Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Death Dis ; 11(10): 875, 2020 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-33070156

RESUMO

Since metastatic colorectal cancer (CRC) is a leading cause of cancer-related death, therapeutic approaches overcoming primary and acquired therapy resistance are an urgent medical need. In this study, the efficacy and toxicity of high-affinity inhibitors targeting antiapoptotic BCL-2 proteins (BCL-2, BCL-XL, and MCL-1) were evaluated. By RNA sequencing analysis of a pan-cancer cohort comprising >1500 patients and subsequent prediction of protein activity, BCL-XL was identified as the only antiapoptotic BCL-2 protein that is overactivated in CRC. Consistently, pharmacologic and genetic inhibition of BCL-XL induced apoptosis in human CRC cell lines. In a combined treatment approach, targeting BCL-XL augmented the efficacy of chemotherapy in vitro, in a murine CRC model, and in human ex vivo derived CRC tissue cultures. Collectively, these data show that targeting of BCL-XL is efficient and safe in preclinical CRC models, observations that pave the way for clinical translation.


Assuntos
Antineoplásicos/farmacologia , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/metabolismo , Proteína bcl-X/metabolismo , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Neoplasias do Colo/tratamento farmacológico , Neoplasias Colorretais/patologia , Humanos , Proteína de Sequência 1 de Leucemia de Células Mieloides/efeitos dos fármacos , Proteína de Sequência 1 de Leucemia de Células Mieloides/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteína bcl-X/efeitos dos fármacos
2.
Int J Mol Sci ; 21(3)2020 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-32046105

RESUMO

Autophagy is a catabolic process that enables cells to degrade obsolete content and refuel energy depots. In colorectal cancer (CRC) autophagy has been shown to promote tumorigenesis through energy delivery in the condition of uncontrolled proliferation. With this study, we aimed at evaluating whether autophagy sustains CRC cell viability and if it impacts therapy resistance. Initially, a colorectal cancer tissue micro array, containing mucosa (n = 10), adenoma (n = 18) and adenocarcinoma (n = 49) spots, was stained for expression of essential autophagy proteins LC3b, Atg7, p62 and Beclin-1. Subsequently, central autophagy proteins were downregulated in CRC cells using siRNA technology. Viability assays, flow cytometry and immunoblotting were performed and three-dimensional cell culture was utilized to study autophagy in a tissue mimicking environment. In our study we found an upregulation of Atg7 in CRC. Furthermore, we identified Atg7 as crucial factor within the autophagy network for CRC cell viability. Its disruption induced cell death via triggering apoptosis and in combination with conventional chemotherapy it exerted synergistic effects in inducing CRC cell death. Cell death was strictly dependent on nuclear LC3b, since simultaneous knockdown of Atg7 and LC3b completely restored viability. This study unravels a novel cell death preventing function of Atg7 in interaction with LC3b, thereby unmasking a promising therapeutic target in CRC.


Assuntos
Adenocarcinoma/metabolismo , Apoptose , Proteína 7 Relacionada à Autofagia/metabolismo , Neoplasias Colorretais/metabolismo , Resistencia a Medicamentos Antineoplásicos , Adenocarcinoma/genética , Antineoplásicos/farmacologia , Autofagia , Proteína 7 Relacionada à Autofagia/genética , Proteína Beclina-1/genética , Proteína Beclina-1/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Neoplasias Colorretais/genética , Fluoruracila/farmacologia , Células HT29 , Humanos , Irinotecano/farmacologia , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo
3.
Gastroenterology ; 156(4): 1190-1205.e14, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30445013

RESUMO

BACKGROUND & AIMS: Cholangiocyte proliferation and ductular reaction contribute to the onset and progression of liver diseases. Little is known about the role of the transcription factor nuclear factor-κB (NF-κB) in this process. We investigated the activities of the RELB proto-oncogene NF-κB subunit in human cholangiocytes and in mouse models of liver disease characterized by a ductular reaction. METHODS: We obtained liver tissue samples from patients with primary sclerosing cholangitis, primary biliary cholangitis, hepatitis B or C virus infection, autoimmune hepatitis, alcoholic liver disease, or without these diseases (controls) from a tissue bank in Germany. Tissues were analyzed by immunohistochemistry for levels of RELB and lymphotoxin ß (LTB). We studied mice with liver parenchymal cell (LPC)-specific disruption of the cylindromatosis (CYLD) lysine 63 deubiquitinase gene (Cyld), with or without disruption of Relb (CyldΔLPC mice and Cyld/RelbΔLPC mice) and compared them with C57BL/6 mice (controls). Mice were fed 5-diethoxycarbonyl-1,4-dihydrocollidine (DDC) or standard chow diets to induce biliary injury or were given injections of CCl4 to induce non-cholestatic liver fibrosis. Liver tissues were analyzed by histology, immunohistochemistry, immunoblots, in situ hybridization, and quantitative real-time polymerase chain reaction. Cholangiocytes were isolated from normal human liver, incubated with LTB receptor agonist, and transfected with small interfering RNAs to knock down RELB. RESULTS: In liver tissues from patients with primary sclerosing cholangitis, primary biliary cholangitis, chronic infection with hepatitis B or C virus, autoimmune hepatitis, or alcoholic liver disease, we detected increased nuclear translocation of RELB and increased levels of LTB in cholangiocytes that formed reactive bile ducts compared with control liver tissues. Human cholangiocytes, but not those with RELB knockdown, proliferated with exposure to LTB. The phenotype of CyldΔLPC mice, which included ductular reaction, oval cell activation, and biliary fibrosis, was completely lost from Cyld/RelbΔLPC mice. Compared with livers from control mice, livers from CyldΔLPC mice (but not Cyld/RelbΔLPC mice) had increased levels of mRNAs encoding cytokines (LTB; CD40; and tumor necrosis factor superfamily [TNFSF] members TNFSF11 [RANKL], TNFSF13B [BAFF], and TNFSF14 [LIGHT]) produced by reactive cholangiocytes. However, these strains of mice developed similar levels of liver fibrosis in response to CCl4 exposure. CyldΔLPC mice and Cyld/RelbΔLPC mice had improved liver function on the DDC diet compared with control mice fed the DDC diet. CONCLUSION: Reactive bile ducts in patients with chronic liver diseases have increased levels of LTB and nuclear translocation of RELB. RELB is required for the ductular reaction and development of biliary fibrosis in CyldΔLPC mice. Deletion of RELB and CYLD from LPCs protects mice from DDC-induced cholestatic liver fibrosis.


Assuntos
Ductos Biliares/metabolismo , Ductos Biliares/patologia , Colangite Esclerosante/metabolismo , Citocinas/genética , Hepatopatias/metabolismo , Fator de Transcrição RelB/metabolismo , Adolescente , Adulto , Idoso , Animais , Tetracloreto de Carbono , Núcleo Celular , Proliferação de Células , Células Cultivadas , Cisteína Endopeptidases/genética , Enzima Desubiquitinante CYLD , Dicarbetoxi-Di-Hidrocolidina , Células Epiteliais/metabolismo , Feminino , Fibrose , Técnicas de Silenciamento de Genes , Humanos , Fígado/patologia , Cirrose Hepática/induzido quimicamente , Cirrose Hepática/metabolismo , Cirrose Hepática/patologia , Receptor beta de Linfotoxina/agonistas , Linfotoxina-beta/metabolismo , Masculino , Camundongos , Pessoa de Meia-Idade , Tecido Parenquimatoso/patologia , Transporte Proteico , Proto-Oncogene Mas , RNA Mensageiro/metabolismo , Fator de Transcrição RelB/genética , Adulto Jovem
4.
EBioMedicine ; 32: 125-133, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29884457

RESUMO

A higher capacity to grow under hypoxic conditions can lead to a more aggressive behavior of tumor cells. Determining tumor activity under hypoxia may identify chronic lymphocytic leukemia (CLL) with aggressive clinical course and predict response to chemo-immunotherapy (CIT). A metabolic score was generated by determining pyruvate kinase and lactate dehydrogenase, key enzymes of glycolysis, ex vivo in primary CLL samples under normoxic and hypoxic conditions. This score was further correlated with clinical endpoints and response to CIT in 96 CLL patients. 45 patients were classified as metabolic high risk (HR), 51 as low risk (LR). Treatment-free survival (TFS) was significantly shorter in HR patients (median 394 vs 723 days, p = .021). 15 HR patients and 14 LR patients received CIT after sample acquisition. HR patients had a significantly shorter progression-free survival after treatment compared to LR patients (median 216 days vs not reached, p = .008). Multivariate analysis evaluating age, IGHV, TP53 deletion or mutation and 11q22-23 deletion besides the capacity of tumor cells to grow under severe hypoxic conditions identified the metabolic profile as the strongest independent risk factor for shorter TFS (hazard ratio 2.37, p = .011). The metabolic risk can provide prognostic and predictive information complementary to genetic biomarkers and identify patients who might benefit from alternative treatment approaches.


Assuntos
Biomarcadores Tumorais/genética , Imunoterapia , Leucemia Linfocítica Crônica de Células B/tratamento farmacológico , Prognóstico , Adulto , Idoso , Idoso de 80 Anos ou mais , Intervalo Livre de Doença , Feminino , Glicólise/genética , Humanos , Leucemia Linfocítica Crônica de Células B/genética , Leucemia Linfocítica Crônica de Células B/imunologia , Leucemia Linfocítica Crônica de Células B/patologia , Masculino , Pessoa de Meia-Idade , Mutação , Modelos de Riscos Proporcionais , Fatores de Risco , Hipóxia Tumoral/genética , Hipóxia Tumoral/imunologia
6.
Mol Cell Oncol ; 3(4): e1175538, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27652323

RESUMO

Targeting tumor glycolysis would hit the main energy source of cancer. We show that natural killer (NK) cells pursue this strategy by employing high mobility group box 1 (HMGB1) protein-a well-known proinflammatory cytokine-to specifically target glycolysis in cancer cells. This opens up new perspectives for cancer immunotherapy.

7.
Alcohol Clin Exp Res ; 40(10): 2094-2101, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27581253

RESUMO

BACKGROUND: Chronic alcohol consumption is a risk factor for colorectal cancer. The mechanisms by which ethanol (EtOH) exerts its carcinogenic effect on the colorectal mucosa are not clear and may include oxidative stress with the action of reactive oxygen species (ROS) generated through EtOH metabolism via cytochrome P4502E1 (CYP2E1) leading to carcinogenic etheno-DNA adducts. ROS may also induce apoptosis. However, the effect of chronic EtOH consumption on CYP2E1, etheno-DNA adducts as well as anti-apoptotic proteins in the colorectal mucosa of heavy drinkers without colorectal inflammation is still not known. METHODS: Rectal biopsies from 32 alcoholics (>60 g EtOH/d) and from 12 controls (<20 g EtOH/d) were histologically examined, and immunohistochemistry for CYP2E1 and etheno-DNA adducts was performed. Apoptosis (cleaved PARP) as well as anti-apoptotic proteins including Bcl-xL , Bcl-2, and Mcl-1 were immunohistochemically determined. RESULTS: No significant difference in mucosal CYP2E1 or etheno-DNA adducts was observed between alcoholics and control patients. However, CYP2E1 and etheno-DNA adducts correlated significantly when both groups were combined (p < 0.001). In addition, although apoptosis was found not to be significantly affected by EtOH, the anti-apoptotic protein Mcl-1, but neither Bcl-xL nor Bcl-2, was found to be significantly increased in heavy drinkers as compared to controls (p = 0.014). CONCLUSIONS: Although colorectal CYP2E1 was not found to be significantly increased in alcoholics, CYP2E1 correlated overall with the level of etheno-DNA adducts in the colorectal mucosa, which identifies CYP2E1 as an important factor in colorectal carcinogenesis. Most importantly, however, is the up-regulation of the anti-apoptotic protein Mcl-1 in heavy drinkers counteracting apoptosis and possibly stimulating cancer development.


Assuntos
Alcoolismo/metabolismo , Carcinogênese/induzido quimicamente , Citocromo P-450 CYP2E1/metabolismo , Adutos de DNA/metabolismo , Etanol/toxicidade , Proteína de Sequência 1 de Leucemia de Células Mieloides/metabolismo , Reto/metabolismo , Idoso , Alcoolismo/complicações , Apoptose , Proteínas Reguladoras de Apoptose/metabolismo , Estudos de Casos e Controles , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Reto/efeitos dos fármacos
8.
Cell Death Dis ; 7(8): e2342, 2016 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-27537525

RESUMO

Colorectal cancer (CRC) is the second most common malignant neoplasia in women and men worldwide. The B-cell lymphoma 2 (Bcl-2) protein family is mainly known for its pivotal role in the regulation of the mitochondrial death pathway. Anti-apoptotic Bcl-2 proteins may provide survival benefits and induce therapy resistance in cancer cells. Among anti-apoptotic Bcl-2 proteins, we found solely Bcl-xL strongly upregulated in human CRC specimens. In order to study protein function in the context of tumor initiation and progression in vivo, we generated a mouse model lacking Bcl-xL in intestinal epithelial cells (Bcl-xL(IEC-KO)). If challenged in an inflammation-driven tumor model, Bcl-xL(IEC-KO) mice showed a significantly reduced tumor burden with lower tumor numbers per animal and decreased tumor sizes. Analysis of cell death events by immunohistochemistry and immunoblotting revealed a striking increase of apoptosis in Bcl-xL-negative tumors. qRT-PCR and immunohistochemistry excluded changes in proliferative capacity and immune cell infiltration as reasons for the reduced tumor load and thereby identify apoptosis as key mechanism. Human CRC tissue was cultured ex vivo and treated with the small molecule compound ABT-737, which inhibits Bcl-xL and Bcl-2. Under ABT-737 treatment, the amount of apoptotic tumor cells significantly increased compared with controls, whereas proliferation levels remained unaltered. In summary, our findings identify Bcl-xL as a driver in colorectal tumorigenesis and cancer progression, making it a valuable target for clinical application.


Assuntos
Neoplasias Colorretais/genética , Oncogenes , Proteína bcl-X/genética , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Compostos de Bifenilo/farmacologia , Carcinogênese/efeitos dos fármacos , Carcinogênese/genética , Carcinogênese/patologia , Técnicas de Cultura de Células , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Neoplasias Colorretais/patologia , Modelos Animais de Doenças , Enterócitos/efeitos dos fármacos , Enterócitos/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Humanos , Inflamação/genética , Inflamação/patologia , Linfócitos do Interstício Tumoral/efeitos dos fármacos , Linfócitos do Interstício Tumoral/metabolismo , Camundongos , Camundongos Knockout , Nitrofenóis/farmacologia , Especificidade de Órgãos , Fenótipo , Piperazinas/farmacologia , Sulfonamidas/farmacologia , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética , Proteína bcl-X/metabolismo
9.
Am J Med Genet A ; 170(6): 1502-9, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27016154

RESUMO

Intellectual disability (ID) with cerebellar ataxia comprises a genetically heterogeneous group of neurodevelopmental disorders. We identified a homozygous frameshift mutation in CWF19L1 (c.467delC; p.(P156Hfs*33)) by a combination of linkage analysis and Whole Exome Sequencing in a consanguineous Turkish family with a 9-year-old boy affected by early onset cerebellar ataxia and mild ID. Serial MRI showed mildly progressive cerebellar atrophy. Absent C19L1 protein expression in lymphoblastoid cell lines strongly suggested that c.467delC is a disease-causing alteration. One further pregnancy of the mother had been terminated at 22 weeks of gestation because of a small cerebellum and agenesis of corpus callosum. The homozygous CWF19L1 variant was also present in the fetus. Postmortem examination of the fetus in addition showed unilateral hexadactyly and vertebral malformations. These features have not been reported and may represent an expansion of the CWF19L1-related phenotypic spectrum, but could also be due to another, possibly autosomal recessive disorder. The exact function of the evolutionarily highly conserved C19L1 protein is unknown. So far, homozygous or compound heterozygous mutations in CWF19L1 have been identified in two Turkish siblings and a Dutch girl, respectively, affected by cerebellar ataxia and ID. A zebrafish model showed that CWF19L1 loss-of-function mutations result in abnormal cerebellar morphology and movement disorders. Our report corroborates that loss-of-function mutations in CWF19Ll lead to early onset cerebellar ataxia and (progressive) cerebellar atrophy. © 2016 Wiley Periodicals, Inc.


Assuntos
Proteínas de Ciclo Celular/genética , Cerebelo/anormalidades , Exoma , Sequenciamento de Nucleotídeos em Larga Escala , Deficiência Intelectual/diagnóstico , Deficiência Intelectual/genética , Mutação , Malformações do Sistema Nervoso/diagnóstico , Malformações do Sistema Nervoso/genética , Encéfalo/anormalidades , Criança , Hibridização Genômica Comparativa , Consanguinidade , Deficiências do Desenvolvimento/diagnóstico , Deficiências do Desenvolvimento/genética , Estudos de Associação Genética , Ligação Genética , Humanos , Imageamento por Ressonância Magnética , Masculino , Linhagem , Fenótipo , Polimorfismo de Nucleotídeo Único , Radiografia
10.
Nat Commun ; 7: 10764, 2016 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-26948869

RESUMO

The high-mobility group box 1 (HMGB1) protein has a central role in immunological antitumour defense. Here we show that natural killer cell-derived HMGB1 directly eliminates cancer cells by triggering metabolic cell death. HMGB1 allosterically inhibits the tetrameric pyruvate kinase isoform M2, thus blocking glucose-driven aerobic respiration. This results in a rapid metabolic shift forcing cells to rely solely on glycolysis for the maintenance of energy production. Cancer cells can acquire resistance to HMGB1 by increasing glycolysis using the dimeric form of PKM2, and employing glutaminolysis. Consistently, we observe an increase in the expression of a key enzyme of glutaminolysis, malic enzyme 1, in advanced colon cancer. Moreover, pharmaceutical inhibition of glutaminolysis sensitizes tumour cells to HMGB1 providing a basis for a therapeutic strategy for treating cancer.


Assuntos
Neoplasias do Colo/metabolismo , Neoplasias do Colo/fisiopatologia , Proteína HMGB1/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Morte Celular , Linhagem Celular Tumoral , Respiração Celular , Neoplasias do Colo/enzimologia , Neoplasias do Colo/genética , Glucose/metabolismo , Glicólise , Proteína HMGB1/genética , Humanos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Hormônios Tireóideos/genética , Hormônios Tireóideos/metabolismo , Proteínas de Ligação a Hormônio da Tireoide
11.
Int J Oncol ; 46(2): 667-76, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25434832

RESUMO

The HMGB1 protein has multiple functions in tumor biology and can act both as a transcription factor and as a cytokine. HMGB1 is released during cell death, and in our previous studies we demonstrated that HMGB1 induces a distinct, necrosis-like cell death in glioblastoma. In epithelial malignant tumors such as colorectal cancer (CRC), the HMGB1-dependent effects show cross-talk with apoptotic signal transduction. Treatment of CRC cells with low concentrations of recombinant HMGB1 results in dose-dependent cytotoxicity which is morphologically characterized by the formation of giant mitochondria and does not share features of apoptosis. HMGB1-triggered cell death is associated with intracellular ROS release, and overexpression of Bcl-2 blocks both the increase of ROS as well as HMGB1-dependent cell death. Importantly, treatment with recombinant HMGB1 or overexpression of endogenous HMGB1 strongly sensitizes CRC cells to the cytotoxic activity of the pro-apoptotic death ligand TRAIL as well as the small molecule Bcl-2 family inhibitor ABT­737. Moreover, treatment of CRC cells with TRAIL or ABT­737 induces a release of endogenous HMGB1 into the extracellular space, and preincubation with glycyrrhizin, an HMGB1 inhibitor, significantly inhibits induction of cell death by TRAIL and ABT­737, suggesting that HMGB1 functionally contributes to the execution of cell death triggered by pro-apoptotic agents. Finally, we investigated the expression of HMGB1 in human CRC tumor samples and found that loss of HMGB1 expression is associated with a more aggressive phenotype and a more advanced stage of disease in patients with CRC. Altogether, our findings demonstrate a functional link between cytotoxic signaling cascades triggered by HMGB1 and pro-apoptotic agents leading to an HMGB1-dependent sensitization to CRC cell death. Thus, a further evaluation of recombinant HMGB1 as part of an experimental combination treatment of CRC seems warranted.


Assuntos
Neoplasias do Colo/genética , Proteína HMGB1/genética , Proteínas Recombinantes/genética , Ligante Indutor de Apoptose Relacionado a TNF/genética , Apoptose/genética , Proteínas Reguladoras de Apoptose/administração & dosagem , Compostos de Bifenilo/administração & dosagem , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Ácido Glicirrízico/administração & dosagem , Células HCT116 , Proteína HMGB1/administração & dosagem , Proteína HMGB1/metabolismo , Humanos , Nitrofenóis/administração & dosagem , Piperazinas/administração & dosagem , Proteínas Proto-Oncogênicas c-bcl-2 , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/metabolismo , Sulfonamidas/administração & dosagem , Ligante Indutor de Apoptose Relacionado a TNF/administração & dosagem , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo
12.
Neuro Oncol ; 14(1): 64-78, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22015597

RESUMO

The role of microglia, the brain-resident macrophages, in glioma biology is still a matter of debate. Clinical observations and in vitro studies in the mouse model indicate that microglia and macrophages that infiltrate the brain tumor tissue in high numbers play a tumor-supportive role. Here, we provide evidence that human microglia isolated from brain tumors indeed support tumor cell growth, migration, and invasion. However, after stimulation with the Toll-like receptor 3 agonist poly (I:C), microglia secrete factors that exerted toxic and suppressive effects on different glioblastoma cell lines, as assessed in cytotoxicity, migration, and tumor cell spheroid invasion assays. Remarkably, these effects were tumor-specific because the microglial factors impaired neither growth nor viability of astrocytes and neurons. Culture supernatants of tumor cells inhibited the poly (I:C) induction of this microglial M1-like, oncotoxic profile. Microglia stimulation before coculture with tumor cells circumvented the tumor-mediated suppression, as demonstrated by the ability to kill and phagocytose glioma cells. Our results show, for the first time to our knowledge, that human microglia exert tumor-supporting functions that are overridden by tumor-suppressing activities gained after poly (I:C) stimulation.


Assuntos
Antineoplásicos/farmacologia , Neoplasias Encefálicas/metabolismo , Glioma/metabolismo , Microglia/fisiologia , Poli I-C/farmacologia , Receptor 3 Toll-Like/agonistas , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Células Cultivadas , Técnicas de Cocultura , Humanos , Microglia/efeitos dos fármacos , Invasividade Neoplásica
13.
Virchows Arch ; 458(3): 371-6, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21234762

RESUMO

The main cause of death from novel (swine origin) influenza A/H1N1 infection is acute respiratory distress syndrome. Most fatal cases are immunocompromised patients or patients with a severe underlying disease. Here, we report a fatal case of acute interstitial myocarditis associated with novel influenza A/H1N1 infection in an immunocompetent young woman. A previously healthy 18-year-old woman experienced malaise, diarrhea, and fever for several days prior to a sudden collapse at home. Autopsy revealed a predominantly lymphocytic myocarditis in the absence of a significant respiratory tract infection. Infection with novel (swine origin) influenza A/H1N1 was confirmed by PCR analysis of blood as well as myocardial tissue. Influenza-caused diarrhea with consecutive hypokalemia potentially contributed to the fatal outcome of the myocarditis, characterized by ventricular fibrillation. In conclusion, sudden death by myocarditis may be a rare complication of novel influenza A/H1N1 infection in otherwise healthy individuals, even in the absence of significant respiratory tract infection.


Assuntos
Morte Súbita Cardíaca/etiologia , Vírus da Influenza A Subtipo H1N1/isolamento & purificação , Influenza Humana/virologia , Miocardite/patologia , Adolescente , DNA Viral/análise , Morte Súbita Cardíaca/patologia , Evolução Fatal , Feminino , Humanos , Imunocompetência , Vírus da Influenza A Subtipo H1N1/genética , Influenza Humana/patologia , Miocardite/virologia , Reação em Cadeia da Polimerase
14.
Cancer Res ; 70(21): 8558-68, 2010 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-20959471

RESUMO

Cells dying by necrosis release the high-mobility group box 1 (HMGB1) protein, which has immunostimulatory effects. However, little is known about the direct actions of extracellular HMGB1 protein on cancer cells. Here, we show that recombinant human HMGB1 (rhHMGB1) exerts strong cytotoxic effects on malignant tumor cells. The rhHMGB1-induced cytotoxicity depends on the presence of mitochondria and leads to fast depletion of mitochondrial DNA, severe damage of the mitochondrial proteome by toxic malondialdehyde adducts, and formation of giant mitochondria. The formation of giant mitochondria is independent of direct nuclear signaling events, because giant mitochondria are also observed in cytoplasts lacking nuclei. Further, the reactive oxygen species scavenger N-acetylcysteine as well as c-Jun NH(2)-terminal kinase blockade inhibited the cytotoxic effect of rhHMGB1. Importantly, glioblastoma cells, but not normal astrocytes, were highly susceptible to rhHMGB1-induced cell death. Systemic treatment with rhHMGB1 results in significant growth inhibition of xenografted tumors in vivo. In summary, rhHMGB1 induces a distinct form of cell death in cancer cells, which differs from the known forms of apoptosis, autophagy, and senescence, possibly representing an important novel mechanism of specialized necrosis. Further, our findings suggest that rhHMGB1 may offer therapeutic applications in treatment of patients with malignant brain tumors.


Assuntos
Apoptose , Glioblastoma/patologia , Proteína HMGB1/metabolismo , Mitocôndrias/patologia , Acetilcisteína/farmacologia , Animais , Astrócitos/citologia , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Western Blotting , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Proliferação de Células/efeitos dos fármacos , Eletroforese em Gel Bidimensional , Feminino , Imunofluorescência , Sequestradores de Radicais Livres/farmacologia , Glioblastoma/tratamento farmacológico , Glioblastoma/metabolismo , Proteína HMGB1/genética , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Potencial da Membrana Mitocondrial , Camundongos , Camundongos Nus , Mitocôndrias/metabolismo , Proteínas Mitocondriais/metabolismo , Necrose , Proteoma/análise , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Células Tumorais Cultivadas
15.
Hepatology ; 52(6): 2023-33, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20979053

RESUMO

UNLABELLED: The A kinase anchor protein 12 (AKAP12) is a central mediator of protein kinase A and protein kinase C signaling. Although AKAP12 has been described to act as a tumor suppressor and its expression is frequently down-regulated in several human malignancies, the underlying molecular mechanisms responsible for the AKAP12 reduction are poorly understood. We therefore analyzed the expression of AKAP12 and its genetic and epigenetic regulatory mechanisms in human hepatocarcinogenesis. Based on tissue microarray analyses (n = 388) and western immunoblotting, we observed a significant reduction of AKAP12 in cirrhotic liver (CL), premalignant lesions (DN), and hepatocellular carcinomas (HCCs) compared to histologically normal liver specimens (NL). Analyses of array comparative genomic hybridization data (aCGH) from human HCCs revealed chromosomal losses of AKAP12 in 36% of cases but suggested additional mechanisms underlying the observed reduction of AKAP12 expression in hepatocarcinogenesis. Quantitative methylation analysis by MassARRAY of NL, CL, DN, and HCC tissues, as well as of various tumorigenic and nontumorigenic liver cell lines revealed specific hypermethylation of the AKAP12α promoter but not of the AKAP12ß promoter in HCC specimens and in HCC cell lines. Consequently, restoration experiments performed with 5-aza-2'deoxycytidine drastically increased AKAP12α mRNA levels in a HCC cell line (AKN1) paralleled by AKAP12α promoter demethylation. As hypermethylation is not observed in CL and DN, we investigated microRNA-mediated posttranscriptional regulation as an additional mechanism to explain reduced AKAP12 expression. We found that miR-183 and miR-186 are up-regulated in CL and DN and are able to target AKAP12. CONCLUSION: In addition to genetic alterations, epigenetic mechanisms are responsible for the reduction of the tumor suppressor gene AKAP12 in human hepatocarcinogenesis.


Assuntos
Proteínas de Ancoragem à Quinase A/genética , Carcinoma Hepatocelular/metabolismo , Proteínas de Ciclo Celular/genética , Epigênese Genética , Neoplasias Hepáticas/metabolismo , Azacitidina/análogos & derivados , Azacitidina/farmacologia , Carcinoma Hepatocelular/genética , Linhagem Celular Tumoral , Decitabina , Regulação para Baixo , Humanos , Neoplasias Hepáticas/genética , MicroRNAs/metabolismo , Regiões Promotoras Genéticas/fisiologia , Análise Serial de Proteínas
16.
Clin Cancer Res ; 16(10): 2715-28, 2010 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-20442299

RESUMO

PURPOSE: Stem-like tumor cells comprise a highly tumorigenic and therapy-resistant tumor subpopulation, which is believed to substantially influence tumor initiation and therapy resistance in glioma. Currently, therapeutic, drug-induced differentiation is considered as a promising approach to eradicate this tumor-driving cell population; retinoic acid is well known as a potent modulator of differentiation and proliferation in normal stem cells. In glioma, knowledge about the efficacy of retinoic acid-induced differentiation to target the stem-like tumor cell pool could have therapeutic implications. EXPERIMENTAL DESIGN: Stem-like glioma cells (SLGC) were differentiated with all-trans retinoic acid-containing medium to study the effect of differentiation on angiogenesis, invasive growth, as well as radioresistance and chemoresistance of SLGCs. In vivo effects were studied using live microscopy in a cranial window model. RESULTS: Our data suggest that in vitro differentiation of SLGCs induces therapy-sensitizing effects, impairs the secretion of angiogenic cytokines, and disrupts SLGCs motility. Further, ex vivo differentiation reduces tumorigenicity of SLGCs. Finally, we show that all-trans retinoic acid treatment alone can induce antitumor effects in vivo. CONCLUSIONS: Altogether, these results highlight the potential of differentiation treatment to target the stem-like cell population in glioblastoma.


Assuntos
Antineoplásicos/farmacologia , Neoplasias Encefálicas/tratamento farmacológico , Diferenciação Celular/efeitos dos fármacos , Glioma/tratamento farmacológico , Células-Tronco Neoplásicas/efeitos dos fármacos , Tretinoína/farmacologia , Animais , Western Blotting , Neoplasias Encefálicas/patologia , Separação Celular , Citometria de Fluxo , Glioma/patologia , Humanos , Imuno-Histoquímica , Hibridização in Situ Fluorescente , Camundongos , Camundongos Endogâmicos NOD , Células-Tronco Neoplásicas/patologia , Reação em Cadeia da Polimerase , Ensaios Antitumorais Modelo de Xenoenxerto
17.
Mol Carcinog ; 49(2): 175-82, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19852062

RESUMO

Approximately 15% of small intestinal adenocarcinomas show inactivation of DNA-mismatch repair (MMR) and display high-level microsatellite instability (MSI-H). MSI-H tumors progress as a result of mutations affecting coding microsatellites (coding microsatellite instability, cMSI) that may result in a functional inactivation of the encoded proteins and provide a selective growth advantage for the affected cell. To investigate the cMSI selection in small intestinal carcinogenesis 56 adenocarcinomas were tested for MSI. Eleven MSI-H carcinomas (19.6%) were identified and subjected to cMSI analysis in 24 potentially tumor relevant genes. Mutation frequencies were similar to those observed in colorectal cancer (CRC). Beside high frequencies of cMSI in TGFbetaR2, ACVR2, and AIM2 we detected MARCKS mutations in 10 out of 11 (91%) tumors with a 30% share of biallelic mutations. Since little is known about MARCKS expression in the intestine, we analyzed MARCKS protein expression in 31 carcinomas. In non-neoplastic mucosa, MARCKS was found to be expressed with a concentration gradient along the crypt-villus axis. In line with cMSI induced functional inactivation of MARCKS, 8 out of 11 MSI-H adenocarcinomas showed regional or complete loss of the protein. In microsatellite stable (MSS) small bowel adenocarcinoma, loss of MARCKS expression was seen in 2 out of 20 tumors (10%). In conclusion, we herein present a cMSI profile of MSI-H small intestinal adenocarcinomas identifying MARCKS as a frequent target of mutation. Loss of MARCKS protein expression suggests a significant role of MARCKS inactivation in the pathogenesis of small intestinal adenocarcinomas.


Assuntos
Adenocarcinoma/genética , Neoplasias Intestinais/genética , Intestino Delgado/patologia , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas de Membrana/genética , Repetições de Microssatélites/genética , Metilação de DNA , Humanos , Imuno-Histoquímica , Mutação , Substrato Quinase C Rico em Alanina Miristoilada , Regiões Promotoras Genéticas
18.
Int J Oncol ; 35(1): 149-58, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19513562

RESUMO

Chemotherapy of non-Hodgkin's lymphoma is frequently hampered by drug resistance. The monoclonal antibody rituximab specifically targets the CD20 antigen and sensitizes B-cell lymphoma cells to standard anticancer drugs. In the present investigation, we analyzed, whether a combination of rituximab and artesunate may act in a complementary manner and eventually synergize in tumor cell killing. Artesunate is an anti-malarial drug, which also exerts profound activity towards cancer cells. While rituximab alone was minimally cytotoxic, rituximab increased cytotoxicity to artesunate in Ramos cells. Artesunate induced apoptosis, induced Fas/CD95 expression and the formation of reactive oxygen species (ROS) and resulted in a breakdown of mitochondrial membrane potential. This argues for the involvement of both receptor-driven extrinsic and mitochondrial intrinsic routes of apoptosis. Rituximab increased Fas/CD95 expression and ROS formation and decreased mitochondrial membrane potential ultimately leading to increased apoptosis induced by artesunate. The transcription factors YY1 and Sp1 are upstream regulators of apoptosis by controlling the expression of apoptosis-regulating genes. YY1 and Sp1 were down-regulated and Fas/CD95 was up-regulated by rituximab and artesunate indicating that artesunate activated the Fas/CD95 pathway and that rituximab increased the susceptibility of tumor cells to artesunate-induced apoptosis. Furthermore, rituximab affected the expression of antioxidant genes. The antibody decreased artesunate-induced up-regulation of catalase expression and increased artesunate-induced down-regulation of glutathione S-transferase-phi expression. Manganese-dependent superoxide dismutase expression was not changed by artesunate. Antioxidant proteins may help to detoxify artesunate-induced ROS. Rituximab reversed the artesunate-induced expression changes of antioxidant genes and, hence, reduced the detoxification capacity of Ramos cells. The effects of rituximab on antioxidant genes represent a novel mechanism of rituximab for chemosensitization.


Assuntos
Antígenos CD20/imunologia , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Apoptose/efeitos dos fármacos , Linfoma de Células B/patologia , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais Murinos , Antimaláricos/administração & dosagem , Artemisininas/administração & dosagem , Artesunato , Proteína Agonista de Morte Celular de Domínio Interatuante com BH3/metabolismo , Catalase/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Glutationa S-Transferase pi/metabolismo , Humanos , Linfoma de Células B/imunologia , Linfoma de Células B/metabolismo , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Rituximab , Fator de Transcrição Sp1/metabolismo , Superóxido Dismutase/metabolismo , Fator de Transcrição YY1/metabolismo , Receptor fas/metabolismo
19.
Cancer Res ; 69(6): 2234-43, 2009 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-19258502

RESUMO

Dynamic instability of the microtubule network modulates processes such as cell division and motility, as well as cellular morphology. Overexpression of the microtubule-destabilizing phosphoprotein stathmin is frequent in human malignancies and represents a promising therapeutic target. Although stathmin inhibition gives rise to antineoplastic effects, additional and functionally redundant microtubule-interacting proteins may attenuate the efficiency of this therapeutic approach. We have systematically analyzed the expression and potential protumorigenic effects of stathmin family members in human non-small cell lung cancer (NSCLC). Both stathmin and stathmin-like 3 (SCLIP) were overexpressed in adenocarcinoma as well as squamous cell carcinoma (SCC) tissues and induced tumor cell proliferation, migration, and matrix invasion in respective cell lines. Accordingly, reduced stathmin and SCLIP levels affected cell morphology and were associated with a less malignant phenotype. Combined inhibition of both factors caused additive effects on tumor cell motility, indicating partial functional redundancy. Because stathmin and SCLIP expression significantly correlated in NSCLC tissues, we searched for common upstream regulators and identified the far upstream sequence element-binding protein-1 (FBP-1) as a pivotal inducer of several stathmin family members. Our results indicate that the coordinated overexpression of microtubule-destabilizing factors by FBP-1 is a critical step to facilitate microtubule dynamics and subsequently increases proliferation and motility of tumor cells.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/metabolismo , DNA Helicases/metabolismo , Proteínas de Ligação a DNA/metabolismo , Neoplasias Pulmonares/metabolismo , Estatmina/biossíntese , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Processos de Crescimento Celular/fisiologia , Movimento Celular/fisiologia , Sobrevivência Celular/fisiologia , DNA Helicases/genética , DNA de Cadeia Simples/metabolismo , Proteínas de Ligação a DNA/genética , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Invasividade Neoplásica , RNA Interferente Pequeno/genética , Proteínas de Ligação a RNA , Estatmina/genética , Transfecção
20.
Apoptosis ; 13(3): 437-47, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18188704

RESUMO

The HIPPI (HIP-1 protein interactor) protein is a multifunctional protein that is involved in the regulation of apoptosis. The interaction partners of HIPPI include HIP-1 (Huntingtin-interacting protein-1), Apoptin, Homer1c, Rybp/DEDAF, and BAR (bifunctional apoptosis regulator). In search for other binding partners of HIPPI, we performed a yeast two hybrid screen and identified BLOC1S2 (Biogenesis of lysosome-related organelles complex-1 subunit 2) as a novel HIPPI-interacting protein. In co-immunoprecipitation assays, BLOC1S2 specifically associates with HIPPI, but not with HIP-1. To study the expression of BLOC1S2 on the protein level, we generated a mouse monoclonal antibody specific for BLOC1S2 and a multiple tissue array comprising 70 normal and cancer tissue samples of diverse origin. BLOC1S2 protein is widely expressed in normal tissue as well as in malignant tumors with a tendency towards lower expression levels in certain subtypes of tumors. On the subcellular level, BLOC1S2 is expressed in an organellar-like pattern and co-localizes with mitochondria. Over-expression of BLOC1S2 in the presence or absence of HIPPI does not induce apoptosis. However, BLOC1S2 and HIPPI sensitize NCH89 glioblastoma cells to the pro-apoptotic actions of staurosporine and the death ligand TRAIL by enhancing caspase activation, cytochrome c release, and disruption of the mitochondrial membrane potential. Given its interaction with HIPPI and its pro-apoptotic activity, BLOC1S2 might play an important functional role in cancer and neurodegenerative diseases.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Apoptose/efeitos dos fármacos , Glioblastoma/patologia , Proteínas/fisiologia , Adulto , Sequência de Aminoácidos , Animais , Caspase 3/metabolismo , Caspase 8/metabolismo , Linhagem Celular Tumoral , Citocromos c/metabolismo , Feminino , Células HeLa , Humanos , Imuno-Histoquímica , Masculino , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias/metabolismo , Dados de Sequência Molecular , Neoplasias/patologia , Ligação Proteica , Alinhamento de Sequência , Estaurosporina/farmacologia , Ligante Indutor de Apoptose Relacionado a TNF/fisiologia , Distribuição Tecidual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...