Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
J Biol Chem ; 289(29): 20170-81, 2014 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-24898246

RESUMO

Cobra cardiotoxins (CTX) are a family of three-fingered basic polypeptides known to interact with diverse targets such as heparan sulfates, sulfatides, and integrins on cell surfaces. After CTX bind to the membrane surface, they are internalized to intracellular space and exert their cytotoxicity via an unknown mechanism. By the combined in vitro kinetic binding, three-dimensional x-ray structure determination, and cell biology studies on the naturally abundant CTX homologues from the Taiwanese cobra, we showed that slight variations on the spatial distribution of positively charged or hydrophobic domains among CTX A2, A3, and A4 could lead to significant changes in their endocytotic pathways and action mechanisms via distinct sulfated glycoconjugate-mediated processes. The intracellular locations of these structurally similar CTX after internalization are shown to vary between the mitochondria and lysosomes via either dynamin2-dependent or -independent processes with distinct membrane cholesterol sensitivity. Evidence is presented to suggest that the shifting between the sulfated glycoconjugates as distinct targets of CTX A2, A3, and A4 might play roles in the co-evolutionary arms race between venomous snake toxins to cope with different membrane repair mechanisms at the cellular levels. The sensitivity of endocytotic routes to the spatial distribution of positively charged or hydrophobic domains may provide an explanation for the diverse endocytosis pathways of other cell-penetrating basic polypeptides.


Assuntos
Proteínas Cardiotóxicas de Elapídeos/química , Proteínas Cardiotóxicas de Elapídeos/farmacocinética , Sequência de Aminoácidos , Animais , Sítios de Ligação , Células CHO , Linhagem Celular , Membrana Celular/metabolismo , Proteínas Cardiotóxicas de Elapídeos/genética , Cricetinae , Cricetulus , Cristalografia por Raios X , Elapidae/genética , Elapidae/metabolismo , Endocitose , Evolução Molecular , Glicoconjugados/química , Glicoconjugados/metabolismo , Heparitina Sulfato/química , Heparitina Sulfato/metabolismo , Interações Hidrofóbicas e Hidrofílicas , Modelos Moleculares , Dados de Sequência Molecular , Domínios e Motivos de Interação entre Proteínas , Ratos , Eletricidade Estática , Homologia Estrutural de Proteína
2.
J Biol Chem ; 288(19): 13522-33, 2013 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-23536183

RESUMO

BACKGROUND: CXCL4L1 is a highly potent anti-angiogenic and anti-tumor chemokine, and its structural information is unknown. RESULTS: CXCL4L1 x-ray structure is determined, and it reveals a previously unrecognized chemokine structure adopting a novel C-terminal helix conformation. CONCLUSION: The alternative helix conformation enhances the anti-angiogenic activity of CXCL4L1 by reducing the glycosaminoglycan binding ability. SIGNIFICANCE: Chemokine C-terminal helix orientation is critical in regulating their functions. Chemokines, a subfamily of cytokines, are small, secreted proteins that mediate a variety of biological processes. Various chemokines adopt remarkable conserved tertiary structure comprising an anti-parallel ß-sheet core domain followed by a C-terminal helix that packs onto the ß-sheet. The conserved structural feature has been considered critical for chemokine function, including binding to cell surface receptor. The recently isolated variant, CXCL4L1, is a homologue of CXCL4 chemokine (or platelet factor 4) with potent anti-angiogenic activity and differed only in three amino acid residues of P58L, K66E, and L67H. In this study we show by x-ray structural determination that CXCL4L1 adopts a previously unrecognized structure at its C terminus. The orientation of the C-terminal helix protrudes into the aqueous space to expose the entire helix. The alternative helix orientation modifies the overall chemokine shape and surface properties. The L67H mutation is mainly responsible for the swing-out effect of the helix, whereas mutations of P58L and K66E only act secondarily. This is the first observation that reports an open conformation of the C-terminal helix in a chemokine. This change leads to a decrease of its glycosaminoglycan binding properties and to an enhancement of its anti-angiogenic and anti-tumor effects. This unique structure is recent in evolution and has allowed CXCL4L1 to gain novel functional properties.


Assuntos
Fator Plaquetário 4/química , Sequência de Aminoácidos , Substituição de Aminoácidos , Proteínas Angiogênicas/química , Cristalografia por Raios X , Cistina/química , Ditiotreitol/química , Heparina/química , Humanos , Ligação de Hidrogênio , Interações Hidrofóbicas e Hidrofílicas , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Oxirredução , Fator Plaquetário 4/genética , Fator Plaquetário 4/fisiologia , Ligação Proteica , Dobramento de Proteína , Domínios e Motivos de Interação entre Proteínas , Estabilidade Proteica , Estrutura Quaternária de Proteína , Estrutura Secundária de Proteína , Substâncias Redutoras/química
3.
Anal Chem ; 86(17): 8742-50, 2014 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-25138527

RESUMO

Snake venom consists of toxin proteins with multiple disulfide linkages to generate unique structures and biological functions. Determination of these cysteine connections usually requires the purification of each protein followed by structural analysis. In this study, dimethyl labeling coupled with LC-MS/MS and RADAR algorithm was developed to identify the disulfide bonds in crude snake venom. Without any protein separation, the disulfide linkages of several cytotoxins and PLA2 could be solved, including more than 20 disulfide bonds. The results show that this method is capable of analyzing protein mixture. In addition, the approach was also used to compare native cytotoxin 3 (CTX III) and its scrambled isomer, another category of protein mixture, for unknown disulfide bonds. Two disulfide-linked peptides were observed in the native CTX III, and 10 in its scrambled form, X-CTX III. This is the first study that reports a platform for the global cysteine connection analysis on a protein mixture. The proposed method is simple and automatic, offering an efficient tool for structural and functional studies of venom proteins.


Assuntos
Dissulfetos/análise , Venenos de Serpentes/química , Espectrometria de Massas em Tandem , Algoritmos , Sequência de Aminoácidos , Cromatografia Líquida de Alta Pressão , Venenos de Cnidários/análise , Venenos de Cnidários/química , Bases de Dados de Proteínas , Isomerismo , Peptídeos/análise
4.
Exp Dermatol ; 23(11): 843-9, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25236603

RESUMO

Anchorage-independent survival is one of the key features for malignant tumor cells. Whether specific gene alterations contributed by anchorage independency would further affect metastatic phenotypes of melanoma cells was unclear. We adapted suspension culture of melanoma cells to establish anchorage independency. The suspended melanoma cells lost their invasive abilities in vitro. Specific loss of laminin-binding ability in suspended melanoma cells was observed, which was correlated with downregulation of syndecan-1 as revealed by microarray and validated by qPCR and Western blot. Modulation of syndecan-1 expression level affected laminin binding, transwell migration and matrix metalloproteinase-2 secretion in melanoma cells. SDC1 expression and transwell migration were correlated with activity or level of protein kinase Cδ as evidence by specific inhibitors and shRNA transfection. In this study, we compared metastatic phenotypes and gene expressions of adherent and suspended melanoma cells. The anchorage independency led to protein kinase Cδ-mediated syndecan-1 downregulation, which contributed to loss of laminin-binding ability, reduced metalloproteinase-2 secretion and loss of invasiveness.


Assuntos
Regulação Neoplásica da Expressão Gênica , Melanoma/metabolismo , Proteína Quinase C-delta/metabolismo , Neoplasias Cutâneas/metabolismo , Sindecana-1/metabolismo , Adesão Celular , Linhagem Celular Tumoral , Movimento Celular , Regulação para Baixo , Expressão Gênica , Humanos , Laminina/metabolismo , Metaloproteinase 2 da Matriz/metabolismo , Invasividade Neoplásica , Metástase Neoplásica , Fenótipo , RNA Interferente Pequeno/metabolismo
5.
Biochim Biophys Acta ; 1818(5): 1378-85, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22387431

RESUMO

Cobra CTX A3, the major cardiotoxin (CTX) from Naja atra, is a cytotoxic, basic ß-sheet polypeptide that is known to induce a transient membrane leakage of cardiomyocytes through a sulfatide-dependent CTX membrane pore formation and internalization mechanism. The molecular specificity of CTX A3-sulfatide interaction at atomic levels has also been shown by both nuclear magnetic resonance (NMR) and X-ray diffraction techniques to reveal a role of CTX-induced sulfatide conformational changes for CTX A3 binding and dimer formation. In this study, we investigate the role of sulfatide lipid domains in CTX pore formation by various biophysical methods, including fluorescence imaging and atomic force microscopy, and suggest an important role of liquid-disordered (ld) and solid-ordered (so) phase boundary in lipid domains to facilitate the process. Fluorescence spectroscopic studies on the kinetics of membrane leakage and CTX oligomerization further reveal that, although most CTXs can oligomerize on membranes, only a small fraction of CTXs oligomerizations form leakage pores. We therefore suggest that CTX binding at the boundary between the so and so/ld phase coexistence sulfatide lipid domains could form effective pores to significantly enhance the CTX-induced membrane leakage of sulfatide-containing phosphatidylcholine vesicles. The model is consistent with our earlier observations that CTX may penetrate and lyse the bilayers into small aggregates at a lipid/protein molar ratio of about 20 in the ripple P(ß)' phase of phosphatidylcholine bilayers and suggest a novel mechanism for the synergistic action of cobra secretary phospholipase A2 and CTXs.


Assuntos
Membrana Celular/química , Proteínas Cardiotóxicas de Elapídeos/química , Elapidae , Bicamadas Lipídicas/química , Membranas Artificiais , Multimerização Proteica , Animais , Membrana Celular/metabolismo , Proteínas Cardiotóxicas de Elapídeos/metabolismo , Bicamadas Lipídicas/metabolismo , Miócitos Cardíacos/química , Miócitos Cardíacos/metabolismo , Ressonância Magnética Nuclear Biomolecular , Fosfatidilcolinas/química , Fosfatidilcolinas/metabolismo , Estrutura Secundária de Proteína , Difração de Raios X
6.
Cell Death Discov ; 9(1): 340, 2023 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-37696858

RESUMO

The tumor suppressor p53 primarily functions as a mediator of DNA damage-induced cell death, thereby contributing to the efficacy of genotoxic anticancer therapeutics. Here, we show, on the contrary, that cancer cells can employ genotoxic stress-induced p53 to acquire treatment resistance through the production of the pleiotropic cytokine interleukin (IL)-6. Mechanistically, DNA damage, either repairable or irreparable, activates p53 and stimulates Caspase-2-mediated cleavage of its negative regulator mouse double minute 2 (MDM2) creating a positive feedback loop that leads to elevated p53 protein accumulation. p53 transcriptionally controls the major adenosine triphosphate (ATP) release channel pannexin 1 (Panx1), which directs IL-6 induction via a mechanism dependent on the extracellular ATP-activated purinergic P2 receptors as well as their downstream intracellular calcium (iCa2+)/PI3K/Akt/NF-ĸB signaling pathway. Thus, p53 silencing impairs Panx1 and IL-6 expression and renders cancer cells sensitive to genotoxic stress. Moreover, we confirm that IL-6 hampers the effectiveness of genotoxic anticancer agents by mitigating DNA damage, driving the expression of anti-apoptotic Bcl-2 family genes, and maintaining the migratory and invasive properties of cancer cells. Analysis of patient survival and relevant factors in lung cancer and pan-cancer cohorts supports the prognostic and clinical values of Panx1 and IL-6. Notably, IL-6 secreted by cancer cells during genotoxic treatments promotes the polarization of monocytic THP-1-derived macrophages into an alternative (M2-like) phenotype that exhibits impaired anti-survival activities but enhanced pro-metastatic effects on cancer cells as compared to nonpolarized macrophages. Our study reveals the precise mechanism for genotoxic-induced IL-6 and suggests that targeting p53-mediated IL-6 may improve the responsiveness of cancer cells to genotoxic anticancer therapy.

7.
Redox Biol ; 64: 102791, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37385076

RESUMO

Snake venom l-amino acid oxidases (svLAAOs) have been recognized as promising candidates for anticancer therapeutics. However, multiple aspects of their catalytic mechanism and the overall responses of cancer cells to these redox enzymes remain ambiguous. Here, we present an analysis of the phylogenetic relationships and active site-related residues among svLAAOs and reveal that the previously proposed critical catalytic residue His 223 is highly conserved in the viperid but not the elapid svLAAO clade. To gain further insight into the action mechanism of the elapid svLAAOs, we purify and characterize the structural, biochemical, and anticancer therapeutic potentials of the Thailand elapid snake Naja kaouthia LAAO (NK-LAAO). We find that NK-LAAO, with Ser 223, exhibits high catalytic activity toward hydrophobic l-amino acid substrates. Moreover, NK-LAAO induces substantial oxidative stress-mediated cytotoxicity with the magnitude relying on both the levels of extracellular hydrogen peroxide (H2O2) and intracellular reactive oxygen species (ROS) generated during the enzymatic redox reactions, but not being influenced by the N-linked glycans on its surface. Unexpectedly, we discover a tolerant mechanism deployed by cancer cells to dampen the anticancer activities of NK-LAAO. NK-LAAO treatment amplifies interleukin (IL)-6 expression via the pannexin 1 (Panx1)-directed intracellular calcium (iCa2+) signaling pathway to confer adaptive and aggressive phenotypes on cancer cells. Accordingly, IL-6 silencing renders cancer cells vulnerable to NK-LAAO-induced oxidative stress together with abrogating NK-LAAO-stimulated metastatic acquisition. Collectively, our study urges caution when using svLAAOs in cancer treatment and identifies the Panx1/iCa2+/IL-6 axis as a therapeutic target for improving the effectiveness of svLAAOs-based anticancer therapies.


Assuntos
Interleucina-6 , Neoplasias , Humanos , Interleucina-6/genética , L-Aminoácido Oxidase/química , L-Aminoácido Oxidase/metabolismo , L-Aminoácido Oxidase/farmacologia , Peróxido de Hidrogênio/metabolismo , Filogenia , Venenos de Serpentes , Neoplasias/tratamento farmacológico , Aminoácidos
8.
Elife ; 122023 04 17.
Artigo em Inglês | MEDLINE | ID: mdl-37067034

RESUMO

For decades, studies of snake venoms focused on the venom-ome-specific toxins (VSTs). VSTs are dominant soluble proteins believed to contribute to the main venomous effects and emerged into gene clusters for fast adaptation and diversification of snake venoms. However, the conserved minor venom components, such as snake venom phosphodiesterase (svPDE), remain largely unexplored. Here, we focus on svPDE by genomic and transcriptomic analysis across snake clades and demonstrate that soluble svPDE is co-opted from the ancestral membrane-attached ENPP3 (ectonucleotide pyrophosphatase/phosphodiesterase 3) gene by replacing the original 5' exon with the exon encoding a signal peptide. Notably, the exons, promoters, and transcription/translation starts have been replaced multiple times during snake evolution, suggesting the evolutionary necessity of svPDE. The structural and biochemical analyses also show that svPDE shares the similar functions with ENPP family, suggesting its perturbation to the purinergic signaling and insulin transduction in venomous effects.


Assuntos
Venenos de Serpentes , Toxinas Biológicas , Animais , Venenos de Serpentes/genética , Venenos de Serpentes/química , Venenos de Serpentes/metabolismo , Serpentes , Fosfodiesterase I
9.
Blood ; 116(22): 4703-11, 2010 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-20688960

RESUMO

CXCL4 and CXCL4L1 are 2 closely related CXC chemokines that exhibit potent antiangiogenic activity. Because interactions with glycosaminoglycans play a crucial role in chemokines activity, we determined the binding parameters of CXCL4 and CXCL4L1 for heparin, heparan sulfate, and chondroitin sulfate B. We further demonstrated that the Leu67/His67 substitution is critical for the decrease in glycan binding of CXCL4L1 but also for the increase of its angiostatic activities. Using a set of mutants, we show that glycan affinity and angiostatic properties are not completely related. These data are reinforced using a monoclonal antibody that specifically recognizes structural modifications in CXCL4L1 due to the presence of His67 and that blocks its biologic activity. In vivo, half-life and diffusibility of CXCL4L1 compared with CXCL4 is strongly increased. As opposed to CXCL4L1, CXCL4 is preferentially retained at its site of expression. These findings establish that, despite small differences in the primary structure, CXCL4L1 is highly distinct from CXCL4. These observations are not only of great significance for the antiangiogenic activity of CXCL4L1 and for its potential use in clinical development but also for other biologic processes such as inflammation, thrombosis or tissue repair.


Assuntos
Aminoácidos/metabolismo , Dermatan Sulfato/metabolismo , Heparina/metabolismo , Heparitina Sulfato/metabolismo , Fator Plaquetário 4/metabolismo , Sequência de Aminoácidos , Aminoácidos/análise , Aminoácidos/genética , Animais , Bovinos , Linhagem Celular , Proliferação de Células , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Mutação , Neovascularização Fisiológica , Fator Plaquetário 4/análise , Fator Plaquetário 4/genética , Ligação Proteica , Proteínas Recombinantes/análise , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
10.
Biochem J ; 433(1): 127-38, 2011 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-20964630

RESUMO

HDGF (hepatoma-derived growth factor) stimulates cell proliferation by functioning on both sides of the plasma membrane as a ligand for membrane receptor binding to trigger cell signalling and as a stimulator for DNA synthesis in the nucleus. Although HDGF was initially identified as a secretory heparin-binding protein, the biological significance of its heparin-binding ability remains to be determined. In the present study we demonstrate that cells devoid of surface HS (heparan sulfate) were unable to internalize HDGF, HATH (N-terminal domain of HDGF consisting of amino acid residues 1-100, including the PWWP motif) and HATH(K96A) (single-site mutant form of HATH devoid of receptor binding activity), suggesting that the binding of HATH to surface HS is important for HDGF internalization. We further demonstrate that both HATH and HATH(K96A) could be internalized through macropinocytosis after binding to the cell surface HS. Interestingly, HS-mediated HATH(K96A) internalization is found to exhibit an inhibitory effect on cell migration and proliferation in contrast with that observed for HATH action on NIH 3T3 cells, suggesting that HDGF exploits the innate properties of both cell surface HS and membrane receptor via the HATH domain to affect related cell signalling processes. The present study indicates that MAPK (mitogen-activated protein kinase) signalling pathways could be affected by the HS-mediated HATH internalization to regulate cell migration in NIH 3T3 fibroblasts, as judged from the differential effect of HATH and HATH(K96A) treatment on the expression level of matrix metalloproteases.


Assuntos
Movimento Celular , Fibroblastos/fisiologia , Heparitina Sulfato/fisiologia , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Pinocitose , Transdução de Sinais/fisiologia , Células 3T3 , Animais , Membrana Celular/química , Proliferação de Células , Fibroblastos/citologia , Metaloproteinases da Matriz/biossíntese , Camundongos , Estrutura Terciária de Proteína
11.
Toxins (Basel) ; 14(12)2022 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-36548757

RESUMO

Naja nivea (Cape Cobra) is endemic to southern Africa. Envenoming by N. nivea is neurotoxic, resulting in fatal paralysis. Its venom composition, however, has not been studied in depth, and specific antivenoms against it remain limited in supply. Applying a protein decomplexation approach, this study unveiled the venom proteome of N. nivea from South Africa. The major components in the venom are cytotoxins/cardiotoxins (~75.6% of total venom proteins) and alpha-neurotoxins (~7.4%), which belong to the three-finger toxin family. Intriguingly, phospholipase A2 (PLA2) was undetected-this is a unique venom phenotype increasingly recognized in the African cobras of the Uraeus subgenus. The work further showed that VINS African Polyvalent Antivenom (VAPAV) exhibited cross-reactivity toward the venom and immunorecognized its toxin fractions. In mice, VAPAV was moderately efficacious in cross-neutralizing the venom lethality with a potency of 0.51 mg/mL (amount of venom completely neutralized per milliliter of antivenom). In the challenge-rescue model, VAPAV prevented death in 75% of experimentally envenomed mice, with slow recovery from neurotoxicity up to 24 h. The finding suggests the potential para-specific utility of VAPAV for N. nivea envenoming, although a higher dose or repeated administration of the antivenom may be required to fully reverse the neurotoxic effect of the venom.


Assuntos
Naja , Síndromes Neurotóxicas , Camundongos , Animais , Antivenenos/farmacologia , Antivenenos/metabolismo , Venenos Elapídicos/toxicidade , Venenos Elapídicos/metabolismo , África do Sul , Elapidae/metabolismo
12.
Toxins (Basel) ; 14(9)2022 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-36136582

RESUMO

Reports of bite from Protobothrops mucrosquamatus (Pmu) are frequent in Taiwan, and its wide-spread distribution and diverse habitats drove us to investigate its envenoming effects and relevant venom variations. We used reversed-phase high-performance liquid chromatography and mass spectrometry to analyze 163 Pmu venom samples collected from northern and southeastern Taiwan. Twenty-two major protein fractions were separated and analyzed, and their contents were determined semi-quantitatively. The results showed that despite the trivial differences in the protein family, there is an existing variation in acidic phospholipases A2s, serine proteinases, metalloproteinases, C-type lectin-like proteins, and other less abundant components in the Pmu venoms. Moreover, clinical manifestations of 209 Pmu envenomed patients hospitalized in northern or southeastern Taiwan revealed significant differences in local symptoms, such as ecchymosis and blistering. The mechanism of these local effects and possibly relevant venom components were examined. Further analysis showed that certain venom components with inter-population variation might work alone or synergistically with others to aggravate the local effects. Therefore, our findings of the venom variation may help one to improve antivenom production and better understand and manage Pmu bites.


Assuntos
Mordeduras de Serpentes , Trimeresurus , Animais , Antivenenos/química , Humanos , Lectinas Tipo C , Metaloproteases , Fosfolipases A2 , Serina Proteases , Taiwan
13.
J Biol Chem ; 285(41): 31603-15, 2010 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-20685646

RESUMO

Chitinases hydrolyze chitin, an insoluble linear polymer of N-acetyl-d-glucosamine (NAG)(n), into nutrient sources. Bacillus cereus NCTU2 chitinase (ChiNCTU2) predominantly produces chitobioses and belongs to glycoside hydrolase family 18. The crystal structure of wild-type ChiNCTU2 comprises only a catalytic domain, unlike other chitinases that are equipped with additional chitin binding and insertion domains to bind substrates into the active site. Lacking chitin binding and chitin insertion domains, ChiNCTU2 utilizes two dynamic loops (Gly-67-Thr-69 and Ile-106-Val-112) to interact with (NAG)(n), generating novel substrate binding and distortion for catalysis. Gln-109 is crucial for direct binding with substrates, leading to conformational changes of two loops with a maximum shift of ∼4.6 Šalong the binding cleft. The structures of E145Q, E145Q/Y227F, and E145G/Y227F mutants complexed with (NAG)(n) reveal (NAG)(2), (NAG)(2), and (NAG)(4) in the active site, respectively, implying various stages of reaction: before hydrolysis, E145G/Y227F with (NAG)(4); in an intermediate state, E145Q/Y227F with a boat-form NAG at the -1 subsite, -1-(NAG); after hydrolysis, E145Q with a chair form -1-(NAG). Several residues were confirmed to play catalytic roles: Glu-145 in cleavage of the glycosidic bond between -1-(NAG) and +1-(NAG); Tyr-227 in the conformational change of -1-(NAG); Asp-143 and Gln-225 in stabilizing the conformation of -1-(NAG). Additionally, Glu-190 acts in the process of product release, and Tyr-193 coordinates with water for catalysis. Residues Asp-143, E145Q, Glu-190, and Tyr-193 exhibit multiple conformations for functions. The inhibitors zinc ions and cyclo-(l-His-l-Pro) are located at various positions and confirm the catalytic-site topology. Together with kinetics analyses of related mutants, the structures of ChiNCTU2 and its mutant complexes with (NAG)(n) provide new insights into its substrate binding and the mechanistic action.


Assuntos
Bacillus cereus/enzimologia , Proteínas de Bactérias/química , Quitinases/química , Substituição de Aminoácidos , Bacillus cereus/genética , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Sítios de Ligação , Catálise , Quitinases/genética , Quitinases/metabolismo , Cristalografia por Raios X , Dissacarídeos/química , Dissacarídeos/genética , Dissacarídeos/metabolismo , Cinética , Mutação de Sentido Incorreto , Estrutura Secundária de Proteína , Relação Estrutura-Atividade
14.
J Biol Chem ; 285(48): 37872-83, 2010 Nov 26.
Artigo em Inglês | MEDLINE | ID: mdl-20889969

RESUMO

Cysteine-rich secretory proteins (CRISPs) have been identified as a toxin family in most animal venoms with biological functions mainly associated with the ion channel activity of cysteine-rich domain (CRD). CRISPs also bind to Zn(2+) at their N-terminal pathogenesis-related (PR-1) domain, but their function remains unknown. Interestingly, similar the Zn(2+)-binding site exists in all CRISP family, including those identified in a wide range of organisms. Here, we report that the CRISP from Naja atra (natrin) could induce expression of vascular endothelial cell adhesion molecules, i.e. intercellular adhesion molecule-1, vascular adhesion molecule-1, and E-selectin, to promote monocytic cell adhesion in a heparan sulfate (HS)- and Zn(2+)-dependent manner. Using specific inhibitors and small interfering RNAs, the activation mechanisms are shown to involve both mitogen-activated protein kinases and nuclear factor-κB. Biophysical characterization of natrin by using fluorescence, circular dichroism, and x-ray crystallographic methods further reveals the presence of two Zn(2+)-binding sites for natrin. The strong binding site is located near the putative Ser-His-Glu catalytic triad of the N-terminal domain. The weak binding site remains to be characterized, but it may modulate HS binding by enhancing its interaction with long chain HS. Our results strongly suggest that natrin may serve as an inflammatory modulator that could perturb the wound-healing process of the bitten victim by regulating adhesion molecule expression in endothelial cells. Our finding uncovers a new aspect of the biological role of CRISP family in immune response and is expected to facilitate future development of new therapeutic strategy for the envenomed victims.


Assuntos
Moléculas de Adesão Celular/genética , Venenos Elapídicos/farmacologia , Elapidae , Células Endoteliais/imunologia , Heparitina Sulfato/imunologia , Mediadores da Inflamação/farmacologia , Transcrição Gênica/efeitos dos fármacos , Zinco/imunologia , Animais , Sítios de Ligação , Adesão Celular , Moléculas de Adesão Celular/imunologia , Linhagem Celular , Células Cultivadas , Venenos Elapídicos/química , Venenos Elapídicos/imunologia , Células Endoteliais/efeitos dos fármacos , Expressão Gênica/efeitos dos fármacos , Humanos , Mediadores da Inflamação/química , Mediadores da Inflamação/imunologia , Conformação Molecular , Ligação Proteica , Estrutura Terciária de Proteína , Difração de Raios X
15.
Mol Microbiol ; 78(5): 1101-16, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21059110

RESUMO

The crystal structures of two active forms of dissimilatory sulphite reductase (Dsr) from Desulfovibrio gigas, Dsr-I and Dsr-II, are compared at 1.76 and 2.05 Å resolution respectively. The dimeric α2ß2γ2 structure of Dsr-I contains eight [4Fe-4S] clusters, two saddle-shaped sirohaems and two flat sirohydrochlorins. In Dsr-II, the [4Fe-4S] cluster associated with the sirohaem in Dsr-I is replaced by a [3Fe-4S] cluster. Electron paramagnetic resonance (EPR) of the active Dsr-I and Dsr-II confirm the co-factor structures, whereas EPR of a third but inactive form, Dsr-III, suggests that the sirohaem has been demetallated in addition to its associated [4Fe-4S] cluster replaced by a [3Fe-4S] centre. In Dsr-I and Dsr-II, the sirohydrochlorin is located in a putative substrate channel connected to the sirohaem. The γ-subunit C-terminus is inserted into a positively charged channel formed between the α- and ß-subunits, with its conserved terminal Cys104 side-chain covalently linked to the CHA atom of the sirohaem in Dsr-I. In Dsr-II, the thioether bond is broken, and the Cys104 side-chain moves closer to the bound sulphite at the sirohaem pocket. These different forms of Dsr offer structural insights into a mechanism of sulphite reduction that can lead to S3O6(2-), S2O3(2-) and S2-.


Assuntos
Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Desulfovibrio gigas/enzimologia , Sulfito de Hidrogênio Redutase/química , Sulfito de Hidrogênio Redutase/metabolismo , Sequência de Aminoácidos , Proteínas de Bactérias/genética , Catálise , Domínio Catalítico , Desulfovibrio gigas/química , Desulfovibrio gigas/genética , Sulfito de Hidrogênio Redutase/genética , Conformação Molecular , Dados de Sequência Molecular
16.
Mol Cell Proteomics ; 8(9): 2034-50, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19505873

RESUMO

Transcription factor activating enhancer-binding protein 4 (AP-4) is a basic helix-loop-helix protein that binds to E-box elements. AP-4 has received increasing attention for its regulatory role in cell growth and development, including transcriptional repression of the human homolog of murine double minute 2 (HDM2), an important oncoprotein controlling cell growth and survival, by an unknown mechanism. Here we demonstrate that AP-4 binds to an E-box located in the HDM2-P2 promoter and represses HDM2 transcription in a p53-independent manner. Incremental truncations of AP-4 revealed that the C-terminal Gln/Pro-rich domain was essential for transcriptional repression of HDM2. To further delineate the molecular mechanism(s) of AP-4 transcriptional control and its potential implications, we used DNA-affinity purification followed by complementary quantitative proteomics, cICAT and iTRAQ labeling methods, to identify a previously unknown E-box-bound AP-4 protein complex containing 75 putative components. The two labeling methods complementarily quantified differentially AP-4-enriched proteins, including the most significant recruitment of DNA damage response proteins, followed by transcription factors, transcriptional repressors/corepressors, and histone-modifying proteins. Specific interaction of AP-4 with CCCTC binding factor, stimulatory protein 1, and histone deacetylase 1 (an AP-4 corepressor) was validated using AP-4 truncation mutants. Importantly, inclusion of trichostatin A did not alleviate AP-4-mediated repression of HDM2 transcription, suggesting a previously unidentified histone deacetylase-independent repression mechanism. In contrast, the complementary quantitative proteomics study suggested that transcription repression occurs via coordination of AP-4 with other transcription factors, histone methyltransferases, and/or a nucleosome remodeling SWI.SNF complex. In addition to previously known functions of AP-4, our data suggest that AP-4 participates in a transcriptional-regulating complex at the HDM2-P2 promoter in response to DNA damage.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Elementos E-Box/genética , Proteômica , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Proteínas Repressoras/metabolismo , Fatores de Transcrição/metabolismo , Transcrição Gênica , Sequência de Bases , Sítios de Ligação , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/química , Histona Desacetilases/metabolismo , Humanos , Ácidos Hidroxâmicos/farmacologia , Modelos Biológicos , Dados de Sequência Molecular , Proteínas Mutantes/metabolismo , Proteínas Nucleares/metabolismo , Regiões Promotoras Genéticas/genética , Ligação Proteica/efeitos dos fármacos , Estrutura Terciária de Proteína , Reprodutibilidade dos Testes , Fatores de Transcrição/química , Transcrição Gênica/efeitos dos fármacos , Proteína Supressora de Tumor p53/metabolismo
17.
J Struct Biol ; 169(3): 294-303, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19932752

RESUMO

The structures of snake venom metalloproteases (SVMPs) are proposed to be useful models to understand the structural and functional relationship of ADAM (a disintegrin and metalloprotease) which are membrane-anchored proteins involved in multiple human diseases. We have purified, sequenced and determined the structures of two new P-III SVMPs - atragin and kaouthiagin-like (K-like) from Naja atra. Atragin exhibits a known C-shaped topology, whereas K-like adopts an I-shaped conformation because of the distinct disulfide pattern in the disintegrin-like (D) domain. K-like exhibits an enzymatic specificity toward pro-TNFalpha with less inhibition of cell migration, but atragin shows the opposite effect. The specificity of the enzymatic activity is indicated to be dominated mainly by the local structures of SVMP in the metalloprotease (M) domain, whereas the hyper-variable region (HVR) in the cysteine-rich (C) domain is involved in a cell-migration activity. We demonstrate also a pH-dependent enzymatic activity of atragin that we correlate with the structural dynamics of a Zn(2+)-binding motif and the Met-turn based on the structures determined with a pH-jump method. The structural variations between the C- and I-shapes highlight the disulfide bond patterns in the D domain of the ADAM/adamalysin/reprolysins family proteins.


Assuntos
Venenos Elapídicos/enzimologia , Metaloproteases/química , Metaloproteases/metabolismo , Serpentes/metabolismo , Motivos de Aminoácidos/genética , Motivos de Aminoácidos/fisiologia , Animais , Cristalografia por Raios X , Concentração de Íons de Hidrogênio , Metaloendopeptidases/química , Metaloendopeptidases/genética , Metaloendopeptidases/metabolismo , Metaloproteases/genética , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína/genética , Estrutura Terciária de Proteína/fisiologia , Espectrometria de Fluorescência , Especificidade por Substrato/genética , Especificidade por Substrato/fisiologia
18.
Adv Exp Med Biol ; 677: 143-9, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20687487

RESUMO

Cobra venom contains cardiotoxins (CTXs) that induce tissue necrosis and systolic heart arrest in bitten victims. CTX-induced membrane pore formation is one of the major mechanisms responsible for the venom's designated cytotoxicity. This chapter examines how glycoconjugates such as heparan sulfates (HS) and glycosphingolipids, located respectively in the extracellular matrix and lipid bilayers of the cell membranes, facilitate CTX pore formation. Evidences for HS-facilitated cell surface retention and glycosphingolipid-facilitated membrane bilayer insertion of CTX are reviewed. We suggest that similar physical steps could play a role in the mediation of other pore forming toxins (PFT). The membrane pores formed by PFT are expected to have limited lifetime on biological cell surface as a result of membrane dynamics during endocytosis and/or rearrangement of lipid rafts.


Assuntos
Proteínas Cardiotóxicas de Elapídeos/química , Elapidae , Heparitina Sulfato/química , Bicamadas Lipídicas/química , Microdomínios da Membrana/química , Proteínas Citotóxicas Formadoras de Poros/química , Esfingolipídeos/química , Animais , Proteínas Cardiotóxicas de Elapídeos/metabolismo , Parada Cardíaca/metabolismo , Heparitina Sulfato/metabolismo , Humanos , Bicamadas Lipídicas/metabolismo , Microdomínios da Membrana/metabolismo , Necrose/metabolismo , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Esfingolipídeos/metabolismo
19.
Chemphyschem ; 10(3): 549-58, 2009 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-19142925

RESUMO

Previous nanoscale investigations of the gel-state membrane surface structure under the action of phospholipase A(2) (PLA(2)) suggest that single enzymes at work scoot on the membrane surface from the observed defects, which creates nanosized channels oriented along the lipid crystal-packing structure. To date, however, there have been no reports of direct observation of PLA(2) at the single-molecule level focusing on how the enzymes interact with the defects. Herein, we report a single-molecule fluorescence microscopy study on the action of enzymatically active rhodamine B-labeled cobra PLA(2) on a supported lipid membrane with visible packing defects on a glass substrate. Working with a gel-state phospholipid bilayer, the low-activity period (lag phase) of PLA(2) action is followed by the burst binding of PLA(2) molecules from aqueous solution on a few newly created active sites. These active sites are distinguished by a spatial resolution of approximately 40 nm, which is below the diffraction limit. The rate of active-site propagation as reflected by new PLA(2) binding on the membrane surface is estimated to be approximately 5 nm min(-1). This rate is about two orders of magnitude slower than the propagation rate of hydrolyzed channels estimated by AFM studies on bee venom PLA(2) on a similar membrane surface. This direct observation of PLA(2) molecules allows the visualization of different PLA(2) binding modes on the membrane surface and on the membrane boundary.


Assuntos
Venenos Elapídicos/química , Elapidae/metabolismo , Bicamadas Lipídicas/química , Fosfolipases A2/química , Fosfolipídeos/química , Animais , Fluorescência , Géis/química , Modelos Biológicos , Transição de Fase
20.
Vaccine ; 37(13): 1897-1903, 2019 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-30857635

RESUMO

Human infections of novel avian influenza A virus (H7N9) emerged in early 2013 and caused about 40% case-fatality through 2017. Therefore, development of influenza H7N9 vaccines is critical for pandemic preparedness. Currently, there are three means of production of commercial influenza vaccines: egg-based, mammalian cell-based, and insect cell-based platforms. The insect cell-based platform has the advantage of high speed in producing recombinant protein. In this study, we evaluate the stability and immunogenicity of two different influenza H7 HA expression constructs generated using the baculovirus system, including membrane-based full-length HA (mH7) and secreted ectodomain-based H7 (sH7). The mH7 construct could form an oligomer-rosette structure and had a high hemagglutinin (HA) titer 8192. In contrast to mH7, the sH7 construct could not form an oligomer-rosette structure and did not have HA titer before cross-linking with anti-His antibody. Thermal stability tests showed that the sH7 and mH7 constructs were unstable at 43 °C and 52 °C, respectively. In a mice immunization study, the mH7 construct but not the sH7 construct could induce robust HI and neutralizing antibody titers. In conclusion, further development of the mH7 vaccine candidate is desirable.


Assuntos
Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Imunogenicidade da Vacina , Subtipo H7N9 do Vírus da Influenza A/imunologia , Vacinas contra Influenza/imunologia , Influenza Humana/imunologia , Influenza Humana/prevenção & controle , Proteínas Recombinantes/imunologia , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Feminino , Expressão Gênica , Testes de Inibição da Hemaglutinação , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Humanos , Vacinas contra Influenza/administração & dosagem , Camundongos , Proteínas Recombinantes/genética
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa