Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Bioorg Med Chem ; 72: 116974, 2022 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-36108470

RESUMO

Human galectin 3 (Gal-3) has been implicated to play important roles in different biological recognition processes such as tumor growth and cancer metastasis. High-affinity Gal-3 ligands are desirable for functional studies and as inhibitors for potential therapeutic development. We report here a facile synthesis of ß-cyclodextrin (CD)-based Tn and TF antigen-containing multivalent ligands via a click reaction. Binding studies indicated that the synthetic multivalent glycan ligands demonstrated a clear clustering effect in binding to human Gal-3, with up to 153-fold enhanced relative affinity in comparison with the monomeric glycan ligand. The GalNAc (Tn antigen) containing heptavalent ligand showed the highest affinity for human Gal-3 among the synthetic ligands tested, with an EC50 of 1.4 µM in binding to human Gal-3. A cell-based assay revealed that the synthetic CD-based multivalent ligands could efficiently inhibit Gal-3 binding to human airway epithelial cells, with an inhibitory capacity consistent with their binding affinity measured by SPR. The synthetic cyclodextrin-based ligands described in this study should be valuable for functional studies of human Gal-3 and potentially for therapeutic applications.


Assuntos
Ciclodextrinas , beta-Ciclodextrinas , Galectina 3/metabolismo , Humanos , Ligantes , Ligação Proteica , beta-Ciclodextrinas/farmacologia
2.
Glycobiology ; 30(11): 895-909, 2020 10 21.
Artigo em Inglês | MEDLINE | ID: mdl-32280962

RESUMO

Pregnancy-specific beta 1 glycoprotein (PSG1) is secreted from trophoblast cells of the human placenta in increasing concentrations as pregnancy progresses, becoming one of the most abundant proteins in maternal serum in the third trimester. PSG1 has seven potential N-linked glycosylation sites across its four domains. We carried out glycomic and glycoproteomic studies to characterize the glycan composition of PSG1 purified from serum of pregnant women and identified the presence of complex N-glycans containing poly LacNAc epitopes with α2,3 sialyation at four sites. Using different techniques, we explored whether PSG1 can bind to galectin-1 (Gal-1) as these two proteins were previously shown to participate in processes required for a successful pregnancy. We confirmed that PSG1 binds to Gal-1 in a carbohydrate-dependent manner with an affinity of the interaction of 0.13 µM. In addition, we determined that out of the three N-glycosylation-carrying domains, only the N and A2 domains of recombinant PSG1 interact with Gal-1. Lastly, we observed that the interaction between PSG1 and Gal-1 protects this lectin from oxidative inactivation and that PSG1 competes the ability of Gal-1 to bind to some but not all of its glycoprotein ligands.


Assuntos
Galectina 1/metabolismo , Polissacarídeos/metabolismo , Glicoproteínas beta 1 Específicas da Gravidez/metabolismo , Feminino , Galectina 1/química , Humanos , Ligantes , Polissacarídeos/química , Gravidez , Glicoproteínas beta 1 Específicas da Gravidez/química , Glicoproteínas beta 1 Específicas da Gravidez/isolamento & purificação
3.
Biochemistry ; 54(30): 4711-30, 2015 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-26158802

RESUMO

Galectins are highly conserved lectins that are key to multiple biological functions, including pathogen recognition and regulation of immune responses. We previously reported that CvGal1, a galectin expressed in phagocytic cells (hemocytes) of the eastern oyster (Crassostrea virginica), is hijacked by the parasite Perkinsus marinus to enter the host, where it causes systemic infection and death. Screening of an oyster hemocyte cDNA library revealed a novel galectin, which we designated CvGal2, with four tandemly arrayed carbohydrate recognition domains (CRDs). Phylogentic analysis of the CvGal2 CRDs suggests close relationships with homologous CRDs from CvGal1. Glycan array analysis, however, revealed that, unlike CvGal1 which preferentially binds to the blood group A tetrasaccharide, CvGal2 recognizes both blood group A and B tetrasaccharides and related structures, suggesting that CvGal2 has broader binding specificity. Furthermore, SPR analysis demonstrated significant differences in the binding kinetics of CvGal1 and CvGal2, and structural modeling revealed substantial differences in their interactions with the oligosaccharide ligands. CvGal2 is homogeneously distributed in the hemocyte cytoplasm, is released to the extracellular space, and binds to the hemocyte surface. CvGal2 binds to P. marinus trophozoites in a dose-dependent and ß-galactoside-specific manner. Strikingly, negligible binding of CvGal2 was observed for Perkinsus chesapeaki, a sympatric parasite species mostly prevalent in the clams Mya arenaria and Macoma balthica. The differential recognition of Perkinsus species by the oyster galectins is consistent with their relative prevalence in oyster and clam species and supports their role in facilitating parasite entry and infectivity in a host-preferential manner.


Assuntos
Alveolados , Antígenos de Grupos Sanguíneos , Crassostrea , Galectinas , Oligossacarídeos , Filogenia , Alveolados/química , Alveolados/genética , Alveolados/metabolismo , Animais , Antígenos de Grupos Sanguíneos/química , Antígenos de Grupos Sanguíneos/genética , Antígenos de Grupos Sanguíneos/metabolismo , Crassostrea/química , Crassostrea/genética , Crassostrea/metabolismo , Crassostrea/parasitologia , Galectinas/química , Galectinas/genética , Galectinas/metabolismo , Hemócitos/química , Hemócitos/metabolismo , Hemócitos/parasitologia , Oligossacarídeos/química , Oligossacarídeos/genética , Oligossacarídeos/metabolismo
4.
J Immunol ; 191(9): 4828-37, 2013 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-24068662

RESUMO

We previously reported that removal of sialyl residues primed PBMCs to respond to bacterial LPS stimulation in vitro. Therefore, we speculated that prior desialylation can sensitize the host to generate an enhanced inflammatory response upon exposure to a TLR ligand, such as LPS, in a murine model of acute lung injury. Intratracheal instillation of neuraminidase (NA) 30 min prior to intratracheal administration of LPS increased polymorphonuclear leukocytes (PMNs) in the bronchoalveolar lavage fluid and the wet-to-dry lung weight ratio, a measure of pulmonary edema, compared with mice that received LPS alone. Administration of NA alone resulted in desialylation of bronchiolar and alveolar surfaces and induction of TNF-α, IL-1ß, and chemokines in lung homogenates and bronchoalveolar lavage fluid; however, PMN recruitment in mice treated with NA alone did not differ from that of PBS-administered controls. NA pretreatment alone induced apoptosis and markedly enhanced LPS-induced endothelial apoptosis. Administration of recombinant Bcl-2, an antiapoptotic molecule, abolished the effect of NA treatment on LPS-induced PMN recruitment and pulmonary edema formation. We conclude that NA pretreatment potentiates LPS-induced lung injury through enhanced PMN recruitment, pulmonary edema formation, and endothelial and myeloid cell apoptosis. A similar "reprogramming" of immune responses with desialylation may occur during respiratory infection with NA-expressing microbes and contribute to severe lung injury.


Assuntos
Lesão Pulmonar Aguda/imunologia , Neuraminidase/metabolismo , Neutrófilos/imunologia , Lesão Pulmonar Aguda/induzido quimicamente , Animais , Apoptose/imunologia , Líquido da Lavagem Broncoalveolar/citologia , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Lipopolissacarídeos/administração & dosagem , Pulmão/citologia , Pulmão/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Mieloides/metabolismo , Ácido N-Acetilneuramínico/metabolismo , Neuraminidase/administração & dosagem , Proteínas Proto-Oncogênicas c-bcl-2/administração & dosagem , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Edema Pulmonar/imunologia
5.
J Biol Chem ; 288(34): 24394-409, 2013 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-23824193

RESUMO

The galectin CvGal1 from the eastern oyster (Crassostrea virginica), which possesses four tandemly arrayed carbohydrate recognition domains, was previously shown to display stronger binding to galactosamine and N-acetylgalactosamine relative to d-galactose. CvGal1 expressed by phagocytic cells is "hijacked" by the parasite Perkinsus marinus to enter the host, where it proliferates and causes systemic infection and death. In this study, a detailed glycan array analysis revealed that CvGal1 preferentially recognizes type 2 blood group A oligosaccharides. Homology modeling of the protein and its oligosaccharide ligands supported this preference over type 1 blood group A and B oligosaccharides. The CvGal ligand models were further validated by binding, inhibition, and competitive binding studies of CvGal1 and ABH-specific monoclonal antibodies with intact and deglycosylated glycoproteins, hemocyte extracts, and intact hemocytes and by surface plasmon resonance analysis. A parallel glycomic study carried out on oyster hemocytes (Kurz, S., Jin, C., Hykollari, A., Gregorich, D., Giomarelli, B., Vasta, G. R., Wilson, I. B. H., and Paschinger, K. (2013) J. Biol. Chem. 288) determined the structures of oligosaccharides recognized by CvGal1. Proteomic analysis of the hemocyte glycoproteins identified ß-integrin and dominin as CvGal1 "self"-ligands. Despite strong CvGal1 binding to P. marinus trophozoites, no binding of ABH blood group antibodies was observed. Thus, parasite glycans structurally distinct from the blood group A oligosaccharides on the hemocyte surface may function as potentially effective ligands for CvGal1. We hypothesize that carbohydrate-based mimicry resulting from the host/parasite co-evolution facilitates CvGal1-mediated cross-linking to ß-integrin, located on the hemocyte surface, leading to cell activation, phagocytosis, and host infection.


Assuntos
Sistema ABO de Grupos Sanguíneos/química , Crassostrea/química , Galectinas/química , Hemócitos/química , Oligossacarídeos/química , Sistema ABO de Grupos Sanguíneos/genética , Sistema ABO de Grupos Sanguíneos/metabolismo , Animais , Crassostrea/genética , Crassostrea/metabolismo , Crassostrea/parasitologia , Galectinas/genética , Galectinas/metabolismo , Hemócitos/metabolismo , Hemócitos/parasitologia , Oligossacarídeos/genética , Oligossacarídeos/metabolismo , Ligação Proteica , Proteômica/métodos
6.
Am J Physiol Lung Cell Mol Physiol ; 306(9): L876-86, 2014 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-24658138

RESUMO

Sialic acids on glycoconjugates play a pivotal role in many biological processes. In the airways, sialylated glycoproteins and glycolipids are strategically positioned on the plasma membranes of epithelia to regulate receptor-ligand, cell-cell, and host-pathogen interactions at the molecular level. We now demonstrate, for the first time, sialidase activity for ganglioside substrates in human airway epithelia. Of the four known mammalian sialidases, NEU3 has a substrate preference for gangliosides and is expressed at mRNA and protein levels at comparable abundance in epithelia derived from human trachea, bronchi, small airways, and alveoli. In small airway and alveolar epithelia, NEU3 protein was immunolocalized to the plasma membrane, cytosolic, and nuclear subcellular fractions. Small interfering RNA-induced silencing of NEU3 expression diminished sialidase activity for a ganglioside substrate by >70%. NEU3 immunostaining of intact human lung tissue could be localized to the superficial epithelia, including the ciliated brush border, as well as to nuclei. However, NEU3 was reduced in subepithelial tissues. These results indicate that human airway epithelia express catalytically active NEU3 sialidase.


Assuntos
Membrana Celular/metabolismo , Núcleo Celular/metabolismo , Citosol/metabolismo , Epitélio/metabolismo , Neuraminidase/metabolismo , Sistema Respiratório/metabolismo , Biotinilação , Western Blotting , Catálise , Células Cultivadas , Citometria de Fluxo , Gangliosídeos/metabolismo , Humanos , Técnicas Imunoenzimáticas , Neuraminidase/genética , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ácidos Siálicos/metabolismo , Frações Subcelulares
7.
J Clin Invest ; 134(9)2024 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-38483511

RESUMO

In lung, thromboxane A2 (TXA2) activates the TP receptor to induce proinflammatory and bronchoconstrictor effects. Thus, TP receptor antagonists and TXA2 synthase inhibitors have been tested as potential asthma therapeutics in humans. Th9 cells play key roles in asthma and regulate the lung immune response to allergens. Herein, we found that TXA2 reduces Th9 cell differentiation during allergic lung inflammation. Th9 cells were decreased approximately 2-fold and airway hyperresponsiveness was attenuated in lungs of allergic mice treated with TXA2. Naive CD4+ T cell differentiation to Th9 cells and IL-9 production were inhibited dose-dependently by TXA2 in vitro. TP receptor-deficient mice had an approximately 2-fold increase in numbers of Th9 cells in lungs in vivo after OVA exposure compared with wild-type mice. Naive CD4+ T cells from TP-deficient mice exhibited increased Th9 cell differentiation and IL-9 production in vitro compared with CD4+ T cells from wild-type mice. TXA2 also suppressed Th2 and enhanced Treg differentiation both in vitro and in vivo. Thus, in contrast to its acute, proinflammatory effects, TXA2 also has longer-lasting immunosuppressive effects that attenuate the Th9 differentiation that drives asthma progression. These findings may explain the paradoxical failure of anti-thromboxane therapies in the treatment of asthma.


Assuntos
Asma , Diferenciação Celular , Linfócitos T Reguladores , Células Th2 , Tromboxano A2 , Animais , Camundongos , Células Th2/imunologia , Células Th2/patologia , Tromboxano A2/metabolismo , Tromboxano A2/imunologia , Linfócitos T Reguladores/imunologia , Asma/imunologia , Asma/patologia , Asma/tratamento farmacológico , Asma/genética , Camundongos Knockout , Interleucina-9/imunologia , Interleucina-9/genética , Interleucina-9/metabolismo , Pneumonia/imunologia , Pneumonia/patologia , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos BALB C , Pulmão/imunologia , Pulmão/patologia , Ovalbumina/imunologia , Feminino , Linfócitos T Auxiliares-Indutores/imunologia
8.
J Biol Chem ; 287(19): 15966-80, 2012 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-22403397

RESUMO

The microvascular endothelial surface expresses multiple molecules whose sialylation state regulates multiple aspects of endothelial function. To better regulate these sialoproteins, we asked whether endothelial cells (ECs) might express one or more catalytically active sialidases. Human lung microvascular EC lysates contained heat-labile sialidase activity for a fluorogenic substrate, 2'-(4-methylumbelliferyl)-α-D-N-acetylneuraminic acid (4-MU-NANA), that was dose-dependently inhibited by the competitive sialidase inhibitor, 2,3-dehydro-2-deoxy-N-acetylneuraminic acid but not its negative control. The EC lysates also contained sialidase activity for a ganglioside mixture. Using real time RT-PCR to detect mRNAs for the four known mammalian sialidases, NEU1, -2, -3, and -4, NEU1 mRNA was expressed at levels 2700-fold higher that those found for NEU2, -3, or -4. Western analyses indicated NEU1 and -3 protein expression. Using confocal microscopy and flow cytometry, NEU1 was immunolocalized to both the plasma membrane and the perinuclear region. NEU3 was detected both in the cytosol and nucleus. Prior siRNA-mediated knockdown of NEU1 and NEU3 each decreased EC sialidase activity for 4-MU-NANA by >65 and >17%, respectively, and for the ganglioside mixture by 0 and 40%, respectively. NEU1 overexpression in ECs reduced their migration into a wound by >40%, whereas NEU3 overexpression did not. Immunohistochemical studies of normal human tissues immunolocalized NEU1 and NEU3 proteins to both pulmonary and extrapulmonary vascular endothelia. These combined data indicate that human lung microvascular ECs as well as other endothelia express catalytically active NEU1 and NEU3. NEU1 restrains EC migration, whereas NEU3 does not.


Assuntos
Movimento Celular , Células Endoteliais/enzimologia , Neuraminidase/metabolismo , Aorta/enzimologia , Artérias Carótidas/enzimologia , Linhagem Celular , Membrana Celular/enzimologia , Núcleo Celular/enzimologia , Artérias Cerebrais/enzimologia , Citosol/enzimologia , Células Endoteliais/metabolismo , Citometria de Fluxo , Regulação Enzimológica da Expressão Gênica , Humanos , Himecromona/análogos & derivados , Himecromona/farmacologia , Immunoblotting , Rim/enzimologia , Pulmão/enzimologia , Microscopia Confocal , Ácido N-Acetilneuramínico/análogos & derivados , Ácido N-Acetilneuramínico/metabolismo , Neuraminidase/antagonistas & inibidores , Neuraminidase/genética , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Especificidade por Substrato
9.
J Biol Chem ; 287(11): 8214-31, 2012 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-22247545

RESUMO

Epithelial cells (ECs) lining the airways provide a protective barrier between the external environment and the internal host milieu. These same airway epithelia express receptors that respond to danger signals and initiate repair programs. Because the sialylation state of a receptor can influence its function and is dictated in part by sialidase activity, we asked whether airway epithelia express catalytically active sialidase(s). Human primary small airway and A549 ECs expressed NEU1 sialidase at the mRNA and protein levels, and NEU1 accounted for >70% of EC sialidase activity. Blotting with Maackia amurensis and peanut agglutinin lectins established epidermal growth factor receptor (EGFR) and MUC1 as in vivo substrates for NEU1. NEU1 associated with EGFR and MUC1, and NEU1-EGFR association was regulated by EGF stimulation. NEU1 overexpression diminished EGF-stimulated EGFR Tyr-1068 autophosphorylation by up to 44% but enhanced MUC1-dependent Pseudomonas aeruginosa adhesion by 1.6-1.7-fold and flagellin-stimulated ERK1/2 activation by 1.7-1.9-fold. In contrast, NEU1 depletion increased EGFR activation (1.5-fold) and diminished MUC1-mediated bacterial adhesion (38-56%) and signaling (73%). These data indicate for the first time that human airway epithelia express catalytically active NEU1 sialidase that regulates EGFR- and MUC1-dependent signaling and bacterial adhesion. NEU1 catalytic activity may offer an additional level of regulation over the airway epithelial response to ligands, pathogens, and injurious stimuli.


Assuntos
Receptores ErbB/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Mucina-1/metabolismo , Neuraminidase/biossíntese , Mucosa Respiratória/metabolismo , Linhagem Celular Transformada , Fator de Crescimento Epidérmico/genética , Fator de Crescimento Epidérmico/metabolismo , Receptores ErbB/genética , Regulação Enzimológica da Expressão Gênica/genética , Humanos , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Mucina-1/genética , Neuraminidase/genética , Infecções por Pseudomonas/genética , Infecções por Pseudomonas/metabolismo , Infecções por Pseudomonas/microbiologia , Pseudomonas aeruginosa/metabolismo , Mucosa Respiratória/microbiologia
10.
Mol Immunol ; 156: 127-135, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36921487

RESUMO

The lung airway epithelial surface is heavily covered with sialic acids as the terminal carbohydrate on most cell surface glycoconjugates and can be removed by microbial neuraminidases or endogenous sialidases. By desialylating the lung epithelial surface, neuraminidase acts as an important virulence factor in many mucosal pathogens, such as influenza and S. pneumoniae. Desialylation exposes the subterminal galactosyl moieties - the binding glycotopes for galectins, a family of carbohydrate-recognition proteins playing important roles in various aspects of immune responses. Galectin-1 and galectin-3 have been extensively studied in their roles related to host immune responses, but some questions about their role(s) in leukocyte recruitment during lung bacterial infection remain unanswered. In this study, we found that both galectin-1 and galectin-3 bind to polymorphonuclear leukocytes (PMNs) and enhance the interaction of endothelial intercellular adhesion molecule-1 (ICAM-1) with PMNs, which is further increased by PMN desialylation. In addition, we observed that in vitro galectin-1 mediates the binding of PMNs, particularly desialylated PMNs, onto the endothelial cells. Finally, in a murine model for LPS-mediated acute lung injury, we observed that galectin-1 modulates PMN infiltration to the lung without altering the expression of chemoattractant cytokines. We conclude that galectins, particularly galectin-1, may function as adhesion molecules that mediate PMN-endothelial cell interactions, and modulate PMN infiltration during acute lung injury.


Assuntos
Lesão Pulmonar Aguda , Neutrófilos , Humanos , Camundongos , Animais , Lipopolissacarídeos/farmacologia , Células Endoteliais , Galectina 1 , Galectina 3 , Adesão Celular , Pulmão , Lesão Pulmonar Aguda/induzido quimicamente , Lesão Pulmonar Aguda/metabolismo , Streptococcus pneumoniae , Molécula 1 de Adesão Intercelular/metabolismo
11.
BMC Microbiol ; 12: 2, 2012 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-22230035

RESUMO

BACKGROUND: A common finding amongst patients with inhalational anthrax is a paucity of polymorphonuclear leukocytes (PMNs) in infected tissues in the face of abundant circulating PMNs. A major virulence determinant of anthrax is edema toxin (ET), which is formed by the combination of two proteins produced by the organism, edema factor (EF), which is an adenyl cyclase, and protective antigen (PA). Since cAMP, a product of adenyl cyclase, is known to enhance endothelial barrier integrity, we asked whether ET might decrease extravasation of PMNs into tissues through closure of the paracellular pathway through which PMNs traverse. RESULTS: Pretreatment of human microvascular endothelial cell(EC)s of the lung (HMVEC-L) with ET decreased interleukin (IL)-8-driven transendothelial migration (TEM) of PMNs with a maximal reduction of nearly 60%. This effect required the presence of both EF and PA. Conversely, ET did not diminish PMN chemotaxis in an EC-free system. Pretreatment of subconfluent HMVEC-Ls decreased transendothelial 14 C-albumin flux by ~ 50% compared to medium controls. Coadministration of ET with either tumor necrosis factor-α or bacterial lipopolysaccharide, each at 100 ng/mL, attenuated the increase of transendothelial 14 C-albumin flux caused by either agent alone. The inhibitory effect of ET on TEM paralleled increases in protein kinase A (PKA) activity, but could not be blocked by inhibition of PKA with either H-89 or KT-5720. Finally, we were unable to replicate the ET effect with either forskolin or 3-isobutyl-1-methylxanthine, two agents known to increase cAMP. CONCLUSIONS: We conclude that ET decreases IL-8-driven TEM of PMNs across HMVEC-L monolayers independent of cAMP/PKA activity.


Assuntos
Antígenos de Bactérias/toxicidade , Bacillus anthracis/patogenicidade , Toxinas Bacterianas/toxicidade , Movimento Celular , Células Endoteliais/efeitos dos fármacos , Substâncias Macromoleculares/metabolismo , Neutrófilos/fisiologia , Radioisótopos de Carbono/metabolismo , Células Cultivadas , Células Endoteliais/fisiologia , Humanos , Interleucina-8/metabolismo , Marcação por Isótopo
12.
Stat Med ; 31(7): 619-27, 2012 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-21344469

RESUMO

Traditional Chinese medicine (TCM), used in China and other Asian counties for thousands of years, is increasingly utilized in Western countries. However, due to inherent differences in how Western medicine and this ancient modality are practiced, employing the so-called Western medicine-based gold standard research methods to evaluate TCM is challenging. This paper is a discussion of the obstacles inherent in the design and statistical analysis of clinical trials of TCM. It is based on our experience in designing and conducting a randomized controlled clinical trial of acupuncture for post-operative dental pain control in which acupuncture was shown to be statistically and significantly better than placebo in lengthening the median survival time to rescue drug. We demonstrate here that PH assumptions in the common Cox model did not hold in that trial and that TCM trials warrant more thoughtful modeling and more sophisticated models of statistical analysis. TCM study design entails all the challenges encountered in trials of drugs, devices, and surgical procedures in the Western medicine. We present possible solutions to some but leave many issues unresolved.


Assuntos
Terapia por Acupuntura/estatística & dados numéricos , Interpretação Estatística de Dados , Medicina Tradicional Chinesa , Ensaios Clínicos Controlados Aleatórios como Assunto/estatística & dados numéricos , Projetos de Pesquisa/estatística & dados numéricos , Humanos , Odontalgia/terapia
13.
Methods Mol Biol ; 2442: 425-443, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35320539

RESUMO

Techniques for disrupting gene expression are invaluable tools for the analysis of the biological role of a gene product. Because of its genetic tractability and multiple advantages over conventional mammalian models, the zebrafish (Danio rerio) is recognized as a powerful system for gaining new insight into diverse aspects of human health and disease. Among the multiple mammalian gene families for which the zebrafish has shown promise as an invaluable model for functional studies, the galectins have attracted great interest due to their participation in early development, regulation of immune homeostasis, and recognition of microbial pathogens. Galectins are ß-galactosyl-binding lectins with a characteristic sequence motif in their carbohydrate recognition domains (CRDs), that constitute an evolutionary conserved family ubiquitous in eukaryotic taxa. Galectins are emerging as key players in the modulation of many important pathological processes, which include acute and chronic inflammatory diseases, autoimmunity and cancer, thus making them potential molecular targets for innovative drug discovery. Here, we provide a review of the current methods available for the manipulation of gene expression in the zebrafish, with a focus on gene knockdown [morpholino (MO)-derived antisense oligonucleotides] and knockout (CRISPR-Cas) technologies.


Assuntos
Galectinas , Peixe-Zebra , Animais , Galectinas/metabolismo , Técnicas de Silenciamento de Genes , Mamíferos/genética , Morfolinos/genética , Morfolinos/metabolismo , RNA/metabolismo , Peixe-Zebra/metabolismo
14.
Sci Rep ; 12(1): 14173, 2022 08 19.
Artigo em Inglês | MEDLINE | ID: mdl-35986080

RESUMO

To gain insight into sialic acid biology and sialidase/neuraminidase (NEU) expression in mature human neutrophil (PMN)s, we studied NEU activity and expression in PMNs and the HL60 promyelocytic leukemic cell line, and changes that might occur in PMNs undergoing apoptosis and HL60 cells during their differentiation into PMN-like cells. Mature human PMNs contained NEU activity and expressed NEU2, but not NEU1, the NEU1 chaperone, protective protein/cathepsin A(PPCA), NEU3, and NEU4 proteins. In proapoptotic PMNs, NEU2 protein expression increased > 30.0-fold. Granulocyte colony-stimulating factor protected against NEU2 protein upregulation, PMN surface desialylation and apoptosis. In response to 3 distinct differentiating agents, dimethylformamide, dimethylsulfoxide, and retinoic acid, total NEU activity in differentiated HL60 (dHL60) cells was dramatically reduced compared to that of nondifferentiated cells. With differentiation, NEU1 protein levels decreased > 85%, PPCA and NEU2 proteins increased > 12.0-fold, and 3.0-fold, respectively, NEU3 remained unchanged, and NEU4 increased 1.7-fold by day 3, and then returned to baseline. In dHL60 cells, lectin blotting revealed decreased α2,3-linked and increased α2,6-linked sialylation. dHL60 cells displayed increased adhesion to and migration across human bone marrow-derived endothelium and increased bacterial phagocytosis. Therefore, myeloid apoptosis and differentiation provoke changes in NEU catalytic activity and protein expression, surface sialylation, and functional responsiveness.


Assuntos
Ácido N-Acetilneuramínico , Neuraminidase , Apoptose , Diferenciação Celular , Humanos , Ácido N-Acetilneuramínico/metabolismo , Neuraminidase/metabolismo , Neutrófilos/metabolismo
15.
J Exp Med ; 218(2)2021 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-33216117

RESUMO

Two cosegregating single-nucleotide polymorphisms (SNPs) in human TLR4, an A896G transition at SNP rs4986790 (D299G) and a C1196T transition at SNP rs4986791 (T399I), have been associated with LPS hyporesponsiveness and differential susceptibility to many infectious or inflammatory diseases. However, many studies failed to confirm these associations, and transfection experiments resulted in conflicting conclusions about the impact of these SNPs on TLR4 signaling. Using advanced protein modeling from crystallographic data of human and murine TLR4, we identified homologous substitutions of these SNPs in murine Tlr4, engineered a knock-in strain expressing the D298G and N397I TLR4 SNPs homozygously, and characterized in vivo and in vitro responses to TLR4 ligands and infections in which TLR4 is implicated. Our data provide new insights into cellular and molecular mechanisms by which these SNPs decrease the TLR4 signaling efficiency and offer an experimental approach to confirm or refute human data possibly confounded by variables unrelated to the direct effects of the SNPs on TLR4 functionality.


Assuntos
Lipopolissacarídeos/genética , Polimorfismo de Nucleotídeo Único/genética , Receptor 4 Toll-Like/genética , Animais , Modelos Animais de Doenças , Feminino , Predisposição Genética para Doença/genética , Humanos , Masculino , Camundongos , Transdução de Sinais/genética
16.
Methods Mol Biol ; 2132: 225-239, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32306331

RESUMO

F-type lectins (FTLs) are characterized by a fucose recognition domain (F-type lectin domain; FTLD) that displays a novel jellyroll fold ("F-type" fold) and unique carbohydrate- and calcium-binding sequence motifs. This novel lectin family comprises widely distributed proteins exhibiting single, double, or greater multiples of the FTLD, either tandemly arrayed or combined with other structurally and functionally distinct domains. Further, differences in carbohydrate specificity among tandemly arrayed FTLDs present in any FTL polypeptide subunit, together with the expression of multiple FTL isoforms in a single individual supports a striking diversity in ligand recognition. Functions of FTLs in self/nonself recognition include innate immunity, fertilization, microbial adhesion, and pathogenesis, among others, revealing an extensive structural/functional diversification. The taxonomic distribution of FTLDs is surprisingly discontinuous, suggesting that this lectin family has been subject to secondary loss, lateral transfer, and functional co-option along evolutionary lineages.


Assuntos
Fucose/metabolismo , Lectinas/química , Lectinas/metabolismo , Sítios de Ligação , Sequência de Carboidratos , Evolução Molecular , Lectinas/genética , Modelos Moleculares , Família Multigênica , Ligação Proteica , Conformação Proteica , Domínios Proteicos
17.
Methods Mol Biol ; 2132: 241-255, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32306332

RESUMO

The purification of fucose-binding lectins from the liver of striped bass (Morone saxatilis), a teleost fish, and the identification of a novel lectin sequence motif led to the identification of a new family of lectins, the F-type lectins (FTLs) (see overview of the FTL family in Chapter 23 ). Isolation and purification of these proteins from liver extracts of striped bass was accomplished by affinity chromatography and size exclusion, and their identification as FTLs, by direct Edman sequencing, and protein, transcript, and gene sequence analysis. The development of specific antibodies against the M. saxatilis FTL provided an additional tool for the identification of FTLs. These methods have been successfully used for the purification of the FTL family members from tissues and body fluids of various animal species. Production and characterization of FTLs has been facilitated by the expression of the recombinant proteins. In this chapter, the biochemical characterization of FTLs is focused on the analysis of their carbohydrate specificity.


Assuntos
Bass/metabolismo , Lectinas/genética , Lectinas/isolamento & purificação , Fígado/metabolismo , Animais , Bass/genética , Cromatografia de Afinidade , Cromatografia em Gel , Proteínas de Peixes/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Lectinas/metabolismo , Família Multigênica , Proteínas Recombinantes/metabolismo
18.
Front Chem ; 8: 98, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32161746

RESUMO

Both vertebrates and invertebrates display active innate immune mechanisms for defense against microbial infection, including diversified repertoires of soluble and cell-associated lectins that can effect recognition and binding to potential pathogens, and trigger downstream effector pathways that clear them from the host internal milieu. Galectins are widely distributed and highly conserved lectins that have key regulatory effects on both innate and adaptive immune responses. In addition, galectins can bind to exogenous ("non-self") carbohydrates on the surface of bacteria, enveloped viruses, parasites, and fungi, and function as recognition receptors and effector factors in innate immunity. Like most invertebrates, eastern oysters (Crassostrea virginica) and softshell clams (Mya arenaria) can effectively respond to most immune challenges through soluble and hemocyte-associated lectins. The protozoan parasite Perkinsus marinus, however, can infect eastern oysters and cause "Dermo" disease, which is highly detrimental to both natural and farmed oyster populations. The sympatric Perkinsus chesapeaki, initially isolated from infected M. arenaria clams, can also be present in oysters, and there is little evidence of pathogenicity in either clams or oysters. In this review, we discuss selected observations from our studies on the mechanisms of Perkinsus recognition that are mediated by galectin-carbohydrate interactions. We identified in the oyster two galectins that we designated CvGal1 and CvGal2, which strongly recognize P. marinus trophozoites. In the clam we also identified galectin sequences, and focused on one (that we named MaGal1) that also recognizes Perkinsus species. Here we describe the biochemical characterization of CvGal1, CvGal2, and MaGal1 with focus on the detailed study of the carbohydrate specificity, and the glycosylated moieties on the surfaces of the oyster hemocytes and the two Perkinsus species (P. marinus and P. chesapeaki). Our goal is to gain further understanding of the biochemical basis for the interactions that lead to recognition and opsonization of the Perkinsus trophozoites by the bivalve hemocytes. These basic studies on the biology of host-parasite interactions may contribute to the development of novel intervention strategies for parasitic diseases of biomedical interest.

19.
Dev Comp Immunol ; 78: 61-70, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28939483

RESUMO

LvMKK4, a homologue of the mammalian mitogen-activated protein kinase kinase 4 (MKK4), was isolated and identified from Litopenaeus vannamei in the present study. The full-length cDNA of LvMKK4 is 1947 bp long, with an open reading frame (ORF) of 1185 bp encoding a putative protein of 388 amino acids. LvMKK4 contains several characteristic domains such as D domain, SIAKT motif and kinase domain, all of which are conserved in MAP kinase kinase family. Like mammalian MKK4 but not Drosophila MKK4, LvMKK4 could bind to, phosphorylate and activate p38 MAPK, which provided some insights into the signal transduction mechanism of MKK4-p38 cascade in invertebrates. Our real-time PCR data indicated that LvMKK4 was ubiquitously expressed in all tested tissues and extraordinarily abundant in muscle. Dual luciferase reporter assays in Drosophila S2 cells revealed that LvMKK4 activated the transcription of antimicrobial peptide genes (AMPs), including Drosophila Attacin A, Drosomycin, and shrimp Penaeidins. Additionally, LvMKK4 was up-regulated in both intestine and hepatopancreas by a variety of inflammatory stimuli including LPS, Vibrio parahaemolyticus, Staphhylococcu saureus, Poly (I: C) and white spot syndrome virus. Furthermore, RNAi-mediated knockdown of LvMKK4 enhanced the sensitivity of L. vannamei to V. parahaemolyticus infection. These findings suggested that LvMKK4 played an important role in anti-bacterial response and could be a potential target for inflammation treatment.


Assuntos
Antibacterianos/metabolismo , Proteínas de Artrópodes/metabolismo , Infecções por Vírus de DNA/imunologia , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Penaeidae/imunologia , Infecções Estafilocócicas/imunologia , Staphylococcus aureus/imunologia , Vibrioses/imunologia , Vibrio parahaemolyticus/imunologia , Vírus da Síndrome da Mancha Branca 1/imunologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Proteínas de Artrópodes/genética , Linhagem Celular , Clonagem Molecular , Drosophila/genética , Drosophila/imunologia , Regulação da Expressão Gênica , Humanos , Imunidade Inata/genética , MAP Quinase Quinase 4/genética , Mamíferos , Quinases de Proteína Quinase Ativadas por Mitógeno/genética , Penaeidae/genética , Poli I-C/imunologia , Ligação Proteica , RNA Interferente Pequeno/genética
20.
Pathog Dis ; 75(5)2017 07 31.
Artigo em Inglês | MEDLINE | ID: mdl-28449072

RESUMO

Carbohydrate structures on the cell surface encode complex information that through specific recognition by carbohydrate-binding proteins (lectins) modulates interactions between cells, cells and the extracellular matrix, or mediates recognition of potential microbial pathogens. Galectins are a family of ß-galactoside-binding lectins, which are evolutionary conserved and have been identified in most organisms, from fungi to invertebrates and vertebrates, including mammals. Since their discovery in the 1970s, their biological roles, initially understood as limited to recognition of endogenous carbohydrate ligands in embryogenesis and development, have expanded in recent years by the discovery of their roles in tissue repair and regulation of immune homeostasis. More recently, evidence has accumulated to support the notion that galectins can also bind glycans on the surface of potentially pathogenic microbes, and function as recognition and effector factors in innate immunity, thus establishing a new paradigm. Furthermore, some parasites 'subvert' the recognition roles of the vector/host galectins for successful attachment or invasion. These recent findings have revealed a striking functional diversification in this structurally conserved lectin family.


Assuntos
Galectinas/metabolismo , Interações Hospedeiro-Patógeno , Evasão da Resposta Imune , Imunidade Inata , Receptores Imunológicos/metabolismo , Animais , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA