Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 85
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nature ; 626(8001): 1116-1124, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38355802

ABSTRACT

Transposable elements (TEs) are a major constituent of human genes, occupying approximately half of the intronic space. During pre-messenger RNA synthesis, intronic TEs are transcribed along with their host genes but rarely contribute to the final mRNA product because they are spliced out together with the intron and rapidly degraded. Paradoxically, TEs are an abundant source of RNA-processing signals through which they can create new introns1, and also functional2 or non-functional chimeric transcripts3. The rarity of these events implies the existence of a resilient splicing code that is able to suppress TE exonization without compromising host pre-mRNA processing. Here we show that SAFB proteins protect genome integrity by preventing retrotransposition of L1 elements while maintaining splicing integrity, via prevention of the exonization of previously integrated TEs. This unique dual role is possible because of L1's conserved adenosine-rich coding sequences that are bound by SAFB proteins. The suppressive activity of SAFB extends to tissue-specific, giant protein-coding cassette exons, nested genes and Tigger DNA transposons. Moreover, SAFB also suppresses LTR/ERV elements in species in which they are still active, such as mice and flies. A significant subset of splicing events suppressed by SAFB in somatic cells are activated in the testis, coinciding with low SAFB expression in postmeiotic spermatids. Reminiscent of the division of labour between innate and adaptive immune systems that fight external pathogens, our results uncover SAFB proteins as an RNA-based, pattern-guided, non-adaptive defence system against TEs in the soma, complementing the RNA-based, adaptive Piwi-interacting RNA pathway of the germline.


Subject(s)
DNA Transposable Elements , Introns , RNA Precursors , RNA Splicing , RNA, Messenger , Animals , Humans , Male , Mice , DNA Transposable Elements/genetics , Drosophila melanogaster/genetics , Exons/genetics , Genome/genetics , Introns/genetics , Organ Specificity/genetics , Piwi-Interacting RNA/genetics , Piwi-Interacting RNA/metabolism , RNA Precursors/genetics , RNA Precursors/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Spermatids/cytology , Spermatids/metabolism , RNA Splicing/genetics , Testis , Meiosis
2.
Mol Cell ; 81(17): 3589-3603.e13, 2021 09 02.
Article in English | MEDLINE | ID: mdl-34324863

ABSTRACT

Transcription elongation has emerged as a regulatory hub in gene expression of metazoans. A major control point occurs during early elongation before RNA polymerase II (Pol II) is released into productive elongation. Prior research has linked BRD4 with transcription elongation. Here, we use rapid BET protein and BRD4-selective degradation along with quantitative genome-wide approaches to investigate direct functions of BRD4 in Pol II transcription regulation. Notably, as an immediate consequence of acute BRD4 loss, promoter-proximal pause release is impaired, and transcriptionally engaged Pol II past this checkpoint undergoes readthrough transcription. An integrated proteome-wide analysis uncovers elongation and 3'-RNA processing factors as core BRD4 interactors. BRD4 ablation disrupts the recruitment of general 3'-RNA processing factors at the 5'-control region, which correlates with RNA cleavage and termination defects. These studies, performed in human cells, reveal a BRD4-mediated checkpoint and begin to establish a molecular link between 5'-elongation control and 3'-RNA processing.


Subject(s)
Cell Cycle Proteins/physiology , Nuclear Proteins/physiology , Transcription Elongation, Genetic/physiology , Transcription Factors/physiology , Animals , Cell Cycle Proteins/metabolism , Cell Line , Gene Expression , Histones/metabolism , Humans , Mice , Nuclear Proteins/metabolism , Promoter Regions, Genetic/genetics , RNA Polymerase II/metabolism , RNA Polymerase II/physiology , Transcription Factors/metabolism , Transcription Termination, Genetic/physiology , Transcription, Genetic/genetics , Transcription, Genetic/physiology
3.
EMBO Rep ; 25(4): 1814-1834, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38413733

ABSTRACT

Stress granules are an integral part of the stress response that are formed from non-translating mRNAs aggregated with proteins. While much is known about stress granules, the factors that drive their mRNA localization are incompletely described. Modification of mRNA can alter the properties of the nucleobases and affect processes such as translation, splicing and localization of individual transcripts. Here, we show that the RNA modification N4-acetylcytidine (ac4C) on mRNA associates with transcripts enriched in stress granules and that stress granule localized transcripts with ac4C are specifically translationally regulated. We also show that ac4C on mRNA can mediate localization of the protein NOP58 to stress granules. Our results suggest that acetylation of mRNA regulates localization of both stress-sensitive transcripts and RNA-binding proteins to stress granules and adds to our understanding of the molecular mechanisms responsible for stress granule formation.


Subject(s)
Cytidine , Cytidine/analogs & derivatives , Stress Granules , RNA, Messenger/genetics , RNA, Messenger/metabolism , Cytidine/genetics , Cytidine/metabolism , RNA-Binding Proteins/metabolism
4.
Mol Syst Biol ; 20(7): 767-798, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38755290

ABSTRACT

Static gene expression programs have been extensively characterized in stem cells and mature human cells. However, the dynamics of RNA isoform changes upon cell-state-transitions during cell differentiation, the determinants and functional consequences have largely remained unclear. Here, we established an improved model for human neurogenesis in vitro that is amenable for systems-wide analyses of gene expression. Our multi-omics analysis reveals that the pronounced alterations in cell morphology correlate strongly with widespread changes in RNA isoform expression. Our approach identifies thousands of new RNA isoforms that are expressed at distinct differentiation stages. RNA isoforms mainly arise from exon skipping and the alternative usage of transcription start and polyadenylation sites during human neurogenesis. The transcript isoform changes can remodel the identity and functions of protein isoforms. Finally, our study identifies a set of RNA binding proteins as a potential determinant of differentiation stage-specific global isoform changes. This work supports the view of regulated isoform changes that underlie state-transitions during neurogenesis.


Subject(s)
Cell Differentiation , Neurogenesis , Neurons , RNA Isoforms , Humans , Neurogenesis/genetics , Cell Differentiation/genetics , RNA Isoforms/genetics , RNA Isoforms/metabolism , Neurons/metabolism , Neurons/cytology , Alternative Splicing , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Protein Isoforms/metabolism , Protein Isoforms/genetics , Exons/genetics
5.
Genet Med ; 24(9): 1927-1940, 2022 09.
Article in English | MEDLINE | ID: mdl-35670808

ABSTRACT

PURPOSE: In this study we aimed to identify the molecular genetic cause of a progressive multisystem disease with prominent lipodystrophy. METHODS: In total, 5 affected individuals were investigated using exome sequencing. Dermal fibroblasts were characterized using RNA sequencing, proteomics, immunoblotting, immunostaining, and electron microscopy. Subcellular localization and rescue studies were performed. RESULTS: We identified a lipodystrophy phenotype with a typical facial appearance, corneal clouding, achalasia, progressive hearing loss, and variable severity. Although 3 individuals showed stunted growth, intellectual disability, and died within the first decade of life (A1, A2, and A3), 2 are adults with normal intellectual development (A4 and A5). All individuals harbored an identical homozygous nonsense variant affecting the retention and splicing complex component BUD13. The nucleotide substitution caused alternative splicing of BUD13 leading to a stable truncated protein whose expression positively correlated with disease expression and life expectancy. In dermal fibroblasts, we found elevated intron retention, a global reduction of spliceosomal proteins, and nuclei with multiple invaginations, which were more pronounced in A1, A2, and A3. Overexpression of both BUD13 isoforms normalized the nuclear morphology. CONCLUSION: Our results define a hitherto unknown syndrome and show that the alternative splice product converts a loss-of-function into a hypomorphic allele, thereby probably determining the severity of the disease and the survival of affected individuals.


Subject(s)
Alternative Splicing , Lipodystrophy , RNA-Binding Proteins/genetics , Child , Developmental Disabilities/genetics , Humans , Introns , Lipodystrophy/genetics , RNA Splicing
6.
Hepatology ; 73(2): 795-810, 2021 02.
Article in English | MEDLINE | ID: mdl-32286709

ABSTRACT

BACKGROUND AND AIMS: Zone-dependent differences in expression of metabolic enzymes along the portocentral axis of the acinus are a long-known feature of liver metabolism. A prominent example is the preferential localization of the enzyme, glutamine synthetase, in pericentral hepatocytes, where it converts potentially toxic ammonia to the valuable amino acid, glutamine. However, with the exception of a few key regulatory enzymes, a comprehensive and quantitative assessment of zonal differences in the abundance of metabolic enzymes and, much more important, an estimation of the associated functional differences between portal and central hepatocytes is missing thus far. APPROACH AND RESULTS: We addressed this problem by establishing a method for the separation of periportal and pericentral hepatocytes that yields sufficiently pure fractions of both cell populations. Quantitative shotgun proteomics identified hundreds of differentially expressed enzymes in the two cell populations. We used zone-specific proteomics data for scaling of the maximal activities to generate portal and central instantiations of a comprehensive kinetic model of central hepatic metabolism (Hepatokin1). CONCLUSIONS: The model simulations revealed significant portal-to-central differences in almost all metabolic pathways involving carbohydrates, fatty acids, amino acids, and detoxification.


Subject(s)
Hepatocytes/enzymology , Liver/metabolism , Amino Acids/metabolism , Ammonia/metabolism , Animals , Arginase/metabolism , Carbohydrate Metabolism , Cells, Cultured , Fatty Acids , Glucokinase/metabolism , Glutaminase/metabolism , L-Lactate Dehydrogenase/metabolism , Liver/cytology , Male , Mice , Models, Animal , Primary Cell Culture , Proteomics , Spatial Analysis
7.
J Pathol ; 255(3): 270-284, 2021 11.
Article in English | MEDLINE | ID: mdl-34309874

ABSTRACT

Activation of the mechanistic target of rapamycin (mTOR) pathway is frequently found in cancer, but mTOR inhibitors have thus far failed to demonstrate significant antiproliferative efficacy in the majority of cancer types. Besides cancer cell-intrinsic resistance mechanisms, it is conceivable that mTOR inhibitors impact on non-malignant host cells in a manner that ultimately supports resistance of cancer cells. Against this background, we sought to analyze the functional consequences of mTOR inhibition in hepatocytes for the growth of metastatic colon cancer. To this end, we established liver epithelial cell (LEC)-specific knockout (KO) of mTOR (mTORLEC ) mice. We used these mice to characterize the growth of colorectal liver metastases with or without partial hepatectomy to model different clinical settings. Although the LEC-specific loss of mTOR remained without effect on metastasis growth in intact liver, partial liver resection resulted in the formation of larger metastases in mTORLEC mice compared with wildtype controls. This was accompanied by significantly enhanced inflammatory activity in LEC-specific mTOR KO livers after partial liver resection. Analysis of NF-ĸB target gene expression and immunohistochemistry of p65 displayed a significant activation of NF-ĸB in mTORLEC mice, suggesting a functional importance of this pathway for the observed inflammatory phenotype. Taken together, we show an unexpected acceleration of liver metastases upon deletion of mTOR in LECs. Our results support the notion that non-malignant host cells can contribute to resistance against mTOR inhibitors and encourage testing whether anti-inflammatory drugs are able to improve the efficacy of mTOR inhibitors for cancer therapy. © 2021 The Authors. The Journal of Pathology published by John Wiley & Sons, Ltd. on behalf of The Pathological Society of Great Britain and Ireland.


Subject(s)
Colonic Neoplasms/pathology , Hepatocytes/metabolism , Liver Neoplasms/secondary , TOR Serine-Threonine Kinases/metabolism , Animals , Liver Neoplasms/metabolism , Mice , Mice, Knockout , Neoplasm Metastasis/pathology
8.
Int J Mol Sci ; 23(19)2022 Sep 21.
Article in English | MEDLINE | ID: mdl-36232372

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) is the most common chronic liver disease in children and is associated with overweight and insulin resistance (IR). Almost nothing is known about in vivo alterations of liver metabolism in NAFLD, especially in the early stages of non-alcoholic steatohepatitis (NASH). Here, we used a complex mathematical model of liver metabolism to quantify the central hepatic metabolic functions of 71 children with biopsy-proven NAFLD. For each patient, a personalized model variant was generated based on enzyme abundances determined by mass spectroscopy. Our analysis revealed statistically significant alterations in the hepatic carbohydrate, lipid, and ammonia metabolism, which increased with the degree of obesity and severity of NAFLD. Histologic features of NASH and IR displayed opposing associations with changes in carbohydrate and lipid metabolism but synergistically decreased urea synthesis in favor of the increased release of glutamine, a driver of liver fibrosis. Taken together, our study reveals already significant alterations in the NASH liver of pediatric patients, which, however, are differently modulated by the simultaneous presence of IR.


Subject(s)
Insulin Resistance , Non-alcoholic Fatty Liver Disease , Ammonia , Carbohydrates , Child , Glutamine , Humans , Lipids , Liver/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Prevalence , Urea
9.
Arch Toxicol ; 94(2): 401-415, 2020 02.
Article in English | MEDLINE | ID: mdl-32020249

ABSTRACT

The principle of dynamic liver function breath tests is founded on the administration of a 13C-labeled drug and subsequent monitoring of 13CO2 in the breath, quantified as time series delta over natural baseline 13CO2 (DOB) liberated from the drug during hepatic CYP-dependent detoxification. One confounding factor limiting the diagnostic value of such tests is that only a fraction of the liberated 13CO2 is immediately exhaled, while another fraction is taken up by body compartments from which it returns with delay to the plasma. The aims of this study were to establish a novel variant of the methacetin-based breath test LiMAx that allows to estimate and to eliminate the confounding effect of systemic 13CO2 distribution on the DOB curve and thus enables a more reliable assessment of the hepatic detoxification capacity compared with the conventional LiMAx test. We designed a new test variant (named "2DOB") consisting of two consecutive phases. Phase 1 is initiated by the intravenous administration of 13C-bicarbonate. Phase 2 starts about 30 min later with the intravenous administration of the 13C-labelled test drug. Using compartment modelling, the resulting 2-phasic DOB curve yields the rate constants for the irreversible elimination and the reversible exchange of plasma 13CO2 with body compartments (phase 1) and for the detoxification and exchange of the drug with body compartments (phase 2). We carried out the 2DOB test with the test drug 13C-methacetin in 16 subjects with chronic liver pathologies and 22 normal subjects, who also underwent the conventional LiMAx test. Individual differences in the systemic CO2 kinetics can lead to deviations up to a factor of 2 in the maximum of DOB curves (coefficient of variation CV ≈ 0.2) which, in particular, may hamper the discrimination between subjects with normal or mildly impaired detoxification capacities. The novel test revealed that a significant portion of the drug is not immediately metabolized, but transiently taken up into a storage compartment. Intriguingly, not only the hepatic detoxification rate but also the storage capacity of the drug, turned out to be indicative for a normal liver function. We thus used both parameters to define a scoring function which yielded an excellent disease classification (AUC = 0.95) and a high correlation with the MELD score (RSpearman = 0.92). The novel test variant 2DOB promises a significant improvement in the assessment of impaired hepatic detoxification capacity. The suitability of the test for the reliable characterization of the natural history of chronic liver diseases (fatty liver-fibrosis-cirrhosis) has to be assessed in further studies.


Subject(s)
Breath Tests/methods , Carbon Dioxide/metabolism , Liver Diseases/physiopathology , Liver Function Tests/methods , Acetamides/administration & dosage , Acetamides/blood , Acetaminophen/blood , Administration, Oral , Adult , Age Factors , Carbon Isotopes/analysis , Carbon Isotopes/blood , Case-Control Studies , Drug Monitoring , Female , Humans , Liver Diseases/diagnosis , Male , Middle Aged , Models, Biological
10.
Neurobiol Dis ; 132: 104559, 2019 12.
Article in English | MEDLINE | ID: mdl-31376479

ABSTRACT

Spinocerebellar ataxia type 2 (SCA2) is an autosomal dominant neurodegenerative disorder caused by CAG-expansion mutations in the ATXN2 gene, mainly affecting motor neurons in the spinal cord and Purkinje neurons in the cerebellum. While the large expansions were shown to cause SCA2, the intermediate length expansions lead to increased risk for several atrophic processes including amyotrophic lateral sclerosis and Parkinson variants, e.g. progressive supranuclear palsy. Intense efforts to pioneer a neuroprotective therapy for SCA2 require longitudinal monitoring of patients and identification of crucial molecular pathways. The ataxin-2 (ATXN2) protein is mainly involved in RNA translation control and regulation of nutrient metabolism during stress periods. The preferential mRNA targets of ATXN2 are yet to be determined. In order to understand the molecular disease mechanism throughout different prognostic stages, we generated an Atxn2-CAG100-knock-in (KIN) mouse model of SCA2 with intact murine ATXN2 expression regulation. Its characterization revealed somatic mosaicism of the expansion, with shortened lifespan, a progressive spatio-temporal pattern of pathology with subsequent phenotypes, and anomalies of brain metabolites such as N-acetylaspartate (NAA), all of which mirror faithfully the findings in SCA2 patients. Novel molecular analyses from stages before the onset of motor deficits revealed a strong selective effect of ATXN2 on Nat8l mRNA which encodes the enzyme responsible for NAA synthesis. This metabolite is a prominent energy store of the brain and a well-established marker for neuronal health. Overall, we present a novel authentic rodent model of SCA2, where in vivo magnetic resonance imaging was feasible to monitor progression and where the definition of earliest transcriptional abnormalities was possible. We believe that this model will not only reveal crucial insights regarding the pathomechanism of SCA2 and other ATXN2-associated disorders, but will also aid in developing gene-targeted therapies and disease prevention.


Subject(s)
Acetyltransferases/genetics , Aspartic Acid/analogs & derivatives , Ataxin-2/genetics , Gene Knock-In Techniques/methods , Spinocerebellar Ataxias/genetics , Trinucleotide Repeats/genetics , Acetyltransferases/biosynthesis , Animals , Aspartic Acid/genetics , Aspartic Acid/metabolism , Ataxin-2/biosynthesis , Brain/metabolism , Brain/pathology , Female , Male , Mice , Mice, Transgenic , Spinocerebellar Ataxias/metabolism , Spinocerebellar Ataxias/pathology
11.
Am J Hum Genet ; 98(3): 473-489, 2016 Mar 03.
Article in English | MEDLINE | ID: mdl-26924529

ABSTRACT

Transcriptional signal cointegrators associate with transcription factors or nuclear receptors and coregulate tissue-specific gene transcription. We report on recessive loss-of-function mutations in two genes (TRIP4 and ASCC1) that encode subunits of the nuclear activating signal cointegrator 1 (ASC-1) complex. We used autozygosity mapping and whole-exome sequencing to search for pathogenic mutations in four families. Affected individuals presented with prenatal-onset spinal muscular atrophy (SMA), multiple congenital contractures (arthrogryposis multiplex congenita), respiratory distress, and congenital bone fractures. We identified homozygous and compound-heterozygous nonsense and frameshift TRIP4 and ASCC1 mutations that led to a truncation or the entire absence of the respective proteins and cosegregated with the disease phenotype. Trip4 and Ascc1 have identical expression patterns in 17.5-day-old mouse embryos with high expression levels in the spinal cord, brain, paraspinal ganglia, thyroid, and submandibular glands. Antisense morpholino-mediated knockdown of either trip4 or ascc1 in zebrafish disrupted the highly patterned and coordinated process of α-motoneuron outgrowth and formation of myotomes and neuromuscular junctions and led to a swimming defect in the larvae. Immunoprecipitation of the ASC-1 complex consistently copurified cysteine and glycine rich protein 1 (CSRP1), a transcriptional cofactor, which is known to be involved in spinal cord regeneration upon injury in adult zebrafish. ASCC1 mutant fibroblasts downregulated genes associated with neurogenesis, neuronal migration, and pathfinding (SERPINF1, DAB1, SEMA3D, SEMA3A), as well as with bone development (TNFRSF11B, RASSF2, STC1). Our findings indicate that the dysfunction of a transcriptional coactivator complex can result in a clinical syndrome affecting the neuromuscular system.


Subject(s)
Fractures, Bone/genetics , Gene Expression Regulation, Developmental , Muscular Atrophy, Spinal/genetics , Transcription Factors/genetics , Amino Acid Sequence , Animals , Arthrogryposis/diagnosis , Arthrogryposis/genetics , Carrier Proteins/genetics , Cells, Cultured , Fibroblasts/cytology , Fibroblasts/metabolism , Fractures, Bone/diagnosis , Gene Expression Profiling , Homozygote , Humans , LIM Domain Proteins/genetics , Mice , Molecular Sequence Data , Muscular Atrophy, Spinal/diagnosis , Mutation , Nuclear Proteins/genetics , Pedigree , Phenotype , Zebrafish , Zebrafish Proteins/genetics
12.
Ann Surg ; 270(5): 783-790, 2019 11.
Article in English | MEDLINE | ID: mdl-31592808

ABSTRACT

OBJECTIVE: The aim of this study was to determine the predictive value of machine perfusate analysis on graft outcome. BACKGROUND: Ex situ machine perfusion (MP) is gaining increasing interest to potentially repair injured organs and to assess organ function. In the field of liver transplantation, however, no studies exist on reliable prediction of graft function during MP. METHODS: We have used hypothermic oxygenated perfusion (HOPE) for donation after circulatory death (DCD) or extended criteria donation after brain death (DBD) human liver grafts during the last 7 years. Our series includes 100 HOPE-treated liver-transplanted patients with an overall tumor-censored 5-year graft survival of 89%. We monitored 54 livers during HOPE in terms of fluorometric analysis of released mitochondrial flavin (flavin mononucleotide, FMN) in the machine perfusate. RESULTS: Real-time optical measurement of mitochondrial FMN release in machine perfusates of livers disclosed a strong correlation with lactate clearance and coagulation factors at day 1 and 2 after transplantation. Receiver-operating characteristic curve analysis revealed an area under the curve (AUROC) of 0.79 [95% confidence interval (CI), 0.62-0.97] for severe allograft dysfunction and for early graft loss (AUROC 0.93, 95% CI, 0.84-1.0). CONCLUSIONS: Assessment of flavin, a marker of mitochondrial complex I injury, in the perfusate provides a fast prediction of liver graft function and loss during ex situ MP before implantation. This finding may have high clinical relevance, as liver grafts from extended DBD or DCD donors carry considerable risks for recipients. On-line estimation of outcome before implantation would therefore substantially increase safe utilization of liver grafts.


Subject(s)
Hypothermia, Induced/methods , Liver Transplantation/methods , Organ Preservation/methods , Oxygen/administration & dosage , Postoperative Complications/mortality , Cohort Studies , Female , Graft Rejection , Graft Survival , Humans , Kaplan-Meier Estimate , Liver Transplantation/adverse effects , Male , Middle Aged , Perfusion/methods , Postoperative Complications/physiopathology , Predictive Value of Tests , Preoperative Care/methods , Retrospective Studies , Risk Assessment , Survival Analysis , Tissue Donors/statistics & numerical data , Transplant Recipients/statistics & numerical data , Treatment Outcome
13.
Liver Int ; 39(3): 540-556, 2019 03.
Article in English | MEDLINE | ID: mdl-30444569

ABSTRACT

BACKGROUND AND AIMS: Non-alcoholic fatty liver disease (NAFLD) is the most common chronic liver disease in children and adolescents today. In comparison with adult disease, paediatric NAFLD may show a periportal localization, which is associated with advanced fibrosis. This study aimed to assess the role of genetic risk variants for histological disease pattern and severity in childhood NAFLD. METHODS: We studied 14 single nucleotide polymorphisms (SNP) in a cohort of 70 adolescents with biopsy-proven NAFLD. Genotype was compared to an adult control cohort (n = 200) and analysed in relation to histological disease severity and liver tissue proteomics. RESULTS: Three of the 14 SNPs were significantly associated with paediatric NAFLD after FDR adjustment, rs738409 (PNPLA3, P = 2.80 × 10-06 ), rs1044498 (ENPP1, P = 0.0091) and rs780094 (GCKR, P = 0.0281). The severity of steatosis was critically associated with rs738409 (OR=3.25; 95% CI: 1.72-6.52, FDR-adjusted P = 0.0070). The strongest variants associated with severity of fibrosis were rs1260326, rs780094 (both GCKR) and rs659366 (UCP2). PNPLA3 was associated with a portal pattern of steatosis, inflammation and fibrosis. Proteome profiling revealed decreasing levels of GCKR protein with increasing carriage of the rs1260326/rs780094 minor alleles and downregulation of the retinol pathway in rs738409 G/G carriers. Computational metabolic modelling highlighted functional relevance of PNPLA3, GCKR and UCP2 for NAFLD development. CONCLUSIONS: This study provides evidence for the role of PNPLA3 as a determinant of portal NAFLD localization and severity of portal fibrosis in children and adolescents, the risk variant being associated with an impaired hepatic retinol metabolism.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Lipase/genetics , Liver Cirrhosis/genetics , Membrane Proteins/genetics , Non-alcoholic Fatty Liver Disease/genetics , Polymorphism, Single Nucleotide , Uncoupling Protein 1/genetics , Adolescent , Age Factors , Case-Control Studies , Child , Disease Progression , Female , Genetic Predisposition to Disease , Humans , Liver/enzymology , Liver Cirrhosis/diagnosis , Liver Cirrhosis/enzymology , Male , Non-alcoholic Fatty Liver Disease/diagnosis , Non-alcoholic Fatty Liver Disease/enzymology , Phenotype , Risk Assessment , Risk Factors , Severity of Illness Index , Time Factors , Vitamin A/metabolism
14.
Int J Mol Sci ; 20(15)2019 Jul 26.
Article in English | MEDLINE | ID: mdl-31357507

ABSTRACT

A significantly increased level of the reactive oxygen species (ROS) scavenger glutathione (GSH) has been identified as a hallmark of renal cell carcinoma (RCC). The proposed mechanism for increased GSH levels is to counteract damaging ROS to sustain the viability and growth of the malignancy. Here, we review the current knowledge about the three main RCC subtypes, namely clear cell RCC (ccRCC), papillary RCC (pRCC), and chromophobe RCC (chRCC), at the genetic, transcript, protein, and metabolite level and highlight their mutual influence on GSH metabolism. A further discussion addresses the question of how the manipulation of GSH levels can be exploited as a potential treatment strategy for RCC.


Subject(s)
Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Glutathione/metabolism , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Animals , Biomarkers, Tumor , Carcinoma, Renal Cell/etiology , Carcinoma, Renal Cell/therapy , Disease Models, Animal , Disease Progression , Humans , Kidney Neoplasms/etiology , Kidney Neoplasms/therapy , Metabolic Networks and Pathways , Molecular Targeted Therapy , Oxidative Stress , Reactive Oxygen Species , Tumor Microenvironment/immunology
15.
Int J Mol Sci ; 20(23)2019 Nov 21.
Article in English | MEDLINE | ID: mdl-31766565

ABSTRACT

Ataxin-2 (human gene symbol ATXN2) acts during stress responses, modulating mRNA translation and nutrient metabolism. Ataxin-2 knockout mice exhibit progressive obesity, dyslipidemia, and insulin resistance. Conversely, the progressive ATXN2 gain of function due to the fact of polyglutamine (polyQ) expansions leads to a dominantly inherited neurodegenerative process named spinocerebellar ataxia type 2 (SCA2) with early adipose tissue loss and late muscle atrophy. We tried to understand lipid dysregulation in a SCA2 patient brain and in an authentic mouse model. Thin layer chromatography of a patient cerebellum was compared to the lipid metabolome of Atxn2-CAG100-Knockin (KIN) mouse spinocerebellar tissue. The human pathology caused deficits of sulfatide, galactosylceramide, cholesterol, C22/24-sphingomyelin, and gangliosides GM1a/GD1b despite quite normal levels of C18-sphingomyelin. Cerebellum and spinal cord from the KIN mouse showed a consistent decrease of various ceramides with a significant elevation of sphingosine in the more severely affected spinal cord. Deficiency of C24/26-sphingomyelins contrasted with excess C18/20-sphingomyelin. Spinocerebellar expression profiling revealed consistent reductions of CERS protein isoforms, Sptlc2 and Smpd3, but upregulation of Cers2 mRNA, as prominent anomalies in the ceramide-sphingosine metabolism. Reduction of Asah2 mRNA correlated to deficient S1P levels. In addition, downregulations for the elongase Elovl1, Elovl4, Elovl5 mRNAs and ELOVL4 protein explain the deficit of very long-chain sphingomyelin. Reduced ASMase protein levels correlated to the accumulation of long-chain sphingomyelin. Overall, a deficit of myelin lipids was prominent in SCA2 nervous tissue at prefinal stage and not compensated by transcriptional adaptation of several metabolic enzymes. Myelination is controlled by mTORC1 signals; thus, our human and murine observations are in agreement with the known role of ATXN2 yeast, nematode, and mouse orthologs as mTORC1 inhibitors and autophagy promoters.


Subject(s)
Ataxin-2/genetics , Ceramides/metabolism , Sphingomyelins/metabolism , Spinocerebellar Ataxias/genetics , Trinucleotide Repeat Expansion/genetics , Animals , Ataxin-2/metabolism , Disease Models, Animal , Eye Proteins/genetics , Eye Proteins/metabolism , Humans , Lipid Metabolism/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Knockout , Sphingomyelin Phosphodiesterase/genetics , Sphingomyelin Phosphodiesterase/metabolism , Sphingosine N-Acyltransferase/genetics , Sphingosine N-Acyltransferase/metabolism , Spinocerebellar Ataxias/metabolism , Spinocerebellar Ataxias/pathology
16.
Am J Hum Genet ; 97(3): 483-92, 2015 Sep 03.
Article in English | MEDLINE | ID: mdl-26320891

ABSTRACT

Progeroid disorders overlapping with De Barsy syndrome (DBS) are collectively denoted as autosomal-recessive cutis laxa type 3 (ARCL3). They are caused by biallelic mutations in PYCR1 or ALDH18A1, encoding pyrroline-5-carboxylate reductase 1 and pyrroline-5-carboxylate synthase (P5CS), respectively, which both operate in the mitochondrial proline cycle. We report here on eight unrelated individuals born to non-consanguineous families clinically diagnosed with DBS or wrinkly skin syndrome. We found three heterozygous mutations in ALDH18A1 leading to amino acid substitutions of the same highly conserved residue, Arg138 in P5CS. A de novo origin was confirmed in all six probands for whom parental DNA was available. Using fibroblasts from affected individuals and heterologous overexpression, we found that the P5CS-p.Arg138Trp protein was stable and able to interact with wild-type P5CS but showed an altered sub-mitochondrial distribution. A reduced size upon native gel electrophoresis indicated an alteration of the structure or composition of P5CS mutant complex. Furthermore, we found that the mutant cells had a reduced P5CS enzymatic activity leading to a delayed proline accumulation. In summary, recurrent de novo mutations, affecting the highly conserved residue Arg138 of P5CS, cause an autosomal-dominant form of cutis laxa with progeroid features. Our data provide insights into the etiology of cutis laxa diseases and will have immediate impact on diagnostics and genetic counseling.


Subject(s)
Corneal Opacity/genetics , Corneal Opacity/pathology , Cutis Laxa/genetics , Cutis Laxa/pathology , Intellectual Disability/genetics , Intellectual Disability/pathology , Mutation, Missense/genetics , Ornithine-Oxo-Acid Transaminase/genetics , Amino Acid Sequence , Base Sequence , Genes, Dominant/genetics , Humans , Molecular Sequence Data , Pedigree , Proline/metabolism , Sequence Alignment , Sequence Analysis, DNA , Skin/pathology , Species Specificity
17.
Mol Cell Proteomics ; 15(5): 1728-39, 2016 05.
Article in English | MEDLINE | ID: mdl-26850065

ABSTRACT

Human Ataxin-2 (ATXN2) gene locus variants have been associated with obesity, diabetes mellitus type 1,and hypertension in genome-wide association studies, whereas mouse studies showed the knock-out of Atxn2 to lead to obesity, insulin resistance, and dyslipidemia. Intriguingly, the deficiency of ATXN2 protein orthologs in yeast and flies rescues the neurodegeneration process triggered by TDP-43 and Ataxin-1 toxicity. To understand the molecular effects of ATXN2 deficiency by unbiased approaches, we quantified the global proteome and metabolome of Atxn2-knock-out mice with label-free mass spectrometry. In liver tissue, significant downregulations of the proteins ACADS, ALDH6A1, ALDH7A1, IVD, MCCC2, PCCA, OTC, together with bioinformatic enrichment of downregulated pathways for branched chain and other amino acid metabolism, fatty acids, and citric acid cycle were observed. Statistical trends in the cerebellar proteome and in the metabolomic profiles supported these findings. They are in good agreement with recent claims that PBP1, the yeast ortholog of ATXN2, sequestrates the nutrient sensor TORC1 in periods of cell stress. Overall, ATXN2 appears to modulate nutrition and metabolism, and its activity changes are determinants of growth excess or cell atrophy.


Subject(s)
Amino Acids, Branched-Chain/metabolism , Ataxin-2/genetics , Cerebellum/metabolism , Fatty Acids/metabolism , Liver/metabolism , Proteomics/methods , Animals , Ataxin-2/metabolism , Atrophy , Computational Biology/methods , Down-Regulation , Gene Regulatory Networks , Humans , Metabolic Networks and Pathways , Mice, Knockout , Protein Interaction Maps
18.
Mol Cell Proteomics ; 15(5): 1526-38, 2016 05.
Article in English | MEDLINE | ID: mdl-26852163

ABSTRACT

Hundreds of genes have been associated with respiratory chain disease (RCD), the most common inborn error of metabolism so far. Elimination of the respiratory electron chain by depleting the entire mitochondrial DNA (mtDNA, ρ(0) cells) has therefore one of the most severe impacts on the energy metabolism in eukaryotic cells. In this study, proteomic data sets including the post-translational modifications (PTMs) phosphorylation and ubiquitination were integrated with metabolomic data sets and selected enzyme activities in the osteosarcoma cell line 143B.TK(-) A shotgun based SILAC LC-MS proteomics and a targeted metabolomics approach was applied to elucidate the consequences of the ρ(0) state. Pathway and protein-protein interaction (PPI) network analyses revealed a nonuniform down-regulation of the respiratory electron chain, the tricarboxylic acid (TCA) cycle, and the pyruvate metabolism in ρ(0) cells. Metabolites of the TCA cycle were dysregulated, such as a reduction of citric acid and cis-aconitic acid (six and 2.5-fold), and an increase of lactic acid, oxalacetic acid (both twofold), and succinic acid (fivefold) in ρ(0) cells. Signaling pathways such as GPCR, EGFR, G12/13 alpha, and Rho GTPases were up-regulated in ρ(0) cells, which could be indicative for the mitochondrial retrograde response, a pathway of communication from mitochondria to the nucleus. This was supported by our phosphoproteome data, which revealed two main processes, GTPase-related signal transduction and cytoskeleton organization. Furthermore, a general de-ubiquitination in ρ(0) cells was observed, for example, 80S ribosomal proteins were in average threefold and SLC amino acid transporters fivefold de-ubiquitinated. The latter might cause the observed significant increase of amino acid levels in ρ(0) cells. We conclude that an elimination of the respiratory electron chain, e.g. mtDNA depletion, not only leads to an uneven down-regulation of mitochondrial energy pathways, but also triggers the retrograde response.


Subject(s)
Citric Acid Cycle , DNA, Mitochondrial/genetics , Energy Metabolism , Metabolomics/methods , Proteomics/methods , Pyruvic Acid/metabolism , Amino Acids/metabolism , Cell Line, Tumor , Down-Regulation , Gas Chromatography-Mass Spectrometry , Humans , Mitochondria/genetics , Mitochondria/metabolism , Phosphorylation , Protein Interaction Mapping , Proteome/metabolism , Sequence Deletion , Ubiquitination
19.
Nucleic Acids Res ; 44(13): 6142-56, 2016 07 27.
Article in English | MEDLINE | ID: mdl-27016732

ABSTRACT

Out of the myriad of potential DNA binding sites of the glucocorticoid receptor (GR) found in the human genome, only a cell-type specific minority is actually bound, indicating that the presence of a recognition sequence alone is insufficient to specify where GR binds. Cooperative interactions with other transcription factors (TFs) are known to contribute to binding specificity. Here, we reasoned that sequence signals preventing GR recruitment to certain loci provide an alternative means to confer specificity. Motif analyses uncovered candidate Negative Regulatory Sequences (NRSs) that interfere with genomic GR binding. Subsequent functional analyses demonstrated that NRSs indeed prevent GR binding to nearby response elements. We show that NRS activity is conserved across species, found in most tissues and that they also interfere with the genomic binding of other TFs. Interestingly, the effects of NRSs appear not to be a simple consequence of changes in chromatin accessibility. Instead, we find that NRSs interact with proteins found at sub-nuclear structures called paraspeckles and that these proteins might mediate the repressive effects of NRSs. Together, our studies suggest that the joint influence of positive and negative sequence signals partition the genome into regions where GR can bind and those where it cannot.


Subject(s)
Base Sequence/genetics , DNA-Binding Proteins/genetics , Receptors, Glucocorticoid/genetics , Response Elements/genetics , Binding Sites/genetics , Chromatin/genetics , DNA-Binding Proteins/metabolism , Humans , Promoter Regions, Genetic , Receptors, Glucocorticoid/metabolism , Transcription Factors/genetics
20.
Nucleic Acids Res ; 44(6): 2538-53, 2016 Apr 07.
Article in English | MEDLINE | ID: mdl-26582913

ABSTRACT

DPF3 (BAF45c) is a member of the BAF chromatin remodeling complex. Two isoforms have been described, namely DPF3a and DPF3b. The latter binds to acetylated and methylated lysine residues of histones. Here, we elaborate on the role of DPF3a and describe a novel pathway of cardiac gene transcription leading to pathological cardiac hypertrophy. Upon hypertrophic stimuli, casein kinase 2 phosphorylates DPF3a at serine 348. This initiates the interaction of DPF3a with the transcriptional repressors HEY, followed by the release of HEY from the DNA. Moreover, BRG1 is bound by DPF3a, and is thus recruited to HEY genomic targets upon interaction of the two components. Consequently, the transcription of downstream targets such as NPPA and GATA4 is initiated and pathological cardiac hypertrophy is established. In human, DPF3a is significantly up-regulated in hypertrophic hearts of patients with hypertrophic cardiomyopathy or aortic stenosis. Taken together, we show that activation of DPF3a upon hypertrophic stimuli switches cardiac fetal gene expression from being silenced by HEY to being activated by BRG1. Thus, we present a novel pathway for pathological cardiac hypertrophy, whose inhibition is a long-term therapeutic goal for the treatment of the course of heart failure.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Cardiomegaly/genetics , Chromatin Assembly and Disassembly , Chromatin/chemistry , DNA Helicases/genetics , DNA-Binding Proteins/genetics , Nuclear Proteins/genetics , Transcription Factors/genetics , Animals , Atrial Natriuretic Factor/genetics , Atrial Natriuretic Factor/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cardiomegaly/metabolism , Cardiomegaly/pathology , Casein Kinase II/genetics , Casein Kinase II/metabolism , Cell Differentiation , Chromatin/metabolism , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , GATA4 Transcription Factor/genetics , GATA4 Transcription Factor/metabolism , Gene Expression Regulation , HEK293 Cells , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Mice , Myoblasts/cytology , Myoblasts/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Nuclear Proteins/metabolism , Phosphorylation , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Signal Transduction , Transcription Factors/metabolism , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL