Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Ocul Immunol Inflamm ; : 1-4, 2023 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-37364062

RESUMO

AIM: To study parasitic eye diseases in a tertiary institute of North-east India by live examination of parasites, rapid staining, and scanning electron microscopy (SEM). METHODS: A 12-year retrospective analysis was performed and all patients diagnosed with ocular parasitic diseases were identified. Examination under a compound microscope, fluorescein staining, and scanning electron microscopy were done. RESULTS: A total of 160 ocular parasitosis cases were identified. The cases for which rapid staining and SEM studies were done included Cysticercosis (n = 18, 11.25%), Hydatidosis (n = 5, 3.13%), Dirofilariasis (n = 5, 3.13%), Thelaziasis (n = 3, 1.87%), and Gnathostomiasis (n = 2, 1.25%). Live examination was performed in 11 cases (6.63%) and 8 cases (4.82%) underwent scanning electron microscopy. . CONCLUSION: Fluorescein staining for identification of parasites and SEM study helped in detailing microscopic and ultrastructural findings.

2.
Front Immunol ; 11: 617767, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33679705

RESUMO

The inhibition of Fcγ receptors (FcγR) is an attractive strategy for treating diseases driven by IgG immune complexes (IC). Previously, we demonstrated that an engineered tri-valent arrangement of IgG1 Fc domains (SIF1) potently inhibited FcγR activation by IC, whereas a penta-valent Fc molecule (PentX) activated FcγR, potentially mimicking ICs and leading to Syk phosphorylation. Thus, a precise balance exists between the number of engaged FcγRs for inhibition versus activation. Here, we demonstrate that Fc valency differentially controls FcγR activation and inhibition within distinct subcellular compartments. Large Fc multimer clusters consisting of 5-50 Fc domains predominately recruited Syk-mScarlet to patches on the plasma membrane, whereas PentX exclusively recruited Syk-mScarlet to endosomes in human monocytic cell line (THP-1 cells). In contrast, SIF1, similar to monomeric Fc, spent longer periods docked to FcγRs on the plasma membrane and did not accumulate and recruit Syk-mScarlet within large endosomes. Single particle tracking (SPT) of fluorescent engineered Fc molecules and Syk-mScarlet at the plasma membrane imaged by total internal reflection fluorescence microscopy (SPT-TIRF), revealed that Syk-mScarlet sampled the plasma membrane was not recruited to FcγR docked with any of the engineered Fc molecules at the plasma membrane. Furthermore, the motions of FcγRs docked with recombinant Fc (rFc), SIF1 or PentX, displayed similar motions with D ~ 0.15 µm2/s, indicating that SIF1 and PentX did not induce reorganization or microclustering of FcγRs beyond the ligating valency. Multicolor SPT-TIRF and brightness analysis of docked rFc, SIF1 and PentX also indicated that FcγRs were not pre-assembled into clusters. Taken together, activation on the plasma membrane requires assembly of more than 5 FcγRs. Unlike rFc or SIF1, PentX accumulated Syk-mScarlet on endosomes indicating that the threshold for FcγR activation on endosomes is lower than on the plasma membrane. We conclude that the inhibitory effects of SIF1 are mediated by stabilizing a ligated and inactive FcγR on the plasma membrane. Thus, FcγR inhibition can be achieved by low valency ligation with SIF1 that behaves similarly to FcγR docked with monomeric IgG.


Assuntos
Fragmentos Fc das Imunoglobulinas/imunologia , Imunoglobulina G/imunologia , Fagocitose/imunologia , Receptores de IgG/metabolismo , Complexo Antígeno-Anticorpo/imunologia , Endossomos/imunologia , Humanos , Macrófagos/imunologia , Transdução de Sinais/imunologia
3.
Oncotarget ; 5(17): 7272-84, 2014 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-25261375

RESUMO

Mucins are high molecular weight O-glycoproteins that are predominantly expressed at the apical surface of epithelial cells and have wide range of functions. The functional diversity is attributed to their structure that comprises of a peptide chain with unique domains and multiple carbohydrate moieties added during posttranslational modifications. Tumor cells aberrantly overexpress mucins, and thereby promote proliferation, differentiation, motility, invasion and metastasis. Along with their aberrant expression, accumulating evidence suggest the critical role of altered subcellular localization of mucins under pathological conditions due to altered endocytic processes. The mislocalization of mucins and their interactions result in change in the density and activity of important cell membrane proteins (like, receptor tyrosine kinases) to facilitate various signaling, which help cancer cells to proliferate, survive and progress to more aggressive phenotype. In this review article, we summarize studies on mucins trafficking and provide a perspective on its importance to pathological conditions and to answer critical questions including its use for therapeutic interventions.


Assuntos
Mucinas/metabolismo , Neoplasias/metabolismo , Animais , Humanos , Neoplasias/patologia , Transporte Proteico
4.
PLoS One ; 8(4): e61857, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23626741

RESUMO

The formation and maintenance of cell-cell junctions, both under physiological and pathological conditions, requires the targeting and trafficking of junctional proteins. Proteins of the syntaxin (Stx)-family localize to a variety of subcellular membranes and contribute to intracellular transport of cargo by regulating vesicle fusion events at these sites. Unlike plasma membrane localized Stxs, the roles of endosome- and Golgi-localized stx proteins in epithelial morphogenesis are less understood. Here we show that Stx16- an endosome- and Golgi-localized target-membrane soluble N-ethylmaleimide attachment protein receptor (t-SNARE) that plays a role in membrane trafficking between these compartments - is essential for lumen development. In cultured Madin Darby Canine Kidney (MDCK) cells, Stx16 was selectively upregulated as sparsely plated cells attained confluency. Stx16-depleted confluent monolayers consistently showed lower transepithelial resistance than control monolayers, and failed to maintain endogenous and ectopically expressed E-cadherin at the adherens junctions due to decreased recycling. We further found that whereas cysts formed by MDCK cells cultured in Matrigel have a single hollow lumen, those formed by stx16-depleted counterparts had multiple lumens, due to abnormal orientiation of the mitotic spindle. Finally, a similar role for stx16 function in vivo is indicated by our analysis of pronephric-duct development in zebrafish expressing the claudinB:lynGFP transgene; lack of stx16 function in this structure (in stx16-morphant embryos) led to the development of enlarged, torturous pronephric ducts with more than one lumen. Taken together, our in vitro and in vivo studies establish a role for Stx16 in maintaining the integrity of cell-cell junctions, and thereby in morphogenesis of the kidney epithelial lumen.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Junções Intercelulares/metabolismo , Rim/metabolismo , Sintaxina 16/metabolismo , Animais , Caderinas/genética , Caderinas/metabolismo , Contagem de Células , Membrana Celular/metabolismo , Membrana Celular/ultraestrutura , Colágeno , Cães , Combinação de Medicamentos , Embrião não Mamífero , Endossomos/metabolismo , Endossomos/ultraestrutura , Complexo de Golgi/metabolismo , Complexo de Golgi/ultraestrutura , Junções Intercelulares/genética , Junções Intercelulares/ultraestrutura , Rim/crescimento & desenvolvimento , Rim/ultraestrutura , Laminina , Células Madin Darby de Rim Canino , Transporte Proteico , Proteoglicanas , Proteínas SNARE/genética , Proteínas SNARE/metabolismo , Transdução de Sinais , Fuso Acromático/metabolismo , Fuso Acromático/ultraestrutura , Sintaxina 16/genética , Transgenes , Peixe-Zebra
5.
PLoS One ; 7(9): e44572, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22962618

RESUMO

The soluble form of vascular endothelial growth factor receptor 1 (sVEGFR-1/sFlt1) is generated by alternative splicing of the FLT1 gene. Secretion of sFlt1 from endothelial cells plays an important role in blood vessel sprouting and morphogenesis. However, excess sFlt1 secretion is associated with diseases such as preeclampsia and chronic kidney disease. To date, the secretory transport process involved in the secretion of sFlt1 is poorly understood. In the present study, we investigated the itinerary of sFlt1 trafficking along the secretory pathway. To understand the timecourse of sFlt1 secretion, endothelial cells stably expressing sFlt1 were metabolically radiolabeled with [(35)S]-methionine and cysteine. Our results indicate that after initial synthesis the levels of secreted [(35)S]-sFlt1 in the extracellular medium peaks at 8 hours. Treatment with brefeldin A (BFA), a drug which blocks trafficking between the endoplasmic reticulum (ER) and the Golgi complex, inhibited extracellular release of sFlt1 suggesting that ER to Golgi and intra-Golgi trafficking of sFlt1 are essential for its secretion. Furthermore, we show that ectopic expression of dominant-negative mutant forms of Arf1, Arf6, and Rab11 as well as siRNA-mediated knockdown of these GTPases block secretion of sFlt1 during normoxic and hypoxic conditions suggesting role for these small GTPases. This work is the first to report role of regulatory proteins involved in sFlt1 trafficking along the secretory pathway and may provide insights and new molecular targets for the modulation of sFlt-1 release during physiological and pathological conditions.


Assuntos
Fator 1 de Ribosilação do ADP/genética , Fatores de Ribosilação do ADP/genética , Retículo Endoplasmático/metabolismo , Complexo de Golgi/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/genética , Proteínas rab de Ligação ao GTP/genética , Fator 1 de Ribosilação do ADP/antagonistas & inibidores , Fator 1 de Ribosilação do ADP/metabolismo , Fator 6 de Ribosilação do ADP , Fatores de Ribosilação do ADP/antagonistas & inibidores , Fatores de Ribosilação do ADP/metabolismo , Aminoácidos/metabolismo , Brefeldina A/farmacologia , Linhagem Celular Tumoral , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/genética , Expressão Gênica/efeitos dos fármacos , Complexo de Golgi/efeitos dos fármacos , Complexo de Golgi/genética , Células Endoteliais da Veia Umbilical Humana , Humanos , Plasmídeos , Estrutura Terciária de Proteína , Inibidores da Síntese de Proteínas/farmacologia , Transporte Proteico/efeitos dos fármacos , RNA Interferente Pequeno/genética , Transdução de Sinais/efeitos dos fármacos , Solubilidade , Radioisótopos de Enxofre , Transfecção , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Proteínas rab de Ligação ao GTP/antagonistas & inibidores , Proteínas rab de Ligação ao GTP/metabolismo
6.
Biochem J ; 444(3): 515-27, 2012 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-22471522

RESUMO

IL (interleukin)-6, an established growth factor for multiple myeloma cells, induces myeloma therapy resistance, but the resistance mechanisms remain unclear. The present study determines the role of IL-6 in re-establishing intracellular redox homoeostasis in the context of myeloma therapy. IL-6 treatment increased myeloma cell resistance to agents that induce oxidative stress, including IR (ionizing radiation) and Dex (dexamethasone). Relative to IR alone, myeloma cells treated with IL-6 plus IR demonstrated reduced annexin/propidium iodide staining, caspase 3 activation, PARP [poly(ADP-ribose) polymerase] cleavage and mitochondrial membrane depolarization with increased clonogenic survival. IL-6 combined with IR or Dex increased early intracellular pro-oxidant levels that were causally related to activation of NF-κB (nuclear factor κB) as determined by the ability of N-acetylcysteine to suppress both pro-oxidant levels and NF-κB activation. In myeloma cells, upon combination with hydrogen peroxide treatment, relative to TNF (tumour necrosis factor)-α, IL-6 induced an early perturbation in reduced glutathione level and increased NF-κB-dependent MnSOD (manganese superoxide dismutase) expression. Furthermore, knockdown of MnSOD suppressed the IL-6-induced myeloma cell resistance to radiation. MitoSOX Red staining showed that IL-6 treatment attenuated late mitochondrial oxidant production in irradiated myeloma cells. The present study provides evidence that increases in MnSOD expression mediate IL-6-induced resistance to Dex and radiation in myeloma cells. The results of the present study indicate that inhibition of antioxidant pathways could enhance myeloma cell responses to radiotherapy and/or chemotherapy.


Assuntos
Resistencia a Medicamentos Antineoplásicos/fisiologia , Interleucina-6/fisiologia , Mieloma Múltiplo/enzimologia , Mieloma Múltiplo/terapia , Estresse Oxidativo/fisiologia , Superóxido Dismutase/biossíntese , Regulação para Cima/fisiologia , Linhagem Celular , Linhagem Celular Tumoral , Humanos , Mieloma Múltiplo/tratamento farmacológico , Espécies Reativas de Oxigênio/metabolismo
7.
Blood ; 117(4): 1425-35, 2011 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-21063020

RESUMO

Vascular endothelial growth factor receptor 2 (VEGFR2) plays a key role in physiologic and pathologic angiogenesis. Plasma membrane (PM) levels of VEGFR2 are regulated by endocytosis and secretory transport through the Golgi apparatus. To date, the mechanism whereby the VEGFR2 traffics through the Golgi apparatus remains incompletely characterized. We show in human endothelial cells that binding of VEGF to the cell surface localized VEGFR2 stimulates exit of intracellular VEGFR2 from the Golgi apparatus. Brefeldin A treatment reduced the level of surface VEGFR2, confirming that VEGFR2 traffics through the Golgi apparatus en route to the PM. Mechanistically, we show that inhibition of syntaxin 6, a Golgi-localized target membrane-soluble N-ethylmaleimide attachment protein receptor (t-SNARE) protein, interferes with VEGFR2 trafficking to the PM and facilitates lysosomal degradation of the VEGFR2. In cell culture, inhibition of syntaxin 6 also reduced VEGF-induced cell proliferation, cell migration, and vascular tube formation. Furthermore, in a mouse ear model of angiogenesis, an inhibitory form of syntaxin 6 reduced VEGF-induced neovascularization and permeability. Our data demonstrate the importance of syntaxin 6 in the maintenance of cellular VEGFR2 levels, and suggest that the inhibitory form of syntaxin 6 has good potential as an antiangiogenic agent.


Assuntos
Complexo de Golgi/metabolismo , Neovascularização Fisiológica/fisiologia , Proteínas Qa-SNARE/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Animais , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Células Cultivadas , Humanos , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Camundongos , Camundongos Nus , Neovascularização Fisiológica/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Proteínas Qa-SNARE/antagonistas & inibidores , Proteínas Qa-SNARE/genética , Proteínas Qa-SNARE/fisiologia , Proteínas SNARE/antagonistas & inibidores , Proteínas SNARE/genética , Proteínas SNARE/metabolismo , Proteínas SNARE/fisiologia , Transfecção , Fator A de Crescimento do Endotélio Vascular/farmacologia
8.
Neoplasia ; 12(12): 980-92, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21170263

RESUMO

Dexamethasone (Dex) and radiation therapy are established modalities in multiple myeloma. In this study, we propose a novel combination of Dex plus radiation that shows superior clonogenic cell killing and apoptosis of myeloma cells and selectively eliminates myeloma cells when cocultured with bone marrow stromal cells (BMSCs). Dex was found to inhibit the release of interleukin-6 from irradiated BMSCs, which is an established myeloma cell proproliferative cytokine. In 5TGM1 model, the combination of Dex with skeletal targeted radiotherapy (153-Sm-EDTMP) prolonged median survival time and inhibited radiation-induced myelosuppression. A two-cycle treatment of Dex plus 153-Sm-EDTMP was well tolerated and further improved median survival time. Mechanistically, Dex increased superoxide and hydrogen peroxide production and augmented radiation-induced oxidative stress and cell death of myeloma cells. In contrast, Dex inhibited radiation-induced increase in pro-oxidant levels and enhanced the clonogenic survival in normal hematopoietic stem and progenitor cells. Treatment with either N-acetylcysteine or the combination of polyethylene glycol (PEG)-conjugated copper, zinc-superoxide dismutase, and PEG-catalase significantly protected myeloma cells from Dex-induced clonogenic death. Overall, these results demonstrate that Dex in combination with radiotherapy enhances the killing of myeloma cells while protecting normal bone marrow hematopoiesis through a mechanism that involves selective increases in oxidative stress.


Assuntos
Dexametasona/farmacologia , Hematopoese , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/radioterapia , Estresse Oxidativo , Células Estromais/metabolismo , Acetilcisteína/metabolismo , Acetilcisteína/uso terapêutico , Animais , Apoptose , Western Blotting , Células da Medula Óssea/metabolismo , Terapia Combinada , Dexametasona/metabolismo , Dexametasona/uso terapêutico , Ensaio de Imunoadsorção Enzimática , Hematopoese/efeitos dos fármacos , Hematopoese/efeitos da radiação , Interleucina-6/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/efeitos da radiação , Plasmócitos/efeitos dos fármacos , Polietilenoglicóis/uso terapêutico , Tolerância a Radiação/efeitos dos fármacos , Protetores contra Radiação/farmacologia , Espécies Reativas de Oxigênio , Células Estromais/efeitos dos fármacos , Células Estromais/efeitos da radiação , Superóxido Dismutase/metabolismo , Superóxido Dismutase/uso terapêutico , Células Tumorais Cultivadas
9.
J Med Case Rep ; 2: 110, 2008 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-18423008

RESUMO

INTRODUCTION: A duodenal Gastrointestinal Stromal Tumour (GIST) is a rare finding and until recently advanced disease had a poor prognosis. A PubMed search revealed no reports of more than five years survival of inoperable GIST on chemotherapy with WHO performance status zero. CASE PRESENTATION: A 68 year old female was diagnosed with unresectable GIST in the duodenum with metastasis to liver, pancreas and omentum in November 2001. She was commenced on imatinib mesylate (Glivec) chemotherapy. This case report was prepared from the medical records and radiology reports. She had good tolerance with stable disease. After six years her CT scan showed no disease progression and her WHO performance status was zero. CONCLUSION: This report supports the view that imatinib is a safe and effective drug in controlling disease progression in advanced metastatic GIST and plays an important role in improving the patient's quality of life.

10.
J Cell Sci ; 120(Pt 3): 447-55, 2007 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-17213331

RESUMO

Most patients with cystic fibrosis (CF) have a single codon deletion (DeltaF508) in the gene encoding the cystic fibrosis transmembrane conductance regulator (CFTR) that impairs assembly of the multidomain glycoprotein. The mutant protein escapes endoplasmic reticulum (ER) quality control at low temperature, but is rapidly cleared from the distal secretory pathway and degraded in lysosomes. CF cells accumulate free cholesterol similar to Niemann-Pick disease type C cells. We show that this lipid alteration is caused by the presence of misassembled mutant CFTR proteins, including DeltaF508, in the distal secretory pathway rather than the absence of functional CFTR. By contrast, cholesterol distribution is not changed by either D572N CFTR, which does not mature even at low temperature, or G551D, which is processed normally but is inactive. On expression of the DeltaF508 mutant, cholesterol and glycosphingolipids accumulate in punctate endosomal structures and cholesterol esters are reduced, indicating a block in the translocation of cholesterol to the ER for esterification. This is overcome by Rab9 overexpression, resulting in clearance of accumulating intracellular cholesterol. Similar but less pronounced alterations in intracellular cholesterol distribution are observed on expression of a temperature-rescued mutant variant of the related ATP-binding cassette (ABC) protein multidrug resistance-associated protein 1 (MRP1). Thus, on escape from ER quality control, misassembled mutants of CFTR and MRP1 impair lipid homeostasis in endocytic compartments.


Assuntos
Membrana Celular/metabolismo , Colesterol/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Endocitose/fisiologia , Animais , Células CHO , Linhagem Celular , Cricetinae , Cricetulus , Endossomos/metabolismo , Deleção de Genes , Glicoesfingolipídeos/metabolismo , Mutagênese Sítio-Dirigida , Mutação , Transfecção
11.
Am J Gastroenterol ; 101(10): 2319-29, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17032197

RESUMO

OBJECTIVE: The human MUC4 mucin plays an important role in the pathogenesis of pancreatic cancer. Recently, we have demonstrated that MUC4 expression in pancreatic tumor cells is regulated by interferon-gamma (IFNgamma) and by retinoic acid via transforming growth factor beta 2 (TGFbeta-2). In the present study, we established the pathobiological association of various cytokines and MUC4 in pancreatic tumor tissues and tumor cell lines. METHODS: Using semiquantitative reverse transcription-polymerase chain reaction (RT-PCR) and/or immunohistochemical analyses, we examined the expression of MUC4, IFNgamma, TGFbetas, and several immunologically relevant cytokines in a panel of 11 pancreatic adenocarcinomas (PA), three normal pancreatic (NP) tissue specimens, and 11 pancreatic tumor cell lines. RESULTS: Our data revealed that both MUC4 and IFNgamma were expressed at moderate to high levels in the majority of PA, while being undetectable in NP. Moreover, transcript for interleukin 2 (IL-2), a known marker of activated T helper 1 (TH1) lymphocytes, exhibited an expression profile similar to IFNgamma, suggesting a role of these immune effector cells as a potential source of IFNgamma in PA. Of note, IFNgamma protein was detected in the inflamed tissues neighboring tumor areas. Furthermore, TGFbetas were expressed by most cell lines and frequently upregulated in PA compared with NP. Interestingly, both IL-12 and IL-10, two key cytokines of the TH1 and TH2 pathways, respectively, were expressed at higher levels in PA relative to NP. CONCLUSIONS: These observations support the potential implication of IFNgamma and TGFbetas in MUC4 regulation in vivo and suggest a complex interaction of TH1 and TH2 signaling in the pancreatic tumor microenvironment. These findings may provide useful insights into the pathobiology of pancreatic cancer.


Assuntos
Adenocarcinoma/metabolismo , Interferon gama/metabolismo , Mucinas/metabolismo , Neoplasias Pancreáticas/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Adenocarcinoma/patologia , Estudos de Casos e Controles , Linhagem Celular Tumoral , Humanos , Interleucinas/metabolismo , Mucina-4 , Neoplasias Pancreáticas/patologia
12.
Cancer Res ; 66(18): 9045-53, 2006 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-16982746

RESUMO

A disintegrin and metalloproteinase (ADAM) molecules are known for their unique potential to combine adhesion, proteolysis, and signaling. To understand the role of ADAM17/tumor necrosis factor-alpha (TNF-alpha) converting enzyme (TACE) in pancreatic ductal adenocarcinoma (PDAC), we investigated its expression, function, and in vitro regulation. ADAM17/TACE mRNA was expressed in 3 of 10 normal pancreatic tissues, 6 of 8 samples from patients with chronic pancreatitis, 10 of 10 PDAC tissues, and 9 of 9 pancreatic cancer cell lines, but it was absent in primary duct epithelial cells. Immunohistochemical staining revealed positive cancer cells in 8 of 10 PDACs but no staining of ducts in normal pancreas. ADAM17/TACE was found in 0 of 16 pancreatic intraepithelial neoplasia (PanIN)-1A lesions, 1 of 30 PanIN-1B lesions, 2 of 13 PanIN-2 lesions but, in 13 of 15 PanIN-3 lesions, associated with PDAC. Western blot, flow cytometry, and confocal microscopy analyses showed the aberrant expression of ADAM17/TACE protein in pancreatic cancer cell lines. The proteolytic activity of ADAM17/TACE, assessed by the release of TNF-alpha, was inhibited by TNF-alpha protease inhibitor. ADAM17/TACE gene silencing using small interfering RNA technique in vitro reduced invasion behavior dramatically, whereas proliferation was unaffected. Furthermore, ADAM17/TACE mRNA expression was down-regulated in pancreatic cancer cells arrested in G2-M phase as well as in a time-dependent manner after TNF-alpha and interleukin-6 incubation. In conclusion, our findings provide evidence of aberrant expression of the proteolytically active ADAM17/TACE in advanced precursor lesions (PanIN-3) and PDAC while identifying its critical involvement in the invasion process.


Assuntos
Proteínas ADAM/biossíntese , Carcinoma Ductal Pancreático/enzimologia , Carcinoma Ductal Pancreático/patologia , Neoplasias Pancreáticas/enzimologia , Neoplasias Pancreáticas/patologia , Proteínas ADAM/genética , Proteína ADAM17 , Carcinoma Ductal Pancreático/genética , Ciclo Celular/genética , Processos de Crescimento Celular/genética , Linhagem Celular Tumoral , Progressão da Doença , Regulação Neoplásica da Expressão Gênica , Inativação Gênica , Humanos , Imuno-Histoquímica , Invasividade Neoplásica , Neoplasias Pancreáticas/genética , Pancreatite Crônica/enzimologia , Pancreatite Crônica/genética , RNA Mensageiro/biossíntese , RNA Mensageiro/genética
13.
Cell Microbiol ; 8(12): 1932-45, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16848787

RESUMO

Cryptosporidium parvum attaches to intestinal and biliary epithelial cells via specific molecules on host-cell surface membranes including Gal/GalNAc-associated glycoproteins. Subsequent cellular entry of this parasite depends on host-cell membrane alterations to form a parasitophorous vacuole via activation of phosphatidylinositol 3-kinase (PI-3K)/Cdc42-associated actin remodelling. How C. parvum hijacks these host-cell processes to facilitate its infection of target epithelia is unclear. Using specific probes to known components of sphingolipid-enriched membrane microdomains (SEMs), we detected aggregation of host-cell SEM components at infection sites during C. parvum infection of cultured human biliary epithelial cells (i.e. cholangiocytes). Activation and membrane translocation of acid-sphingomyelinase (ASM), an enzyme involved in SEM membrane aggregation, were also observed in infected cells. Pharmacological disruption of SEMs and knockdown of ASM via a specific small interfering RNA (siRNA) significantly decreased C. parvum attachment (by approximately 84%) and cellular invasion (by approximately 88%). Importantly, knockdown of ASM and disruption of SEMs significantly blocked C. parvum-induced accumulation of Gal/GalNAc-associated glycoproteins at infection sites by approximately 90%. Disruption of SEMs and knockdown of ASM also significantly blocked C. parvum-induced activation of host-cell PI-3K and subsequent accumulation of Cdc42 and actin by up to 75%. Our results suggest an important role of SEMs for C. parvum attachment to and entry of host cells, likely via clustering of membrane-binding molecules and facilitating of C. parvum-induced actin remodelling at infection sites through activation of the PI-3K/Cdc42 signalling pathway.


Assuntos
Ductos Biliares Intra-Hepáticos/parasitologia , Criptosporidiose/metabolismo , Cryptosporidium parvum/patogenicidade , Células Epiteliais/parasitologia , Microdomínios da Membrana/metabolismo , Esfingolipídeos/metabolismo , Actinas/metabolismo , Animais , Ductos Biliares Intra-Hepáticos/citologia , Linhagem Celular , Humanos , Quinases Associadas a Receptores de Interleucina-1/metabolismo , Esfingolipídeos/química , Proteína cdc42 de Ligação ao GTP/metabolismo
14.
Nat Cell Biol ; 8(4): 317-28, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16565709

RESUMO

Caveolar endocytosis has an important function in the cellular uptake of some bacterial toxins, viruses and circulating proteins. However, the molecular machinery involved in regulating caveolar uptake is poorly defined. Here, we demonstrate that caveolar endocytosis is regulated by syntaxin 6, a target membrane soluble N-ethylmaleimide attachment protein receptor (t-SNARE) involved in membrane fusion events along the secretory pathway. When syntaxin 6 function was inhibited, internalization through caveolae was dramatically reduced, whereas other endocytic mechanisms were unaffected. Syntaxin 6 inhibition also reduced the presence of caveolin-1 and caveolae at the plasma membrane. In addition, syntaxin 6 inhibition decreased the delivery of GM1 ganglioside (GM1) and glycosylphosphatidylinositol (GPI)-GFP (but not vesicular stomatitis virus-glycoprotein G; VSV-G) protein from the Golgi complex to the plasma membrane. Addition of GM1 to syntaxin 6-inhibited cells resulted in the reappearance of caveolin-1 and caveolae at the plasma membrane, and restored caveolar uptake. These results suggest that syntaxin 6 regulates the delivery of microdomain-associated lipids and proteins to the cell surface, which are required for caveolar endocytosis.


Assuntos
Cavéolas/metabolismo , Membrana Celular/metabolismo , Endocitose , Gangliosídeo G(M1)/metabolismo , Glicosilfosfatidilinositóis/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas Qa-SNARE/metabolismo , Proteínas do Envelope Viral/metabolismo , Caveolina 1/metabolismo , Células Cultivadas , Fibroblastos/citologia , Fibroblastos/metabolismo , Complexo de Golgi , Humanos , Oligonucleotídeos/farmacologia , Transporte Proteico , Proteínas Qa-SNARE/antagonistas & inibidores , Proteínas Qa-SNARE/genética , Células-Tronco/citologia , Células-Tronco/metabolismo
15.
FASEB J ; 19(11): 1558-60, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15972801

RESUMO

Niemann-Pick disease type C (NPC) is a genetic disorder in which patient cells exhibit lysosomal accumulation of cholesterol and sphingolipids (SLs) caused by defects in either NPC1 or NPC2 proteins. We previously demonstrated that NPC1 human skin fibroblasts overexpressing endosomal Rab proteins (Rab7 or Rab9) showed a correction in the storage disease phenotype. In the current study, we used protein transduction to further investigate Rab9-mediated reduction of stored lipids in NPC cells. Recombinant human Rab9 fused with the herpes simplex virus VP22 protein fragment was overexpressed, purified, and added to culture medium to induce protein transduction. When VP22-Rab9 was transduced into NPC1 fibroblasts, nearly all cells showed significant reduction in cellular free cholesterol levels, with no cytotoxicity up to 5 microM. A fraction of the VP22-Rab9 that was transduced into the cells was shown to bind to rab GDP dissociation inhibitor, suggesting that this pool of VP22-Rab9 had become prenylated. The reduction in cellular free cholesterol was associated with correction of abnormal intracellular trafficking of BODIPY-lactosylceramide and an increase of sterols in the culture media. The clearance of lysosomal free cholesterol was also associated with a decrease in LDL-receptor levels. In addition, we demonstrated reduction of intracellular cholesterol by VP22-Rab9 transduction in NPC2 fibroblasts and in cultured mouse NPC1 neurons. These observations provide important new information about the correction of membrane traffic in NPC cells by Rab9 overexpression and may lead to new therapeutic approaches for treatment of this disease.


Assuntos
Colesterol/metabolismo , Doenças de Niemann-Pick/tratamento farmacológico , Proteínas Recombinantes de Fusão/farmacologia , Proteínas rab de Ligação ao GTP/farmacologia , Transporte Biológico , Proteínas de Transporte , Células Cultivadas , Retículo Endoplasmático/metabolismo , Glicoproteínas/deficiência , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Glicoproteínas de Membrana/deficiência , Proteína C1 de Niemann-Pick , Doenças de Niemann-Pick/metabolismo , Proteínas de Transporte Vesicular , Proteínas Estruturais Virais
16.
Mol Biol Cell ; 15(9): 4166-78, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15229286

RESUMO

Proteins in the transforming growth factor-beta (TGF-beta) family recognize transmembrane serine/threonine kinases known as type I and type II receptors. Binding of TGF-beta to receptors results in receptor down-regulation and signaling. Whereas previous work has focused on activities controlling TGF-beta signaling, more recent studies have begun to address the trafficking properties of TGF-beta receptors. In this report, it is shown that receptors undergo recycling both in the presence and absence of ligand activation, with the rates of internalization and recycling being unaffected by ligand binding. Recycling occurs as receptors are most likely internalized through clathrin-coated pits, and then returned to the plasma membrane via a rab11-dependent, rab4-independent mechanism. Together, the results suggest a mechanism wherein activated TGF-beta receptors are directed to a distinct endocytic pathway for down-regulation and clathrin-dependent degradation after one or more rounds of recycling.


Assuntos
Clatrina/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo , Animais , Transporte Biológico Ativo , Linhagem Celular , Endocitose , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Ligantes , Vison , Modelos Biológicos , Monensin/farmacologia , Interferência de RNA , Receptores de Fatores de Crescimento Transformadores beta/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta1 , Proteínas rab de Ligação ao GTP/genética , Proteínas rab4 de Ligação ao GTP/genética , Proteínas rab4 de Ligação ao GTP/metabolismo
17.
Mol Biol Cell ; 15(6): 2684-96, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15075371

RESUMO

Intracellular trafficking of cystic fibrosis transmembrane conductance regulator (CFTR) is a focus of attention because it is defective in most patients with cystic fibrosis. DeltaF508 CFTR, which does not mature conformationally, normally does not exit the endoplasmic reticulum, but if induced to do so at reduced temperature is short-lived at the surface. We used external epitope-tagged constructs to elucidate the itinerary and kinetics of wild type and DeltaF508 CFTR in the endocytic pathway and visualized movement of CFTR from the surface to intracellular compartments. Modulation of different endocytic steps with low temperature (16 degrees C) block, protease inhibitors, and overexpression of wild type and mutant Rab GTPases revealed that surface CFTR enters several different routes, including a Rab5-dependent initial step to early endosomes, then either Rab11-dependent recycling back to the surface or Rab7-regulated movement to late endosomes or alternatively Rab9-mediated transit to the trans-Golgi network. Without any of these modulations DeltaF508 CFTR rapidly disappears from and does not return to the cell surface, confirming that its altered structure is detected in the distal as well as proximal secretory pathway. Importantly, however, the mutant protein can be rescued at the plasma membrane by Rab11 overexpression, proteasome inhibitors, or inhibition of Rab5-dependent endocytosis.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Endocitose/fisiologia , Animais , Linhagem Celular , Membrana Celular/metabolismo , Cricetinae , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Endocitose/efeitos dos fármacos , Epitopos/química , Epitopos/genética , Epitopos/metabolismo , Humanos , Inibidores de Proteases/farmacologia , Transporte Proteico/efeitos dos fármacos , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Deleção de Sequência/genética , Temperatura , Proteínas rab de Ligação ao GTP/genética , Proteínas rab de Ligação ao GTP/metabolismo
18.
J Biochem ; 131(1): 21-9, 2002 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11754731

RESUMO

Pancreatic mucins consist of core proteins that are decorated with carbohydrate structures. Previous studies have identified at least two physically distinct populations of mucins produced by a pancreatic adenocarcinoma cell line (HPAF); one is the MUC1 core protein, which includes an oligosaccharide structure identified by a monoclonal antibody (MAb) recognizing the DU-PAN-2 epitope. In this study, we purified and characterized a second mucin fraction, which also shows reactivity with the DU-PAN-2 antibody, but which has an amino acid composition that is not consistent with the MUC1 core protein. This new mucin was purified by ammonium sulfate precipitation, molecular sieve chromatography, and density gradient centrifugation. It eluted in the void volume of a Sepharose 4B column together with an associated low molecular weight protein, which could be further resolved. The mucin is highly polyanionic due to numerous sulfated and sialylated saccharide chains. Carbohydrate analyses of the purified mucin showed the presence of galactose, glucosamine, galactosamine, and sialic acid, but no mannose, glucose, or uronic acid. The purified and deglycosylated mucin shows no reactivity with anti-MUC1 apomucin antibody, but reacts with antiserum against deglycosylated tracheal mucins and antiserum against the MUC4 tandem repeat peptide. Analysis of mucin expression in HPAF cells revealed high levels of MUC1 and MUC4 mRNA, and moderate levels of MUC5AC and MUC5B mRNA. The amino acid composition of the purified mucin shows a high degree of similarity to the MUC4 core protein.


Assuntos
Adenocarcinoma/metabolismo , Mucina-1/isolamento & purificação , Mucinas/isolamento & purificação , Neoplasias Pancreáticas/metabolismo , Anticorpos/imunologia , Antígenos de Neoplasias/química , Antígenos de Neoplasias/imunologia , Carboidratos/química , Glicoproteínas/química , Glicoproteínas/isolamento & purificação , Humanos , Mucina-1/química , Mucina-4 , Mucinas/química , Mucinas/genética , RNA Mensageiro/biossíntese , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA