Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
J Autoimmun ; 135: 102983, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36640636

RESUMO

Myasthenia gravis (MG) is a debilitating autoimmune disease characterized by muscle fatigue and weakness caused by autoantibody- and complement-mediated damage to the neuromuscular junction. This study sought to compare the efficacy of unique sets of monoclonal antibody-siRNA conjugates, individually (mono) or in combination (duo), against the crucial receptors predominantly or solely expressed on two subsets of B cells-plasma B cells and their precursor (transitional mature B) cells in a mouse model of MG. At the optimized doses, the conjugates, likely due to the combined activities of mAb and siRNA, substantially decreased the expression levels of CD268 (B cell-activating factor receptor) in mature B cells and CD269 (B-cell maturation antigen) in plasma cells concomitantly with reducing the levels of acetylcholine receptor (AChR)-specific autoantibodies. PEGylation, but not pretreatment with an antibody against type 1 interferon receptor, further improved duoconjugate-induced reduction in the autoantibody levels. Our results show that the duoconjugate treatment significantly improved the clinical symptoms of MG, consistent with the preservation of bungarotoxin-bound functional AChRs. In the future, developing similar target-specific combination molecules can potentially turn into a new and effective therapeutic approach for MG.


Assuntos
Miastenia Gravis Autoimune Experimental , Camundongos , Animais , RNA Interferente Pequeno , Receptores Colinérgicos , Anticorpos Monoclonais , Autoanticorpos
2.
Virol J ; 17(1): 128, 2020 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-32831108

RESUMO

BACKGROUND: Heterozygosity at HLA class I loci is generally considered beneficial for host defense. We report here an element of HLA class I homozygosity that may or may not help preserve its existence in populations but which could indicate a new avenue for antiviral research. METHODS: Lymphocytes from serologically HLA-homozygous or -heterozygous donors were examined for synthesis of influenza virus proteins and RNA after exposure to virus as peripheral blood mononuclear cells. The virus-exposed lymphocytes were also examined for internalization of the virus after exposure, and for susceptibility to virus-specific cytotoxic T lymphocytes in comparison with virus-exposed monocytes/macrophages and unseparated peripheral blood mononuclear cells. Results were compared using two-tailed Fisher's exact test. RESULTS: Serologically-defined HLA-A2-homozygous lymphocytes, in contrast to heterozygous lymphocytes, did not synthesize detectable influenza virus RNA or protein after exposure to the virus. HLA-A2-homozygous lymphocytes, including both homozygous and heterozygous donors by genetic sequence subtyping, did internalize infectious virus but were not susceptible to lysis by autologous virus-specific cytotoxic T lymphocytes ("fratricide"). Similar intrinsic resistance to influenza virus infection was observed with HLA-A1- and HLA-A11-homozygous lymphocytes and with HLA-B-homozygous lymphocytes. CONCLUSIONS: A significant proportion of individuals within a population that is characterized by common expression of HLA class I alleles may possess lymphocytes that are not susceptible to influenza virus infection and thus to mutual virus-specific lysis. Further study may identify new approaches to limit influenza virus infection.


Assuntos
Genes MHC Classe I/imunologia , Influenza Humana/genética , Influenza Humana/imunologia , Macrófagos/virologia , Linfócitos T Citotóxicos/imunologia , Alelos , Feminino , Antígeno HLA-A1/imunologia , Antígeno HLA-A11/imunologia , Antígeno HLA-A2/imunologia , Homozigoto , Humanos , Leucócitos Mononucleares/virologia , Macrófagos/imunologia , Masculino
3.
Clin Immunol ; 176: 122-130, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28099879

RESUMO

We investigated potential therapeutic effects of a conjugate of BAFF receptor specific-monoclonal antibody and short interference RNA in a mouse model of myasthenia gravis (EAMG). Whereas high-dose siRNA conjugate resulted in significant accumulation of Fas expressing CD19+/B220+ cells and concurrent expression of type 1 interferon in lymph nodes, low-dose conjugate did not induce FAS expression but caused marked BAFF receptor deficiency in lymph nodes that was further associated with improved MG symptoms. Unexpectedly, despite inhibiting BAFF receptor significantly in PBMCs and secondary lymphoid organs, conjugate treatment did not reduce the levels of autoantibody. Rather, at high dose, it caused robust increase in high affinity anti-AChR antibody and increased levels of serum IL10 and IL-4 cytokines. Our findings reveal a previously undocumented, dose dependent, immunomodulatory distant effect resulting from BAFF receptor specific mAb-siRNA conjugate treatment in an in vivo model of autoimmune disease.


Assuntos
Anticorpos Monoclonais/imunologia , Autoanticorpos/imunologia , Receptor do Fator Ativador de Células B/imunologia , Linfócitos B/imunologia , Linfonodos/imunologia , Miastenia Gravis/imunologia , RNA Interferente Pequeno/imunologia , Receptor fas/imunologia , Animais , Antígenos CD19/imunologia , Autoanticorpos/sangue , Linhagem Celular , Modelos Animais de Doenças , Interferon gama/imunologia , Interleucina-10/imunologia , Interleucina-4/imunologia , Antígenos Comuns de Leucócito/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Miastenia Gravis/sangue , Ratos , Receptores Colinérgicos/imunologia
4.
Clin Immunol ; 151(2): 155-63, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24589747

RESUMO

Myasthenia gravis (MG) is an autoimmune disease characterized by muscle weakness associated with acetylcholine receptor (AChR), muscle-specific receptor kinase (MuSK) or low-density lipoprotein receptor-related protein 4 (LRP4)-antibodies. MuSK-antibodies are predominantly of the non-complement fixing IgG4 isotype. The MuSK associated experimental autoimmune myasthenia gravis (EAMG) model was established in mice to investigate immunoglobulin (Ig) and cytokine responses related with MuSK immunity. C57BL/6 (B6) mice immunized with 30µg of recombinant human MuSK in incomplete or complete Freund's adjuvant (CFA) showed significant EAMG susceptibility (>80% incidence). Although mice immunized with 10µg of MuSK had lower EAMG incidence (14.3%), serum MuSK-antibody levels were comparable to mice immunized with 30µg MuSK. While MuSK immunization stimulated production of all antibody isotypes, non-complement fixing IgG1 was the dominant anti-MuSK Ig isotype in both sera and neuromuscular junctions. Moreover, MuSK immunized IgG1 knockout mice showed very low serum MuSK-antibody levels. Sera and MuSK-stimulated lymph node cell supernatants of MuSK immunized mice showed significantly higher levels of IL-4 and IL-10 (but not IFN-γ and IL-12), than those of CFA immunized mice. Our results suggest that through activation of Th2-type cells, anti-MuSK immunity promotes production of IL-4, which in turn activates anti-MuSK IgG1, the mouse analog of human IgG4. These findings might provide clues for the pathogenesis of other IgG4-related diseases as well as development of disease specific treatment methods (e.g. specific IgG4 inhibitors) for MuSK-related MG.


Assuntos
Imunoglobulina G/metabolismo , Interleucina-10/metabolismo , Interleucina-4/metabolismo , Miastenia Gravis/imunologia , Receptores Proteína Tirosina Quinases/imunologia , Animais , Especificidade de Anticorpos , Células Cultivadas , Ensaio de Imunoadsorção Enzimática , Regulação da Expressão Gênica/imunologia , Imunização , Imunoglobulina G/genética , Interleucina-10/genética , Interleucina-4/genética , Linfonodos/citologia , Linfonodos/imunologia , Camundongos , Camundongos Knockout
5.
Rev Neurosci ; 25(4): 575-83, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24731953

RESUMO

While the complement system is desired for protective immunity, antibody- and complement-mediated neuromuscular junction (NMJ) destruction, a hallmark of myasthenia gravis (MG) or experimental autoimmune MG (EAMG), is a significant concern. Evidence suggests that the binding of complement factors to the pathogenic anti-acetylcholine receptor (AChR) autoantibody induces the formation of membrane attack complexes (MAC), which ultimately lead to NMJ destruction and muscle weakness. Studies corroborating the evidence show that the complement (C3-C6)-deficient or complement inhibitor (anti-C1q, soluble CR1, anti-C6, and C5 inhibiting peptide)-treated animals are highly resistant to EAMG induction, whereas the deficiency of the naturally occurring complement inhibitors, such as the decay-accelerating factor (DAF), increases EAMG susceptibility. Notably, the complement-inhibited animals do not exhibit significant immunosuppression but only a marginal reduction in the production of certain cytokines and immunoglobulin isotypes. A preliminary clinical trial using antibody-based C5 inhibitor eculizumab has been shown to be of potential use for MG treatment. The inhibition of the classic complement pathway (CCP) alone appears to be enough to suppress EAMG, suggesting that the complement inhibitors targeting specifically the classic pathway could effectively treat MG without causing immunosuppressive and other side effects. For instance, a recent non-antibody-based therapeutic approach selectively targeting the CCP component C2 by small interfering RNA (siRNA) has proven useful in EAMG treatment. The treatment strategies developed for MG might also be beneficial for other complement-mediated autoimmune diseases.


Assuntos
Inativadores do Complemento/uso terapêutico , Miastenia Gravis/terapia , Animais , Anticorpos Monoclonais/uso terapêutico , Humanos , Terapia de Imunossupressão , Miastenia Gravis/imunologia
6.
J Autoimmun ; 42: 94-104, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23410585

RESUMO

Activation of complement components is crucial in the progression and severity of myasthenia gravis and experimental autoimmune myasthenia gravis (EAMG). Mice deficient in complement component C4 or treated with monoclonal antibody to C1q are resistant to EAMG. In this study, we show that inhibition of complement cascade activation by suppressing the expression of a critical low-abundant protein, C2, in the classical complement pathway, significantly improved clinical and immunopathological manifestations of EAMG. Two weeks after a second booster immunization with acetylcholine receptor, when mice exhibit muscle weakness, i.p. injection of C2 siRNA significantly suppressed C2 mRNA in the blood cells and liver of EAMG mice. Treatment of EAMG mice with C2 siRNA, once a week for 5 weeks, significantly improved muscle strength, which was further evidenced by functional AChR preservation in muscle, reduction in number of C3 and membrane-attack complexes at neuro-muscular junctions in forelimb muscle sections, and a transient decrease in serum IgG2b levels. Our study shows for the first time that siRNA-mediated suppression of C2, a component of the classical complement system, following established disease, can effectively contribute to the remission of EAMG. Therefore, C2 siRNA mediated therapy can be applied in all complement mediated autoimmune diseases.


Assuntos
Complemento C2/antagonistas & inibidores , Fígado/imunologia , Músculos/imunologia , Miastenia Gravis Autoimune Experimental/terapia , RNA Interferente Pequeno/genética , Animais , Ativação do Complemento/genética , Complemento C2/genética , Complexo de Ataque à Membrana do Sistema Complemento/metabolismo , Feminino , Humanos , Imunoglobulina G/sangue , Camundongos , Camundongos Endogâmicos C57BL , Músculos/patologia , Receptores Colinérgicos/imunologia
7.
Front Immunol ; 14: 1110499, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36793733

RESUMO

Myasthenia gravis (MG) is a neuromuscular autoimmune disorder characterized by chronic but intermittent fatigue of the eye- and general body muscles. Muscle weakness is caused primarily by the binding of an autoantibody to the acetylcholine receptors, resulting in blockage of normal neuromuscular signal transmission. Studies revealed substantial contributions of different proinflammatory or inflammatory mediators in the pathogenesis of MG. Despite these findings, compared to therapeutic approaches that target autoantibody and complements, only a few therapeutics against key inflammatory molecules have been designed or tested in MG clinical trials. Recent research focuses largely on identifying unknown molecular pathways and novel targets involved in inflammation associated with MG. A well-designed combination or adjunct treatment utilizing one or more selective and validated promising biomarkers of inflammation as a component of targeted therapy may yield better treatment outcomes. This review briefly discusses some preclinical and clinical findings of inflammation associated with MG and current therapy approaches and suggest the potential of targeting important inflammatory marker(s) along with current monoclonal antibody or antibody fragment based targeted therapies directed to a variety of cell surface receptors.


Assuntos
Miastenia Gravis , Humanos , Miastenia Gravis/terapia , Receptores Colinérgicos , Autoanticorpos , Músculos , Inflamação
8.
J Autoimmun ; 37(2): 136-43, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21636248

RESUMO

Myasthenia gravis (MG) is a T cell-dependent and antibody-mediated disease in which the target antigen is the skeletal muscle acetylcholine receptor (AChR). In the last few decades, several immunological factors involved in MG pathogenesis have been discovered mostly by studies utilizing the experimental autoimmune myasthenia gravis (EAMG) model. Nevertheless, MG patients are still treated with non-specific global immunosuppression that is associated with severe chronic side effects. Due to the high heterogeneity of AChR epitopes and antibody responses involved in MG pathogenesis, the specific treatment of MG symptoms have to be achieved by inhibiting the complement factors and cytokines involved in anti-AChR immunity. EAMG studies have clearly shown that inhibition of the classical and common complement pathways effectively and specifically diminish the neuromuscular junction destruction induced by anti-AChR antibodies. The inborn or acquired deficiencies of IL-6, TNF-α and TNF receptor functions are associated with the lowest EAMG incidences. Th17-type immunity has recently emerged as an important contributor of EAMG pathogenesis. Overall, these results suggest that inhibition of the complement cascade and the cytokine networks alone or in combination might aid in development of future treatment models that would reduce MG symptoms with highest efficacy and lowest side effect profile.


Assuntos
Proteínas do Sistema Complemento/imunologia , Citocinas/imunologia , Miastenia Gravis/tratamento farmacológico , Miastenia Gravis/imunologia , Receptores Colinérgicos/imunologia , Animais , Autoanticorpos/imunologia , Ativação do Complemento/efeitos dos fármacos , Modelos Animais de Doenças , Humanos , Camundongos , Terapia de Alvo Molecular/tendências , Células Th17/imunologia
9.
Front Neurol ; 12: 804113, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35222229

RESUMO

Myasthenia gravis (MG) is an autoimmune disease characterized by chronic muscle fatigue and weakness caused by autoantibodies and complement-mediated damage at neuromuscular junctions. Histone deacetylases (HDACs) are crucial epigenetic regulators of proinflammatory gene expression; however, it is unclear whether HDACs modulate chronic inflammation or autoantibody production associated with MG pathogenesis. We examined expression profiles and serum levels of key inflammatory cytokines (IL-6 and IL-21) and acetylcholine receptor (AChR)-specific autoantibodies following pharmacological inhibition of key HDAC isoforms in a mouse model of MG. We found that HDAC inhibition significantly reduced the production of IL-6, but not IL-21, in AChR-stimulated PBMCs and splenocytes (n = 5 per group). Trichostatin (pan-HDAC inhibitor) treatment of MG-PBMCs (n = 2) also exhibited reduced production of induced IL-6. Although HDAC1 inhibition lowered IL-6 levels the most, HDAC2 inhibition depleted intracellular IL-6 and markedly reduced serum anti-AChR IgG2b in EAMG mice. The transcriptomic profiling and pathway mapping also revealed that autoimmunity-linked, major cell signaling pathways were differentially altered by HDAC1/2 inhibition. HDAC inhibition-mediated reduction in IL-6 and autoantibody levels also correlated with milder disease and preservation of muscle AChR in the treated mice. Overall, our findings revealed isoform-specific functional variance of HDACs in reducing inflammation and identified HDAC-regulated many genes underlying specific inflammatory and autoantibody pathways in EAMG. Thus, the study provides a rationale for further research to evaluate the HDACs or their gene targets as a potential adjunct treatment for MG.

10.
Front Immunol ; 11: 240, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32153573

RESUMO

Current therapies for myasthenia gravis (MG) are limited, and many investigations have recently focused on target-specific therapies. B cell-targeting monoclonal antibody (mAb) therapies for MG are increasingly attractive due to their specificity and efficacy. The targeted B cell biomarkers are mainly the cluster of differentiation (CD) proteins that mediate maturation, differentiation, or survival of pathogenic B cells. Additional B cell-directed therapies include non-specific peptide inhibitors that preferentially target specific B cell subsets. The primary goals of such therapies are to intercept autoantibodies and prevent the generation of an inflammatory response that contributes to the pathogenesis of MG. Treatment of patients with MG using B cell-directed mAbs, antibody fragments, or selective inhibitors have exhibited moderate to high efficacy in early studies, and some of these therapies appear to be highly promising for further drug development. Numerous other biologics targeting various B cell surface molecules have been approved for the treatment of other conditions or are either in clinical trials or preclinical development stages. These approaches remain to be tested in patients with MG or animal models of the disease. This review article provides an overview of B cell-targeted treatments for MG, including those already available and those still in preclinical and clinical development. We also discuss the potential benefits as well as the shortcomings of these approaches to development of new therapies for MG and future directions in the field.


Assuntos
Linfócitos B/imunologia , Imunoterapia/métodos , Miastenia Gravis/imunologia , Animais , Autoanticorpos/metabolismo , Humanos
11.
J Clin Neurol ; 16(3): 376-382, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32657057

RESUMO

Acute flaccid myelitis (AFM) is a sudden-onset polio-like neuromuscular disability found commonly in young children. There is an increasing incidence of confirmed AFM cases in the USA and other countries in recent years, and in association with nonpolio enterovirus infection. This represents a significant challenge to clinicians and causes significant concern to the general public. Acute flaccid paralysis (AFP) is the long-known limb paralytic syndrome caused by a viral pathogen. AFM is a subset of AFP that is also characterized by a limb paralytic condition, but it has certain distinct features such as lesions in magnetic resonance imaging of the spinal cord gray matter. AFM leads to spinal cord, brainstem, or motor neuron dysfunction. The clinical phenotypes, pathology, and patient presentation of AFM closely mimic AFP. This article provides a concise overview of our current understanding of AFM and the clinical features that distinguish AFM from AFP and similar other neurological infectious and autoimmune diseases or disorders. We also discuss the diagnosis, clinical pathology, possible pathogenetic mechanisms, and currently available therapies.

12.
Shock ; 29(5): 642-9, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-17885644

RESUMO

Methicillin-resistant Staphylococcus aureus (MRSA)-related pneumonia and/or sepsis are a frequent serious menace. The aim of the study was to establish a standardized and reproducible model of MRSA-induced septic pneumonia to evaluate new therapies. Sheep were operatively prepared for chronic study. After 5 days' recovery, tracheostomy was performed under anesthesia, and smoke injury was induced by inhalation of cotton smoke (48 breaths, <40 degrees C). Methicillin-resistant S. aureus (AW6) (approximately 2.5x10(11) colony-forming units) was instilled into the airway by a bronchoscope. After the injury, animals were awakened and maintained on mechanical ventilation by 100% oxygen for first 3 h, and thereafter, oxygen concentration was adjusted according to blood gases. The sheep were resuscitated by lactated Ringer solution with an initial rate of 2 mL kg(-1) h(-1) that was further adjusted according to hematocrit. Study groups include (1) sham (noninjured, nontreated; n=6), (2) S+MRSA (exposed to smoke inhalation and MRSA, nontreated; n=6), and (3) smoke (exposed to smoke inhalation alone; n=6). Injured (S+MRSA) animals showed the signs of severe sepsis-related multiple organ failure 3 h after insult. Cardiovascular morbidity was evidenced by severe hypotension, with increased heart rate, cardiac output, left atrial pressure and severely decreased systemic vascular resistance index, and left ventricle stroke work index. Pulmonary dysfunction was characterized by deteriorated gas exchange (PaO2/FIO2 and pulmonary shunt) and increased ventilatory pressures. The S+MRSA group showed significantly greater lung tissue water content, myeloperoxidase activity, and cytokine production compared with uninjured sham animals. Microvascular hyperpermeability was evidenced by marked fluid retention (fluid net balance), decreased plasma protein with decreased plasma oncotic pressure, and increased pulmonary microvascular pressure. All these changes were accompanied by 6- to 7-fold increase in plasma nitrite/nitrate and increased production of reactive nitrogen species in lung. The smoke inhalation alone had a little or no effect on these variables. This model closely mimics hyperdynamic human sepsis. The excessive production of NO may be extensively involved in the pathogenic process.


Assuntos
Resistência a Meticilina , Meticilina/farmacologia , Pneumonia/microbiologia , Sepse/microbiologia , Staphylococcus aureus/metabolismo , Animais , Modelos Animais de Doenças , Feminino , Hematócrito , Hemodinâmica , Pulmão/patologia , Ovinos , Lesão por Inalação de Fumaça , Fatores de Tempo , Tirosina/análogos & derivados , Tirosina/metabolismo
13.
Intensive Care Med ; 33(7): 1219-1227, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17503018

RESUMO

OBJECTIVE: To investigate ceftazidime in acute lung injury (ALI) and sepsis. DESIGN AND SETTING: Prospective, randomized, controlled animal study in an investigational ICU at a university hospital. INTERVENTIONS: Eighteen female Merino sheep were prepared for chronic study and subjected to smoke inhalation and septic challenge according to an established protocol. MEASUREMENTS AND RESULTS: Whereas global hemodynamics and oxygenation remained stable in sham animals (no injury, no treatment), the injury contributed to a hypotensive-hyperdynamic circulation in the control group (smoke inhalation and sepsis, no treatment), as indicated by a significant increase in cardiac index) and heart rate and a drop in mean arterial pressure. Treatment with ceftazidime (smoke inhalation and sepsis, treatment group) stabilized cardiac index and heart rate and attenuated the decrease in mean arterial pressure. The deterioration in PaO2/FiO2 ratio and pulmonary shunt fraction (Qs/Qt) was significantly delayed and blunted by ceftazidime. At 24 h after injury a significant increase in airway obstruction scores of bronchi and bronchioles in both injured groups was observed. Ceftazidime significantly reduced airway obstruction vs. control animals. Whereas plasma nitrate/nitrite levels increased similarly in the two injured groups, lung 3-nitrotyrosine content remained at the baseline level in the ceftazidime group. CONCLUSIONS: In ovine lung injury ceftazidime improves global hemodynamics and oxygenation not only by bacterial clearance but also via reduction in toxic nitrogen species such as 3-nitrotyrosine. Therefore ceftazidime appears as a clinically relevant adjunct in the common setting of sepsis-associated lung injury.


Assuntos
Antibacterianos/farmacologia , Ceftazidima/farmacologia , Hemodinâmica/efeitos dos fármacos , Consumo de Oxigênio/efeitos dos fármacos , Choque Séptico/fisiopatologia , Lesão por Inalação de Fumaça/fisiopatologia , Animais , Pressão Sanguínea/efeitos dos fármacos , Modelos Animais de Doenças , Feminino , Frequência Cardíaca/efeitos dos fármacos , Pulmão/química , Nitratos/sangue , Nitritos/sangue , Distribuição Aleatória , Ovinos , Tirosina/análogos & derivados , Tirosina/análise
14.
J Neuroimmunol ; 295-296: 84-92, 2016 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-27235354

RESUMO

Sera of myasthenia gravis (MG) patients with muscle-specific receptor kinase-antibody (MuSK-Ab) predominantly display the non-complement fixing IgG4 isotype. Similarly, mouse IgG1, which is the analog of human IgG4, is the predominant isotype in mice with experimental autoimmune myasthenia gravis (EAMG) induced by MuSK immunization. The present study was performed to determine whether IgG1 anti-MuSK antibody is required for immunized mice to develop EAMG. Results demonstrated a significant correlation between clinical severity of EAMG and levels of MuSK-binding IgG1+, IgG2+ and IgG3+ peripheral blood B cells in MuSK-immunized wild-type (WT) mice. Moreover, MuSK-immunized IgG1 knockout (KO) and WT mice showed similar EAMG severity, serum MuSK-Ab levels, muscle acetylcholine receptor concentrations, neuromuscular junction immunoglobulin and complement deposit ratios. IgG1 and IgG3 were the predominant anti-MuSK isotypes in WT and IgG1 KO mice, respectively. These observations demonstrate that non-IgG1 isotypes can mediate MuSK-EAMG pathogenesis.


Assuntos
Imunoglobulina G/imunologia , Miastenia Gravis Autoimune Experimental/induzido quimicamente , Miastenia Gravis Autoimune Experimental/imunologia , Receptores Proteína Tirosina Quinases/toxicidade , Animais , Antígenos CD/imunologia , Antígenos CD/metabolismo , Autoanticorpos/sangue , Linfócitos B/metabolismo , Linfócitos B/patologia , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Adjuvante de Freund/toxicidade , Imunização , Imunoglobulina G/genética , Imunoglobulina G/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miastenia Gravis Autoimune Experimental/patologia , Junção Neuromuscular/imunologia , Junção Neuromuscular/metabolismo , Junção Neuromuscular/patologia , Receptores Colinérgicos , Índice de Gravidade de Doença , Estatísticas não Paramétricas
15.
J Neuroimmunol ; 281: 68-72, 2015 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-25867470

RESUMO

Myasthenia gravis is an autoimmune disease characterized by muscle weakness due to neuromuscular junction (NMJ) damage by anti-acetylcholine receptor (AChR) auto-antibodies and complement. In experimental autoimmune myasthenia gravis (EAMG), which is induced by immunization with Torpedo AChR in CFA, anti-AChR IgG2b and IgG1 are the predominant isotypes in the circulation. Complement activation by isotypes such as IgG2b plays a crucial role in EAMG pathogenesis; this suggested the possibility that IgG1, which does not activate complement through the classical pathway, may suppress EAMG. In this study, we show that AChR-immunized BALB/c mice genetically deficient for IgG1 produce higher levels of complement-activating isotypes of anti-AChR, especially IgG3 and IgG2a, and develop increased IgG3/IgG2a deposits at the NMJ, as compared to wild type (WT) BALB/c mice. Consistent with this, AChR-immunized IgG1(-/-) BALB/c mice lose muscle strength and muscle AChR to a greater extent than AChR-immunized WT mice. These observations demonstrate that IgG1 deficiency leads to increased severity of EAMG associated with an increase in complement activating IgG isotypes. Further studies are needed to dissect the specific role or mechanism of IgG1 in limiting EAMG and that of EAMG exacerbating role of complement activating IgG3 and IgG2a in IgG1 deficiency.


Assuntos
Imunoglobulina G/imunologia , Imunoglobulina G/metabolismo , Miastenia Gravis Autoimune Experimental/imunologia , Miastenia Gravis Autoimune Experimental/metabolismo , Animais , Feminino , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Torpedo
16.
Immunol Lett ; 168(2): 306-12, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26493475

RESUMO

Extraocular muscles (EOM) are preferentially involved in myasthenia gravis (MG) and acetylcholine receptor (AChR) antibody positive MG patients may occasionally present with isolated ocular symptoms. Although experimental autoimmune myasthenia gravis (EAMG) induced by whole AChR immunization closely mimics clinical and immunopathological aspects of MG, EOM are usually not affected. We have previously developed an EAMG model, which imitates EOM symptoms of MG by immunization of human leukocyte antigen (HLA) transgenic mice with α or γ-subunits of human AChR (H-AChR). To investigate the significance of the ϵ-subunit in ocular MG, we immunized HLA-DR3 and HLA-DQ8 transgenic mice with recombinant H-AChR ϵ-subunit expressed in Escherichia coli. HLA-DR3 transgenic mice showed significantly higher clinical ocular and generalized MG severity scores and lower grip strength values than HLA-DQ8 mice. H-AChR ϵ-subunit-immunized HLA-DR3 transgenic mice had higher serum anti-AChR antibody (IgG, IgG1, IgG2b, IgG2c and IgM) levels, neuromuscular junction IgG and complement deposit percentages than ϵ-subunit-immunized HLA-DQ8 transgenic mice. Control mice immunized with E. coli extract or complete Freund adjuvant (CFA) did not show clinical and immunopathological features of ocular and generalized EAMG. Lymph node cells of ϵ-subunit-immunized HLA-DR3 mice showed significantly higher proliferative responses than those of ϵ-subunit-immunized HLA-DQ8 mice, crude E. coli extract-immunized and CFA-immunized transgenic mice. Our results indicate that the human AChR ϵ-subunit is capable of inducing myasthenic muscle weakness. Diversity of the autoimmune responses displayed by mice expressing different HLA class II molecules suggests that the interplay between HLA class II alleles and AChR subunits might have a profound impact on the clinical course of MG.


Assuntos
Antígenos HLA-DQ/imunologia , Antígeno HLA-DR3/imunologia , Miastenia Gravis Autoimune Experimental/imunologia , Músculos Oculomotores/imunologia , Animais , Proliferação de Células , Ensaio de Imunoadsorção Enzimática , Adjuvante de Freund/imunologia , Antígenos HLA-DQ/genética , Antígeno HLA-DR3/genética , Humanos , Imunização/métodos , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Imunoglobulina M/sangue , Imunoglobulina M/imunologia , Linfócitos/imunologia , Linfócitos/patologia , Camundongos Transgênicos , Microscopia de Fluorescência , Miastenia Gravis Autoimune Experimental/induzido quimicamente , Miastenia Gravis Autoimune Experimental/genética , Músculos Oculomotores/metabolismo , Músculos Oculomotores/patologia , Subunidades Proteicas/imunologia , Receptores Colinérgicos/imunologia
17.
Shock ; 21(6): 500-4, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15167677

RESUMO

Circulatory neutrophils are known to be critical mediators of inflammation and oxidative stress during ischemia reperfusion (I/R) injury. Recent studies have shown an important role for protein kinase C (PKC) in neutrophil survival and function. Activation of specific isotypes of PKC are known to be involved in membrane alteration and motility, oxidative phosphorylation, and apoptosis modulation of neutrophils. However, the role of PKC in neutrophil responses to I/R in the clinical setting has not been studied. In this study, we examined the neutrophil activation of PKC induced by tourniquet-controlled I/R of skeletal muscle in humans. We found that I/R rapidly activates and translocates PKC delta, but not any of the classical forms of PKC (alpha or beta) from cytosol to the particulate fraction of neutrophils. Particulate translocation of PKC delta is sustained up to 4 h after reperfusion and is associated with kinase activity. Postreperfusion activation of PKC delta in neutrophils signals proapoptosis, but does not cause immediate cell death (as revealed by neutrophil morphology study and DNA-laddering assay). This study indicates that calcium-independent novel PKC delta (nPKC delta) might be predominantly involved in regulating membrane functions and survival of neutrophils associated with post-I/R-induced inflammatory oxidative stress.


Assuntos
Músculo Esquelético/irrigação sanguínea , Neutrófilos/enzimologia , Proteína Quinase C/metabolismo , Traumatismo por Reperfusão/enzimologia , Reperfusão , Acetofenonas/farmacologia , Apoptose/fisiologia , Benzopiranos/farmacologia , Células Cultivadas , Citosol/metabolismo , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Humanos , Neutrófilos/citologia , Neutrófilos/efeitos dos fármacos , Fosforilação , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C-delta , Transporte Proteico
18.
Curr Protoc Immunol ; Chapter 15: Unit 15.8., 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23392639

RESUMO

Myasthenia gravis (MG) is a T cell-dependent antibody-mediated autoimmune neuromuscular disease. Antibodies to the nicotinic acetylcholine receptor (AChR) destroy the AChR, thus leading to defective neuromuscular transmission of electrical impulse and to muscle weakness. This unit is a practical guide to the induction and evaluation of experimental autoimmune myasthenia gravis (EAMG) in the mouse, the animal model for MG. Protocols are provided for the extraction and purification of AChR from the electric organs of Torpedo californica, or the electric ray. The purified receptor is used as an immunogen to induce autoimmunity to AChR, thus causing EAMG. The defect in neuromuscular transmission can also be measured quantitatively by electromyography. In addition, EAMG is frequently characterized by the presence of serum antibodies to AChR, which are measured by radioimmunoassay and by a marked antibody-mediated reduction in the number of muscle AChRs. AChR extracted from mouse muscle is used in measuring serum antibody levels and for quantifying muscle AChR content. Another hallmark of the disease is complement and IgG deposits located at the neuromuscular junction, which can be visualized by immunofluorescence techniques.


Assuntos
Proteínas de Peixes/imunologia , Músculo Esquelético/imunologia , Miastenia Gravis Autoimune Experimental/imunologia , Junção Neuromuscular/efeitos dos fármacos , Receptores Nicotínicos/imunologia , Transmissão Sináptica/efeitos dos fármacos , Animais , Autoanticorpos/biossíntese , Autoanticorpos/imunologia , Proteínas do Sistema Complemento/imunologia , Eletromiografia , Feminino , Proteínas de Peixes/administração & dosagem , Proteínas de Peixes/isolamento & purificação , Imunoglobulina G/imunologia , Masculino , Camundongos , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiopatologia , Miastenia Gravis Autoimune Experimental/metabolismo , Miastenia Gravis Autoimune Experimental/fisiopatologia , Junção Neuromuscular/fisiopatologia , Radioimunoensaio , Receptores Nicotínicos/administração & dosagem , Receptores Nicotínicos/isolamento & purificação , Torpedo/fisiologia
19.
Curr Protoc Immunol ; Chapter 15: Unit 15.23, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22048803

RESUMO

Myasthenia gravis (MG) is a T cell-dependent antibody-mediated autoimmune neuromuscular disease. Antibodies to the nicotinic acetylcholine receptor (AChR) destroy the AChR, thus leading to defective neuromuscular transmission of electrical impulse and to muscle weakness. This unit is a practical guide to the induction and evaluation of experimental autoimmune myasthenia gravis (EAMG) in the mouse, the animal model for MG. Protocols are provided for the extraction and purification of AChR from the electric organs of Torpedo californica, or the electric ray. The purified receptor is used as an immunogen to induce autoimmunity to AChR, thus causing EAMG. The defect in neuromuscular transmission can also be measured quantitatively by electromyography. In addition, EAMG is frequently characterized by the presence of serum antibodies to AChR, which are measured by radioimmunoassay and by a marked antibody-mediated reduction in the number of muscle AChRs. AChR extracted from mouse muscle is used in measuring serum antibody levels and for quantifying muscle AChR content. Another hallmark of the disease is complement and IgG deposits located at the neuromuscular junction, which can be visualized by immunofluorescence techniques.


Assuntos
Miastenia Gravis Autoimune Experimental/imunologia , Receptores Nicotínicos/imunologia , Animais , Órgão Elétrico/imunologia , Eletromiografia , Imunoglobulina G/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Músculo Esquelético/imunologia , Junção Neuromuscular/imunologia , Receptores Nicotínicos/isolamento & purificação , Torpedo/imunologia
20.
J Interferon Cytokine Res ; 30(12): 917-21, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20973681

RESUMO

Interleukin-6⁻¹74 (IL-6⁻¹74) and tumor necrosis factor α⁻³°8 (TNFα⁻³°8) are high-cytokine-producing genotypes that are known to increase the susceptibility to infectious diseases, but their influence on cytokine production induced by respiratory viruses is unknown. We exposed human monocyte-derived macrophages from IL-6⁻¹74, TNFα⁻³°8, and normal genotype donors to different respiratory viruses. Respiratory syncytial virus (RSV) stimulation was associated with higher IL-6 concentrations in IL-6⁻¹74 donors than in normal donors (P = 0.015); 2 of 7 (29%) polymorphic donors were poor responders compared with 6 of 7 (86%) normal donors (P = 0.002). Adenovirus, influenza virus, and RSV stimulations were associated with higher TNFα concentrations in TNFα⁻³°8 donors than in normal donors (P = 0.03, <0.01, <0.01). A similar trend was seen with rhinovirus stimulation, but this was not significant. These results show that IL-6⁻¹74 and TNFα⁻³°8 gene polymorphisms lead to enhanced production of the respective cytokines when exposed to specific respiratory viruses. This, in turn, may influence the susceptibility to, severity of, and recovery from respiratory virus infections, or influence the immune response to and reactogenicity of viral vaccines.


Assuntos
Interleucina-6/biossíntese , Interleucina-6/genética , Macrófagos/virologia , Polimorfismo de Nucleotídeo Único/genética , Vírus Sinciciais Respiratórios/imunologia , Fator de Necrose Tumoral alfa/biossíntese , Fator de Necrose Tumoral alfa/genética , Humanos , Interleucina-6/imunologia , Macrófagos/imunologia , Macrófagos/metabolismo , Polimorfismo de Nucleotídeo Único/imunologia , Vírus Sinciciais Respiratórios/isolamento & purificação , Fator de Necrose Tumoral alfa/imunologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa