Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
MAbs ; 12(1): 1831880, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33183151

RESUMO

CXCR1 and CXCR2 signaling play a critical role in neutrophil migration, angiogenesis, and tumorigenesis and are therefore an attractive signaling axis to target in a variety of indications. In human, a total of seven chemokines signal through these receptors and comprise the ELR+CXC chemokine family, so named because of the conserved ELRCXC N-terminal motif. To fully antagonize CXCR1 and CXCR2 signaling, an effective therapeutic should block either both receptors or all seven ligands, yet neither approach has been fully realized clinically. In this work, we describe the generation and characterization of LY3041658, a humanized monoclonal antibody that binds and neutralizes all seven human and cynomolgus monkey ELR+CXC chemokines and three of five mouse and rat ELR+CXC chemokines with high affinity. LY3041658 is able to block ELR+CXC chemokine-induced Ca2+ mobilization, CXCR2 internalization, and chemotaxis in vitro as well as neutrophil mobilization in vivo without affecting other neutrophil functions. In addition to the in vitro and in vivo activity, we characterized the epitope and structural basis for binding in detail through alanine scanning, crystallography, and mutagenesis. Together, these data provide a robust preclinical characterization of LY3041658 for which the efficacy and safety is being evaluated in human clinical trials for neutrophilic skin diseases.


Assuntos
Anticorpos Monoclonais/imunologia , Anticorpos Neutralizantes/imunologia , Receptores de Interleucina-8A/antagonistas & inibidores , Receptores de Interleucina-8B/antagonistas & inibidores , Animais , Anticorpos Monoclonais/química , Anticorpos Neutralizantes/química , Afinidade de Anticorpos , Quimiotaxia de Leucócito/imunologia , Humanos , Macaca fascicularis , Camundongos , Neutrófilos/imunologia , Ratos
2.
Biopharm Drug Dispos ; 41(3): 111-125, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-32080869

RESUMO

The immunogenicity of biotherapeutics presents a major challenge during the clinical development of new protein drugs including monoclonal antibodies. To address this, multiple humanization and de-immunization techniques that employ in silico algorithms and in vitro test systems have been proposed and implemented. However, the success of these approaches has been variable and to date, the ability of these techniques to predict immunogenicity has not been systematically tested in humans or other primates. This study tested whether antibody humanization and de-immunization strategies reduce the risk of anti-drug antibody (ADA) development using cynomolgus macaque as a surrogate for human. First human-cyno chimeric antibodies were constructed by grafting the variable domains of the adalimumab and golimumab monoclonal antibodies onto cynomolgus macaque IgG1 and Igκ constant domains followed by framework germlining to cyno to reduce the xenogenic content. Next, B and T cell epitopes and aggregation-prone regions were identified using common in silico methods to select domains with an ADA risk for additional modification. The resultant engineered antibodies had a comparable affinity for TNFα, demonstrated similar biophysical properties, and exhibited significantly reduced ADA levels in cynomolgus macaque compared with the parental antibodies, with a corresponding improvement in the pharmacokinetic profile. Notably, plasma concentrations of the engineered antibodies were quantifiable through 504 hours (chimeric) and 840 hours (germlined/de-immunized), compared with only 336 hours (adalimumab) or 336-672 hours (golimumab). The results point to the significant value in the investment in these engineering strategies as an important guide for monoclonal antibody optimization that can contribute to improved clinical outcomes.


Assuntos
Adalimumab/imunologia , Anticorpos Monoclonais/imunologia , Imunoglobulina G/imunologia , Animais , Feminino , Humanos , Imunização , Macaca fascicularis , Masculino , Fator de Necrose Tumoral alfa/imunologia
3.
Biotechnol J ; 14(3): e1800007, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29802766

RESUMO

There is a rapidly growing reinvigoration of the investigation of small proteins, cyclic peptides, and mAb derived domains as biotherapies. The drugability of these structures are challenged by fast peripheral clearance properties that can reduce their potential to be realized as medicines. Engineering strategies have been of limited value because mechanistically the half-life benefit is manifested by increasing the molecular weight and/or the hydrodyanimc radius which slows the molecule's renal elimination, but can result in the inherent loss of activity and target accessibility. The present work evaluated an alternative approach using smaller peptide sequences which bind to the neonatal Fc receptor (FcRn). Results revealed, small linear and cyclic FcRn binding peptides (FcRnBPs) fused to a combination of the N- and C-termini of a Fab can significantly improve the pharmacokinetics of the protein in cynomolgus monkeys relative to the parental Fab. The linear and cyclic conformations, as well as, the number of FcRnBPs fused to the Fab both influence the clearance and the extent of pharmacokinetic benefit. FcRnBP fusion protein kinetics were also affected by a combination of post-translation modifications and non-specific binding properties. The results in this report lay some foundation in fostering the advent of newer technologies toward successfully improving the pharmacokinetics of proteins, peptides, and mAb-derived domains. Additional work in the integration of a variety of factors including the intended site of action, tissue disposition, metabolism, toxicity and pharmacokinetic, and pharmacodynamics relationship of the intended therapeutic modality are key areas for advancement of these approaches.


Assuntos
Antígenos de Histocompatibilidade Classe I/metabolismo , Fragmentos Fab das Imunoglobulinas/metabolismo , Peptídeos/metabolismo , Receptores Fc/metabolismo , Animais , Anticorpos Monoclonais/metabolismo , Células CACO-2 , Linhagem Celular , Linhagem Celular Tumoral , Células HEK293 , Meia-Vida , Humanos , Cinética , Macaca fascicularis , Masculino , Ligação Proteica/fisiologia
4.
MAbs ; 10(6): 913-921, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29953319

RESUMO

Immunomodulatory monoclonal IgG1 antibodies developed for cancer and autoimmune disease have an inherent risk of systemic release of pro-inflammatory cytokines. In vitro cytokine release assays are currently used to predict cytokine release syndrome (CRS) risk, but the validation of these preclinical tools suffers from the limited number of characterized CRS-inducing IgG1 antibodies and the poor understanding of the mechanisms regulating cytokine release. Here, we incubated human whole blood from naïve healthy volunteers with four monoclonal IgG1 antibodies with different proven or predicted capacity to elicit CRS in clinic and measured cytokine release using a multiplex assay. We found that, in contrast to anti-CD52 antibodies (Campath-1H homolog) that elicited high level of multiple inflammatory cytokines from human blood cells in vitro, other IgG1 antibodies with CRS-inducing potential consistently induced release of a single tested cytokine, interferon (IFN)-γ, with a smaller magnitude than Campath. IFN-γ expression was observed as early as 2-4 h after incubation, mediated by natural killer cells, and dependent upon tumor necrosis factor and FcγRIII. Importantly, the magnitude of the IFN-γ response elicited by IgG1 antibodies with CRS-inducing potential was determined by donor FcγRIIIa-V158F polymorphism. Overall, our results highlight the importance of FcγRIIIa-dependent IFN-γ release in preclinical cytokine release assay for the prediction of CRS risk associated with therapeutic IgG1 antibodies.


Assuntos
Anticorpos Monoclonais/imunologia , Imunoglobulina G/imunologia , Interferon gama/imunologia , Receptores de IgG/imunologia , Alemtuzumab/imunologia , Alemtuzumab/uso terapêutico , Anticorpos Monoclonais/uso terapêutico , Citocinas/sangue , Citocinas/imunologia , Citocinas/metabolismo , Humanos , Imunoensaio/métodos , Imunoglobulina G/uso terapêutico , Interferon gama/sangue , Interferon gama/metabolismo , Neoplasias/tratamento farmacológico , Neoplasias/imunologia , Polimorfismo Genético/imunologia , Prognóstico , Receptores de IgG/genética , Síndrome
5.
Biotechnol Bioeng ; 115(3): 705-718, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29150961

RESUMO

Cross-linking of the Fcγ receptors expressed on the surface of hematopoietic cells by IgG immune complexes triggers the activation of key immune effector mechanisms, including antibody-dependent cell mediated cytotoxicity (ADCC). A conserved N-glycan positioned at the N-terminal region of the IgG CH 2 domain is critical in maintaining the quaternary structure of the molecule for Fcγ receptor engagement. The removal of a single core fucose residue from the N-glycan results in a considerable increase in affinity for FcγRIIIa leading to an enhanced receptor-mediated immunoeffector function. The enhanced potency of the molecule translates into a number of distinct advantages in the development of IgG antibodies for cancer therapy. In an effort to significantly increase the potency of an anti-CD20, IgG1 molecule, we selectively targeted the de novo GDP-fucose biosynthesis pathway of the host CHO cell line to generate >80% afucosylated IgG1 resulting in enhanced FcγRIIIa binding (13-fold) and in vitro ADCC cell-based activity (11-fold). In addition, this effective glycoengineering strategy also allowed for the utilization of the alternate GDP-fucose salvage pathway to provide a fast and efficient mechanism to manipulate the N-glycan fucosylation level to modulate IgG immune effector function.


Assuntos
Cricetulus/metabolismo , Imunoglobulina G/biossíntese , Engenharia de Proteínas , Rituximab/biossíntese , Animais , Cricetulus/genética , Glicosilação , Imunoglobulina G/genética , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Rituximab/genética
6.
Am J Physiol Renal Physiol ; 312(6): F951-F962, 2017 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-28249836

RESUMO

Transforming growth factor-alpha (TGFA) has been shown to play a role in experimental chronic kidney disease associated with nephron reduction, while its role in diabetic kidney disease (DKD) is unknown. We show here that intrarenal TGFA mRNA expression, as well as urine and serum TGFA, are increased in human DKD. We used a TGFA neutralizing antibody to determine the role of TGFA in two models of renal disease, the remnant surgical reduction model and the uninephrectomized (uniNx) db/db DKD model. In addition, the contribution of TGFA to DKD progression was examined using an adeno-associated virus approach to increase circulating TGFA in experimental DKD. In vivo blockade of TGFA attenuated kidney disease progression in both nondiabetic 129S6 nephron reduction and Type 2 diabetic uniNx db/db models, whereas overexpression of TGFA in uniNx db/db model accelerated renal disease. Therapeutic activity of the TGFA antibody was enhanced with renin angiotensin system inhibition with further improvement in renal parameters. These findings suggest a pathologic contribution of TGFA in DKD and support the possibility that therapeutic administration of neutralizing antibodies could provide a novel treatment for the disease.


Assuntos
Nefropatias Diabéticas/metabolismo , Rim/metabolismo , Fator de Crescimento Transformador alfa/metabolismo , Idoso , Inibidores da Enzima Conversora de Angiotensina/farmacologia , Animais , Anticorpos Monoclonais/farmacologia , Anticorpos Neutralizantes/farmacologia , Pressão Sanguínea , Células Cultivadas , Dependovirus/genética , Diabetes Mellitus Tipo 2/complicações , Nefropatias Diabéticas/tratamento farmacológico , Nefropatias Diabéticas/genética , Nefropatias Diabéticas/fisiopatologia , Modelos Animais de Doenças , Progressão da Doença , Receptores ErbB/metabolismo , Matriz Extracelular/metabolismo , Feminino , Técnicas de Transferência de Genes , Vetores Genéticos , Taxa de Filtração Glomerular , Humanos , Hipertensão/complicações , Hipertensão/fisiopatologia , Rim/efeitos dos fármacos , Rim/fisiopatologia , Rim/cirurgia , Masculino , Camundongos da Linhagem 129 , Camundongos Knockout , Pessoa de Meia-Idade , Nefrectomia , Fosforilação , Sistema Renina-Angiotensina , Transdução de Sinais , Fatores de Tempo , Fator de Crescimento Transformador alfa/antagonistas & inibidores , Fator de Crescimento Transformador alfa/deficiência , Fator de Crescimento Transformador alfa/genética
7.
J Inflamm Res ; 7: 121-31, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25258549

RESUMO

B-cell activating factor (BAFF) is a B-cell survival factor with a key role in B-cell homeostasis and tolerance. Dysregulated BAFF expression may contribute to autoimmune diseases or B-cell malignancies via effects on abnormal B-lymphocyte activation, proliferation, survival, and immunoglobulin secretion. Monoclonal antibodies were generated against human BAFF, characterized for species specificity and affinity, and screened for the ability to neutralize both membrane-bound and soluble BAFF. In addition, studies were undertaken to determine the relative potency of membrane-bound and soluble BAFF. Tabalumab has a high affinity for human, cynomolgus monkey, and rabbit BAFF. No binding to mouse BAFF was detected. Tabalumab was able to neutralize soluble human, cynomolgus monkey, or rabbit BAFF with equal potency. Our data demonstrate that membrane-bound BAFF can be a more potent stimulus for B-cells than soluble BAFF, and tabalumab also neutralized membrane-bound BAFF. Tabalumab prevented BAFF from binding to BAFF receptors and demonstrated pharmacodynamic effects in human BAFF transgenic mice. Tabalumab is a high-affinity human antibody with neutralizing activity against membrane-bound and soluble BAFF. Given our findings that membrane-bound BAFF can have greater in vitro potency than soluble BAFF, neutralization of both forms of BAFF is likely to be important for optimal therapeutic effect.

8.
Haematologica ; 99(9): 1516-24, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24895335

RESUMO

Anemia of chronic disease is a multifactorial disorder, resulting mainly from inflammation-driven reticuloendothelial iron retention, impaired erythropoiesis, and reduced biological activity of erythropoietin. Erythropoiesis-stimulating agents have been used for the treatment of anemia of chronic disease, although with varying response rates and potential adverse effects. Serum concentrations of hepcidin, a key regulator of iron homeostasis, are increased in patients with anemia of chronic disease and linked to the pathogenesis of this disease, because hepcidin blocks cellular iron egress, thus limiting availability of iron for erythropoiesis. We tested whether serum hepcidin levels can predict and affect the therapeutic efficacy of erythropoiesis-stimulating agent treatment using a well-established rat model of anemia of chronic disease. We found that high pre-treatment hepcidin levels correlated with an impaired hematologic response to an erythropoiesis-stimulating agent in rats with anemia of chronic disease. Combined treatment with an erythropoiesis-stimulating agent and an inhibitor of hepcidin expression, LDN-193189, significantly reduced serum hepcidin levels, mobilized iron from tissue stores, increased serum iron levels and improved hemoglobin levels more effectively than did the erythropoiesis-stimulating agent or LDN-193189 monotherapy. In parallel, both the erythropoiesis-stimulating agent and erythropoiesis-stimulating agent/LDN-193189 combined reduced the expression of cytokines known to inhibit erythropoiesis. We conclude that serum hepcidin levels can predict the hematologic responsiveness to erythropoiesis-stimulating agent therapy in anemia of chronic disease. Pharmacological inhibition of hepcidin formation improves the erythropoiesis-stimulating agent's therapeutic efficacy, which may favor a reduction of erythropoiesis-stimulating agent dosages, costs and side effects.


Assuntos
Anemia/tratamento farmacológico , Eritropoetina/farmacologia , Hematínicos/farmacologia , Hepcidinas/genética , Ferro/sangue , RNA Mensageiro/genética , Anemia/sangue , Anemia/induzido quimicamente , Anemia/patologia , Animais , Biomarcadores/sangue , Doença Crônica , Combinação de Medicamentos , Sinergismo Farmacológico , Eritropoese/efeitos dos fármacos , Feminino , Expressão Gênica , Hepcidinas/antagonistas & inibidores , Hepcidinas/sangue , Humanos , Interferon gama/antagonistas & inibidores , Interferon gama/biossíntese , Ferro/agonistas , Polissacarídeos Bacterianos , Prognóstico , Pirazóis/farmacologia , Pirimidinas/farmacologia , RNA Mensageiro/antagonistas & inibidores , RNA Mensageiro/sangue , Ratos , Ratos Endogâmicos Lew , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/biossíntese
9.
Gut ; 63(12): 1951-9, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24598129

RESUMO

OBJECTIVE: Hypoxia affects body iron homeostasis; however, the underlying mechanisms are incompletely understood. DESIGN: Using a standardised hypoxia chamber, 23 healthy volunteers were subjected to hypoxic conditions, equivalent to an altitude of 5600 m, for 6 h. Subsequent experiments were performed in C57BL/6 mice, CREB-H knockout mice, primary hepatocytes and HepG2 cells. RESULTS: Exposure of subjects to hypoxia resulted in a significant decrease of serum levels of the master regulator of iron homeostasis hepcidin and elevated concentrations of platelet derived growth factor (PDGF)-BB. Using correlation analysis, we identified PDGF-BB to be associated with hypoxia mediated hepcidin repression in humans. We then exposed mice to hypoxia using a standardised chamber and observed downregulation of hepatic hepcidin mRNA expression that was paralleled by elevated serum PDGF-BB protein concentrations and higher serum iron levels as compared with mice housed under normoxic conditions. PDGF-BB treatment in vitro and in vivo resulted in suppression of both steady state and BMP6 inducible hepcidin expression. Mechanistically, PDGF-BB inhibits hepcidin transcription by downregulating the protein expression of the transcription factors CREB and CREB-H, and pharmacological blockade or genetic ablation of these pathways abrogated the effects of PDGF-BB toward hepcidin expression. CONCLUSIONS: Hypoxia decreases hepatic hepcidin expression by a novel regulatory pathway exerted via PDGF-BB, leading to increased availability of circulating iron that can be used for erythropoiesis.


Assuntos
Hepcidinas/metabolismo , Hipóxia/metabolismo , Ferro/metabolismo , Fator de Crescimento Derivado de Plaquetas/metabolismo , Proteínas Proto-Oncogênicas c-sis/farmacologia , Adulto , Animais , Becaplermina , Modelos Animais de Doenças , Regulação para Baixo , Eritropoese/fisiologia , Feminino , Voluntários Saudáveis , Fármacos Hematológicos/farmacologia , Células Hep G2 , Humanos , Hipóxia/etiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL
10.
J Pharmacol Exp Ther ; 349(2): 330-43, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24518034

RESUMO

At least seven distinct epidermal growth factor (EGF) ligands bind to and activate the EGF receptor (EGFR). This activation plays an important role in the embryo and in the maintenance of adult tissues. Importantly, pharmacologic EGFR inhibition also plays a critical role in the pathophysiology of diverse disease states, especially cancer. The roles of specific EGFR ligands are poorly defined in these disease states. Accumulating evidence suggests a role for transforming growth factor α (TGFα) in skin, lung, and kidney disease. To explore the role of Tgfa, we generated a monoclonal antibody (mAb41) that binds to and neutralizes human Tgfa with high affinity (KD = 36.5 pM). The antibody also binds human epiregulin (Ereg) (KD = 346.6 pM) and inhibits ligand induced myofibroblast cell proliferation (IC50 values of 0.52 and 1.12 nM for human Tgfa and Ereg, respectively). In vivo, a single administration of the antibody to pregnant mice (30 mg/kg s.c. at day 14 after plug) or weekly administration to neonate mice (20 mg/kg s.c. for 4 weeks) phenocopy Tgfa knockout mice with curly whiskers, stunted growth, and expansion of the hypertrophic zone of growth plate cartilage. Humanization of this monoclonal antibody to a human IgG4 antibody (LY3016859) enables clinical development. Importantly, administration of the humanized antibody to cynomolgus monkeys is absent of the skin toxicity observed with current EGFR inhibitors used clinically and no other pathologies were noted, indicating that neutralization of Tgfa could provide a relatively safe profile as it advances in clinical development.


Assuntos
Anticorpos Monoclonais Humanizados/farmacologia , Fator de Crescimento Epidérmico/metabolismo , Receptores ErbB/metabolismo , Fator de Crescimento Transformador alfa/metabolismo , Sequência de Aminoácidos , Animais , Animais Recém-Nascidos , Anticorpos Monoclonais Humanizados/metabolismo , Anticorpos Monoclonais Humanizados/farmacocinética , Anticorpos Neutralizantes/metabolismo , Anticorpos Neutralizantes/farmacologia , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Epirregulina , Humanos , Imunoglobulina G/imunologia , Macaca fascicularis , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Dados de Sequência Molecular , Miofibroblastos/citologia , Miofibroblastos/efeitos dos fármacos , Miofibroblastos/metabolismo , Ligação Proteica , Fator de Crescimento Transformador alfa/genética
11.
Thorax ; 67(11): 985-92, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22735687

RESUMO

BACKGROUND: Therapeutic strategies to modulate the host response to bacterial pneumonia are needed to improve outcomes during community-acquired pneumonia. This study used mice with impaired Fas signalling to examine susceptibility to pneumococcal pneumonia and decoy receptor 3 analogue (DcR3-a) to correct factors associated with increased susceptibility. METHODS: Wild-type mice and those with varying degrees of impairment of Fas (lpr) or Fas ligand signalling (gld) were challenged with Streptococcus pneumoniae and microbiological and immunological outcomes measured in the presence or absence of DcR3-a. RESULTS: During established pneumonia, neutrophils became the predominant cell in the airway and gld mice were less able to clear bacteria from the lungs, demonstrating localised impairment of pulmonary neutrophil function in comparison to lpr or wild-type mice. T-cells from gld mice had enhanced activation and reduced apoptosis in comparison to wild-type and lpr mice during established pneumonia. Treatment with DcR3-a reduced T-cell activation and corrected the defect in pulmonary bacterial clearance in gld mice. CONCLUSIONS: The results suggest that imbalance in tumour necrosis factor superfamily signalling and excessive T-cell activation can impair bacterial clearance in the lung but that DcR3-a treatment can reduce T-cell activation, restore optimal pulmonary neutrophil function and enhance bacterial clearance during S pneumoniae infection.


Assuntos
Proteína Ligante Fas/metabolismo , Neutrófilos/imunologia , Fagócitos/imunologia , Pneumonia Pneumocócica/imunologia , Membro 6b de Receptores do Fator de Necrose Tumoral/farmacologia , Animais , Modelos Animais de Doenças , Proteína Ligante Fas/farmacologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Neutrófilos/efeitos dos fármacos , Fagócitos/efeitos dos fármacos , Pneumonia Pneumocócica/metabolismo , Pneumonia Pneumocócica/microbiologia , Pneumonia Pneumocócica/terapia , Síndrome do Desconforto Respiratório/imunologia , Síndrome do Desconforto Respiratório/prevenção & controle , Transdução de Sinais/efeitos dos fármacos , Streptococcus pneumoniae/imunologia
12.
J Nutr Biochem ; 23(12): 1600-8, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22444869

RESUMO

Obesity is often associated with disorders of iron homeostasis; however, the underlying mechanisms are not fully understood. Hepcidin is a key regulator of iron metabolism and may be responsible for obesity-driven iron deficiency. Herein, we used an animal model of diet-induced obesity to study high-fat-diet-induced changes in iron homeostasis. C57BL/6 mice were fed a standard (SD) or high-fat diet (HFD) for 8 weeks, and in addition, half of the mice received high dietary iron (Fe+) for the last 2 weeks. Surprisingly, HFD led to systemic iron deficiency which was traced back to reduced duodenal iron absorption. The mRNA and protein expressions of the duodenal iron transporters Dmt1 and Tfr1 were significantly higher in HFD- than in SD-fed mice, indicating enterocyte iron deficiency, whereas the mRNA levels of the duodenal iron oxidoreductases Dcytb and hephaestin were lower in HFD-fed mice. Neither hepatic and adipose tissue nor serum hepcidin concentrations differed significantly between SD- and HFD-fed mice, whereas dietary iron supplementation resulted in increased hepatic hepcidin mRNA expression and serum hepcidin levels in SD as compared to HFD mice. Our study suggests that HFD results in iron deficiency which is neither due to intake of energy-dense nutrient poor food nor due to increased sequestration in the reticulo-endothelial system but is the consequence of diminished intestinal iron uptake. We found that impaired iron absorption is independent of hepcidin but rather results from reduced metal uptake into the mucosa and discordant oxidoreductases expressions despite enterocyte iron deficiency.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Duodeno/metabolismo , Deficiências de Ferro , Obesidade/metabolismo , Adipocinas/genética , Tecido Adiposo/efeitos dos fármacos , Tecido Adiposo/metabolismo , Animais , Peptídeos Catiônicos Antimicrobianos/sangue , Peptídeos Catiônicos Antimicrobianos/genética , Proteínas de Transporte de Cátions/genética , Duodeno/efeitos dos fármacos , Eritrócitos/efeitos dos fármacos , Eritrócitos/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Hepcidinas , Absorção Intestinal , Ferro/sangue , Ferro/metabolismo , Ferro da Dieta/farmacocinética , Ferro da Dieta/farmacologia , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/etiologia , Receptores da Transferrina/genética
13.
Hum Mol Genet ; 20(6): 1232-40, 2011 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-21208937

RESUMO

The genetic determinants of variation in iron status are actively sought, but remain incompletely understood. Meta-analysis of two genome-wide association (GWA) studies and replication in three independent cohorts was performed to identify genetic loci associated in the general population with serum levels of iron and markers of iron status, including transferrin, ferritin, soluble transferrin receptor (sTfR) and sTfR-ferritin index. We identified and replicated a novel association of a common variant in the type-2 transferrin receptor (TFR2) gene with iron levels, with effect sizes highly consistent across samples. In addition, we identified and replicated an association between the HFE locus and ferritin and confirmed previously reported associations with the TF, TMPRSS6 and HFE genes. The five replicated variants were tested for association with expression levels of the corresponding genes in a publicly available data set of human liver samples, and nominally statistically significant expression differences by genotype were observed for all genes, although only rs3811647 in the TF gene survived the Bonferroni correction for multiple testing. In addition, we measured for the first time the effects of the common variant in TMPRSS6, rs4820268, on hepcidin mRNA in peripheral blood (n = 83 individuals) and on hepcidin levels in urine (n = 529) and observed an association in the same direction, though only borderline significant. These functional findings require confirmation in further studies with larger sample sizes, but they suggest that common variants in TMPRSS6 could modify the hepcidin-iron feedback loop in clinically unaffected individuals, thus making them more susceptible to imbalances of iron homeostasis.


Assuntos
Variação Genética , Ferro/sangue , Receptores da Transferrina/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Estudos Transversais , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Polimorfismo de Nucleotídeo Único , Receptores da Transferrina/metabolismo , Adulto Jovem
14.
Clin Chem ; 56(11): 1725-32, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20847326

RESUMO

BACKGROUND: Hepcidin, a key regulator of iron metabolism, binds to the iron transporter ferroportin to cause its degradation. In humans, hepcidin deficiency has been linked to hemochromatosis and iron overload, whereas increased concentrations have been reported in anemia of cancer and chronic disease. There is currently an unmet clinical need for a specific immunoassay with a low limit of quantification to measure serum concentrations of hepcidin-25, the active form of the protein. METHODS: We generated 2 antihepcidin-25 monoclonal antibodies and used them to build a sandwich ELISA. We correlated ELISA results to hepcidin-25 measurements by LC-MS and used ELISA to measure serum hepcidin-25 concentrations in normal individuals, cancer patients, and patients with rheumatoid arthritis. RESULTS: The sandwich ELISA was highly specific for hepcidin-25, having a limit of quantification of 0.01 µg/L (10 pg/mL). Serum concentrations of hepcidin-25 measured by ELISA correlated with hepcidin-25 concentrations measured by using an independent LC-MS assay (r = 0.98, P < 0.001). Hepcidin-25 concentrations were increased in patients with cancer (median 54.8 µg/L, 25%-75% range 23.2-93.5 µg/L, n = 34) and rheumatoid arthritis (median 10.6 µg/L, 25%-75% range 5.9-18.4 µg/L, n = 76) compared with healthy individuals (median 1.20 µg/L, 25%-75% range 0.42-3.07 µg/L, n = 100). CONCLUSIONS: The use of 2 monoclonal antibodies in a sandwich ELISA format provides a robust and convenient method for measuring concentrations of the active form of hepcidin. This ELISA should help to improve our understanding of the role of hepcidin in regulating iron metabolism.


Assuntos
Anticorpos Monoclonais , Peptídeos Catiônicos Antimicrobianos/sangue , Adolescente , Adulto , Peptídeos Catiônicos Antimicrobianos/imunologia , Artrite Reumatoide/sangue , Ensaio de Imunoadsorção Enzimática , Feminino , Hepcidinas , Humanos , Masculino , Pessoa de Meia-Idade , Neoplasias/sangue , Grupos Raciais , Valores de Referência , Fatores Sexuais , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Adulto Jovem
15.
Blood ; 113(21): 5277-86, 2009 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-19293425

RESUMO

The anemia of chronic disease (ACD) is characterized by macrophage iron retention induced by cytokines and the master regulator hepcidin. Hepcidin controls cellular iron efflux on binding to the iron export protein ferroportin. Many patients, however, present with both ACD and iron deficiency anemia (ACD/IDA), the latter resulting from chronic blood loss. We used a rat model of ACD resulting from chronic arthritis and mimicked ACD/IDA by additional phlebotomy to define differing iron-regulatory pathways. Iron retention during inflammation occurs in macrophages and the spleen, but not in the liver. In rats and humans with ACD, serum hepcidin concentrations are elevated, which is paralleled by reduced duodenal and macrophage expression of ferroportin. Individuals with ACD/IDA have significantly lower hepcidin levels than ACD subjects, and ACD/IDA persons, in contrast to ACD subjects, were able to absorb dietary iron from the gut and to mobilize iron from macrophages. Circulating hepcidin levels affect iron traffic in ACD and ACD/IDA and are more responsive to the erythropoietic demands for iron than to inflammation. Hepcidin determination may aid to differentiate between ACD and ACD/IDA and in selecting appropriate therapy for these patients.


Assuntos
Anemia Ferropriva/patologia , Anemia/patologia , Homeostase , Ferro/metabolismo , Anemia/sangue , Anemia/metabolismo , Anemia Ferropriva/sangue , Anemia Ferropriva/metabolismo , Animais , Peptídeos Catiônicos Antimicrobianos/sangue , Estudos de Casos e Controles , Proteínas de Transporte de Cátions/análise , Doença Crônica , Modelos Animais de Doenças , Duodeno/metabolismo , Hepcidinas , Humanos , Inflamação , Macrófagos/metabolismo , Macrófagos/patologia , Ratos , Ratos Endogâmicos Lew , Baço/metabolismo , Baço/patologia
16.
Br J Haematol ; 144(5): 789-93, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19120353

RESUMO

Iron absorption is inadequately increased in patients with chronic haemolytic anaemia, which is commonly complicated by iron overload. Growth differentiation factor 15 (GDF15) has been identified as a bone marrow-derived factor that abrogates hepcidin-mediated protection from iron overload under conditions of increased erythropoiesis. Increased concentrations of GDF15 have been reported in beta-thalassaemia patients and GDF15 has been found to suppress hepcidin expression in vitro. To further study the interdependencies of iron metabolism and erythropoiesis in vivo, the concentrations of hepcidin and GDF15 were determined in sera from 22 patients with pyruvate kinase deficiency (PKD) and 21 healthy control subjects. In PKD patients, serum hepcidin levels were 13-fold lower than in controls (2.0 ng/ml vs. 26.2 ng/ml) and GDF15 was significantly higher (859 pg/ml vs. 528 pg/ml). Serum hepcidin concentrations correlated positively with haemoglobin and negatively with serum GDF15. These results suggest that GDF15 contributes to low hepcidin expression and iron loading in PKD.


Assuntos
Peptídeos Catiônicos Antimicrobianos/sangue , Fator 15 de Diferenciação de Crescimento/sangue , Sobrecarga de Ferro/sangue , Ferro/metabolismo , Piruvato Quinase/deficiência , Adolescente , Adulto , Idoso , Peptídeos Catiônicos Antimicrobianos/metabolismo , Estudos de Casos e Controles , Eritropoese/fisiologia , Feminino , Fator 15 de Diferenciação de Crescimento/metabolismo , Hemoglobinas/análise , Hepcidinas , Humanos , Masculino , Pessoa de Meia-Idade , Contagem de Reticulócitos , Estatísticas não Paramétricas , Adulto Jovem
17.
Transfusion ; 48(10): 2197-204, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18657084

RESUMO

BACKGROUND: Individuals donating whole blood 13 times in a 2-year period without development of iron deficiency anemia (superdonors) are a self-selected population that is deferred for low hematocrit (Hct) level less frequently than other donors. STUDY DESIGN AND METHODS: Iron metabolism was assessed in 138 superdonors through a questionnaire and measurement of Hct, serum ferritin, serum hepcidin, and serum growth differentiation factor 15 (GDF15). Genetic testing for HFE and JAK-2 mutations was also performed. RESULTS AND CONCLUSIONS: Iron deficiency (ferritin level, <30 microg/L) is present in more than 60 percent of superdonors. Behaviors altering iron status included casual use of iron supplements in males, but not in females, and cigarette smoking that produced increased Hct associated with decreased ferritin. The striking biochemical characteristic of superdonors is greatly decreased serum hepcidin, consistent with their need to absorb maximal amounts of dietary iron to replace that lost from blood donation. GDF15 is normal in most superdonors, indicating that GDF15 overexpression arising from the expanded erythroid pool necessary to replace donated red cells is not the biochemical mechanism for the decreased serum hepcidin. Mutations in JAK-2 were not found, indicating that undiagnosed polycythemia vera is not a common cause for successful repeated blood donation by superdonors. Mutations in HFE associated with hemochromatosis were present in superdonors at the same frequency as the normal population. However, superdonors heterozygous for the H63D mutation in HFE had significantly decreased hepcidin : ferritin ratios demonstrating for the first time that the heterozygous state for HFE mutations is associated with alterations in hepcidin expression.


Assuntos
Anemia Ferropriva/sangue , Anemia Ferropriva/genética , Doadores de Sangue , Citocinas/genética , Antígenos de Histocompatibilidade Classe I/genética , Janus Quinase 2/genética , Proteínas de Membrana/genética , Anemia Ferropriva/tratamento farmacológico , Peptídeos Catiônicos Antimicrobianos/sangue , Feminino , Ferritinas/sangue , Fator 15 de Diferenciação de Crescimento , Hematócrito , Proteína da Hemocromatose , Hepcidinas , Humanos , Ferro/farmacologia , Ferro da Dieta/farmacologia , Masculino , Pessoa de Meia-Idade , Policitemia Vera/genética , Fumar/sangue , Inquéritos e Questionários
18.
Proc Natl Acad Sci U S A ; 105(17): 6320-5, 2008 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-18443287

RESUMO

The peptide hormone ghrelin is the only known protein modified with an O-linked octanoyl side group, which occurs on its third serine residue. This modification is crucial for ghrelin's physiological effects including regulation of feeding, adiposity, and insulin secretion. Despite the crucial role for octanoylation in the physiology of ghrelin, the lipid transferase that mediates this novel modification has remained unknown. Here we report the identification and characterization of human GOAT, the ghrelin O-acyl transferase. GOAT is a conserved orphan membrane-bound O-acyl transferase (MBOAT) that specifically octanoylates serine-3 of the ghrelin peptide. Transcripts for both GOAT and ghrelin occur predominantly in stomach and pancreas. GOAT is conserved across vertebrates, and genetic disruption of the GOAT gene in mice leads to complete absence of acylated ghrelin in circulation. The occurrence of ghrelin and GOAT in stomach and pancreas tissues demonstrates the relevance of GOAT in the acylation of ghrelin and further implicates acylated ghrelin in pancreatic function.


Assuntos
Aciltransferases/metabolismo , Grelina/metabolismo , Acilação , Aciltransferases/genética , Animais , Caprilatos/metabolismo , Linhagem Celular Tumoral , Membrana Celular/enzimologia , Sequência Conservada , Perfilação da Expressão Gênica , Regulação Enzimológica da Expressão Gênica , Grelina/sangue , Grelina/genética , Humanos , Dados de Sequência Molecular , Pâncreas/enzimologia , Peptídeos/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Serina/metabolismo , Estômago/enzimologia
19.
Cancer Immunol Immunother ; 57(6): 777-87, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17962943

RESUMO

Studies in murine models of cancer as well as in cancer patients have demonstrated that the immune response to cancer is often compromised. This paradigm is viewed as one of the major mechanisms of tumor escape. Many therapies focus on employing the professional antigen presenting dendritic cells (DC) as a strategy to overcome immune inhibition in cancer patients. Death receptor 6 (DR6) is an orphan member of the tumor necrosis factor receptor superfamily (TNFRSF21). It is overexpressed on many tumor cells and DR6(-/-) mice display altered immunity. We investigated whether DR6 plays a role in tumorigenesis by negatively affecting the generation of anti-tumor activity. We show that DR6 is uniquely cleaved from the cell surface of tumor cell lines by the membrane-associated matrix metalloproteinase (MMP)-14, which is often overexpressed on tumor cells and is associated with malignancy. We also demonstrate that >50% of monocytes differentiating into DC die when the extracellular domain of DR6 is present. In addition, DR6 affects the cell surface phenotype of the resulting immature DC and changes their cytokine production upon stimulation with LPS/IFN-gamma. The effects of DR6 are mostly amended when these immature DC are matured with IL-1beta/TNF-alpha, as measured by cell surface phenotype and their ability to present antigen. These results implicate MMP-14 and DR6 as a mechanism tumor cells can employ to actively escape detection by the immune system by affecting the generation of antigen presenting cells.


Assuntos
Metaloproteinase 14 da Matriz/metabolismo , Receptores do Fator de Necrose Tumoral/metabolismo , Animais , Diferenciação Celular , Linhagem Celular , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Células Dendríticas/citologia , Células Dendríticas/metabolismo , Humanos , Interferon gama/metabolismo , Interleucina-1beta/metabolismo , Linfócitos/citologia , Linfócitos/metabolismo , Camundongos , Monócitos/citologia , Monócitos/metabolismo , Fenótipo
20.
Drug Metab Dispos ; 35(1): 86-94, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17050651

RESUMO

It is well established that the neonatal Fc receptor (FcRn) plays a critical role in regulating IgG homeostasis in vivo. As such, modification of the interaction of IgG with FcRn has been the focus of protein-engineering strategies designed to generate therapeutic antibodies with improved pharmacokinetic properties. In the current work, we characterized differences in interaction of IgG between mouse and primate receptors using three humanized anti-tumor necrosis factor alpha antibodies with variant IgG(1) Fc regions. The wild-type and variant IgG showed a differential combination of improved affinity, modified dissociation kinetics, and altered pH-dependent complex dissociation when evaluated on the primate and murine receptors. The observed in vitro binding differences within and between species allowed us to more completely relate these parameters to their influence on the in vivo pharmacokinetics in mice and cynomolgus monkeys. The variant antibodies have different pharmacokinetic behavior in cynomolgus monkeys and mice, which appears to be related to the unique binding characteristics observed with the murine receptor. However, we did not observe a direct relationship between increased binding affinity to the receptor and improved pharmacokinetic properties for these molecules in either species. This work provides further insights into how the FcRn/IgG interaction may be modulated to develop monoclonal antibodies with improved therapeutic properties.


Assuntos
Anticorpos/metabolismo , Antígenos de Histocompatibilidade Classe I/metabolismo , Imunoglobulina G/genética , Imunoglobulina G/metabolismo , Receptores Fc/metabolismo , Fator de Necrose Tumoral alfa/imunologia , Animais , Células CHO , Cricetinae , Cricetulus , Humanos , Macaca fascicularis , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Ligação Proteica , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA