Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Exp Eye Res ; 201: 108326, 2020 12.
Article in English | MEDLINE | ID: mdl-33147472

ABSTRACT

The Descemet's membrane (DM) and the lens capsule (LC) are two ocular basement membranes (BMs) that are essential in maintaining stability and structure of the cornea and lens. In this study, we investigated the proteomes and biomechanical properties of these two materials to uncover common and unique properties. We also screened for possible protein changes during diabetes. LC-MS/MS was used to determine the proteomes of both BMs. Biomechanical measurements were conducted by atomic force microscopy (AFM) in force spectroscopy mode, and complemented with immunofluorescence microscopy. Proteome analysis showed that all six existing collagen IV chains represent 70% of all LC-protein, and are thus the dominant components of the LC. The DM on the other hand is predominantly composed of a single protein, TGF-induced protein, which accounted for around 50% of all DM-protein. Four collagen IV-family members in DM accounted for only 10% of the DM protein. Unlike the retinal vascular BMs, the LC and DM do not undergo significant changes in their protein compositions during diabetes. Nanomechanical measurements showed that the endothelial/epithelial sides of both BMs are stiffer than their respective stromal/anterior-chamber sides, and both endothelial and stromal sides of the DM were stiffer than the epithelial and anterior-chamber sides of the LC. Long-term diabetes did not change the stiffness of the DM and LC. In summary, our analyses show that the protein composition and biomechanical properties of the DM and LC are different, i.e., the LC is softer than DM despite a significantly higher concentration of collagen IV family members. This finding is unexpected, as collagen IV members are presumed to be responsible for BM stiffness. Diabetes had no significant effect on the protein composition and the biomechanical properties of both the DM and LC.


Subject(s)
Basement Membrane/metabolism , Cornea/metabolism , Descemet Membrane/metabolism , Eye Proteins/metabolism , Lens Capsule, Crystalline/metabolism , Aged , Basement Membrane/cytology , Chromatography, Liquid , Descemet Membrane/cytology , Elasticity , Female , Humans , Lens Capsule, Crystalline/cytology , Male , Microscopy, Atomic Force , Middle Aged , Tandem Mass Spectrometry
2.
Sci Rep ; 9(1): 16720, 2019 11 13.
Article in English | MEDLINE | ID: mdl-31723180

ABSTRACT

Previous studies in model eukaryotes have demonstrated that phosphorylation of heterochromatin protein 1 (HP1) is important for dynamically regulating its various functions. However, in the malaria parasite Plasmodium falciparum both the function of HP1 phosphorylation and the identity of the protein kinases targeting HP1 are still elusive. In order to functionally analyze phosphorylation of P. falciparum HP1 (PfHP1), we first mapped PfHP1 phosphorylation sites by liquid chromatography tandem mass spectrometry (LC-MS/MS) analysis of native PfHP1, which identified motifs from which potential kinases could be predicted; in particular, several phosphorylated residues were embedded in motifs rich in acidic residues, reminiscent of targets for P. falciparum casein kinase 2 (PfCK2). Secondly, we tested recombinant PfCK2 and a number of additional protein kinases for their ability to phosphorylate PfHP1 in in vitro kinase assays. These experiments validated our prediction that PfHP1 acts as a substrate for PfCK2. Furthermore, LC-MS/MS analysis showed that PfCK2 phosphorylates three clustered serine residues in an acidic motif within the central hinge region of PfHP1. To study the role of PfHP1 phosphorylation in live parasites we used CRISPR/Cas9-mediated genome editing to generate a number of conditional PfHP1 phosphomutants based on the DiCre/LoxP system. Our studies revealed that neither PfCK2-dependent phosphorylation of PfHP1, nor phosphorylation of the hinge domain in general, affect PfHP1's ability to localize to heterochromatin, and that PfHP1 phosphorylation in this region is dispensable for the proliferation of P. falciparum blood stage parasites.


Subject(s)
Casein Kinase II/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Heterochromatin/metabolism , Malaria, Falciparum/parasitology , Plasmodium falciparum/isolation & purification , Plasmodium falciparum/metabolism , Protozoan Proteins/metabolism , Amino Acid Sequence , Casein Kinase II/genetics , Chromobox Protein Homolog 5 , Chromosomal Proteins, Non-Histone/genetics , Humans , Malaria, Falciparum/metabolism , Mutation , Phosphorylation , Protozoan Proteins/genetics
3.
EBioMedicine ; 46: 32-41, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31331834

ABSTRACT

BACKGROUND: Gliomas are the most frequent and aggressive malignancies of the central nervous system. Decades of molecular analyses have demonstrated that gliomas accumulate genetic alterations that culminate in enhanced activity of receptor tyrosine kinases and downstream mediators. While the genetic alterations, like gene amplification or loss, have been well characterized, little information exists about changes in the proteome of gliomas of different grades. METHODS: We performed unbiased quantitative proteomics of human glioma biopsies by mass spectrometry followed by bioinformatic analysis. FINDINGS: Various pathways were found to be up- or downregulated. In particular, endocytosis as pathway was affected by a vast and concomitant reduction of multiple machinery components involved in initiation, formation, and scission of endocytic carriers. Both clathrin-dependent and -independent endocytosis were changed, since not only clathrin, AP-2 adaptins, and endophilins were downregulated, but also dynamin that is shared by both pathways. The reduction of endocytic machinery components caused increased receptor cell surface levels, a prominent phenotype of defective endocytosis. Analysis of additional biopsies revealed that depletion of endocytic machinery components was a common trait of various glioma grades and subclasses. INTERPRETATION: We propose that impaired endocytosis creates a selective advantage in glioma tumor progression due to prolonged receptor tyrosine kinase signaling from the cell surface. FUND: This work was supported by Grants 316030-164105 (to P. Jenö), 31003A-162643 (to M. Spiess) and PP00P3-176974 (to G. Hutter) from the Swiss National Science Foundation. Further funding was received by the Department of Surgery from the University Hospital Basel.


Subject(s)
Endocytosis , Glioma/metabolism , Proteome , Proteomics , Biopsy , Computational Biology/methods , Glioma/genetics , Glioma/pathology , Humans , Mass Spectrometry , Neoplasm Grading , Neoplasm Staging , Neoplastic Cells, Circulating , Proteomics/methods
4.
Brain Pathol ; 29(3): 336-350, 2019 05.
Article in English | MEDLINE | ID: mdl-30403311

ABSTRACT

Diffuse gliomas progress by invading neighboring brain tissue to promote postoperative relapse. Transcription factor SOX2 is highly expressed in invasive gliomas and maps to chromosome region 3q26 together with the genes for PI3K/AKT signaling activator PIK3CA and effector molecules of mitochondria fusion and cell invasion, MFN1 and OPA1. Gene copy number analysis at 3q26 from 129 glioma patient biopsies revealed mutually exclusive SOX2 amplifications (26%) and OPA1 losses (19%). Both forced SOX2 expression and OPA1 inactivation increased LN319 glioma cell invasion in vitro and promoted cell dispersion in vivo in xenotransplanted D. rerio embryos. While PI3 kinase activity sustained SOX2 expression, pharmacological PI3K/AKT pathway inhibition decreased invasion and resulted in SOX2 nucleus-to-cytoplasm translocation in an mTORC1-independent manner. Chromatin immunoprecipitation and luciferase reporter gene assays together demonstrated that SOX2 trans-activates PIK3CA and OPA1. Thus, SOX2 activates PI3K/AKT signaling in a positive feedback loop, while OPA1 deletion is interpreted to counteract OPA1 trans-activation. Remarkably, neuroimaging of human gliomas with high SOX2 or low OPA1 genomic imbalances revealed significantly larger necrotic tumor zone volumes, corresponding to higher invasive capacities of tumors, while autologous necrotic cells are capable of inducing higher invasion in SOX2 overexpressing or OPA1 knocked-down relative to parental LN319. We thus propose necrosis volume as a surrogate marker for the assessment of glioma invasive potential. Whereas glioma invasion is activated by a PI3K/AKT-SOX2 loop, it is reduced by a cryptic invasion suppressor SOX2-OPA1 pathway. Thus, PI3K/AKT-SOX2 and mitochondria fission represent connected signaling networks regulating glioma invasion.


Subject(s)
Chromosomes, Human, Pair 3 , Class I Phosphatidylinositol 3-Kinases/genetics , GTP Phosphohydrolases/genetics , Glioma/genetics , SOXB1 Transcription Factors/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Class I Phosphatidylinositol 3-Kinases/metabolism , DNA Copy Number Variations , GTP Phosphohydrolases/metabolism , Glioma/metabolism , Glioma/pathology , HEK293 Cells , Humans , Necrosis/genetics , Neoplasm Invasiveness , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/pathology , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , SOXB1 Transcription Factors/metabolism , Signal Transduction
5.
Hypertension ; 73(2): 469-480, 2019 02.
Article in English | MEDLINE | ID: mdl-30580688

ABSTRACT

Primary aldosteronism is a disease of excessive production of adrenal steroid hormones and the most common cause of endocrine hypertension. Primary aldosteronism results mainly from bilateral adrenal hyperplasia or unilateral aldosterone-producing adenoma (APA). Primary aldosteronism cause at the molecular level is incompletely understood and a targeted treatment preventing excessive adrenal steroid production is not available. Here, we perform deep quantitative proteomic and phosphoproteomic profiling of 6 pairs of APA and adjacent nontumoral adrenal cortex. We show that increased steroidogenesis in APA is accompanied by upregulation of steroidogenic enzymes (HSD3B2, CYP21A2, CYP11B2) and of proteins involved in cholesterol uptake (LSR). We demonstrate that HSD3B2 is phosphorylated at Ser95 or 96 and identify a novel phosphorylation site, Ser489, in CYP21A2, suggesting that steroidogenic enzymes are regulated by phosphorylation. Our analysis also reveals altered ECM (extracellular matrix) composition in APA that affects ECM-cell surface interactions and actin cytoskeleton rearrangements. We show that RHOC, a GTPase controlling actin organization in response to extracellular stimuli, is upregulated in APA and promotes expression of the aldosterone synthase gene CYP11B2. Our data also indicate deregulation of protein N-glycosylation and GABAergic signaling in APAs. Finally, we find that mTORC1 (mammalian target of rapamycin complex 1) signaling is the major pathway deregulated in APA. Our study provides a rich resource for future research on the molecular mechanisms of primary aldosteronism.


Subject(s)
Adenoma/metabolism , Aldosterone/biosynthesis , Proteomics/methods , Extracellular Matrix/metabolism , Humans , Mechanistic Target of Rapamycin Complex 1/physiology , Signal Transduction , gamma-Aminobutyric Acid/metabolism
6.
Invest Ophthalmol Vis Sci ; 59(8): 3521-3530, 2018 07 02.
Article in English | MEDLINE | ID: mdl-30025099

ABSTRACT

Purpose: To date, no biomarkers for ocular graft versus host disease (GvHD), a frequent complication following allogeneic hematopoietic cell transplantation (HCT), exist. In this prospective study, we evaluated the potential of human tear proteins as biomarkers for ocular GvHD. Methods: Tears from 10 patients with moderate-to-severe ocular GvHD were compared to 10 patients without ocular GvHD. After a full ocular surface clinical examination, tears were collected onto Schirmer strips and protein composition was analyzed by liquid chromatography tandem mass spectrometry. Statistical evaluation was performed using the Mann-Whitney U test to compare means and the false discovery rate method to adjust for multiple comparisons. Functional annotation of differentially expressed proteins was done with the PANTHER classification system. Results: We identified 282 proteins in tryptic digests of Schirmer strips; 79 proteins were significantly differentially expressed between the two groups, from which 54 were up- and 25 downregulated. The most upregulated proteins were classified as nucleic acid binding and cytoskeletal proteins, while the most extensively downregulated proteins belong to an array of classes including transfer and receptor proteins, enzyme modulators, and hydrolases. In addition to proteins already confirmed as differentially expressed in dry eye disease, we report changes in 36 novel proteins. Conclusions: This study reports the proteomic profile of tears in ocular GvHD for the first time and identifies a number of unique differentially expressed proteins. Further studies with a higher number of participants are necessary to confirm these results and to evaluate the reliability of these expression patterns in longitudinal studies.


Subject(s)
Biomarkers/metabolism , Eye Diseases/metabolism , Eye Proteins/metabolism , Graft vs Host Disease/metabolism , Hematopoietic Stem Cell Transplantation/adverse effects , Tears/metabolism , Adult , Aged , Chromatography, Liquid , Eye Diseases/etiology , Female , Graft vs Host Disease/etiology , Humans , Male , Middle Aged , Prospective Studies , Proteomics , Tandem Mass Spectrometry , Transplantation, Homologous , Young Adult
8.
Nature ; 555(7698): 678-682, 2018 03 29.
Article in English | MEDLINE | ID: mdl-29562234

ABSTRACT

Histidine phosphorylation, the so-called hidden phosphoproteome, is a poorly characterized post-translational modification of proteins. Here we describe a role of histidine phosphorylation in tumorigenesis. Proteomic analysis of 12 tumours from an mTOR-driven hepatocellular carcinoma mouse model revealed that NME1 and NME2, the only known mammalian histidine kinases, were upregulated. Conversely, expression of the putative histidine phosphatase LHPP was downregulated specifically in the tumours. We demonstrate that LHPP is indeed a protein histidine phosphatase. Consistent with these observations, global histidine phosphorylation was significantly upregulated in the liver tumours. Sustained, hepatic expression of LHPP in the hepatocellular carcinoma mouse model reduced tumour burden and prevented the loss of liver function. Finally, in patients with hepatocellular carcinoma, low expression of LHPP correlated with increased tumour severity and reduced overall survival. Thus, LHPP is a protein histidine phosphatase and tumour suppressor, suggesting that deregulated histidine phosphorylation is oncogenic.


Subject(s)
Histidine/metabolism , Inorganic Pyrophosphatase/metabolism , Liver Neoplasms/enzymology , Liver Neoplasms/pathology , Tumor Suppressor Proteins/metabolism , Animals , Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/pathology , Disease Models, Animal , Humans , Inorganic Pyrophosphatase/deficiency , Inorganic Pyrophosphatase/genetics , Male , Mice , Phosphorylation , Proteomics , Survival Analysis , TOR Serine-Threonine Kinases/metabolism , Tumor Burden , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics
9.
J Clin Invest ; 128(4): 1538-1550, 2018 04 02.
Article in English | MEDLINE | ID: mdl-29528335

ABSTRACT

Obesity is a major risk factor for insulin resistance and type 2 diabetes. In adipose tissue, obesity-mediated insulin resistance correlates with the accumulation of proinflammatory macrophages and inflammation. However, the causal relationship of these events is unclear. Here, we report that obesity-induced insulin resistance in mice precedes macrophage accumulation and inflammation in adipose tissue. Using a mouse model that combines genetically induced, adipose-specific insulin resistance (mTORC2-knockout) and diet-induced obesity, we found that insulin resistance causes local accumulation of proinflammatory macrophages. Mechanistically, insulin resistance in adipocytes results in production of the chemokine monocyte chemoattractant protein 1 (MCP1), which recruits monocytes and activates proinflammatory macrophages. Finally, insulin resistance (high homeostatic model assessment of insulin resistance [HOMA-IR]) correlated with reduced insulin/mTORC2 signaling and elevated MCP1 production in visceral adipose tissue from obese human subjects. Our findings suggest that insulin resistance in adipose tissue leads to inflammation rather than vice versa.


Subject(s)
Insulin Resistance , Intra-Abdominal Fat/metabolism , Macrophages/metabolism , Obesity/metabolism , Panniculitis/metabolism , Signal Transduction , 3T3-L1 Cells , Animals , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Humans , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Intra-Abdominal Fat/pathology , Macrophages/pathology , Mechanistic Target of Rapamycin Complex 2/genetics , Mechanistic Target of Rapamycin Complex 2/metabolism , Mice , Mice, Knockout , Obesity/genetics , Obesity/pathology , Panniculitis/genetics , Panniculitis/pathology
10.
Science ; 359(6381): 1259-1263, 2018 03 16.
Article in English | MEDLINE | ID: mdl-29590075

ABSTRACT

Malaria is caused by Plasmodium parasites that proliferate in the bloodstream. During each replication cycle, some parasites differentiate into gametocytes, the only forms able to infect the mosquito vector and transmit malaria. Sexual commitment is triggered by activation of AP2-G, the master transcriptional regulator of gametocytogenesis. Heterochromatin protein 1 (HP1)-dependent silencing of ap2-g prevents sexual conversion in proliferating parasites. In this study, we identified Plasmodium falciparum gametocyte development 1 (GDV1) as an upstream activator of sexual commitment. We found that GDV1 targeted heterochromatin and triggered HP1 eviction, thus derepressing ap2-g Expression of GDV1 was responsive to environmental triggers of sexual conversion and controlled via a gdv1 antisense RNA. Hence, GDV1 appears to act as an effector protein that induces sexual differentiation by antagonizing HP1-dependent gene silencing.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , Gametogenesis/genetics , Gene Silencing , Malaria, Falciparum/parasitology , Plasmodium falciparum/growth & development , Sex Differentiation/genetics , Animals , Chromobox Protein Homolog 5 , Plasmodium falciparum/genetics
11.
Cancer Cell ; 32(6): 807-823.e12, 2017 Dec 11.
Article in English | MEDLINE | ID: mdl-29232555

ABSTRACT

Dysregulated mammalian target of rapamycin (mTOR) promotes cancer, but underlying mechanisms are poorly understood. We describe an mTOR-driven mouse model that displays hepatosteatosis progressing to hepatocellular carcinoma (HCC). Longitudinal proteomic, lipidomics, and metabolomic analyses revealed that hepatic mTORC2 promotes de novo fatty acid and lipid synthesis, leading to steatosis and tumor development. In particular, mTORC2 stimulated sphingolipid (glucosylceramide) and glycerophospholipid (cardiolipin) synthesis. Inhibition of fatty acid or sphingolipid synthesis prevented tumor development, indicating a causal effect in tumorigenesis. Increased levels of cardiolipin were associated with tubular mitochondria and enhanced oxidative phosphorylation. Furthermore, increased lipogenesis correlated with elevated mTORC2 activity and HCC in human patients. Thus, mTORC2 promotes cancer via formation of lipids essential for growth and energy production.


Subject(s)
Carcinogenesis/metabolism , Carcinoma, Hepatocellular/metabolism , Fatty Liver/metabolism , Lipogenesis/physiology , Liver Neoplasms/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , Animals , Carcinoma, Hepatocellular/etiology , Cell Transformation, Neoplastic/metabolism , Fatty Liver/complications , Humans , Lipids/biosynthesis , Liver Neoplasms/etiology , Mice , Mice, Knockout
12.
PLoS One ; 12(12): e0189857, 2017.
Article in English | MEDLINE | ID: mdl-29284024

ABSTRACT

Basement membranes (BMs) are specialized sheets of extracellular matrix that outline epithelial cell layers, muscle fibers, blood vessels, and peripheral nerves. A well-documented histological hallmark of progressing diabetes is a major increase in vascular BM thickness. In order to investigate whether this structural change is accompanied by a change in the protein composition, we compared the proteomes of retinal vascular BMs from diabetic and non-diabetic donors by using LC-MS/MS. Data analysis showed that seventeen extracellular matrix (ECM)-associated proteins were more abundant in diabetic than non-diabetic vascular BMs. Four ECM proteins were more abundant in non-diabetic than in diabetic BMs. Most of the over-expressed proteins implicate a complement-mediated chronic inflammatory process in the diabetic retinal vasculature. We also found an up-regulation of norrin, a protein that is known to promote vascular proliferation, possibly contributing to the vascular remodeling during diabetes. Many of the over-expressed proteins were localized to microvascular aneurisms. Further, the overall stoichiometry of proteins was changed, such that the relative abundance of collagens in BMs from diabetic patients was higher than normal. Biomechanical measurements of vascular BM flat mounts using AFM showed that their outer surface was softer than normal.


Subject(s)
Basement Membrane/metabolism , Diabetes Mellitus/metabolism , Eye Proteins/metabolism , Retinal Vessels/metabolism , Case-Control Studies , Chromatography, Liquid , Humans , Microscopy, Atomic Force , Proteome , Retinal Vessels/pathology , Tandem Mass Spectrometry
13.
Nat Microbiol ; 2: 17033, 2017 Mar 13.
Article in English | MEDLINE | ID: mdl-28288093

ABSTRACT

Telomere repeat-binding factors (TRFs) are essential components of the molecular machinery that regulates telomere function. TRFs are widely conserved across eukaryotes and bind duplex telomere repeats via a characteristic MYB-type domain. Here, we identified the telomere repeat-binding protein PfTRZ in the malaria parasite Plasmodium falciparum, a member of the Alveolate phylum for which TRFs have not been described so far. PfTRZ lacks an MYB domain and binds telomere repeats via a C2H2-type zinc finger domain instead. In vivo, PfTRZ binds with high specificity to the telomeric tract and to interstitial telomere repeats upstream of subtelomeric virulence genes. Conditional depletion experiments revealed that PfTRZ regulates telomere length homeostasis and is required for efficient cell cycle progression. Intriguingly, we found that PfTRZ also binds to and regulates the expression of 5S rDNA genes. Combined with detailed phylogenetic analyses, our findings identified PfTRZ as a remote functional homologue of the basic transcription factor TFIIIA, which acquired a new function in telomere maintenance early in the apicomplexan lineage. Our work sheds unexpected new light on the evolution of telomere repeat-binding proteins and paves the way for dissecting the presumably divergent mechanisms regulating telomere functionality in one of the most deadly human pathogens.


Subject(s)
Evolution, Molecular , Malaria, Falciparum/genetics , Telomere-Binding Proteins/genetics , Protein Binding , Telomere/metabolism , Transcription Factor TFIIIA/genetics , Zinc Fingers
14.
Sci Adv ; 2(12): e1601756, 2016 Dec.
Article in English | MEDLINE | ID: mdl-28028542

ABSTRACT

We report that the anticancer activity of the widely used diabetic drug metformin is strongly potentiated by syrosingopine. Synthetic lethality elicited by combining the two drugs is synergistic and specific to transformed cells. This effect is unrelated to syrosingopine's known role as an inhibitor of the vesicular monoamine transporters. Syrosingopine binds to the glycolytic enzyme α-enolase in vitro, and the expression of the γ-enolase isoform correlates with nonresponsiveness to the drug combination. Syrosingopine sensitized cancer cells to metformin and its more potent derivative phenformin far below the individual toxic threshold of each compound. Thus, combining syrosingopine and codrugs is a promising therapeutic strategy for clinical application for the treatment of cancer.


Subject(s)
Metformin/pharmacology , Reserpine/analogs & derivatives , Animals , Cell Line, Tumor , Drug Synergism , Glycolysis , Humans , Mice , Mice, Knockout , Phenformin/pharmacology , Phosphopyruvate Hydratase/chemistry , Reserpine/pharmacology
15.
Bioanalysis ; 8(12): 1279-96, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27187191

ABSTRACT

BACKGROUND: Uromodulin is the most abundant protein in healthy human urine. Recently it has been suggested as a specific biomarker of renal tubular damage. We have developed a novel pseudo multiple reaction monitoring (pseudo MRM) for the protein's quantification in human urine. RESULTS: Selection of two peptides allowed quantification of uromodulin in human urine. The pseudo MRM quantified uromodulin in healthy individuals between 21 and 1344 nM and in autosomal dominant tubulointerstitial kidney disease-UMOD patients between 2 and 25 nM. CONCLUSION: The pseudo MRM allows greater confidence in assay specificity than traditional MRM methods and quantified uromodulin at concentrations higher than achievable by ELISA. Differences in urinary uromodulin concentration related to the rs4293393 promoter variant in the UMOD gene was confirmed. This method will be used to further investigate uromodulin as a biomarker of renal injury.


Subject(s)
Urinalysis/methods , Uromodulin/urine , Adult , Amino Acid Sequence , Biomarkers/urine , Chromatography, Liquid/methods , Humans , Isotope Labeling/methods , Limit of Detection , Male , Middle Aged , Proteolysis , Tandem Mass Spectrometry/methods , Uromodulin/analysis
16.
Proc Natl Acad Sci U S A ; 113(5): 1381-6, 2016 Feb 02.
Article in English | MEDLINE | ID: mdl-26787912

ABSTRACT

Compensatory signaling pathways in tumors confer resistance to targeted therapy, but the pathways and their mechanisms of activation remain largely unknown. We describe a procedure for quantitative proteomics and phosphoproteomics on snap-frozen biopsies of hepatocellular carcinoma (HCC) and matched nontumor liver tissue. We applied this procedure to monitor signaling pathways in serial biopsies taken from an HCC patient before and during treatment with the multikinase inhibitor sorafenib. At diagnosis, the patient had an advanced HCC. At the time of the second biopsy, abdominal imaging revealed progressive disease despite sorafenib treatment. Sorafenib was confirmed to inhibit MAPK signaling in the tumor, as measured by reduced ribosomal protein S6 kinase phosphorylation. Hierarchical clustering and enrichment analysis revealed pathways broadly implicated in tumor progression and resistance, such as epithelial-to-mesenchymal transition and cell adhesion pathways. Thus, we describe a protocol for quantitative analysis of oncogenic pathways in HCC biopsies and obtained first insights into the effect of sorafenib in vivo. This protocol will allow elucidation of mechanisms of resistance and enable precision medicine.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , Niacinamide/analogs & derivatives , Phenylurea Compounds/therapeutic use , Phosphoproteins/metabolism , Proteomics , Biopsy , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Niacinamide/therapeutic use , Phosphorylation , Sorafenib
18.
Cell Host Microbe ; 17(6): 741-51, 2015 Jun 10.
Article in English | MEDLINE | ID: mdl-26067602

ABSTRACT

During red-blood-cell-stage infection of Plasmodium falciparum, the parasite undergoes repeated rounds of replication, egress, and invasion. Erythrocyte invasion involves specific interactions between host cell receptors and parasite ligands and coordinated expression of genes specific to this step of the life cycle. We show that a parasite-specific bromodomain protein, PfBDP1, binds to chromatin at transcriptional start sites of invasion-related genes and directly controls their expression. Conditional PfBDP1 knockdown causes a dramatic defect in parasite invasion and growth and results in transcriptional downregulation of multiple invasion-related genes at a time point critical for invasion. Conversely, PfBDP1 overexpression enhances expression of these same invasion-related genes. PfBDP1 binds to acetylated histone H3 and a second bromodomain protein, PfBDP2, suggesting a potential mechanism for gene recognition and control. Collectively, these findings show that PfBDP1 critically coordinates expression of invasion genes and indicate that targeting PfBDP1 could be an invaluable tool in malaria eradication.


Subject(s)
Gene Expression Regulation , Plasmodium falciparum/pathogenicity , Protozoan Proteins/metabolism , Acetylation , Cells, Cultured , Chromatin/metabolism , Erythrocytes/parasitology , Gene Knockdown Techniques , Histones/metabolism , Host-Parasite Interactions , Plasmodium falciparum/genetics , Promoter Regions, Genetic , Protozoan Proteins/genetics
19.
FEBS J ; 282(7): 1167-81, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25645340

ABSTRACT

The induction of macropinocytosis in macrophages during an inflammatory response is important for clearance of pathogenic microbes as well as the generation of appropriate immune responses. Recent data suggest that cytokine stimulation of macrophages induces macropinocytosis through phosphorylation of the protein coronin 1, thereby redistributing coronin 1 from the cell cortex to the cytoplasm followed by the activation of phosphoinositol-3 (PI-3) kinase. However, how coronin 1 phosphorylation regulates these processes remains unclear. We here define an essential role for 14-3-3ζ in cytokine-induced and coronin-1-dependent macropinocytosis in macrophages. We found that, upon stimulation, phosphorylated coronin 1 transiently associated with 14-3-3ζ and receptor of activated C kinase 1 (RACK1). Importantly, downregulation of 14-3-3ζ, but not RACK1, prevented relocation of coronin 1, as well as the induction of PI-3 kinase activity and thereby macropinocytosis upon cytokine stimulation. Together these data define an essential role for 14-3-3ζ in the regulation of macropinocytosis in macrophages upon cytokine stimulation through modulation of the localization of coronin 1.


Subject(s)
14-3-3 Proteins/metabolism , Cytokines/physiology , Macrophages/metabolism , Microfilament Proteins/metabolism , Pinocytosis , Animals , Cell Line , Macrophage Activation , Macrophages/microbiology , Mice, Transgenic , Mycobacterium bovis/physiology , Neuropeptides/metabolism , Phosphoproteins/metabolism , Phosphorylation , Protein Processing, Post-Translational , Protein Transport , Receptors for Activated C Kinase
20.
Free Radic Biol Med ; 83: 361-72, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25697776

ABSTRACT

Oxidative folding in the endoplasmic reticulum (ER) involves ER oxidoreductin 1 (Ero1)-mediated disulfide formation in protein disulfide isomerase (PDI). In this process, Ero1 consumes oxygen (O2) and releases hydrogen peroxide (H2O2), but none of the published Ero1 crystal structures reveal any potential pathway for entry and exit of these reactants. We report that additional mutation of the Cys(208)-Cys(241) disulfide in hyperactive Ero1α (Ero1α-C104A/C131A) potentiates H2O2 production, ER oxidation, and cell toxicity. This disulfide clamps two helices that seal the flavin cofactor where O2 is reduced to H2O2. Through its carboxyterminal active site, PDI unlocks this seal by forming a Cys(208)/Cys(241)-dependent mixed-disulfide complex with Ero1α. The H2O2-detoxifying glutathione peroxidase 8 also binds to the Cys(208)/Cys(241) loop region. Supported by O2 diffusion simulations, these data describe the first enzymatically controlled O2 access into a flavoprotein active site, provide molecular-level understanding of Ero1α regulation and H2O2 production/detoxification, and establish the deleterious consequences of constitutive Ero1 activity.


Subject(s)
Apoptosis , Disulfides/metabolism , Hydrogen Peroxide/metabolism , Membrane Glycoproteins/metabolism , Oxidoreductases/metabolism , Oxygen/metabolism , Protein Disulfide-Isomerases/metabolism , Blotting, Western , Catalysis , Catalytic Domain , Cell Proliferation , Cells, Cultured , Endoplasmic Reticulum , Flavin-Adenine Dinucleotide/metabolism , Fluorescent Antibody Technique , HeLa Cells , Humans , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/genetics , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Membrane Proteins/metabolism , Molecular Chaperones/antagonists & inhibitors , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Oxidation-Reduction , Oxidoreductases/chemistry , Oxidoreductases/genetics , Protein Conformation , Protein Disulfide-Isomerases/chemistry , Protein Disulfide-Isomerases/genetics , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...