Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
PLoS One ; 17(10): e0275901, 2022.
Article in English | MEDLINE | ID: mdl-36260611

ABSTRACT

Non-invasive biomarkers of non-alcoholic fatty liver disease (NAFLD) supporting diagnosis and monitoring disease progression are urgently needed. The present study aimed to establish a bioinformatics pipeline capable of defining and validating NAFLD biomarker candidates based on paired hepatic global gene expression and plasma bioanalysis from individuals representing different stages of histologically confirmed NAFLD (no/mild, moderate, more advanced NAFLD). Liver secretome gene signatures were generated in a patient cohort of 26 severely obese individuals with the majority having no or mild fibrosis. To this end, global gene expression changes were compared between individuals with no/mild NAFLD and moderate/advanced NAFLD with subsequent filtering for candidate gene products with liver-selective expression and secretion. Four candidate genes, including LPA (lipoprotein A), IGFBP-1 (insulin-like growth factor-binding protein 1), SERPINF2 (serpin family F member 2) and MAT1A (methionine adenosyltransferase 1A), were differentially expressed in moderate/advanced NAFLD, which was confirmed in three independent RNA sequencing datasets from large, publicly available NAFLD studies. The corresponding gene products were quantified in plasma samples but could not discriminate among different grades of NAFLD based on NAFLD activity score. Conclusion: We demonstrate a novel approach based on the liver transcriptome allowing for identification of secreted hepatic gene products as potential circulating diagnostic biomarkers of NAFLD. Using this approach in larger NAFLD patient cohorts may yield potential circulating biomarkers for NAFLD severity.


Subject(s)
Non-alcoholic Fatty Liver Disease , Serpins , Somatomedins , Humans , Non-alcoholic Fatty Liver Disease/diagnosis , Non-alcoholic Fatty Liver Disease/genetics , Insulin-Like Growth Factor Binding Protein 1/metabolism , Methionine Adenosyltransferase/genetics , Secretome , Serpins/metabolism , Biomarkers , Somatomedins/metabolism , Lipoprotein(a)/metabolism
2.
Dis Model Mech ; 14(10)2021 10 01.
Article in English | MEDLINE | ID: mdl-34494644

ABSTRACT

The current understanding of molecular mechanisms driving diabetic kidney disease (DKD) is limited, partly due to the complex structure of the kidney. To identify genes and signalling pathways involved in the progression of DKD, we compared kidney cortical versus glomerular transcriptome profiles in uninephrectomized (UNx) db/db mouse models of early-stage (UNx only) and advanced [UNxplus adeno-associated virus-mediated renin-1 overexpression (UNx-Renin)] DKD using RNAseq. Compared to normoglycemic db/m mice, db/db UNx and db/db UNx-Renin mice showed marked changes in their kidney cortical and glomerular gene expression profiles. UNx-Renin mice displayed more marked perturbations in gene components associated with the activation of the immune system and enhanced extracellular matrix remodelling, supporting histological hallmarks of progressive DKD in this model. Single-nucleus RNAseq enabled the linking of transcriptome profiles to specific kidney cell types. In conclusion, integration of RNAseq at the cortical, glomerular and single-nucleus level provides an enhanced resolution of molecular signalling pathways associated with disease progression in preclinical models of DKD, and may thus be advantageous for identifying novel therapeutic targets in DKD.


Subject(s)
Diabetic Nephropathies/etiology , Diabetic Nephropathies/genetics , Gene Expression Profiling , Hypertension/complications , Animals , Dependovirus/metabolism , Disease Models, Animal , Female , Gene Expression Regulation , Kidney Cortex/metabolism , Kidney Cortex/pathology , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , Mice, Inbred C57BL , Renin/metabolism
3.
Mol Metab ; 47: 101171, 2021 05.
Article in English | MEDLINE | ID: mdl-33529728

ABSTRACT

OBJECTIVE: The development of effective anti-obesity therapeutics relies heavily on the ability to target specific brain homeostatic and hedonic mechanisms controlling body weight. To obtain further insight into neurocircuits recruited by anti-obesity drug treatment, the present study aimed to determine whole-brain activation signatures of six different weight-lowering drug classes. METHODS: Chow-fed C57BL/6J mice (n = 8 per group) received acute treatment with lorcaserin (7 mg/kg; i.p.), rimonabant (10 mg/kg; i.p.), bromocriptine (10 mg/kg; i.p.), sibutramine (10 mg/kg; p.o.), semaglutide (0.04 mg/kg; s.c.) or setmelanotide (4 mg/kg; s.c.). Brains were sampled two hours post-dosing and whole-brain neuronal activation patterns were analysed at single-cell resolution using c-Fos immunohistochemistry and automated quantitative three-dimensional (3D) imaging. RESULTS: The whole-brain analysis comprised 308 atlas-defined mouse brain areas. To enable fast and efficient data mining, a web-based 3D imaging data viewer was developed. All weight-lowering drugs demonstrated brain-wide responses with notable similarities in c-Fos expression signatures. Overlapping c-Fos responses were detected in discrete homeostatic and non-homeostatic feeding centres located in the dorsal vagal complex and hypothalamus with concurrent activation of several limbic structures as well as the dopaminergic system. CONCLUSIONS: Whole-brain c-Fos expression signatures of various weight-lowering drug classes point to a discrete set of brain regions and neurocircuits which could represent key neuroanatomical targets for future anti-obesity therapeutics.


Subject(s)
Anti-Obesity Agents/pharmacology , Brain/diagnostic imaging , Brain/metabolism , Pharmaceutical Preparations/metabolism , Animals , Body Weight , Cyclobutanes , Homeostasis , Imaging, Three-Dimensional , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Neurons/metabolism , Obesity/metabolism , Obesity/therapy , Proto-Oncogene Proteins c-fos/metabolism
4.
J Clin Endocrinol Metab ; 106(2): e966-e981, 2021 01 23.
Article in English | MEDLINE | ID: mdl-33135737

ABSTRACT

CONTEXT: The mechanisms underlying Roux-en-Y gastric bypass (RYGB) surgery-induced weight loss and the immediate postoperative beneficial metabolic effects associated with the operation remain uncertain. Enteroendocrine cell (EEC) secretory function has been proposed as a key factor in the marked metabolic benefits from RYGB surgery. OBJECTIVE: To identify novel gut-derived peptides with therapeutic potential in obesity and/or diabetes by profiling EEC-specific molecular changes in obese patients following RYGB-induced weight loss. SUBJECTS AND METHODS: Genome-wide expression analysis was performed in isolated human small intestinal EECs obtained from 20 gut-biopsied obese subjects before and after RYGB. Targets of interest were profiled for preclinical and clinical metabolic effects. RESULTS: Roux-en-Y gastric bypass consistently increased expression levels of the inverse ghrelin receptor agonist, liver-expressed antimicrobial peptide 2 (LEAP2). A secreted endogenous LEAP2 fragment (LEAP238-47) demonstrated robust insulinotropic properties, stimulating insulin release in human pancreatic islets comparable to the gut hormone glucagon-like peptide-1. LEAP238-47 showed reciprocal effects on growth hormone secretagogue receptor (GHSR) activity, suggesting that the insulinotropic action of the peptide may be directly linked to attenuation of tonic GHSR activity. The fragment was infused in healthy human individuals (n = 10), but no glucoregulatory effect was observed in the chosen dose as compared to placebo. CONCLUSIONS: Small intestinal LEAP2 expression was upregulated after RYGB. The corresponding circulating LEAP238-47 fragment demonstrated strong insulinotropic action in vitro but failed to elicit glucoregulatory effects in healthy human subjects.


Subject(s)
Antimicrobial Cationic Peptides/metabolism , Blood Proteins/metabolism , Gastric Bypass/methods , Gastrointestinal Tract/metabolism , Islets of Langerhans/metabolism , Obesity/surgery , Peptide Fragments/metabolism , Transcriptome , Adolescent , Adult , Antimicrobial Cationic Peptides/genetics , Biomarkers/analysis , Blood Proteins/genetics , Case-Control Studies , Cross-Over Studies , Double-Blind Method , Enteroendocrine Cells/metabolism , Enteroendocrine Cells/pathology , Female , Follow-Up Studies , Humans , Islets of Langerhans/pathology , Male , Obesity/pathology , Peptide Fragments/genetics , Prognosis , Prospective Studies , Young Adult
5.
BMC Gastroenterol ; 20(1): 210, 2020 Jul 06.
Article in English | MEDLINE | ID: mdl-32631250

ABSTRACT

BACKGROUND: Animal models of non-alcoholic steatohepatitis (NASH) are important tools in preclinical research and drug discovery. Gubra-Amylin NASH (GAN) diet-induced obese (DIO) mice represent a model of fibrosing NASH. The present study directly assessed the clinical translatability of the model by head-to-head comparison of liver biopsy histological and transcriptome changes in GAN DIO-NASH mouse and human NASH patients. METHODS: C57Bl/6 J mice were fed chow or the GAN diet rich in saturated fat (40%), fructose (22%) and cholesterol (2%) for ≥38 weeks. Metabolic parameters as well as plasma and liver biomarkers were assessed. Liver biopsy histology and transcriptome signatures were compared to samples from human lean individuals and patients diagnosed with NASH. RESULTS: Liver lesions in GAN DIO-NASH mice showed similar morphological characteristics compared to the NASH patient validation set, including macrosteatosis, lobular inflammation, hepatocyte ballooning degeneration and periportal/perisinusoidal fibrosis. Histomorphometric analysis indicated comparable increases in markers of hepatic lipid accumulation, inflammation and collagen deposition in GAN DIO-NASH mice and NASH patient samples. Liver biopsies from GAN DIO-NASH mice and NASH patients showed comparable dynamics in several gene expression pathways involved in NASH pathogenesis. Consistent with the clinical features of NASH, GAN DIO-NASH mice demonstrated key components of the metabolic syndrome, including obesity and impaired glucose tolerance. CONCLUSIONS: The GAN DIO-NASH mouse model demonstrates good clinical translatability with respect to the histopathological, transcriptional and metabolic aspects of the human disease, highlighting the suitability of the GAN DIO-NASH mouse model for identifying therapeutic targets and characterizing novel drug therapies for NASH.


Subject(s)
Non-alcoholic Fatty Liver Disease , Animals , Diet, High-Fat/adverse effects , Disease Models, Animal , Humans , Liver , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity/complications
6.
Sci Rep ; 10(1): 1148, 2020 01 24.
Article in English | MEDLINE | ID: mdl-31980690

ABSTRACT

To improve the understanding of the complex biological processes underlying the development of non-alcoholic steatohepatitis (NASH), a multi-omics approach combining bulk RNA-sequencing based transcriptomics, quantitative proteomics and single-cell RNA-sequencing was used to characterize tissue biopsies from histologically validated diet-induced obese (DIO) NASH mice compared to chow-fed controls. Bulk RNA-sequencing and proteomics showed a clear distinction between phenotypes and a good correspondence between mRNA and protein level regulations, apart from specific regulatory events discovered by each technology. Transcriptomics-based gene set enrichment analysis revealed changes associated with key clinical manifestations of NASH, including impaired lipid metabolism, increased extracellular matrix formation/remodeling and pro-inflammatory responses, whereas proteomics-based gene set enrichment analysis pinpointed metabolic pathway perturbations. Integration with single-cell RNA-sequencing data identified key regulated cell types involved in development of NASH demonstrating the cellular heterogeneity and complexity of NASH pathogenesis.


Subject(s)
Diet, High-Fat/adverse effects , Liver/metabolism , Non-alcoholic Fatty Liver Disease/etiology , Obesity/etiology , Proteomics/methods , Transcriptome , Animals , Chromatography, Liquid , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Obesity/genetics , RNA/genetics , RNA/isolation & purification , Sequence Alignment , Sequence Analysis, RNA , Single-Cell Analysis , Tandem Mass Spectrometry
7.
Genome Res ; 30(1): 127-137, 2020 01.
Article in English | MEDLINE | ID: mdl-31831592

ABSTRACT

Bone marrow-derived mesenchymal stem cells (MSCs) differentiate into osteoblasts upon stimulation by signals present in their niche. Because the global signaling cascades involved in the early phases of MSCs osteoblast (OB) differentiation are not well-defined, we used quantitative mass spectrometry to delineate changes in human MSCs proteome and phosphoproteome during the first 24 h of their OB lineage commitment. The temporal profiles of 6252 proteins and 15,059 phosphorylation sites suggested at least two distinct signaling waves: one peaking within 30 to 60 min after stimulation and a second upsurge after 24 h. In addition to providing a comprehensive view of the proteome and phosphoproteome dynamics during early MSCs differentiation, our analyses identified a key role of serine/threonine protein kinase D1 (PRKD1) in OB commitment. At the onset of OB differentiation, PRKD1 initiates activation of the pro-osteogenic transcription factor RUNX2 by triggering phosphorylation and nuclear exclusion of the histone deacetylase HDAC7.


Subject(s)
Cell Differentiation , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Osteoblasts/cytology , Osteoblasts/metabolism , Phosphoproteins/metabolism , Proteome , Proteomics , Humans , Phylogeny , Proteomics/methods
8.
Sci Rep ; 9(1): 16161, 2019 11 06.
Article in English | MEDLINE | ID: mdl-31695063

ABSTRACT

The central mechanisms underlying the marked beneficial metabolic effects of bariatric surgery are unclear. Here, we characterized global gene expression in the hypothalamic arcuate nucleus (Arc) in diet-induced obese (DIO) rats following Roux-en-Y gastric bypass (RYGB). 60 days post-RYGB, the Arc was isolated by laser-capture microdissection and global gene expression was assessed by RNA sequencing. RYGB lowered body weight and adiposity as compared to sham-operated DIO rats. Discrete transcriptome changes were observed in the Arc following RYGB, including differential expression of genes associated with inflammation and neuropeptide signaling. RYGB reduced gene expression of glial cell markers, including Gfap, Aif1 and Timp1, confirmed by a lower number of GFAP immunopositive astrocyte profiles in the Arc. Sham-operated weight-matched rats demonstrated a similar glial gene expression signature, suggesting that RYGB and dietary restriction have common effects on hypothalamic gliosis. Considering that RYGB surgery also led to increased orexigenic and decreased anorexigenic gene expression, this may signify increased hunger-associated signaling at the level of the Arc. Hence, induction of counterregulatory molecular mechanisms downstream from the Arc may play an important role in RYGB-induced weight loss.


Subject(s)
Arcuate Nucleus of Hypothalamus/metabolism , Diet, Reducing , Gastric Bypass , Gene Expression Profiling , Gene Expression Regulation , Gliosis/genetics , Adiposity , Animals , Astrocytes/metabolism , Biomarkers , Diet, High-Fat , Eating , Glial Fibrillary Acidic Protein/analysis , Glucagon-Like Peptide 1/blood , Inflammation/genetics , Laser Capture Microdissection , Male , Neuropeptides/biosynthesis , Neuropeptides/genetics , Obesity/etiology , Obesity/surgery , Peptide YY/blood , Rats , Rats, Sprague-Dawley , Sequence Analysis, RNA , Weight Loss
9.
Sci Rep ; 9(1): 9046, 2019 06 21.
Article in English | MEDLINE | ID: mdl-31227742

ABSTRACT

Obeticholic acid (OCA) and elafibranor (ELA) are selective and potent agonists for the farnesoid X receptor (FXR) and dual peroxisome proliferator-activated receptor α/δ (PPAR-α/δ), respectively. Both agents have demonstrated clinical efficacy in nonalcoholic steatohepatitis (NASH). The present study used OCA and ELA to compare the effects of mono- and combination therapies on metabolic and histological endpoints in Lepob/ob mice with established diet-induced and biopsy-confirmed NASH (ob/ob-NASH). ob/ob-NASH mice were fed the AMLN diet high in trans-fat, fructose and cholesterol for 15 weeks, whereafter they received vehicle, OCA (30 mg/kg, PO, QD), ELA (3, 10 mg/kg, PO, QD), or combinations (OCA + ELA) for eight weeks. Within-subject comparisons were performed on histomorphometric changes, including fractional area of liver fat, galectin-3 and Col1a1. OCA and ELA monotherapies improved all quantitative histopathological parameters and OCA + ELA combinations exerted additive effects on metabolic and histological endpoints. In agreement with their different molecular mechanisms of action, OCA and ELA monotherapies elicited distinct hepatic gene expression profiles and their combination led to profound transcriptome changes associated with further improvements in lipid handling and insulin signaling, suppression of immune responses and reduced extracellular matrix formation. In conclusion, these findings provide preclinical proof-of-concept for combined FXR and PPAR-α/δ agonist-based therapies in NASH.


Subject(s)
Chalcones/therapeutic use , Chenodeoxycholic Acid/analogs & derivatives , Liver Cirrhosis/drug therapy , Propionates/therapeutic use , Animals , Biopsy , Chenodeoxycholic Acid/therapeutic use , Disease Models, Animal , Liver Cirrhosis/pathology , Mice , PPAR alpha/metabolism , Proof of Concept Study , Transcription, Genetic
10.
Lipids ; 54(1): 109-115, 2019 01.
Article in English | MEDLINE | ID: mdl-30723896

ABSTRACT

Simple steatosis is the hallmark of nonalcoholic fatty liver disease, with lipid accumulating as either microvesicular or macrovesicular lipid droplets within hepatocytes. The present study used a combination of laser capture microdissection and RNAseq to characterize murine gene expression in nonsteatotic, microsteatotic, and macrosteatotic compartments collected from the same liver. The data indicate that microvesicular steatosis is intermediate to macrovesicular steatosis, showing a widespread and pronounced metabolic gene regulation of lipid export, gluconeogenesis, and de novo lipogenesis. Key enzymes, such as fatty acid synthase and fructose 1,6-bisphosphatase as well as apolipoprotein C-III, were identified to show clear expression differences between the compartments. Furthermore, increased expression of lipid particle formation genes provided a molecular description of the fusion of microsteatotic lipid compartments to produce macrosteatotic cells with a single enlarged lipid droplet.


Subject(s)
Metabolic Networks and Pathways/physiology , Non-alcoholic Fatty Liver Disease/metabolism , Animals , Gluconeogenesis/physiology , Hepatocytes/metabolism , Lipid Metabolism/physiology , Lipogenesis/physiology , Liver/metabolism , Male , Mice , Mice, Inbred C57BL
11.
Am J Physiol Gastrointest Liver Physiol ; 316(4): G462-G472, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30653341

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) represents a spectrum of conditions ranging from simple steatosis (NAFL), over nonalcoholic steatohepatitis (NASH) with or without fibrosis, to cirrhosis with end-stage disease. The hepatic molecular events underlying the development of NAFLD and transition to NASH are poorly understood. The present study aimed to determine hepatic transcriptome dynamics in patients with NAFL or NASH compared with healthy normal-weight and obese individuals. RNA sequencing and quantitative histomorphometry of liver fat, inflammation and fibrosis were performed on liver biopsies obtained from healthy normal-weight ( n = 14) and obese ( n = 12) individuals, NAFL ( n = 15) and NASH ( n = 16) patients. Normal-weight and obese subjects showed normal liver histology and comparable gene expression profiles. Liver transcriptome signatures were largely overlapping in NAFL and NASH patients, however, clearly separated from healthy normal-weight and obese controls. Most marked pathway perturbations identified in both NAFL and NASH were associated with markers of lipid metabolism, immunomodulation, extracellular matrix remodeling, and cell cycle control. Interestingly, NASH patients with positive Sonic hedgehog hepatocyte staining showed distinct transcriptome and histomorphometric changes compared with NAFL. In conclusion, application of immunohistochemical markers of hepatocyte injury may serve as a more objective tool for distinguishing NASH from NAFL, facilitating improved resolution of hepatic molecular changes associated with progression of NAFLD. NEW & NOTEWORTHY Nonalcoholic fatty liver disease (NAFLD) is the most common liver disease in Western countries. NAFLD is associated with the metabolic syndrome and can progress to the more serious form, nonalcoholic steatohepatitis (NASH), and ultimately lead to irreversible liver damage. Using gold standard molecular and histological techniques, this study demonstrates that the currently used diagnostic tools are problematic for differentiating mild NAFLD from NASH and emphasizes the marked need for developing improved histological markers of NAFLD progression.


Subject(s)
Adipose Tissue , Gene Expression Profiling/methods , Inflammation , Liver Cirrhosis , Liver , Non-alcoholic Fatty Liver Disease , Obesity , Adipose Tissue/metabolism , Adipose Tissue/pathology , Body Mass Index , Disease Progression , Female , Humans , Immunohistochemistry , Inflammation/immunology , Inflammation/pathology , Liver/metabolism , Liver/pathology , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Male , Middle Aged , Non-alcoholic Fatty Liver Disease/diagnosis , Non-alcoholic Fatty Liver Disease/metabolism , Obesity/diagnosis , Obesity/metabolism
12.
Dig Dis Sci ; 64(5): 1238-1256, 2019 05.
Article in English | MEDLINE | ID: mdl-30511198

ABSTRACT

BACKGROUND: There is a marked need for improved animal models of nonalcoholic steatohepatitis (NASH) to facilitate the development of more efficacious drug therapies for the disease. METHODS: Here, we investigated the development of fibrotic NASH in male Wistar rats fed a choline-deficient L-amino acid-defined (CDAA) diet with or without cholesterol supplementation for subsequent assessment of drug treatment efficacy in NASH biopsy-confirmed rats. The metabolic profile and liver histopathology were evaluated after 4, 8, and 12 weeks of dieting. Subsequently, rats with biopsy-confirmed NASH were selected for pharmacological intervention with vehicle, elafibranor (30 mg/kg/day) or obeticholic acid (OCA, 30 mg/kg/day) for 5 weeks. RESULTS: The CDAA diet led to marked hepatomegaly and fibrosis already after 4 weeks of feeding, with further progression of collagen deposition and fibrogenesis-associated gene expression during the 12-week feeding period. Cholesterol supplementation enhanced the stimulatory effect of CDAA on gene transcripts associated with fibrogenesis without significantly increasing collagen deposition. Pharmacological intervention with elafibranor, but not OCA, significantly reduced steatohepatitis scores, and fibrosis-associated gene expression, however, was unable to prevent progression in fibrosis scores. CONCLUSION: CDAA-fed rats develop early-onset progressive NASH, which offers the opportunity to probe anti-NASH compounds with potential disease-modifying properties.


Subject(s)
Chalcones/therapeutic use , Chenodeoxycholic Acid/analogs & derivatives , Cholesterol/toxicity , Non-alcoholic Fatty Liver Disease/chemically induced , Non-alcoholic Fatty Liver Disease/drug therapy , Nutrients/deficiency , Propionates/therapeutic use , Animals , Chenodeoxycholic Acid/therapeutic use , Cholesterol/administration & dosage , Disease Progression , Male , Non-alcoholic Fatty Liver Disease/pathology , Rats , Rats, Wistar
13.
Autophagy ; 13(6): 1064-1075, 2017 Jun 03.
Article in English | MEDLINE | ID: mdl-28453381

ABSTRACT

Macroautophagy is regarded as a nonspecific bulk degradation process of cytoplasmic material within the lysosome. However, the process has mainly been studied by nonspecific bulk degradation assays using radiolabeling. In the present study we monitor protein turnover and degradation by global, unbiased approaches relying on quantitative mass spectrometry-based proteomics. Macroautophagy is induced by rapamycin treatment, and by amino acid and glucose starvation in differentially, metabolically labeled cells. Protein dynamics are linked to image-based models of autophagosome turnover. Depending on the inducing stimulus, protein as well as organelle turnover differ. Amino acid starvation-induced macroautophagy leads to selective degradation of proteins important for protein translation. Thus, protein dynamics reflect cellular conditions in the respective treatment indicating stimulus-specific pathways in stress-induced macroautophagy.


Subject(s)
Amino Acids/deficiency , Autophagy , Protein Biosynthesis , Proteolysis , Autophagosomes/metabolism , Humans , Isotope Labeling , MCF-7 Cells
14.
Nat Struct Mol Biol ; 23(6): 608-18, 2016 06.
Article in English | MEDLINE | ID: mdl-27136326

ABSTRACT

A fascinating conundrum in cell signaling is how stimulation of the same receptor tyrosine kinase with distinct ligands generates specific outcomes. To decipher the functional selectivity of EGF and TGF-α, which induce epidermal growth factor receptor (EGFR) degradation and recycling, respectively, we devised an integrated multilayered proteomics approach (IMPA). We analyzed dynamic changes in the receptor interactome, ubiquitinome, phosphoproteome, and late proteome in response to both ligands in human cells by quantitative MS and identified 67 proteins regulated at multiple levels. We identified RAB7 phosphorylation and RCP recruitment to EGFR as switches for EGF and TGF-α outputs, controlling receptor trafficking, signaling duration, proliferation, and migration. By manipulating RCP levels or phosphorylation of RAB7 in EGFR-positive cancer cells, we were able to switch a TGF-α-mediated response to an EGF-like response or vice versa as EGFR trafficking was rerouted. We propose IMPA as an approach to uncover fine-tuned regulatory mechanisms in cell signaling.


Subject(s)
Epidermal Growth Factor/metabolism , ErbB Receptors/metabolism , Transforming Growth Factor alpha/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , HeLa Cells , Humans , Membrane Proteins/metabolism , Phosphorylation , Protein Interaction Maps , Protein Transport , Proteolysis , Proteomics , Signal Transduction , rab GTP-Binding Proteins/metabolism , rab7 GTP-Binding Proteins
15.
J Pept Sci ; 21(2): 85-94, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25521062

ABSTRACT

Neuromedin U (NMU) is a 25 amino acid peptide expressed and secreted in the brain and gastrointestinal tract. Data have shown that peripheral administration of human NMU decreases food intake and body weight and improves glucose tolerance in mice, suggesting that NMU receptors constitute a possible anti-diabetic and anti-obesity drug target. However, the clinical use of native NMU is hampered by a poor pharmacokinetic profile. In the current study, we report in vitro and in vivo data from a series of novel lipidated NMU analogs. In vitro plasma stability studies of native NMU were performed to investigate the proteolytic stability and cleavage sites using LC-MS. Native NMU was found to be rapidly cleaved at the C-terminus between Arg(24) and Asn(25) , followed by cleavage between Arg(16) and Gly(17) . Lipidated NMU analogs were generated using solid-phase peptide synthesis, and in vitro potency was investigated using a human embryonic kidney 293-based inositol phosphate accumulation assay. All lipidated analogs had preserved in vitro activity on both NMU receptors with potency improving as the lipidation site was moved away from the receptor-interacting C-terminal octapeptide segment. In vivo efficacy was assessed in lean mice as reduction in food intake after acute subcutaneous administration of 1, 0.3, 0.1, and 0.03 µmol/kg. These lipidated NMU analogs prolonged the anorectic effect of NMU in a dose-dependent manner. This was likely an effect of improved pharmacokinetic properties because of improved vitro plasma stability. Accordingly, the data demonstrate that lipidated NMU analogs may represent drug candidates for the treatment of obesity.


Subject(s)
Eating/drug effects , Neuropeptides/chemical synthesis , Neuropeptides/pharmacology , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Drug Stability , HEK293 Cells , Humans , Male , Mice , Neuropeptides/blood , Neuropeptides/chemistry , Protein Stability
16.
J Proteome Res ; 13(9): 4192-204, 2014 Sep 05.
Article in English | MEDLINE | ID: mdl-25093938

ABSTRACT

Post-translational modification of proteins with the small polypeptide ubiquitin plays a pivotal role in many cellular processes, altering protein lifespan, location, and function and regulating protein-protein interactions. Ubiquitination exerts its diverse functions through complex mechanisms by formation of different polymeric chains and subsequent recognition of the ubiquitin signal by specific protein interaction domains. Despite some recent advances in the analytical tools for the analysis of ubiquitination by mass spectrometry, there is still a need for additional strategies suitable for investigation of cellular ubiquitination at the proteome level. Here, we present a stable tagged ubiquitin exchange (StUbEx) cellular system in which endogenous ubiquitin is replaced with an epitope-tagged version, thereby allowing specific and efficient affinity purification of ubiquitinated proteins for global analyses of protein ubiquitination. Importantly, the overall level of ubiquitin in the cell remains virtually unchanged, thus avoiding ubiquitination artifacts associated with overexpression. The efficiency and reproducibility of the method were assessed through unbiased analysis of epidermal growth factor (EGF) signaling by quantitative mass spectrometry, covering over 3400 potential ubiquitinated proteins. The StUbEx system is applicable to virtually any cell line and can be readily adapted to any of the ubiquitin-like post-translational modifications.


Subject(s)
Isotope Labeling/methods , Proteomics/methods , Ubiquitin/chemistry , Ubiquitin/metabolism , Chromatography, Affinity/methods , Databases, Protein , HeLa Cells , Histidine , Humans , Oligopeptides , Recombinant Fusion Proteins , Reproducibility of Results , Ubiquitination
17.
PLoS One ; 9(8): e104240, 2014.
Article in English | MEDLINE | ID: mdl-25147952

ABSTRACT

In adaptive immune responses, T-cell receptor (TCR) signaling impacts multiple cellular processes and results in T-cell differentiation, proliferation, and cytokine production. Although individual protein-protein interactions and phosphorylation events have been studied extensively, we lack a systems-level understanding of how these components cooperate to control signaling dynamics, especially during the crucial first seconds of stimulation. Here, we used quantitative proteomics to characterize reshaping of the T-cell phosphoproteome in response to TCR/CD28 co-stimulation, and found that diverse dynamic patterns emerge within seconds. We detected phosphorylation dynamics as early as 5 s and observed widespread regulation of key TCR signaling proteins by 30 s. Development of a computational model pointed to the presence of novel regulatory mechanisms controlling phosphorylation of sites with central roles in TCR signaling. The model was used to generate predictions suggesting unexpected roles for the phosphatase PTPN6 (SHP-1) and shortcut recruitment of the actin regulator WAS. Predictions were validated experimentally. This integration of proteomics and modeling illustrates a novel, generalizable framework for solidifying quantitative understanding of a signaling network and for elucidating missing links.


Subject(s)
Receptors, Antigen, T-Cell/metabolism , Signal Transduction , T-Lymphocytes/metabolism , Cell Line , Humans , Models, Biological , Phosphoproteins/metabolism , Phosphorylation , Protein Interaction Maps , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Proteome , Proteomics/methods , Tyrosine/metabolism , Wiskott-Aldrich Syndrome Protein Family/metabolism
18.
Nat Commun ; 4: 2789, 2013.
Article in English | MEDLINE | ID: mdl-24240771

ABSTRACT

Mitophagy, the autophagic degradation of mitochondria, is an important housekeeping function in eukaryotic cells, and defects in mitophagy correlate with ageing phenomena and with several neurodegenerative disorders. A central mechanistic question regarding mitophagy is whether mitochondria are consumed en masse, or whether an active process segregates defective molecules from functional ones within the mitochondrial network, thus allowing a more efficient culling mechanism. Here we combine a proteomic study with a molecular genetics and cell biology approach to determine whether such a segregation process occurs in yeast mitochondria. We find that different mitochondrial matrix proteins undergo mitophagic degradation at distinctly different rates, supporting the active segregation hypothesis. These differential degradation rates depend on mitochondrial dynamics, suggesting a mechanism coupling weak physical segregation with mitochondrial dynamics to achieve a distillation-like effect. In agreement, the rates of mitophagic degradation strongly correlate with the degree of physical segregation of specific matrix proteins.


Subject(s)
Mitochondrial Dynamics , Mitochondrial Proteins/metabolism , Mitophagy , Proteomics , Schizosaccharomyces
19.
Mol Cell ; 51(6): 707-22, 2013 Sep 26.
Article in English | MEDLINE | ID: mdl-24011590

ABSTRACT

The stimulation of fibroblast growth factor receptors (FGFRs) with distinct FGF ligands generates specific cellular responses. However, the mechanisms underlying this paradigm have remained elusive. Here, we show that FGF-7 stimulation leads to FGFR2b degradation and, ultimately, cell proliferation, whereas FGF-10 promotes receptor recycling and cell migration. By combining mass-spectrometry-based quantitative proteomics with fluorescence microscopy and biochemical methods, we find that FGF-10 specifically induces the rapid phosphorylation of tyrosine (Y) 734 on FGFR2b, which leads to PI3K and SH3BP4 recruitment. This complex is crucial for FGFR2b recycling and responses, given that FGF-10 stimulation of either FGFR2b_Y734F mutant- or SH3BP4-depleted cells switches the receptor endocytic route to degradation, resulting in decreased breast cancer cell migration and the inhibition of epithelial branching in mouse lung explants. Altogether, these results identify an intriguing ligand-dependent mechanism for the control of receptor fate and cellular outputs that may explain the pathogenic role of deregulated FGFR2b, thus offering therapeutic opportunities.


Subject(s)
Fibroblast Growth Factor 10/metabolism , Fibroblast Growth Factor 7/metabolism , Proteomics , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Animals , Cell Movement , Ligands , Mice , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proteolysis , Tyrosine/metabolism
20.
Mol Syst Biol ; 9: 657, 2013 Apr 16.
Article in English | MEDLINE | ID: mdl-23591773

ABSTRACT

The mammalian cellular microenvironment is shaped by soluble factors and structural components, the extracellular matrix, providing physical support, regulating adhesion and signalling. A global, quantitative mass spectrometry strategy, combined with bioinformatics data processing, was developed to assess proteome differences in the microenvironment of primary human fibroblasts. We studied secreted proteins of fibroblasts from normal and pathologically altered skin and their post-translational modifications. The influence of collagen VII, an important structural component, which is lost in genetic skin fragility, was used as model. Loss of collagen VII had a global impact on the cellular microenvironment and was associated with proteome alterations highly relevant for disease pathogenesis including decrease in basement membrane components, increase in dermal matrix proteins, TGF-ß and metalloproteases, but not higher protease activity. The definition of the proteome of fibroblast microenvironment and its plasticity in health and disease identified novel disease mechanisms and potential targets of intervention.


Subject(s)
Basement Membrane/metabolism , Cellular Microenvironment/genetics , Collagen Type VII/genetics , Dermis/metabolism , Epidermolysis Bullosa Dystrophica/genetics , Extracellular Matrix/genetics , Fibroblasts/metabolism , Basement Membrane/pathology , Case-Control Studies , Cell Communication , Collagen Type VII/deficiency , Dermis/pathology , Epidermolysis Bullosa Dystrophica/metabolism , Epidermolysis Bullosa Dystrophica/pathology , Extracellular Matrix/pathology , Female , Fibroblasts/pathology , Gene Expression , Humans , Infant , Infant, Newborn , Male , Metalloproteases/genetics , Metalloproteases/metabolism , Primary Cell Culture , Protein Processing, Post-Translational , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...