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











Intervalo de año de publicación
1.
Chem Phys Lipids ; 243: 105178, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35122738

RESUMEN

Lipid nanoparticles (LNPs) mediated mRNA delivery has gained prominence due to the success of mRNA vaccines against Covid-19, without which it would not have been possible. However, there is little clinical validation of this technology for other mRNA-based therapeutic approaches. Systemic administration of LNPs predominantly targets the liver, but delivery to other organs remains a challenge. Local approaches remain a viable option for some disease indications, such as Cystic Fibrosis, where aerosolized delivery to airway epithelium is the preferred route of administration. With this in mind, novel cationic lipids (L1-L4) have been designed, synthesized and co-formulated with a proprietary ionizable lipid. These LNPs were further nebulized, along with baseline control DOTAP-based LNP (DOTAP+), and tested in vitro for mRNA integrity and encapsulation efficiency, as well as transfection efficiency and cytotoxicity in cell cultures. Improved biodegradability and potentially superior elimination profiles of L1-L4, in part due to physicochemical characteristics of putative metabolites, are thought to be advantageous for prospective therapeutic lung delivery applications using these lipids.


Asunto(s)
Liposomas/química , Pulmón , Nanopartículas/química , ARN Mensajero/administración & dosificación , Humanos
2.
Sci Adv ; 6(26): eaba4353, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32637608

RESUMEN

Fibroblast-like synoviocytes (FLS) are joint-lining cells that promote rheumatoid arthritis (RA) pathology. Current disease-modifying antirheumatic agents (DMARDs) operate through systemic immunosuppression. FLS-targeted approaches could potentially be combined with DMARDs to improve control of RA without increasing immunosuppression. Here, we assessed the potential of immunoglobulin-like domains 1 and 2 (Ig1&2), a decoy protein that activates the receptor tyrosine phosphatase sigma (PTPRS) on FLS, for RA therapy. We report that PTPRS expression is enriched in synovial lining RA FLS and that Ig1&2 reduces migration of RA but not osteoarthritis FLS. Administration of an Fc-fusion Ig1&2 attenuated arthritis in mice without affecting innate or adaptive immunity. Furthermore, PTPRS was down-regulated in FLS by tumor necrosis factor (TNF) via a phosphatidylinositol 3-kinase-mediated pathway, and TNF inhibition enhanced PTPRS expression in arthritic joints. Combination of ineffective doses of TNF inhibitor and Fc-Ig1&2 reversed arthritis in mice, providing an example of synergy between FLS-targeted and immunosuppressive DMARD therapies.


Asunto(s)
Antirreumáticos , Artritis Reumatoide , Sinoviocitos , Animales , Antirreumáticos/uso terapéutico , Células Cultivadas , Fibroblastos/metabolismo , Ratones , Sinoviocitos/metabolismo , Sinoviocitos/patología , Factor de Necrosis Tumoral alfa/metabolismo
3.
Ann Rheum Dis ; 78(5): 600-609, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30808624

RESUMEN

OBJECTIVE: We aimed to understand the role of the tyrosine phosphatase PTPN14-which in cancer cells modulates the Hippo pathway by retaining YAP in the cytosol-in fibroblast-like synoviocytes (FLS) from patients with rheumatoid arthritis (RA). METHODS: Gene/protein expression levels were measured by quantitative PCR and/or Western blotting. Gene knockdown in RA FLS was achieved using antisense oligonucleotides. The interaction between PTPN14 and YAP was assessed by immunoprecipitation. The cellular localisation of YAP and SMAD3 was examined via immunofluorescence. SMAD reporter studies were carried out in HEK293T cells. The RA FLS/cartilage coimplantation and passive K/BxN models were used to examine the role of YAP in arthritis. RESULTS: RA FLS displayed overexpression of PTPN14 when compared with FLS from patients with osteoarthritis (OA). PTPN14 knockdown in RA FLS impaired TGFß-dependent expression of MMP13 and potentiation of TNF signalling. In RA FLS, PTPN14 formed a complex with YAP. Expression of PTPN14 or nuclear YAP-but not of a non-YAP-interacting PTPN14 mutant-enhanced SMAD reporter activity. YAP promoted TGFß-dependent SMAD3 nuclear localisation in RA FLS. Differences in epigenetic marks within Hippo pathway genes, including YAP, were found between RA FLS and OA FLS. Inhibition of YAP reduced RA FLS pathogenic behaviour and ameliorated arthritis severity. CONCLUSION: In RA FLS, PTPN14 and YAP promote nuclear localisation of SMAD3. YAP enhances a range of RA FLS pathogenic behaviours which, together with epigenetic evidence, points to the Hippo pathway as an important regulator of RA FLS behaviour.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Proteínas Tirosina Fosfatasas no Receptoras/fisiología , Transducción de Señal/fisiología , Sinoviocitos/metabolismo , Factores de Transcripción/fisiología , Factor de Crecimiento Transformador beta/fisiología , Animales , Artritis Reumatoide/metabolismo , Proteínas de Ciclo Celular/fisiología , Humanos , Ratones , Proteínas Señalizadoras YAP
4.
J Clin Invest ; 129(3): 1193-1210, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30620725

RESUMEN

Genetic variants at the PTPN2 locus, which encodes the tyrosine phosphatase PTPN2, cause reduced gene expression and are linked to rheumatoid arthritis (RA) and other autoimmune diseases. PTPN2 inhibits signaling through the T cell and cytokine receptors, and loss of PTPN2 promotes T cell expansion and CD4- and CD8-driven autoimmunity. However, it remains unknown whether loss of PTPN2 in FoxP3+ regulatory T cells (Tregs) plays a role in autoimmunity. Here we aimed to model human autoimmune-predisposing PTPN2 variants, the presence of which results in a partial loss of PTPN2 expression, in mouse models of RA. We identified that reduced expression of Ptpn2 enhanced the severity of autoimmune arthritis in the T cell-dependent SKG mouse model and demonstrated that this phenotype was mediated through a Treg-intrinsic mechanism. Mechanistically, we found that through dephosphorylation of STAT3, PTPN2 inhibits IL-6-driven pathogenic loss of FoxP3 after Tregs have acquired RORγt expression, at a stage when chromatin accessibility for STAT3-targeted IL-17-associated transcription factors is maximized. We conclude that PTPN2 promotes FoxP3 stability in mouse RORγt+ Tregs and that loss of function of PTPN2 in Tregs contributes to the association between PTPN2 and autoimmunity.


Asunto(s)
Artritis Reumatoide/inmunología , Proteína Tirosina Fosfatasa no Receptora Tipo 2/inmunología , Linfocitos T Reguladores/inmunología , Animales , Artritis Reumatoide/genética , Artritis Reumatoide/patología , Modelos Animales de Enfermedad , Femenino , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/inmunología , Interleucina-17/genética , Interleucina-17/inmunología , Interleucina-6/genética , Interleucina-6/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/inmunología , Proteína Tirosina Fosfatasa no Receptora Tipo 2/genética , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/inmunología , Linfocitos T Reguladores/patología
5.
Nat Commun ; 8(1): 1060, 2017 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-29057934

RESUMEN

Systemic sclerosis (SSc) is an autoimmune disease characterized by fibrosis of skin and internal organs. Protein tyrosine phosphatases have received little attention in the study of SSc or fibrosis. Here, we show that the tyrosine phosphatase PTP4A1 is highly expressed in fibroblasts from patients with SSc. PTP4A1 and its close homolog PTP4A2 are critical promoters of TGFß signaling in primary dermal fibroblasts and of bleomycin-induced fibrosis in vivo. PTP4A1 promotes TGFß signaling in human fibroblasts through enhancement of ERK activity, which stimulates SMAD3 expression and nuclear translocation. Upstream from ERK, we show that PTP4A1 directly interacts with SRC and inhibits SRC basal activation independently of its phosphatase activity. Unexpectedly, PTP4A2 minimally interacts with SRC and does not promote the SRC-ERK-SMAD3 pathway. Thus, in addition to defining PTP4A1 as a molecule of interest for TGFß-dependent fibrosis, our study provides information regarding the functional specificity of different members of the PTP4A subclass of phosphatases.


Asunto(s)
Proteínas Inmediatas-Precoces/metabolismo , Sistema de Señalización de MAP Quinasas , Proteínas Tirosina Fosfatasas/metabolismo , Esclerodermia Sistémica/enzimología , Factor de Crecimiento Transformador beta/fisiología , Animales , Línea Celular , Dermis/patología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Fibroblastos/enzimología , Fibroblastos/metabolismo , Humanos , Proteínas Inmediatas-Precoces/antagonistas & inhibidores , Proteínas Inmediatas-Precoces/genética , Proteínas Inmediatas-Precoces/fisiología , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Tirosina Fosfatasas/antagonistas & inhibidores , Proteínas Tirosina Fosfatasas/genética , Proteínas Tirosina Fosfatasas/fisiología , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo , Esclerodermia Sistémica/metabolismo , Esclerodermia Sistémica/patología , Proteína smad3/metabolismo
6.
Curr Rheumatol Rep ; 19(5): 28, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28397126

RESUMEN

PURPOSE OF REVIEW: The pathogenesis of systemic sclerosis depends on a complex interplay between autoimmunity, vasculopathy, and fibrosis. Reversible phosphorylation on tyrosine residues, in response to growth factors and other stimuli, critically regulates each one of these three key pathogenic processes. Protein tyrosine kinases, the enzymes that catalyze addition of phosphate to tyrosine residues, are known players in systemic sclerosis, and tyrosine kinase inhibitors are undergoing clinical trials for treatment of this disease. Until recently, the role of tyrosine phosphatases-the enzymes that counteract the action of tyrosine kinases by removing phosphate from tyrosine residues-in systemic sclerosis has remained largely unknown. Here, we review the function of tyrosine phosphatases in pathways relevant to the pathogenesis of systemic sclerosis and their potential promise as therapeutic targets to halt progression of this debilitating rheumatic disease. RECENT FINDINGS: Protein tyrosine phosphatases are emerging as important regulators of a multitude of signaling pathways and undergoing validation as molecular targets for cancer and other common diseases. Recent advances in drug discovery are paving the ways to develop new classes of tyrosine phosphatase modulators to treat human diseases. Although so far only few reports have focused on tyrosine phosphatases in systemic sclerosis, these enzymes play a role in multiple pathways relevant to disease pathogenesis. Further studies in this field are warranted to explore the potential of tyrosine phosphatases as drug targets for systemic sclerosis.


Asunto(s)
Terapia Molecular Dirigida/métodos , Proteínas Tirosina Fosfatasas/fisiología , Esclerodermia Sistémica/enzimología , Endotelio Vascular/fisiopatología , Inhibidores Enzimáticos/uso terapéutico , Fibrosis , Sustancias de Crecimiento/fisiología , Humanos , Proteínas Tirosina Fosfatasas/antagonistas & inhibidores , Receptores de Interleucina/inmunología , Esclerodermia Sistémica/tratamiento farmacológico , Esclerodermia Sistémica/inmunología , Transducción de Señal/inmunología , Investigación Biomédica Traslacional/métodos
7.
Arthritis Rheumatol ; 68(2): 359-69, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26414708

RESUMEN

OBJECTIVE: During rheumatoid arthritis (RA), fibroblast-like synoviocytes (FLS) critically promote disease pathogenesis by aggressively invading the extracellular matrix of the joint. The focal adhesion kinase (FAK) signaling pathway is emerging as a contributor to the anomalous behavior of RA FLS. The receptor protein tyrosine phosphatase α (RPTPα), which is encoded by the PTPRA gene, is a key promoter of FAK signaling. The aim of this study was to investigate whether RPTPα mediates FLS aggressiveness and RA pathogenesis. METHODS: Through RPTPα knockdown, we assessed FLS gene expression by quantitative polymerase chain reaction analysis and enzyme-linked immunosorbent assay, invasion and migration by Transwell assays, survival by annexin V and propidium iodide staining, adhesion and spreading by immunofluorescence microscopy, and activation of signaling pathways by Western blotting of FLS lysates. Arthritis development was examined in RPTPα-knockout (KO) mice using the K/BxN serum-transfer model. The contribution of radiosensitive and radioresistant cells to disease was evaluated by reciprocal bone marrow transplantation. RESULTS: RPTPα was enriched in the RA synovial lining. RPTPα knockdown impaired RA FLS survival, spreading, migration, invasiveness, and responsiveness to platelet-derived growth factor, tumor necrosis factor, and interleukin-1 stimulation. These phenotypes correlated with increased phosphorylation of Src on inhibitory Y(527) and decreased phosphorylation of FAK on stimulatory Y(397) . Treatment of RA FLS with an inhibitor of FAK phenocopied the knockdown of RPTPα. RPTPα-KO mice were protected from arthritis development, which was due to radioresistant cells. CONCLUSION: By regulating the phosphorylation of Src and FAK, RPTPα mediates proinflammatory and proinvasive signaling in RA FLS, correlating with the promotion of disease in an FLS-dependent model of RA.


Asunto(s)
Artritis Experimental/genética , Artritis Reumatoide/genética , Fibroblastos/metabolismo , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Proteínas Tirosina Fosfatasas Clase 4 Similares a Receptores/genética , Familia-src Quinasas/metabolismo , Animales , Articulación del Tobillo , Apoptosis/efectos de los fármacos , Apoptosis/genética , Artritis Experimental/metabolismo , Artritis Reumatoide/metabolismo , Western Blotting , Adhesión Celular/efectos de los fármacos , Adhesión Celular/genética , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Progresión de la Enfermedad , Ensayo de Inmunoadsorción Enzimática , Fibroblastos/efectos de los fármacos , Perfilación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Interleucina-1/farmacología , Ratones , Ratones Noqueados , Fosforilación/efectos de los fármacos , Fosforilación/genética , Factor de Crecimiento Derivado de Plaquetas/farmacología , Reacción en Cadena de la Polimerasa , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Membrana Sinovial/citología , Factor de Necrosis Tumoral alfa/farmacología , Familia-src Quinasas/efectos de los fármacos
8.
Ann Rheum Dis ; 75(1): 295-302, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25378349

RESUMEN

OBJECTIVE: In rheumatoid arthritis (RA), fibroblast-like synoviocytes (FLS) that line joint synovial membranes aggressively invade the extracellular matrix, destroying cartilage and bone. As signal transduction in FLS is mediated through multiple pathways involving protein tyrosine phosphorylation, we sought to identify protein tyrosine phosphatases (PTPs) regulating the invasiveness of RA FLS. We describe that the transmembrane receptor PTPκ (RPTPκ), encoded by the transforming growth factor (TGF) ß-target gene, PTPRK, promotes RA FLS invasiveness. METHODS: Gene expression was quantified by quantitative PCR. PTP knockdown was achieved using antisense oligonucleotides. FLS invasion and migration were assessed in transwell or spot assays. FLS spreading was assessed by immunofluorescence microscopy. Activation of signalling pathways was analysed by Western blotting of FLS lysates using phosphospecific antibodies. In vivo FLS invasiveness was assessed by intradermal implantation of FLS into nude mice. The RPTPκ substrate was identified by pull-down assays. RESULTS: PTPRK expression was higher in FLS from patients with RA versus patients with osteoarthritis, resulting from increased TGFB1 expression in RA FLS. RPTPκ knockdown impaired RA FLS spreading, migration, invasiveness and responsiveness to platelet-derived growth factor, tumour necrosis factor and interleukin 1 stimulation. Furthermore, RPTPκ deficiency impaired the in vivo invasiveness of RA FLS. Molecular analysis revealed that RPTPκ promoted RA FLS migration by dephosphorylation of the inhibitory residue Y527 of SRC. CONCLUSIONS: By regulating phosphorylation of SRC, RPTPκ promotes the pathogenic action of RA FLS, mediating cross-activation of growth factor and inflammatory cytokine signalling by TGFß in RA FLS.


Asunto(s)
Artritis Reumatoide/patología , Fibroblastos/patología , Proteínas Tirosina Fosfatasas Clase 2 Similares a Receptores/fisiología , Membrana Sinovial/patología , Factor de Crecimiento Transformador beta1/fisiología , Animales , Artritis Reumatoide/metabolismo , Movimiento Celular/genética , Movimiento Celular/fisiología , Fibroblastos/metabolismo , Fibroblastos/fisiología , Fibroblastos/trasplante , Regulación Enzimológica de la Expresión Génica/fisiología , Técnicas de Silenciamiento del Gen , Xenoinjertos , Humanos , Ratones Desnudos , Proteínas Tirosina Fosfatasas/genética , Proteínas Tirosina Fosfatasas/fisiología , ARN Mensajero/genética , Proteínas Tirosina Fosfatasas Clase 2 Similares a Receptores/genética , Membrana Sinovial/metabolismo , Membrana Sinovial/trasplante , Regulación hacia Arriba
9.
J Nanosci Nanotechnol ; 14(1): 98-114, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24730253

RESUMEN

The recent advent of nanomedicine holds potential to revolutionize cancer therapy. This innovative discipline has paved the way for the emergence of a new class of drugs based on nanoengineered particles. These "nanodrugs" are designed to greatly enhance drug therapeutic indices. First-generation nanodrugs consisted of conventional anti-cancer drugs loaded into/onto nanoengineered particles (nanocarriers) devoid of targeting features (non-targeted nanodrugs). Non-targeted nanodrugs have provided the opportunity to carry large amounts of drugs, including poorly water-soluble and/or permeable drugs, to several types of tumors, improving the therapeutic index with respect to comparable free drugs. Although effective, the primary delivery mechanism of non-targeted nanodrugs was through passive tissue accumulation, due to pathophysiological differences between tumor-associated and healthy vessels, and through non-specific targeting of cell subsets, posing the danger of off-target binding and effects. Recently, the therapeutic indices of certain anti-cancer drugs were further improved by attaching targeting ligands to nanodrugs (targeted-nanodrugs). Targeted-nanodrugs selectively bind to cognate receptors expressed on target cells and enter cells more efficiently than non-targeted formulations. Although these advancements have been sufficiently beneficial to place targeted-nanodrugs into clinical development for use in cancer therapy, they also come at a price. The addition of ligands to drug-loaded nanocarriers often leads to additional synthesis steps and costs, and more complex biological performance relative to ligand-devoid nanodrugs. Here, we will discuss the benefits and challenges facing the addition of targeting features to nanodrugs for cancer therapy.


Asunto(s)
Antineoplásicos/administración & dosificación , Antineoplásicos/farmacocinética , Terapia Molecular Dirigida/métodos , Nanocápsulas/administración & dosificación , Nanocápsulas/química , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Animales , Antineoplásicos/química , Diseño de Fármacos , Humanos
10.
Bioconjug Chem ; 24(6): 852-8, 2013 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-23682992

RESUMEN

Recent evidence regarding the role of regulatory T cells (Treg) in tumor development has suggested that the manipulation of Treg function selectively in the tumor microenvironment would be a desirable immunotherapy approach. Targeting intratumor immune populations would reduce side effects on peripheral healthy cells and increase antitumor efficacy of immunotherapies. However, no current approaches are available which enable selective in vivo targeting of intratumor Treg or other immune cell subpopulations. Herein, we investigated the ability of ligands against Treg-specific receptors to drive selective internalization of PEG-modified single-walled carbon nanotubes (PEG-SWCNTs) into Treg residing in the tumor microenvironment. We focused our attention on the glucocorticoid-induced TNFR-related receptor (GITR), as it showed higher overexpression on intratumor vs peripheral (i.e., splenic) Treg compared to other reported Treg-specific markers (folate receptor 4, CD103, and CD39). Ex vivo investigations showed that the Treg targeting efficiency and selectivity of PEG-SWCNTs depended on incubation time, dose, number of ligands per nanotube, and targeted surface marker. In vivo investigations showed that PEG-SWCNTs armed with GITR ligands targeted Treg residing in a B16 melanoma more efficiently then intratumor non-Treg or splenic Treg. The latter result was achieved by exploiting a combination of passive tumor targeting due to enhanced tumor vascular permeability, naturally increased intratumor Treg vs effector T cell (Teff) ratio, and active targeting of markers that are enriched in intratumor vs splenic Treg. We also found that PEG-SWCNTs loaded with GITR ligands were internalized by Treg through receptor-mediated endocytosis and transported into the cytoplasm and nucleus ex vivo and in vivo. This is the first example of intratumor immune cell targeting and we hope it will pave the way to innovative immunotherapies against cancer.


Asunto(s)
Nanotubos de Carbono/química , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/inmunología , Polietilenglicoles/farmacología , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Animales , Relación Dosis-Respuesta a Droga , Humanos , Ratones , Ratones Endogámicos C57BL , Polietilenglicoles/química , Receptores de Antígenos de Linfocitos T/antagonistas & inhibidores , Receptores de Antígenos de Linfocitos T/inmunología , Relación Estructura-Actividad , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA