Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
1.
Cell Immunol ; 347: 104020, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31767118

RESUMO

High-mobility group box 1 (HMGB1) concentration in serum or plasma has been proposed as an important biological marker in various inflammation-related pathologies. We previously showed that low titer autoantibodies against HMGB1 could emerge during the course of sepsis. Importantly their presence was positively related with patients' survival. In this study, we focused on plasma samples from 2 patients who survived sepsis and exhibited high titer antibodies to HMGB1. These antibodies were proved to be specific for HMGB1 since they did not bind to HMGB2 or to human serum albumin. Following IgG purification, it has shown that both patients secreted HMGB1-hydrolyzing autoantibodies in vitro. These findings suggested that proteolytic antibodies directed against HMGB1 can be produced in patients surviving septic shock.


Assuntos
Autoanticorpos/imunologia , Proteína HMGB1/imunologia , Choque Séptico/imunologia , Autoanticorpos/sangue , Proteína HMGB2/imunologia , Humanos , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Proteólise , Albumina Sérica Humana/imunologia , Choque Séptico/mortalidade , Choque Séptico/patologia
2.
J Immunol ; 196(10): 4075-81, 2016 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-27067006

RESUMO

Renal transplant is the treatment of choice for patients with terminal end-stage renal disease. We have previously identified low levels of catalytic IgG as a potential prognosis marker for chronic allograft rejection. The origin and physiopathological relevance of catalytic Abs is not well understood, owing to the fact that catalytic Abs have been studied in relatively small cohorts of patients with rare diseases and/or without systematic follow-up. In the current study, we have followed the evolution of the levels of catalytic IgG in a large cohort of renal transplant patients over a 2-y period. Our results demonstrate that, prior to transplant, patients with renal failure present with heterogeneous levels of IgG hydrolyzing the generic proline-phenylalanine-arginine-methylcoumarinamide (PFR-MCA) substrate. PFR-MCA hydrolysis was greater for patients' IgG than for a therapeutic preparation of pooled IgG from healthy donors. Renal transplant was marked by a drastic decrease in levels of catalytic IgG over 3 mo followed by a steady increase during the next 21 mo. Patients who displayed high levels of catalytic IgG pretransplant recovered high levels of catalytic Abs 2 y posttransplant. Interestingly, IgG-mediated hydrolysis of a model protein substrate, procoagulant factor VIII, did not correlate with that of PFR-MCA prior transplantation, whereas it did 12 mo posttransplant. Taken together, our results suggest that the level of circulating catalytic IgG under pathological conditions is an intrinsic property of each individual's immune system and that recovery of pretransplant levels of catalytic IgG is accompanied by changes in the repertoire of target Ags.


Assuntos
Biomarcadores/metabolismo , Rejeição de Enxerto/imunologia , Sistema Imunitário , Imunoglobulina G/metabolismo , Transplante de Rim , Adulto , Idoso , Idoso de 80 Anos ou mais , Anticorpos Catalíticos , Autoanticorpos/metabolismo , Coagulação Sanguínea , Doença Crônica , Fator VIII/metabolismo , Feminino , Seguimentos , Rejeição de Enxerto/diagnóstico , Humanos , Masculino , Pessoa de Meia-Idade , Transplantados , Adulto Jovem
3.
J Neuroinflammation ; 13(1): 94, 2016 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-27126523

RESUMO

BACKGROUND: Whereas demyelination underlies early neurological symptoms in multiple sclerosis (MS), axonal damage is considered critical for permanent chronic deficits. Intracerebral infection of susceptible mouse strains with Theiler's murine encephalomyelitis virus (TMEV) results in chronic induced demyelinating disease (TMEV-IDD) with progressive axonal loss and neurologic dysfunction similar to progressive forms of MS. We previously reported that treatment of chronic TMEV-IDD mice with a neurite outgrowth-promoting natural human antibody, HIgM12, improved brainstem NAA concentrations and preserved functional motor activity. In order to translate this antibody toward clinical trial, we generated a fully human recombinant form of HIgM12, rHIgM12, determined the optimal in vivo dose for functional improvement in TMEV-IDD, and evaluated the functional preservation of descending spinal cord axons by retrograde labeling. FINDINGS: SJL/J mice at 45 to 90 days post infection (dpi) were studied. A single intraperitoneal dose of 0.25 mg/kg of rHIgM12 per mouse is sufficient to preserve motor function in TMEV-IDD. The optimal dose was 10 mg/kg. rHIgM12 treatment protected the functional transport in spinal cord axons and led to 40 % more Fluoro-Gold-labeled brainstem neurons in retrograde transport studies. This suggests that axons are not only present but also functionally competent. rHIgM12-treated mice also contained more mid-thoracic (T6) spinal cord axons than controls. CONCLUSIONS: This study confirms that a fully human recombinant neurite outgrowth-promoting monoclonal IgM is therapeutic in a model of progressive MS using multiple reparative readouts. The minimum effective dose is similar to that of a remyelination-promoting monoclonal human IgM discovered by our group that is presently in clinical trials for MS.


Assuntos
Anticorpos Monoclonais/farmacologia , Axônios/efeitos dos fármacos , Imunoglobulina M/farmacologia , Esclerose Múltipla/patologia , Fármacos Neuroprotetores/farmacologia , Animais , Axônios/patologia , Tronco Encefálico/patologia , Modelos Animais de Doenças , Humanos , Camundongos , Medula Espinal/patologia , Theilovirus
4.
J Neuroinflammation ; 13(1): 293, 2016 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-27855706

RESUMO

BACKGROUND: We investigated the role of human HLA class I molecules in persistent central nervous system (CNS) injury versus repair following virus infection of the CNS. METHODS: Human class I A11+ and B27+ transgenic human beta-2 microglobulin positive (Hß2m+) mice of the H-2 b background were generated on a combined class I-deficient (mouse beta-2 microglobulin deficient, ß2m0) and class II-deficient (mouse Aß0) phenotype. Intracranial infection with Theiler's murine encephalomyelitis virus (TMEV) in susceptible SJL mice results in acute encephalitis with prominent injury in the hippocampus, striatum, and cortex. RESULTS: Following infection with TMEV, a picornavirus, the Aß0.ß2m0 mice lacking active immune responses died within 18 to 21 days post-infection. These mice showed severe encephalomyelitis due to rapid replication of the viral genome. In contrast, transgenic Hß2m mice with insertion of a single human class I MHC gene in the absence of human or mouse class II survived the acute infection. Both A11+ and B27+ mice significantly controlled virus RNA expression by 45 days and did not develop late-onset spinal cord demyelination. By 45 days post-infection (DPI), B27+ transgenic mice showed almost complete repair of the virus-induced brain injury, but A11+ mice conversely showed persistent severe hippocampal and cortical injury. CONCLUSIONS: The findings support the hypothesis that the expression of a single human class I MHC molecule, independent of persistent virus infection, influences the extent of sub frequent chronic neuronal injury or repair in the absence of a class II MHC immune response.


Assuntos
Infecções por Cardiovirus/patologia , Sistema Nervoso Central/patologia , Sistema Nervoso Central/virologia , Antígenos de Histocompatibilidade Classe I/metabolismo , Theilovirus/fisiologia , Análise de Variância , Animais , Anticorpos/metabolismo , Modelos Animais de Doenças , Citometria de Fluxo , Antígeno HLA-A11/metabolismo , Antígeno HLA-B27/metabolismo , Antígenos de Histocompatibilidade Classe I/genética , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , RNA Mensageiro , Proteínas Virais/genética , Proteínas Virais/imunologia , Proteínas Virais/metabolismo
5.
J Neurochem ; 134(5): 865-78, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25866077

RESUMO

CNS regeneration is a desirable goal for diseases of brain and spinal cord. Current therapeutic strategies for the treatment of multiple sclerosis (MS) aim to eliminate detrimental effects of the immune system, so far without reversing disability or affecting long-term prognosis in patients. Approachable molecular targets that stimulate CNS repair are not part of the clinical praxis or have not been identified yet. The purpose of this study was to identify the molecular target of the human monoclonal antibody HIgM12. HIgM12 reverses motor deficits in chronically demyelinated mice, a model of MS. Here, we identified polysialic acid (PSA) attached to the neural cell adhesion molecule (NCAM) as the antigen for HIgM12 by using different NCAM knockout strains and through PSA removal from the NCAM protein core. Antibody binding to CNS tissue and primary cells, antibody-mediated cell adhesion, and neurite outgrowth on HIgM12-coated nitrocellulose was detected only in the presence of PSA as assessed by western blotting, immunoprecipitation, immunocytochemistry, and histochemistry. We conclude that HIgM12 mediates its in vivo and in vitro effects through binding to PSA and has the potential to be an effective therapy for MS and neurodegenerative diseases. The human antibody HIgM12 stimulates neurite outgrowth in vitro and promotes function in chronically demyelinated mice, a model of multiple sclerosis. The cellular antigen for HIgM12 was undetermined. Here, we identified polysialic acid attached to NCAM (neural cell adhesion molecule) as the cellular target for HIgM12. This includes glial fibrillary acidic protein (GFAP)-positive mouse astrocytes (GFAP, red; HIgM12, green; DAPI, blue) among other cell types of the central nervous system. These findings indicate a new strategy for the treatment of neuro-motor disorders including multiple sclerosis.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Antígenos/imunologia , Antígeno CD56/imunologia , Doenças Autoimunes Desmielinizantes do Sistema Nervoso Central/tratamento farmacológico , Esclerose Múltipla/tratamento farmacológico , Doenças Neurodegenerativas/tratamento farmacológico , Ácidos Siálicos/imunologia , Animais , Anticorpos Monoclonais/imunologia , Especificidade de Anticorpos , Reações Antígeno-Anticorpo , Antígeno CD56/química , Antígeno CD56/genética , Adesão Celular , Células Cultivadas , Cerebelo/citologia , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Glicosilação/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Esclerose Múltipla/imunologia , Regeneração Nervosa , Neuraminidase/farmacologia , Neuritos/efeitos dos fármacos , Doenças Neurodegenerativas/imunologia , Neuroglia/citologia , Neurônios/efeitos dos fármacos , Neurônios/imunologia , Neurônios/ultraestrutura , Ratos , Ratos Sprague-Dawley
6.
J Neuroinflammation ; 12: 83, 2015 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-25924771

RESUMO

BACKGROUND: Intracerebral infection of susceptible mouse strains with Theiler's murine encephalomyelitis virus (TMEV) results in chronic demyelinating disease with progressive axonal loss and neurologic dysfunction similar to progressive forms of multiple sclerosis (MS). We previously showed that as the disease progresses, a marked decrease in brainstem N-acetyl aspartate (NAA; metabolite associated with neuronal integrity) concentrations, reflecting axon health, is measured. We also demonstrated stimulation of neurite outgrowth by a neuron-binding natural human antibody, IgM12. Treatment with either the serum-derived or recombinant human immunoglobulin M 12 (HIgM12) preserved functional motor activity in the TMEV model. In this study, we examined IgM-mediated changes in brainstem NAA concentrations and central nervous system (CNS) pathology. FINDINGS: (1)H-magnetic resonance spectroscopy (MRS) showed that treatment with HIgM12 significantly increased brainstem NAA concentrations compared to controls in TMEV-infected mice. Pathologic analysis demonstrated a significant preservation of axons in the spinal cord of animals treated with HIgM12. CONCLUSIONS: This study links drug efficacy of slowing deficits with axon preservation and NAA concentrations in the brainstem in a model of progressive MS. HIgM12-mediated changes of NAA concentrations in the brainstem are a surrogate marker of axon injury/preservation throughout the spinal cord. This study provides proof-of-concept that a neuron-reactive human IgM can be therapeutic and provides a biomarker for clinical trials.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Ácido Aspártico/análogos & derivados , Tronco Encefálico/metabolismo , Glicoproteínas de Membrana/imunologia , Esclerose Múltipla/tratamento farmacológico , Esclerose Múltipla/patologia , Proteínas do Envelope Viral/imunologia , Animais , Ácido Aspártico/metabolismo , Axônios/efeitos dos fármacos , Encéfalo/patologia , Tronco Encefálico/efeitos dos fármacos , Modelos Animais de Doenças , Humanos , Espectroscopia de Ressonância Magnética , Camundongos , Esclerose Múltipla/etiologia , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo , Medula Espinal/patologia , Estatísticas não Paramétricas , Theilovirus/imunologia
7.
Sci Rep ; 14(1): 19561, 2024 08 22.
Artigo em Inglês | MEDLINE | ID: mdl-39174596

RESUMO

There is a critical need to non-invasively assess the PD-L1 expression in tumors as a predictive biomarker for determining the efficacy of anti-PD-1/PD-L1 immunotherapies. Non-invasive imaging modality like positron emission tomography (PET) can be a powerful tool to assess the PD-L1 expression in the whole body including multiple metastases as a patient selection criterion for the anti-PD-1/PD-L1 immunotherapy. In this study, we synthesized B11-nanobody, B11-scFv and B11-diabody fragments from the full-length anti-PD-L1 B11 IgG. Out of the three antibody fragments, B11-diabody showed higher nM affinity towards PD-L1 antigen as compared to B11-scFv and B11-nanobody. All three antibody fragments were successfully radiolabeled with 64Cu, a PET radioisotope. For radiolabeling, the antibody fragments were first conjugated with p-SCN-Bn-NOTA followed by chelation with 64Cu. All three radiolabeled antibody fragments were found to be stable in mouse and human sera for up to 24 h. Additionally, all three [64Cu]Cu-NOTA-B11-antibody fragments were evaluated in PD-L1 negative and human PD-L1 expressing cancer cells and subcutaneous tumor models. Based on the results, [64Cu]Cu-NOTA-B11-diabody has potential to be used as a PET imaging probe for assessing PD-L1 expression in tumors as early as 4 h post-injection, allowing faster assessment compared to the full length IgG based PET imaging probe.


Assuntos
Antígeno B7-H1 , Neoplasias da Mama , Tomografia por Emissão de Pósitrons , Tomografia por Emissão de Pósitrons/métodos , Antígeno B7-H1/metabolismo , Antígeno B7-H1/imunologia , Animais , Humanos , Feminino , Camundongos , Neoplasias da Mama/diagnóstico por imagem , Neoplasias da Mama/imunologia , Linhagem Celular Tumoral , Melanoma/diagnóstico por imagem , Melanoma/imunologia , Melanoma/metabolismo , Anticorpos de Cadeia Única/imunologia , Radioisótopos de Cobre , Fragmentos de Imunoglobulinas/imunologia
8.
Blood ; 117(7): 2257-64, 2011 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-21131590

RESUMO

Acquired hemophilia is a rare bleeding disorder characterized by the spontaneous occurrence of inhibitory antibodies against endogenous factor VIII (FVIII). IgG from some patients with acquired hemophilia hydrolyze FVIII. Because of the complex etiology of the disease, no clinical parameter, including the presence of FVIII-hydrolyzing IgG, has been associated with patient's survival or death. Here, we demonstrate the presence of anti-FIX antibodies in acquired hemophilia patients. IgG from some patients were found to hydrolyze FIX. In most cases, IgG-mediated FIX-hydrolysis resulted in FIX activation. IgG-mediated hydrolysis of FIX thus led to the significant generation of activated FIX in 25 of 65 patients. Based on the estimated kinetic parameters, patients' IgG activated up to 0.3nM FIX in 24 hours, an amount that restored thrombin generation in vitro provided the presence of more than or equal to 3% residual FVIII activity in plasma. This work identifies proteolytic IgG as novel molecules able to activate FIX under pathologic conditions. IgG-mediated FIX activation is a prevalent phenomenon among acquired hemophilia patients. The presence of FIX-activating IgG may partly compensate for the antibody-mediated inhibition of endogenous FVIII in restoring thrombin generation. This clinical trial was registered at www.clinicaltrials.gov as #NCT00213473.


Assuntos
Autoanticorpos/sangue , Fator IX/imunologia , Fator IX/metabolismo , Imunoglobulina G/sangue , Adulto , Idoso , Idoso de 80 Anos ou mais , Fator VIII/imunologia , Fator VIII/metabolismo , Feminino , Hemofilia A/sangue , Hemofilia A/imunologia , Humanos , Hidrólise , Técnicas In Vitro , Cinética , Masculino , Pessoa de Meia-Idade , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/metabolismo
9.
Proc Natl Acad Sci U S A ; 107(30): 13526-31, 2010 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-20624970

RESUMO

Tissue kallikrein (TK) is a serine protease synthetized in renal tubular cells located upstream from the collecting duct where renal potassium balance is regulated. Because secretion of TK is promoted by K+ intake, we hypothesized that this enzyme might regulate plasma K+ concentration ([K+]). We showed in wild-type mice that renal K+ and TK excretion increase in parallel after a single meal, representing an acute K+ load, whereas aldosterone secretion is not modified. Using aldosterone synthase-deficient mice, we confirmed that the control of TK secretion is aldosterone-independent. Mice with TK gene disruption (TK-/-) were used to assess the impact of the enzyme on plasma [K+]. A single large feeding did not lead to any significant change in plasma [K+] in TK+/+, whereas TK-/- mice became hyperkalemic. We next examined the impact of TK disruption on K+ transport in isolated cortical collecting ducts (CCDs) microperfused in vitro. We found that CCDs isolated from TK-/- mice exhibit net transepithelial K+ absorption because of abnormal activation of the colonic H+,K+-ATPase in the intercalated cells. Finally, in CCDs isolated from TK-/- mice and microperfused in vitro, the addition of TK to the perfusate but not to the peritubular bath caused a 70% inhibition of H+,K+-ATPase activity. In conclusion, we have identified the serine protease TK as a unique kalliuretic factor that protects against hyperkalemia after a dietary K+ load.


Assuntos
Adaptação Fisiológica/fisiologia , Rim/fisiologia , Potássio/metabolismo , Calicreínas Teciduais/metabolismo , Adaptação Fisiológica/efeitos dos fármacos , Aldosterona/metabolismo , Aldosterona/urina , Animais , Transporte Biológico , Citocromo P-450 CYP11B2/deficiência , Citocromo P-450 CYP11B2/genética , ATPase Trocadora de Hidrogênio-Potássio/genética , ATPase Trocadora de Hidrogênio-Potássio/metabolismo , Concentração de Íons de Hidrogênio , Técnicas In Vitro , Rim/metabolismo , Túbulos Renais Coletores/metabolismo , Túbulos Renais Coletores/fisiologia , Camundongos , Camundongos Knockout , Potássio/sangue , Potássio/urina , Potássio na Dieta/administração & dosagem , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sódio/metabolismo , Calicreínas Teciduais/genética
10.
Anal Chem ; 84(14): 6031-9, 2012 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-22762372

RESUMO

Multiple sclerosis is a progressive neurological disorder that results in the degradation of myelin sheaths that insulate axons in the central nervous system. Therefore promotion of myelin repair is a major thrust of multiple sclerosis treatment research. Two mouse monoclonal natural autoantibodies, O1 and O4, promote myelin repair in several mouse models of multiple sclerosis. Natural autoantibodies are generally polyreactive and predominantly of the IgM isotype. The prevailing paradigm is that because they are polyreactive, these antibodies bind antigens with low affinities. Despite their wide use in neuroscience and glial cell research, however, the affinities and kinetic constants of O1 and O4 antibodies have not been measured to date. In this work, we developed a membrane biosensing platform based on surface plasmon resonance in gold nanohole arrays with a series of surface modification techniques to form myelin-mimicking lipid bilayer membranes to measure both the association and dissociation rate constants for O1 and O4 antibodies binding to their myelin lipid antigens. The ratio of rate constants shows that O1 and O4 bind to galactocerebroside and sulfated galactocerebroside, respectively, with unusually small apparent dissociation constants (K(D) ≈ 0.9 nM) for natural autoantibodies. This is approximately one to 2 orders of magnitude lower than typically observed for the highest affinity natural autoantibodies. We propose that the unusually high affinity of O1 and O4 to their targets in myelin contributes to the mechanism by which they signal oligodendrocytes and induce central nervous system repair.


Assuntos
Autoanticorpos/metabolismo , Materiais Biomiméticos/metabolismo , Bicamadas Lipídicas/metabolismo , Bainha de Mielina/metabolismo , Nanotecnologia/métodos , Ressonância de Plasmônio de Superfície/métodos , Animais , Materiais Biomiméticos/química , Imunoglobulina M/metabolismo , Bicamadas Lipídicas/química , Camundongos , Bainha de Mielina/fisiologia , Oligodendroglia/metabolismo
11.
J Autoimmun ; 37(2): 144-50, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21624820

RESUMO

Immunoglobulins (Ig) or antibodies are heavy plasma proteins, with sugar chains added to amino acid residues by N-linked glycosylation and occasionally by O-linked glycosylation. The versatility of antibodies is demonstrated by the various functions that they mediate such as neutralization, agglutination, fixation with activation of complement and activation of effector cells. In addition to this plethora of functions, some antibodies express enzymatic activity. Antibodies endowed with enzymatic properties have been described in human autoimmune manifestations in a variety of disorders such as autoimmune thyroiditis, systemic erythematosus (SLE), scleroderma, rheumatoid arthritis (RA), multiple sclerosis (MS) and acquired hemophilia (AH). Antibodies isolated from these conditions were able to specifically hydrolyze thyroglobulin, DNA, RNA, myelin basic protein (MBP), and factor VIII (FVIII) or factor IX (FIX), respectively. The therapeutic relevance of these findings is discussed.


Assuntos
Anticorpos Catalíticos/metabolismo , Autoanticorpos/metabolismo , Autoantígenos/metabolismo , Doenças Autoimunes/enzimologia , Doenças Autoimunes/imunologia , Animais , Anticorpos Catalíticos/imunologia , Autoanticorpos/imunologia , Autoantígenos/imunologia , Doenças Autoimunes/tratamento farmacológico , Humanos , Hidrólise , Imunoterapia/tendências
12.
J Autoimmun ; 36(1): 9-15, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20970960

RESUMO

Common variable immunodeficiency (CVID) is associated with low serum immunoglobulin concentrations and an increased susceptibility to infections and autoimmune diseases. The treatment of choice for CVID patients is replacement intravenous immunoglobulin (IVIg) therapy. IVIg has been beneficial in preventing or alleviating the severity of infections and autoimmune and inflammatory process in majority of CVID patients. Although the mechanisms of action of IVIg given as 'therapeutic high dose' in patients with autoimmune diseases are well studied, the underlying mechanisms of beneficial effects of IVIg in primary immunodeficiencies are not completely understood. Therefore we investigated the effect of 'replacement dose' of IVIg by probing its action on B cells from CVID patients. We demonstrate that IVIg at low doses induces proliferation and immunoglobulin synthesis from B cells of CVID patients. Interestingly, B cell stimulation by IVIg is not associated with induction of B cell effector cytokine IFN-γ and of transcription factor T-bet. Together, our results indicate that in some CVID patients, IVIg rectifies the defective signaling of B cells normally provided by T cells and delivers T-independent signaling for B cells to proliferate. IVIg 'replacement therapy' in primary immunodeficiencies is therefore not a merepassive transfer of antibodies to prevent exclusively the recurrent infections; rather it has an active role in regulating autoimmune and inflammatory responses through modulating B cell functions and thus imposing dynamic equilibrium of the immune system.


Assuntos
Formação de Anticorpos/efeitos dos fármacos , Linfócitos B/efeitos dos fármacos , Imunodeficiência de Variável Comum/tratamento farmacológico , Imunoglobulinas Intravenosas/farmacologia , Adulto , Idoso , Linfócitos B/imunologia , Proliferação de Células/efeitos dos fármacos , Separação Celular , Imunodeficiência de Variável Comum/imunologia , Feminino , Citometria de Fluxo , Humanos , Masculino , Pessoa de Meia-Idade , Adulto Jovem
13.
Oncoimmunology ; 9(1): 1744980, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32363112

RESUMO

ADAM10 and ADAM17 expression and soluble PD-L1 (sPD-L1) predict poor prognosis in many malignancies, including in patients treated with PD-(L)1 inhibitors. The mechanism of soluble PD-L1 production and its effects are unknown. Here we uncover a novel mechanism of ADAM10- and ADAM17-mediated resistance to PD-(L)1 inhibitors. ADAM10 and ADAM17 cleave PD-L1 from the surface of malignant cells and extracellular vesicles. This cleavage produces an active sPD-L1 fragment that induces apoptosis in CD8 + T cells and compromises the killing of tumor cells by CD8 + T cells. Reduced tumor site PD-L1 protein-to-mRNA ratios predict poor outcomes and are correlated with elevated ADAM10 and ADAM17 expression in multiple cancers. These results may explain the discordance between PD-L1 immunohistochemistry and PD-(L)1 inhibitor response. Thus, including ADAM10 and ADAM17 tissue staining may improve therapy selection. Furthermore, treatment with an ADAM10/ADAM17 inhibitor may abrogate PD-(L)1 inhibitor resistance and improve clinical responses to PD-(L)1 immunotherapy.


Assuntos
Secretases da Proteína Precursora do Amiloide , Antígeno B7-H1 , Proteína ADAM10 , Proteína ADAM17 , Apoptose , Antígeno B7-H1/genética , Humanos , Proteínas de Membrana/genética
14.
Haematologica ; 93(1): 83-9, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18166789

RESUMO

BACKGROUND: The development of factor VIII (FVIII) inhibitors remains the major hurdle in the clinical management of patients with hemophilia A. FVIII uptake by professional antigen-presenting cells (APC) is the first step involved in initiation of immune responses to FVIII. Studies on FVIII catabolism have highlighted the role played by CD91/LRP as a potential target for increasing FVIII half-life in patients and prolonging treatment efficiency. We investigated the involvement of CD91 in FVIII endocytosis by human dendritic cells (DC), a model of professional APC. DESIGN AND METHODS: Immature DC were generated from circulating monocytes from healthy donors. Surface expression of CD91 was assessed by flow cytometry. Uptake of fluorescein isothiocyanate-conjugated ligands by immature DC was studied in the presence of various blocking agents. RESULTS: CD91 was expressed on approximately 20% of DC and mediated the internalization of its model ligand, alpha2-macroglobulin. DC internalized FVIII and activated a human FVIII-specific T-cell clone in a dose-dependent manner. FVIII uptake by DC and subsequent T-cell activation were not inhibited by receptor-associated protein. CONCLUSIONS: Our results indicate that CD91 and other members of the LDL receptor family are not strongly implicated in FVIII internalization by monocyte-derived DC, and suggest the involvement of alternative divalent ion-dependent endocytic receptors.


Assuntos
Antígenos CD/biossíntese , Células Dendríticas/citologia , Fator VIII/biossíntese , Linfócitos T/citologia , Animais , Separação Celular , Endocitose , Fator VIII/metabolismo , Hemofilia A/genética , Humanos , Leucócitos Mononucleares/citologia , Ligantes , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade , Ativação Linfocitária , Camundongos , Monócitos/citologia , Monócitos/metabolismo
15.
Expert Opin Biol Ther ; 18(5): 545-560, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29460650

RESUMO

INTRODUCTION: Multiple sclerosis (MS) is a chronic and progressive inflammatory demyelinating disease of the human central nervous system (CNS) and is the most common disabling neurological condition in young adults, resulting in severe neurological defects. No curative or long-term progression-inhibiting therapy has yet been developed. However, recent investigation has revealed potential strategies that do not merely modulate potentially pathogenic autoimmune responses, but stimulate remyelination within CNS lesions. AREAS COVERED: We discuss the history and development of natural human IgM-isotype immunoglobulins (HIgMs) and recently-identified aptamer-conjugates that have been shown to enhance endogenous myelin repair in animal models of demyelination by acting on myelin-producing oligodendrocytes (OLs) or oligodendrocyte progenitor cells (OPCs) within CNS lesions. We also discuss future development aims and applications for these important novel technologies. EXPERT OPINION: Aptamer conjugate Myaptavin-3064 and recombinant human IgM-isotype antibody rHIgM22 regenerate CNS myelin, thereby reducing axonal degeneration and offering the potential of recovery from MS relapses, reversal of disability and prevention of disease progression. Advancement of these technologies into the clinic for MS treatment is therefore a top priority. It remains unclear to what extent the therapeutic modalities of remyelinating antibodies and aptamers may synergize with other currently-approved therapies to yield enhanced therapeutic effects.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Produtos Biológicos/uso terapêutico , Doenças do Sistema Nervoso Central/tratamento farmacológico , Imunoconjugados/uso terapêutico , Remielinização/efeitos dos fármacos , Adulto , Animais , Aptâmeros de Peptídeos/uso terapêutico , Doenças Desmielinizantes/tratamento farmacológico , Humanos , Esclerose Múltipla/tratamento farmacológico , Regeneração/efeitos dos fármacos , Remielinização/fisiologia , Adulto Jovem
16.
Thromb Haemost ; 98(1): 138-47, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17598006

RESUMO

The development of antibodies directed against factor VIII (FVIII) represents a major hurdle in the treatment of hemophilia A. Most anti-FVIII antibodies are identified through their ability to inhibit the FVIII procoagulant activity. Many of them, however, do not interfere with the functional properties of FVIII. Antibodies directed against the B domain belong to this latter category. Here, we characterized B domain-specific human monoclonal Abs (mAbs) at the molecular level. A series of human mAbs directed against FVIII was produced upon immunization of transgenic XenoMouse mice with human recombinant FVIII (rFVIII). Selection of the hybridoma with epitope specificity for the B domain was performed by differential recognition of full-length and B domain-deleted rFVIII. None of the anti-B domain mAbs demonstrated inhibitory activity against FVIII. Three of the mAbs recognized linear epitopes: mAb 25H3 bound to the (1014)HIDGPSLLIEN(1024) sequence; mAbs 8E3 and 22B6 shared the same epitope, composed of residues (1534)KWNEANR(1540). The corresponding soluble peptides inhibited the binding of their respective mAbs to FVIII. mAbs 8E3 and 22B6 displaced the binding of FVIII to vonWillebrand factor. Moreover, some of them (in particular mAbs 4G6 and 8E3) were able to compete for binding to the B domain with the anti-FVIII Abs from hemophilia A patients without inhibitor or with low Bethesda titers. Further investigation will allow to better characterize their clinical relevance.


Assuntos
Anticorpos Monoclonais/imunologia , Epitopos , Fator VIII/imunologia , Adolescente , Adulto , Idoso , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/biossíntese , Anticorpos Monoclonais/farmacologia , Ligação Competitiva , Criança , Pré-Escolar , Fator VIII/metabolismo , Hemofilia A/imunologia , Humanos , Hibridomas/imunologia , Lactente , Camundongos , Camundongos Transgênicos , Pessoa de Meia-Idade , Fragmentos de Peptídeos/imunologia , Fator de von Willebrand/metabolismo
17.
Immunol Lett ; 110(1): 23-8, 2007 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-17467813

RESUMO

A number of diseases are treated by passive administration of human proteins. Human coagulation factor VIII (FVIII) is one such protein which is administered to hemophilia A patients in order to manage and treat hemorrhagic incidences. This mode of therapy suffers from the side effect of generating anti-FVIII antibodies (inhibitors) which neutralizes the function of the infused FVIII. At a time when efficient viral screening procedures are at place, development of inhibitors poses the greatest threat to such a therapy. Various predisposing factors, both patient and product-related, are responsible for the development of inhibitory antibodies. A proper understanding of these "risk-factors" would eventually help to better design therapeutic regimen to tackle hemophilia A.


Assuntos
Anticorpos/sangue , Fator VIII/imunologia , Hemofilia A/tratamento farmacológico , Animais , Anticorpos/imunologia , Fator VIII/efeitos adversos , Fator VIII/uso terapêutico , Hemofilia A/imunologia , Humanos , Fatores de Risco , Resultado do Tratamento
18.
Hum Antibodies ; 25(3-4): 121-129, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28269761

RESUMO

A single peripheral dose of CNS-binding IgMs promote remyelination and preserve axons in a number of animal models of neurologic disease. A myelin-binding recombinant human IgM (rHIgM22) is presently in a safety trial in MS patients following an acute MS exacerbation. rHIgM22 (directed against oligodendrocytes) or rHIgM12 (directed against neurons) were administered to mice with MOG-induced experimental autoimmune encephalomyelitis (EAE) with study endpoints: clinical deficits and brain and spinal cord pathology. IgMs were administered at a therapeutic dose of 100 µ g intra peritoneal at the time of immunization (day -1, 0, +$1), disease onset (15 days) or peak of the disease (28 days). Disease course was not worsened by either human IgM regardless of the time of treatment. Of note, the human IgM that recognizes a carbohydrate epitope on gangliosides and NCAM, rHIgM12, reduced brain pathology when given at time of immunization or at onset of disease, but did not reduce clinical deficits or spinal cord disease burden. Hence, treatment with rHIgM12 resulted in marked reduction in meningeal inflammation. Data consistent with the hypothesis that in the EAE model this molecule has an immune-modulatory effect. Treatment with an anti-CD4 blocking IgG prevented both clinical course and CNS pathology. This pre-clinical study further supports the safety of therapeutic CNS-binding human IgMs in the presence of autoimmunity and clearly differentiates them from IgGs directed against MOG or aquaporin-4 that worsen neurologic disease.


Assuntos
Disfunção Cognitiva/tratamento farmacológico , Doenças Desmielinizantes/tratamento farmacológico , Encefalomielite Autoimune Experimental/tratamento farmacológico , Imunoglobulina M/farmacologia , Fatores Imunológicos/farmacologia , Molécula L1 de Adesão de Célula Nervosa/imunologia , Fármacos Neuroprotetores/farmacologia , Ácidos Siálicos/imunologia , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/imunologia , Encéfalo/patologia , Disfunção Cognitiva/induzido quimicamente , Disfunção Cognitiva/imunologia , Disfunção Cognitiva/patologia , Doenças Desmielinizantes/induzido quimicamente , Doenças Desmielinizantes/imunologia , Doenças Desmielinizantes/patologia , Esquema de Medicação , Encefalomielite Autoimune Experimental/induzido quimicamente , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/patologia , Feminino , Adjuvante de Freund/administração & dosagem , Humanos , Injeções Intraperitoneais , Camundongos , Camundongos Endogâmicos C57BL , Bainha de Mielina/efeitos dos fármacos , Bainha de Mielina/imunologia , Bainha de Mielina/patologia , Glicoproteína Mielina-Oligodendrócito/administração & dosagem , Molécula L1 de Adesão de Célula Nervosa/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/imunologia , Neurônios/patologia , Oligodendroglia/efeitos dos fármacos , Oligodendroglia/imunologia , Oligodendroglia/patologia , Fragmentos de Peptídeos/administração & dosagem , Ligação Proteica , Proteínas Recombinantes/farmacologia , Ácidos Siálicos/metabolismo , Medula Espinal/efeitos dos fármacos , Medula Espinal/imunologia , Medula Espinal/patologia
19.
Immunol Lett ; 103(1): 3-7, 2006 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-16427704

RESUMO

Immunoglobulins have initially been illustrated as proteins produced by the immune system for binding and neutralizing foreign molecules potentially harmful to the organism. The number of V(H), D(H), J(H), V(L) and J(L) genes that encode the variable regions of immunoglobulins and the junctional diversity that occurs at the time of somatic rearrangement determine the extent of the repertoire of antibodies that may be potentially produced by an organism. This potential repertoire includes antibodies the antigen binding site of which may recognize external as well as autologous antigens, or may structurally resemble the active site of enzymes and be endowed with enzymatic activity. Under physiological conditions, B cell clones that produce antibodies naturally endowed with catalytic activity are negatively regulated and subjected to apoptosis. Catalytic antibodies are expressed only following active immunization, or if the physiological regulatory mechanisms that control the expression of catalytic antibody-producing B cell clones are perturbed, e.g. in the context of pregnancy or in the course of autoimmune diseases.


Assuntos
Anticorpos Catalíticos , Doenças Autoimunes/imunologia , Doenças Autoimunes/fisiopatologia , Doença de Hashimoto/imunologia , Doença de Hashimoto/fisiopatologia , Humanos , Imunização
20.
Blood Coagul Fibrinolysis ; 17(4): 229-34, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16651863

RESUMO

Antibodies that are able to catalyze the antigen for which they are specific are produced spontaneously by the immune system. Catalytic immunoglobulins (Igs) both of the IgM and IgG isotypes have been detected in the serum of healthy donors, where they have been proposed to participate in the removal of metabolic waste and in the defense of the organism against invading pathogens. Conversely, antigen-specific hydrolytic IgG have been reported in a number of inflammatory, autoimmune and neoplastic disorders: their pathogenic effects have been demonstrated occasionally. The pathophysiological relevance of catalytic antibodies thus remains an elusive issue. Through the description of the pro-coagulation factor VIII as a model target antigen for catalytic antibodies, we propose that catalytic antibodies have either a beneficial or a deleterious role depending on the physiopathological context. Physiology thus relies on a delicate equilibrium between the levels of soluble target antigen and that of antigen-specific hydrolyzing immunoglobulins. Indeed, in patients with hemophilia A, in whom endogenous factor VIII is deficient or missing and exogenous factor VIII needs to be administered to treat hemorrhagic events, the development of factor VIII-hydrolyzing IgG that inactivate the therapeutically administered factor VIII, may reveal deleterious. In contrast, in a situation in which excess factor VIII may be detrimental and lead to excessive coagulation, disseminated thrombosis and organ ischemia, as seen in severe sepsis, our recent data suggest that the presence of factor VIII-hydrolyzing IgG may be beneficial to the patient.


Assuntos
Anticorpos Catalíticos/imunologia , Fator VIII/imunologia , Hemofilia A/fisiopatologia , Imunoglobulina G/imunologia , Complexo Antígeno-Anticorpo , Hemofilia A/imunologia , Humanos , Hidrólise
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA