Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros











Base de dados
Tipo de estudo
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 121(26): e2319322121, 2024 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-38900789

RESUMO

Thymocyte selection-associated high-mobility group box (TOX) is a transcription factor that is crucial for T cell exhaustion during chronic antigenic stimulation, but its role in inflammation is poorly understood. Here, we report that TOX extracellularly mediates drastic inflammation upon severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection by binding to the cell surface receptor for advanced glycation end-products (RAGE). In various diseases, including COVID-19, TOX release was highly detectable in association with disease severity, contributing to lung fibroproliferative acute respiratory distress syndrome (ARDS). Recombinant TOX-induced blood vessel rupture, similar to a clinical signature in patients experiencing a cytokine storm, further exacerbating respiratory function impairment. In contrast, disruption of TOX function by a neutralizing antibody and genetic removal of RAGE diminished TOX-mediated deleterious effects. Altogether, our results suggest an insight into TOX function as an inflammatory mediator and propose the TOX-RAGE axis as a potential target for treating severe patients with pulmonary infection and mitigating lung fibroproliferative ARDS.


Assuntos
COVID-19 , Receptor para Produtos Finais de Glicação Avançada , SARS-CoV-2 , Humanos , Receptor para Produtos Finais de Glicação Avançada/metabolismo , COVID-19/imunologia , COVID-19/metabolismo , COVID-19/patologia , COVID-19/complicações , COVID-19/virologia , Animais , Camundongos , Inflamação/metabolismo , Inflamação/patologia , Síndrome do Desconforto Respiratório/imunologia , Síndrome do Desconforto Respiratório/metabolismo , Síndrome do Desconforto Respiratório/patologia , Síndrome do Desconforto Respiratório/virologia , Lesão Pulmonar/imunologia , Lesão Pulmonar/metabolismo , Lesão Pulmonar/patologia , Proteínas de Grupo de Alta Mobilidade/metabolismo , Proteínas de Grupo de Alta Mobilidade/genética , Masculino , Pulmão/patologia , Pulmão/metabolismo , Pulmão/imunologia , Feminino
2.
J Nanobiotechnology ; 21(1): 310, 2023 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-37658367

RESUMO

BACKGROUND: Basic fibroblast growth factor (bFGF) is one of the critical components accelerating angiogenesis and tissue regeneration by promoting the migration of dermal fibroblasts and endothelial cells associated with matrix formation and remodeling in wound healing process. However, clinical applications of bFGF are substantially limited by its unstable nature due to rapid decomposition under physiological microenvironment. RESULTS: In this study, we present the bFGF-loaded human serum albumin nanoparticles (HSA-bFGF NPs) as a means of enhanced stability and sustained release platform during tissue regeneration. Spherical shape of the HSA-bFGF NPs with uniform size distribution (polydispersity index < 0.2) is obtained via a simple desolvation and crosslinking process. The HSA-bFGF NPs securely load and release the intact soluble bFGF proteins, thereby significantly enhancing the proliferation and migration activity of human dermal fibroblasts. Myofibroblast-related genes and proteins were also significantly down-regulated, indicating decrease in risk of scar formation. Furthermore, wound healing is accelerated while achieving a highly organized extracellular matrix and enhanced angiogenesis in vivo. CONCLUSION: Consequently, the HSA-bFGF NPs are suggested not only as a delivery vehicle but also as a protein stabilizer for effective wound healing and tissue regeneration.


Assuntos
Fator 2 de Crescimento de Fibroblastos , Nanopartículas , Humanos , Fator 2 de Crescimento de Fibroblastos/farmacologia , Células Endoteliais , Albumina Sérica Humana , Cicatrização
3.
Biomater Adv ; 144: 213221, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36459949

RESUMO

Recently, it has been revealed that the physical microenvironment can be translated into cellular mechanosensing to direct human mesenchymal stem cell (hMSC) differentiation. Graphene oxide (GO), a major derivative of graphene, has been regarded as a promising material for stem cell lineage specification due to its biocompatibility and unique physical properties to interact with stem cells. Especially, the lateral size of GO flakes is regarded as the key factor regulating cellular response caused by GO. In this work, GO that had been mechanically created and had an average diameter of 0.9, 1.1, and 1.7 m was produced using a ball-mill process. When size-controlled GO flakes were applied to hMSCs, osteogenic differentiation was enhanced by GO with a specific average diameter of 1.7 µm. It was confirmed that osteogenic differentiation was increased due to the enhanced expression of focal adhesion and the development of focal adhesion subordinate signals via extracellular signal-regulated kinase (ERK)-mitogen-activated protein kinase (MEK) pathway. These results suggest that size-controlled GO flakes could be efficient materials for promoting osteogenesis of hMSCs. Results of this study could also improve our understanding of the correlation between hMSCs and cellular responses to GO.


Assuntos
Grafite , Células-Tronco Mesenquimais , Humanos , Osteogênese , Células-Tronco Mesenquimais/metabolismo , Diferenciação Celular , MAP Quinases Reguladas por Sinal Extracelular/metabolismo
4.
Appl Microbiol Biotechnol ; 106(22): 7531-7545, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36227339

RESUMO

Arginine deiminase (ADI) is a microbial-derived enzyme which catalyzes the conversion of L-arginine into L-citrulline. ADI originating from Mycoplasma has been reported to present anti-tumor activity against arginine-auxotrophic tumors, including melanoma. Melanoma cells are sensitive to arginine depletion due to reduced expression of argininosuccinate synthase 1 (ASS1), a key enzyme for arginine biosynthesis. However, clinical applications of recombinant ADI for melanoma treatment present some limitations. Since recombinant ADI is not human-derived, it shows instability, proteolytic degradation, and antigenicity in human serum. In addition, there is a problem of drug resistance issue due to the intracellular expression of once-silenced ASS1. Moreover, recombinant ADI proteins are mainly expressed as inclusion body forms in Escherichia coli and require a time-consuming refolding process to turn them back into active form. Herein, we propose fusion of recombinant ADI from Mycoplasma hominis and 30Kc19α, a cell-penetrating protein which also increases stability and soluble expression of cargo proteins, to overcome these problems. We inserted matrix metalloproteinase-2 cleavable linker between ADI and 30Kc19α to increase enzyme activity in melanoma cells. Compared to ADI, ADI-LK-30Kc19α showed enhanced solubility, stability, and cell penetration. The fusion protein demonstrated selective cytotoxicity and reduced drug resistance in melanoma cells, thus would be a promising strategy for the improved efficacy in melanoma treatment. KEY POINTS: • Fusion of ADI with 30Kc19α enhances soluble expression and productivity of recombinant ADI in E. coli • 30Kc19α protects ADI from the proteolytic degradation by shielding effect, helping ADI to remain active • Intracellular delivery of ADI by 30Kc19α overcomes ADI resistance in melanoma cells by degrading intracellularly expressed arginine.


Assuntos
Metaloproteinase 2 da Matriz , Melanoma , Humanos , Escherichia coli/genética , Escherichia coli/metabolismo , Polietilenoglicóis , Argininossuccinato Sintase/metabolismo , Hidrolases/genética , Hidrolases/farmacologia , Hidrolases/metabolismo , Melanoma/tratamento farmacológico , Arginina/metabolismo , Linhagem Celular Tumoral
5.
Artif Cells Nanomed Biotechnol ; 50(1): 278-285, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36191335

RESUMO

As the acute lymphoblastic leukaemia (ALL) develops, expression of L-asparaginase (ASNase) protein is known to decrease. Therefore, deficiency of the ASNase protein would be regarded as one of the significant indications of the ALL. For the treatment of ALL, recombinant ASNase protein derived from bacterial origin is used which causes cytotoxicity by deprivation of Asn. However, short half-life of the protein is an obstacle for medical use. In order to overcome this limit, recombinant ASNase was fused to 30Kc19 with protein-stabilizing and cell-penetrating properties. As the 30Kc19 protein may induce steric hindrance, we further added a PLGLAG linker sequence (LK) between the ASNase and 30Kc19. The treatment of ASNase-LK-30Kc19 fusion protein demonstrated enhanced stability, cell-penetrating property, and anti-cancer activity. Intracellular delivery of both the non-cleaved and cleaved forms of the protein were observed, suggesting that ASNase acted both internally and externally, performing high anti-cancer activity by effective depletion of intracellular Asn. Additionally, ASNase-LK-30Kc19 showed high selectivity towards cancer cells. In terms of the dosage, releasable ASNase from ASNase-LK-30Kc19 reached the same half-maximal inhibitory concentration at a concentration five times lower than non-releasable ASNase-30Kc19. Altogether, the findings suggest that this fusion approach has potential applications in the treatment of ALL.


Assuntos
Antineoplásicos , Leucemia-Linfoma Linfoblástico de Células Precursoras , Antineoplásicos/uso terapêutico , Asparaginase/genética , Asparaginase/farmacologia , Humanos , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo
6.
Biomater Adv ; 139: 213028, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-35882121

RESUMO

Cells can 'sense' physical cues in the surrounding microenvironment and 'react' by changing their function. Previous studies have focused on regulating the physical properties of the matrix, such as stiffness and topography, thus changing the tension 'felt' by the cell as a result. In this study, by directly applying a quantified magnetic force to the cell, a correlation between differentiation and tension was shown. The magnetic force, quantified by magnetic tweezers, was applied by incorporating magnetotactic bacteria-isolated magnetic nanoparticles (MNPs) in human mesenchymal stem cells. As the applied tension increased, the expression levels of osteogenic differentiation marker genes and proteins were proportionally upregulated. Additionally, the translocation of YAP and RUNX2, deformation of nucleus, and activation of the MAPK signaling pathway were observed in tension-based osteogenic differentiation. Our findings provide a platform for the quantitative control of tension, a key factor in stem cell differentiation, between cells and the matrix using MNPs. Furthermore, these findings improve the understanding of osteogenic differentiation by mechanotransduction.


Assuntos
Nanopartículas de Magnetita , Células-Tronco Mesenquimais , Diferenciação Celular/genética , Humanos , Mecanotransdução Celular/genética , Osteogênese/genética
7.
Adv Sci (Weinh) ; 9(26): e2201883, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35751470

RESUMO

Severe infectious diseases, such as coronavirus disease 2019 (COVID-19), can induce hypercytokinemia and multiple organ failure. In spite of the growing demand for peptide therapeutics against infectious diseases, current small molecule-based strategies still require frequent administration due to limited half-life and enzymatic digestion in blood. To overcome this challenge, a strategy to continuously express multi-level therapeutic peptide drugs on the surface of immune cells, is established. Here, chimeric T cells stably expressing therapeutic peptides are presented for treatment of severe infectious diseases. Using lentiviral system, T cells are engineered to express multi-level therapeutic peptides with matrix metallopeptidases- (MMP-) and tumor necrosis factor alpha converting enzyme- (TACE-) responsive cleavage sites on the surface. The enzymatic cleavage releases γ-carboxyglutamic acid of protein C (PC-Gla) domain and thrombin receptor agonist peptide (TRAP), which activate endothelial protein C receptor (EPCR) and protease-activated receptor-1 (PAR-1), respectively. These chimeric T cells prevent vascular damage in tissue-engineered blood vessel and suppress hypercytokinemia and lung tissue damages in vivo, demonstrating promise for use of engineered T cells against sepsis and other infectious-related diseases.


Assuntos
COVID-19 , Doenças Transmissíveis , Antígenos CD/metabolismo , Antígenos CD/farmacologia , Síndrome da Liberação de Citocina , Células Endoteliais/metabolismo , Humanos , Peptídeos/metabolismo , Receptor PAR-1/metabolismo , Receptores de Superfície Celular/metabolismo , Linfócitos T/metabolismo
8.
Adv Healthc Mater ; 4(9): 1339-47, 2015 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-25846518

RESUMO

Chondrogenic commitments of mesenchymal stem cells (MSCs) require 3D cellular organization. Furthermore, recent progresses in bioreactor technology have contributed to the development of various biophysical stimulation platforms for efficient cartilage tissue formation. Here, an approach is reported to drive 3D cellular organization and enhance chondrogenic commitment of bone-marrow-derived human mesenchymal stem cells (BM-hMSCs) via magnetic nanoparticle (MNP)-mediated physical stimuli. MNPs isolated from Magnetospirillum sp. AMB-1 are endocytosed by the BM-hMSCs in a highly efficient manner. MNPs-incorporated BM-hMSCs are pelleted and then subjected to static magnetic field and/or magnet-derived shear stress. Magnetic-based stimuli enhance level of sulfated glycosaminoglycan (sGAG) and collagen synthesis, and facilitate the chondrogenic differentiation of BM-hMSCs. In addition, both static magnetic field and magnet-derived shear stress applied for the chondrogenic differentiation of BM-hMSCs do not show increament of hypertrophic differentiation. This MNP-mediated physical stimulation platform demonstrates a promising strategy for efficient cartilage tissue engineering.


Assuntos
Diferenciação Celular , Condrócitos/metabolismo , Condrogênese , Campos Magnéticos , Nanopartículas de Magnetita/química , Células-Tronco Mesenquimais/metabolismo , Condrócitos/citologia , Humanos , Magnetospirillum/química , Células-Tronco Mesenquimais/citologia , Resistência ao Cisalhamento
9.
Biomaterials ; 34(34): 8555-63, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23937911

RESUMO

Various attempts have been made to develop three-dimensional (3-D) cell culture methods because 3-D cells mimic the structures and functional properties of real tissue compared with those of monolayer cultures. Here, we report on a highly simple and efficient 3-D spheroid generation method based on a magnetic pin-array system to concentrate magnetic nanoparticle-incorporated cells in a focal direction. This system was comprised only of external magnets and magnetically induced iron pins to generate a concentrated magnetic field for attracting cells in a focused direction. 3-D spheroid generation was achieved simply by adding magnetic nanoparticle-incorporated cells into a well and covering the plate with a magnetic lid. Cell clustering occurred rapidly within 5 min and created more compact cells with time through the focused magnetic force. This system ensured not only reproducible and size-controlled generation of spheroids but also versatile types of spheroids such as random mixed, core-shell, and fused spheroids, providing a very useful tool for various biological applications.


Assuntos
Técnicas de Cultura de Células/métodos , Magnetismo , Nanopartículas/química , Esferoides Celulares/citologia , Células da Medula Óssea , Sobrevivência Celular , Simulação por Computador , Humanos , Campos Magnéticos , Microscopia Confocal
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA