Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Comput Struct Biotechnol J ; 18: 464-481, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32180905

RESUMO

Obesity is characterized by a state of chronic, unresolved inflammation in insulin-targeted tissues. Obesity-induced inflammation causes accumulation of proinflammatory macrophages in adipose tissue and liver. Proinflammatory cytokines released from tissue macrophages inhibits insulin sensitivity. Obesity also leads to inflammation-induced endoplasmic reticulum (ER) stress and insulin resistance. In this scenario, based on the data (specifically patterns) generated by our in vivo experiments on both diet-induced obese (DIO) and normal chow diet (NCD) mice, we developed an in silico state space model to integrate ER stress and insulin signaling pathways. Computational results successfully followed the experimental results for both DIO and NCD conditions. Chromogranin A (CgA) peptide catestatin (CST: hCgA 352 - 372 ) improves obesity-induced hepatic insulin resistance by reducing inflammation and inhibiting proinflammatory macrophage infiltration. We reasoned that the anti-inflammatory effects of CST would alleviate ER stress. CST decreased obesity-induced ER dilation in hepatocytes and macrophages. On application of Proportional-Integral-Derivative (PID) controllers on the in silico model, we checked whether the reduction of phosphorylated PERK resulting in attenuation of ER stress, resembling CST effect, could enhance insulin sensitivity. The simulation results clearly pointed out that CST not only decreased ER stress but also enhanced insulin sensitivity in mammalian cells. In vivo experiment validated the simulation results by depicting that CST caused decrease in phosphorylation of UPR signaling molecules and increased phosphorylation of insulin signaling molecules. Besides simulation results predicted that enhancement of AKT phosphorylation helps in both overcoming ER stress and achieving insulin sensitivity. These effects of CST were verified in hepatocyte culture model.

2.
Cell Tissue Res ; 376(1): 51-70, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30467710

RESUMO

We have previously shown that the chromogranin A (CgA)-derived peptide catestatin (CST: hCgA352-372) inhibits nicotine-induced secretion of catecholamines from the adrenal medulla and chromaffin cells. In the present study, we seek to determine whether CST regulates dense core (DC) vesicle (DCV) quanta (catecholamine and chromogranin/secretogranin proteins) during acute (0.5-h treatment) or chronic (24-h treatment) cholinergic (nicotine) or peptidergic (PACAP, pituitary adenylyl cyclase activating polypeptide) stimulation of PC12 cells. In acute experiments, we found that both nicotine (60 µM) and PACAP (0.1 µM) decreased intracellular norepinephrine (NE) content and increased 3H-NE secretion, with both effects markedly inhibited by co-treatment with CST (2 µM). In chronic experiments, we found that nicotine and PACAP both reduced DCV and DC diameters and that this effect was likewise prevented by CST. Nicotine or CST alone increased expression of CgA protein and together elicited an additional increase in CgA protein, implying that nicotine and CST utilize separate signaling pathways to activate CgA expression. In contrast, PACAP increased expression of CgB and SgII proteins, with a further potentiation by CST. CST augmented the expression of tyrosine hydroxylase (TH) but did not increase intracellular NE levels, presumably due to its inability to cause post-translational activation of TH through serine phosphorylation. Co-treatment of CST with nicotine or PACAP increased quantal size, plausibly due to increased synthesis of CgA, CgB and SgII by CST. We conclude that CST regulates DCV quanta by acutely inhibiting catecholamine secretion and chronically increasing expression of CgA after nicotinic stimulation and CgB and SgII after PACAPergic stimulation.


Assuntos
Catecolaminas/metabolismo , Cromogranina A/fisiologia , Cromograninas/metabolismo , Nicotina/farmacologia , Fragmentos de Peptídeos/fisiologia , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/farmacologia , Animais , Cromogranina A/farmacologia , Subunidade alfa de Hormônios Glicoproteicos/metabolismo , Humanos , Norepinefrina/metabolismo , Células PC12 , Fragmentos de Peptídeos/farmacologia , Ratos , Proteínas Secretadas pela Vesícula Seminal/metabolismo , Transdução de Sinais/efeitos dos fármacos , Tirosina 3-Mono-Oxigenase/metabolismo
3.
BMC Microbiol ; 17(1): 226, 2017 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-29202699

RESUMO

BACKGROUND: Helicobacter pylori (H. pylori) is a Gram-negative, microaerophilic bacterium that is recognized as a major cause of chronic gastritis, peptic ulcers, and gastric cancer. Comparable to other Gram-negative bacteria, lipopolysaccharides (LPS) are an important cellular component of the outer membrane of H. pylori. The LPS of this organism plays a key role in its colonization and persistence in the stomach. In addition, H. pylori LPS modulates pathogen-induced host inflammatory responses resulting in chronic inflammation within the gastrointestinal tract. Very little is known about the comparative LPS compositions of different strains of H. pylori with varied degree of virulence in human. Therefore, LPS was analyzed from two strains of H. pylori with differing potency in inducing inflammatory responses (SS1 and G27). LPS were extracted from aqueous and phenol layer of hot-phenol water extraction method and subjected for composition analysis by gas chromatography - mass spectrometry (GC-MS) to sugar and fatty acid compositions. RESULTS: The major difference between the two strains of H. pylori is the presence of Rhamnose, Fucose and GalNAc in the SS1 strain, which was either not found or with low abundance in the G27 strain. On the other hand, high amount of Mannose was present in G27 in comparison to SS1. Fatty acid composition of lipid-A portion also showed considerable amount of differences between the two strains, phenol layer of SS1 had enhanced amount of 3 hydroxy decanoic acid (3-OH-C10:0) and 3-hydroxy dodecanoic acid (3-OH-C12:0) which were not present in G27, whereas myristic acid (C14:0) was present in G27 in relatively high amount. CONCLUSION: The composition analysis of H. pylori LPS, revealed differences in sugars and fatty acids composition between a mouse adapted strain SS1 and G27. This knowledge provides a novel way to dissect out their importance in host-pathogen interaction in further studies.


Assuntos
Helicobacter pylori/química , Lipopolissacarídeos/química , Ácidos Graxos/química , Cromatografia Gasosa-Espectrometria de Massas , Helicobacter pylori/metabolismo , Monossacarídeos/química , Especificidade da Espécie
4.
Cell Mol Immunol ; 13(3): 337-46, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26972772

RESUMO

Natural killer T (NKT) cells are innate-like lymphocytes that generally recognize lipid antigens and are enriched in microvascular compartments of the liver. NKT cells can be activated by self- or microbial-lipid antigens and by signaling through toll-like receptors. Following activation, NKT cells rapidly secrete pro-inflammatory or anti-inflammatory cytokines and chemokines, and thereby determine the milieu for subsequent immunity or tolerance. It is becoming clear that two different subsets of NKT cells-type I and type II-have different modes of antigen recognition and have opposing roles in inflammatory liver diseases. Here we focus mainly on the roles of both NKT cell subsets in the maintenance of immune tolerance and inflammatory diseases in liver. Furthermore, how the differential activation of type I and type II NKT cells influences other innate cells and adaptive immune cells to result in important consequences for tissue integrity is discussed. It is crucial that better reagents, including CD1d tetramers, be used in clinical studies to define the roles of NKT cells in liver diseases in patients.


Assuntos
Fígado/imunologia , Fígado/patologia , Subpopulações de Linfócitos/imunologia , Células T Matadoras Naturais/imunologia , Animais , Antígenos/imunologia , Humanos , Ativação Linfocitária/imunologia
5.
PLoS One ; 8(3): e58835, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23555599

RESUMO

In addition to its well-characterized function as a tumor suppressor, p14ARF (ARF) is a positive regulator of topoisomerase I (topo I), a central enzyme in DNA metabolism and a target for cancer therapy. We previously showed that topo I hyperphosphorylation, a cancer-associated event mediated by elevated levels of the protein kinase CK2, increases topo I activity and the cellular sensitivity to topo I-targeted drugs. Topo I hyperphosphorylation also increases its interaction with ARF. Because the ARF-topo I interaction could be highly relevant to DNA metabolism and cancer treatment, we identified the regions of topo I involved in ARF binding and characterized the effects of ARF binding on topo I function. Using a series of topo I deletion constructs, we found that ARF interacted with the topo I core domain, which encompasses most of the catalytic and DNA-interacting residues. ARF binding increased the DNA relaxation activity of hyperphosphorylated topo I by enhancing its association with DNA, but did not affect the topo I catalytic rate. In cells, ARF promoted the chromatin association of hyperphosphorylated, but not basal phosphorylated, topo I, and increased topo I-mediated DNA nicking under conditions of oxidative stress. The aberrant nicking was found to correlate with increased formation of DNA double-strand breaks, which are precursors of many genome destabilizing events. The results suggest that the convergent actions of oxidative stress and elevated CK2 and ARF levels, which are common features of cancer cells, lead to a dysregulation of topo I that may contribute both to the cellular response to topo I-targeted drugs and to genome instability.


Assuntos
DNA Topoisomerases Tipo I/metabolismo , Fases de Leitura Aberta , Proteína Supressora de Tumor p14ARF/genética , Proteína Supressora de Tumor p14ARF/metabolismo , Caseína Quinase II/metabolismo , Linhagem Celular , DNA Topoisomerases Tipo I/química , Ativação Enzimática , Humanos , Fosforilação , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Serina/metabolismo
6.
PLoS One ; 7(11): e50427, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23185622

RESUMO

Topoisomerase I is the target for a potent class of chemotherapeutic drugs derived from the plant alkaloid camptothecin that includes irinotecan and topotecan. In this study we have identified a novel site of CK2-mediated topoisomerase I (topo I) phosphorylation at serine 506 (PS506) that is relevant to topo I function and to cellular responses to these topo I-targeted drugs. CK2 treatment induced hyperphosphorylation of recombinant topo I and expression of the PS506 epitope, and resulted in increased binding of topo I to supercoiled plasmid DNA. Hyperphosphorylated topo I was approximately three times more effective than the basal phosphorylated enzyme at relaxing plasmid supercoils but had similar DNA cleavage activity once bound to DNA. The PS506 epitope was expressed in cancer cell lines with elevated CK2 activity, hyperphosphorylated topo I, and increased sensitivity to camptothecin. In contrast, PS506 was not detected in normal cells or cancer cell lines with lower levels of CK2 activity. By experimentally manipulating CK2 activity in cancer cell lines, we demonstrate a cause and effect relationship between CK2 activity, PS506 expression, camptothecin-induced cellular DNA damage, and cellular camptothecin sensitivity. Our results show that the PS506 epitope is an indicator of dysregulated, hyperphosphorylated topo I in cancer cells, and may thus serve as a diagnostic or prognostic biomarker and predict tumor responsiveness to widely used topo I-targeted therapies.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Biomarcadores Tumorais/genética , Camptotecina/farmacologia , Caseína Quinase II/genética , DNA Topoisomerases Tipo I/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Serina/metabolismo , Inibidores da Topoisomerase I/farmacologia , Biomarcadores Tumorais/metabolismo , Caseína Quinase II/metabolismo , Linhagem Celular Tumoral , Fragmentação do DNA/efeitos dos fármacos , DNA Topoisomerases Tipo I/metabolismo , DNA Super-Helicoidal/metabolismo , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Epitopos , Humanos , Fosforilação/efeitos dos fármacos , Plasmídeos , Serina/genética
7.
Biochemistry ; 50(5): 704-14, 2011 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-21182307

RESUMO

Topoisomerase I (topo I) is required to unwind DNA during synthesis and provides the unique target for camptothecin-derived chemotherapeutic agents, including Irinotecan and Topotecan. While these agents are highly effective anticancer agents, some tumors do not respond due to intrinsic or acquired resistance, a process that remains poorly understood. Because of treatment toxicity, there is interest in identifying cellular factors that regulate tumor sensitivity and might serve as predictive biomarkers of therapy sensitivity. Here we identify the serine kinase, protein kinase CK2, as a central regulator of topo I hyperphosphorylation and activity and cellular sensitivity to camptothecin. In nine cancer cell lines and three normal tissue-derived cell lines we observe a consistent correlation between CK2 levels and camptothecin responsiveness. Two other topo I-targeted serine kinases, protein kinase C and cyclin-dependent kinase 1, do not show this correlation. Camptothecin-sensitive cancer cell lines display high CK2 activity, hyperphosphorylation of topo I, elevated topo I activity, and elevated phosphorylation-dependent complex formation between topo I and p14ARF, a topo I activator. Camptothecin-resistant cancer cell lines and normal cell lines display lower CK2 activity, lower topo I phosphorylation, lower topo I activity, and undetectable topo I/p14ARF complex formation. Experimental inhibition or activation of CK2 demonstrates that CK2 is necessary and sufficient for regulating these topo I properties and altering cellular responses to camptothecin. The results establish a cause and effect relationship between CK2 activity and camptothecin sensitivity and suggest that CK2, topo I phosphorylation, or topo I/p14ARF complex formation could provide biomarkers of therapy-responsive tumors.


Assuntos
Antineoplásicos/farmacologia , Camptotecina/farmacologia , Caseína Quinase II/metabolismo , DNA Topoisomerases Tipo I/metabolismo , Neoplasias/enzimologia , Caseína Quinase II/genética , Linhagem Celular Tumoral , DNA Topoisomerases Tipo I/química , DNA Topoisomerases Tipo I/genética , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/genética , Neoplasias/metabolismo , Fosforilação , Estrutura Terciária de Proteína
8.
Cell Cycle ; 8(17): 2779-88, 2009 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-19652528

RESUMO

Acetyl group turnover on specific lysine epsilon-amino groups of the core chromosomal histones regulates DNA accessibility function, and the acetylating and deacetylating enzymes that govern the turnover provide important targets for the development of anti-cancer drugs. Histone deacetylase (HDAC) inhibitors have been developed and evaluated extensively in clinical trials, while the development of inhibitors of histone acetyltransferase (HAT) has proceeded more slowly. Here we have examined the cellular effects of an S-substituted coenzyme A (CoA) inhibitor of histone acetylation, consisting of spermidine (Spd) linked to the S-terminus of CoA through a thioglycolic acid linkage (adduct abbreviated as Spd-CoA), as well as the effects of a truncated Spd-CoA derivative lacking the negatively charged portion of the CoA moiety. While exposure of cancer cells to Spd-CoA has little effect on cell viability, it causes a rapid inhibition of histone acetylation that correlates with a transient arrest of DNA synthesis, a transient delay in S-phase progression, and an inhibition of nucleotide excision repair and DNA double strand break repair. These effects correlate with increased cellular sensitivity to the DNA-targeted chemotherapeutic drugs, cisplatin (Platinol()) and 5-fluorouracil, to the DNA damaging drug, camptothecin, and to UV-C irradiation. The sensitization effects of Spd-CoA are not observed in normal cells due to a barrier to uptake. The truncated Spd-CoA derivative displays similar but enhanced chemosensitization effects, suggesting that further modifications of the Spd-CoA structure could further improve potency. The results demonstrate that Spd-CoA and its truncated version are efficiently and selectively internalized into cancer cells, and suggest that the resulting inhibition of acetylation-dependent DNA repair enhances cellular sensitivity to DNA damage. These and related inhibitors of histone acetylation could therefore constitute a novel class of potent therapy sensitizers applicable to a broad range of conventional cancer treatments.


Assuntos
Antineoplásicos/farmacologia , Coenzima A/farmacologia , Reparo do DNA , Histona Acetiltransferases/antagonistas & inibidores , Histonas/metabolismo , Espermidina/análogos & derivados , Espermidina/farmacologia , Acetilação , Antineoplásicos/química , Linhagem Celular Tumoral , Coenzima A/química , Histona Acetiltransferases/metabolismo , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Fase S , Espermidina/química
9.
Biochemistry ; 46(49): 14325-34, 2007 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-18004878

RESUMO

p14ARF (ARF) and topoisomerase I play central roles in cancer and have recently been shown to interact. The interaction activates topoisomerase I, an important target for camptothecin-like chemotherapeutic drugs, but the regulation of the interaction is poorly understood. We have used the H358 and H23 lung cancer cell lines and purified recombinant human topoisomerase I to demonstrate that the ARF/topoisomerase I interaction is regulated by topoisomerase I serine phosphorylation, a modification that regulates topoisomerase I activity. Both cell lines express wild-type ARF and topoisomerase I proteins at equivalent levels, but H23 topoisomerase I, unlike that of H358 cells, is largely devoid of serine phosphorylation, has low activity, and complexes poorly with ARF. The ability of H23 topoisomerase I to complex with ARF can be restored by treatment with the serine kinase, casein kinase II. Consistent with these observations, we show that the response of H23 cells to camptothecin treatment is unaffected by changes in intracellular levels of ARF. However, in H358 and PC-3 cells, which express a serine phosphorylated topoisomerase I that complexes with ARF, ectopic overexpression of ARF causes sensitization to camptothecin, and siRNA-mediated down-regulation of endogenous ARF causes desensitization to camptothecin. These biological responses correlate with increased and decreased levels, respectively, of ARF/topoisomerase I complex and DNA-bound topoisomerase I. Thus, ARF is a serine phosphorylation-dependent coregulator of topoisomerase I in vivo, and it regulates cellular sensitivity to camptothecin by interacting with topoisomerase I. Certain cancer associated defects affecting ARF/topoisomerase I complex formation could contribute to cellular resistance to camptothecin.


Assuntos
DNA Topoisomerases Tipo I/fisiologia , Serina/metabolismo , Proteína Supressora de Tumor p14ARF/fisiologia , Camptotecina/farmacologia , Carcinoma Pulmonar de Células não Pequenas , Linhagem Celular Tumoral , Dano ao DNA , Regulação da Expressão Gênica , Humanos , Neoplasias Pulmonares , Fosforilação , Fosfosserina/metabolismo
10.
Cell Cycle ; 5(7): 686-90, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16627991

RESUMO

The p53-mediated pathway cell cycle arrest and apoptosis is central to cancer and an important point of focus for therapeutics development. The p14ARF ("ARF") tumor suppressor induces the p53 pathway in response to oncogene activation or DNA damage. However, ARF is predominantly nucleolar in localization and engages in several interactions with nucleolar proteins, whereas p53 is nucleoplasmic. This raises the question as to how ARF initiates its involvement in the p53 pathway. We have found that UV irradiation of cells disrupts the interaction of ARF with two of its nucleolar binding partners, B23 (NPM, nucleophosmin, NO38, numatrin) and topoisomerase I, and promotes an immediate and transient subnuclear redistribution of ARF to the nucleoplasm, where it can engage the p53 pathway (Lee et al, Cancer Res 65:9834-42; 2005). The results support a model in which the nucleolus serves as a p53 upstream sensor of cellular stress, and add to a growing body of evidence that nucleolar sequestration of ARF prevents activation of p53. The results also have therapeutic implications for therapies based on exploiting p53 and other cellular stress response pathways to suppress cancer.


Assuntos
Compartimento Celular , Núcleo Celular/metabolismo , Regulação da Expressão Gênica , Proteína Supressora de Tumor p14ARF/metabolismo , Linhagem Celular Tumoral , Humanos , Proteínas Nucleares/metabolismo , Nucleofosmina , Ligação Proteica , Proteínas Proto-Oncogênicas c-mdm2/metabolismo
11.
Cancer Res ; 65(21): 9834-42, 2005 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-16267006

RESUMO

The p14 alternate reading frame (ARF) tumor suppressor plays a central role in cancer by binding to mdm2 (Hdm2 in humans) and enhancing p53-mediated apoptosis following DNA damage and oncogene activation. It is unclear, however, how ARF initiates its involvement in the p53/mdm2 pathway, as p53 and mdm2 are located in the nucleoplasm, whereas ARF is largely nucleolar in tumor cells. We have used immunofluorescence and coimmunoprecipitation to examine how the subnuclear distribution and protein-protein interactions of ARF change immediately after DNA damage and over the time course of the DNA damage response in human tumor cells. We find that DNA damage disrupts the interaction of ARF with the nucleolar protein B23(nucleophosmin) and promotes a transient p53-independent translocation of ARF to the nucleoplasm, resulting in a masking of the ARF NH2 terminus that correlates with the appearance of ARF-Hdm2 complexes. The translocation also results in an unmasking of the ARF COOH terminus, suggesting that redistribution disrupts a nucleolar interaction of ARF involving this region. By 24 hours after irradiation, DNA repair has ceased and the pretreatment immunofluorescence patterns and complexes of ARF have been restored. Although the redistribution of ARF is independent of p53 and likely to be regulated by interactions other than Hdm2, ARF does not promote UV sensitization unless p53 is expressed. The results implicate the nucleolus and nucleolar interactions of the ARF, including potentially novel interactions involving its COOH terminus as sites for early DNA damage and stress-mediated cellular events.


Assuntos
Nucléolo Celular/metabolismo , Dano ao DNA/fisiologia , Proteínas Nucleares/metabolismo , Proteína Supressora de Tumor p14ARF/metabolismo , Sequência de Aminoácidos , Linhagem Celular Tumoral , Sobrevivência Celular/fisiologia , Sobrevivência Celular/efeitos da radiação , Humanos , Microscopia de Fluorescência , Dados de Sequência Molecular , Nucleofosmina , Nucleoplasminas , Fosfoproteínas/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Raios Ultravioleta
12.
Eur J Biochem ; 270(18): 3806-13, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12950264

RESUMO

Earlier we presented several lines of evidence that a 67-kDa laminin binding protein (LBP) in Leishmania donovani, that is different from the putative mammalian 67-kDa laminin receptor, may play an important role in the onset of leishmaniasis, as these parasites invade macrophages in various organs after migrating through the extracellular matrix. Here we describe the membrane orientation of this Leishmania laminin receptor. Flow cytometric analysis using anti-LBP Ig revealed its surface localization, which was further confirmed by enzymatic radiolabeling of Leishmania surface proteins, autoradiography and Western blotting. Efficient incorporation of LBP into artificial lipid bilayer, as well as its presence in the detergent phase after Triton X-114 membrane extraction, suggests that it may be an integral membrane protein. Limited trypsinization of intact parasite and subsequent immunoblotting of trypsin released material using laminin as primary probe revealed that a major part of this protein harbouring the laminin binding site is oriented extracellularly. Carboxypeptidase Y treatment of the whole cell, as well as the membrane preparation, revealed that a small part of the C-terminal is located in the cytosol. A 34-kDa transmembrane part of LBP could be identified using the photoactive probe, 3-(trifluoromethyl)-3-(m-iodophenyl)diazirine (TID). Partial sequence comparison of the intact protein to that with the trypsin-released fragment indicated that N-terminal may be located extracellularly. Together, these results suggest that LBP may be an integral membrane protein, having significant portion of N-terminal end as well as the laminin binding site oriented extracellularly, a membrane spanning domain and a C-terminal cytosolic end.


Assuntos
Matriz Extracelular/química , Matriz Extracelular/metabolismo , Leishmania donovani/química , Proteínas de Membrana/química , Proteínas de Membrana/metabolismo , Receptores de Laminina/química , Receptores de Laminina/metabolismo , Animais , Azirinas/química , Sítios de Ligação , Carboxipeptidases/metabolismo , Citometria de Fluxo , Humanos , Radioisótopos do Iodo , Laminina/química , Laminina/metabolismo , Leishmania donovani/metabolismo , Lipossomos/química , Lipossomos/metabolismo , Fragmentos de Peptídeos/análise , Fragmentos de Peptídeos/química , Marcadores de Fotoafinidade/química , Polietilenoglicóis/química , Ligação Proteica , Estrutura Terciária de Proteína
13.
Eur J Biochem ; 269(6): 1622-9, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11895432

RESUMO

In the course of trying to understand the pathogenesis of leishmaniasis in relation to extracellular matrix (ECM) elements, laminin, a major ECM protein, has been found to bind saturably and with high affinity to a 67-kDa cell surface protein of Leishmania donovani. This interaction involves a single class of binding sites, which are ionic in nature, conformation-dependent and possibly involves sulfhydryls. Binding activity was significantly enhanced by Zn2+, an effect possibly mediated through Cys-rich zinc finger-like sequences on laminin. Inhibition studies with monoclonals against polypeptide chains and specific peptides with adhesive properties revealed that the binding site was localized in one of the nested zinc finger consensus sequences of B1 chain containing the specific pentapeptide sequence, YIGSR. Furthermore, incubation of L. donovani promastigotes with C(YIGSR)3-NH2 peptide amide or antibody directed against the 67-kDa laminin-binding protein (LBP) induced tyrosine phosphorylation of proteins with a molecular mass ranging from 115 to 130 kDa. These studies suggest a role for LBP in the interaction of parasites with ECM elements, which may mediate one or more downstream signalling events necessary for establishment of infection.


Assuntos
Laminina/metabolismo , Leishmania donovani/metabolismo , Proteínas de Membrana/metabolismo , Dedos de Zinco , Zinco/metabolismo , Sequência de Aminoácidos , Animais , Humanos , Dados de Sequência Molecular , Fosforilação , Tirosina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...