Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Int J Nanomedicine ; 19: 2529-2552, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38505170

RESUMO

The blood-brain barrier (BBB) and blood-tumor barrier (BTB) pose substantial challenges to efficacious drug delivery for glioblastoma multiforme (GBM), a primary brain tumor with poor prognosis. Nanoparticle-based combinational strategies have emerged as promising modalities to overcome these barriers and enhance drug penetration into the brain parenchyma. This review discusses various nanoparticle-based combinatorial approaches that combine nanoparticles with cell-based drug delivery, viral drug delivery, focused ultrasound, magnetic field, and intranasal drug delivery to enhance drug permeability across the BBB and BTB. Cell-based drug delivery involves using engineered cells as carriers for nanoparticles, taking advantage of their intrinsic migratory and homing capabilities to facilitate the transport of therapeutic payloads across BBB and BTB. Viral drug delivery uses engineered viral vectors to deliver therapeutic genes or payloads to specific cells within the GBM microenvironment. Focused ultrasound, coupled with microbubbles or nanoparticles, can temporarily disrupt the BBB to increase drug permeability. Magnetic field-guided drug delivery exploits magnetic nanoparticles to facilitate targeted drug delivery under an external magnetic field. Intranasal drug delivery offers a minimally invasive avenue to bypass the BBB and deliver therapeutic agents directly to the brain via olfactory and trigeminal pathways. By combining these strategies, synergistic effects can enhance drug delivery efficiency, improve therapeutic efficacy, and reduce off-target effects. Future research should focus on optimizing nanoparticle design, exploring new combination strategies, and advancing preclinical and clinical investigations to promote the translation of nanoparticle-based combination therapies for GBM.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Nanopartículas , Humanos , Barreira Hematoencefálica , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/metabolismo , Sistemas de Liberação de Medicamentos , Encéfalo/metabolismo , Nanopartículas/metabolismo , Glioblastoma/tratamento farmacológico , Glioblastoma/metabolismo , Microambiente Tumoral
2.
Adv Mater ; 36(11): e2304333, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38096399

RESUMO

Current clinical therapeutic efficacy for the treatment of osteo- and rheumatoid-arthritis is obviously limited. Although mesenchymal stem cells (MSCs) are considered as a source of promising regenerative therapy, un-modified or genetically engineered MSCs injected in vivo restrict their clinical utility because of the low drug efficacy and unpredicted side effect, respectively. Herein, a strategy to enhance the migration efficacy of MSCs to inflamed joints via an inflammation-mediated education process is demonstrated. To reinforce the limited anti-inflammatory activity of MSCs, gold nanostar loaded with triamcinolone is conjugated to MSC. Furthermore, near-infrared laser-assisted photothermal therapy (PTT) induced by gold nanostar significantly elevates the anti-inflammatory efficacy of the developed drugs, even in advanced stage arthritis model. An immunological regulation mechanism study of PTT is first suggested in this study; the expression of the interleukin 22 receptor, implicated in the pathogenesis of arthritis, is downregulated in T lymphocytes by PTT, and Th17 differentiation from naïve CD4 T cell is inhibited. Collectively, inflammation-targeting MSCs conjugated with triamcinolone-loaded gold nanostar (Edu-MSCs-AuS-TA) promote the repolarization of macrophages and decrease neutrophil recruitment in joints. In addition, Edu-MSCs-AuS-TA significantly alleviate arthritis-associated pain, improve general locomotor activity, and more importantly, induce cartilage regeneration even for severe stages of arthritis model.


Assuntos
Artrite Reumatoide , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Humanos , Inflamação/metabolismo , Triancinolona/metabolismo , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Anti-Inflamatórios/metabolismo , Ouro
3.
Cells ; 12(11)2023 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-37296614

RESUMO

(1) Background: This study aims to elucidate a novel non-transcriptional action of IRF3 in addition to its role as a transcription factor in mast cell activation and associated allergic inflammation; (2) Methods: For in vitro experiments, mouse bone-marrow-derived mast cells (mBMMCs) and a rat basophilic leukemia cell line (RBL-2H3) were used for investigating the underlying mechanism of IRF3 in mast-cell-mediated allergic inflammation. For in vivo experiments, wild-type and Irf3 knockout mice were used for evaluating IgE-mediated local and systemic anaphylaxis; (3) Results: Passive cutaneous anaphylaxis (PCA)-induced tissues showed highly increased IRF3 activity. In addition, the activation of IRF3 was observed in DNP-HSA-treated mast cells. Phosphorylated IRF3 by DNP-HSA was spatially co-localized with tryptase according to the mast cell activation process, and FcεRI-mediated signaling pathways directly regulated that activity. The alteration of IRF3 affected the production of granule contents in the mast cells and the anaphylaxis responses, including PCA- and ovalbumin-induced active systemic anaphylaxis. Furthermore, IRF3 influenced the post-translational processing of histidine decarboxylase (HDC), which is required for granule maturation; and (4) Conclusion: Through this study, we demonstrated the novel function of IRF3 as an important factor inducing mast cell activation and as an upstream molecule for HDC activity.


Assuntos
Anafilaxia , Receptores de IgE , Ratos , Camundongos , Animais , Receptores de IgE/metabolismo , Mastócitos/metabolismo , Linhagem Celular , Inflamação/metabolismo
4.
Adv Mater ; 35(30): e2300934, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37114740

RESUMO

Owing to the intrinsic ability of stem cells to target the tumor environment, stem-cell-membrane-functionalized nanocarriers can target and load active anticancer drugs. In this work, a strategy that focuses on stem cells that self-target pancreatic cancer cells is developed. In particular, malignant deep tumors such as pancreatic cancer cells, one of the intractable tumors that currently have no successful clinical strategy, are available for targeting and destruction. By gaining the targeting ability of stem cells against pancreatic tumor cells, stem cell membranes can encapsulate nano-polylactide-co-glycolide loaded with doxorubicin to target and reduce deep pancreatic tumor tissues. Considering the lack of known target proteins on pancreatic tumor cells, the suggested platform technology can be utilized for targeting any malignant tumors in which surface target receptors are unavailable.


Assuntos
Antineoplásicos , Nanopartículas , Neoplasias Pancreáticas , Humanos , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/patologia , Doxorrubicina/farmacologia , Doxorrubicina/uso terapêutico , Células-Tronco , Linhagem Celular Tumoral , Neoplasias Pancreáticas
5.
J Nanobiotechnology ; 21(1): 12, 2023 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-36635755

RESUMO

Despite the development of therapeutic modalities to treat cancer, multidrug resistance (MDR) and incomplete destruction of deeply embedded lung tumors remain long-standing problems responsible for tumor recurrence and low survival rates. Therefore, developing therapeutic approaches to treat MDR tumors is necessary. In this study, nanodrugs with enhanced intracellular drug internalization were identified by the covalent bonding of carbon nanotubes of a specific nano size and doxorubicin (DOX). In addition, carbon nanotube conjugated DOX (CNT-DOX) sustained in the intracellular environment in multidrug-resistant tumor cells for a long time causes mitochondrial damage, suppresses ATP production, and results in the effective therapeutic effect of drug-resistant tumors. This study identified that H69AR lung cancer cells, an adriamycin (DOX) drug-resistant tumor cell line, did not activate drug resistance function on designed nano-anticancer drugs with a specific nano size. In summary, this study identified that the specific size of the nanodrug in combination with DOX overcame multidrug-resistant tumors by inducing selective accumulation in tumor cells and inhibiting ATP by mitochondrial damage.


Assuntos
Neoplasias Pulmonares , Nanopartículas , Nanotubos de Carbono , Humanos , Resistencia a Medicamentos Antineoplásicos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/patologia , Doxorrubicina/farmacologia , Doxorrubicina/uso terapêutico , Linhagem Celular Tumoral , Nanopartículas/uso terapêutico , Trifosfato de Adenosina
6.
Mol Med Rep ; 24(6)2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34664681

RESUMO

Psoriasis, a chronic inflammatory skin disease, is characterized by the excessive proliferation and impaired differentiation of epidermal keratinocytes and is accompanied by the increased infiltration of inflammatory cells. The condition requires long­term treatment and has no definitive cure. Hence, supplements and therapeutic agents have been intensely investigated. Gomisin M2 (GM2), a lignan extracted from Schisandra chinensis (Turcz). Baill. (Schisandraceae; S. chinensis), has demonstrated diverse pharmacological properties, including anticancer, anti­inflammatory and antiallergic effects. Based on these findings, the present study examined the effects of GM2 on an imiquimod (IMQ)­induced psoriasis mouse model and on keratinocytes stimulated by tumor necrosis factor (TNF)­α and interferon­Î³. IMQ was topically applied to the back skin of mice for 7 consecutive days, and the mice were orally administered CD. These results showed that the oral administration of GM2 suppressed the symptoms of psoriasis, as evidenced by reductions in skin thickness, psoriasis area severity index scores for psoriasis lesions, transepidermal water loss and myeloperoxidase (MPO)­associated cell infiltration. Furthermore, GM2 reduced the pathologically increased levels of immunoglobulin G2a, MPO and TNF­α in the serum and T helper (Th)1 and Th17 cell populations in the spleen. GM2 decreased the gene expression of inflammatory­related cytokines and chemokines and inhibited the expression of signal transducer and activator of transcription 1 and nuclear factor­κB in the activated keratinocytes. These results suggested that GM2 from S. chinensis is a potential therapeutic candidate to alleviate psoriasis­like skin inflammation.


Assuntos
Anti-Inflamatórios/farmacologia , Medicamentos de Ervas Chinesas/farmacologia , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Lignanas/farmacologia , Psoríase/tratamento farmacológico , Psoríase/metabolismo , Animais , Anti-Inflamatórios/uso terapêutico , Linhagem Celular , Citocinas/metabolismo , Modelos Animais de Doenças , Medicamentos de Ervas Chinesas/uso terapêutico , Feminino , Humanos , Imiquimode/toxicidade , Inflamação/induzido quimicamente , Inflamação/genética , Interferon gama/toxicidade , Queratinócitos/efeitos dos fármacos , Queratinócitos/metabolismo , Lignanas/uso terapêutico , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Psoríase/induzido quimicamente , Psoríase/patologia , Fator de Transcrição STAT1/metabolismo , Transdução de Sinais/efeitos dos fármacos , Células Th1/efeitos dos fármacos , Células Th17/efeitos dos fármacos , Fator de Necrose Tumoral alfa/toxicidade
7.
Adv Sci (Weinh) ; 8(8): 2004979, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33898204

RESUMO

The plasma proteins around nanoparticles (NPs) form an outer protein corona, significantly influencing the subsequent immune response. However, it was uncertain whether the protein corona around NPs influences immune response. This study clarified that the immune response mediated by the protein corona is greatly dependent on the type of plasma proteins surrounding the NPs. Structural changes in the unfolded protein corona elevated reactive oxygen species (ROS) levels and induced major proinflammatory cytokine release in both murine and human macrophage cell lines. In contrast, negligible structural changes in the protein corona provoke neither ROS production nor proinflammatory cytokine release. Furthermore, in vivo analysis confirms that a stimulated immune response by an unfolded protein corona triggers selective activation of innate and adaptive immunity in the spleen. Specifically, neutrophils, natural killer cells, and CD8+ T cells are overpopulated by unfolded protein corona structures surrounding nanotubes, whereas innate and adaptive immunologic responses are not triggered by a normal protein corona. In conclusion, highly unfolded protein corona structures are strongly correlated with subsequent activation of proinflammatory cytokines and innate immune responses; thus, the protein corona can be used in immune-enhancing therapy.


Assuntos
Imunidade Adaptativa/imunologia , Imunidade Inata/imunologia , Coroa de Proteína/imunologia , Animais , Linfócitos T CD8-Positivos/imunologia , Citocinas/imunologia , Feminino , Humanos , Células Matadoras Naturais , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Modelos Animais , Nanotubos , Neutrófilos/imunologia , Espécies Reativas de Oxigênio/imunologia
8.
Biomed Pharmacother ; 137: 111359, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33761595

RESUMO

Atopic dermatitis (AD) is a chronic inflammatory skin disorder that affects 10-20% of the world's population. Therefore, the discovery of drugs for the treatment of AD is important for human health. Hispidulin (HPD; also known as scutellarein 6-methyl ether or dinatin) is a natural flavone that exerts anti-inflammatory effects. In the present study, the effectiveness of HPD on AD-like skin inflammation was investigated. We used a mouse AD model through repeated exposure of mice to 2,4-dinitrochlorobenzene and house dust mite extract (Dermatophagoides farinae extract, DFE) to the ears. In addition, tumor necrosis factor-α and interferon-γ-activated keratinocytes (HaCaT cells) were used to investigate the underlying mechanism of HPD action. Oral administration of HPD alleviated AD-like skin inflammations: it reduced ear thickness; serum immunoglobulin (Ig)E, DFE-specific IgE, and IgG2a levels; and inflammatory cell infiltration. HPD reduced the expression of pro-inflammatory cytokines and chemokines through inhibition of signal transducer and activator of transcription 1 nuclear factor-κB in HaCaT cells. Taken together, these results suggest that HPD could be a potential drug candidate for the treatment of AD.


Assuntos
Antialérgicos/uso terapêutico , Dermatite Atópica/tratamento farmacológico , Dinitroclorobenzeno , Flavonas/uso terapêutico , Pyroglyphidae/imunologia , Pele/patologia , Animais , Antígenos de Dermatophagoides , Dermatite Atópica/imunologia , Dermatite Atópica/patologia , Eosinófilos/efeitos dos fármacos , Feminino , Imunoglobulinas/metabolismo , Queratinócitos/efeitos dos fármacos , Mastócitos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C
9.
Int J Nanomedicine ; 16: 8485-8507, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35002240

RESUMO

Mesenchymal stem cells (MSCs) are considered a promising regenerative therapy due to their ability to migrate toward damaged tissues. The homing ability of MSCs is unique compared with that of non-migrating cells and MSCs are considered promising therapeutic vectors for targeting major cells in many pathophysiological sites. MSCs have many advantages in the treatment of malignant diseases, particularly rheumatoid arthritis (RA). RA is a representative autoimmune disease that primarily affects joints, and secreted chemokines in the joints are well recognized by MSCs following their migration to the joints. Furthermore, MSCs can regulate the inflammatory process and repair damaged cells in the joints. However, the functionality and migration ability of MSCs injected in vivo still show insufficient. The targeting ability and migration efficiency of MSCs can be enhanced by genetic engineering or modification, eg, overexpressing chemokine receptors or migration-related genes, thus maximizing their therapeutic effect. However, there are concerns about genetic changes due to the increased probability of oncogenesis resulting from genome integration of the viral vector, and thus, clinical application is limited. Furthermore, it is suspected that administering MSCs can promote tumor growth and metastasis in xenograft and orthotopic models. For this reason, MSC mimicking nanoencapsulations are an alternative strategy that does not involve using MSCs or bioengineered MSCs. MSC mimicking nanoencapsulations consist of MSC membrane-coated nanoparticles, MSC-derived exosomes and artificial ectosomes, and MSC membrane-fused liposomes with natural or genetically engineered MSC membranes. MSC mimicking nanoencapsulations not only retain the targeting ability of MSCs but also have many advantages in terms of targeted drug delivery. Specifically, MSC mimicking nanoencapsulations are capable of encapsulating drugs with various components, including chemotherapeutic agents, nucleic acids, and proteins. Furthermore, there are fewer concerns over safety issues on MSC mimicking nanoencapsulations associated with mutagenesis even when using genetically engineered MSCs, because MSC mimicking nanoencapsulations use only the membrane fraction of MSCs. Genetic engineering is a promising route in clinical settings, where nano-encapsulated technology strategies are combined. In this review, the mechanism underlying MSC homing and the advantages of MSC mimicking nanoencapsulations are discussed. In addition, genetic engineering of MSCs and MSC mimicking nanoencapsulation is described as a promising strategy for the treatment of immune-related diseases.


Assuntos
Artrite Reumatoide , Exossomos , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Nanopartículas , Sistemas de Liberação de Medicamentos , Humanos
10.
ACS Appl Mater Interfaces ; 12(35): 38936-38949, 2020 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-32805872

RESUMO

Understanding the crosstalk between synoviocytes and macrophages is very important for the development of strategies to regulate inflammatory responses in an inflamed synovium. Simultaneous regulation of the pro- and anti-inflammatory responses of synoviocytes and macrophages (repolarization) is critical for the treatment of arthritis. Thus, the immune regulatory functions of an ideal nanodrug should not only decrease the pro-inflammatory response but also effectively increase the anti-inflammatory response. In this study, crosstalk between synoviocytes and macrophages was found to be significantly involved in the activation and deactivation of inflammatory responses in the synovium. Interestingly, a developed triamcinolone-gold nanoparticle (Triam-AuNP) complex both decreased the pro-inflammatory responses and increased the anti-inflammatory responses of fibroblast-like synoviocytes (FLSs) and macrophages via repolarization of macrophages from the M1 to the M2 phenotype. In contrast, triamcinolone alone only decreased the pro-inflammatory responses of FLSs and macrophages without upregulating their anti-inflammatory responses. In vitro (human), ex vivo (human), and in vivo (mouse) analyses clearly indicated that Triam-AuNPs effectively regulated the expression of both pro- and anti-inflammatory cytokines in FLSs and effectively repolarized activity of macrophages in the inflamed synovium. Furthermore, Triam-AuNPs significantly promoted cartilage regeneration, whereas triamcinolone alone did not induce either FLS anti-inflammatory activity or macrophage repolarization.


Assuntos
Ouro/química , Nanopartículas Metálicas/química , Triancinolona/química , Animais , Artrite Experimental/induzido quimicamente , Artrite Experimental/tratamento farmacológico , Artrite Experimental/patologia , Artrite Reumatoide/induzido quimicamente , Artrite Reumatoide/tratamento farmacológico , Sobrevivência Celular/efeitos dos fármacos , Citocinas/metabolismo , Regulação para Baixo/efeitos dos fármacos , Humanos , Ativação de Macrófagos/efeitos dos fármacos , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos DBA , Espécies Reativas de Oxigênio/metabolismo , Sinoviócitos/citologia , Sinoviócitos/efeitos dos fármacos , Sinoviócitos/metabolismo , Triancinolona/farmacologia
11.
Int J Nanomedicine ; 15: 5745-5765, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32821099

RESUMO

Glaucoma is a group of diseases characterized by progressive degeneration of retinal ganglion cells, leading to irreversible blindness. Currently, intraocular pressure reduction is the only established treatment available for glaucoma. With this treatment, the progression of the disease can only be delayed and there is no recovery. In addition, the commercially available eye drops have the disadvantage of low compliance and short therapeutic time, while glaucoma surgery always has the risk of failure due to wound fibrosis. Nanotechnology can overcome the limitations of the current treatment through the encapsulation and conjugation of drugs used for lowering intraocular pressure and antifibrotic agents using biodegradable or biocompatible nanoparticles for the sustained release of the drugs to protect the damaged ocular cells. Furthermore, using nanotechnology, treatment can be administered in various forms, including eye drops, contact lens, and ocular inserts, according to the convenience of the patients. Despite the promising results of delaying the progression of glaucoma, the regeneration of damaged ocular cells, including trabecular meshwork and retinal ganglion cells, is another critical hurdle to overcome. Bone marrow-derived mesenchymal stem cells and Müller glia cells can secrete neurogenic factors that trigger the regeneration of associated cells, including trabecular meshwork and retinal ganglion cells. In conclusion, this review highlights the potential therapeutic applications of nanotechnology- and stem cell-based methods that can be employed for the protection and regeneration of ocular cells.


Assuntos
Glaucoma/terapia , Nanomedicina , Animais , Lentes de Contato , Sistemas de Liberação de Medicamentos , Glaucoma/etiologia , Humanos , Nanotecnologia , Malha Trabecular/patologia
12.
Int J Nanomedicine ; 15: 5719-5743, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32821098

RESUMO

This review aims to summarize the methods that have been used till today, highlight methods that are currently being developed, and predict the future roadmap for anticancer therapy. In the beginning of this review, established approaches for anticancer therapy, such as conventional chemotherapy, hormonal therapy, monoclonal antibodies, and tyrosine kinase inhibitors are summarized. To counteract the side effects of conventional chemotherapy and to increase limited anticancer efficacy, nanodrug- and stem cell-based therapies have been introduced. However, current level of understanding and strategies of nanodrug and stem cell-based therapies have limitations that make them inadequate for clinical application. Subsequently, this manuscript reviews methods with fewer side effects compared to those of the methods mentioned above which are currently being investigated and are already being applied in the clinic. The newer strategies that are already being clinically applied include cancer immunotherapy, especially T cell-mediated therapy and immune checkpoint inhibitors, and strategies that are gaining attention include the manipulation of the tumor microenvironment or the activation of dendritic cells. Tumor-associated macrophage repolarization is another potential strategy for cancer immunotherapy, a method which activates macrophages to immunologically attack malignant cells. At the end of this review, we discuss combination therapies, which are the future of cancer treatment. Nanoparticle-based anticancer immunotherapies seem to be effective, in that they effectively use nanodrugs to elicit a greater immune response. The combination of these therapies with others, such as photothermal or tumor vaccine therapy, can result in a greater anticancer effect. Thus, the future of anticancer therapy aims to increase the effectiveness of therapy using various therapies in a synergistic combination rather than individually.


Assuntos
Antineoplásicos/farmacologia , Nanomedicina/tendências , Vacinas Anticâncer/imunologia , Humanos , Imunoterapia , Transplante de Células-Tronco , Microambiente Tumoral
13.
Int Immunopharmacol ; 83: 106398, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32197228

RESUMO

The functional inhibition of mast cells, which serve as a key effector cells in allergic reactions may be a specific target for treating immunoglobulin (Ig)E-mediated allergic reactions, which occur in various allergic diseases including anaphylaxis, asthma, and atopic dermatitis. In this study, we demonstrated the effects of dabrafenib, a therapeutic agent used to treat metastatic melanoma, with a focus on mast cell activation and local cutaneous anaphylaxis. In two types of mast cells (RBL-2H3 and mouse bone marrow-derived mast cells), dabrafenib (0.01, 0.1, 1 µM) pretreatment significantly decreased IgE-induced degranulation, intracellular calcium influx, and the activity of intracellular signaling molecules, such as Lyn, Syk, Akt, and PLCγ. Dabrafenib ameliorated mRNA and protein expression levels of interleukin-4 and tumor necrosis factor-α by the reduction of nuclear localization of nuclear factor-κB and nuclear factor of activated T-cells. In passive cutaneous anaphylaxis, oral administration of dabrafenib (0.1, 1, 10 mg/kg) reduced local pigmentation and ear thickness in a dose-dependent manner. Taken together, these results suggest that dabrafenib is a therapeutic drug candidate that controls IgE-mediated allergic inflammatory diseases through suppression of mast cell activity.


Assuntos
Anafilaxia/tratamento farmacológico , Antineoplásicos/uso terapêutico , Imidazóis/uso terapêutico , Mastócitos/imunologia , Melanoma/tratamento farmacológico , Oximas/uso terapêutico , Pele/patologia , Linfócitos T/imunologia , Animais , Sinalização do Cálcio , Degranulação Celular/efeitos dos fármacos , Modelos Animais de Doenças , Humanos , Imunoglobulina E/metabolismo , Interleucina-4/metabolismo , Masculino , Mastócitos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos ICR , NF-kappa B/metabolismo , Metástase Neoplásica
14.
Pharmacol Rep ; 72(4): 1002-1010, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32048267

RESUMO

BACKGROUND: Mast cells are immune effector cells mediating allergic inflammation by the secretion of inflammatory mediators such as histamine and pro-inflammatory cytokines. Orientin is a naturally occurring bioactive flavonoid that possesses diverse biological properties, including anti-inflammation, anti-oxidative, anti-tumor, and cardio protection. The objective of this study was to rule out the effectiveness of orientin in mast cell-mediated allergic inflammation. METHODS: In this study, in vitro effects of orientin were evaluated in RBL-2H3, mouse bone marrow-derived mast cells, rat peritoneal mast cells, and in vivo effects were evaluated by inducing passive cutaneous anaphylaxis (PCA) in Imprinting Control Region (ICR) mice. RESULTS: Findings show that orientin suppressed the immunoglobulin E (IgE)-mediated mast cell degranulation by reducing intracellular calcium level in a concentration-dependent manner. Orientin suppressed the secretion of pro-inflammatory cytokines in mast cells. This inhibitory effects of orientin was through inhibition of FcεRI-mediated signaling proteins. In addition, oral administration of orientin suppressed the IgE-mediated PCA reactions in a dose-dependent manner, which was evidenced by reduced Evan's blue pigmentation and ear swelling. CONCLUSIONS: Based on these findings, we suggest that orientin might have potential to alleviate allergic reaction and mast cell-mediated allergic disease.


Assuntos
Antialérgicos/uso terapêutico , Anti-Inflamatórios/uso terapêutico , Flavonoides/uso terapêutico , Glucosídeos/uso terapêutico , Hipersensibilidade/prevenção & controle , Mediadores da Inflamação/antagonistas & inibidores , Mastócitos/efeitos dos fármacos , Animais , Antialérgicos/farmacologia , Anti-Inflamatórios/farmacologia , Células Cultivadas , Relação Dose-Resposta a Droga , Flavonoides/farmacologia , Glucosídeos/farmacologia , Hipersensibilidade/imunologia , Hipersensibilidade/metabolismo , Mediadores da Inflamação/imunologia , Mediadores da Inflamação/metabolismo , Masculino , Mastócitos/imunologia , Mastócitos/metabolismo , Camundongos , Camundongos Endogâmicos ICR , Ratos , Ratos Sprague-Dawley
15.
Nanoscale ; 11(42): 19980-19993, 2019 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-31603160

RESUMO

Despite the lysosomal "proton sponge hypothesis" being considered to be an additional factor for stimulating the cellular toxicity of nanoparticle-based drug delivery systems, a clear relationship between the massive influx of calcium ions and the proton sponge effect, both of which are associated with cancer cell apoptosis, has still not been elucidated. Cetrimonium bromide (CTAB: cationic quaternary amino group based) gold nanorods possessed a more effective electric surface charge for inducing the lysosomal proton sponge effect than anionic gold nanoparticles. In this aspect, identifying released cytoplasmic Cl-, arising from the ruptured lysosomal compartment, in the cytoplasm is critical for supporting the "proton sponge hypothesis". This study clarified that the burst release of Cl-, as a result of lysosomal swelling by CTAB gold nanorods, stimulates the transient receptor potential channels melastatin 2 (TRPM2) channels, and subsequently induces a massive Ca2+ influx, which independently increases apoptosis of cancer cells. Although the previous concept of elevated cancer apoptosis acting through the proton sponge effect is unclear, this study supports the evidence that a massive Ca2+ influx mediated in response to a burst release of Cl- significantly influenced cytotoxicity of cancer cells in tumor tissues.


Assuntos
Apoptose/efeitos dos fármacos , Ouro , Lisossomos , Nanopartículas Metálicas , Nanotubos/química , Proteínas de Neoplasias , Neoplasias Experimentais , Canais de Cátion TRPM , Animais , Linhagem Celular Tumoral , Cetrimônio/química , Cetrimônio/farmacologia , Feminino , Ouro/química , Ouro/farmacologia , Humanos , Lisossomos/genética , Lisossomos/metabolismo , Lisossomos/patologia , Nanopartículas Metálicas/química , Nanopartículas Metálicas/uso terapêutico , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/genética , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Canais de Cátion TRPM/genética , Canais de Cátion TRPM/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
16.
Int J Nanomedicine ; 14: 5925-5942, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31534331

RESUMO

Mesenchymal stem cells (MSCs) intrinsically possess unique features that not only help in their migration towards the tumor-rich environment but they also secrete versatile types of secretomes to induce nerve regeneration and analgesic effects at inflammatory sites. As a matter of course, engineering MSCs to enhance their intrinsic abilities is growing in interest in the oncology and regenerative field. However, the concern of possible tumorigenesis of genetically modified MSCs prompted the development of non-viral transfected MSCs armed with nanotechnology for more effective cancer and regenerative treatment. Despite the fact that a large number of successful studies have expanded our current knowledge in tumor-specific targeting, targeting damaged brain site remains enigmatic due to the presence of a blood-brain barrier (BBB). A BBB is a barrier that separates blood from brain, but MSCs with intrinsic features of transmigration across the BBB can efficiently deliver desired drugs to target sites. Importantly, MSCs, when mediated by nanoparticles, can further enhance tumor tropism and can regenerate the damaged neurons in the central nervous system through the promotion of axon growth. This review highlights the homing and nerve regenerative abilities of MSCs in order to provide a better understanding of potential cell therapeutic applications of non-genetically engineered MSCs with the aid of nanotechnology.


Assuntos
Neoplasias Encefálicas/terapia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Nanotecnologia/métodos , Regeneração Nervosa , Animais , Neoplasias Encefálicas/irrigação sanguínea , Neoplasias Encefálicas/patologia , Humanos , Tropismo
17.
Sci Rep ; 9(1): 6884, 2019 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-31053741

RESUMO

Mast cells play a crucial role in allergic diseases via the release of inflammatory mediators, particularly histamine and pro-inflammatory cytokines. Avenanthramide (Avn) C, a polyphenol found mainly in oats, is known to exhibit various biological properties. In this study, we aimed to evaluate the effectiveness of Avn C from germinated oats against mast cell-mediated allergic inflammation. For the in vitro study, RBL-2H3, mouse bone marrow-derived mast cells and rat peritoneal mast cells were used. Avn C (1-100 nM) inhibited the immunoglobulin (Ig)E-stimulated mast cells degranulation by suppressing phosphorylation of phosphoinositide 3-kinase and phospholipase Cγ1 and decreasing intracellular calcium levels. It inhibited IgE-stimulated secretion of inflammatory cytokines via suppression of FcεRI-mediated signaling proteins Lyn, Syk, Akt, and nuclear factor-κB. To verify the effects of Avn C in vivo, ovalbumin-induced active systemic anaphylaxis (ASA) and IgE-mediated passive cutaneous anaphylaxis (PCA) models were used. Oral administration of Avn C dose-dependently attenuated the ASA reactions, as evidenced by the inhibition of hypothermia and reduction of elevated serum histamine, IgE, and interleukin-4 levels. Avn C also inhibited the PCA reactions, such as ear swelling and plasma extravasation. Our results suggested that Avn C from germinated oats might be a possible therapeutic candidate for mast cell-mediated allergic inflammation.


Assuntos
Antialérgicos/farmacologia , Anti-Inflamatórios/farmacologia , Avena/química , Avena/crescimento & desenvolvimento , Germinação , Mastócitos/efeitos dos fármacos , ortoaminobenzoatos/farmacologia , Animais , Degranulação Celular/efeitos dos fármacos , Citocinas/metabolismo , Masculino , Mastócitos/imunologia , Camundongos , Camundongos Endogâmicos ICR
18.
Chem Biol Interact ; 298: 1-7, 2019 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-30392763

RESUMO

Mast cells play a major role in immunoglobulin E-mediated allergic inflammation, which is involved in asthma, atopic dermatitis, and allergic rhinitis. Nothofagin has been shown to ameliorate various inflammatory responses such as the septic response and vascular inflammation. In this study, we assessed the inhibitory effect of nothofagin on allergic inflammation using cultured/isolated mast cells and an anaphylaxis mouse model. Nothofagin treatment prevented histamine and ß-hexosaminidase release by reducing the influx of calcium into the cytosol in a concentration-dependent manner. Nothofagin also inhibited the gene expression and secretion of pro-inflammatory cytokines such as tumor necrosis factor-α and interleukin-4 by downregulating the phosphorylation of Lyn, Syk, Akt and nuclear translocation of nuclear factor-κB. To confirm these effects of nothofagin in vivo, we used a passive cutaneous anaphylaxis mouse model. Topical administration of nothofagin suppressed local pigmentation and ear thickness. Taken together, these results suggest nothofagin as a potential candidate for the treatment of mast cell-involved allergic inflammatory diseases.


Assuntos
Chalconas/farmacologia , Hipersensibilidade/tratamento farmacológico , Inflamação/prevenção & controle , Mastócitos/efeitos dos fármacos , Anafilaxia/etiologia , Anafilaxia/prevenção & controle , Animais , Cálcio/metabolismo , Degranulação Celular/efeitos dos fármacos , Citocinas/genética , Citocinas/metabolismo , Modelos Animais de Doenças , Regulação da Expressão Gênica/efeitos dos fármacos , Hipersensibilidade/complicações , Hipersensibilidade/patologia , Inflamação/etiologia , Inflamação/patologia , Masculino , Mastócitos/fisiologia , Camundongos Endogâmicos ICR , NF-kappa B/antagonistas & inibidores , NF-kappa B/metabolismo , Ratos Sprague-Dawley
19.
Front Pharmacol ; 9: 591, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29930511

RESUMO

Mast cells are major effector cells for allergic responses that act by releasing inflammatory mediators, such as histamine and pro-inflammatory cytokines. Accordingly, different strategies have been pursued to develop anti-allergic and anti-inflammatory candidates by regulating the function of mast cells. The purpose of this study was to determine the effectiveness of elaeocarpusin (EL) on mast cell-mediated allergic inflammation. We isolated EL from Elaeocarpus sylvestris L. (Elaeocarpaceae), which is known to possess anti-inflammatory properties. For this study, various sources of mast cells and mouse anaphylaxis models were used. EL suppressed the induction of markers for mast cell degranulation, such as histamine and ß-hexosaminidase, by reducing intracellular calcium levels. Expression of pro-inflammatory cytokines, such as tumor necrosis factor-α and IL-4, was significantly decreased in activated mast cells by EL. This inhibitory effect was related to inhibition of the phosphorylation of Fyn, Lyn, Syk, and Akt, and the nuclear translocation of nuclear factor-κB. To confirm the effect of EL in vivo, immunoglobulin E-mediated passive cutaneous anaphylaxis (PCA) and ovalbumin-induced active systemic anaphylaxis (ASA) models were induced. EL reduced the PCA reaction in a dose dependent manner. In addition, EL attenuated ASA reactions such as hypothemia, histamine release, and IgE production. Our results suggest that EL is a potential therapeutic candidate for allergic inflammatory diseases that acts via the inhibition of mast cell degranulation and expression of proinflammatory cytokines.

20.
Adv Sci (Weinh) ; 5(5): 1700860, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-29876212

RESUMO

Lung cancer is a highly malignant tumor, and targeted delivery of anti-cancer drugs to deep lung tumor tissue remains a challenge in drug design. Here, it is demonstrated that bone marrow mesenchymal stem cells armed with nanodrugs are highly targeted and mutually destructive with malignant lung cancer cells and successfully eradicate lung tumors tissues. Using this approach, the current clinical dose of anti-cancer drugs for the treatment of malignant lung tumors can be decreased by more than 100-fold without triggering immunotoxicity.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA