Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters











Publication year range
1.
Cancers (Basel) ; 14(17)2022 Aug 23.
Article in English | MEDLINE | ID: mdl-36077614

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC), due to its genomic heterogeneity and lack of effective treatment, despite decades of intensive research, will become the second leading cause of cancer-related deaths by 2030. Step-wise acquisition of mutations, due to genomic instability, is considered to drive the development of PDAC; the KRAS mutation occurs in 95 to 100% of human PDAC, and is already detectable in early premalignant lesions designated as pancreatic intraepithelial neoplasia (PanIN). This mutation is possibly the key event leading to genomic instability and PDAC development. Our study aimed to investigate the role of the error-prone DNA double-strand breaks (DSBs) repair pathway, alt-EJ, in the presence of the KRAS G12D mutation in pancreatic cancer development. Our findings show that oncogenic KRAS contributes to increasing the expression of Polθ, Lig3, and Mre11, key components of alt-EJ in both mouse and human PDAC models. We further confirm increased catalytic activity of alt-EJ in a mouse and human model of PDAC bearing the KRAS G12D mutation. Subsequently, we focused on estimating the impact of alt-EJ inactivation by polymerase theta (Polθ) deletion on pancreatic cancer development, and survival in genetically engineered mouse models (GEMMs) and cancer patients. Here, we show that even though Polθ deficiency does not fully prevent the development of pancreatic cancer, it significantly delays the onset of PanIN formation, prolongs the overall survival of experimental mice, and correlates with the overall survival of pancreatic cancer patients in the TCGA database. Our study clearly demonstrates the role of alt-EJ in the development of PDAC, and alt-EJ may be an attractive therapeutic target for pancreatic cancer patients.

2.
Heliyon ; 6(1): e03187, 2020 Jan.
Article in English | MEDLINE | ID: mdl-32042960

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest cancers. Acquired inherited and/or somatic mutations drive its development. In order to prevent the formation of these mutations, precise and immediate repair of any DNA damage is indispensable. Non-homologous end-joining (NHEJ) is the key mechanism of DNA double-strand break repair. Here, we report that miR-502 targets two components in pancreatic cell lines, Ku70 and XLF of the C-NHEJ. Interestingly, we also observed an attenuated cell cycle response to gamma ionizing radiation (γ-IR) via diminished phosphorylation of checkpoint kinase 1 (Chk1) on serine 345 in these cell lines. Altogether, pancreatic cells showed increased susceptibility to γ-IR via direct inhibition of DNA double-strand break repair and attenuation of the cell cycle response.

3.
Clin Anat ; 33(3): 431-439, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31883163

ABSTRACT

INTRODUCTION: Transabdominal ultrasound (US) and magnetic resonance imaging (MRI) are commonly used for the examination of the pancreas in clinical routine. We therefore were interested in the concordance of these two imaging methods for the size measurement of the pancreas and how age, gender, and body mass index (BMI) affect the organ size. METHODS: A total of 342 participants from the Study of Health in Pomerania underwent whole-body MRI and transabdominal US on the same day, and the diameter of the pancreatic head, body, and tail were measured. The agreement between US and MRI measurements was assessed by Bland and Altman plots. Intraclass correlation coefficients were used to compare observers. A multivariable regression model was applied using the independent variables age, gender, and body mass index. RESULTS: Compared to MRI, abdominal US returned smaller values for each segment of the pancreas, with a high level of inconsistency between these two methods. The mean difference was 0.39, 0.18, and 0.54 cm for the head, body, and tail, respectively. A high interobserver variability was detected for US. Multivariable analysis showed that pancreatic size in all three segments increased with BMI in both genders whereas pancreatic head and tail size decreased with age, an effect more marked in women. CONCLUSIONS: Agreement of pancreatic size measurements is poor between US and MRI. These limitations should be considered when evaluating morphologic features for pathologic conditions or setting limits of normal size. Adjustments for BMI, gender, and age may also be warranted.


Subject(s)
Magnetic Resonance Imaging , Pancreas/anatomy & histology , Pancreas/diagnostic imaging , Ultrasonography , Adult , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Observer Variation , Organ Size , Reproducibility of Results
4.
Dig Dis ; 34(5): 525-31, 2016.
Article in English | MEDLINE | ID: mdl-27332960

ABSTRACT

BACKGROUND: Pancreatic ductal adenocarcinoma (PDAC) is the 4th leading cause of cancer death worldwide and compared to other malignancies its share in cancer mortality is expected to rise further. This is due to a lack of sensitive diagnostic tools that would permit earlier detection in a potentially curable stage and the very slow progress in finding effective drug treatments for pancreatic cancer. KEY MESSAGES: Aside from genetic predispositions and environmental agents, chronic pancreatitis is by far the greatest risk factor for PDAC. It also shares several etiological factors with pancreatic cancer and represents its most challenging differential diagnosis. Biomarkers that can distinguish between chronic pancreatitis and PDAC may therefore be suitable for the latter's early detection. Moreover, targeting the natural history of chronic pancreatitis would be one approach to prevent PDAC. Targeting tumor-cell signaling directly by interfering with receptor tyrosine kinases has shown some efficacy, although the results in clinical trials were less encouraging than for other cancers. Other compounds developed have targeted the formation of extracellular matrix around the tumor, the proteolytic activity in the tumor environment, histone deacetylases, hedgehog signaling and heat shock proteins, but none has yet found its way into routine patient care. Attempts to individualize treatment according to the tumor's somatic mutation profile are novel but so far impractical. CONCLUSIONS: Progress in the treatment of pancreatic cancer has been exceedingly slow and mostly dependent on improved pharmaceutical preparations or combinations of established chemotherapeutic agents. The promise of major breakthroughs implied in targeting tumor signal transduction events has so far not materialized.


Subject(s)
Antineoplastic Agents/therapeutic use , Early Detection of Cancer/methods , Pancreatic Neoplasms/diagnosis , Pancreatitis, Chronic/complications , Biomarkers, Tumor , Humans , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/therapy , Precision Medicine/methods , Risk Factors , Signal Transduction
5.
Sci Transl Med ; 7(287): 287ra69, 2015 May 13.
Article in English | MEDLINE | ID: mdl-25972001

ABSTRACT

Accidental radiation exposure is a threat to human health that necessitates effective clinical planning and diagnosis. Minimally invasive biomarkers that can predict long-term radiation injury are urgently needed for optimal management after a radiation accident. We have identified serum microRNA (miRNA) signatures that indicate long-term impact of total body irradiation (TBI) in mice when measured within 24 hours of exposure. Impact of TBI on the hematopoietic system was systematically assessed to determine a correlation of residual hematopoietic stem cells (HSCs) with increasing doses of radiation. Serum miRNA signatures distinguished untreated mice from animals exposed to radiation and correlated with the impact of radiation on HSCs. Mice exposed to sublethal (6.5 Gy) and lethal (8 Gy) doses of radiation were indistinguishable for 3 to 4 weeks after exposure. A serum miRNA signature detectable 24 hours after radiation exposure consistently segregated these two cohorts. Furthermore, using either a radioprotective agent before, or radiation mitigation after, lethal radiation, we determined that the serum miRNA signature correlated with the impact of radiation on animal health rather than the radiation dose. Last, using humanized mice that had been engrafted with human CD34(+) HSCs, we determined that the serum miRNA signature indicated radiation-induced injury to the human bone marrow cells. Our data suggest that serum miRNAs can serve as functional dosimeters of radiation, representing a potential breakthrough in early assessment of radiation-induced hematopoietic damage and timely use of medical countermeasures to mitigate the long-term impact of radiation.


Subject(s)
Biomarkers/blood , Hematopoietic Stem Cells/radiation effects , MicroRNAs/blood , Whole-Body Irradiation , Animals , Gene Expression Profiling , Humans , Male , Mice , Mice, Inbred C57BL , MicroRNAs/genetics
6.
Mol Cancer Res ; 12(12): 1767-78, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25256711

ABSTRACT

UNLABELLED: The efficacy of radiotherapy in many tumor types is limited by normal tissue toxicity and by intrinsic or acquired radioresistance. Therefore, it is essential to understand the molecular network responsible for regulating radiosensitivity/resistance. Here, an unbiased functional screen identified four microRNAs (miR1, miR125a, miR150, and miR425) that induce radioresistance. Considering the clinical importance of radiotherapy for patients with glioblastoma, the impact of these miRNAs on glioblastoma radioresistance was investigated. Overexpression of miR1, miR125a, miR150, and/or miR425 in glioblastoma promotes radioresistance through upregulation of the cell-cycle checkpoint response. Conversely, antagonizing with antagomiRs sensitizes glioblastoma cells to irradiation, suggesting their potential as targets for inhibiting therapeutic resistance. Analysis of glioblastoma datasets from The Cancer Genome Atlas (TCGA) revealed that these miRNAs are expressed in glioblastoma patient specimens and correlate with TGFß signaling. Finally, it is demonstrated that expression of miR1 and miR125a can be induced by TGFß and antagonized by a TGFß receptor inhibitor. Together, these results identify and characterize a new role for miR425, miR1, miR125, and miR150 in promoting radioresistance in glioblastomas and provide insight into the therapeutic application of TGFß inhibitors in radiotherapy. IMPLICATIONS: Systematic identification of miRs that cause radioresistance in gliomas is important for uncovering predictive markers for radiotherapy or targets for overcoming radioresistance.


Subject(s)
Glioblastoma/genetics , MicroRNAs/genetics , Radiation Tolerance , Cell Cycle Checkpoints/radiation effects , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/radiation effects , Glioblastoma/radiotherapy , Humans , Signal Transduction/radiation effects , Transforming Growth Factor beta/metabolism
7.
Mol Cell ; 41(2): 210-20, 2011 Jan 21.
Article in English | MEDLINE | ID: mdl-21195000

ABSTRACT

Expression of BRCA1 is commonly decreased in sporadic breast tumors, and this correlates with poor prognosis of breast cancer patients. Here we show that BRCA1 transcripts are selectively enriched in the Argonaute/miR-182 complex and miR-182 downregulates BRCA1 expression. Antagonizing miR-182 enhances BRCA1 protein levels and protects them from IR-induced cell death, while overexpressing miR-182 reduces BRCA1 protein, impairs homologous recombination-mediated repair, and render cells hypersensitive to IR. The impaired DNA repair phenotype induced by miR-182 overexpression can be fully rescued by overexpressing miR-182-insensitive BRCA1. Consistent with a BRCA1-deficiency phenotype, miR-182-overexpressing breast tumor cells are hypersensitive to inhibitors of poly (ADP-ribose) polymerase 1 (PARP1). Conversely, antagonizing miR-182 enhances BRCA1 levels and induces resistance to PARP1 inhibitor. Finally, a clinical-grade PARP1 inhibitor impacts outgrowth of miR-182-expressing tumors in animal models. Together these results suggest that miR-182-mediated downregulation of BRCA1 impedes DNA repair and may impact breast cancer therapy.


Subject(s)
Antineoplastic Agents/pharmacology , BRCA1 Protein/genetics , DNA Repair/drug effects , MicroRNAs/physiology , Phthalazines/pharmacology , Piperazines/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors , Animals , Cell Differentiation , Cell Line, Tumor , DNA Breaks, Double-Stranded/radiation effects , Down-Regulation , Humans , K562 Cells , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Poly (ADP-Ribose) Polymerase-1
8.
J Cell Biol ; 191(1): 31-43, 2010 Oct 04.
Article in English | MEDLINE | ID: mdl-20876283

ABSTRACT

The complexity of chromatin architecture presents a significant barrier to the ability of the DNA repair machinery to access and repair DNA double-strand breaks (DSBs). Consequently, remodeling of the chromatin landscape adjacent to DSBs is vital for efficient DNA repair. Here, we demonstrate that DNA damage destabilizes nucleosomes within chromatin regions that correspond to the γ-H2AX domains surrounding DSBs. This nucleosome destabilization is an active process requiring the ATPase activity of the p400 SWI/SNF ATPase and histone acetylation by the Tip60 acetyltransferase. p400 is recruited to DSBs by a mechanism that is independent of ATM but requires mdc1. Further, the destabilization of nucleosomes by p400 is required for the RNF8-dependent ubiquitination of chromatin, and for the subsequent recruitment of brca1 and 53BP1 to DSBs. These results identify p400 as a novel DNA damage response protein and demonstrate that p400-mediated alterations in nucleosome and chromatin structure promote both chromatin ubiquitination and the accumulation of brca1 and 53BP1 at sites of DNA damage.


Subject(s)
Chromatin/metabolism , DNA Helicases/physiology , DNA Repair , DNA-Binding Proteins/physiology , Nucleosomes/metabolism , Chromatin Assembly and Disassembly , DNA Damage , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Histone Acetyltransferases/metabolism , Histone Acetyltransferases/physiology , Histones/metabolism , Humans , Lysine Acetyltransferase 5 , Protein Stability , Ubiquitination
9.
Am J Respir Crit Care Med ; 175(2): 174-83, 2007 Jan 15.
Article in English | MEDLINE | ID: mdl-17082494

ABSTRACT

RATIONALE: The respiratory tract is constantly exposed to airborne microorganisms. Nevertheless, normal airways remain sterile without recruiting phagocytes. This innate immune activity has been attributed to mucociliary clearance and antimicrobial polypeptides of airway surface liquid. Defective airway immunity characterizes cystic fibrosis (CF), a disease caused by mutations in the CF transmembrane conductance regulator, a chloride channel. The pathophysiology of defective immunity in CF remains to be elucidated. OBJECTIVE: We investigated the ability of non-CF and CF airway epithelia to kill bacteria through the generation of reactive oxygen species (ROS). METHODS: ROS production and ROS-mediated bactericidal activity were determined on the apical surfaces of human and rat airway epithelia and on cow tracheal explants. MEASUREMENTS AND MAIN RESULTS: Dual oxidase enzyme of airway epithelial cells generated sufficient H(2)O(2) to support production of bactericidal hypothiocyanite (OSCN(-)) in the presence of airway surface liquid components lactoperoxidase and thiocyanate (SCN(-)). This OSCN(-) formation eliminated Staphylococcus aureus and Pseudomonas aeruginosa on airway mucosal surfaces, whereas it was nontoxic to the host. In contrast to normal epithelia, CF epithelia failed to secrete SCN(-), thereby rendering the oxidative antimicrobial system inactive. CONCLUSIONS: These data indicate a novel innate defense mechanism of airways that kills bacteria via ROS and suggest a new cellular and molecular basis for defective airway immunity in CF.


Subject(s)
Cystic Fibrosis/immunology , Flavoproteins/metabolism , Lactoperoxidase/metabolism , Lung Diseases/immunology , Respiratory Mucosa/immunology , Animals , Cattle , Cells, Cultured , Cystic Fibrosis/enzymology , Cystic Fibrosis/microbiology , Dual Oxidases , Flavoproteins/analysis , Flavoproteins/genetics , Humans , Hydrogen Peroxide/metabolism , Immunity, Innate/genetics , Immunity, Mucosal , Lactoperoxidase/analysis , Lactoperoxidase/genetics , Lung Diseases/enzymology , Lung Diseases/microbiology , Pseudomonas aeruginosa/immunology , RNA, Small Interfering/pharmacology , Rats , Reactive Oxygen Species/metabolism , Respiratory Mucosa/enzymology , Respiratory Mucosa/microbiology , Staphylococcus aureus/immunology , Thiocyanates/metabolism , Trachea/cytology , Trachea/immunology , Trachea/microbiology
10.
J Biol Chem ; 280(8): 6716-20, 2005 Feb 25.
Article in English | MEDLINE | ID: mdl-15596440

ABSTRACT

Interaction of p50 Rho GTPase-activating protein (p50RhoGAP) with Rho family small GTPases was investigated in a yeast two-hybrid system, by radioactive GAP assay, and in a Rac-regulated enzymatic reaction, through superoxide production by the phagocytic NADPH oxidase. The yeast two-hybrid system revealed an interaction between the C-terminal GAP domain and the N-terminal part of p50RhoGAP. The first 48 amino acids play a special role both in the stabilization of the intramolecular interaction and in recognition of the prenyl tail of small GTPases. The GAP assay and the NADPH oxidase activity indicate that the GTPase-activating effect of full-length p50RhoGAP is lower on non-prenylated than on prenylated small GTPase. Removal of amino acids 1-48 and 169-197 of p50RhoGAP increases the GAP effect on non-prenylated Rac, whereas prenylated Rac reacts equally well with the full-length and the truncated proteins. We suggest that p50RhoGAP is in an autoinhibited conformation stabilized by the stretches 1-48 and 169-197 and the prenyl group of the small GTPase plays a role in releasing this intramolecular restraint.


Subject(s)
GTPase-Activating Proteins/antagonists & inhibitors , Monomeric GTP-Binding Proteins/physiology , Protein Prenylation , Amino Acid Sequence , Animals , Cell-Free System , GTPase-Activating Proteins/metabolism , Humans , Monomeric GTP-Binding Proteins/metabolism , NADPH Oxidases/metabolism , Protein Binding , Protein Structure, Tertiary , Superoxides/metabolism , Two-Hybrid System Techniques , rac1 GTP-Binding Protein/metabolism
11.
Biochim Biophys Acta ; 1688(3): 197-203, 2004 Apr 05.
Article in English | MEDLINE | ID: mdl-15062869

ABSTRACT

We reported earlier that monocytes and macrophages from patients with type I Gaucher disease have a decreased capacity to generate superoxide anion (O(2)(-)) on stimulation with opsonized S. aureus or formyl-methionyl-leucyl-phenylalanine. In this study, various forms of the cell-free assay system were used to probe the hypothesis that glucocerebroside (GC) accumulating in Gaucher patients' phagocytes may interfere with the activation of NADPH oxidase. Xanthine/xanthine oxidase assay was applied to explore the possibility that GC may scavenge O(2)(-). We found that addition of GC to the crude, semirecombinant or fully purified cell-free systems inhibited activation of NADPH oxidase in a concentration-dependent manner. The inhibitory effect of GC could be overcome by increased concentrations of p47(phox) and p67(phox). In contrast, O(2)(-) generation was not decreased by GC added to the assembled, catalytically active enzyme complex. In the xanthine/xanthine oxidase system, GC had no effect on the generation of O(2)(-). These data indicate that assembly of the respiratory burst oxidase of phagocytic cells may be a possible target of the pathologic actions of GC.


Subject(s)
Glucosylceramides/pharmacology , NADPH Oxidases/blood , Neutrophils/enzymology , Cell-Free System , Cloning, Molecular , Enzyme Activation/drug effects , Humans , Kinetics , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/drug effects , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism , Superoxides/blood , Xanthine Oxidase/blood
12.
J Biol Chem ; 279(18): 18583-91, 2004 Apr 30.
Article in English | MEDLINE | ID: mdl-14982937

ABSTRACT

NADPH oxidase 5 (NOX5) is a homologue of the gp91(phox) subunit of the phagocyte NADPH oxidase. NOX5 is expressed in lymphoid organs and testis and distinguished from the other NADPH oxidases by its unique N terminus, which contains three canonical EF-hands, Ca(2+)-binding domains. Upon heterologous expression, NOX5 was shown to generate superoxide in response to intracellular Ca(2+) elevations. In this study, we have analyzed the mechanism of Ca(2+) activation of NOX5. In a cell-free system, Ca(2+) elevations triggered superoxide production by NOX5 (K(m) = 1.06 microm) in an NADPH- and FAD-dependent but cytosol-independent manner. That result indicated a role for the N-terminal EF-hands in NOX5 activation. Therefore, we generated recombinant proteins of NOX5 N terminus and investigated their interactions with Ca(2+). Flow dialysis experiments showed that NOX5 N terminus contained four Ca(2+)-binding sites and allowed us to define the hitherto unidentified fourth, non-canonical EF-hand. The EF-hands of NOX5 formed two pairs: the very N-terminal pair had relatively low affinity for Ca(2+), whereas the more C-terminal pair bound Ca(2+) with high affinity. Ca(2+) binding caused a marked conformation change in the N terminus, which exposed its hydrophobic core, and became able to bind melittin, a model peptide for calmodulin targets. Using a pull-down assay, we demonstrate that the regulatory N terminus and the catalytic C terminus of NOX5 interact in a Ca(2+)-dependent way. Our results indicate that the Ca(2+)-induced conformation change of NOX5 N terminus led to enzyme activation through an intra-molecular interaction. That represents a novel mechanism of activation among NAD(P)H oxidases and Ca(2+)-activated enzymes.


Subject(s)
Calcium/pharmacology , Membrane Proteins/metabolism , NADPH Oxidases/metabolism , Amino Acid Sequence , Binding Sites , Cell Line , Cell-Free System , Enzyme Activation/drug effects , Humans , Kinetics , Melitten/metabolism , Melitten/pharmacology , NADPH Oxidase 5 , Peptide Fragments/metabolism , Protein Conformation , Superoxides/metabolism
13.
Biochemistry ; 41(34): 10710-6, 2002 Aug 27.
Article in English | MEDLINE | ID: mdl-12186557

ABSTRACT

The aim of the present study was to investigate possible mechanisms that could be involved in the deactivation of the assembled, catalytically active NADPH oxidase of phagocytic cells and thereby lead to termination of O(2)(.-) production. Our major findings are the following: (1) Addition of GDP to the active oxidase is able to reduce O(2)(.-) production both in the fully purified and in a semi-recombinant cell-free activation system. (2) p67(phox) inhibits GTP hydrolysis on Rac whereas p47(phox) has no effect on Rac GTPase activity. (3) Soluble regulatory proteins (GTPase activating protein, guanine nucleotide dissociation inhibitor, and the Rac-binding domain of the target protein p21-activated kinase) inhibit activation of the NADPH oxidase but have no effect on electron transfer via the assembled enzyme complex. (4) Membrane-associated GTPase activating proteins (GAPs) have access also to the assembled, catalytically active oxidase. Taken together, we propose that the GTP-bound active form of Rac is required for sustained enzyme activity and that membrane-localized GAPs have a role in the deactivation of NADPH oxidase.


Subject(s)
NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/metabolism , Neutrophils/enzymology , rac GTP-Binding Proteins/metabolism , Catalysis/drug effects , Enzyme Activation/drug effects , Guanosine Diphosphate/metabolism , Guanosine Diphosphate/pharmacology , Guanosine Triphosphate/metabolism , Humans , Phagocytosis , Phosphoproteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL