Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Front Immunol ; 14: 1266668, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38077343

RESUMO

An antibody molecule that can bind to multiple distinct antigens is defined as polyreactive. In the present study, we performed statistical analyses to assess sequence correlates of polyreactivity of >600 antibodies cloned from different B-cell types of healthy humans. The data revealed several sequence patterns of variable regions of heavy and light immunoglobulin chains that determine polyreactivity. The most prominent identified patterns were increased number of basic amino acid residues, reduced frequency of acidic residues, increased number of aromatic and hydrophobic residues, and longer length of CDR L1. Importantly, our study revealed that antibodies isolated from different B-cell populations used distinct sequence patterns (or combinations of them) for polyreactive antigen binding. Furthermore, we combined the data from sequence analyses with molecular modeling of selected polyreactive antibodies and demonstrated that human antibodies can use multiple pathways for achieving antigen-binding promiscuity. These data reconcile some contradictions in the literature regarding the determinants of antibody polyreactivity. Moreover, our study demonstrates that the mechanism of polyreactivity of antibodies evolves during immune response and might be tailored to specific functional properties of different B-cell compartments. Finally, these data can be of use for efforts in the development and engineering of therapeutic antibodies.


Assuntos
Anticorpos , Região Variável de Imunoglobulina , Humanos , Região Variável de Imunoglobulina/genética , Região Variável de Imunoglobulina/química , Linfócitos B , Imunidade Adaptativa
2.
J Thromb Haemost ; 21(9): 2405-2417, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37271431

RESUMO

BACKGROUND: Transplacental delivery of maternal immunoglobulin G (IgG) provides humoral protection during the first months of life until the newborn's immune system reaches maturity. The maternofetal interface has been exploited therapeutically to replace missing enzymes in the fetus, as shown in experimental mucopolysaccharidoses, or to shape adaptive immune repertoires during fetal development and induce tolerance to self-antigens or immunogenic therapeutic molecules. OBJECTIVES: To investigate whether proteins that are administered to pregnant mice or endogenously present in their circulation may be delivered through the placenta. METHODS: We engineered monovalent immunoglobulin G (FabFc) specific for different domains of human factor VIII (FVIII), a therapeutically relevant model antigen. FabFc was injected with exogenous FVIII into pregnant severe hemophilia A mice or pregnant mice expressing human FVIII following AAV8-mediated gene therapy. FabFc and FVIII were detected in the pregnant mice and/or fetuses by enzyme-linked immunosorbent assay and immunohistochemistry. RESULTS: Administration of FabFc to pregnant mice allowed the maternofetal delivery of FVIII in a FcRn-dependent manner. FVIII antigen levels achieved in the fetuses represented 10% of normal plasma levels in the human. We identified antigen/FabFc complex stability, antigen size, and shielding of promiscuous protein patches as key parameters to foster optimal antigen delivery. CONCLUSION: Our results pave the way toward the development of novel strategies for the in utero delivery of endogenous maternal proteins to replace genetically deficient fetal proteins or to educate the immune system and favor active immune tolerance upon protein encounter later in life.


Assuntos
Hemofilia A , Imunoglobulina G , Gravidez , Feminino , Camundongos , Humanos , Animais , Fator VIII , Hemofilia A/genética , Hemofilia A/terapia , Placenta , Terapia Genética , Tolerância Imunológica
3.
Commun Biol ; 6(1): 168, 2023 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-36774392

RESUMO

Intravascular hemolysis occurs in diverse pathological conditions. Extracellular hemoglobin and heme have strong pro-oxidative and pro-inflammatory potentials that can contribute to the pathology of hemolytic diseases. However, many of the effects of extracellular hemoglobin and heme in hemolytic diseases are still not well understood. Here we demonstrate that oxidized hemoglobin (methemoglobin) can modify the antigen-binding characteristics of human immunoglobulins. Thus, incubation of polyclonal or some monoclonal human IgG in the presence of methemoglobin results in an appearance of binding reactivities towards distinct unrelated self-proteins, including the protein constituent of hemoglobin i.e., globin. We demonstrate that a transfer of heme from methemoglobin to IgG is indispensable for this acquisition of antibody polyreactivity. Our data also show that only oxidized form of hemoglobin have the capacity to induce polyreactivity of antibodies. Site-directed mutagenesis of a heme-sensitive human monoclonal IgG1 reveals details about the mechanism of methemoglobin-induced antigen-binding polyreactivity. Further here we assess the kinetics and thermodynamics of interaction of a heme-induced polyreactive human antibody with hemoglobin and myoglobin. Taken together presented data contribute to a better understanding of the functions of extracellular hemoglobin in the context of hemolytic diseases.


Assuntos
Heme , Metemoglobina , Humanos , Heme/metabolismo , Metemoglobina/metabolismo , Hemoglobinas/metabolismo , Imunoglobulina G , Anticorpos Monoclonais , Hemólise
4.
Int J Mol Sci ; 24(4)2023 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-36834827

RESUMO

The interaction of some human antibodies with heme results in posttranslational acquisition of binding to various self- and pathogen-derived antigens. The previous studies on this phenomenon were performed with oxidized heme (Fe3+). In the present study, we elucidated the effect of other pathologically relevant species of heme, i.e., species that were formed after contact of heme with oxidizing agents such as hydrogen peroxide, situations in which heme's iron could acquire higher oxidation states. Our data reveal that hyperoxidized species of heme have a superior capacity to heme (Fe3+) in triggering the autoreactivity of human IgG. Mechanistic studies demonstrated that oxidation status of iron was of critical importance for the heme's effect on antibodies. We also demonstrated that hyperoxidized heme species interacted at higher affinities with IgG and that this binding occurred through a different mechanism as compared to heme (Fe3+). Regardless of their profound functional impact on the antigen-binding properties of antibodies, hyperoxidized species of heme did not affect Fc-mediated functions of IgG, such as binding to the neonatal Fc receptor. The obtained data contribute to a better understanding of the pathophysiological mechanism of hemolytic diseases and of the origin of elevated antibody autoreactivity in patients with some hemolytic disorders.


Assuntos
Heme , Imunoglobulina G , Recém-Nascido , Humanos , Heme/metabolismo , Oxirredução , Imunidade Adaptativa , Ferro
5.
Protein Sci ; 31(11): e4447, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36305765

RESUMO

SARS-CoV-2 infects cells by attachment to its receptor-the angiotensin converting enzyme 2 (ACE2). Regardless of the wealth of structural data, little is known about the physicochemical mechanism of interactions of the viral spike (S) protein with ACE2 and how this mechanism has evolved during the pandemic. Here, we applied experimental and computational approaches to characterize the molecular interaction of S proteins from SARS-CoV-2 variants of concern (VOC). Data on kinetics, activation-, and equilibrium thermodynamics of binding of the receptor binding domain (RBD) from VOC with ACE2 as well as data from computational protein electrostatics revealed a profound remodeling of the physicochemical characteristics of the interaction during the evolution. Thus, as compared to RBDs from Wuhan strain and other VOC, Omicron RBD presented as a unique protein in terms of conformational dynamics and types of non-covalent forces driving the complex formation with ACE2. Viral evolution resulted in a restriction of the RBD structural dynamics, and a shift to a major role of polar forces for ACE2 binding. Further, we investigated how the reshaping of the physicochemical characteristics of interaction affects the binding specificity of S proteins. Data from various binding assays revealed that SARS-CoV-2 Wuhan and Omicron RBDs manifest capacity for promiscuous recognition of unrelated human proteins, but they harbor distinct reactivity patterns. These findings might contribute for mechanistic understanding of the viral tropism and capacity to evade immune responses during evolution.


Assuntos
COVID-19 , SARS-CoV-2 , Humanos , SARS-CoV-2/genética , Enzima de Conversão de Angiotensina 2 , Glicoproteína da Espícula de Coronavírus/genética , Glicoproteína da Espícula de Coronavírus/química , Peptidil Dipeptidase A/química , Peptidil Dipeptidase A/metabolismo , Ligação Proteica
6.
Immunobiology ; 227(3): 152213, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35429697

RESUMO

Previous studies have shown that polyreactive antibodies play an important role in the frontline defense against the dissemination of pathogens in the pre-immune host. Interestingly, antigen-binding polyreactivity can not only be inherent, but also acquired post-translationally. The ability of individual monoclonal IgG and IgE antibodies to acquire polyreactivity following contact with various agents that destabilize protein structure (urea, low pH) or have a pro-oxidative potential (heme, ferrous ions) has been studied in detail. However, to the best of our knowledge this property of human IgA has previously been described only cursorily. In the present study pooled human serum IgA and two human monoclonal IgA antibodies were exposed to buffers with acidic pH, to free heme or to ferrous ions, and the antigen-binding behavior of the native and modified IgA to viral and bacterial antigens were compared using immunoblot and ELISA. We observed a dose-dependent increase in reactivity to several bacterial extracts and to pure viral antigens. This newly described property of IgA may have therapeutic potential as has already been shown for pooled IgG with induced polyreactivity.


Assuntos
Anticorpos Monoclonais , Imunoglobulina G , Especificidade de Anticorpos , Heme , Humanos , Imunoglobulina A , Íons
7.
Antibodies (Basel) ; 11(1)2022 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-35225869

RESUMO

Therapeutic monoclonal antibodies have exerted a transformative impact on clinical practice in last two decades. However, development of a therapeutic antibody remains a complex process. Various physiochemical and functional liabilities can compromise the production or the therapeutic efficacy of antibodies. One of these liabilities is the susceptibility to oxidation. In the present study, we portrayed an oxidation-dependent vulnerability of immunoglobulins that can be of concern for therapeutic antibodies. By using a library of 119 monoclonal IgG1 molecules, containing variable domain matching clinical-stage antibodies, we demonstrated that a substantial number of these molecules acquired antigen-binding polyreactivity upon exposure to ferrous ions. Statistical analyses revealed that the potential for induction of polyreactivity by the redox-active metal ions correlated with a higher number of somatic mutations in V genes encoding variable domains of heavy and light immunoglobulin chains. Moreover, the sensitive antibodies used with biased frequencies particular V gene families encoding variable domains of their light chains. Besides the exposure to ferrous ions the induction of polyreactivity of therapeutic antibodies occurred after contact with an unrelated pro-oxidative substance-hypochlorite ions. Our data also revealed that induction of polyreactivity by pro-oxidative agents did not impact the binding of antibodies to their cognate antigens. The results from this study may contribute for better selection of antibody therapeutics with suitable developability profiles.

8.
Mol Immunol ; 140: 233-239, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34773862

RESUMO

Therapeutic antibodies should cover particular physicochemical and functional requirements for successful entry into clinical practice. Numerous experimental and computational approaches have been developed for early identification of different unfavourable features of antibodies. Immune repertoires of healthy humans contain a fraction of antibodies that recognize nitroarenes. These antibodies have been demonstrated to manifest antigen-binding polyreactivity. Here we observed that >20 % of 112 clinical stage therapeutic antibodies show pronounced binding to 2,4-dinitrophenol conjugated to albumin. This interaction predicts a number of unfavourable functional and physicochemical features of antibodies such as polyreactivity, tendency for self-association, stability and expression yields. Based on these findings we proposed a simple approach that may add to the armamentarium of assays for early identification of developability liabilities of antibodies intended for therapeutic use.


Assuntos
2,4-Dinitrofenol/metabolismo , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/uso terapêutico , Dinitrofenóis/metabolismo , Humanos , Imunoglobulina G/metabolismo , Ligação Proteica , Estabilidade Proteica , Soroalbumina Bovina/metabolismo
9.
Commun Biol ; 4(1): 391, 2021 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-33758329

RESUMO

Immunoglobulin repertoires contain a fraction of antibodies that recognize low molecular weight compounds, including some enzymes' cofactors, such as heme. Here, by using a set of 113 samples with variable region sequences matching clinical-stage antibodies, we demonstrated that a considerable number of these antibodies interact with heme. Antibodies that interact with heme possess specific sequence traits of their antigen-binding regions. Moreover they manifest particular physicochemical and functional qualities i.e. increased hydrophobicity, higher propensity of self-binding, higher intrinsic polyreactivity and reduced expression yields. Thus, interaction with heme is a strong predictor of different molecular and functional qualities of antibodies. Notably, these qualities are of high importance for therapeutic antibodies, as their presence was associated with failure of drug candidates to reach clinic. Our study reveled an important facet of information about relationship sequence-function in antibodies. It also offers a convenient tool for detection of liabilities of therapeutic antibodies.


Assuntos
Anticorpos Monoclonais/metabolismo , Heme/metabolismo , Região Variável de Imunoglobulina/metabolismo , Especificidade de Anticorpos , Sítios de Ligação de Anticorpos , Epitopos , Heme/imunologia , Simulação de Acoplamento Molecular , Ligação Proteica
10.
J Immunol ; 205(10): 2850-2860, 2020 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-33077645

RESUMO

Neonatal Fc receptor (FcRn) has a key role in the homeostasis of IgG. Despite its physiological and clinical importance, the interaction of IgG and FcRn remains not completely comprehended. Thus, IgG molecules with identical constant portions but with minor differences in their V regions have been demonstrated to interact with FcRn with a considerable heterogeneity in the binding affinity. To understand this discrepancy, we dissected the physicochemical mechanism of the interaction of 10 human IgG1 to human FcRn. The interactions of two Abs in the presence of their cognate Ags were also examined. Data from activation and equilibrium thermodynamics analyses as well as pH dependence of the kinetics revealed that the V region of IgG could modulate a degree of conformational changes and binding energy of noncovalent contacts at the FcRn binding interface. These results suggest that the V domains modulate FcRn binding site in Fc by allosteric effects. These findings contribute for a deeper understanding of the mechanism of IgG-FcRn interaction. They might also be of relevance for rational engineering of Abs for optimizing their pharmacokinetic properties.


Assuntos
Anticorpos Monoclonais/metabolismo , Antígenos de Histocompatibilidade Classe I/metabolismo , Imunoglobulina G/metabolismo , Domínios Proteicos/imunologia , Receptores Fc/metabolismo , Regulação Alostérica/imunologia , Anticorpos Monoclonais/química , Antígenos/metabolismo , Sítios de Ligação , Antígenos de Histocompatibilidade Classe I/química , Humanos , Concentração de Íons de Hidrogênio , Imunoglobulina G/química , Ligação Proteica/imunologia , Receptores Fc/química , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Termodinâmica
11.
Anal Biochem ; 607: 113865, 2020 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-32738209

RESUMO

The standard assay for characterization of interaction of heme with proteins is absorbance spectroscopy. However, this approach demands relatively large quantities of proteins and it is difficult to perform in high-throughput manner. Here, we describe an immunosorbent assay based on the covalent in situ conjugation of heme to a pre-coated carrier. Advantage of this assay is that it allows both identification of heme-binding proteins and quantification of their binding avidity, using only minimal amounts of protein (1-10 µg). Importantly, the same approach can be used for covalent linkage of other natural or synthetic compounds and analyzing their interactions with proteins.


Assuntos
Heme/química , Hemoglobinas/análise , Imunoglobulina G/química , Técnicas Biossensoriais , Ensaios de Triagem em Larga Escala , Humanos , Limite de Detecção , Ligação Proteica , Espectrofotometria , Propriedades de Superfície
12.
Front Immunol ; 11: 810, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32477339

RESUMO

In humans, maternal IgGs are transferred to the fetus from the second trimester of pregnancy onwards. The transplacental delivery of maternal IgG is mediated by its binding to the neonatal Fc receptor (FcRn) after endocytosis by the syncytiotrophoblast. IgGs present in the maternal milk are also transferred to the newborn through the digestive epithelium upon binding to the FcRn. Importantly, the binding of IgGs to the FcRn is also responsible for the recycling of circulating IgGs that confers them with a long half-life. Maternally delivered IgG provides passive immunity to the newborn, for instance by conferring protective anti-flu or anti-pertussis toxin IgGs. It may, however, lead to the development of autoimmune manifestations when pathological autoantibodies from the mother cross the placenta and reach the circulation of the fetus. In recent years, strategies that exploit the transplacental delivery of antigen/IgG complexes or of Fc-fused proteins have been validated in mouse models of human diseases to impose antigen-specific tolerance, particularly in the case of Fc-fused factor VIII (FVIII) domains in hemophilia A mice or pre-pro-insulin (PPI) in the case of preclinical models of type 1 diabetes (T1D). The present review summarizes the mechanisms underlying the FcRn-mediated transcytosis of IgGs, the physiopathological relevance of this phenomenon, and the repercussion for drug delivery and shaping of the immune system during its ontogeny.


Assuntos
Antígenos/imunologia , Tolerância Imunológica , Troca Materno-Fetal/imunologia , Animais , Autoanticorpos/metabolismo , Feminino , Feto/imunologia , Antígenos de Histocompatibilidade Classe I/metabolismo , Humanos , Sistema Imunitário/embriologia , Sistema Imunitário/metabolismo , Imunoglobulina G/metabolismo , Camundongos , Placenta/imunologia , Gravidez , Transporte Proteico/imunologia , Receptores Fc/metabolismo , Transcitose/imunologia
13.
Cell Death Dis ; 11(1): 50, 2020 01 23.
Artigo em Inglês | MEDLINE | ID: mdl-31974400

RESUMO

Autophagy plays an important role in the regulation of autoimmune and autoinflammatory responses of the immune cells. Defective autophagy process is associated with various autoimmune and inflammatory diseases. Moreover, in many of these diseases, the therapeutic use of normal immunoglobulin G or intravenous immunoglobulin (IVIG), a pooled normal IgG preparation, is well documented. Therefore, we explored if IVIG immunotherapy exerts therapeutic benefits via induction of autophagy in the immune cells. Here we show that IVIG induces autophagy in peripheral blood mononuclear cells (PBMCs). Further dissection of this process revealed that IVIG-induced autophagy is restricted to inflammatory cells like monocytes, dendritic cells, and M1 macrophages but not in cells associated with Th2 immune response like M2 macrophages. IVIG induces autophagy by activating AMP-dependent protein kinase, beclin-1, class III phosphoinositide 3-kinase and p38 mitogen-activated protein kinase and by inhibiting mammalian target of rapamycin. Mechanistically, IVIG-induced autophagy is F(ab')2-dependent but sialylation independent, and requires endocytosis of IgG by innate cells. Inhibition of autophagy compromised the ability of IVIG to suppress the inflammatory cytokines in innate immune cells. Moreover, IVIG therapy in inflammatory myopathies such as dermatomyositis, antisynthetase syndrome and immune-mediated necrotizing myopathy induced autophagy in PBMCs and reduced inflammatory cytokines in the circulation, thus validating the translational importance of these results. Our data provide insight on how circulating normal immunoglobulins maintain immune homeostasis and explain in part the mechanism by which IVIG therapy benefits patients with autoimmune and inflammatory diseases.


Assuntos
Anti-Inflamatórios/farmacologia , Autofagia/efeitos dos fármacos , Imunoglobulinas Intravenosas/farmacologia , Leucócitos Mononucleares/metabolismo , Adenilato Quinase/metabolismo , Proteína Beclina-1/metabolismo , Linhagem Celular , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/metabolismo , Células Dendríticas/ultraestrutura , Endocitose/efeitos dos fármacos , Humanos , Imunidade Inata/efeitos dos fármacos , Fragmentos Fab das Imunoglobulinas/metabolismo , Leucócitos Mononucleares/efeitos dos fármacos , Lipopolissacarídeos/farmacologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Organelas/efeitos dos fármacos , Organelas/metabolismo , Organelas/ultraestrutura , Fosfatidilinositol 3-Quinase/metabolismo , Fosforilação/efeitos dos fármacos , Serina-Treonina Quinases TOR/metabolismo , Doadores de Tecidos , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
14.
ACS Omega ; 4(24): 20450-20458, 2019 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-31858028

RESUMO

In a healthy immune repertoire, there exists a fraction of polyreactive antibodies that can bind to a variety of unrelated self- and foreign antigens. Apart from naturally polyreactive antibodies, in every healthy individual, there is a fraction of antibody that can gain polyreactivity upon exposure to porphyrin cofactor heme. Molecular mechanisms and biological significance of the appearance of cryptic polyreactivity are not well understood. It is believed that heme acts as an interfacial cofactor between the antibody and the newly recognized antigens. To further test this claim and gain insight into the types of interactions involved in heme binding, we herein investigated the influence of a group of aromatic guanylhydrazone molecules on the heme-induced antibody polyreactivity. From the analysis of SAR and the results of UV-vis absorbance spectroscopy, it was concluded that the most probable mechanism by which the studied molecules inhibit heme-mediated polyreactivity of the antibody is the direct binding to heme, thus preventing heme from binding to antibody and/or antigen. The inhibitory capacity of the most potent compounds was substantially higher than that of chloroquine, a well-known heme binder. Some of the guanylhydrazone molecules were able to induce polyreactivity of the studied antibody themselves, possibly by a mechanism similar to heme. Results described here point to the conclusion that heme indeed must bind to an antibody to induce its polyreactivity, and that both π-stacking interactions and iron coordination contribute to the binding affinity, while certain structures, such as guanylhydrazones, can interfere with these processes.

15.
Mol Immunol ; 112: 338-346, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31254774

RESUMO

Therapeutic antibodies have transformed the clinical practice. Not surprisingly, development of antibody therapeutics is currently the main focus of the biotechnology industry. Nonetheless, the development process is complex, and many antibodies do not reach the clinic. Reasons for the failures include, undesired binding behavior (polyreactivity), low stability, poor expression yields, unfavorable pharmacokinetics etc. Numerous studies have proposed different analytical methods for assessment of physicochemical parameters of antibodies and identification of problematic molecules at early stages of the development process. These studies, however, have not addressed the basic mechanistic question of how sequence features of variable regions determinate the different biophysical characteristics and the binding behavior of the antibodies. In a recent study, Jain et al assessed 12 biophysical qualities of 137 monoclonal therapeutic antibodies. We used the raw data from this comprehensive study to perform correlation analyses of different biophysical measurables with various sequence features of variable regions of the antibodies - number of mutations, length of hypervariable loops, and frequency of amino acid residue types. The obtained data reveled significant relationships between the sequence characteristics of the variable domains and different physiochemical properties of antibodies. The data from this study can assist in design of a set of criteria for early identification of antibodies with developability issues. Moreover, our findings provide novel fundamental insights into the sequence-function relationship of antibodies.


Assuntos
Anticorpos Monoclonais/imunologia , Especificidade de Anticorpos/imunologia , Regiões Determinantes de Complementaridade/imunologia , Região Variável de Imunoglobulina/imunologia , Aminoácidos/imunologia , Humanos
16.
J Pharm Biomed Anal ; 172: 311-319, 2019 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-31085393

RESUMO

Heme serves as a prosthetic group of numerous proteins involved in the oxidative metabolism. As result of various pathological conditions associated with hemolysis or tissue damage, large quantities of hemoproteins and heme can be released extracellularly. Extracellular heme has pronounced pathogenic effects in hemolytic diseases, mediated by its pro-oxidative and pro-inflammatory activities. The pathogenic potential of heme is mostly expressed when the molecule is in protein unbound form. The pathological relevance of free heme deems it necessary to develop reliable approaches for its assessment. Here we developed a technique based on UV-vis absorbance spectroscopy, where cysteine was used as a spectroscopy probe to distinguish between heme-bound to plasma proteins or hemoglobin from free heme. This technique allowed estimation of the heme-binding capacity of human serum, of particular heme scavenging proteins (albumin, hemopexin) or of immunoglobulins. The main advantage of the proposed approach is that it can distinguish free heme from heme associated with proteins with a wide range of affinities. The described strategy can be used for evaluation of heme-binding capacity of human plasma or serum following intravascular hemolysis or for estimation of stoichiometry of interaction of heme with a given protein.


Assuntos
Proteínas Sanguíneas/metabolismo , Cisteína/metabolismo , Heme/metabolismo , Hemoglobinas/metabolismo , Hemólise/fisiologia , Hemopexina/metabolismo , Humanos , Oxirredução , Ligação Proteica/fisiologia , Análise Espectral/métodos
17.
Mol Immunol ; 111: 205-208, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31078967

RESUMO

Therapeutic intravenous immunoglobulin preparations (IVIg) are used for treatment of wide range of autoimmune and inflammatory diseases. Versatile mechanisms have been reported to contribute to the immunomodulatory effects of IVIg. Here we demonstrate that IVIg has a strong potential to inhibit pro-inflammatory effect of extracellular heme. Indeed, the presence of immunoglobulins reduced the potential of heme to activate the complement system on the surface of human endothelial cells. Since extracellular heme is considered as one of the principal pathogenic factors in hemolytic disorders, its therapeutic scavenging by IVIg may have significant clinical repercussions.


Assuntos
Anti-Inflamatórios/imunologia , Heme/imunologia , Imunoglobulinas Intravenosas/imunologia , Inflamação/imunologia , Doenças Autoimunes/imunologia , Linhagem Celular , Proteínas do Sistema Complemento/imunologia , Células Endoteliais/imunologia , Células Endoteliais da Veia Umbilical Humana , Humanos
18.
Eur J Rheumatol ; 5(3): 173-178, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30185370

RESUMO

OBJECTIVE: Antibodies with catalytic (hydrolytic) properties to DNA or RNA have been reported in systemic lupus erythematosus (SLE). However, it is well known that ethnicity plays an important role in the presentation of SLE and severity of the disease; hence, these data may not truly represent a general feature of all SLE patients. Therefore, we have analyzed the hydrolyzing activity of immunoglobulin G (IgG) of SLE patients from the Indian population with an aim to decode whether the catalytic antibody response represents part of an active disease process. METHODS: IgGs were isolated from the sera of 72 consecutive patients diagnosed with SLE. As a control, IgGs from healthy donors were used. The catalytic activity of IgG was measured by PFR-MCA and affinity-linked oligonucleotide nuclease assay. RESULTS: IgGs from patients with SLE from the Indian subcontinent displayed significantly higher hydrolysis rates of both the surrogate substrate, PFR-MCA, and the DNA than IgG from healthy individuals. Intergroup comparisons of the IgG-PFR-MCA interactions with clinical manifestations of the disease demonstrated a significantly increased level of hydrolysis among the patients with renal involvement who tested positive for anti-dsDNA antibodies. The PFR-MCA hydrolysis also appears to be associated with the active disease (p=0.0988, vs. inactive group). CONCLUSION: The prevalence of catalytic antibodies represents a general feature of SLE patients, irrespective of their origin.

19.
Methods Mol Biol ; 1643: 135-145, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28667534

RESUMO

An antibody molecule that recognizes multiple unrelated antigens is defined as polyreactive. Polyreactivity is an intrinsic characteristic of immune repertoires. Degenerated antigen binding diversifies the repertoire of specificities, thus contributing to immune defense and immune regulation. Immune repertoire contains also a fraction of immunoglobulins, which acquire polyreactivity only following contact with various protein-destabilizing or pro-oxidative substances. Posttranslational induction of the antibody polyreactivity may have important repercussion for laboratory practice, as well as in cases of pathological conditions accompanied by liberation of large quantities of pro-oxidative substances such as heme, labile iron, or reactive oxygen species. Antibodies with induced polyreactivity have been demonstrated to exert pathogen neutralization and immune regulatory potential in inflammatory conditions, suggesting that this phenomenon may be exploited for design of therapeutic strategies. In this article, we provide description of the basic procedures for uncovering of the cryptic polyreactivity of antibodies by heme, ferrous ions, and acid pH solution.


Assuntos
Anticorpos/imunologia , Anticorpos/metabolismo , Especificidade de Anticorpos/imunologia , Reações Cruzadas/imunologia , Processamento de Proteína Pós-Traducional , Heme/imunologia , Hemina/imunologia , Humanos , Concentração de Íons de Hidrogênio , Imunoglobulina G/imunologia , Imunoglobulina G/metabolismo , Imunoglobulinas/imunologia , Imunoglobulinas/metabolismo
20.
Front Immunol ; 7: 465, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27867382

RESUMO

The programed death-1 (PD-1)-programed death ligand-1 (PD-L1) and PD-L2 co-inhibitory pathway has been implicated in the evasion strategies of Mycobacterium tuberculosis. Specifically, M. tuberculosis-induced PD-L1 orchestrates expansion of regulatory T cells and suppression of Th1 response. However, the role of PD pathway in regulating Th17 response to M. tuberculosis has not been investigated. In the present report, we demonstrate that M. tuberculosis and M. tuberculosis-derived antigen fractions have differential abilities to mediate human monocyte- and dendritic cell (DC)-mediated Th17 response and were independent of expression of PD-L1 or PD-L2 on aforementioned antigen-presenting cells. Importantly, we observed that blockade of PD-L1 or PD-1 did not significantly modify either the frequencies of Th17 cells or the production of IL-17 from CD4+ T cells though IFN-γ response was significantly enhanced. On the contrary, IL-1ß from monocytes and DCs were critical for the Th17 response to M. tuberculosis. Together, our results indicate that IL-1ß, but not members of the programed death pathway, is critical for human Th17 response to M. tuberculosis.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...