Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Front Cell Dev Biol ; 12: 1359451, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38694823

RESUMO

Immunotherapy has changed the landscape of treatment options for patients with hepatocellular cancer. Checkpoint inhibitors are now standard of care for patients with advanced tumours, yet the majority remain resistant to this therapy and urgent approaches are needed to boost the efficacy of these agents. Targeting the liver endothelial cells, as the orchestrators of immune cell recruitment, within the tumour microenvironment of this highly vascular cancer could potentially boost immune cell infiltration. We demonstrate the successful culture of primary human liver endothelial cells in organ-on-a-chip technology followed by perfusion of peripheral blood mononuclear cells. We confirm, with confocal and multiphoton imaging, the capture and adhesion of immune cells in response to pro-inflammatory cytokines in this model. This multicellular platform sets the foundation for testing the efficacy of new therapies in promoting leukocyte infiltration across liver endothelium as well as a model for testing cell therapy, such as chimeric antigen receptor (CAR)-T cell, capture and migration across human liver endothelium.

2.
Nat Commun ; 14(1): 4895, 2023 08 14.
Artigo em Inglês | MEDLINE | ID: mdl-37580395

RESUMO

The opportunistic fungal pathogen Cryptococcus neoformans causes lethal infections in immunocompromised patients. Macrophages are central to the host response to cryptococci; however, it is unclear how C. neoformans is recognised and phagocytosed by macrophages. Here we investigate the role of TLR4 in the non-opsonic phagocytosis of C. neoformans. We find that loss of TLR4 function unexpectedly increases phagocytosis of non-opsonised cryptococci by murine and human macrophages. The increased phagocytosis observed in Tlr4-/- cells was dampened by pre-treatment of macrophages with oxidised-LDL, a known ligand of scavenger receptors. The scavenger receptor, macrophage scavenger receptor 1 (MSR1) (also known as SR-A1 or CD204) was upregulated in Tlr4-/- macrophages. Genetic ablation of MSR1 resulted in a 75% decrease in phagocytosis of non-opsonised cryptococci, strongly suggesting that it is a key non-opsonic receptor for this pathogen. We go on to show that MSR1-mediated uptake likely involves the formation of a multimolecular signalling complex involving FcγR leading to SYK, PI3K, p38 and ERK1/2 activation to drive actin remodelling and phagocytosis. Altogether, our data indicate a hitherto unidentified role for TLR4/MSR1 crosstalk in the non-opsonic phagocytosis of C. neoformans.


Assuntos
Criptococose , Fagocitose , Receptores Depuradores Classe A , Receptor 4 Toll-Like , Animais , Humanos , Camundongos , Cryptococcus neoformans , Macrófagos/microbiologia , Receptor 4 Toll-Like/genética , Receptores Depuradores Classe A/metabolismo
3.
Semin Liver Dis ; 42(1): 61-76, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34553345

RESUMO

The scavenger receptor superfamily represents a highly diverse collection of evolutionarily-conserved receptors which are known to play key roles in host homeostasis, the most prominent of which is the clearance of unwanted endogenous macromolecules, such as oxidized low-density lipoproteins, from the systemic circulation. Members of this family have also been well characterized in their binding and internalization of a vast range of exogenous antigens and, consequently, are generally considered to be pattern recognition receptors, thus contributing to innate immunity. Several studies have implicated scavenger receptors in the pathophysiology of several inflammatory diseases, such as Alzheimer's and atherosclerosis. Hepatic resident cellular populations express a diverse complement of scavenger receptors in keeping with the liver's homeostatic functions, but there is gathering interest in the contribution of these receptors to hepatic inflammation and its complications. Here, we review the expression of scavenger receptors in the liver, their functionality in liver homeostasis, and their role in inflammatory liver disease and cancer.


Assuntos
Aterosclerose , Hepatite , Neoplasias , Aterosclerose/etiologia , Humanos , Inflamação , Receptores Depuradores/metabolismo
5.
Eur J Pharmacol ; 913: 174618, 2021 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-34762934

RESUMO

Fibrosis is the formation of scar tissue due to injury or long-term inflammation and is a leading cause of morbidity and mortality. Activation of the pro-fibrotic cytokine transforming growth factor-ß (TGFß) via the alpha-V beta-6 (αvß6) integrin has been identified as playing a key role in the development of fibrosis. Therefore, a drug discovery programme to identify an orally bioavailable small molecule αvß6 arginyl-glycinyl-aspartic acid (RGD)-mimetic was initiated. As part of a medicinal chemistry programme GSK3335103 was identified and profiled in a range of pre-clinical in vitro and in vivo systems. GSK3335103 was shown to bind to the αvß6 with high affinity and demonstrated fast binding kinetics. In primary human lung epithelial cells, GSK3335103-induced concentration- and time-dependent internalisation of αvß6 with a rapid return of integrin to the cell surface observed after washout. Following sustained engagement of the αvß6 integrin in vitro, lysosomal degradation was induced by GSK3335103. GSK3335103 was shown to engage with the αvß6 integrin and inhibit the activation of TGFß in both ex vivo IPF tissue and in a murine model of bleomycin-induced lung fibrosis, as measured by αvß6 engagement, TGFß signalling and collagen deposition, with a prolonged duration of action observed in vivo. In summary, GSK3335103 is a potent αvß6 inhibitor that attenuates TGFß signalling in vitro and in vivo with a well-defined pharmacokinetic/pharmacodynamic relationship. This translates to a significant reduction of collagen deposition in vivo and therefore GSK3335103 represents a potential novel oral therapy for fibrotic disorders.


Assuntos
Antifibróticos/farmacologia , Integrinas/antagonistas & inibidores , Fibrose Pulmonar/tratamento farmacológico , Administração Oral , Animais , Antifibróticos/química , Antifibróticos/uso terapêutico , Antígenos de Neoplasias/química , Antígenos de Neoplasias/metabolismo , Disponibilidade Biológica , Bleomicina/administração & dosagem , Bleomicina/toxicidade , Células Cultivadas , Modelos Animais de Doenças , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/patologia , Humanos , Integrinas/química , Integrinas/metabolismo , Pulmão/efeitos dos fármacos , Pulmão/patologia , Lisossomos/metabolismo , Masculino , Camundongos , Oligopeptídeos/química , Cultura Primária de Células , Proteólise/efeitos dos fármacos , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/patologia , Fator de Crescimento Transformador beta/metabolismo
6.
J Vis Exp ; (175)2021 09 30.
Artigo em Inglês | MEDLINE | ID: mdl-34661571

RESUMO

The aggressiveness and lack of well-tolerated and widely effective treatments for advanced hepatocellular carcinoma (HCC), the predominant form of liver cancer, rationalize its rank as the second most common cause of cancer-related death. Preclinical models need to be adapted to recapitulate the human conditions to select the best therapeutic candidates for clinical development and aid the delivery of personalized medicine. Three-dimensional (3D) cellular spheroid models show promise as an emerging in vitro alternative to two-dimensional (2D) monolayer cultures. Here, we describe a 3D tumor spheroid model which exploits the ability of individual cells to aggregate when maintained in hanging droplets, and is more representative of an in vivo environment than standard monolayers. Furthermore, 3D spheroids can be produced by combining homotypic or heterotypic cells, more reflective of the cellular heterogeneity in vivo, potentially enabling the study of environmental interactions that can influence progression and treatment responses. The current research optimized the cell density to form 3D homotypic and heterotypic tumor spheroids by immobilizing cell suspensions on the lids of standard 10 cm3 Petri dishes. Longitudinal analysis was performed to generate growth curves for homotypic versus heterotypic tumor/fibroblasts spheroids. Finally, the proliferative impact of fibroblasts (COS7 cells) and liver myofibroblasts (LX2) on homotypic tumor (Hep3B) spheroids was investigated. A seeding density of 3,000 cells (in 20 µL media) successfully yielded Huh7/COS7 heterotypic spheroids, which displayed a steady increase in size up to culture day 8, followed by growth retardation. This finding was corroborated using Hep3B homotypic spheroids cultured in LX2 (human hepatic stellate cell line) conditioned medium (CM). LX2 CM triggered the proliferation of Hep3B spheroids compared to control tumor spheroids. In conclusion, this protocol has shown that 3D tumor spheroids can be used as a simple, economical, and prescreen in vitro tool to study tumor-stromal interactions more comprehensively.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Linhagem Celular , Humanos , Esferoides Celulares
7.
J Pharmacol Exp Ther ; 376(2): 273-280, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33318076

RESUMO

The arginyl-glycinyl-aspartic acid (RGD) integrin alpha-v beta-6 (αvß6) has been identified as playing a key role in the activation of transforming growth factor-ß (TGFß) that is hypothesized to be pivotal in the development of fibrosis and other diseases. In this study, αvß6 small molecule inhibitors were characterized in a range of in vitro systems to determine affinity, kinetics, and duration of TGFß inhibition. High αvß6 binding affinity was shown to be correlated with slow dissociation kinetics. Compound 1 (high αvß6 affinity, slow dissociation) and SC-68448 (low αvß6 affinity, fast dissociation) induced concentration- and time-dependent internalization of αvß6 in normal human bronchial epithelial (NHBE) cells. After washout, the αvß6 cell surface repopulation was faster for SC-68448 compared with compound 1 In addition, αvß6-dependent release of active TGFß from NHBE cells was inhibited by compound 1 and SC-68448. After washout of SC-68448, release of active TGFß was restored, whereas after washout of compound 1 the inhibition of TGFß activation was maintained and only reversible in the presence of a lysosomal inhibitor (chloroquine). However, SC-68448 was able to reduce total levels of αvß6 in NHBE cells if present continuously. These observations suggest αvß6 can be degraded after high affinity RGD binding that sorts the integrin for lysosomal degradation after internalization, likely due to sustained engagement as a result of slow dissociation kinetics. In addition, the αvß6 integrin can also be downregulated after sustained engagement of the RGD binding site with low affinity ligands that do not sort the integrin for immediate lysosomal degradation. SIGNIFICANCE STATEMENT: The fate of RGD integrin after ligand binding has not been widely investigated. Using the αvß6 integrin as a case study, we have demonstrated that RGD-induced downregulation of αvß6 is both affinity and time dependent. High affinity ligands induced downregulation via lysosomal degradation, likely due to slow dissociation, whereas sustained low affinity ligand engagement was only able to decrease αvß6 expression over longer periods of time. Our study provides a potential unique mechanism for obtaining duration of action for drugs targeting integrins.


Assuntos
Antígenos de Neoplasias/metabolismo , Regulação para Baixo , Integrinas/metabolismo , Fator de Crescimento Transformador beta/antagonistas & inibidores , Antígenos de Neoplasias/química , Sítios de Ligação , Células Cultivadas , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Humanos , Integrinas/química , Cinética , Lisossomos/metabolismo , Oligopeptídeos/metabolismo , Fenilpropionatos/farmacologia , Ligação Proteica , Proteólise , Mucosa Respiratória/citologia , Fator de Crescimento Transformador beta/metabolismo
8.
Front Physiol ; 11: 990, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32982772

RESUMO

Liver sinusoidal endothelial cells (LSEC) form a unique barrier between the liver sinusoids and the underlying parenchyma, and thus play a crucial role in maintaining metabolic and immune homeostasis, as well as actively contributing to disease pathophysiology. Whilst their endocytic and scavenging function is integral for nutrient exchange and clearance of waste products, their capillarisation and dysfunction precedes fibrogenesis. Furthermore, their ability to promote immune tolerance and recruit distinct immunosuppressive leukocyte subsets can allow persistence of chronic viral infections and facilitate tumour development. In this review, we present the immunological and barrier functions of LSEC, along with their role in orchestrating fibrotic processes which precede tumourigenesis. We also summarise the role of LSEC in modulating the tumour microenvironment, and promoting development of a pre-metastatic niche, which can drive formation of secondary liver tumours. Finally, we summarise closely inter-linked disease pathways which collectively perpetuate pathogenesis, highlighting LSEC as novel targets for therapeutic intervention.

9.
Front Immunol ; 11: 650, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32528462

RESUMO

The liver is our largest internal organ and it plays major roles in drug detoxification and immunity, where the ingestion of extracellular material through phagocytosis is a critical pathway. Phagocytosis is the deliberate endocytosis of large particles, microbes, dead cells or cell debris and can lead to cell-in-cell structures. Various types of cell endocytosis have been recently described for hepatic epithelia (hepatocytes), which are non-professional phagocytes. Given that up to 80% of the liver comprises hepatocytes, the biological impact of cell-in-cell structures in the liver can have profound effects in liver regeneration, inflammation and cancer. This review brings together the latest reports on four types of endocytosis in the liver -efferocytosis, entosis, emperipolesis and enclysis, with a focus on hepatocyte biology.


Assuntos
Compartimento Celular/fisiologia , Emperipolese/fisiologia , Endocitose/fisiologia , Entose/fisiologia , Hepatócitos/fisiologia , Fígado/citologia , Animais , Humanos , Imunidade , Inativação Metabólica , Fígado/metabolismo , Regeneração Hepática , Fagocitose
10.
Front Oncol ; 10: 565950, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-34354939

RESUMO

Scavenger receptor class F member 1 (SCARF1) is thought to play an important role in the selective recruitment of CD4+ T cells to liver sinusoidal endothelial cells during chronic liver disease. However, the contribution of SCARF1 to hepatocellular carcinoma (HCC) is currently unknown. We utilized publically-available RNA-sequencing data from The Cancer Genome Atlas (TGCA) to explore SCARF1 expression in HCC and correlated it with a number of clinicopathological features. Flow adhesion assays were used to determine the role of SCARF1 in CD4+ T cell subset recruitment. SCARF1 expression was downregulated in HCC tumor tissues, compared to non-tumoral tissues, and loss of SCARF1 expression was associated with poorly differentiated/aggressive tumors. Additionally, higher SCARF1 expression in HCC tumor tissues was highly prognostic of better overall, disease-free and progression-free survival. SCARF1 within HCC was largely associated with tumor endothelial cells and adhesion studies suggested that it played a role in the specific recruitment of proinflammatory CD4+ T cells (CD4+CD25-) to HCC tumor tissues. Endothelial SCARF1 expression in tumor biopsies may provide critical prognostic information. Additionally, SCARF1 may also be a novel endothelial target that could help re-programme the microenvironment of HCC by promoting effector T cell tumor infiltration.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA