Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
J Hepatol ; 76(5): 1001-1012, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-34942286

RESUMO

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.


Assuntos
Hepatopatia Gordurosa não Alcoólica , Animais , Anticorpos Monoclonais , Dieta Hiperlipídica/efeitos adversos , Estudo de Associação Genômica Ampla , Humanos , Inflamação/metabolismo , Lipídeos , Fígado/patologia , Camundongos , Camundongos Endogâmicos C57BL , Hepatopatia Gordurosa não Alcoólica/genética , Hepatopatia Gordurosa não Alcoólica/metabolismo , Obesidade/metabolismo
2.
Respir Res ; 22(1): 172, 2021 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-34098956

RESUMO

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.


Assuntos
Bronquite Crônica/metabolismo , Resistência a Medicamentos , Glucocorticoides/farmacologia , Pulmão/patologia , Macrófagos Alveolares/metabolismo , Mucina-5B/biossíntese , Muco/metabolismo , Idoso , Biomarcadores/metabolismo , Bronquite Crônica/tratamento farmacológico , Bronquite Crônica/patologia , Progressão da Doença , Feminino , Seguimentos , Humanos , Inflamação/metabolismo , Contagem de Leucócitos , Macrófagos Alveolares/patologia , Masculino , Pessoa de Meia-Idade
3.
Life Sci Alliance ; 6(4)2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36697252

RESUMO

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.


Assuntos
Catepsina K , Macrófagos , Fagossomos , Humanos , Catepsina K/metabolismo , Colágeno/metabolismo , Pulmão , Macrófagos/metabolismo , Fagossomos/metabolismo
4.
Am J Physiol Endocrinol Metab ; 300(1): E211-20, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20959533

RESUMO

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.


Assuntos
Gorduras na Dieta/administração & dosagem , Hiperglicemia/prevenção & controle , Hipoglicemia/prevenção & controle , Obesidade/prevenção & controle , Receptores Acoplados a Proteínas G/fisiologia , Tecido Adiposo Marrom/patologia , Tecido Adiposo Branco/patologia , Animais , Composição Corporal , Regulação da Temperatura Corporal , Dieta , Dislipidemias/sangue , Dislipidemias/prevenção & controle , Metabolismo Energético , Fígado Gorduroso/metabolismo , Fígado Gorduroso/prevenção & controle , Homeostase , Resistência à Insulina , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Knockout , Obesidade/sangue , Obesidade/genética , Obesidade/patologia , Receptores Acoplados a Proteínas G/genética
5.
BMC Cancer ; 10: 249, 2010 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-20515481

RESUMO

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.


Assuntos
Núcleo Celular/metabolismo , Fibrossarcoma/metabolismo , Lipossarcoma Mixoide/metabolismo , Proteínas de Fusão Oncogênica/metabolismo , Proteínas da Gravidez/metabolismo , Proteína FUS de Ligação a RNA/metabolismo , Transdução de Sinais , Fator de Transcrição CHOP/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Animais , Western Blotting , Linhagem Celular Tumoral , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/patologia , Proliferação de Células , Relação Dose-Resposta a Droga , Fibrossarcoma/genética , Fibrossarcoma/patologia , Imunofluorescência , Humanos , Imuno-Histoquímica , Ligantes , Lipossarcoma Mixoide/genética , Lipossarcoma Mixoide/patologia , Camundongos , Camundongos SCID , Transplante de Neoplasias , Proteínas de Fusão Oncogênica/genética , Fator de Crescimento Placentário , Proteínas da Gravidez/genética , Inibidores de Proteínas Quinases/farmacologia , Interferência de RNA , Proteína FUS de Ligação a RNA/genética , Proteínas Recombinantes de Fusão/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição CHOP/genética , Transfecção , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fator B de Crescimento do Endotélio Vascular/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/genética
6.
PLoS One ; 15(12): e0243807, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33332401

RESUMO

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.


Assuntos
Células Dendríticas/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Macrófagos/citologia , Transcrição Gênica , Diferenciação Celular , Linhagem da Célula , Células Dendríticas/metabolismo , Humanos , Macrófagos/metabolismo
7.
Int J Oncol ; 25(5): 1349-55, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15492825

RESUMO

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.


Assuntos
Proteínas Estimuladoras de Ligação a CCAAT/genética , Proteínas de Ciclo Celular/biossíntese , Proteínas de Ciclo Celular/farmacologia , Perfilação da Expressão Gênica , Lipossarcoma Mixoide/patologia , Proteínas de Fusão Oncogênica/genética , Proteína FUS de Ligação a RNA/genética , Adulto , Idoso , Western Blotting , Proteínas de Ciclo Celular/genética , Feminino , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Análise de Sequência com Séries de Oligonucleotídeos , Fator de Transcrição CHOP , Transfecção , Células Tumorais Cultivadas
8.
PLoS One ; 8(5): e64721, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23700488

RESUMO

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.


Assuntos
Metabolismo Energético , Fígado Gorduroso/metabolismo , Glucose/metabolismo , Receptores Citoplasmáticos e Nucleares/deficiência , Tecido Adiposo Branco/metabolismo , Tecido Adiposo Branco/patologia , Envelhecimento , Animais , Glicemia , Composição Corporal , Peso Corporal , Tamanho Celular , Ingestão de Energia , Feminino , Metabolismo dos Lipídeos , Fígado/metabolismo , Fígado/patologia , Fígado/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Atividade Motora , Hepatopatia Gordurosa não Alcoólica , Tamanho do Órgão , Receptores Citoplasmáticos e Nucleares/genética , Pele/patologia , Triglicerídeos/metabolismo
9.
Biochem Biophys Res Commun ; 363(3): 633-8, 2007 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-17904108

RESUMO

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.


Assuntos
Metabolismo Energético/fisiologia , Obesidade/fisiopatologia , Receptores Acoplados a Proteínas G/fisiologia , Tecido Adiposo/metabolismo , Animais , Glicemia/metabolismo , Temperatura Corporal , Peso Corporal/genética , Peso Corporal/fisiologia , Calorimetria/métodos , Ingestão de Alimentos/genética , Ingestão de Alimentos/fisiologia , Metabolismo Energético/genética , Fezes/química , Feminino , Genótipo , Lipoproteínas HDL/sangue , Lipoproteínas LDL/sangue , Masculino , Camundongos , Camundongos Knockout , Obesidade/genética , Receptores Acoplados a Proteínas G/deficiência , Receptores Acoplados a Proteínas G/genética , Fatores Sexuais , Fatores de Tempo
10.
Am J Pathol ; 168(5): 1642-53, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16651630

RESUMO

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.


Assuntos
Fibrossarcoma/irrigação sanguínea , Regulação Neoplásica da Expressão Gênica , Lipossarcoma Mixoide/irrigação sanguínea , Proteína FUS de Ligação a RNA/fisiologia , Fator de Transcrição CHOP/fisiologia , Adipogenia , Animais , Análise por Conglomerados , Regulação para Baixo , Feminino , Fibrossarcoma/metabolismo , Humanos , Lipossarcoma Mixoide/metabolismo , Camundongos , Camundongos SCID , Análise de Sequência com Séries de Oligonucleotídeos , Proteínas de Fusão Oncogênica/fisiologia , Proteína FUS de Ligação a RNA/genética , Proteína FUS de Ligação a RNA/metabolismo , Fator de Transcrição CHOP/metabolismo , Transfecção , Regulação para Cima
11.
Int J Cancer ; 115(4): 556-60, 2005 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-15688424

RESUMO

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.


Assuntos
Proteína beta Intensificadora de Ligação a CCAAT/fisiologia , Proteínas Estimuladoras de Ligação a CCAAT/genética , Fibrossarcoma/genética , Interleucina-6/genética , Lipossarcoma Mixoide/genética , Proteínas de Fusão Oncogênica/genética , Proteína FUS de Ligação a RNA/genética , Fatores de Transcrição/genética , Animais , Linhagem Celular Tumoral , Primers do DNA , Fibrossarcoma/imunologia , Humanos , Interleucina-6/sangue , Lipossarcoma Mixoide/sangue , Lipossarcoma Mixoide/imunologia , Camundongos , Camundongos SCID , NF-kappa B/metabolismo , Reação em Cadeia da Polimerase , Regiões Promotoras Genéticas , RNA Interferente Pequeno/genética , Fator de Transcrição CHOP , Transplante Heterólogo
12.
Exp Cell Res ; 278(2): 125-32, 2002 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-12169268

RESUMO

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.


Assuntos
Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Compartimento Celular/fisiologia , Núcleo Celular/metabolismo , Proteínas de Fusão Oncogênica/metabolismo , Proteína FUS de Ligação a RNA , Ribonucleoproteínas , Sequência de Aminoácidos , Proteínas Estimuladoras de Ligação a CCAAT/genética , Núcleo Celular/ultraestrutura , Humanos , Mutagênese Sítio-Dirigida , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares/metabolismo , Proteínas de Fusão Oncogênica/genética , Fatores de Processamento de Serina-Arginina , Spliceossomos/metabolismo , Temperatura , Fator de Transcrição CHOP , Fatores de Transcrição/metabolismo , Transfecção , Células Tumorais Cultivadas
13.
Int J Cancer ; 97(4): 446-50, 2002 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-11802205

RESUMO

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.


Assuntos
Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Núcleo Celular/química , Lipossarcoma Mixoide/genética , Proteínas Nucleares , Proteínas de Fusão Oncogênica/metabolismo , Proteína FUS de Ligação a RNA , Animais , Células COS , Divisão Celular , Fracionamento Celular , Núcleo Celular/ultraestrutura , Chlorocebus aethiops , Cromossomos Humanos Par 12/genética , Cromossomos Humanos Par 12/ultraestrutura , Cromossomos Humanos Par 16/genética , Cromossomos Humanos Par 16/ultraestrutura , Citoplasma/química , Dimerização , Fibroblastos/metabolismo , Fibrossarcoma/patologia , Genes Reporter , Proteínas de Fluorescência Verde , Humanos , Antígeno Ki-67/análise , Leucemia Promielocítica Aguda/genética , Lipossarcoma Mixoide/ultraestrutura , Proteínas Luminescentes/análise , Proteínas Luminescentes/genética , Proteínas de Neoplasias/análise , Reação em Cadeia da Polimerase , Antígeno Nuclear de Célula em Proliferação/análise , Proteína da Leucemia Promielocítica , Proteínas Recombinantes de Fusão/metabolismo , Fator de Transcrição CHOP , Fatores de Transcrição/análise , Transfecção , Translocação Genética , Proteínas Supressoras de Tumor
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA