast Neoplasms/metabolism , Mitogen-Activated Protein Kinases/metabolism , Proteomics/methods , Biopsy , Breast Neoplasms/pathology , Cell Cycle/physiology , Cell Line, Tumor , Feedback, Physiological/physiology , Female , Humans , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase 2/metabolism , MAP Kinase Signaling System/physiology , MCF-7 Cells , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Mitogen-Activated Protein Kinases/physiology , Phosphorylation , Protein Processing, Post-Translational/physiology , Signal Transduction/physiology
14.
Sci Rep ; 8(1): 3606, 2018 02 26.
Article in English | MEDLINE | ID: mdl-29483581

ABSTRACT

Current assessment of non-alcoholic fatty liver disease (NAFLD) with histology is time-consuming, insensitive to early-stage detection, qualitative, and lacks information on etiology. This study explored alternative methods for fast and quantitative assessment of NAFLD with hyperspectral stimulated Raman scattering (SRS) microscopy and nanofluidic proteomics. Hyperspectral SRS microscopy quantitatively measured liver composition of protein, DNA, and lipid without labeling and sensitively detected early-stage steatosis in a few minutes. On the other hand, nanofluidic proteomics quantitatively measured perturbations to the post-translational modification (PTM) profiles of selective liver proteins to identify affected cellular signaling and metabolic pathways in a few hours. Perturbations to the PTM profiles of Akt, 4EBP1, BID, HMGCS2, FABP1, and FABP5 indicated abnormalities in multiple cellular processes including cell cycle regulation, PI3K/Akt/mTOR signaling cascade, autophagy, ketogenesis, and fatty acid transport. The integrative deployment of hyperspectral SRS microscopy and nanofluidic proteomics provided fast, sensitive, and quantitative assessment of liver steatosis and affected pathways that overcame the limitations of histology.


Subject(s)
Fatty Liver/metabolism , Molecular Imaging/methods , Proteomics/methods , BH3 Interacting Domain Death Agonist Protein/metabolism , Fatty Acid-Binding Proteins/metabolism , Humans , Hydroxymethylglutaryl-CoA Synthase/metabolism , Immunoassay , Nonlinear Optical Microscopy , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/physiology
15.
PLoS One ; 11(1): e0146994, 2016.
Article in English | MEDLINE | ID: mdl-26789264

ABSTRACT

Uridine is a pyrimidine nucleoside that exerts restorative functions in tissues under stress. Short-term co-administration of uridine with multiple unrelated drugs prevents drug-induced liver lipid accumulation. Uridine has the ability to modulate liver metabolism; however, the precise mechanism has not been delineated. In this study, long-term effects of uridine on liver metabolism were examined in both HepG2 cell cultures and C57BL/6J mice. We report that uridine administration was associated with O-GlcNAc modification of FOXO1, increased gluconeogenesis, reduced insulin signaling activity, and reduced expression of a liver-specific fatty acid binding protein FABP1. Long-term uridine feeding induced systemic glucose intolerance and severe liver lipid accumulation in mice. Our findings suggest that the therapeutic potentials of uridine should be designed for short-term acute administration.


Subject(s)
Fatty Liver , Forkhead Transcription Factors/metabolism , Gluconeogenesis/drug effects , Glucose Intolerance , Prediabetic State , Uridine/adverse effects , Animals , Fatty Liver/chemically induced , Fatty Liver/metabolism , Fatty Liver/pathology , Forkhead Box Protein O1 , Glucose Intolerance/chemically induced , Glucose Intolerance/metabolism , Glucose Intolerance/pathology , Hep G2 Cells , Humans , Mice , Prediabetic State/chemically induced , Prediabetic State/metabolism , Prediabetic State/pathology , Uridine/pharmacology
16.
Stroke ; 36(11): 2457-62, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16210561

ABSTRACT

BACKGROUND AND PURPOSE: Some proteins involved in self-repair after stroke in the adult brain are primarily expressed during embryonic development and strongly down-regulated during the early postnatal phase. Neuronal precursor cell-expressed, developmentally down-regulated gene (Nedd) 9 was recognized to be identical to Crk-associated substrate lymphocyte type (Cas-L), a docking protein that associates with a variety of signaling molecules, such as focal adhesion kinase (FAK), proline-rich tyrosine kinase 2 (Pyk2), and Crk. We investigated the involvement of these proteins in the pathophysiology of global cerebral ischemia. METHODS: The mouse Cas-L/Nedd9 cDNAs were cloned. The expression and function of Cas-L/Nedd9 protein in the pathogenesis of global ischemia in rats was investigated by RT-PCR, Western blot analysis, and immunohistochemistry. The neurite outgrowth of the transfectants of Nedd9 deletion mutants in PC-12 cells was also assessed to clarify the function of the Nedd9 protein. RESULTS: Nedd9 was a splicing variant of Cas-L and was selectively induced in neurons of the cerebral cortex and hippocampus 1 to 14 days after the ischemia. Induced Nedd9 protein was tyrosine phosphorylated and was bound to FAK in dendrite and soma of neurons after the ischemia. Finally, it was demonstrated that Nedd9 promoted neurite outgrowth of PC-12 cells. CONCLUSIONS: Our study may support the potential of Nedd9 for participation in the differentiation of neurons after global ischemia in rats.


Subject(s)
Down-Regulation , Ischemia/pathology , Neurons/pathology , Phosphoproteins/biosynthesis , Phosphoproteins/genetics , Up-Regulation , Adaptor Proteins, Signal Transducing , Alternative Splicing , Amino Acid Sequence , Animals , Blotting, Western , Cell Differentiation , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Cloning, Molecular , Cytoskeletal Proteins/biosynthesis , DNA, Complementary/metabolism , Dendrites/pathology , Focal Adhesion Kinase 2/metabolism , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Gene Deletion , Gene Expression Regulation, Developmental , Gene Transfer Techniques , Hippocampus/metabolism , Hippocampus/pathology , Immunoblotting , Immunohistochemistry , Immunoprecipitation , Intracellular Signaling Peptides and Proteins , LIM Domain Proteins , Male , Mice , Microfilament Proteins , Mixed Function Oxygenases , Models, Genetic , Molecular Sequence Data , Mutation , Nerve Growth Factor/metabolism , Neurons/metabolism , Oncogene Protein v-crk/metabolism , PC12 Cells , Phosphorylation , Protein Structure, Tertiary , Rats , Rats, Sprague-Dawley , Retroviridae/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Transcription, Genetic , Transfection , Tyrosine/chemistry
17.
PLoS One ; 10(7): e0132105, 2015.
Article in English | MEDLINE | ID: mdl-26132171

ABSTRACT

Profiling cellular proteome is critical to understanding signal integration during cell fate determination. In this study, the capability of capillary isoelectric focusing (cIEF) immunoassays to detect post-translational modifications (PTM) of protein isoforms is demonstrated. cIEF immunoassays exhibit protein detection sensitivity at up to 5 orders of magnitude higher than traditional methods. This detection ultra-sensitivity permits proteomic profiling of several nanograms of tissue samples. cIEF immunoassays are employed to simultaneously profile three protein kinases during fat cell differentiation: cGMP-dependent protein kinase type I (PKG-I) of the nitric oxide (NO) signaling pathway, protein kinase B (Akt) of the insulin signaling pathway, and extracellular signal-regulated kinase (ERK) of the mitogen-activated protein kinase (MAPK) signaling pathway. Interestingly, a switch in the expression level of PKG- isoforms is observed during fat cell differentiation. While both PKG-Iα and PKG-Iß isoforms are present in preadipocytes, only PKG-Iß isoform is expressed in adipocytes. On the other hand, the phosphorylation level increases for Akt while decreases for ERK1 and ERK2 following the maturation of preadipocytes into adipocytes. Taken together, cIEF immunoassay provides a highly sensitive means to study fat cell differentiation proteomics. cIEF immunoassay should be a powerful proteomics tool to study complex protein signal integration in biological systems.


Subject(s)
Adipocytes/enzymology , Adipogenesis , Electrophoresis, Capillary/methods , Isoelectric Focusing/methods , Proteomics/methods , Adipocytes/cytology , Blotting, Western , Cell Line , Cells, Cultured , Cyclic GMP-Dependent Protein Kinase Type I/analysis , Cyclic GMP-Dependent Protein Kinase Type I/biosynthesis , HeLa Cells/chemistry , Humans , Microscopy/methods , Mitogen-Activated Protein Kinase 1/analysis , Mitogen-Activated Protein Kinase 3/analysis , Omentum/cytology , Phosphorylation , Protein Isoforms/analysis , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-akt/analysis , Signal Transduction , Spectrum Analysis, Raman
18.
BMC Pharmacol Toxicol ; 15: 27, 2014 May 23.
Article in English | MEDLINE | ID: mdl-24887406

ABSTRACT

BACKGROUND: Tamoxifen, an agonist of estrogen receptor, is widely prescribed for the prevention and long-term treatment of breast cancer. A side effect of tamoxifen is fatty liver, which increases the risk for non-alcoholic fatty liver disease. Prevention of tamoxifen-induced fatty liver has the potential to improve the safety of long-term tamoxifen usage. METHODS: Uridine, a pyrimidine nucleoside with reported protective effects against drug-induced fatty liver, was co-administered with tamoxifen in C57BL/6J mice. Liver lipid levels were evaluated with lipid visualization using coherent anti-Stokes Raman scatting (CARS) microscopy, biochemical assay measurement of triacylglyceride (TAG), and liquid chromatography coupled with mass spectrometry (LC-MS) measurement of membrane phospholipid. Blood TAG and cholesterol levels were measured. Mitochondrial respiration of primary hepatocytes in the presence of tamoxifen and/or uridine was evaluated by measuring oxygen consumption rate with an extracellular flux analyzer. Liver protein lysine acetylation profiles were evaluated with 1D and 2D Western blots. In addition, the relationship between endogenous uridine levels, fatty liver, and tamoxifen administration was evaluated in transgenic mice UPase1-/-and UPase1-TG. RESULTS: Uridine co-administration prevented tamoxifen-induced liver lipid droplet accumulation in mice. The most prominent effect of uridine co-administration with tamoxifen was the stimulation of liver membrane phospholipid biosynthesis. Uridine had no protective effect against tamoxifen-induced impairment to mitochondrial respiration of primary hepatocytes or liver TAG and cholesterol export. Uridine had no effect on tamoxifen-induced changes to liver protein acetylation profile. Transgenic mice UPase1-/-with increased pyrimidine salvage activity were protected against tamoxifen-induced liver lipid droplet accumulation. In contrast, UPase1-TG mice with increased pyrimidine catabolism activity had intrinsic liver lipid droplet accumulation, which was aggravated following tamoxifen administration. CONCLUSION: Uridine co-administration was effective at preventing tamoxifen-induced liver lipid droplet accumulation. The ability of uridine to prevent tamoxifen-induced fatty liver appeared to depend on the pyrimidine salvage pathway, which promotes biosynthesis of membrane phospholipid.


Subject(s)
Antineoplastic Agents, Hormonal/adverse effects , Fatty Liver/prevention & control , Protective Agents/therapeutic use , Tamoxifen/adverse effects , Uridine/therapeutic use , Animals , Fatty Liver/chemically induced , Fatty Liver/metabolism , Female , Lipid Metabolism/drug effects , Lipids/blood , Liver/drug effects , Liver/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Protective Agents/pharmacology , Uridine/pharmacology , Uridine Phosphorylase/deficiency , Uridine Phosphorylase/genetics
19.
PLoS One ; 9(6): e99728, 2014.
Article in English | MEDLINE | ID: mdl-24918436

ABSTRACT

Purines and pyrimidines are complementary bases of the genetic code. The roles of purines and their derivatives in cellular signal transduction and energy metabolism are well-known. In contrast, the roles of pyrimidines and their derivatives in cellular function remain poorly understood. In this study, the roles of uridine, a pyrimidine nucleoside, in liver metabolism are examined in mice. We report that short-term uridine administration in C57BL/6J mice increases liver protein glycosylation profiles, reduces phosphorylation level of insulin signaling proteins, and activates the HRI-eIF-2α-ATF4 heme-deficiency stress response pathway. Short-term uridine administration is also associated with reduced liver hemin level and reduced ability for insulin-stimulated blood glucose removal during an insulin tolerance test. Some of the short-term effects of exogenous uridine in C57BL/6J mice are conserved in transgenic UPase1-/- mice with long-term elevation of endogenous uridine level. UPase1-/- mice exhibit activation of the liver HRI-eIF-2α-ATF4 heme-deficiency stress response pathway. UPase1-/- mice also exhibit impaired ability for insulin-stimulated blood glucose removal. However, other short-term effects of exogenous uridine in C57BL/6J mice are not conserved in UPase1-/- mice. UPase1-/- mice exhibit normal phosphorylation level of liver insulin signaling proteins and increased liver hemin concentration compared to untreated control C57BL/6J mice. Contrasting short-term and long-term consequences of uridine on liver metabolism suggest that uridine exerts transient effects and elicits adaptive responses. Taken together, our data support potential roles of pyrimidines and their derivatives in the regulation of liver metabolism.


Subject(s)
Glycosylation/drug effects , Heme/metabolism , Insulin/metabolism , Liver/drug effects , Protein Biosynthesis/drug effects , Signal Transduction/drug effects , Uridine/administration & dosage , Activating Transcription Factor 4/metabolism , Animals , Blood Glucose/drug effects , Eukaryotic Initiation Factor-2/metabolism , Glucose/metabolism , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phosphorylation/drug effects
20.
PLoS One ; 9(1): e87179, 2014.
Article in English | MEDLINE | ID: mdl-24475249

ABSTRACT

Uridine, a pyrimidine nucleoside, can modulate liver lipid metabolism although its specific acting targets have not been identified. Using mice with fenofibrate-induced fatty liver as a model system, the effects of uridine on liver lipid metabolism are examined. At a daily dosage of 400 mg/kg, fenofibrate treatment causes reduction of liver NAD(+)/NADH ratio, induces hyper-acetylation of peroxisomal bifunctional enzyme (ECHD) and acyl-CoA oxidase 1 (ACOX1), and induces excessive accumulation of long chain fatty acids (LCFA) and very long chain fatty acids (VLCFA). Uridine co-administration at a daily dosage of 400 mg/kg raises NAD(+)/NADH ratio, inhibits fenofibrate-induced hyper-acetylation of ECHD, ACOX1, and reduces accumulation of LCFA and VLCFA. Our data indicates a therapeutic potential for uridine co-administration to prevent fenofibrate-induced fatty liver.


Subject(s)
Fatty Liver/prevention & control , Fenofibrate/adverse effects , Hypolipidemic Agents/adverse effects , Protective Agents/pharmacology , Uridine/pharmacology , Acyl-CoA Oxidase/genetics , Acyl-CoA Oxidase/metabolism , Animals , Biological Transport/drug effects , Fatty Acids/metabolism , Fatty Liver/chemically induced , Fatty Liver/enzymology , Fatty Liver/pathology , Gene Expression Regulation , Lipid Metabolism/drug effects , Liver/drug effects , Liver/enzymology , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NAD/metabolism , Oxidation-Reduction/drug effects , Peroxisomal Bifunctional Enzyme/genetics , Peroxisomal Bifunctional Enzyme/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL