Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
J Biol Chem ; 295(4): 1009-1020, 2020 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-31831622

RESUMO

Cancer remains a leading cause of morbidity and mortality worldwide, requiring ongoing development of targeted therapeutics such as monoclonal antibodies. Carbohydrates on embryonic cells are often highly expressed in cancer and are therefore attractive targets for antibodies. Stage-specific embryonic antigen-4 (SSEA-4) is one such glycolipid target expressed in many cancers, including breast and ovarian carcinomas. Here, we defined the structural basis for recognition of SSEA-4 by a novel monospecific chimeric antibody (ch28/11). Five X-ray structures of ch28/11 Fab complexes with the SSEA-4 glycan headgroup, determined at 1.5-2.7 Å resolutions, displayed highly similar three-dimensional structures indicating a stable binding mode. The structures also revealed that by adopting a horseshoe-shaped conformation in a deep groove, the glycan headgroup likely sits flat against the membrane to allow the antibody to interact with SSEA-4 on cancer cells. Moreover, we found that the terminal sialic acid of SSEA-4 plays a dominant role in dictating the exquisite specificity of the ch28/11 antibody. This observation was further supported by molecular dynamics simulations of the ch28/11-glycan complex, which show that SSEA-4 is stabilized by its terminal sialic acid, unlike SSEA-3, which lacks this sialic acid modification. These high-resolution views of how a glycolipid interacts with an antibody may help to advance a new class of cancer-targeting immunotherapy.


Assuntos
Anticorpos Antineoplásicos/imunologia , Ácido N-Acetilneuramínico/metabolismo , Neoplasias/imunologia , Antígenos Embrionários Estágio-Específicos/metabolismo , Anticorpos Antineoplásicos/química , Especificidade de Anticorpos/imunologia , Configuração de Carboidratos , Humanos , Fragmentos Fab das Imunoglobulinas/metabolismo , Ligantes , Simulação de Dinâmica Molecular , Polissacarídeos/química , Polissacarídeos/metabolismo , Antígenos Embrionários Estágio-Específicos/química
2.
Biochem J ; 477(17): 3219-3235, 2020 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-32789497

RESUMO

Immunotherapy has been successful in treating many tumour types. The development of additional tumour-antigen binding monoclonal antibodies (mAbs) will help expand the range of immunotherapeutic targets. Lewis histo-blood group and related glycans are overexpressed on many carcinomas, including those of the colon, lung, breast, prostate and ovary, and can therefore be selectively targeted by mAbs. Here we examine the molecular and structural basis for recognition of extended Lea and Lex containing glycans by a chimeric mAb. Both the murine (FG88.2) IgG3 and a chimeric (ch88.2) IgG1 mAb variants showed reactivity to colorectal cancer cells leading to significantly reduced cell viability. We determined the X-ray structure of the unliganded ch88.2 fragment antigen-binding (Fab) containing two Fabs in the unit cell. A combination of molecular docking, glycan grafting and molecular dynamics simulations predicts two distinct subsites for recognition of Lea and Lex trisaccharides. While light chain residues were exclusively used for Lea binding, recognition of Lex involved both light and heavy chain residues. An extended groove is predicted to accommodate the Lea-Lex hexasaccharide with adjoining subsites for each trisaccharide. The molecular and structural details of the ch88.2 mAb presented here provide insight into its cross-reactivity for various Lea and Lex containing glycans. Furthermore, the predicted interactions with extended epitopes likely explains the selectivity of this antibody for targeting Lewis-positive tumours.


Assuntos
Anticorpos Monoclonais Murinos , Antineoplásicos Imunológicos , Fragmentos Fab das Imunoglobulinas , Antígenos do Grupo Sanguíneo de Lewis , Antígenos CD15 , Simulação de Acoplamento Molecular , Neoplasias , Oligossacarídeos , Animais , Anticorpos Monoclonais Murinos/química , Anticorpos Monoclonais Murinos/imunologia , Antineoplásicos Imunológicos/química , Antineoplásicos Imunológicos/imunologia , Linhagem Celular Tumoral , Humanos , Fragmentos Fab das Imunoglobulinas/química , Fragmentos Fab das Imunoglobulinas/imunologia , Antígenos do Grupo Sanguíneo de Lewis/química , Antígenos do Grupo Sanguíneo de Lewis/imunologia , Antígenos CD15/química , Antígenos CD15/imunologia , Camundongos , Neoplasias/química , Neoplasias/imunologia , Oligossacarídeos/química , Oligossacarídeos/imunologia
3.
Exp Parasitol ; 130(4): 384-93, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22343045

RESUMO

Murine immunoglobulin G (IgG) plays an important role in mediating protective immune responses to malaria. We still know relatively little about which IgG subclasses protect against this disease in mouse models, although IgG2a and IgG2b are considered to be the most potent and dominate in successful passive transfer experiments in rodent malarias. To explore the mechanism(s) by which the different mouse IgG subclasses may mediate a protective effect, we generated mouse IgG1, IgG2a, IgG2b and IgG3 specific for the C-terminal 19-kDa region of Plasmodium falciparum merozoite surface protein 1 (PfMSP1(19)), and to the homologous antigen from Plasmodium yoelii (P. yoelii), both major targets of protective immune responses. This panel of eight IgGs bound antigen with an affinity comparable to that seen for their epitope-matched parental monoclonal antibodies (mAbs) from which they were derived, although for reasons of yield, we were only able to explore the function of mouse IgG1 recognizing PfMSP1(19) in detail, both in vitro and in vivo. Murine IgG1 was as effective as the parental human IgG from which it was derived at inducing NADPH-mediated oxidative bursts and degranulation from neutrophils. Despite showing efficacy in in vitro functional assays with neutrophils, the mouse IgG1 failed to protect against parasite challenge in vivo. The lack of protection afforded by MSP1(19)-specific IgG1 against parasite challenge in wild type mice suggests that this Ab class does not play a major role in the control of infection with mouse malaria in the Plasmodium berghei transgenic model.


Assuntos
Anticorpos Antiprotozoários/biossíntese , Especificidade de Anticorpos , Imunoglobulina G/biossíntese , Proteína 1 de Superfície de Merozoito/imunologia , Plasmodium falciparum/imunologia , Plasmodium yoelii/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Antiprotozoários/imunologia , Western Blotting , Linhagem Celular , Ensaio de Imunoadsorção Enzimática , Epitopos/imunologia , Imunofluorescência , Imunização Passiva , Imunoglobulina G/classificação , Imunoglobulina G/imunologia , Malária/prevenção & controle , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Plasmodium berghei/imunologia , Alinhamento de Sequência , Organismos Livres de Patógenos Específicos
4.
BMC Biotechnol ; 11: 77, 2011 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-21781305

RESUMO

BACKGROUND: Human immunoglobulin G (IgG) plays an important role in mediating protective immune responses to malaria. Although human serum immunoglobulin A (IgA) is the second most abundant class of antibody in the circulation, its contribution, if any, to protective responses against malaria is not clear. RESULTS: To explore the mechanism(s) by which IgA may mediate a protective effect, we generated fully human IgA specific for the C-terminal 19-kDa region of Plasmodium falciparum merozoite surface protein 1 (PfMSP1 19), a major target of protective immune responses. This novel human IgA bound antigen with an affinity comparable to that seen for an epitope-matched protective human IgG1. Furthermore, the human IgA induced significantly higher NADPH-mediated oxidative bursts and degranulation from human neutrophils than the epitope-matched human IgG1 from which it was derived. Despite showing efficacy in in vitro functional assays, the human IgA failed to protect against parasite challenge in vivo in mice transgenic for the human Fcα receptor (FcαRI/CD89). A minority of the animals treated with IgA, irrespective of FcαRI expression, showed elevated serum TNF-α levels and concomitant mouse anti-human antibody (MAHA) responses. CONCLUSIONS: The lack of protection afforded by MSP1 19-specific IgA against parasite challenge in mice transgenic for human FcαRI suggests that this antibody class does not play a major role in control of infection. However, we cannot exclude the possibility that protective capacity may have been compromised in this model due to rapid clearance and inappropriate bio-distribution of IgA, and differences in FcαRI expression profile between humans and transgenic mice.


Assuntos
Anticorpos Antiprotozoários/imunologia , Imunoglobulina A/imunologia , Proteína 1 de Superfície de Merozoito/imunologia , Plasmodium falciparum/imunologia , Proteínas Recombinantes/imunologia , Animais , Antígenos CD/genética , Modelos Animais de Doenças , Humanos , Imunização Passiva , Imunoglobulina G/metabolismo , Malária/imunologia , Malária/prevenção & controle , Camundongos , Camundongos Transgênicos , NADPH Oxidases/metabolismo , Plasmodium berghei , Receptores Fc/genética , Fator de Necrose Tumoral alfa/metabolismo
5.
Eur J Immunol ; 39(4): 1147-56, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19266484

RESUMO

Here we unravel the structural features of human IgM and IgA that govern their interaction with the human Fcalpha/mu receptor (hFcalpha/muR). Ligand polymerization status was crucial for the interaction, because hFcalpha/muR binding did not occur with monomeric Ab of either class. hFcalpha/muR bound IgM with an affinity in the nanomolar range, whereas the affinity for dimeric IgA (dIgA) was tenfold lower. Panels of mutant IgM and dIgA were used to identify regions critical for hFcalpha/muR binding. IgM binding required contributions from both Cmu3 and Cmu4 Fc domains, whereas for dIgA, an exposed loop in the Calpha3 domain was crucial. This loop, comprising residues Pro440-Phe443, lies at the Fc domain interface and has been implicated in the binding of host receptors FcalphaRI and polymeric Ig receptor (pIgR), as well as IgA-binding proteins produced by certain pathogenic bacteria. Substitutions within the Pro440-Phe443 loop resulted in loss of hFcalpha/muR binding. Furthermore, secretory component (SC, the extracellular portion of pIgR) and bacterial IgA-binding proteins were shown to inhibit the dIgA-hFcalpha/muR interaction. Therefore, we have identified a motif in the IgA-Fc inter-domain region critical for hFcalpha/muR interaction, and highlighted the multi-functional nature of a key site for protein-protein interaction at the IgA Fc domain interface.


Assuntos
Afinidade de Anticorpos , Imunoglobulina A/química , Imunoglobulina M/química , Receptores Fc/imunologia , Motivos de Aminoácidos , Substituição de Aminoácidos , Animais , Afinidade de Anticorpos/genética , Afinidade de Anticorpos/imunologia , Células COS , Chlorocebus aethiops , Humanos , Imunoglobulina A/genética , Imunoglobulina A/imunologia , Imunoglobulina M/genética , Imunoglobulina M/imunologia , Proteínas Mutantes/imunologia , Mutação , Domínios e Motivos de Interação entre Proteínas/imunologia , Multimerização Proteica , Estrutura Terciária de Proteína , Receptores Fc/genética
6.
J Immunol ; 181(3): 1988-2000, 2008 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-18641336

RESUMO

The binding of nonspecific human IgM to the surface of infected erythrocytes is important in rosetting, a major virulence factor in the pathogenesis of severe malaria due to Plasmodium falciparum, and IgM binding has also been implicated in placental malaria. Herein we have identified the IgM-binding parasite ligand from a virulent P. falciparum strain as PfEMP1 (TM284var1 variant), and localized the region within this PfEMP1 variant that binds IgM (DBL4beta domain). We have used this parasite IgM-binding protein to investigate the interaction with human IgM. Interaction studies with domain-swapped Abs, IgM mutants, and anti-IgM mAbs showed that PfEMP1 binds to the Fc portion of the human IgM H chain and requires the IgM Cmu4 domain. Polymerization of IgM was shown to be crucial for the interaction because PfEMP1 binding did not occur with mutant monomeric IgM molecules. These results with PfEMP1 protein have physiological relevance because infected erythrocytes from strain TM284 and four other IgM-binding P. falciparum strains showed analogous results to those seen with the DBL4beta domain. Detailed investigation of the PfEMP1 binding site on IgM showed that some of the critical amino acids in the IgM Cmu4 domain are equivalent to those regions of IgG and IgA recognized by Fc-binding proteins from bacteria, suggesting that this region of Ig molecules may be of major functional significance in host-microbe interactions. We have therefore shown that PfEMP1 is an Fc-binding protein of malaria parasites specific for polymeric human IgM, and that it shows functional similarities with Fc-binding proteins from pathogenic bacteria.


Assuntos
Imunoglobulina M/imunologia , Plasmodium falciparum/imunologia , Proteínas de Protozoários/imunologia , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Sítios de Ligação , Células COS , Chlorocebus aethiops , Eritrócitos/imunologia , Eritrócitos/metabolismo , Humanos , Fragmentos Fc das Imunoglobulinas/imunologia , Imunoglobulina M/química , Ligantes , Modelos Moleculares , Dados de Sequência Molecular , Ligação Proteica , Estrutura Quaternária de Proteína , Proteínas de Protozoários/genética , Proteínas de Protozoários/metabolismo , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Solubilidade
7.
Mol Cancer Ther ; 19(3): 790-801, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31871270

RESUMO

Tumor glycans constitute attractive targets for therapeutic antibodies. The sialylated glycocalyx plays a prominent role in cancer progression and immune evasion. Here, we describe the characterization of the mAb, FG129, which targets tumor-associated sialylated glycan, and demonstrate its potential for multimodal cancer therapy. FG129, obtained through BALB/c mouse immunizations with liposomes containing membrane glycan extracts from the colorectal cancer cell line LS180, is an mIgG1κ that targets sialyl-di-Lewisa-containing glycoproteins. FG129, as well as its chimeric human IgG1 variant, CH129, binds with nanomolar functional affinity to a range of colorectal, pancreatic, and gastric cancer cell lines. FG129 targets 74% (135/182) of pancreatic, 50% (46/92) of gastric, 36% (100/281) of colorectal, 27% (89/327) of ovarian, and 21% (42/201) of non-small cell lung cancers, by IHC. In our pancreatic cancer cohort, high FG129 glyco-epitope expression was significantly associated with poor prognosis (P = 0.004). Crucially, the glyco-epitope displays limited normal tissue distribution, with FG129 binding weakly to a small percentage of cells within gallbladder, ileum, liver, esophagus, pancreas, and thyroid tissues. Owing to glyco-epitope internalization, we validated payload delivery by CH129 through monomethyl auristatin E (MMAE) or maytansinoid (DM1 and DM4) conjugation. All three CH129 drug conjugates killed high-binding colorectal and pancreatic cancer cell lines with (sub)nanomolar potency, coinciding with significant in vivo xenograft tumor control by CH129-vcMMAE. CH129, with its restricted normal tissue distribution, avid tumor binding, and efficient payload delivery, is a promising candidate for the treatment of sialyl-di-Lewisa-expressing solid tumors, as an antibody-drug conjugate or as an alternative cancer immunotherapy modality.


Assuntos
Anticorpos Monoclonais/farmacologia , Antígenos de Neoplasias/imunologia , Neoplasias do Colo/terapia , Glicoproteínas/imunologia , Imunoglobulina G/imunologia , Imunoterapia/métodos , Antígeno Sialil Lewis X/imunologia , Animais , Anticorpos Monoclonais/imunologia , Apoptose , Proliferação de Células , Neoplasias do Colo/imunologia , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias/imunologia , Neoplasias/metabolismo , Neoplasias/patologia , Neoplasias/terapia , Polissacarídeos/imunologia , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Cancer Res ; 80(16): 3399-3412, 2020 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-32532823

RESUMO

Murine IgG3 glycan-targeting mAb often induces direct cell killing in the absence of immune effector cells or complement via a proinflammatory mechanism resembling oncotic necrosis. This cancer cell killing is due to noncovalent association between Fc regions of neighboring antibodies, resulting in enhanced avidity. Human isotypes do not contain the residues underlying this cooperative binding mode; consequently, the direct cell killing of mouse IgG3 mAb is lost upon chimerization or humanization. Using the Lewisa/c/x -targeting 88mAb, we identified the murine IgG3 residues underlying the direct cell killing and increased avidity via a series of constant region shuffling and subdomain swapping approaches to create improved ("i") chimeric mAb with enhanced tumor killing in vitro and in vivo. Constant region shuffling identified a major CH3 and a minor CH2 contribution, which was further mapped to discontinuous regions among residues 286-306 and 339-378 that, when introduced in 88hIgG1, recapitulated the direct cell killing and avidity of 88mIgG3. Of greater interest was the creation of a sialyl-di-Lewisa-targeting i129G1 mAb via introduction of these selected residues into 129hIgG1, converting it into a direct cell killing mAb with enhanced avidity and significant in vivo tumor control. The human iG1 mAb, termed Avidimabs, retained effector functions, paving the way for the proinflammatory direct cell killing to promote antibody-dependent cellular cytotoxicity and complement-dependent cytotoxicity through relief of immunosuppression. Ultimately, Fc engineering of human glycan-targeting IgG1 mAb confers proinflammatory direct cell killing and enhanced avidity, an approach that could be used to improve the avidity of other mAb with therapeutic potential. SIGNIFICANCE: Fc engineering enhances avidity and direct cell killing of cancer-targeting anti-glycan antibodies to create superior clinical candidates for cancer immunotherapy.


Assuntos
Anticorpos Monoclonais/imunologia , Afinidade de Anticorpos/imunologia , Morte Celular/imunologia , Neoplasias Colorretais/terapia , Fragmentos Fc das Imunoglobulinas/imunologia , Imunoglobulina G/imunologia , Polissacarídeos/imunologia , Animais , Citotoxicidade Celular Dependente de Anticorpos , Linhagem Celular Tumoral , Neoplasias Colorretais/imunologia , Proteínas do Sistema Complemento , Feminino , Engenharia Genética , Humanos , Imunoterapia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Distribuição Aleatória
9.
Infect Immun ; 77(12): 5659-67, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19805526

RESUMO

Antigen-specific antibodies (Abs) to the 19-kDa carboxy-terminal region of Plasmodium falciparum merozoite surface protein 1 (MSP1(19)) play an important role in protective immunity to malaria. Mouse monoclonal Abs (MAbs) 12.10 and 12.8 recognizing MSP1(19) can inhibit red cell invasion by interfering with MSP1 processing on the merozoite surface. We show here that this ability is dependent on the intact Ab since Fab and F(ab')(2) fragments derived from MAb 12.10, although capable of binding MSP1 with high affinity and competing with the intact antibody for binding to MSP1, were unable to inhibit erythrocyte invasion or MSP1 processing. The DNA sequences of the variable (V) regions of both MAbs 12.8 and 12.10 were obtained, and partial amino acid sequences of the same regions were confirmed by mass spectrometry. Human chimeric Abs constructed by using these sequences, which combine the original mouse V regions with human gamma1 and gamma3 constant regions, retain the ability to bind to both parasites and recombinant MSP1(19), and both chimeric human immunoglobulin G1s (IgG1s) were at least as good at inhibiting erythrocyte invasion as the parental murine MAbs 12.8 and 12.10. Furthermore, the human chimeric Abs of the IgG1 class (but not the corresponding human IgG3), induced significant NADPH-mediated oxidative bursts and degranulation from human neutrophils. These chimeric human Abs will enable investigators to examine the role of human Fcgamma receptors in immunity to malaria using a transgenic parasite and mouse model and may prove useful in humans for neutralizing parasites as an adjunct to antimalarial drug therapy.


Assuntos
Anticorpos Antiprotozoários/imunologia , Eritrócitos/parasitologia , Imunoglobulina G/imunologia , Proteína 1 de Superfície de Merozoito/antagonistas & inibidores , Plasmodium falciparum/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/metabolismo , Anticorpos Antiprotozoários/metabolismo , Sequência de Bases , Humanos , Fragmentos Fab das Imunoglobulinas/metabolismo , Proteína 1 de Superfície de Merozoito/imunologia , Proteína 1 de Superfície de Merozoito/metabolismo , Camundongos , Dados de Sequência Molecular , Neutrófilos/imunologia , Ligação Proteica , RNA , Explosão Respiratória
10.
PLoS Pathog ; 3(5): e72, 2007 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-17511516

RESUMO

The success of passive immunization suggests that antibody-based therapies will be effective at controlling malaria. We describe the development of fully human antibodies specific for Plasmodium falciparum by antibody repertoire cloning from phage display libraries generated from immune Gambian adults. Although these novel reagents bind with strong affinity to malaria parasites, it remains unclear if in vitro assays are predictive of functional immunity in humans, due to the lack of suitable animal models permissive for P. falciparum. A potentially useful solution described herein allows the antimalarial efficacy of human antibodies to be determined using rodent malaria parasites transgenic for P. falciparum antigens in mice also transgenic for human Fc-receptors. These human IgG1s cured animals of an otherwise lethal malaria infection, and protection was crucially dependent on human FcgammaRI. This important finding documents the capacity of FcgammaRI to mediate potent antimalaria immunity and supports the development of FcgammaRI-directed therapy for human malaria.


Assuntos
Malária/imunologia , Receptores Fc , Animais , Anticorpos Antiprotozoários/biossíntese , Anticorpos Antiprotozoários/isolamento & purificação , Anticorpos Antiprotozoários/uso terapêutico , Antígenos de Protozoários , Antimaláricos , Mapeamento de Epitopos , Humanos , Imunoglobulina G , Malária/terapia , Camundongos , Camundongos Transgênicos , Plasmodium falciparum/imunologia
11.
Mol Immunol ; 45(2): 567-74, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17631966

RESUMO

Parasitic infections, including schistosomiasis, are associated with high titres of specific and non-specific IgE antibody, and many reports show an in vitro role for IgE in parasite killing. Despite an active immune response, schistosomes survive for long periods in the human bloodstream, implying that the parasite is able to overcome or evade the IgE response mounted against it. One such mechanism is through cleavage of IgE into non-functional fragments by potent parasite derived enzymes. Using domain swap antibodies, recombinant Fcepsilon, and C-terminally tagged Cepsilon4 domains, we have narrowed down the principal cleavage sites to the Cepsilon2/Cepsilon3 and Cepsilon3/Cepsilon4 interdomain region of the IgE-Fc. Two serine proteases, one chymotrypsin-like and the second trypsin-like, have been proposed to be involved. Inhibition assays using selective inhibitors confirmed that both proteases contribute to Fc cleavage, although the chymotrypsin-like enzyme makes the greater contribution. Protein sequencing of IgE fragments cleaved by highly pure preparations of the chymotrypsin-like enzyme revealed that cleavage also occurred post Lys residues within kappa light chain dimers (LELK/GA). Related sequences are found in myosin, thrombospondin, collagen and actin-related proteins; macromolecules present in the skin and through which cercariae must penetrate to initiate an infection. Chemical knockout experiments using specific inhibitors and chromogenic substrates allowed us to show that the trypsin-like enzyme was responsible for light chain cleavage. The finding that pathogenic proteases can cleave the Fc of IgE may provide a useful biochemical tool for the further analysis of IgE structure. Indeed, the finding may raise new possibilities for treatment of IgE-mediated allergic reactions mediated through Fcepsilon-receptors.


Assuntos
Imunoglobulina E/química , Imunoglobulina E/metabolismo , Elastase Pancreática/metabolismo , Schistosoma mansoni/enzimologia , Solventes/metabolismo , Sequência de Aminoácidos , Animais , Anticorpos , Proteína de Bence Jones/metabolismo , Biologia Computacional , Dimerização , Inibidores Enzimáticos/farmacologia , Humanos , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Elastase Pancreática/isolamento & purificação , Estrutura Terciária de Proteína , Ratos , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Schistosoma mansoni/efeitos dos fármacos , Especificidade por Substrato/efeitos dos fármacos
12.
Invest Ophthalmol Vis Sci ; 47(10): 4316-22, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17003421

RESUMO

PURPOSE: Amniotic membrane (AM) transplantation is an accepted procedure in ocular surgery. However, little is known of the interdonor and intradonor variability within the membrane. In addition, the effects of the methods of processing, storage, and preoperative preparation on the membrane are not fully elucidated. The purpose of this study was to use TGF-beta as an example to investigate interdonor and intradonor variability and to determine the effect of "handling " on TGF-beta1 within fresh, processed and stored, and transplantation-ready AM (TRAM). METHODS: Seventeen human AMs, both fresh and handled, were analyzed for TGF-beta1 by real-time polymerase chain reaction, immunohistochemistry, SDS-PAGE, and Western blotting. RESULTS: TGF-beta1 was the highest normalized expressed isoform of TGF-beta in all samples, but it varied between membranes of different donors and at different sites within the same membrane. The highest concentration was noted in the spongy layer. Removal of the spongy layer successfully removed the bulk of TGF-beta1 from TRAM. Latency-associated protein (LAP) and a latent TGF-beta-binding protein (LTBP) were also detected. CONCLUSION: TGF-beta1 is present in various regulatory forms in the AM. A degree of intermembrane and intramembrane variation is modified by handling. Unless a standardized protocol is adopted that delivers a membrane with consistent constituents, clinical outcomes may vary and comparisons may be invalid.


Assuntos
Âmnio/metabolismo , Manejo de Espécimes , Doadores de Tecidos , Fator de Crescimento Transformador beta/metabolismo , Âmnio/transplante , Antígenos Virais/metabolismo , Western Blotting , Doenças da Córnea/cirurgia , Eletroforese em Gel de Poliacrilamida , Humanos , Imuno-Histoquímica , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Ligação a TGF-beta Latente , Proteínas Nucleares/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Crescimento Transformador beta1
13.
Invest Ophthalmol Vis Sci ; 46(4): 1379-85, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15790905

RESUMO

PURPOSE: Antimicrobial peptides are the eukaryotic analogues of antibiotics. In addition to their antimicrobial activity, these peptides can signal to host cells and are therefore intermediaries between the innate and adaptive immune systems. Results in a prior study showed that beta-defensins-1 and -2 are made by ocular surface epithelial cells. In the present study, a survey was made of antimicrobial peptide expression, including 17 previously described members of the beta-defensin family, at the surface of the human eye. METHODS: Total RNA was obtained from 43 fresh and cultured corneal and conjunctival samples, including 9 samples from patients with clinical infections. The expression of 21 antimicrobial peptides was determined using reverse transcription-PCR. Where detected, relative expression was quantitated using real-time PCR. RESULTS: Expression of 7 of the 21 antimicrobial peptides investigated, beta-defensin-1 to -4, liver expressed antimicrobial peptide (LEAP)-1 and -2, and LL37/cathelicidin, were detected frequently in samples of ocular surface epithelia. Distinct but overlapping profiles of expression were detected in cornea and conjunctiva, with expression of beta-defensin-3 and -4 and LEAP1 and -2 most common in cultured corneal epithelia. Expression of beta-defensin-3 was detected in a greater percentage of corneal and conjunctival samples with infection. CONCLUSIONS: Together with known lacrimal antimicrobial activities, these results extend the knowledge of antimicrobial activity at an important mucosal site, the ocular surface, allowing synergistic interactions to be investigated. The findings has significant implications both for the understanding of the normal homeostasis of mucosal surfaces and for antimicrobial and anti-inflammatory therapies.


Assuntos
Antibacterianos/metabolismo , Peptídeos Catiônicos Antimicrobianos/metabolismo , Proteínas Sanguíneas/metabolismo , Túnica Conjuntiva/metabolismo , Córnea/metabolismo , Defensinas/metabolismo , Proteínas/metabolismo , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Peptídeos Catiônicos Antimicrobianos/genética , Proteínas Sanguíneas/genética , Catelicidinas , Defensinas/genética , Células Epiteliais/metabolismo , Feminino , Hepcidinas , Humanos , Masculino , Pessoa de Meia-Idade , Proteínas/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
14.
Clin Cancer Res ; 21(13): 2963-74, 2015 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-25779947

RESUMO

PURPOSE: To produce antitumor monoclonal antibodies (mAbs) targeting glycans as they are aberrantly expressed in tumors and are coaccessory molecules for key survival pathways. EXPERIMENTAL DESIGN: Two mAbs (FG88.2 and FG88.7) recognizing novel tumor-associated Lewis (Le) glycans were produced by immunizations with plasma membrane lipid extracts of the COLO205 cell line. RESULTS: Glycan array analysis showed that both mAbs bound Le(c)Le(x), di-Le(a), and Le(a)Le(x), as well as Le(a)-containing glycans. These glycans are expressed on both lipids and proteins. Both mAbs showed strong tumor reactivity, binding to 71% (147 of 208) of colorectal, 81% (155 of 192) of pancreatic, 54% (52 of 96) of gastric, 23% (62 of 274) of non-small cell lung, and 31% (66 of 217) of ovarian tumor tissue in combination with a restricted normal tissue distribution. In colorectal cancer, high FG88 glyco-epitope expression was significantly associated with poor survival. The mAbs demonstrated excellent antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC), in addition to direct tumor cell killing via a caspase-independent mechanism. Scanning electron microscopy revealed antibody-induced pore formation. In addition, the mAbs internalized, colocalized with lysosomes, and delivered saporin that killed cells with subnanomolar potency. In vivo, the mAbs demonstrated potent antitumor efficacy in a metastatic colorectal tumor model, leading to significant long-term survival. CONCLUSIONS: The mAbs direct and immune-assisted tumor cell killing, pan-tumor reactivity, and potent in vivo antitumor efficacy indicate their potential as therapeutic agents for the treatment of multiple solid tumors. In addition, internalization of saporin conjugates and associated tumor cell killing suggests their potential as antibody drug carriers.


Assuntos
Anticorpos Monoclonais Murinos/farmacologia , Citotoxicidade Celular Dependente de Anticorpos , Antineoplásicos/farmacologia , Neoplasias Hepáticas Experimentais/tratamento farmacológico , Animais , Anticorpos Monoclonais Murinos/metabolismo , Antineoplásicos/metabolismo , Biomarcadores Tumorais/metabolismo , Linhagem Celular Tumoral , Humanos , Antígenos do Grupo Sanguíneo de Lewis/imunologia , Neoplasias Hepáticas Experimentais/secundário , Masculino , Camundongos Endogâmicos BALB C , Camundongos Nus , Polissacarídeos/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto
15.
Invest Ophthalmol Vis Sci ; 44(11): 4689-92, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14578387

RESUMO

PURPOSE: To investigate the expression of CD34, a hematopoietic stem cell marker and an adhesion molecule, and its ligand L-selectin in the human cornea. METHODS: Seventeen normal adult human corneal specimens were studied by immunohistochemistry using a panel of monoclonal antibodies against all three classes of the hematopoietic stem cell marker CD34 and its ligand L-selectin. An additional six corneal specimens were used for protein extraction and analysis by Western blotting, using the CD34 and L-selectin antibodies. PCR was used to determine expression of mRNA for CD34 and L-selectin in the corneal specimens. RESULTS: Only corneal keratocytes showed positive immunostaining for all three classes of CD34. Western blotting confirmed the expression of CD34 by these cells and mRNA expression for CD34 in the corneal stroma was demonstrated by PCR. For L-selectin, positive staining around keratocytes was noted on immunohistochemistry but L-selectin could not be detected either by Western blotting or PCR. CONCLUSIONS: Normal human corneal keratocytes express all three classes of CD34. The expression of this adhesion molecule on corneal keratocytes suggests that it may have a role in keeping the keratocytes anchored in their microniche, between the collagen lamellae. The positive staining for L-selectin found by immunohistochemistry but not by Western blotting or PCR would indicate the presence of either another ligand from the selectin family or a cross-reactive epitope on corneal keratocytes.


Assuntos
Antígenos CD34/metabolismo , Substância Própria/metabolismo , Fibroblastos/metabolismo , Selectina L/metabolismo , Anticorpos Monoclonais , Antígenos CD34/genética , Western Blotting , Humanos , Técnicas Imunoenzimáticas , Selectina L/genética , Reação em Cadeia da Polimerase , RNA Mensageiro/metabolismo
16.
Sci Rep ; 1: 124, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22355641

RESUMO

The success of Fc-fusion bio-therapeutics has spurred the development of other Fc-fusion products for treating and/or vaccinating against a range of diseases. We describe a method to modulate their function by converting them into well-defined stable polymers. This strategy resulted in cylindrical hexameric structures revealed by tapping mode atomic force microscopy (AFM). Polymeric Fc-fusions were significantly less immunogenic than their dimeric or monomeric counterparts, a result partly owing to their reduced ability to interact with critical Fc-receptors. However, in the absence of the fusion partner, polymeric IgG1-Fc molecules were capable of binding selectively to FcγRs, with significantly increased affinity owing to their increased valency, suggesting that these reagents may prove of immediate utility in the development of well-defined replacements for intravenous immunoglobulin (IVIG) therapy. Overall, these findings establish an effective IgG Fc-fusion based polymeric platform with which the therapeutic and vaccination applications of Fc-fusion immune-complexes can now be explored.


Assuntos
Fragmentos Fc das Imunoglobulinas/metabolismo , Animais , Linfócitos B/imunologia , Linfócitos B/metabolismo , Sequência de Bases , Terapia Biológica , Proteínas do Sistema Complemento/metabolismo , Primers do DNA/genética , Feminino , Antígenos de Histocompatibilidade Classe I/metabolismo , Humanos , Imunização , Fragmentos Fc das Imunoglobulinas/química , Fragmentos Fc das Imunoglobulinas/genética , Fragmentos Fc das Imunoglobulinas/uso terapêutico , Imunoglobulina G/química , Imunoglobulina G/genética , Imunoglobulina G/metabolismo , Malária/imunologia , Malária/parasitologia , Malária/terapia , Camundongos , Camundongos Endogâmicos BALB C , Modelos Moleculares , Plasmodium berghei , Ligação Proteica , Multimerização Proteica , Receptores Fc/metabolismo , Receptores de IgG/metabolismo , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes de Fusão/uso terapêutico
17.
Clin Sci (Lond) ; 110(1): 11-9, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16336201

RESUMO

Abs (antibodies) are complex glycoproteins that play a crucial role in protective immunity to malaria, but their effectiveness in mediating resistance can be enhanced by genetically engineered modifications that improve on nature. These Abs also aid investigation of immune mechanisms operating to control the disease and are valuable tools in developing neutralization assays for vaccine design. This review explores how this might be achieved.


Assuntos
Anticorpos Antiprotozoários/uso terapêutico , Imunização Passiva/métodos , Malária/terapia , Receptores Fc/imunologia , Animais , Anticorpos Antiprotozoários/imunologia , Humanos , Malária/imunologia , Vacinas Antimaláricas/imunologia , Plasmodium/imunologia , Polimorfismo Genético , Receptores Fc/genética , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/uso terapêutico
18.
J Proteome Res ; 5(9): 2226-35, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16944934

RESUMO

Amniotic membrane is commonly exploited in several surgical procedures. Despite a freeze preservation period, it is reported to retain wound healing, anti-angiogenic, antiinflammatory and anti-scarring properties; however, little is known about the active protein content. 2-DE analysis of transplant-ready amniotic membrane (TRAM) was performed. The effects of preservation and processing on amnion proteome were investigated, and the major proteins in the TRAM characterized using mass spectrometry and immunoblotting. This identified a spectrum of proteins including thrombospondin, mimecan, BIG-H3, and integrin alpha 6. Preservation compromises cellular viability resulting in selective elution of soluble cellular proteins, leaving behind extracellular matrix-associated and cell structural proteins. A number of key architectural proteins common to the architecture of the ocular surface were demonstrated in AM, which are involved in homeostasis and wound healing. Handling procedures alter the protein composition of amniotic membrane prepared for transplantation. Without standardization, there will be inter-membrane variation, which may compromise the desired therapeutic effect of transplant ready amniotic membrane.


Assuntos
Âmnio/química , Proteínas/análise , Manejo de Espécimes/métodos , Âmnio/transplante , Eletroforese em Gel Bidimensional , Traumatismos Oculares/terapia , Humanos , Immunoblotting , Espectrometria de Massas , Proteômica/métodos
19.
Proteomics ; 5(7): 1967-79, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15816006

RESUMO

The protein analysis of structural tissues is typically highly problematic. Amniotic membrane displays unique wound healing and anti-scarring properties; however, little is known concerning its active protein content. The structural nature of amniotic membrane necessitated development and extensive optimisation of the entire two-dimensional (2-D) workflow. Proteins were extracted using powerful solubilisation buffers and analysis carried out using 2-D electrophoresis followed by mass spectrometry (MS) identification. Preservation and processing resulted in prefractionation of soluble from structural and membrane-associated proteins. Enhanced protein solubility was achieved by cysteine blocking using both N,N-dimethylacrylamide (DMA) alkylation and bis(2-hydroxyethyl) disulphide (HED); an alternative procedure for the effective application of HED is demonstrated. The benefits of precipitation and cup-loading versus in-gel rehydration were also assessed, with procedures for the employment of HED with the latter described. Following optimisation, a representative sample 21 proteins were identified from amniotic membrane using MS verify procedures were MS-compatible. Our results demonstrate that techniques for the reproducible separation of proteins from a proteinaceous structural tissue have been optimised. Briefly, proteins are extracted using a thiourea/urea extraction buffer containing carrier ampholytes, dithiothreitol (DTT), and 3-(cyclohexylamino)-1-propanesulfonic acid (CHAPS). After DMA alkylation, proteins were precipitated (using the 2-D clean-up kit from Amersham Biosciences) and resolubilised in extraction buffer containing a lower concentration of DTT. Samples were either cup-loaded onto rehydrated HED-containing strips or rebuffered into HED-containing buffer followed by in-gel rehydration.


Assuntos
Eletroforese em Gel Bidimensional , Análise Serial de Proteínas , Proteínas/química , Proteínas/isolamento & purificação , Âmnio/química , Âmnio/metabolismo , Cisteína , Feminino , Humanos , Especificidade de Órgãos , Análise Serial de Proteínas/métodos , Proteínas/metabolismo , Sulfetos
20.
Wound Repair Regen ; 13(3): 295-302, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15953049

RESUMO

Glaucoma drainage surgery in diabetic patients is associated with a relatively poor prognosis due to increased scarring at the site of surgery, secondary to increased proliferation of human Tenon's capsule fibroblasts (hTCF). This is in marked contrast to diabetic wound healing at other sites, where it is generally impaired. The aim of this study was to determine why diabetics show an increased ocular scarring response in comparison to that found at other sites. Under normoglycemic conditions, hTCF isolated from diabetics showed a mean reduction in short-term proliferation (95% CI) of 45 +/- 12% compared with normal controls (p < 0.001). Under hyperglycemic conditions, proliferation of diabetic hTCF was reduced by 21 +/- 11% (p < 0.01) compared with nondiabetic controls. When exposed to transforming growth factor-beta2 (1-10,000 pg/ml) and platelet-derived growth factor-BB (0.5-500 ng/ml) under both normo- and hyperglycemic conditions, there was a dose-related increase in proliferation of both diabetic and nondiabetic controls. There was no significant difference in response to cytokine stimulation between the two groups at any of the cytokine concentrations used. Western blot analysis did not show any apparent difference in the expression of platelet-derived growth factor receptor alpha, mitogen-activated protein kinase/ERK2, or transforming growth factor-beta receptor II to account for the reduced proliferation of diabetic hTCF. These results suggest that hTCF behave in a manner similar to fibroblasts from other nonocular sites and that the increased proliferation and scarring response found in vivo may be secondary to the previously noted elevated cytokine concentrations in the aqueous and vitreous of diabetics.


Assuntos
Proliferação de Células/efeitos dos fármacos , Cicatriz/fisiopatologia , Diabetes Mellitus/fisiopatologia , Cicatrização/efeitos dos fármacos , Adulto , Idoso , Células Cultivadas , Cicatriz/etiologia , Feminino , Fibroblastos , Humanos , Hiperglicemia/complicações , Hiperglicemia/fisiopatologia , Masculino , Pessoa de Meia-Idade , Proteínas Proto-Oncogênicas c-sis/farmacologia , Fator de Crescimento Transformador beta/farmacologia , Fator de Crescimento Transformador beta2 , Cicatrização/fisiologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa