Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Theranostics ; 13(6): 1921-1948, 2023.
Article in English | MEDLINE | ID: mdl-37064875

ABSTRACT

Lysophosphatidic acid (LPA) species accumulate in the ascites of ovarian high-grade serous cancer (HGSC) and are associated with short relapse-free survival. LPA is known to support metastatic spread of cancer cells by activating a multitude of signaling pathways via G-protein-coupled receptors of the LPAR family. Systematic unbiased analyses of the LPA-regulated signal transduction network in ovarian cancer cells have, however, not been reported to date. Methods: LPA-induced signaling pathways were identified by phosphoproteomics of both patient-derived and OVCAR8 cells, RNA sequencing, measurements of intracellular Ca2+ and cAMP as well as cell imaging. The function of LPARs and downstream signaling components in migration and entosis were analyzed by selective pharmacological inhibitors and RNA interference. Results: Phosphoproteomic analyses identified > 1100 LPA-regulated sites in > 800 proteins and revealed interconnected LPAR1, ROCK/RAC, PKC/D and ERK pathways to play a prominent role within a comprehensive signaling network. These pathways regulate essential processes, including transcriptional responses, actomyosin dynamics, cell migration and entosis. A critical component of this signaling network is MYPT1, a stimulatory subunit of protein phosphatase 1 (PP1), which in turn is a negative regulator of myosin light chain 2 (MLC2). LPA induces phosphorylation of MYPT1 through ROCK (T853) and PKC/ERK (S507), which is majorly driven by LPAR1. Inhibition of MYPT1, PKC or ERK impedes both LPA-induced cell migration and entosis, while interference with ROCK activity and MLC2 phosphorylation selectively blocks entosis, suggesting that MYPT1 figures in both ROCK/MLC2-dependent and -independent pathways. We finally show a novel pathway governed by LPAR2 and the RAC-GEF DOCK7 to be indispensable for the induction of entosis. Conclusion: We have identified a comprehensive LPA-induced signal transduction network controlling LPA-triggered cytoskeletal changes, cell migration and entosis in HGSC cells. Due to its pivotal role in this network, MYPT1 may represent a promising target for interfering with specific functions of PP1 essential for HGSC progression.


Subject(s)
Actomyosin , Ovarian Neoplasms , Humans , Female , Actomyosin/metabolism , Entosis , Neoplasm Recurrence, Local , Signal Transduction , Ovarian Neoplasms/metabolism , Cell Movement/physiology
2.
Cancer Gene Ther ; 29(12): 1975-1987, 2022 12.
Article in English | MEDLINE | ID: mdl-35902728

ABSTRACT

Silencing of the Apoptosis associated Tyrosine Kinase gene (AATK) has been described in cancer. In our study, we specifically investigated the epigenetic inactivation of AATK in pancreatic adenocarcinoma, lower grade glioma, lung, breast, head, and neck cancer. The resulting loss of AATK correlates with impaired patient survival. Inhibition of DNA methyltransferases (DNMTs) reactivated AATK in glioblastoma and pancreatic cancer. In contrast, epigenetic targeting via the CRISPR/dCas9 system with either EZH2 or DNMT3A inhibited the expression of AATK. Via large-scale kinomic profiling and kinase assays, we demonstrate that AATK acts a Ser/Thr kinase that phosphorylates TP53 at Ser366. Furthermore, whole transcriptome analyses and mass spectrometry associate AATK expression with the GO term 'regulation of cell proliferation'. The kinase activity of AATK in comparison to the kinase-dead mutant mediates a decreased expression of the key cell cycle regulators Cyclin D1 and WEE1. Moreover, growth suppression through AATK relies on its kinase activity. In conclusion, the Ser/Thr kinase AATK represses growth and phosphorylates TP53. Furthermore, expression of AATK was correlated with a better patient survival for different cancer entities. This data suggests that AATK acts as an epigenetically inactivated tumor suppressor gene.


Subject(s)
Adenocarcinoma , Apoptosis Regulatory Proteins , Pancreatic Neoplasms , Protein-Tyrosine Kinases , Humans , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/genetics , Cyclin D1/genetics , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Pancreatic Neoplasms
3.
Clin Transl Med ; 11(11): e633, 2021 11.
Article in English | MEDLINE | ID: mdl-34841720

ABSTRACT

BACKGROUND: Transcoelomic spread is the major route of metastasis of ovarian high-grade serous carcinoma (HGSC) with the omentum as the major metastatic site. Its unique tumour microenvironment with its large populations of adipocytes, mesothelial cells and immune cells establishes an intercellular signaling network that is instrumental for metastatic growth yet poorly understood. METHODS: Based on transcriptomic analysis of tumour cells, tumour-associated immune and stroma cells we defined intercellular signaling pathways for 284 cytokines and growth factors and their cognate receptors after bioinformatic adjustment for contaminating cell types. The significance of individual components of this network was validated by analysing clinical correlations and potentially pro-metastatic functions, including tumour cell migration, pro-inflammatory signal transduction and TAM expansion. RESULTS: The data show an unexpected prominent role of host cells, and in particular of omental adipocytes, mesothelial cells and fibroblasts (CAF), in sustaining this signaling network. These cells, rather than tumour cells, are the major source of most cytokines and growth factors in the omental microenvironment (n = 176 vs. n = 13). Many of these factors target tumour cells, are linked to metastasis and are associated with a short survival. Likewise, tumour stroma cells play a major role in extracellular-matrix-triggered signaling. We have verified the functional significance of our observations for three exemplary instances. We show that the omental microenvironment (i) stimulates tumour cell migration and adhesion via WNT4 which is highly expressed by CAF; (ii) induces pro-tumourigenic TAM proliferation in conjunction with high CSF1 expression by omental stroma cells and (iii) triggers pro-inflammatory signaling, at least in part via a HSP70-NF-κB pathway. CONCLUSIONS: The intercellular signaling network of omental metastases is majorly dependent on factors secreted by immune and stroma cells to provide an environment that supports ovarian HGSC progression. Clinically relevant pathways within this network represent novel options for therapeutic intervention.


Subject(s)
Gene Regulatory Networks/physiology , Neoplasm Metastasis/physiopathology , Ovarian Neoplasms/physiopathology , Cell Movement/genetics , Cell Movement/physiology , Female , Gene Regulatory Networks/genetics , Humans , Neoplasm Metastasis/immunology , Ovarian Neoplasms/immunology , Signal Transduction/genetics , Signal Transduction/physiology
4.
Dev Biol ; 480: 1-12, 2021 12.
Article in English | MEDLINE | ID: mdl-34363825

ABSTRACT

Cardiac trabeculae are muscular ridge-like structures within the ventricular wall that are crucial for cardiac function. In zebrafish, these structures first form primarily through the delamination of compact wall cardiomyocytes (CMs). Although defects in proteasomal degradation have been associated with decreased cardiac function, whether they also affect cardiac development has not been extensively analyzed. Here we report a role during cardiac wall morphogenesis in zebrafish for the E3 ubiquitin-protein ligase Rbx1, which has been shown to regulate the degradation of key signaling molecules. Although development is largely unperturbed in zebrafish rbx1 mutant larvae, they exhibit CM multi-layering. This phenotype is not affected by blocking ErbB signaling, but fails to manifest itself in the absence of blood flow/cardiac contractility. Surprisingly, rbx1 mutants display ErbB independent Notch reporter expression in the myocardium. We generated tissue-specific rbx1 overexpression lines and found that endothelial, but not myocardial, specific rbx1 expression normalizes the cardiac wall morphogenesis phenotype. In addition, we found that pharmacological activation of Hedgehog signaling ameliorates the multi-layered myocardial wall phenotype in rbx1 mutants. Collectively, our data indicate that endocardial activity of Rbx1 is essential for cardiac wall morphogenesis.


Subject(s)
Myocardium/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Cell Proliferation/genetics , Endocardium/metabolism , Endothelium/metabolism , Gene Expression/genetics , Gene Expression Regulation/genetics , Genes, erbB/genetics , Heart/physiology , Heart Ventricles/metabolism , Hedgehog Proteins/metabolism , Morphogenesis/genetics , Myocardial Contraction , Myocytes, Cardiac/metabolism , Organogenesis/genetics , Receptors, Notch/genetics , Receptors, Notch/metabolism , Signal Transduction/genetics , Ubiquitin-Protein Ligases/genetics , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
5.
Theranostics ; 11(3): 1377-1395, 2021.
Article in English | MEDLINE | ID: mdl-33391540

ABSTRACT

Arachidonic acid (AA) is a polyunsaturated fatty acid present at high concentrations in the ovarian cancer (OC) microenvironment and associated with a poor clinical outcome. In the present study, we have unraveled a potential link between AA and macrophage functions. Methods: AA-triggered signal transduction was studied in primary monocyte-derived macrophages (MDMs) by phosphoproteomics, transcriptional profiling, measurement of intracellular Ca2+ accumulation and reactive oxygen species production in conjunction with bioinformatic analyses. Functional effects were investigated by actin filament staining, quantification of macropinocytosis and analysis of extracellular vesicle release. Results: We identified the ASK1 - p38δ/α (MAPK13/14) axis as a central constituent of signal transduction pathways triggered by non-metabolized AA. This pathway was induced by the Ca2+-triggered activation of calmodulin kinase II, and to a minor extent by ROS generation in a subset of donors. Activated p38 in turn was linked to a transcriptional stress response associated with a poor relapse-free survival. Consistent with the phosphorylation of the p38 substrate HSP27 and the (de)phosphorylation of multiple regulators of Rho family GTPases, AA impaired actin filament organization and inhibited actin-driven macropinocytosis. AA also affected the phosphorylation of proteins regulating vesicle biogenesis, and consistently, AA enhanced the release of tetraspanin-containing exosome-like vesicles. Finally, we identified phospholipase A2 group 2A (PLA2G2A) as the clinically most relevant enzyme producing extracellular AA, providing further potentially theranostic options. Conclusion: Our results suggest that AA contributes to an unfavorable clinical outcome of OC by impacting the phenotype of tumor-associated macrophages. Besides critical AA-regulated signal transduction proteins identified in the present study, PLA2G2A might represent a potential prognostic tool and therapeutic target to interfere with OC progression.


Subject(s)
Arachidonic Acid/pharmacology , Macrophages/drug effects , Ovarian Neoplasms/drug therapy , Phosphorylation/drug effects , Signal Transduction/drug effects , Calcium/metabolism , Extracellular Vesicles/drug effects , Extracellular Vesicles/metabolism , Female , Group II Phospholipases A2/metabolism , Humans , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/metabolism , Ovarian Neoplasms/metabolism , Reactive Oxygen Species/metabolism , Transcription, Genetic/drug effects , Tumor Microenvironment/drug effects
6.
Yeast ; 37(1): 15-25, 2020 01.
Article in English | MEDLINE | ID: mdl-31758572

ABSTRACT

Ergosterol is a prominent component of the yeast plasma membrane and essential for yeast cell viability. It is synthesized in the endoplasmic reticulum and transported to the plasma membrane by nonvesicular mechanisms requiring carrier proteins. Oxysterol-binding protein homologues and yeast StARkin proteins have been proposed to function as sterol carriers. Although many of these proteins are capable of transporting sterols between synthetic lipid vesicles in vitro, they are not essential for ergosterol transport in cells, indicating that they may be functionally redundant with each other or with additional-as yet unidentified-sterol carriers. To address this point, we hypothesized that sterol transport proteins are also sterol-binding proteins (SBPs), and used an in vitro chemoproteomic strategy to identify all cytosolic SBPs. We generated a cytosol fraction enriched in SBPs and captured the proteins with a photoreactive clickable cholesterol analogue. Quantitative proteomics of the captured proteins identified 342 putative SBPs. Analysis of these identified proteins based on their annotated function, reported drug phenotypes, interactions with proteins regulating lipid metabolism, gene ontology, and presence of mammalian orthologues revealed a subset of 62 characterized and nine uncharacterized candidates. Five of the uncharacterized proteins play a role in maintaining plasma membrane integrity as their absence affects the ability of cells to grow in the presence of nystatin or myriocin. We anticipate that the dataset reported here will be a comprehensive resource for functional analysis of sterol-binding/transport proteins and provide insights into novel aspects of non-vesicular sterol trafficking.


Subject(s)
Carrier Proteins/metabolism , Cholesterol/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Carrier Proteins/genetics , Cytosol/metabolism , Ergosterol/metabolism , Protein Transport , Proteomics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics
7.
Clin Epigenetics ; 11(1): 182, 2019 12 04.
Article in English | MEDLINE | ID: mdl-31801617

ABSTRACT

BACKGROUND: Cancer still is one of the leading causes of death and its death toll is predicted to rise further. We identified earlier the potential tumour suppressor zygote arrest 1 (ZAR1) to play a role in lung carcinogenesis through its epigenetic inactivation. RESULTS: We are the first to report that ZAR1 is epigenetically inactivated not only in lung cancer but also across cancer types, and ZAR1 methylation occurs across its complete CpG island. ZAR1 hypermethylation significantly correlates with its expression reduction in cancers. We are also the first to report that ZAR1 methylation and expression reduction are of clinical importance as a prognostic marker for lung cancer and kidney cancer. We further established that the carboxy (C)-terminally present zinc-finger of ZAR1 is relevant for its tumour suppression function and its protein partner binding associated with the mRNA/ribosomal network. Global gene expression profiling supported ZAR1's role in cell cycle arrest and p53 signalling pathway, and we could show that ZAR1 growth suppression was in part p53 dependent. Using the CRISPR-dCas9 tools, we were able to prove that epigenetic editing and reactivation of ZAR1 is possible in cancer cell lines. CONCLUSION: ZAR1 is a novel cancer biomarker for lung and kidney, which is epigenetically silenced in various cancers by DNA hypermethylation. ZAR1 exerts its tumour suppressive function in part through p53 and through its zinc-finger domain. Epigenetic therapy can reactivate the ZAR1 tumour suppressor in cancer.


Subject(s)
Biomarkers, Tumor/genetics , DNA Methylation , Egg Proteins/genetics , Egg Proteins/metabolism , Kidney Neoplasms/diagnosis , Lung Neoplasms/diagnosis , A549 Cells , Binding Sites , Cell Cycle , Cell Line, Tumor , CpG Islands , Down-Regulation , Early Detection of Cancer , Egg Proteins/chemistry , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , HCT116 Cells , HeLa Cells , Humans , Kidney Neoplasms/drug therapy , Kidney Neoplasms/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Prognosis , Signal Transduction , Survival Analysis , Tumor Suppressor Protein p53/metabolism , Zinc Fingers
8.
Proc Natl Acad Sci U S A ; 116(48): 24115-24121, 2019 11 26.
Article in English | MEDLINE | ID: mdl-31704768

ABSTRACT

Atrial fibrillation (AF) is the most common type of cardiac arrhythmia. The major AF susceptibility locus 4q25 establishes long-range interactions with the promoter of PITX2, a transcription factor gene with critical functions during cardiac development. While many AF-linked loci have been identified in genome-wide association studies, mechanistic understanding into how genetic variants, including those at the 4q25 locus, increase vulnerability to AF is mostly lacking. Here, we show that loss of pitx2c in zebrafish leads to adult cardiac phenotypes with substantial similarities to pathologies observed in AF patients, including arrhythmia, atrial conduction defects, sarcomere disassembly, and altered cardiac metabolism. These phenotypes are also observed in a subset of pitx2c+/- fish, mimicking the situation in humans. Most notably, the onset of these phenotypes occurs at an early developmental stage. Detailed analyses of pitx2c loss- and gain-of-function embryonic hearts reveal changes in sarcomeric and metabolic gene expression and function that precede the onset of cardiac arrhythmia first observed at larval stages. We further find that antioxidant treatment of pitx2c-/- larvae significantly reduces the incidence and severity of cardiac arrhythmia, suggesting that metabolic dysfunction is an important driver of conduction defects. We propose that these early sarcomere and metabolic defects alter cardiac function and contribute to the electrical instability and structural remodeling observed in adult fish. Overall, these data provide insight into the mechanisms underlying the development and pathophysiology of some cardiac arrhythmias and importantly, increase our understanding of how developmental perturbations can predispose to functional defects in the adult heart.


Subject(s)
Arrhythmias, Cardiac/metabolism , Homeodomain Proteins/genetics , Sarcomeres/metabolism , Transcription Factors/genetics , Zebrafish Proteins/genetics , Zebrafish/genetics , Acetylcysteine/pharmacology , Animals , Animals, Genetically Modified , Antioxidants/pharmacology , Arrhythmias, Cardiac/drug therapy , Arrhythmias, Cardiac/etiology , Cardiac Conduction System Disease/etiology , Cardiac Conduction System Disease/genetics , Cardiomyopathies/genetics , Cardiomyopathies/physiopathology , Disease Models, Animal , Electrocardiography , Gene Expression Regulation , Homeodomain Proteins/metabolism , Larva/drug effects , Mitochondria, Heart/genetics , Mitochondria, Heart/metabolism , Mitochondria, Heart/pathology , Sarcomeres/genetics , Sarcomeres/pathology , Stress, Physiological/genetics , Transcription Factors/metabolism , Zebrafish Proteins/metabolism
9.
J Clin Invest ; 129(7): 2775-2791, 2019 06 17.
Article in English | MEDLINE | ID: mdl-31205027

ABSTRACT

Hypertension is a primary risk factor for cardiovascular diseases including myocardial infarction and stroke. Major determinants of blood pressure are vasodilatory factors such as nitric oxide (NO) released from the endothelium under the influence of fluid shear stress exerted by the flowing blood. Several endothelial signaling processes mediating fluid shear stress-induced formation and release of vasodilatory factors have been described. It is, however, still poorly understood how fluid shear stress induces these endothelial responses. Here we show that the endothelial mechanosensitive cation channel PIEZO1 mediated fluid shear stress-induced release of adrenomedullin, which in turn activated its Gs-coupled receptor. The subsequent increase in cAMP levels promoted the phosphorylation of endothelial NO synthase (eNOS) at serine 633 through protein kinase A (PKA), leading to the activation of the enzyme. This Gs/PKA-mediated pathway synergized with the AKT-mediated pathways leading to eNOS phosphorylation at serine 1177. Mice with endothelium-specific deficiency of adrenomedullin, the adrenomedullin receptor, or Gαs showed reduced flow-induced eNOS activation and vasodilation and developed hypertension. Our data identify fluid shear stress-induced PIEZO1 activation as a central regulator of endothelial adrenomedullin release and establish the adrenomedullin receptor and subsequent Gs-mediated formation of cAMP as a critical endothelial mechanosignaling pathway regulating basal endothelial NO formation, vascular tone, and blood pressure.


Subject(s)
Adrenomedullin/metabolism , Blood Pressure , Endothelium, Vascular , Second Messenger Systems , Stress, Mechanical , Animals , Cyclic AMP/metabolism , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Endothelium, Vascular/physiopathology , Human Umbilical Vein Endothelial Cells , Humans , Hypertension/metabolism , Hypertension/pathology , Hypertension/physiopathology , Ion Channels/metabolism , Male , Mice , Mice, Knockout , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/metabolism
11.
Elife ; 72018 12 06.
Article in English | MEDLINE | ID: mdl-30520733

ABSTRACT

Pathways modulating glucose homeostasis independently of insulin would open new avenues to combat insulin resistance and diabetes. Here, we report the establishment, characterization, and use of a vertebrate 'insulin-free' model to identify insulin-independent modulators of glucose metabolism. insulin knockout zebrafish recapitulate core characteristics of diabetes and survive only up to larval stages. Utilizing a highly efficient endoderm transplant technique, we generated viable chimeric adults that provide the large numbers of insulin mutant larvae required for our screening platform. Using glucose as a disease-relevant readout, we screened 2233 molecules and identified three that consistently reduced glucose levels in insulin mutants. Most significantly, we uncovered an insulin-independent beneficial role for androgen receptor antagonism in hyperglycemia, mostly by reducing fasting glucose levels. Our study proposes therapeutic roles for androgen signaling in diabetes and, more broadly, offers a novel in vivo model for rapid screening and decoupling of insulin-dependent and -independent mechanisms.


Subject(s)
Glucose/metabolism , Hyperglycemia/genetics , Insulin/genetics , Receptors, Androgen/genetics , Androgen Receptor Antagonists/chemistry , Androgen Receptor Antagonists/metabolism , Animals , Disease Models, Animal , Gene Knockout Techniques , Homeostasis , Hyperglycemia/metabolism , Hyperglycemia/pathology , Insulin Resistance/genetics , Receptors, Androgen/chemistry , Signal Transduction/genetics , Zebrafish/genetics
12.
Nat Commun ; 9(1): 4600, 2018 11 02.
Article in English | MEDLINE | ID: mdl-30389913

ABSTRACT

Impaired alveolar formation and maintenance are features of many pulmonary diseases that are associated with significant morbidity and mortality. In a forward genetic screen for modulators of mouse lung development, we identified the non-muscle myosin II heavy chain gene, Myh10. Myh10 mutant pups exhibit cyanosis and respiratory distress, and die shortly after birth from differentiation defects in alveolar epithelium and mesenchyme. From omics analyses and follow up studies, we find decreased Thrombospondin expression accompanied with increased matrix metalloproteinase activity in both mutant lungs and cultured mutant fibroblasts, as well as disrupted extracellular matrix (ECM) remodeling. Loss of Myh10 specifically in mesenchymal cells results in ECM deposition defects and alveolar simplification. Notably, MYH10 expression is downregulated in the lung of emphysema patients. Altogether, our findings reveal critical roles for Myh10 in alveologenesis at least in part via the regulation of ECM remodeling, which may contribute to the pathogenesis of emphysema.


Subject(s)
Extracellular Matrix/metabolism , Lung Diseases/metabolism , Myosin Heavy Chains/deficiency , Nonmuscle Myosin Type IIB/deficiency , Amino Acid Sequence , Animals , Down-Regulation/genetics , Emphysema/pathology , Ethylnitrosourea , Female , Lung Diseases/pathology , Male , Matrix Metalloproteinase 2/metabolism , Mesoderm/metabolism , Mice, Inbred C57BL , Mutagenesis/genetics , Mutation, Missense/genetics , Myosin Heavy Chains/chemistry , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Nonmuscle Myosin Type IIB/chemistry , Nonmuscle Myosin Type IIB/genetics , Nonmuscle Myosin Type IIB/metabolism , Organogenesis , Phenotype , Pulmonary Alveoli/embryology , Pulmonary Alveoli/metabolism , Up-Regulation/genetics
13.
PLoS Genet ; 14(11): e1007743, 2018 11.
Article in English | MEDLINE | ID: mdl-30457989

ABSTRACT

Development and function of tissues and organs are powered by the activity of mitochondria. In humans, inherited genetic mutations that lead to progressive mitochondrial pathology often manifest during infancy and can lead to death, reflecting the indispensable nature of mitochondrial biogenesis and function. Here, we describe a zebrafish mutant for the gene mia40a (chchd4a), the life-essential homologue of the evolutionarily conserved Mia40 oxidoreductase which drives the biogenesis of cysteine-rich mitochondrial proteins. We report that mia40a mutant animals undergo progressive cellular respiration defects and develop enlarged mitochondria in skeletal muscles before their ultimate death at the larval stage. We generated a deep transcriptomic and proteomic resource that allowed us to identify abnormalities in the development and physiology of endodermal organs, in particular the liver and pancreas. We identify the acinar cells of the exocrine pancreas to be severely affected by mutations in the MIA pathway. Our data contribute to a better understanding of the molecular, cellular and organismal effects of mitochondrial deficiency, important for the accurate diagnosis and future treatment strategies of mitochondrial diseases.

14.
Sci Rep ; 6: 27484, 2016 06 06.
Article in English | MEDLINE | ID: mdl-27265872

ABSTRACT

Disulfide bond formation is crucial for the biogenesis and structure of many proteins that are localized in the intermembrane space of mitochondria. The importance of disulfide bond formation within mitochondrial proteins was extended beyond soluble intermembrane space proteins. Tim22, a membrane protein and core component of the mitochondrial translocase TIM22, forms an intramolecular disulfide bond in yeast. Tim22 belongs to the Tim17/Tim22/Tim23 family of protein translocases. Here, we present evidence of the high evolutionary conservation of disulfide bond formation in Tim17 and Tim22 among fungi and metazoa. Topological models are proposed that include the location of disulfide bonds relative to the predicted transmembrane regions. Yeast and human Tim22 variants that are not oxidized do not properly integrate into the membrane complex. Moreover, the lack of Tim17 oxidation disrupts the TIM23 translocase complex. This underlines the importance of disulfide bond formation for mature translocase assembly through membrane stabilization of weak transmembrane domains.


Subject(s)
Biological Evolution , Disulfides/chemistry , Membrane Transport Proteins/metabolism , Mitochondrial Proteins/metabolism , Humans , Oxidation-Reduction , Saccharomyces cerevisiae/metabolism
15.
Hum Mol Genet ; 24(23): 6640-52, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26376862

ABSTRACT

Autosomal recessively inherited glucocerebrosidase 1 (GBA1) mutations cause the lysosomal storage disorder Gaucher's disease (GD). Heterozygous GBA1 mutations (GBA1(+/-)) are the most common risk factor for Parkinson's disease (PD). Previous studies typically focused on the interaction between the reduction of glucocerebrosidase (enzymatic) activity in GBA1(+/-) carriers and alpha-synuclein-mediated neurotoxicity. However, it is unclear whether other mechanisms also contribute to the increased risk of PD in GBA1(+/-) carriers. The zebrafish genome does not contain alpha-synuclein (SNCA), thus providing a unique opportunity to study pathogenic mechanisms unrelated to alpha-synuclein toxicity. Here we describe a mutant zebrafish line created by TALEN genome editing carrying a 23 bp deletion in gba1 (gba1(c.1276_1298del)), the zebrafish orthologue of human GBA1. Marked sphingolipid accumulation was already detected at 5 days post-fertilization with accompanying microglial activation and early, sustained up-regulation of miR-155, a master regulator of inflammation. gba1(c.1276_1298del) mutant zebrafish developed a rapidly worsening phenotype from 8 weeks onwards with striking reduction in motor activity by 12 weeks. Histopathologically, we observed marked Gaucher cell invasion of the brain and other organs. Dopaminergic neuronal cell count was normal through development but reduced by >30% at 12 weeks in the presence of ubiquitin-positive, intra-neuronal inclusions. This gba1(c.1276_1298del) zebrafish line is the first viable vertebrate model sharing key pathological features of GD in both neuronal and non-neuronal tissue. Our study also provides evidence for early microglial activation prior to alpha-synuclein-independent neuronal cell death in GBA1 deficiency and suggests upregulation of miR-155 as a common denominator across different neurodegenerative disorders.


Subject(s)
Disease Models, Animal , Gaucher Disease/genetics , Glucosylceramidase/genetics , Neurons/pathology , Zebrafish Proteins/genetics , Zebrafish , Animals , Cell Death , Gaucher Disease/pathology , MicroRNAs/genetics , Microglia/metabolism , Microglia/physiology , Neurons/metabolism , Neurons/physiology , Sequence Deletion , Up-Regulation , Zebrafish/genetics , Zebrafish/metabolism , alpha-Synuclein/metabolism
16.
Sci Rep ; 3: 3277, 2013 Nov 20.
Article in English | MEDLINE | ID: mdl-24253929

ABSTRACT

Human cells lacking DNA polymerase η (polη) are sensitive to platinum-based cancer chemotherapeutic agents. Using DNA combing to directly investigate the role of polη in bypass of platinum-induced DNA lesions in vivo, we demonstrate that nascent DNA strands are up to 39% shorter in human cells lacking polη than in cells expressing polη. This provides the first direct evidence that polη modulates replication fork progression in vivo following cisplatin and carboplatin treatment. Severe replication inhibition in individual platinum-treated polη-deficient cells correlates with enhanced phosphorylation of the RPA2 subunit of replication protein A on serines 4 and 8, as determined using EdU labelling and immunofluorescence, consistent with formation of DNA strand breaks at arrested forks in the absence of polη. Polη-mediated bypass of platinum-induced DNA lesions may therefore represent one mechanism by which cancer cells can tolerate platinum-based chemotherapy.


Subject(s)
DNA Damage/drug effects , DNA Damage/physiology , DNA Replication/drug effects , DNA Replication/physiology , DNA-Directed DNA Polymerase/metabolism , Platinum/pharmacology , Carboplatin/pharmacology , Carboplatin/toxicity , Cell Cycle/drug effects , Cell Cycle/physiology , Cell Line , Cisplatin/pharmacology , Cisplatin/toxicity , DNA-Directed DNA Polymerase/deficiency , DNA-Directed DNA Polymerase/genetics , Drug Synergism , Gene Expression , Humans , Phosphorylation , Platinum/toxicity , Replication Protein A/metabolism
17.
Bioorg Med Chem ; 18(8): 2930-6, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20347318

ABSTRACT

A series of new aromatic monoesters of alpha-aminoaralkylphosphonic acids were synthesized by selective hydrolysis of corresponding aromatic diesters of alpha-aminoaralkylphosphonic acids. New potential inhibitors of aminopeptidase N/CD13, an enzyme important in tumour angiogenesis, were developed. Some derivatives of the homophenylalanine and norleucine related monoaryl phosphonates displayed higher inhibition potency than corresponding alpha-aminoaralkylphosphonic acids toward aminopeptidase N/CD13. The effect of one of the new inhibitors on the growth of human PANC-1 and HT-1080 cell lines was examined, either alone or in combination with TNF-alpha.


Subject(s)
Angiogenesis Inhibitors/chemistry , CD13 Antigens/antagonists & inhibitors , Phosphinic Acids/chemistry , Protease Inhibitors/chemistry , Angiogenesis Inhibitors/chemical synthesis , Angiogenesis Inhibitors/pharmacology , CD13 Antigens/metabolism , Cell Line, Tumor , Humans , Phosphinic Acids/chemical synthesis , Phosphinic Acids/pharmacology , Protease Inhibitors/chemical synthesis , Protease Inhibitors/pharmacology , Structure-Activity Relationship , Tumor Necrosis Factor-alpha/metabolism
18.
Subcell Biochem ; 50: 189-209, 2010.
Article in English | MEDLINE | ID: mdl-20012583

ABSTRACT

Genomic DNA is constantly damaged by exposure to exogenous and endogenous agents. Bulky adducts such as UV-induced cyclobutane pyrimidine dimers (CPDs) in the template DNA present a barrier to DNA synthesis by the major eukaryotic replicative polymerases including DNA polymerase delta. Translesion synthesis (TLS) carried out by specialized DNA polymerases is an evolutionarily conserved mechanism of DNA damage tolerance. The Y family of DNA polymerases, including DNA polymerase eta (Pol eta), the subject of this chapter, play a key role in TLS. Mutations in the human POLH gene encoding Pol eta underlie the genetic disease xeroderma pigmentosum variant (XPV), characterized by sun sensitivity, elevated incidence of skin cancer, and at the cellular level, by delayed replication and hypermutability after UV-irradiation. Pol eta is a low fidelity enzyme when copying undamaged DNA, but can carry out error-free TLS at sites of UV-induced dithymine CPDs. The active site of Pol eta has an open conformation that can accommodate CPDs, as well as cisplatin-induced intrastrand DNA crosslinks. Pol eta is recruited to sites of replication arrest in a tightly regulated process through interaction with PCNA. Pol eta-deficient cells show strong activation of downstream DNA damage responses including ATR signaling, and accumulate strand breaks as a result of replication fork collapse. Thus, Pol eta plays an important role in preventing genome instability after UV- and cisplatin-induced DNA damage. Inhibition of DNA damage tolerance pathways in tumors might also represent an approach to potentiate the effects of DNA damaging agents such as cisplatin.


Subject(s)
DNA Damage , DNA Repair , DNA-Directed DNA Polymerase/metabolism , Humans
19.
Cell Cycle ; 8(18): 3039-50, 2009 Sep 15.
Article in English | MEDLINE | ID: mdl-19713747

ABSTRACT

Translesion synthesis by DNA polymerase eta (poleta) is one mechanism by which cancer cells can tolerate DNA damage by platinum-based anti-cancer drugs. Cells lacking poleta are sensitive to these agents. To help define the consequences of poeta-deficiency, we characterized the effects of equitoxic doses of cisplatin and carboplatin on cell cycle progression and activation of DNA damage response pathways in a human cell line lacking poleta. We show that both cisplatin and carboplatin induce strong S-phase arrest in poleta-deficient XP30RO cells, associated with reduced expression of cyclin E and cyclin B. PIK kinase-mediated phosphorylation of Chk1, H2AX and RPA2 was strongly activated by both cisplatin and carboplatin, but phosphorylation of these proteins was induced earlier by cisplatin than by an equitoxic dose of carboplatin. Compared to Chk1 and H2AX phosphorylation, RPA2 hyperphosphorylation on serine4/serine8 is a late event in response to platinum-induced DNA damage. We directly demonstrate, using dual-labeling flow cytometry, that damage-induced phosphorylation of RPA2 on serine4/serine8 occurs primarily in the S and G(2) phases of the cell cycle, and show that the timing of RPA2 phosphorylation can be modulated by inhibition of the checkpoint kinase Chk1. Furthermore, Chk1 inhibition sensitizes poleta-deficient cells to the cytotoxic effects of carboplatin. Both hyperphosphorylated RPA2 and the homologous recombination protein Rad51 are present in nuclear foci after cisplatin treatment, but these are separable events in individual cells. These results provide insight into the relationship between cell cycle regulation and processing of platinum-induced DNA damage in human cells when poleta-mediated TLS is compromised.


Subject(s)
Carboplatin , Cell Cycle , Cisplatin , DNA Repair/drug effects , DNA-Directed DNA Polymerase/deficiency , Antineoplastic Agents/pharmacology , Carboplatin/pharmacology , Cell Cycle/drug effects , Cell Line, Transformed , Cisplatin/pharmacology , DNA Damage , Fibroblasts , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...