Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Life Sci Alliance ; 6(4)2023 04.
Article in English | MEDLINE | ID: mdl-36697252

ABSTRACT

Resident tissue macrophages are organ-specialized phagocytes responsible for the maintenance and protection of tissue homeostasis. It is well established that tissue diversity is reflected by the heterogeneity of resident tissue macrophage origin and phenotype. However, much less is known about tissue-specific phagocytic and proteolytic macrophage functions. Here, using a quantitative proteomics approach, we identify cathepsins as key determinants of phagosome maturation in primary peritoneum-, lung-, and brain-resident macrophages. The data further uncover cathepsin K (CtsK) as a molecular marker for lung phagosomes required for intracellular protein and collagen degradation. Pharmacological blockade of CtsK activity diminished phagosomal proteolysis and collagenolysis in lung-resident macrophages. Furthermore, profibrotic TGF-ß negatively regulated CtsK-mediated phagosomal collagen degradation independently from classical endocytic-proteolytic pathways. In humans, phagosomal CtsK activity was reduced in COPD lung macrophages and non-COPD lung macrophages exposed to cigarette smoke extract. Taken together, this study provides a comprehensive map of how peritoneal, lung, and brain tissue environment shapes phagosomal composition, revealing CtsK as a key molecular determinant of lung phagosomes contributing to phagocytic collagen clearance in lungs.


Subject(s)
Cathepsin K , Macrophages , Phagosomes , Humans , Cathepsin K/metabolism , Collagen/metabolism , Lung , Macrophages/metabolism , Phagosomes/metabolism
2.
J Hepatol ; 76(5): 1001-1012, 2022 05.
Article in English | MEDLINE | ID: mdl-34942286

ABSTRACT

BACKGROUND & AIMS: Obesity-associated inflammation is a key player in the pathogenesis of non-alcoholic fatty liver disease (NAFLD). However, the role of macrophage scavenger receptor 1 (MSR1, CD204) remains incompletely understood. METHODS: A total of 170 NAFLD liver biopsies were processed for transcriptomic analysis and correlated with clinicopathological features. Msr1-/- and wild-type mice were subjected to a 16-week high-fat and high-cholesterol diet. Mice and ex vivo human liver slices were treated with a monoclonal antibody against MSR1. Genetic susceptibility was assessed using genome-wide association study data from 1,483 patients with NAFLD and 430,101 participants of the UK Biobank. RESULTS: MSR1 expression was associated with the occurrence of hepatic lipid-laden foamy macrophages and correlated with the degree of steatosis and steatohepatitis in patients with NAFLD. Mice lacking Msr1 were protected against diet-induced metabolic disorder, showing fewer hepatic foamy macrophages, less hepatic inflammation, improved dyslipidaemia and glucose tolerance, and altered hepatic lipid metabolism. Upon induction by saturated fatty acids, MSR1 induced a pro-inflammatory response via the JNK signalling pathway. In vitro blockade of the receptor prevented the accumulation of lipids in primary macrophages which inhibited the switch towards a pro-inflammatory phenotype and the release of cytokines such as TNF-ɑ. Targeting MSR1 using monoclonal antibody therapy in an obesity-associated NAFLD mouse model and human liver slices resulted in the prevention of foamy macrophage formation and inflammation. Moreover, we identified that rs41505344, a polymorphism in the upstream transcriptional region of MSR1, was associated with altered serum triglycerides and aspartate aminotransferase levels in a cohort of over 400,000 patients. CONCLUSIONS: Taken together, our data suggest that MSR1 plays a critical role in lipid-induced inflammation and could thus be a potential therapeutic target for the treatment of NAFLD. LAY SUMMARY: Non-alcoholic fatty liver disease (NAFLD) is a chronic disease primarily caused by excessive consumption of fat and sugar combined with a lack of exercise or a sedentary lifestyle. Herein, we show that the macrophage scavenger receptor MSR1, an innate immune receptor, mediates lipid uptake and accumulation in Kupffer cells, resulting in liver inflammation and thereby promoting the progression of NAFLD in humans and mice.


Subject(s)
Non-alcoholic Fatty Liver Disease , Animals , Antibodies, Monoclonal , Diet, High-Fat/adverse effects , Genome-Wide Association Study , Humans , Inflammation/metabolism , Lipids , Liver/pathology , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Obesity/metabolism
3.
Respir Res ; 22(1): 172, 2021 Jun 07.
Article in English | MEDLINE | ID: mdl-34098956

ABSTRACT

BACKGROUND: Patients with chronic obstructive pulmonary disease (COPD) frequently suffer from chronic bronchitis (CB) and display steroid-resistant inflammation with increased sputum neutrophils and macrophages. Recently, a causal link between mucus hyper-concentration and disease progression of CB has been suggested. METHODS: In this study, we have evaluated the steroid sensitivity of purified, patient-derived sputum and alveolar macrophages and used a novel mechanistic cross-talk assay to examine how macrophages and bronchial epithelial cells cross-talk to regulate MUC5B production. RESULTS: We demonstrate that sputum plug macrophages isolated from COPD patients with chronic bronchitis (COPD/CB) are chronically activated and only partially respond to ex vivo corticosteroid treatment compared to alveolar macrophages isolated from lung resections. Further, we show that pseudo-stratified bronchial epithelial cells grown in air-liquid-interface are inert to direct bacterial lipopolysaccharide stimulation and that macrophages are able to relay this signal and activate the CREB/AP-1 transcription factor complex and subsequent MUC5B expression in epithelial cells through a soluble mediator. Using recombinant protein and neutralizing antibodies, we identified a key role for TNFα in this cross-talk. CONCLUSIONS: For the first time, we describe ex vivo pharmacology in purified human sputum macrophages isolated from chronic bronchitis COPD patients and identify a possible basis for the steroid resistance frequently seen in this population. Our data pinpoint a critical role for chronically activated sputum macrophages in perpetuating TNFα-dependent signals driving mucus hyper-production. Targeting the chronically activated mucus plug macrophage phenotype and interfering with aberrant macrophage-epithelial cross-talk may provide a novel strategy to resolve chronic inflammatory lung disease.


Subject(s)
Bronchitis, Chronic/metabolism , Drug Resistance , Glucocorticoids/pharmacology , Lung/pathology , Macrophages, Alveolar/metabolism , Mucin-5B/biosynthesis , Mucus/metabolism , Aged , Biomarkers/metabolism , Bronchitis, Chronic/drug therapy , Bronchitis, Chronic/pathology , Disease Progression , Female , Follow-Up Studies , Humans , Inflammation/metabolism , Leukocyte Count , Macrophages, Alveolar/pathology , Male , Middle Aged
4.
PLoS One ; 15(12): e0243807, 2020.
Article in English | MEDLINE | ID: mdl-33332401

ABSTRACT

Induced pluripotent stem cells (iPSC) offer the possibility to generate diverse disease-relevant cell types, from any genetic background with the use of cellular reprogramming and directed differentiation. This provides a powerful platform for disease modeling, drug screening and cell therapeutics. The critical question is how the differentiated iPSC-derived cells translate to their primary counterparts. Our refinement of a published differentiation protocol produces a CD14+ monocytic lineage at a higher yield, in a smaller format and at a lower cost. These iPSC-derived monocytes can be further differentiated into macrophages or dendritic cells (DC), both with similar morphological and functional profiles as compared to their primary counterparts. Transcriptomic analysis of iPSC-derived cells at different stages of differentiation as well as comparison to their blood-derived counterparts demonstrates a complete switch of iPSCs to cells expressing a monocyte, macrophage or DC specific gene profile. iPSC-derived macrophages respond to LPS treatment by inducing expression of classic macrophage pro-inflammatory response markers. Interestingly, though iPSC-derived DC show similarities to monocyte derived DC, they are more similar transcriptionally to a newly described subpopulation of AXL+ DC. Thus, our study provides a detailed and accurate profile of iPSC-derived monocytic lineage cells.


Subject(s)
Dendritic Cells/cytology , Induced Pluripotent Stem Cells/cytology , Macrophages/cytology , Transcription, Genetic , Cell Differentiation , Cell Lineage , Dendritic Cells/metabolism , Humans , Macrophages/metabolism
5.
PLoS One ; 8(5): e64721, 2013.
Article in English | MEDLINE | ID: mdl-23700488

ABSTRACT

Nuclear receptor subfamily 1, group H, member 4 (Nr1h4, FXR) is a bile acid activated nuclear receptor mainly expressed in the liver, intestine, kidney and adrenal glands. Upon activation, the primary function is to suppress cholesterol 7 alpha-hydroxylase (Cyp7a1), the rate-limiting enzyme in the classic or neutral bile acid synthesis pathway. In the present study, a novel Fxr deficient mouse line was created and studied with respect to metabolism and liver function in ageing mice fed chow diet. The Fxr deficient mice were similar to wild type mice in terms of body weight, body composition, energy intake and expenditure as well as behaviours at a young age. However, from 15 weeks of age and onwards, the Fxr deficient mice had almost no body weight increase up to 39 weeks of age mainly because of lower body fat mass. The lower body weight gain was associated with increased energy expenditure that was not compensated by increased food intake. Fasting levels of glucose and insulin were lower and glucose tolerance was improved in old and lean Fxr deficient mice. However, the Fxr deficient mice displayed significantly increased liver weight, steatosis, hepatocyte ballooning degeneration and lobular inflammation together with elevated plasma levels of ALT, bilirubin and bile acids, findings compatible with non-alcoholic steatohepatitis (NASH) and cholestasis. In conclusion, ageing Fxr deficient mice display late onset leanness associated with elevated energy expenditure and improved glucose control but develop severe NASH-like liver pathology.


Subject(s)
Energy Metabolism , Fatty Liver/metabolism , Glucose/metabolism , Receptors, Cytoplasmic and Nuclear/deficiency , Adipose Tissue, White/metabolism , Adipose Tissue, White/pathology , Aging , Animals , Blood Glucose , Body Composition , Body Weight , Cell Size , Energy Intake , Female , Lipid Metabolism , Liver/metabolism , Liver/pathology , Liver/physiopathology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Motor Activity , Non-alcoholic Fatty Liver Disease , Organ Size , Receptors, Cytoplasmic and Nuclear/genetics , Skin/pathology , Triglycerides/metabolism
6.
Am J Physiol Endocrinol Metab ; 300(1): E211-20, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20959533

ABSTRACT

Free fatty acid receptor 2 (Ffar2), also known as GPR43, is activated by short-chain fatty acids (SCFA) and expressed in intestine, adipocytes, and immune cells, suggesting involvement in lipid and immune regulation. In the present study, Ffar2-deficient mice (Ffar2-KO) were given a high-fat diet (HFD) or chow diet and studied with respect to lipid and energy metabolism. On a HFD, Ffar2-KO mice had lower body fat mass and increased lean body mass. The changed body composition was accompanied by improved glucose control and lower HOMA index, indicating improved insulin sensitivity in Ffar2-KO mice. Moreover, the Ffar2-KO mice had higher energy expenditure accompanied by higher core body temperature and increased food intake. The liver weight and content of triglycerides as well as plasma levels of cholesterol were lower in the Ffar2-KO mice fed a HFD. A histological examination unveiled decreased lipid interspersed in brown adipose tissue of the Ffar2-KO mice. Interestingly, no significant differences in white adipose tissue (WAT) cell size were observed, but significantly lower macrophage content was detected in WAT from HFD-fed Ffar2-KO compared with wild-type mice. In conclusion, Ffar2 deficiency protects from HFD-induced obesity and dyslipidemia at least partly via increased energy expenditure.


Subject(s)
Dietary Fats/administration & dosage , Hyperglycemia/prevention & control , Hypoglycemia/prevention & control , Obesity/prevention & control , Receptors, G-Protein-Coupled/physiology , Adipose Tissue, Brown/pathology , Adipose Tissue, White/pathology , Animals , Body Composition , Body Temperature Regulation , Diet , Dyslipidemias/blood , Dyslipidemias/prevention & control , Energy Metabolism , Fatty Liver/metabolism , Fatty Liver/prevention & control , Homeostasis , Insulin Resistance , Macrophages/metabolism , Male , Mice , Mice, 129 Strain , Mice, Knockout , Obesity/blood , Obesity/genetics , Obesity/pathology , Receptors, G-Protein-Coupled/genetics
7.
BMC Cancer ; 10: 249, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20515481

ABSTRACT

BACKGROUND: The FUS-DDIT3 fusion oncogene encodes an abnormal transcription factor that has a causative role in the development of myxoid/round-cell liposarcomas (MLS/RCLS). We have previously identified FLT1 (VEGFR1) as a candidate downstream target gene of FUS-DDIT3. The aim of this study was to investigate expression of FLT1 and its ligands in MLS cells. METHODS: HT1080 human fibrosarcoma cells were transiently transfected with FUS-DDIT3-GFP variant constructs and FLT1 expression was measured by quantitative real-time PCR. In addition, FLT1, PGF, VEGFA and VEGFB expression was measured in MLS/RCLS cell lines, MLS/RCLS tumors and in normal adiopocytes. We analyzed nine cases of MLS/RCLS and one cell line xenografted in mice for FLT1 protein expression using immunohistochemistry. MLS/RCLS cell lines were also analyzed for FLT1 by immunofluorescence and western blot. MLS/RCLS cell lines were additionally treated with FLT1 tyrosine kinase inhibitors and assayed for alterations in proliferation rate. RESULTS: FLT1 expression was dramatically increased in transfected cells stably expressing FUS-DDIT3 and present at high levels in cell lines derived from MLS. The FLT1 protein showed a strong nuclear expression in cells of MLS tissue as well as in cultured MLS cells, which was confirmed by cellular fractionation. Tissue array analysis showed a nuclear expression of the FLT1 protein also in several other tumor and normal cell types including normal adipocytes. The FLT1 ligand coding gene PGF was highly expressed in cultured MLS cells compared to normal adipocytes while the other ligand genes VEGFA and VEGFB were expressed to lower levels. A more heterogeneous expression pattern of these genes were observed in tumor samples. No changes in proliferation rate of MLS cells were detected at concentrations for which the kinase inhibitors have shown specific inhibition of FLT1. CONCLUSIONS: Our results imply that FLT1 is induced as an indirect downstream effect of FUS-DDIT3 expression in MLS. This could be a consequence of the ability of FUS-DDIT3 to hijack parts of normal adipose tissue development and reprogram primary cells to a liposarcoma-like phenotype. The findings of nuclear FLT1 protein and expression of corresponding ligands in MLS and normal tissues may have implications for tissue homeostasis and tumor development through auto- or intracrine signaling.


Subject(s)
Cell Nucleus/metabolism , Fibrosarcoma/metabolism , Liposarcoma, Myxoid/metabolism , Oncogene Proteins, Fusion/metabolism , Pregnancy Proteins/metabolism , RNA-Binding Protein FUS/metabolism , Signal Transduction , Transcription Factor CHOP/metabolism , Vascular Endothelial Growth Factor Receptor-1/metabolism , Animals , Blotting, Western , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/pathology , Cell Proliferation , Dose-Response Relationship, Drug , Fibrosarcoma/genetics , Fibrosarcoma/pathology , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Ligands , Liposarcoma, Myxoid/genetics , Liposarcoma, Myxoid/pathology , Mice , Mice, SCID , Neoplasm Transplantation , Oncogene Proteins, Fusion/genetics , Placenta Growth Factor , Pregnancy Proteins/genetics , Protein Kinase Inhibitors/pharmacology , RNA Interference , RNA-Binding Protein FUS/genetics , Recombinant Fusion Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factor CHOP/genetics , Transfection , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor B/metabolism , Vascular Endothelial Growth Factor Receptor-1/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-1/genetics
8.
Biochem Biophys Res Commun ; 363(3): 633-8, 2007 Nov 23.
Article in English | MEDLINE | ID: mdl-17904108

ABSTRACT

In this study, mice carrying a disrupted gene encoding GPR10 (GPR10 KO) were studied to elucidate the function and importance of this receptor regarding metabolism. Female and male GPR10 KO mice had higher body weight after 11 and 15 weeks of age, respectively. The increased body weight was a result of increased fat mass. The obesity was much more pronounced in female mice, which also had a significant decrease in energy expenditure. In correlation to obesity, higher plasma levels of leptin, total cholesterol, and fractions of LDL and HDL were found in GPR10 KO compared to WT mice. Interestingly, GPR10 KO female mice had decreased relative food intake in correlation to higher hypothalamic expression levels of the anorexic signals CRH and POMC. In conclusion, female mice deficient of the gene encoding GPR10 develop higher body weight and obesity due to lower energy expenditure.


Subject(s)
Energy Metabolism/physiology , Obesity/physiopathology , Receptors, G-Protein-Coupled/physiology , Adipose Tissue/metabolism , Animals , Blood Glucose/metabolism , Body Temperature , Body Weight/genetics , Body Weight/physiology , Calorimetry/methods , Eating/genetics , Eating/physiology , Energy Metabolism/genetics , Feces/chemistry , Female , Genotype , Lipoproteins, HDL/blood , Lipoproteins, LDL/blood , Male , Mice , Mice, Knockout , Obesity/genetics , Receptors, G-Protein-Coupled/deficiency , Receptors, G-Protein-Coupled/genetics , Sex Factors , Time Factors
9.
Am J Pathol ; 168(5): 1642-53, 2006 May.
Article in English | MEDLINE | ID: mdl-16651630

ABSTRACT

Myxoid/round cell liposarcoma (MLS/RCLS) is the most common subtype of liposarcoma. Most MLS/RCLS carry a t(12;16) translocation, resulting in a FUS-DDIT3 fusion gene. We investigated the role of the FUS-DDIT3 fusion in the development of MLS/RCLS in FUS-DDIT3- and DDIT3-transfected human HT1080 sarcoma cells. Cells expressing FUS-DDIT3 and DDIT3 grew as liposarcomas in severe combined immunodeficient mice and exhibited a capillary network morphology that was similar to networks of MLS/RCLS. Microarray-based comparison of HT1080, the transfected cells, and an MLS/RCLS-derived cell line showed that the FUS-DDIT3- and DDIT3-transfected variants shifted toward an MLS/RCLS-like expression pattern. DDIT3-transfected cells responded in vitro to adipogenic factors by accumulation of fat and transformation to a lipoblast-like morphology. In conclusion, because the fusion oncogene FUS-DDIT3 and the normal DDIT3 induce a liposarcoma phenotype when expressed in a primitive sarcoma cell line, MLS/RCLS may develop from cell types other than preadipocytes. This may explain the preferential occurrence of MLS/RCLS in nonadipose tissues. In addition, development of lipoblasts and the typical MLS/RCLS capillary network could be an effect of the DDIT3 transcription factor partner of the fusion oncogene.


Subject(s)
Fibrosarcoma/blood supply , Gene Expression Regulation, Neoplastic , Liposarcoma, Myxoid/blood supply , RNA-Binding Protein FUS/physiology , Transcription Factor CHOP/physiology , Adipogenesis , Animals , Cluster Analysis , Down-Regulation , Female , Fibrosarcoma/metabolism , Humans , Liposarcoma, Myxoid/metabolism , Mice , Mice, SCID , Oligonucleotide Array Sequence Analysis , Oncogene Proteins, Fusion/physiology , RNA-Binding Protein FUS/genetics , RNA-Binding Protein FUS/metabolism , Transcription Factor CHOP/metabolism , Transfection , Up-Regulation
10.
Int J Cancer ; 115(4): 556-60, 2005 Jul 01.
Article in English | MEDLINE | ID: mdl-15688424

ABSTRACT

The myxoid/round cell liposarcoma oncogene FUS-DDIT3 is the result of a translocation derived gene fusion between the splicing factor FUS and DDIT3. In order to investigate the downstream targets of DDIT3, and the transforming effects of the FUS-DDIT3 fusion protein, we have introduced DDIT3-GFP and FUS-DDIT3-GFP constructs into a human fibrosarcoma cell line. The gene expression profiles of stable transfectants were compared to the original fibrosarcoma cell line by microarray analysis. We here report that the NFkappaB and C/EBP beta controlled gene IL6 is upregulated in DDIT3- and FUS-DDIT3-expressing fibrosarcoma cell lines and in myxoid liposarcoma cell lines. Strong expression of the tumor associated multifunctional cytokine interleukin 6 was confirmed both at mRNA and protein level. Knockdown experiments using siRNA against CEBPB transcripts showed that the effect of FUS-DDIT3 on IL6 expression is C/EBP beta dependent. Chromatin immunoprecipitation revealed direct interaction between the IL6 promoter and the C/EBP beta protein. In addition, the effect of DDIT3 and FUS-DDIT3 on the expression of other acute phase genes was examined using real-time PCR. We demonstrate for the first time that DDIT3 and FUS-DDIT3 show opposite transcriptional regulation of IL8 and suggest that FUS-DDIT3 may affect the synergistic activation of promoters regulated by C/EBP beta and NFkappaB.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/physiology , CCAAT-Enhancer-Binding Proteins/genetics , Fibrosarcoma/genetics , Interleukin-6/genetics , Liposarcoma, Myxoid/genetics , Oncogene Proteins, Fusion/genetics , RNA-Binding Protein FUS/genetics , Transcription Factors/genetics , Animals , Cell Line, Tumor , DNA Primers , Fibrosarcoma/immunology , Humans , Interleukin-6/blood , Liposarcoma, Myxoid/blood , Liposarcoma, Myxoid/immunology , Mice , Mice, SCID , NF-kappa B/metabolism , Polymerase Chain Reaction , Promoter Regions, Genetic , RNA, Small Interfering/genetics , Transcription Factor CHOP , Transplantation, Heterologous
11.
Int J Oncol ; 25(5): 1349-55, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15492825

ABSTRACT

Myxoid/round cell liposarcomas (MLS/RCLS) are characterized by chromosome translocations that result in formation of FUS-CHOP or EWSR1-CHOP fusion oncogenes. More than 95% of the tumors carry one of these fusion genes. FUS-CHOP transforms 3T3 cells and causes MLS/RCLS-like tumors in transgenic mice. The fusion oncoproteins act as abnormal transcription factors and are believed to induce abnormal expression of growth controlling genes as part of their transforming activities. The aim of this study was to search for recurrent abnormal expression patterns of cell cycle regulating proteins and growth factor receptors. A series of 14 MLS/RCLS, 2 MLS/RCLS derived cell lines and a FUS-CHOP transfected human sarcoma cell line were analyzed using immunohistochemistry, Western blotting, and cDNA microarray based screening. The results revealed a highly abnormal expression pattern of several growth controlling proteins. The G1 cyclins D1 and E and their associated kinases CDK4 and CDK2 were strongly overexpressed in all of the tumors. High expression levels were also found for Cdk4/6 inhibitor P16 and CDK2 inhibitors P27 and P57. The growth factor tyrosine kinase receptors PDGFRB and EGFR were present in most cells of all investigated tumors. We conclude that deregulation of G1 controlling proteins is common in MLS/RCLS and that aberrant expression of these proteins is of importance in the pathogenesis of this tumor type.


Subject(s)
CCAAT-Enhancer-Binding Proteins/genetics , Cell Cycle Proteins/biosynthesis , Cell Cycle Proteins/pharmacology , Gene Expression Profiling , Liposarcoma, Myxoid/pathology , Oncogene Proteins, Fusion/genetics , RNA-Binding Protein FUS/genetics , Adult , Aged , Blotting, Western , Cell Cycle Proteins/genetics , Female , Humans , Immunohistochemistry , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , Transcription Factor CHOP , Transfection , Tumor Cells, Cultured
12.
Exp Cell Res ; 278(2): 125-32, 2002 Aug 15.
Article in English | MEDLINE | ID: mdl-12169268

ABSTRACT

The myxoid/round cell liposarcoma oncogene TLS-CHOP belongs to a growing family of tumor type specific fusion genes generated by chromosome translocations. We have recently found that the TLS-CHOP fusion protein is localized to well-defined nuclear structures, a pattern distinct from normal TLS or CHOP cellular distribution. Since location and function are intimately coupled in the organized nucleus, the aberrant localization of the fusion protein most certainly reflects the oncogenic activities of TLS-CHOP. We have investigated the role of the functionally unknown, SYGQ-rich, TLS N-terminal in the localization of TLS-CHOP to nuclear structures. Here, we report the temperature-dependent localization of TLS-CHOP to splicing factor compartments and association with Cajal bodies. Further, mutational analysis of the N-terminal part of green fluorescent protein-tagged TLS-CHOP identifies a region within the N-terminal required for colocalization with the splicing factor SC-35.


Subject(s)
CCAAT-Enhancer-Binding Proteins/metabolism , Cell Compartmentation/physiology , Cell Nucleus/metabolism , Oncogene Proteins, Fusion/metabolism , RNA-Binding Protein FUS , Ribonucleoproteins , Amino Acid Sequence , CCAAT-Enhancer-Binding Proteins/genetics , Cell Nucleus/ultrastructure , Humans , Mutagenesis, Site-Directed , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Nuclear Proteins/metabolism , Oncogene Proteins, Fusion/genetics , Serine-Arginine Splicing Factors , Spliceosomes/metabolism , Temperature , Transcription Factor CHOP , Transcription Factors/metabolism , Transfection , Tumor Cells, Cultured
13.
Int J Cancer ; 97(4): 446-50, 2002 Feb 01.
Article in English | MEDLINE | ID: mdl-11802205

ABSTRACT

CHOP in 12q13, also called GADD153 or DDIT3, encodes a transcription factor of the C/EBP type. As a result of t(12;16) translocations, CHOP is rearranged and fused to TLS in 16p11 in about 90% of myxoid liposarcomas/round cell liposarcomas (MLS/RCLS). The TLS-CHOP protein consists of the N-terminal half of TLS juxtaposed to the N-terminal of the entire CHOP. It is capable of forming dimers with the natural dimer partners of CHOP. Here we report that recombinant TLS-CHOP-green fluorescence protein localizes to nuclear structures, similar to, but distinct from, PML nuclear bodies. The TLS-CHOP-green fluorescent protein nuclear structures are resistant to high salt concentration and nuclease treatment. Transfection of TLS-CHOP to normal fibroblasts causes a rapid down regulation and relocation of PML nuclear bodies. An abnormal extra nuclear localization of PML bodies was also found in TLS-CHOP carrying cell lines established from myxoid liposarcomas. Transfection of TLS-CHOP induced a rapid disappearance of PCNA. TLS-CHOP may disturb the nuclear machinery by binding and sequestering important factors from their natural sites.


Subject(s)
CCAAT-Enhancer-Binding Proteins/metabolism , Cell Nucleus/chemistry , Liposarcoma, Myxoid/genetics , Nuclear Proteins , Oncogene Proteins, Fusion/metabolism , RNA-Binding Protein FUS , Animals , COS Cells , Cell Division , Cell Fractionation , Cell Nucleus/ultrastructure , Chlorocebus aethiops , Chromosomes, Human, Pair 12/genetics , Chromosomes, Human, Pair 12/ultrastructure , Chromosomes, Human, Pair 16/genetics , Chromosomes, Human, Pair 16/ultrastructure , Cytoplasm/chemistry , Dimerization , Fibroblasts/metabolism , Fibrosarcoma/pathology , Genes, Reporter , Green Fluorescent Proteins , Humans , Ki-67 Antigen/analysis , Leukemia, Promyelocytic, Acute/genetics , Liposarcoma, Myxoid/ultrastructure , Luminescent Proteins/analysis , Luminescent Proteins/genetics , Neoplasm Proteins/analysis , Polymerase Chain Reaction , Proliferating Cell Nuclear Antigen/analysis , Promyelocytic Leukemia Protein , Recombinant Fusion Proteins/metabolism , Transcription Factor CHOP , Transcription Factors/analysis , Transfection , Translocation, Genetic , Tumor Suppressor Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...