Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Cancer Immunol Res ; 7(5): 841-852, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30940644

RESUMO

Immune checkpoint blockade therapy has changed prognoses for many melanoma patients. However, immune responses that correlate with clinical progression of the disease are still poorly understood. To identify immune responses correlating with melanoma clinical evolution, we analyzed serum cytokines as well as circulating NK and T-cell subpopulations from melanoma patients. The patients' immune profiles suggested that melanoma progression leads to changes in peripheral blood NK and T-cell subsets. Stage IV melanoma was characterized by an increased frequency of CCR7+CD56bright NK cells as well as high serum concentrations of the CCR7 ligand CCL19. CCR7 expression and CCL19 secretion were also observed in melanoma cell lines. The CCR7+ melanoma cell subpopulation coexpressed PD-L1 and Galectin-9 and had stemness properties. Analysis of melanoma-derived cancer stem cells (CSC) showed high CCR7 expression; these CSCs were efficiently recognized and killed by NK cells. An accumulation of CCR7+, PD-L1+, and Galectin-9+ melanoma cells in melanoma metastases was demonstrated ex vivo Altogether, our data identify biomarkers that may mark a CCR7-driven metastatic melanoma pathway.


Assuntos
Células Matadoras Naturais/imunologia , Melanoma/imunologia , Antígeno B7-H1/imunologia , Linhagem Celular , Quimiocina CCL19/imunologia , Técnicas de Cocultura , Citocinas/sangue , Feminino , Galectinas/imunologia , Humanos , Masculino , Melanoma/sangue , Melanoma/patologia , Células-Tronco Neoplásicas/imunologia , Receptores CCR7/imunologia
2.
Int J Cancer ; 145(8): 2238-2248, 2019 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-31018250

RESUMO

Malignant mesothelioma (MM) is a highly aggressive form of cancer with limited treatment options. Although the role of NK cells has been studied in many solid tumors, the pattern of NK-cell subsets and their recognition of mesothelioma cells remain to be explored. We used RNA expression data of MM biopsies derived from the cancer genome atlas to evaluate the immune cell infiltrates. We characterized the phenotype of circulating NK and T cells of 27 MM patients before and after treatment with an anti-CTLA-4 antibody (tremelimumab). These immune cell profiles were compared to healthy controls. The RNA expression data of the MM biopsies indicated the presence of NK cells in a subgroup of patients. We demonstrated that NK cells recognize MM cell lines and that IL-15 stimulation improved NK cell-mediated lysis in vitro. Using multivariate projection models, we found that MM patients had a perturbed ratio of CD56bright and CD56dim NK subsets and increased serum concentrations of the cytokines IL-10, IL-8 and TNF-α. After tremelimumab treatment, the ratio between the CD56bright and CD56dim subsets shifted back towards physiological levels. Furthermore, the improved overall survival was correlated with low TIM-3+ CD8+ T-cell frequency, high DNAM-1+ CD56dim NK-cell frequency and high expression levels of NKp46 on the CD56dim NK cells before and after immune checkpoint blockade. Together, our observations suggest that NK cells infiltrate MM and that they can recognize and kill mesothelioma cells. The disease is associated with distinct lymphocytes patterns, some of which correlate with prognosis or are affected by treatment with tremelimumab.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Células Matadoras Naturais/imunologia , Neoplasias Pulmonares/tratamento farmacológico , Mesotelioma/tratamento farmacológico , Subpopulações de Linfócitos T/imunologia , Antineoplásicos/uso terapêutico , Antígeno CD56/imunologia , Antígeno CD56/metabolismo , Antígeno CTLA-4/antagonistas & inibidores , Antígeno CTLA-4/imunologia , Linhagem Celular Tumoral , Células Cultivadas , Citocinas/sangue , Citocinas/imunologia , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Células K562 , Estimativa de Kaplan-Meier , Células Matadoras Naturais/metabolismo , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/imunologia , Masculino , Mesotelioma/genética , Mesotelioma/imunologia , Mesotelioma Maligno , Pessoa de Meia-Idade , Prognóstico , Subpopulações de Linfócitos T/metabolismo
3.
Front Immunol ; 10: 224, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30873154

RESUMO

The ability of pathogens to sequester iron from their host cells and proteins affects their virulence. Moreover, iron is required for various innate host defense mechanisms as well as for acquired immune responses. Therefore, intracellular iron concentration may influence the interplay between pathogens and immune system. Here, we investigated whether changes in iron concentrations and intracellular ferritin heavy chain (FTH) abundance may modulate the expression of Major Histocompatibility Complex molecules (MHC), and susceptibility to Natural Killer (NK) cell cytotoxicity. FTH downregulation, either by shRNA transfection or iron chelation, led to MHC surface reduction in primary cancer cells and macrophages. On the contrary, mouse embryonic fibroblasts (MEFs) from NCOA4 null mice accumulated FTH for ferritinophagy impairment and displayed MHC class I cell surface overexpression. Low iron concentration, but not FTH, interfered with IFN-γ receptor signaling, preventing the increase of MHC-class I molecules on the membrane by obstructing STAT1 phosphorylation and nuclear translocation. Finally, iron depletion and FTH downregulation increased the target susceptibility of both primary cancer cells and macrophages to NK cell recognition. In conclusion, the reduction of iron and FTH may influence the expression of MHC class I molecules leading to NK cells activation.


Assuntos
Apoferritinas/metabolismo , Citotoxicidade Imunológica/imunologia , Antígenos de Histocompatibilidade Classe I/imunologia , Ferro/metabolismo , Células Matadoras Naturais/imunologia , Animais , Apoferritinas/genética , Linhagem Celular Tumoral , Células Cultivadas , Citotoxicidade Imunológica/genética , Desferroxamina/farmacologia , Embrião de Mamíferos/citologia , Fibroblastos/citologia , Fibroblastos/imunologia , Fibroblastos/metabolismo , Expressão Gênica/efeitos dos fármacos , Células HeLa , Antígenos de Histocompatibilidade Classe I/genética , Antígenos de Histocompatibilidade Classe I/metabolismo , Humanos , Interferon gama/farmacologia , Células K562 , Células Matadoras Naturais/metabolismo , Células MCF-7 , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Coativadores de Receptor Nuclear/genética , Coativadores de Receptor Nuclear/metabolismo , Interferência de RNA , Sideróforos/farmacologia
4.
Protein Sci ; 28(1): 150-166, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30242933

RESUMO

Human MICAL1 is a member of a recently discovered family of multidomain proteins that couple a FAD-containing monooxygenase-like domain to typical protein interaction domains. Growing evidence implicates the NADPH oxidase reaction catalyzed by the flavoprotein domain in generation of hydrogen peroxide as a second messenger in an increasing number of cell types and as a specific modulator of actin filaments stability. Several proteins of the Rab families of small GTPases are emerging as regulators of MICAL activity by binding to its C-terminal helical domain presumably shifting the equilibrium from the free - auto-inhibited - conformation to the active one. We here extend the characterization of the MICAL1-Rab8 interaction and show that indeed Rab8, in the active GTP-bound state, stabilizes the active MICAL1 conformation causing a specific four-fold increase of kcat of the NADPH oxidase reaction. Kinetic data and small-angle X-ray scattering (SAXS) measurements support the formation of a 1:1 complex between full-length MICAL1 and Rab8 with an apparent dissociation constant of approximately 8 µM. This finding supports the hypothesis that Rab8 is a physiological regulator of MICAL1 activity and shows how the protein region preceding the C-terminal Rab-binding domain may mask one of the Rab-binding sites detected with the isolated C-terminal fragment. SAXS-based modeling allowed us to propose the first model of the free full-length MICAL1, which is consistent with an auto-inhibited conformation in which the C-terminal region prevents catalysis by interfering with the conformational changes that are predicted to occur during the catalytic cycle.


Assuntos
Proteínas dos Microfilamentos/química , Oxigenases de Função Mista/química , Complexos Multiproteicos/química , Multimerização Proteica , Proteínas rab de Ligação ao GTP/química , Ativação Enzimática , Humanos , Proteínas dos Microfilamentos/metabolismo , Oxigenases de Função Mista/metabolismo , Complexos Multiproteicos/metabolismo , Espalhamento a Baixo Ângulo , Difração de Raios X , Proteínas rab de Ligação ao GTP/metabolismo
5.
Inflamm Bowel Dis ; 24(9): 1967-1977, 2018 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-29788407

RESUMO

BACKGROUND: Inflammatory bowel disease (IBD) is due to the interaction of genetic and environmental factors that trigger an unbalanced immune response ultimately resulting in the peculiar inflammatory reaction. Experimental models of IBD point to a role of T-cell-derived cytokines (Th17) and to SGK1 as mediator of the Th17 switch. We hypothesize that SGK1, a salt inducible kinase, directs lymphocytic behavior and tissue damage. METHODS: Eleven controls and 32 ulcerative colitis (UC) patients were randomized according to endoscopic Mayo score. Mucosal biopsies from different intestinal tracts were analyzed by immunohistochemistry and quantitative real-time polymerase chain reaction to check the expression of disease markers including SGK1. Peripheral blood mononuclear cells (PBMCs) from patients and controls were analyzed by fluorescence-activated cell sorting. Finally, an in vitro cell model was developed to test the hypothesis. RESULTS: SGK1 mRNA and protein expression in lesional areas of UC patients were lower than in normal peri-lesional areas of the same patients and in normal tissues of healthy controls. SGK1 expression was increased in PBMCs from UC patients, particularly in the CD4+ cell population, enriched in Th17 cells. IL17/IL13 was increased in patients and correlated with SGK1 expression. Genetically engineered Jurkat cells confirmed the effect of SGK1 overexpression on viability of RKO cells. CONCLUSIONS: These observations suggest a pathogenic mechanism whereby SGK1 overexpression in CD4+ T cells induces the secretion of the inflammatory cytokines IL17 and IL13, which downregulate the expression of SGK1 in target tissues. Our data suggest a novel hypothesis in the pathogenesis of UC, integrating colonic epithelial cells and lymphocytes.


Assuntos
Linfócitos T CD4-Positivos/metabolismo , Colite Ulcerativa/genética , Colite Ulcerativa/imunologia , Células Epiteliais/metabolismo , Proteínas Imediatamente Precoces/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Linhagem Celular , Colo/citologia , Colo/patologia , Regulação para Baixo , Humanos , Interleucina-13/metabolismo , Interleucina-17/metabolismo , Mucosa Intestinal/citologia , Leucócitos Mononucleares , RNA Mensageiro/metabolismo , Células Th17/imunologia
6.
Int J Mol Sci ; 18(4)2017 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-28353639

RESUMO

Genetic polymorphisms of the methylenetetrahydrofolate reductase (MTHFR) enzyme may influence DNA methylation. Alterations in DNA methylation patterns of genes involved in the regulation of the cell cycle, DNA repair, cell adherence and metastasis process are known to contribute to cancer development. In this study, the influence of the MTHFR C677T and A1298C gene polymorphisms on global DNA methylation and site-specific methylation on p16 and O6-methylguanine-DNA methyltransferase (MGMT) gene promoters was investigated in patients with oral squamous cell cancer (OSCC). To this aim, methylation studies were carried out by using genomic DNA isolated from saliva samples of 58 OSCC patients and 90 healthy controls. The frequency of the CT/AC and TT/AA genotypes was significantly higher in patients than in controls. Whereas no difference in global DNA methylation levels was observed between patients and controls, a higher frequency of methylation at both p16 and MGMT gene promoters was detected in patients compared with controls. A significant association between MTHFR gene polymorphisms and p16 and MGMT gene promoter methylation was found. The frequency of p16 and MGMT methylation was around 60% in patients with either the CT/AC or TT/AA genotype. Our results suggest that hypermethylation of cancer-related genes may be affected by MTHFR polymorphisms.


Assuntos
Carcinoma de Células Escamosas/genética , Metilação de DNA , Metilases de Modificação do DNA/genética , Enzimas Reparadoras do DNA/genética , Genes p16 , Metilenotetra-Hidrofolato Redutase (NADPH2)/genética , Neoplasias Bucais/genética , Polimorfismo de Nucleotídeo Único , Proteínas Supressoras de Tumor/genética , Adulto , Estudos de Casos e Controles , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Regiões Promotoras Genéticas
7.
Int J Cardiol ; 227: 331-334, 2017 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-27839822

RESUMO

BACKGROUND: Acute Myocardial infarction (AMI), a leading cause of morbidity and mortality worldwide, is a dreadful acute complication of coronary atherosclerosis. Type 2 diabetes mellitus (T2DM) is associated with an increased risk of developing AMI. The architectural transcription factor high-mobility-group AT-hook 1 (HMGA1) has been involved in atherosclerosis, plaque formation, inflammation, and in the pathogenesis of insulin resistance and T2DM. An association of the HMGA1 rs146052672 variant with T2DM has been recently reported. Thus, our aim was to evaluate whether this variant was also associated with AMI. METHODS AND RESULTS: In a case-control study from Calabria (Southern Italy), we enrolled 254 consecutive, unrelated, patients with first diagnosis of AMI, and 508 age, sex-matched controls. Genotyping of the rs146052672 was performed using the TaqMan allelic discrimination method. We found that this variant was present in 7.9% of AMI patients and in 3.1% of controls (p=0.003). Multiple logistic regression confirmed that the rs146052672 was significantly associated with AMI (OR=2.54; p=0.002), and this association was independent of classical cardiovascular risk factors such as gender, hypertension, obesity and T2DM (for all, p<0.05). CONCLUSIONS: Our findings demonstrate that a relationship exists between the HMGA1 rs146052672 variant and AMI, suggesting that defects at the HMGA1 locus may play a pathogenetic role in AMI, in the absence of T2DM and other cardiovascular risk factors.


Assuntos
Predisposição Genética para Doença/genética , Proteína HMGA1a/genética , Infarto do Miocárdio/genética , Idoso , Estudos de Casos e Controles , Feminino , Genótipo , Humanos , Itália , Masculino , Pessoa de Meia-Idade , Polimorfismo Genético/genética
8.
Sci Rep ; 6: 39429, 2016 12 19.
Artigo em Inglês | MEDLINE | ID: mdl-27991577

RESUMO

Diabetic retinopathy (DR) is a major complication of diabetes mellitus, and is the leading cause of blindness in working-age people. Usually, DR progresses from the asymptomatic non-proliferative DR that does not significantly alter vision, to proliferative DR (PDR), which can result in aberrant retinal neovessel formation and blindness. The High-Mobility-Group A1 (HMGA1) protein is a transcriptional master regulator of numerous genes, including metabolic and inflammatory genes, which, by modulating the expression of angiogenic factors, may induce retinal neovascularization, a hallmark of PDR. Herein, we examined the relationship between HMGA1 rs139876191 variant and DR. Results revealed that patients with type 2 diabetes, who were carriers of the HMGA1 rs139876191 variant had a significantly lower risk of developing PDR, compared to non-carrier diabetic patients. From a mechanistic point of view, our findings indicated that, by adversely affecting HMGA1 protein expression and function, the HMGA1 rs139876191 variant played a key role in this protective mechanism by downregulating the expression of vascular endothelial growth factor A (VEGFA), a major activator of neovascularization in DR. These data provide new insights into the pathogenesis and progression of DR, and may offer opportunities for discovering novel biomarkers and therapeutic targets for diagnosis, prevention and treatment of PDR.


Assuntos
Proliferação de Células/genética , Retinopatia Diabética/genética , Proteína HMGA1a/genética , Polimorfismo Genético/genética , Fator A de Crescimento do Endotélio Vascular/genética , Animais , Linhagem Celular Tumoral , Diabetes Mellitus Tipo 2/genética , Regulação para Baixo/genética , Células HEK293 , Células Hep G2 , Células Endoteliais da Veia Umbilical Humana , Humanos , Masculino , Camundongos , Neovascularização Patológica/genética , Retina/patologia , Neovascularização Retiniana/genética
9.
PLoS One ; 8(12): e83969, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24367622

RESUMO

The High-Mobility Group AT-Hook 1 (HMGA1) protein is an architectural transcription factor that binds to AT-rich sequences in the promoter region of DNA and functions as a specific cofactor for gene activation. Previously, we demonstrated that HMGA1 is a key regulator of the insulin receptor (INSR) gene and an important downstream target of the INSR signaling cascade. Moreover, from a pathogenic point of view, overexpression of HMGA1 has been associated with human cancer, whereas functional variants of the HMGA1 gene have been recently linked to type 2 diabetes mellitus and metabolic syndrome. However, despite of this biological and pathological relevance, the mechanisms that control HMGA1 gene expression remain unknown. In this study, to define the molecular mechanism(s) that regulate HMGA1 gene expression, the HMGA1 gene promoter was investigated by transient transfection of different cell lines, either before or after DNA and siRNA cotransfections. An octamer motif was identified as an important element of transcriptional regulation of this gene, the interaction of which with the octamer transcription factors Oct-1 and Oct-2 is crucial in modulating HMGA1 gene and protein expression. Additionally, we demonstrate that HMGA1 binds its own promoter and contributes to its transactivation by Oct-2 (but not Oct-1), supporting its role in an auto-regulatory circuit. Overall, our results provide insight into the transcriptional regulation of the HMGA1 gene, revealing a differential control exerted by both Oct-1 and Oct-2. Furthermore, they consistently support the hypothesis that a putative defect in Oct-1 and/or Oct-2, by affecting HMGA1 expression, may cause INSR dysfunction, leading to defects of the INSR signaling pathway.


Assuntos
Regulação da Expressão Gênica , Proteína HMGA1a/genética , Motivos de Nucleotídeos , Fator 1 de Transcrição de Octâmero/metabolismo , Fator 2 de Transcrição de Octâmero/metabolismo , Transcrição Gênica , Sequência de Bases , Sítios de Ligação , Núcleo Celular/metabolismo , Células HeLa , Células Hep G2 , Humanos , Regiões Promotoras Genéticas/genética , Ligação Proteica , Receptor de Insulina/genética
10.
Sci Rep ; 3: 1491, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23512162

RESUMO

The metabolic syndrome (MetS) is a common disorder, where systemic insulin-resistance is associated with increased risk for type 2 diabetes (T2D) and cardiovascular disease. Identifying genetic traits influencing risk and progression of MetS is important. We and others previously reported a functional HMGA1 gene variant, rs146052672, predisposing to T2D. Here we investigated the association of rs146052672 variant with MetS and related components. In a case-control study from Italy and Turkey, increased risk of MetS was seen among carriers of the HMGA1 variant. In the larger Italian cohort, this variant positively correlated with BMI, hyperglycemia and insulin-resistance, and negatively correlated with serum HDL-cholesterol. Association between rs146052672 variant and MetS occurred independently of T2D, indicating that HMGA1 gene defects play a pathogenetic role in MetS and other insulin-resistance-related conditions. Overall, our results indicate that the rs146052672 variant represents an early predictive marker of MetS, as well as a predictive tool for therapy.


Assuntos
Estudos de Associação Genética , Predisposição Genética para Doença , Proteína HMGA1a/genética , Síndrome Metabólica/genética , Polimorfismo de Nucleotídeo Único/genética , Estudos de Casos e Controles , Demografia , Feminino , Humanos , Resistência à Insulina/genética , Itália , Masculino , Pessoa de Meia-Idade , Fatores de Risco , Turquia
11.
Int J Artif Organs ; 35(9): 679-88, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22865477

RESUMO

OBJECTIVES: Leukocyte depletion (LD) has been reported to reduce inflammatory damage during cardiopulmonary bypass (CPB). We evaluated the role of LD in pulmonary function and inflammatory response. METHODS: Seventy consecutive CABG patients were randomized (1:1) to receive LD on both arterial and cardioplegia lines (Filters) or standard arterial filters (Controls) during CPB. Estimates of pulmonary function, inflammatory and anti-inflammatory cytokines were collected pre-, intra- and postoperatively. RESULTS: Hospital mortality, intensive care and in-hospital lengths of stay were similar. Although duration of ventilation and incidence of pneumonia were comparable, leukodepleted patients showed higher PaO2/FiO2 (p-between groups = 0.005; ICU arrival p = 0.023; 24 hours p = 0.039; 48 hours p<0.001) and lower need for postoperative non-invasive ventilation (NIV), (p = 0.029). Moreover, Filters showed lower inflammatory burst at 24 hours (IL-6 p<0.001; IL-8 p = 0.002) and 48 hours (IL-6 p = 0.015). This was associated with a lower release of the anti-inflammatory IL-10 (p-between groups = 0.030; ICU admission p = 0.002; 24 hours p = 0.003). Furthermore, IL-2 concentration proved higher in Filters (p-between groups = 0.013; ICU arrival p = 0.029; 24 hours p = 0.040; 48 hours p = 0.021) in association with lower leukocyte and platelet counts at ICU admission. CONCLUSIONS: LD resulted in lower inflammatory burst and less need for release of anti-inflammatory cytokines. Although hospital outcomes were similar in terms of mortality and length of stay, improvements in pulmonary function and reduced need for postoperative NIV support the use of LD.


Assuntos
Ponte Cardiopulmonar/métodos , Procedimentos de Redução de Leucócitos/métodos , Pulmão/fisiologia , Ventilação não Invasiva , Adulto , Idoso , Ponte Cardiopulmonar/mortalidade , Citocinas/sangue , Feminino , Mortalidade Hospitalar , Humanos , Tempo de Internação , Masculino , Pessoa de Meia-Idade , Período Pós-Operatório
12.
Mol Endocrinol ; 26(9): 1578-89, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22745191

RESUMO

We previously showed that loss of the high mobility group A1 (HMGA1) protein expression, induced in mice by disrupting the Hmga1 gene, considerably decreased insulin receptor expression in the major target tissues of insulin action, causing a type 2-like diabetic phenotype, in which, however, glucose intolerance was paradoxically associated with increased peripheral insulin sensitivity. Insulin hypersensitivity despite impairment of insulin action supports the existence of molecular adaptation mechanisms promoting glucose disposal via insulin-independent processes. Herein, we provide support for these compensatory pathways/circuits of glucose uptake in vivo, the activation of which under certain adverse metabolic conditions may protect against hyperglycemia. Using chromatin immunoprecipitation combined with protein-protein interaction studies of nuclear proteins in vivo, and transient transcription assays in living cells, we show that HMGA1 is required for gene activation of the IGF-binding proteins 1 (IGFBP1) and 3 (IGFBP3), two major members of the IGF-binding protein superfamily. Furthermore, by using positron emission tomography with (18)F-labeled 2-fluoro-2-deoxy-d-glucose, in combination with the euglycemic clamp with IGF-I, we demonstrated that IGF-I's bioactivity was increased in Hmga1-knockout mice, in which both skeletal muscle Glut4 protein expression and glucose uptake were enhanced compared with wild-type littermates. We propose that, by affecting the expression of both IGFBP protein species, HMGA1 can serve as a modulator of IGF-I activity, thus representing an important novel mediator of glucose disposal.


Assuntos
Proteínas HMGA/metabolismo , Proteínas de Ligação a Fator de Crescimento Semelhante a Insulina/metabolismo , Fator de Crescimento Insulin-Like I/metabolismo , Animais , Western Blotting , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Glucose , Proteínas HMGA/genética , Células Hep G2 , Humanos , Imunoprecipitação , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/genética , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/metabolismo , Proteína 3 de Ligação a Fator de Crescimento Semelhante à Insulina/genética , Proteína 3 de Ligação a Fator de Crescimento Semelhante à Insulina/metabolismo , Proteínas de Ligação a Fator de Crescimento Semelhante a Insulina/genética , Fator de Crescimento Insulin-Like I/genética , Camundongos , Camundongos Knockout , Células NIH 3T3 , Tomografia por Emissão de Pósitrons , Regiões Promotoras Genéticas/genética , Ligação Proteica , Reação em Cadeia da Polimerase Via Transcriptase Reversa
13.
Exp Diabetes Res ; 2012: 789174, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22701472

RESUMO

Insulin resistance is common in individuals with obesity or type 2 diabetes (T2D), in which circulating insulin levels are frequently increased. Recent epidemiological and clinical evidence points to a link between insulin resistance and cancer. The mechanisms for this association are unknown, but hyperinsulinaemia (a hallmark of insulin resistance) and the increase in bioavailable insulin-like growth factor I (IGF-I) appear to have a role in tumor initiation and progression in insulin-resistant patients. Insulin and IGF-I inhibit the hepatic synthesis of sex-hormone binding globulin (SHBG), whereas both hormones stimulate the ovarian synthesis of sex steroids, whose effects, in breast epithelium and endometrium, can promote cellular proliferation and inhibit apoptosis. Furthermore, an increased risk of cancer among insulin-resistant patients can be due to overproduction of reactive oxygen species (ROS) that can damage DNA contributing to mutagenesis and carcinogenesis. On the other hand, it is possible that the abundance of inflammatory cells in adipose tissue of obese and diabetic patients may promote systemic inflammation which can result in a protumorigenic environment. Here, we summarize recent progress on insulin resistance and cancer, focusing on various implicated mechanisms that have been described recently, and discuss how these mechanisms may contribute to cancer initiation and progression.


Assuntos
Regulação Neoplásica da Expressão Gênica , Resistência à Insulina , Neoplasias/patologia , Animais , Apoptose , Proliferação de Células , Dano ao DNA , Diabetes Mellitus Tipo 2/fisiopatologia , Feminino , Humanos , Fator de Crescimento Insulin-Like I/metabolismo , Modelos Biológicos , Neoplasias/diagnóstico , Neoplasias/fisiopatologia , Obesidade/fisiopatologia , Ovário/metabolismo , Espécies Reativas de Oxigênio , Risco , Globulina de Ligação a Hormônio Sexual/metabolismo , Esteroides/metabolismo
14.
Circ J ; 76(5): 1121-9, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22447003

RESUMO

BACKGROUND: We aimed to evaluate if the use of an intra-aortic balloon pump (IABP) during cardioplegic arrest improves organ function and reduces endothelial activation in patients undergoing coronary artery bypass graft (CABG). METHODS AND RESULTS: Five-hundred and one CABG patients were randomized into 2 groups: (Group A n=270) linear cardiopulmonary bypass (CPB); and (Group B n=231) automatic 80 beats/min IABP-induced pulsatile CPB. We evaluated hemodynamic response, coagulation and fibrinolysis, transaminase, bilirubin, amylase, lactate, renal function (estimated glomerular filtration rate [eGFR], creatinine and any possibility of renal insufficiency or failure), respiratory function and endothelial markers (vascular endothelial growth factor [VEGF] and monocyte chemotactic protein-1 [MCP-1]). IABP, which induced surplus hemodynamic energy, was 21,387 ± 4,262 ergs/cm(3). Group B showed lower chest drainage, transfusions, international normalized ratio, and antithrombin III, together with higher platelets, activated partial thromboplastin time, fibrinogen and D-dimer. Transaminases, bilirubin, amylase, lactate were lower in Group B; there were better results for eGFR in Group B from ICU-arrival to 48 h, resulting in lower creatinine from ICU-arrival to 48 h. The necessity for renal replacement therapy was lower in Group B Stage-3. Group B P(a)O(2)/F(i)O(2) and lung compliance improved with aortic de-clamping on the first day with shorter intubation time. Group B showed lower VEGF and MCP-1. CONCLUSIONS: Pulsatile flow by IABP improves whole-body perfusion and reduces endothelial activation during CPB.


Assuntos
Ponte Cardiopulmonar , Ponte de Artéria Coronária , Endotélio Vascular/metabolismo , Endotélio Vascular/fisiopatologia , Balão Intra-Aórtico , Idoso , Biomarcadores/sangue , Feminino , Fibrinólise , Taxa de Filtração Glomerular , Hemodinâmica , Humanos , Masculino
15.
Vaccine ; 28(47): 7551-5, 2010 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-20837078

RESUMO

Burkholderia thailandensis is a less virulent close relative of Burkholderia pseudomallei, a CDC category B biothreat agent. We have previously shown that lipopolysaccharide (LPS) extracted from B. pseudomallei can provide protection against a lethal challenge of B. pseudomallei in a mouse model of melioidosis. Sugar analysis on LPS from B. thailandensis strain E264 confirmed that this polysaccharide has a similar structure to LPS from B. pseudomallei. Mice were immunised with LPS from B. thailandensis or B. pseudomallei and challenged with a lethal dose of B. pseudomallei strain K96243. Similar protection levels were observed when either LPS was used as the immunogen. This data suggests that B. thailandensis LPS has the potential to be used as part of a subunit based vaccine against pathogenic B. pseudomallei.


Assuntos
Burkholderia pseudomallei/patogenicidade , Burkholderia/patogenicidade , Lipopolissacarídeos/imunologia , Melioidose/prevenção & controle , Animais , Anticorpos Antibacterianos/sangue , Antígenos de Bactérias/imunologia , Antígenos de Bactérias/isolamento & purificação , Vacinas Bacterianas/imunologia , Burkholderia/imunologia , Burkholderia pseudomallei/imunologia , Modelos Animais de Doenças , Feminino , Lipopolissacarídeos/isolamento & purificação , Melioidose/imunologia , Camundongos , Camundongos Endogâmicos BALB C
16.
J Bacteriol ; 192(21): 5572-9, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20802039

RESUMO

Like the Eukarya and Bacteria, the Archaea also perform N glycosylation. Using the haloarchaeon Haloferax volcanii as a model system, a series of Agl proteins involved in the archaeal version of this posttranslational modification has been identified. In the present study, the participation of HVO_1517 in N glycosylation was considered, given its homology to a known component of the eukaryal N-glycosylation pathway and because of the genomic proximity of HVO_1517 to agl genes encoding known elements of the H. volcanii N-glycosylation process. By combining the deletion of HVO_1517 with mass spectrometric analysis of both dolichol phosphate monosaccharide-charged carriers and the S-layer glycoprotein, evidence was obtained showing the participation of HVO_1517, renamed AglJ, in adding the first hexose of the N-linked pentasaccharide decorating this reporter glycoprotein. The deletion of aglJ, however, did not fully prevent the attachment of a hexose residue to the S-layer glycoprotein. Moreover, in the absence of AglJ, the level of only one of the three monosaccharide-charged dolichol phosphate carriers detected in the cell was reduced. Nonetheless, in cells lacking AglJ, no further sugar subunits were added to the remaining monosaccharide-charged dolichol phosphate carriers or to the monosaccharide-modified S-layer glycoprotein, pointing to the importance of the sugar added through the actions of AglJ for proper N glycosylation. Finally, while aglJ can be deleted, H. volcanii surface layer integrity is compromised in the absence of the encoded protein.


Assuntos
Proteínas Arqueais/metabolismo , Metabolismo dos Carboidratos , Regulação da Expressão Gênica em Archaea/fisiologia , Haloferax volcanii/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas Arqueais/genética , Proteínas de Transporte/metabolismo , Deleção de Genes , Glicosilação , Haloferax volcanii/genética , Hexoses/metabolismo , Dados de Sequência Molecular , Estrutura Molecular
17.
Mol Microbiol ; 75(4): 1047-58, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20487296

RESUMO

Like Eukarya and Bacteria, Archaea are also capable of performing N-glycosylation. In the halophilic archaeon Haloferax volcanii, N-glycosylation is mediated by the products of the agl gene cluster. In the present report, this gene cluster was expanded to include an additional sequence, aglM, shown to participate in the biosynthesis of hexuronic acids contained within a pentasaccharide decorating the S-layer glycoprotein, a reporter H. volcanii glycoprotein. In response to different growth conditions, changes in the transcription profile of aglM mirrored changes in the transcription profiles of aglF, aglG and aglI, genes encoding confirmed participants in the H. volcanii N-glycosylation pathway, thus offering support to the hypothesis that in H. volcanii, N-glycosylation serves an adaptive role. Following purification, biochemical analysis revealed AglM to function as a UDP-glucose dehydrogenase. In a scoupled reaction with AglF, a previously identified glucose-1-phosphate uridyltransferase, UDP-glucuronic acid was generated from glucose-1-phosphate and UTP in a NAD(+)-dependent manner. These experiments thus represent the first step towards in vitro reconstitution of the archaeal N-glycosylation process.


Assuntos
Proteínas Arqueais/metabolismo , Glicoproteínas/metabolismo , Haloferax volcanii/metabolismo , Glicoproteínas/genética , Glicosilação , Haloferax volcanii/genética , Haloferax volcanii/crescimento & desenvolvimento , Família Multigênica , Transcrição Gênica
18.
Mol Microbiol ; 76(1): 190-9, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20149102

RESUMO

While pathways for N-glycosylation in Eukarya and Bacteria have been solved, considerably less is known of this post-translational modification in Archaea. In the halophilic archaeon Haloferax volcanii, proteins encoded by the agl genes are involved in the assembly and attachment of a pentasaccharide to select asparagine residues of the S-layer glycoprotein. AglP, originally identified based on the proximity of its encoding gene to other agl genes whose products were shown to participate in N-glycosylation, was proposed, based on sequence homology, to serve as a methyltransferase. In the present report, gene deletion and mass spectrometry were employed to reveal that AglP is responsible for adding a 14 Da moiety to a hexuronic acid found at position four of the pentasaccharide decorating the Hfx. volcanii S-layer glycoprotein. Subsequent purification of a tagged version of AglP and development of an in vitro assay to test the function of the protein confirmed that AglP is a S-adenosyl-L-methionine-dependent methyltransferase.


Assuntos
Proteínas Arqueais/metabolismo , Glicosilação , Haloferax volcanii/enzimologia , Metiltransferases/metabolismo , Selenometionina/análogos & derivados , Proteínas Arqueais/genética , Proteínas Arqueais/isolamento & purificação , Deleção de Genes , Haloferax volcanii/genética , Ácidos Hexurônicos/metabolismo , Espectrometria de Massas , Metiltransferases/genética , Metiltransferases/isolamento & purificação , Selenometionina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA