Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Genes Genomics ; 46(3): 333-340, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-37837514

RESUMO

OBJECTIVE: Atopic dermatitis (AD) is an inflammatory skin disease. Naringenin (Nar) possesses an anti-inflammatory property. This paper attempts to discuss the functional mechanism of Nar in AD mice through the Janus kinase 2 (JAK2)/signal transducer and activation of transcription 3 (STAT3) pathway. METHODS: Mouse models of DNFB-induced AD were established and treated with Nar, followed by intraperitoneal injection with the JAK2/STAT3 pathway activator Coumermycin A1. Dermatitis severity was scored and the thickness of right ear was measured. The pathological changes in dorsal skin tissues were observed by HE staining. The number of infiltrated mast cells and eosinophilic granulocytes was counted by TB staining. The serum IgE level and levels of TNF-α, IL-6, IFN-γ, IL-12, and IL-5 in dorsal skin tissues were measured by ELISA. The levels of p-JAK2, JAK2, p-STAT3, and STAT3 were determined by Western blot. RESULTS: Nar decreased dermatitis scores and right ear thickness, alleviated skin lesions, and reduced the number of infiltrated mast cells and eosinophilic granulocytes in AD mice. The serum IgE level and levels of TNF-α, IL-6, IFN-γ, IL-12, and IL-5 in dorsal skin tissues of AD mice were diminished after Nar treatment in a dose-dependent manner. Nar inhibited the activation of the JAK2/STAT3 pathway. The activation of the JAK2/STAT3 pathway partially nullified the therapeutic function of Nar on AD mice. CONCLUSION: Nar protects mice from AD by inhibiting inflammation and promoting immune responses through the inhibition of the JAK2/STAT3 pathway.


Assuntos
Dermatite Atópica , Flavanonas , Camundongos , Animais , Dermatite Atópica/tratamento farmacológico , Dermatite Atópica/patologia , Interleucina-6 , Fator de Necrose Tumoral alfa , Janus Quinase 2/metabolismo , Interleucina-5/efeitos adversos , Citocinas , Inflamação/tratamento farmacológico , Imunoglobulina E/efeitos adversos , Interleucina-12/efeitos adversos
2.
BMC Cancer ; 23(1): 32, 2023 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-36624467

RESUMO

BACKGROUND: The link between immediate hypersensitivity reactions (HSR) following the first cetuximab infusion and the IgE sensitization against anti-galactose-α-1,3-galactose (α-Gal) is now well-established. An automated Fluoroenzyme-Immunoassay (FEIA) is available and may facilitate the screening of patients with anti-α-Gal IgE before treatment. METHODS: This study aimed to evaluate its performances as compared to a previously validated anti-cetuximab IgE ELISA, using 185 samples from two previously studied cohorts. RESULTS: Despite 21.1% of discrepancies between the two techniques, FEIA discriminated better positive patients and similarly negative ones with a ≥ 0.525 kUA/L threshold. Sensitivity was 87.5% for both tests, specificity was better for FEIA (96.3% vs ELISA: 82.1%). FEIA had a higher positive likelihood ratio (23.9 vs ELISA: 4.89) and a similar negative likelihood ratio (0.13 vs ELISA: 0.15). In our population, the risk of severe HSR following a positive test was higher with FEIA (56.7% vs ELISA: 19.6%) and similar following a negative test (0.7% vs ELISA: 0.8%). CONCLUSION: Although the predictive value of the IgE screening before cetuximab infusion remains discussed, this automated commercial test can identify high-risk patients and is suitable for routine use in laboratories. It could help avoiding cetuximab-induced HSR by a systematic anti-α-Gal IgE screening before treatment.


Assuntos
Anafilaxia , Hipersensibilidade Alimentar , Humanos , Anafilaxia/induzido quimicamente , Anafilaxia/diagnóstico , Cetuximab/efeitos adversos , Hipersensibilidade Alimentar/diagnóstico , Galactose/efeitos adversos , Imunoglobulina E/efeitos adversos , Ensaio de Imunoadsorção Enzimática
3.
PLoS One ; 18(1): e0279893, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36598904

RESUMO

Arsenic is a potent environmental toxicant and human carcinogen. Skin lesions are the most common manifestations of chronic exposure to arsenic. Advanced-stage skin lesions, particularly hyperkeratosis have been recognized as precancerous diseases. However, the underlying mechanism of arsenic-induced skin lesions remains unknown. Periostin, a matricellular protein, is implicated in the pathogenesis of many forms of skin lesions. The objective of this study was to examine whether periostin is associated with arsenic-induced skin lesions. A total of 442 individuals from low- (n = 123) and high-arsenic exposure areas (n = 319) in rural Bangladesh were evaluated for the presence of arsenic-induced skin lesions (Yes/No). Participants with skin lesions were further categorized into two groups: early-stage skin lesions (melanosis and keratosis) and advanced-stage skin lesions (hyperkeratosis). Drinking water, hair, and nail arsenic concentrations were considered as the participants' exposure levels. The higher levels of arsenic and serum periostin were significantly associated with skin lesions. Causal mediation analysis revealed the significant effect of arsenic on skin lesions through the mediator, periostin, suggesting that periostin contributes to the development of skin lesions. When skin lesion was used as a three-category outcome (none, early-stage, and advanced-stage skin lesions), higher serum periostin levels were significantly associated with both early-stage and advanced-stage skin lesions. Median (IQR) periostin levels were progressively increased with the increasing severity of skin lesions. Furthermore, there were general trends in increasing serum type 2 cytokines (IL-4, IL-5, IL-13, and eotaxin) and immunoglobulin E (IgE) levels with the progression of the disease. The median (IQR) of IL-4, IL-5, IL-13, eotaxin, and IgE levels were significantly higher in the early-and advanced-stage skin lesions compared to the group of participants without skin lesions. The results of this study suggest that periostin is implicated in the pathogenesis and progression of arsenic-induced skin lesions through the dysregulation of type 2 immune response.


Assuntos
Arsênio , Ceratose Actínica , Dermatopatias , Humanos , Arsênio/toxicidade , Arsênio/análise , Interleucina-13 , Interleucina-4 , Interleucina-5 , Exposição Ambiental , Abastecimento de Água , Dermatopatias/induzido quimicamente , Imunoglobulina E/efeitos adversos
4.
Animal Model Exp Med ; 6(3): 255-265, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-36131559

RESUMO

BACKGROUND: Amarogentin (AMA) is a secoiridoid glycoside extracted from Swertia and Gentiana roots and exhibits many biological effects such as antioxidative, anti-inflammatory, and antitumor activities. Atopic dermatitis (AD) is a chronic inflammatory skin disease caused by disorders in the regulation of multiple inflammatory cytokines. No effective cure has been found for AD now. METHODS: We constructed the HaCat and splenocyte model and tested the inhibitory effect of AMA on IL-4, IL-6, and IL-13 secretions using enzyme-linked immunosorbent assay (ELISA). The AD mouse model was constructed and treated with AMA, the severity of skin lesions was observed, epidermal tissue was collected, and epidermal thickness and mast cell infiltration were observed using hematoxylin and eosin and toluidine blue staining, respectively. The expression of kallikrein-related peptidase 7 (KLK7) and filaggrin (FLG) was detected using immunostaining and Western blot analysis. The mRNA expression of KLK7 and FLG was detected using quantitative polymerase chain reaction (qPCR). Blood immunoglobulin E (IgE) secretion was detected. RESULTS: AMA inhibited IL-6 secreted by tumor necrosis factor (TNF)-α-induced HaCaT cells and reduced IL-4 and IL-13 secreted by phytohemagglutinin (PHA)-induced primary cells in the mice spleen. It was found that the treatment of AMA with 2,4-dinitrochlorobenzene-induced AD-like mice could promote the recovery of dermatitis, reduce the score of dermatitis severity and the scratching frequency, treat the skin lesions, reduce the epidermal thickness, decrease the infiltration of mast cells, reduce the IgE level in serum, decrease the expression levels of AD-related cytokines, increase protein and mRNA expression of FLG, and reduce the protein and mRNA expression of KLK7 in the skin tissues of AD-like mice. CONCLUSION: In conclusion, AMA inhibits inflammatory response at the cellular level, and AMA reduces the validation response of specific dermatitis mice, relieves pruritus, and repairs the damaged skin barrier.


Assuntos
Dermatite Atópica , Animais , Camundongos , Humanos , Dermatite Atópica/induzido quimicamente , Dermatite Atópica/tratamento farmacológico , Dermatite Atópica/metabolismo , Dinitroclorobenzeno/efeitos adversos , Interleucina-13/efeitos adversos , Interleucina-6/efeitos adversos , Células HaCaT/metabolismo , Células HaCaT/patologia , Interleucina-4/efeitos adversos , Citocinas/genética , Citocinas/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Anti-Inflamatórios/efeitos adversos , Imunoglobulina E/efeitos adversos , RNA Mensageiro/efeitos adversos
5.
Front Immunol ; 14: 1241851, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38274796

RESUMO

Background: Perioperative anaphylaxis is a rare and acute systemic manifestation of drug-induced hypersensitivity reactions that occurs following anesthesia induction; the two main classes of drugs responsible for these reactions being neuromuscular blocking agents (NMBA) and antibiotics. The sensitization mechanisms to the drugs are not precisely known, and few risk factors have been described. A growing body of evidence underlines a link between occurrence of allergy and microbiota composition. However, no data exist on microbiota in perioperative anaphylaxis. The aim of this study was to compare circulating microbiota richness and composition between perioperative anaphylaxis patients and matched controls. Methods: Circulating 16s rDNA was quantified and sequenced in serum samples from 20 individuals with fully characterized IgE-mediated NMBA-related anaphylaxis and 20 controls matched on sex, age, NMBA received, type of surgery and infectious status. Microbiota composition was analyzed with a published bioinformatic pipeline and links with patients clinical and biological data investigated. Results: Analysis of microbiota diversity showed that anaphylaxis patients seem to have a richer circulating microbiota than controls, but no major differences of composition could be detected with global diversity indexes. Pairwise comparison showed a difference in relative abundance between patients and controls for Saprospiraceae, Enterobacteriaceae, Veillonellaceae, Escherichia-Shigella, Pseudarcicella, Rhodoferax, and Lewinella. Some taxa were associated with concentrations of mast cell tryptase and specific IgE. Conclusion: We did not find a global difference in terms of microbiota composition between anaphylaxis patient and controls. However, several taxa were associated with anaphylaxis patients and with their biological data. These findings must be further confirmed in different settings to broaden our understanding of drug anaphylaxis pathophysiology and identify predisposition markers.


Assuntos
Anafilaxia , Hipersensibilidade a Drogas , Bloqueadores Neuromusculares , Humanos , Anafilaxia/etiologia , Triptases , Fatores de Risco , Bloqueadores Neuromusculares/efeitos adversos , Imunoglobulina E/efeitos adversos
6.
J Food Biochem ; 46(7): e14135, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35301731

RESUMO

Pinus koraiensis needles (PKN) and cones (PKC) have been shown to protect against inflammation and pathogenic bacteria. We investigated the efficacies and action mechanisms of topical applications of 1,3-butylene glycol (BG) extracts and oral administration of their water extracts on atopic dermatitis (AD) symptoms. After exposing HaCaT cells and Nc/Nga mice dorsal skins to 2,4-dinitrochlorobenzene (DNCB) to induce atopic dermatitis models, they were topically applied BG (AD-control), 30% PKNX, or 30% PKCX to the skin lesions and fed water extracts (0.5%) in high-fat diets for 5 weeks. Normal-control mice had no DNCB exposure. Serum immunoglobulin E (IgE), IL-4, and TNF-α levels and gene expressions of TNF-α, IL-4, IL-6, and IFN-γ in the dorsal skin and HaCaT cells were measured. The AD-control mice elevated TNF-α and IL-6 mRNA levels in HaCaT cells. Both extracts attenuated clinical AD symptoms in AD-induced Nc/Nga mice: PKNX improved hemorrhage, erythema, and lichenification of dorsal skin better than PKCX while both similarly alleviated erythema, edema, excoriation, and itching behavior. PKCX reduced IgE contents and increased filaggrin mRNA expression better than PKNX, but PKNX reduced lipid peroxides and mRNA levels of TNF-α and IL-4 in the dorsal skin. In the histological analysis of the dorsal skin, the administration of both extracts significantly decreased mast cell numbers, immune cell infiltration, gaps between the epidermis and dermis, and abnormal cell and nucleus shapes. In conclusion, both PKCX and PKNX treatment alleviated the DNCB-induced clinical symptoms of AD by alleviating immune-related symptoms and inflammation in partially different pathways. Therefore, PKNX and PKCX may be effective for AD therapy. PRACTICAL APPLICATIONS: Atopic dermatitis (AD) is related to an overly activated immune response, and it has steadily increased last 3 decades. However, no optimal sustainable treatments are available. Pinus koraiensis needles and cones extracts have been used for anti-inflammatory and antimicrobial treatment. The present study demonstrated that their intake and topical administration onto the AD lesion alleviated clinical AD symptoms associated with reduced proinflammatory cytokines, mast cell numbers, and immune cell infiltrates to maintain dermal structure with maintaining filaggrin expression in AD-induced HaCaT cells and Nc/Nga mice. These results suggested that Pinus koraiensis needles and cones extracts can be developed and applied as beneficial alternative therapies for AD.


Assuntos
Dermatite Atópica , Pinus , Animais , Dermatite Atópica/induzido quimicamente , Dermatite Atópica/tratamento farmacológico , Dinitroclorobenzeno/efeitos adversos , Células HaCaT , Humanos , Imunoglobulina E/efeitos adversos , Inflamação , Interleucina-4 , Interleucina-6 , Camundongos , RNA Mensageiro , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo , Água
7.
Phytomedicine ; 79: 153346, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33002828

RESUMO

BACKGROUND: Immunoglobulin E (IgE)-mediated mast cell (MC) activation is crucial in multiple allergic diseases. Parkinson disease protein 7 (DJ-1) and Lyn kinase were reported as the receptor-proximal events in IgE receptor (FcεRI) signals in human MC. Kaempferol, a natural flavonol mainly derived from the rhizome of traditional Chinese herb Kaempferia galanga L. (Zingiberaceae), has been known to inhibit allergic reactions, but it was limited to the receptor-distal signals on rat basophilic leukemia cells. A thorough investigation of the inhibitory effects of kaempferol on human MC has not been done. PURPOSE: To investigate the inhibitory effects of kaempferol on IgE-mediated anaphylaxis in vivo and in human MCs, as well as the mechanism underlying its effects, especially the receptor-proximal signals. METHODS: IgE-mediated passive cutaneous anaphylaxis and systemic anaphylaxis model were applied to elucidate the antiallergic activity of kaempferol in vivo. The degranulation assay, calcium imaging, the release of cytokines and chemokines on the laboratory of allergic disease 2 (LAD2) cells were used to evaluate the antiallergic effect of kaempferol in vitro. Western blot analysis was performed to investigate the DJ-1/Lyn signaling pathway and downstream molecules. Kinase activity assay, immunofluorescence, and molecular docking were conducted to confirm the influence of kaempferol on DJ-1/Lyn molecules. RESULTS: Kaempferol dose-dependently attenuated ovalbumin/IgE-induced mice paw swelling, primary MC activation from paw skin, as well as rehabilitated the hypothermia, and reduced the serum concentrations of histamine, tumor necrosis factor-alpha, interleukin-8, and monocyte chemo-attractant protein-1. Additionally, kaempferol suppressed IgE-mediated LAD2 cell degranulation and calcium fluctuation. Remarkably, kaempferol was found to bind with DJ-1 protein, and initially prevented DJ-1 from translocating to the plasma membrane, thereby inhibited full activation of Lyn, and eventually restrained those receptor-distal signaling molecules, involved Syk, Btk, PLCγ, IP3R, PKC, MAPKs, Akt and NF-κB. CONCLUSION: Kaempferol could be used as a DJ-1 modulator for preventing MC-mediated allergic disorders through attenuating Lyn activation.


Assuntos
Anafilaxia/tratamento farmacológico , Antialérgicos/farmacologia , Quempferóis/farmacologia , Mastócitos/efeitos dos fármacos , Anafilaxia/imunologia , Animais , Degranulação Celular/efeitos dos fármacos , Linhagem Celular , Relação Dose-Resposta a Droga , Humanos , Imunoglobulina E/efeitos adversos , Imunoglobulina E/metabolismo , Quempferóis/química , Masculino , Mastócitos/imunologia , Mastócitos/metabolismo , Camundongos Endogâmicos C57BL , Simulação de Acoplamento Molecular , Ovalbumina/toxicidade , Anafilaxia Cutânea Passiva/efeitos dos fármacos , Fosfolipase C gama/metabolismo , Proteína Desglicase DJ-1/metabolismo , Receptores de IgE/metabolismo , Transdução de Sinais/efeitos dos fármacos , Quinases da Família src/metabolismo
9.
Front Immunol ; 11: 50, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32063904

RESUMO

Background: As the number of allergic disease increases, studies to identify new treatments take on new urgency. Epigallocatechin gallate (EGCG), a major component of green tea, has been shown to possess a wide range of pharmacological properties, including anti-inflammation and anti-viral infection. In previous study, gallic acid (GA), a part of EGCG, has shown anti-allergic inflammatory effect. To improve on preliminary evidence that GA has allergy mitigating effect, we designed SG-SP1 based on GA, and aimed to assess the effects of SG-SP1 on mast cell-mediated allergic inflammation using various animal and in vitro models. Methods: For in vitro experiments, various types of IgE-stimulated mast cells (RBL-2H3: mast cell-like basophilic leukemia cells, and primary cultured peritoneal and bone marrow-derived mast cells) were used to determine the role of SG-SP1 (0.1-1 nM). Immunoglobulin (Ig) E-induced passive cutaneous anaphylaxis and ovalbumin-induced systemic anaphylaxis, standard animal models for immediate-type hypersensitivity were also used. Results: For in vitro, SG-SP1 reduced degranulation of mast cells by down-regulating intracellular calcium levels in a concentration-dependent manner. SG-SP1 decreased expression and secretion of inflammatory cytokines in activated mast cells. This suppressive effect was associated with inhibition of the phosphorylation of Lyn, Syk and Akt, and the nuclear translocation of nuclear factor-κB. Due to the strong inhibitory effect of SG-SP1 on Lyn, the known upstream signaling to FcεRI-dependent pathway, we confirmed the direct binding of SG-SP1 to FcεRI, a high affinity IgE receptor by surface plasmon resonance experiment. Oral administration of SG-SP1 hindered allergic symptoms of both anaphylaxis models evidenced by reduction of hypothermia, serum IgE, ear thickness, and tissue pigmentation. This inhibition was mediated by the reductions in serum histamine and interleukin-4. Conclusions: We determined that SG-SP1 directly interacts with FcεRI and propose SG-SP1 as a therapeutic candidate for mast cell-mediated allergic inflammatory disorders via inhibition of FcεRI signaling.


Assuntos
Anafilaxia/tratamento farmacológico , Anafilaxia/metabolismo , Anti-Inflamatórios/administração & dosagem , Ácido Gálico/análogos & derivados , Ácido Gálico/administração & dosagem , Mastócitos/metabolismo , Anafilaxia Cutânea Passiva/efeitos dos fármacos , Receptores de IgE/antagonistas & inibidores , Anafilaxia/induzido quimicamente , Animais , Anti-Inflamatórios/metabolismo , Cálcio/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Degranulação Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Ácido Gálico/metabolismo , Imunoglobulina E/efeitos adversos , Inflamação/imunologia , Inflamação/metabolismo , Masculino , Mastócitos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos ICR , Ovalbumina/efeitos adversos , Ratos , Ratos Sprague-Dawley , Receptores de IgE/metabolismo
10.
MAbs ; 12(1): 1685349, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31769737

RESUMO

IgE monoclonal antibodies hold great potential for cancer therapy. Preclinical in vivo systems, particularly those in which the antibody recognizes the host species target antigen and binds to cognate Fc receptors, are often the closest approximation to human exposure and represent a key challenge for evaluating the safety of antibody-based therapies. We sought to develop an immunocompetent rat system to assess the safety of a rodent anti-tumor IgE, as a surrogate for the human therapeutic candidate. We generated a rat IgE against the human tumor-associated antigen chondroitin sulfate proteoglycan 4 (CSPG4) and cross-reactive for the rat antigen. We analyzed CSPG4 distribution in normal rat and human tissues and investigated the in vivo safety of the antibody by monitoring clinical signs and molecular biomarkers after systemic administration to immunocompetent rats. Human and rat CSPG4 expression in normal tissues were comparable. Animals receiving antibody exhibited transient mild to moderate adverse events accompanied by mild elevation of serum tryptase, but not of angiotensin II or cytokines implicated in allergic reactions or cytokine storm. In the long term, repeated antibody administration was well tolerated, with no changes in animal body weight, liver and kidney functions or blood cell counts. This model provides preclinical support for the safety profiling of IgE therapeutic antibodies. Due to the comparable antigen tissue distribution in human and rat, this model may also comprise an appropriate tool for proof-of-concept safety evaluations of different treatment approaches targeting CSPG4.


Assuntos
Antígenos de Neoplasias/imunologia , Antineoplásicos Imunológicos/administração & dosagem , Proteoglicanas de Sulfatos de Condroitina/imunologia , Imunoglobulina E/administração & dosagem , Proteínas de Membrana/imunologia , Proteínas Recombinantes de Fusão/administração & dosagem , Animais , Citotoxicidade Celular Dependente de Anticorpos , Antineoplásicos Imunológicos/efeitos adversos , Linhagem Celular Tumoral , Reações Cruzadas , Feminino , Humanos , Imunização Secundária , Imunocompetência , Imunoglobulina E/efeitos adversos , Camundongos , Ratos , Proteínas Recombinantes de Fusão/efeitos adversos
11.
Allergy ; 73(12): 2328-2341, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-29654623

RESUMO

BACKGROUND: Designing biologically informative models for assessing the safety of novel agents, especially for cancer immunotherapy, carries substantial challenges. The choice of an in vivo system for studies on IgE antibodies represents a major impediment to their clinical translation, especially with respect to class-specific immunological functions and safety. Fcε receptor expression and structure are different in humans and mice, so that the murine system is not informative when studying human IgE biology. By contrast, FcεRI expression and cellular distribution in rats mirror that of humans. METHODS: We are developing MOv18 IgE, a human chimeric antibody recognizing the tumour-associated antigen folate receptor alpha. We created an immunologically congruent surrogate rat model likely to recapitulate human IgE-FcεR interactions and engineered a surrogate rat IgE equivalent to MOv18. Employing this model, we examined in vivo safety and efficacy of antitumour IgE antibodies. RESULTS: In immunocompetent rats, rodent IgE restricted growth of syngeneic tumours in the absence of clinical, histopathological or metabolic signs associated with obvious toxicity. No physiological or immunological evidence of a "cytokine storm" or allergic response was seen, even at 50 mg/kg weekly doses. IgE treatment was associated with elevated serum concentrations of TNFα, a mediator previously linked with IgE-mediated antitumour and antiparasitic functions, alongside evidence of substantially elevated tumoural immune cell infiltration and immunological pathway activation in tumour-bearing lungs. CONCLUSION: Our findings indicate safety of MOv18 IgE, in conjunction with efficacy and immune activation, supporting the translation of this therapeutic approach to the clinical arena.


Assuntos
Anticorpos Monoclonais Murinos/efeitos adversos , Anticorpos Monoclonais Murinos/uso terapêutico , Imunoglobulina E/efeitos adversos , Imunoglobulina E/uso terapêutico , Imunoterapia/métodos , Neoplasias/terapia , Receptores de IgE/metabolismo , Animais , Anticorpos Monoclonais Murinos/administração & dosagem , Anticorpos Monoclonais Murinos/metabolismo , Linhagem Celular Tumoral , Receptor 1 de Folato/imunologia , Humanos , Imunoglobulina E/administração & dosagem , Imunoglobulina E/imunologia , Imunoglobulina G/imunologia , Imunoglobulina G/metabolismo , Camundongos , Modelos Animais , Neoplasias/patologia , Ligação Proteica , Ratos , Estatísticas não Paramétricas , Resultado do Tratamento , Fator de Necrose Tumoral alfa/sangue
12.
Curr Top Microbiol Immunol ; 388: 109-49, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25553797

RESUMO

The success of antibody therapy in cancer is consistent with the ability of these molecules to activate immune responses against tumors. Experience in clinical applications, antibody design, and advancement in technology have enabled antibodies to be engineered with enhanced efficacy against cancer cells. This allows re-evaluation of current antibody approaches dominated by antibodies of the IgG class with a new light. Antibodies of the IgE class play a central role in allergic reactions and have many properties that may be advantageous for cancer therapy. IgE-based active and passive immunotherapeutic approaches have been shown to be effective in both in vitro and in vivo models of cancer, suggesting the potential use of these approaches in humans. Further studies on the anticancer efficacy and safety profile of these IgE-based approaches are warranted in preparation for translation toward clinical application.


Assuntos
Imunoglobulina E/uso terapêutico , Neoplasias/terapia , Animais , Anticorpos/uso terapêutico , Humanos , Imunoglobulina E/efeitos adversos , Imunoglobulina E/fisiologia , Imunoterapia , Mucina-1/imunologia , Antígeno Prostático Específico/imunologia , Receptor ErbB-2/imunologia , Vacinação
14.
Methods Mol Biol ; 677: 207-20, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-20941613

RESUMO

Through the release of biologically active products, mast cells function as important effector and immunoregulatory cells in diverse immunological reactions and other biological responses; for example, mast cells promote inflammation and other tissue changes in immunoglobulin E (IgE)-associated allergic disorders, as well as in certain innate and adaptive immune responses that are thought to be independent of IgE. Despite the mast cell's well-deserved reputation as a promoter of inflammation, others and we have used bone marrow-derived cultured mast cell (BMCMC) engrafted mast cell-deficient c-kit-mutant mice (so-called "mast cell knock-in" mice) to show that mast cells can also have important antiinflammatory and immunosuppressive functions in vivo. An early study showed that mast cells can contribute to susceptibility to ultraviolet B (UVB)-induced immunosuppression in one model of contact hypersensitivity (CHS), through effects mediated at least in part by histamine. Subsequently, it was reported that mast cells can mediate negative immunomodulatory effects following Anopheles mosquito bites, and in peripheral tolerance to skin allografts; however, the mechanism(s) by which mast cells mediate immunosuppressive functions in these two studies remains to be elucidated. Finally, we showed that mast cells and mast cell-derived IL-10 can limit the magnitude of and promote the resolution of certain CHS responses, and suppress the inflammation and skin injury associated with innate cutaneous responses to chronic low-dose UVB irradiation. This chapter outlines the generation of BMCMCs, a powerful model system commonly used to: (1) identify potential mast cell mediators in vitro; (2) study the mechanisms of mast cell activation and mediator release in response to specific stimuli in vitro; and (3) engraft mast cell-deficient mice to study the effector and immunoregulatory roles of mast cells or specific mast cell mediators in diverse immunological responses in vivo.


Assuntos
Imunoglobulina E/efeitos adversos , Imunossupressores/imunologia , Interleucina-10/uso terapêutico , Animais , Anti-Inflamatórios/uso terapêutico , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/imunologia , Células Cultivadas , Histamina/farmacologia , Imunoglobulina E/imunologia , Terapia de Imunossupressão , Imunossupressores/efeitos adversos , Inflamação/tratamento farmacológico , Inflamação/imunologia , Interleucina-10/imunologia , Mastócitos/efeitos dos fármacos , Mastócitos/fisiologia , Camundongos , Camundongos Endogâmicos C57BL
15.
Immunopharmacol Immunotoxicol ; 25(1): 73-85, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12675201

RESUMO

The effect of aqueous extract of Phlomis umbrosa Turcz. (Labiatae) root (PUAE) on mast cell-dependent immediate-type allergic reaction by anal therapy was investigated. PUAE (0.01 to 1 g/kg) dose-dependently inhibited systemic anaphylaxis induced by compound 48/80 in mice. When PUAE was pretreated at the same concentrations with systemic anaphylaxis, the plasma histamine levels were reduced in a dose-dependent manner. PUAE (0.1 and 1 g/kg) also significantly inhibited local anaphylaxis activated by anti-DNP IgE. PUAE (0.001 to 1 mg/mL) dose-dependently inhibited the histamine release from rat peritoneal mast cells (RPMC) activated by compound 48/80 or anti-DNP IgE. The level of cyclic AMP (cAMP) in human mast cells (HMC-1 cells) when PUAE (1 mg/mL) was added, transiently and significantly increased compared with that of basal cells. In addition, PUAE (0.1 and 1 mg/mL) inhibited the secretion of tumor necrosis factor-alpha (TNF-alpha), interleukin (IL)-6 in phorbol 12-myristate 13-acetate (PMA) plus calcium ionophore A23187-stimulated HMC-1 cells. These results provide evidence that anal therapy of PUAE may be beneficial in the treatment of allergic diseases.


Assuntos
Medicamentos de Ervas Chinesas/uso terapêutico , Liberação de Histamina/efeitos dos fármacos , Hipersensibilidade Imediata/tratamento farmacológico , Phlomis , Raízes de Plantas , Administração Retal , Animais , Calcimicina/farmacologia , AMP Cíclico/metabolismo , Dinitrobenzenos/imunologia , Relação Dose-Resposta a Droga , Hipersensibilidade Imediata/induzido quimicamente , Hipersensibilidade Imediata/imunologia , Imunoglobulina E/efeitos adversos , Imunoglobulina E/imunologia , Interleucina-6/metabolismo , Mastócitos/efeitos dos fármacos , Mastócitos/metabolismo , Camundongos , Camundongos Endogâmicos ICR , Peritônio/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Acetato de Tetradecanoilforbol/farmacologia , Fator de Necrose Tumoral alfa/metabolismo , p-Metoxi-N-metilfenetilamina
16.
J Pharm Pharmacol ; 51(2): 221-6, 1999 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10217323

RESUMO

We studied the effects of lavender oil on mast cell-mediated immediate-type allergic reactions in mice and rats. Lavender oil (1:500, 1:100, 1:10, 1:1, 1:0) inhibited concentration-dependently mast cell-dependent ear swelling response induced by compound 48/80 in mice by both topical and intradermal application. Lavender oil (1:500, 1:100, 1:10, 1:1, 1:0) inhibited concentration-dependently passive cutaneous anaphylaxis induced by anti-dinitrophenyl (DNP) IgE in rats by both topical and intradermal application. Lavender oil (1:500, 1:100, 1:10, 1:1, 1:0) also inhibited concentration-dependently the histamine release from the peritoneal mast cells by compound 48/80 or anti-DNP IgE. Moreover, lavender oil (1:1000, 1:100, 1:10, 1:0) had a significant inhibitory effect on anti-DNP IgE-induced tumour necrosis factor-alpha secretion from peritoneal mast cells. These results indicate that lavender oil inhibits immediate-type allergic reactions by inhibition of mast cell degranulation in-vivo and in-vitro.


Assuntos
Hipersensibilidade Imediata/prevenção & controle , Óleos Voláteis/uso terapêutico , Extratos Vegetais/uso terapêutico , Óleos de Plantas , Anafilaxia/etiologia , Anafilaxia/prevenção & controle , Animais , Dinitrobenzenos/imunologia , Relação Dose-Resposta a Droga , Feminino , Liberação de Histamina/efeitos dos fármacos , Hipersensibilidade Imediata/induzido quimicamente , Imunoglobulina E/efeitos adversos , Lavandula , Masculino , Mastócitos/efeitos dos fármacos , Mastócitos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Óleos Voláteis/farmacologia , Peritônio/citologia , Extratos Vegetais/farmacologia , Plantas Medicinais/química , Ratos , Ratos Wistar , Dermatopatias/etiologia , Dermatopatias/prevenção & controle , Fator de Necrose Tumoral alfa/efeitos dos fármacos , Fator de Necrose Tumoral alfa/metabolismo , p-Metoxi-N-metilfenetilamina/efeitos adversos
17.
Am J Respir Crit Care Med ; 155(2): 442-8, 1997 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9032176

RESUMO

Events occurring up to 16 d after antigen challenge were characterized using a novel protocol employing four bronchoscopies, two segmental antigen challenge (SAC) procedures (on Days 1 and 2), and six bronchoalveolar lavages (BALs) (on Days 1, 2, 9, and 16) in three groups: ragweed allergic asthmatics with dual phase airway reactions (AA-D), allergic asthmatics with a single early airway reaction (AA-S), and nonallergic nonasthmatic control subjects. In AA-D subjects, SAC produced a marked eosinophilic inflammatory response at 24 h associated with eosinophil degranulation (eosinophil cationic protein [ECP] in BAL fluid) and lung injury, which largely resolved by Day 16. When the second antigen-challenged segment (SAC performed on Day 2) was lavaged 7 d after challenge (Day 9), a persistent pulmonary eosinophilia was noted accompanied by minimal elevations in ECP and albumin. Eosinophil-active cytokines showed unique patterns: interleukin-5 (IL-5) increased in the antigen segment on Day 2 then returned to baseline after 7 d; granulocyte-macrophage colony-stimulating factor (GM-CSF) peaked at Day 2 but was persistently elevated throughout Day 16 in antigen segments, and increased in control segments at late time points; IL-3 levels were constant and similar in antigen and control segments. Changes were specific to AA-D subjects in comparison with control subjects. Elements of the IgE-mediated pulmonary inflammatory response differ markedly in their development and resolution.


Assuntos
Alérgenos/efeitos adversos , Asma/diagnóstico , Imunoglobulina E/efeitos adversos , Eosinofilia Pulmonar/fisiopatologia , Ribonucleases , Adulto , Antígenos , Proteínas Sanguíneas/análise , Testes de Provocação Brônquica , Lavagem Broncoalveolar , Broncoscopia , Proteínas Granulares de Eosinófilos , Feminino , Humanos , Mediadores da Inflamação/análise , Cinética , Masculino , Eosinofilia Pulmonar/etiologia , Testes de Função Respiratória
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA