Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Biomaterials ; 281: 121370, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35032910

RESUMEN

Protein-based biomaterials offer several advantages over synthetic materials, owing to their unique stimuli-responsive properties, biocompatibility and modular nature. Here, we demonstrate that E5C, a recombinant protein block polymer, consisting of five repeats of elastin like polypeptide (E) and a coiled-coil domain of cartilage oligomeric matrix protein (C), is capable of forming a porous networked gel at physiological temperature, making it an excellent candidate for injectable biomaterials. Combination of E5C with Atsttrin, a chondroprotective engineered derivative of anti-inflammatory growth factor progranulin, provides a unique biochemical and biomechanical environment to protect against post-traumatic osteoarthritis (PTOA) onset and progression. E5C gel was demonstrated to provide prolonged release of Atsttrin and inhibit chondrocyte catabolism while facilitating anabolic signaling in vitro. We also provide in vivo evidence that prophylactic and therapeutic application of Atsttrin-loaded E5C gels protected against PTOA onset and progression in a rabbit anterior cruciate ligament transection model. Collectively, we have developed a unique protein-based gel capable of minimally invasive, sustained delivery of prospective therapeutics, particularly the progranulin-derivative Atsttrin, for therapeutic application in OA.


Asunto(s)
Lesiones del Ligamento Cruzado Anterior , Cartílago Articular , Osteoartritis , Animales , Materiales Biocompatibles/uso terapéutico , Cartílago Articular/metabolismo , Geles , Osteoartritis/tratamiento farmacológico , Osteoartritis/metabolismo , Polímeros/uso terapéutico , Progranulinas/metabolismo , Progranulinas/uso terapéutico , Conejos
2.
Arthritis Res Ther ; 24(1): 27, 2022 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-35045889

RESUMEN

BACKGROUND: Penfluridol, isolated from an FDA-approved small-molecule drug library as an inhibitor of tumor necrosis factor α (TNFα)-stimulated NF-κB activation, is clinically used to treat chronic schizophrenia and related disorders. This study is aimed to investigate the therapeutic effect of penfluridol on TNFα-stimulated inflammatory autoimmune diseases, particularly inflammatory arthritis. METHODS: Various in vitro studies to confirm the inhibitory effect of penfluridol on TNFα-induced NF-κB activity in bone marrow-derived macrophages or Raw 264.7 macrophage cell line. In vivo studies assessed the therapeutic effects of penfluridol in various disease models, including TNFα transgenic mice, collagen-induced arthritis, DSS-induced colitis, and TNBS-induced colitis. Identification and characterization of the binding of penfluridol to acid sphingomyelinase using bioinformatics and drug affinity responsive target stability assay. Acid sphingomyelinase activity assays to reveal penfluridol-mediated inhibition of acid sphingomyelinase activity. siRNA knockdown experiments to illustrate the dependence of penfluridol's anti-TNF activity on acid sphingomyelinase. RESULTS: Penfluridol effectively inhibited TNFα-induced NF-κB activation in vitro and alleviated the severity of arthritis and colitis in vivo. Mechanistic studies revealed that penfluridol bound to acid sphingomyelinase and inhibited its activation. In addition, knockdown of acid sphingomyelinase largely abolished the inhibitory effects of penfluridol on TNFα-induced inflammatory cytokine production. Furthermore, penfluridol suppressed the differentiation of spleen naive CD4+T cells to TH1 and TH17 and inhibited M1 macrophage polarization. CONCLUSION: This study provides the rationale for the possible innovative use of penfluridol as a newly identified small-molecule drug for TNFα-driven diseases, such as inflammatory arthritis and colitis.


Asunto(s)
Enfermedades Autoinmunes , Penfluridol , Animales , Enfermedades Autoinmunes/tratamiento farmacológico , Ratones , FN-kappa B/metabolismo , Esfingomielina Fosfodiesterasa , Inhibidores del Factor de Necrosis Tumoral , Factor de Necrosis Tumoral alfa/metabolismo
3.
Ann Rheum Dis ; 80(12): 1615-1627, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34226187

RESUMEN

OBJECTIVES: Osteoarthritis (OA) is the most common joint disease; however, the indeterminate nature of mechanisms by which OA develops has restrained advancement of therapeutic targets. TNF signalling has been implicated in the pathogenesis of OA. TNFR1 primarily mediates inflammation, whereas emerging evidences demonstrate that TNFR2 plays an anti-inflammatory and protective role in several diseases and conditions. This study aims to decipher TNFR2 signalling in chondrocytes and OA. METHODS: Biochemical copurification and proteomics screen were performed to isolate the intracellular cofactors of TNFR2 complex. Bulk and single cell RNA-seq were employed to determine 14-3-3 epsilon (14-3-3ε) expression in human normal and OA cartilage. Transcription factor activity screen was used to isolate the transcription factors downstream of TNFR2/14-3-3ε. Various cell-based assays and genetically modified mice with naturally occurring and surgically induced OA were performed to examine the importance of this pathway in chondrocytes and OA. RESULTS: Signalling molecule 14-3-3ε was identified as an intracellular component of TNFR2 complexes in chondrocytes in response to progranulin (PGRN), a growth factor known to protect against OA primarily through activating TNFR2. 14-3-3ε was downregulated in OA and its deficiency deteriorated OA. 14-3-3ε was required for PGRN regulation of chondrocyte metabolism. In addition, both global and chondrocyte-specific deletion of 14-3-3ε largely abolished PGRN's therapeutic effects against OA. Furthermore, PGRN/TNFR2/14-3-3ε signalled through activating extracellular signal-regulated kinase (ERK)-dependent Elk-1 while suppressing nuclear factor kappa B (NF-κB) in chondrocytes. CONCLUSIONS: This study identifies 14-3-3ε as an inducible component of TNFR2 receptor complex in response to PGRN in chondrocytes and presents a previously unrecognised TNFR2 pathway in the pathogenesis of OA.


Asunto(s)
Proteínas 14-3-3/metabolismo , Cartílago Articular/metabolismo , Condrocitos/metabolismo , Osteoartritis/metabolismo , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Animales , Cartílago Articular/citología , Humanos , Ratones , Ratones Noqueados , FN-kappa B/metabolismo , Progranulinas/metabolismo , Transducción de Señal , Proteína Elk-1 con Dominio ets/metabolismo
4.
J Clin Invest ; 131(16)2021 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-34185706

RESUMEN

TNFR1 and TNFR2 have received prominent attention because of their dominance in the pathogenesis of inflammation and autoimmunity. TNFR1 has been extensively studied and primarily mediates inflammation. TNFR2 remains far less studied, although emerging evidence demonstrates that TNFR2 plays an antiinflammatory and immunoregulatory role in various conditions and diseases. Herein, we report that TNFR2 regulates macrophage polarization, a highly dynamic process controlled by largely unidentified intracellular regulators. Using biochemical copurification and mass spectrometry approaches, we isolated the signaling molecule 14-3-3ε as a component of TNFR2 complexes in response to progranulin stimulation in macrophages. In addition, 14-3-3ε was essential for TNFR2 signaling-mediated regulation of macrophage polarization and switch. Both global and myeloid-specific deletion of 14-3-3ε resulted in exacerbated inflammatory arthritis and counteracted the protective effects of progranulin-mediated TNFR2 activation against inflammation and autoimmunity. TNFR2/14-3-3ε signaled through PI3K/Akt/mTOR to restrict NF-κB activation while simultaneously stimulating C/EBPß activation, thereby instructing macrophage plasticity. Collectively, this study identifies 14-3-3ε as a previously unrecognized vital component of the TNFR2 receptor complex and provides new insights into the TNFR2 signaling, particularly its role in macrophage polarization with therapeutic implications for various inflammatory and autoimmune diseases with activation of the TNFR2/14-3-3ε antiinflammatory pathway.


Asunto(s)
Proteínas 14-3-3/inmunología , Macrófagos/inmunología , Receptores Tipo II del Factor de Necrosis Tumoral/inmunología , Proteínas 14-3-3/química , Proteínas 14-3-3/deficiencia , Proteínas 14-3-3/metabolismo , Animales , Artritis Experimental/inmunología , Artritis Experimental/metabolismo , Artritis Experimental/patología , Autoinmunidad , Humanos , Inflamación/inmunología , Macrófagos/metabolismo , Ratones , Ratones Noqueados , Complejos Multiproteicos/química , Complejos Multiproteicos/inmunología , Complejos Multiproteicos/metabolismo , Progranulinas/inmunología , Progranulinas/metabolismo , Células RAW 264.7 , Receptores Tipo II del Factor de Necrosis Tumoral/química , Receptores Tipo II del Factor de Necrosis Tumoral/deficiencia , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Transducción de Señal/inmunología
5.
Ann N Y Acad Sci ; 1490(1): 77-89, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33543485

RESUMEN

Type 2 diabetes mellitus (T2DM) significantly increases bone fragility and fracture risk. Progranulin (PGRN) promotes bone fracture healing in both physiological and type 1 diabetic conditions. The present study aimed to investigate the role of PGRN in T2DM bone fracture healing. MKR mice (with an FVB/N genetic background) were used as the T2DM model. Drill-hole and Bonnarens and Einhorn models were used to investigate the role of PGRN in T2DM fracture healing in vivo. Primary bone marrow cells were isolated for molecular and signaling studies, and reverse transcription-polymerase chain reaction, immunohistochemical staining, and western blotting were performed to assess PGRN effects in vitro. PGRN mRNA and protein expression were upregulated in the T2DM model. Local administration of recombinant PGRN effectively promoted T2DM bone fracture healing in vivo. Additionally, PGRN could induce anabolic metabolism during endochondral ossification through the TNFR2-Akt and Erk1/2 pathways. Furthermore, PGRN showed anti-inflammatory activity in the T2DM bone regeneration process. These findings suggest that local administration of exogenous PGRN may be an alternative strategy to support bone regeneration in patients with T2DM. Additionally, PGRN might hold therapeutic potential for other TNFR-related metabolic disorders.


Asunto(s)
Regeneración Ósea/efectos de los fármacos , Diabetes Mellitus Tipo 2/patología , Curación de Fractura/efectos de los fármacos , Fracturas Óseas/tratamiento farmacológico , Osteogénesis/efectos de los fármacos , Progranulinas/uso terapéutico , Anabolizantes/uso terapéutico , Animales , Humanos , Ratones , Ratones Transgénicos , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
6.
Methods Mol Biol ; 2248: 109-119, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33185871

RESUMEN

TNFα/TNFR signaling plays a critical role in the pathogenesis of various inflammatory and autoimmune diseases, and anti-TNFα therapies have been accepted as the effective approaches for treating several autoimmune diseases. Progranulin (PGRN), a multi-faced growth factor-like molecule, directly binds to TNFR1 and TNFR2, particularly to the latter with higher affinity than TNFα. PGRN derivative Atsttrin is composed of three TNFR-binding domain of PGRN and exhibits even better therapeutic effects than PGRN in several inflammatory disease models, including collagen-induced arthritis. Herein we describe the detailed methodology of using (1) ELISA-based solid phase protein-protein interaction assay to demonstrate the direct binding of Atsttrin to TNFR2 and its inhibition of TNFα/TNFR2 interaction; and (2) tartrate-resistant acid phosphatase (TRAP) staining of in vitro osteoclastogenesis to reveal the cell-based anti-TNFα activity of Atsttrin. Using the protocol described here, the investigators should be able to reproducibly detect the physical inhibition of TNFα binding to TNFR and the functional inhibition of TNFα activity by Atsttrin and various kinds of TNF inhibitors.


Asunto(s)
Progranulinas/química , Receptores Tipo II del Factor de Necrosis Tumoral/química , Factor de Necrosis Tumoral alfa/química , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática , Osteogénesis/efectos de los fármacos , Osteogénesis/genética , Progranulinas/metabolismo , Unión Proteica , Mapeo de Interacción de Proteínas/métodos , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Relación Estructura-Actividad , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores
7.
Methods Mol Biol ; 2248: 201-210, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33185877

RESUMEN

The NF-κß transcription factor is a molecular mediator crucial to many biological functions and a central regulator of inflammatory and immune responses. NF-κß is activated by multiple immunologically relevant stimuli, including members of the tumor necrosis factor (TNF) superfamily, and targeting TNF/NFκß activity is a therapeutic objective in many inflammatory and autoimmune conditions. Here, we describe the generation of a transgenic reporter mouse model, expressing the human tumor necrosis factor α (TNF-α) transgene (TNF-tg) and carrying the luciferase gene under control of the NFκB-responsive element (NF-κB-Luc). Bioluminescence imaging shows that overexpression of TNF-α effectively activates NF-κB luciferase in vivo. To evaluate this system as a screen for potential therapeutics targeting the TNF/NFκß signaling pathway, we treated double mutant mice with PGRN-derived Atsttrin, an engineered molecule comprising the minimal progranulin (PGRN):TNFR binding fragments previously demonstrated as therapeutic in multiple models of TNF/NFκß-driven disease. Administration of Atsttrin could effectively inhibit luciferase activity in TNF-tg:NF-κB-Luc double mutant mice and demonstrates that this transgenic model can be used to non-invasively monitor the in vivo efficacy of modulators of TNF-activated NF-κB signaling pathway.


Asunto(s)
Mediciones Luminiscentes , Imagen Molecular , FN-kappa B/metabolismo , Progranulinas/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Humanos , Mediciones Luminiscentes/métodos , Ratones , Imagen Molecular/métodos , FN-kappa B/antagonistas & inhibidores , Progranulinas/genética , Factor de Necrosis Tumoral alfa/farmacología
8.
Front Cell Dev Biol ; 8: 577572, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33195216

RESUMEN

BACKGROUND: Cartilage defects account for substantial economic and humanistic burdens and pose a significant clinical problem. The efficacy of clinical approaches to cartilage repair is often inadequate, in part, owing to the restricted proliferative capacity of chondrocytes. Molecules have the capacity to promote the differentiation of multipotent mesenchymal stem cells into chondrocytes and may also gain the ability to repair the damaged cartilage. OBJECTIVE: This study aimed to investigate the role of Atsttrin (progranulin-derived engineered protein) in cartilage repair as well as the signaling pathway involved. METHODS: Primary and mesenchymal stem cell lines were used for the micromass culture. A murine cartilage defect model was used to determine the role of Atsttrin in cartilage repair in vivo. Real-time polymerase chain reaction and Western blot analysis were used to monitor the effect of Atsttrin on the transcriptional and protein levels, respectively, of key anabolic and catabolic signaling molecules. RESULTS: Atsttrin stimulated chondrogenesis in vitro and accelerated cartilage repair in vivo. In addition, Atsttrin-mediated cartilage repair occurred primarily through tumor necrosis factor receptor 2-initiated Akt signaling and downstream JunB transcription factor. CONCLUSION: Atsttrin might serve as a promising therapeutic modality for cartilage regeneration.

9.
Ann N Y Acad Sci ; 1460(1): 43-56, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31423598

RESUMEN

Type 1 diabetes mellitus (T1DM) is an autoimmune disease characterized by insulin deficiency, and patients with diabetes have an increased risk of bone fracture and significantly impaired fracture healing. Proinflammatory cytokine tumor necrosis factor-alpha is significantly upregulated in diabetic fractures and is believed to underlie delayed fracture healing commonly observed in diabetes. Our previous genetic screen for the binding partners of progranulin (PGRN), a growth factor-like molecule that induces chondrogenesis, led to the identification of tumor necrosis factor receptors (TNFRs) as the PGRN-binding receptors. In this study, we employed several in vivo models to ascertain whether PGRN has therapeutic effects in diabetic fracture healing. Here, we report that deletion of PGRN significantly delayed bone fracture healing and aggravated inflammation in the fracture models of mice with T1DM. In contrast, recombinant PGRN effectively promoted diabetic fracture healing by inhibiting inflammation and enhancing chondrogenesis. In addition, both TNFR1 proinflammatory and TNFR2 anti-inflammatory signaling pathways are involved in PGRN-stimulated diabetic fracture healing. Collectively, these findings illuminate a novel understanding concerning the role of PGRN in diabetic fracture healing and may have an application in the development of novel therapeutic intervention strategies for diabetic and other types of impaired fracture healing.


Asunto(s)
Diabetes Mellitus Experimental/patología , Diabetes Mellitus Tipo 1/patología , Curación de Fractura/efectos de los fármacos , Progranulinas/farmacología , Animales , Condrogénesis/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Eliminación de Gen , Humanos , Inflamación/patología , Ratones , Progranulinas/deficiencia , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores del Factor de Necrosis Tumoral/metabolismo , Proteínas Recombinantes/farmacología , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
10.
J Neuroinflammation ; 16(1): 238, 2019 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-31775776

RESUMEN

PURPOSE: Spinal cord injury (SCI) often results in significant and catastrophic dysfunction and disability and imposes a huge economic burden on society. This study aimed to determine whether progranulin (PGRN) plays a role in the progressive damage following SCI and evaluate the potential for development of a PGRN derivative as a new therapeutic target in SCI. METHODS: PGRN-deficient (Gr-/-) and wild-type (WT) littermate mice were subjected to SCI using a weight-drop technique. Local PGRN expression following injury was evaluated by Western blotting and immunofluorescence. Basso Mouse Scale (BMS), inclined grid walking test, and inclined plane test were conducted at indicated time points to assess neurological recovery. Inflammation and apoptosis were examined by histology (Hematoxylin and Eosin (H&E) staining and Nissl staining, TUNEL assays, and immunofluorescence), Western blotting (from whole tissue protein for iNOS/p-p65/Bax/Bcl-2), and ex vivo ELISA (for TNFα/IL-1ß/IL-6/IL-10). To identify the prophylactic and therapeutic potential of targeting PGRN, a PGRN derived small protein, Atsttrin, was conjugated to PLGA-PEG-PLGA thermosensitive hydrogel and injected into intrathecal space prior to SCI. BMS was recorded for neurological recovery and Western blotting was applied to detect the inflammatory and apoptotic proteins. RESULTS: After SCI, PGRN was highly expressed in activated macrophage/microglia and peaked at day 7 post-injury. Grn-/- mice showed a delayed neurological recovery after SCI at day 21, 28, 35, and 42 post-injury relative to WT controls. Histology, TUNEL assay, immunofluorescence, Western blotting, and ELISA all indicated that Grn-/- mice manifested uncontrolled and expanded inflammation and apoptosis. Administration of control-released Atsttrin could improve the neurological recovery and the pro-inflammatory/pro-apoptotic effect of PGRN deficiency. CONCLUSION: PGRN deficiency exacerbates SCI by promoting neuroinflammation and cellular apoptosis, which can be alleviated by Atsttrin. Collectively, our data provide novel evidence of using PGRN derivatives as a promising therapeutic approach to improve the functional recovery for patients with spinal cord injury.


Asunto(s)
Apoptosis/fisiología , Inflamación/metabolismo , Progranulinas/metabolismo , Traumatismos de la Médula Espinal/metabolismo , Animales , Citocinas/metabolismo , Femenino , Inflamación/genética , Ratones , Ratones Noqueados , Óxido Nítrico Sintasa de Tipo II/metabolismo , Progranulinas/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Recuperación de la Función/fisiología , Traumatismos de la Médula Espinal/genética , Regulación hacia Arriba , Proteína X Asociada a bcl-2/metabolismo
11.
Ann Rheum Dis ; 78(11): 1524-1535, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31302596

RESUMEN

OBJECTIVE: Tumour necrosis factor alpha (TNF-α) signalling plays a central role in the pathogenesis of various autoimmune diseases, particularly inflammatory arthritis. This study aimed to repurpose clinically approved drugs as potential inhibitors of TNF-α signalling in treatment of inflammatory arthritis. METHODS: In vitro and in vivo screening of an Food and Drug Administration (FDA)-approved drug library; in vitro and in vivo assays for examining the blockade of TNF actions by fexofenadine: assays for defining the anti-inflammatory activity of fexofenadine using TNF-α transgenic (TNF-tg) mice and collagen-induced arthritis in DBA/1 mice. Identification and characterisation of the binding of fexofenadine to cytosolic phospholipase A2 (cPLA2) using drug affinity responsive target stability assay, proteomics, cellular thermal shift assay, information field dynamics and molecular dynamics; various assays for examining fexofenadine inhibition of cPLA2 as well as the dependence of fexofenadine's anti-TNF activity on cPLA2. RESULTS: Serial screenings of a library composed of FDA-approved drugs led to the identification of fexofenadine as an inhibitor of TNF-α signalling. Fexofenadine potently inhibited TNF/nuclear factor kappa-light-chain-enhancer of activated B cells (NF-ĸB) signalling in vitro and in vivo, and ameliorated disease symptoms in inflammatory arthritis models. cPLA2 was isolated as a novel target of fexofenadine. Fexofenadine blocked TNF-stimulated cPLA2 activity and arachidonic acid production through binding to catalytic domain 2 of cPLA2 and inhibition of its phosphorylation on Ser-505. Further, deletion of cPLA2 abolished fexofenadine's anti-TNF activity. CONCLUSION: Collectively, these findings not only provide new insights into the understanding of fexofenadine action and underlying mechanisms but also provide new therapeutic interventions for various TNF-α and cPLA2-associated pathologies and conditions, particularly inflammatory rheumatic diseases.


Asunto(s)
Artritis Experimental/tratamiento farmacológico , Fosfolipasas A2 Citosólicas/efectos de los fármacos , Terfenadina/análogos & derivados , Inhibidores del Factor de Necrosis Tumoral/farmacología , Animales , Ratones , Ratones Endogámicos DBA , Ratones Transgénicos , Transducción de Señal/efectos de los fármacos , Terfenadina/farmacología , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores
12.
Cytokine Growth Factor Rev ; 45: 53-64, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30733059

RESUMEN

Progranulin (PGRN), a widely expressed glycoprotein with pleiotropic function, has been linked to a host of physiological processes and diverse pathological states. A series of contemporary preclinical disease models and clinical trials have evaluated various therapeutic strategies targeting PGRN, highlighting PGRN as a promising therapeutic target. Herein we summarize available knowledge of PGRN targeting in various kinds of diseases, including common neurological diseases, inflammatory autoimmune diseases, cancer, tissue repair, and rare lysosomal storage diseases, with a focus on the functional domain-oriented drug development strategies. In particular, we emphasize the role of extracellular PGRN as a non-conventional, extracellular matrix bound, growth factor-like conductor orchestrating multiple membrane receptors and intracellular PGRN as a chaperone/co-chaperone that mediates the folding and traffic of its various binding partners.


Asunto(s)
Enfermedades Autoinmunes/tratamiento farmacológico , Enfermedades por Almacenamiento Lisosomal/tratamiento farmacológico , Neoplasias/tratamiento farmacológico , Enfermedades del Sistema Nervioso/tratamiento farmacológico , Progranulinas/fisiología , Animales , Ensayos Clínicos como Asunto , Modelos Animales de Enfermedad , Desarrollo de Medicamentos , Humanos , Inflamación/tratamiento farmacológico , Lisosomas/enzimología , Ratones , Chaperonas Moleculares/fisiología , Terapia Molecular Dirigida , Enfermedades Neurodegenerativas/tratamiento farmacológico
13.
JBJS Rev ; 7(1): e10, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30694878

RESUMEN

BACKGROUND: Teriparatide, a 1-34 fragment of parathyroid hormone (PTH) that maintains most of the biological activities of PTH, has been employed since 2002 as an anabolic agent for osteoporotic individuals who are at high risk of fracture. The purpose of the present review is to provide a systematic summary and timely update on treatment with teriparatide for fracture prevention. METHODS: Electronic databases, including OVID MEDLINE, OVID Embase, and the Cochrane Library, were searched on February 9, 2018, to identify published systematic reviews and meta-analyses addressing treatment with teriparatide for fracture prevention, and A Measurement Tool to Assess Systematic Reviews 2 (AMSTAR 2) was used to assess the quality of included studies. RESULTS: Seventeen studies were included. Of the 17 eligible studies, 3 were rated as high quality, 3 were rated as moderate quality, 6 were rated as low quality, and 5 were rated as critically low quality. Teriparatide reduced vertebral and overall nonvertebral fractures in osteoporotic patients regardless of the existence of precipitating conditions, including postmenopausal status, glucocorticoid treatment, and chronic kidney disease, as compared with placebo, but not the site-specific nonvertebral fractures of the wrist and hip. Teriparatide did not more effectively reduce fracture risks when compared with other medications, such as bisphosphonates, selective estrogen receptor modulators, RANKL (receptor activator of nuclear factor kappa-beta ligand) inhibitor, or strontium ranelate. CONCLUSIONS: Teriparatide was safe and was not associated with an increased rate of adverse events when compared with other drugs. Teriparatide was effective for the prevention of vertebral and overall nonvertebral fractures in osteoporotic patients but not for the prevention of site-specific nonvertebral fractures at the wrist and hip. LEVEL OF EVIDENCE: Therapeutic Level I. See Instructions for Authors for a complete description of levels of evidence.


Asunto(s)
Conservadores de la Densidad Ósea/uso terapéutico , Fracturas Óseas/prevención & control , Teriparatido/uso terapéutico , Humanos
14.
Ann N Y Acad Sci ; 1442(1): 5-16, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30008173

RESUMEN

Over the past two decades, considerable advances in our understanding of inflammatory and immune pathways have allowed for the growing use of targeted biologic therapy. Most notably, the introduction of tumor necrosis factor (TNF) inhibitors has dramatically changed the management of autoimmune inflammatory disorders, including ankylosing spondylitis (AS). Despite the efficacy of TNF inhibitors documented in multiple clinical trials, anti-TNF therapy in AS is far from foolproof; it is associated with serious adverse effects and limited response to therapy in some patients. Moreover, specific questions regarding the role of TNF as a mediator of AS remain unanswered. Therefore, additional efforts are needed in order to better understand the role of TNF in the pathogenesis of AS and to develop safer and more effective treatment strategies. The purpose of this review is to better the understanding of the physiologic and pathogenic roles of TNF signaling in the course of AS. Relevant TNF biology and novel approaches to TNF blockade in AS are discussed.


Asunto(s)
Receptores del Factor de Necrosis Tumoral/antagonistas & inhibidores , Espondilitis Anquilosante/tratamiento farmacológico , Animales , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Humanos , Receptores del Factor de Necrosis Tumoral/metabolismo , Transducción de Señal , Espondilitis Anquilosante/metabolismo , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/metabolismo
15.
Sci Rep ; 8(1): 6495, 2018 04 24.
Artículo en Inglés | MEDLINE | ID: mdl-29691417

RESUMEN

p204, a murine member of the interferon-inducible p200 protein family, and its human analogue, IFI16, have been shown to function as tumor suppressors in vitro, but the molecular events involved, in particular in vivo, remain unclear. Herein we induced the Lewis Lung carcinoma (LLC) murine model of human lung cancer in p204 null mice (KO) and their control littermates (WT). We compared the transcriptome in spleen from WT and p204 KO mice using a high-throughput RNA-sequencing array. A total 30.02 Gb of clean data were obtained, and overall Q30% was greater than 90.54%. More than 75% of clean data from 12 transcriptome samples were mapped to exons. The results showed that only 11 genes exhibited altered expression in untreated p204 KO mice relative to untreated WT mice, while 393 altered genes were identified in tumor-bearing p204 KO mice when compared with tumor-bearing WT mice. Further differentially expressed gene cluster and gene ontology consortium classification revealed that numerous cytokines and their receptors, chemoattractant molecules, and adhesion molecules were significantly induced in p204 KO mice. This study provides novel insights to the p204 network in anti-tumor immune response and also presents a foundation for future work concerning p204-mediated gene expressions and pathways.


Asunto(s)
Carcinoma Pulmonar de Lewis/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Animales , Carcinoma Pulmonar de Lewis/metabolismo , Diferenciación Celular/fisiología , Expresión Génica/genética , Perfilación de la Expresión Génica/métodos , Interferones/metabolismo , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neoplasias/genética , Neoplasias/metabolismo , Análisis de Secuencia de ARN/métodos , Transcriptoma/genética
16.
EBioMedicine ; 29: 78-91, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29472103

RESUMEN

p204, a murine member of an interferon-inducible p200 family, was reported to recognize intracellular viral and bacterial DNAs, however, its role in the innate immunity in vivo remains unknown due to the lack of p204-deficient animal models. In this study we first generated the p204-/- mice. Unexpectedly, p204 deficiency led to significant defect in extracellular LPS signaling in macrophages, as demonstrated by dramatic reductions of LPS-mediated IFN-ß and pro-inflammatory cytokines. The serum levels of IFN-ß and pro-inflammatory cytokines were also significantly reduced in p204-/- mice following LPS challenge. In addition, p204-/- mice were resistant to LPS-induced shock. LPS-activated NF-ĸB and IRF-3 pathways were all defective in p204-deficient macrophages. p204 binds to TLR4 through its Pyrin domain, and it is required for the dimerization of TLR4 following LPS-challenge. Collectively, p204 is a critical component of canonical LPS-TLR4 signaling pathway, and these studies also suggest that p204 could be a potential target to prevent and treat inflammatory and infectious diseases.


Asunto(s)
Lipopolisacáridos/inmunología , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Transducción de Señal , Receptor Toll-Like 4/metabolismo , Animales , Células Cultivadas , Citocinas/metabolismo , Genotipo , Inmunidad Innata , Inflamasomas/inmunología , Inflamasomas/metabolismo , Mediadores de Inflamación/metabolismo , Factor 3 Regulador del Interferón/metabolismo , Interferón beta/metabolismo , Activación de Macrófagos/inmunología , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/microbiología , Macrófagos/virología , Ratones , Ratones Noqueados , Modelos Biológicos , FN-kappa B/metabolismo , Proteínas Nucleares/genética , Fosfoproteínas/genética , Unión Proteica , Multimerización de Proteína , Células RAW 264.7 , Choque Séptico/etiología , Choque Séptico/metabolismo , Choque Séptico/mortalidad , Receptor Toll-Like 4/química
17.
Pharmacol Ther ; 181: 34-48, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28723413

RESUMEN

The protein disulfide isomerase (PDI) gene family is a protein family classically characterized by endoplasmic reticulum (ER) localization and isomerase and redox activity. ERp57, a prominent multifunctional member of the PDI family, is detected at various levels in multiple cellular localizations outside of the ER. ERp57 has been functionally linked to a host of physiological processes and numerous studies have demonstrated altered expression and aberrant functionality of ERp57 in association with diverse pathological states. Here, we summarize available knowledge of ERp57's functions in subcellular compartments and the roles of dysregulated ERp57 in various diseases toward an emphasis on the potential utility of therapeutic development of ERp57.


Asunto(s)
Neoplasias/fisiopatología , Enfermedades Neurodegenerativas/fisiopatología , Proteína Disulfuro Isomerasas/metabolismo , Animales , Humanos
18.
Cytokine ; 101: 48-55, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-27527809

RESUMEN

Autoimmune disease encompasses an array of conditions with a variety of presentations and the involvement of multiple organs. Though the etiologies of many autoimmune conditions are unclear, uncontrolled inflammatory immune response is believed to be a major cause of disease development and progression. Progranulin (PGRN), an anti-inflammatory molecule with therapeutic effect in inflammatory arthritis, was identified as an endogenous antagonist of TNFα by competitively binding to TNFR. PGRN exerts its anti-inflammatory activity through multiple pathways, including induction of Treg differentiation and IL-10 expression and inhibition of chemokine release from macrophages. In addition, the protective role of PGRN has also been demonstrated in osteoarthritis, inflammatory bowel disease, and psoriasis. Intriguingly, PGRN was reported to contribute to development of insulin resistance in high-fat diet induced diabetes. Emerging evidences indicate that PGRN may also be associated with various autoimmune diseases, including systemic lupus erythematous, systemic sclerosis, multiple sclerosis and Sjogren's syndrome. This review summarizes recent studies of PGRN as a novel target molecule in the field of autoimmune disease, and provides updated information to inspire future studies.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/fisiopatología , Péptidos y Proteínas de Señalización Intercelular/inmunología , Animales , Artritis/inmunología , Artritis/fisiopatología , Artritis/terapia , Enfermedades Autoinmunes/complicaciones , Enfermedades Autoinmunes/terapia , Humanos , Inflamación/inmunología , Inflamación/terapia , Enfermedades Inflamatorias del Intestino/inmunología , Enfermedades Inflamatorias del Intestino/fisiopatología , Enfermedades Inflamatorias del Intestino/terapia , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Péptidos y Proteínas de Señalización Intercelular/uso terapéutico , Interleucina-10/genética , Interleucina-10/inmunología , Macrófagos/inmunología , Ratones , Osteoartritis/inmunología , Osteoartritis/fisiopatología , Osteoartritis/terapia , Progranulinas , Unión Proteica , Psoriasis/inmunología , Psoriasis/fisiopatología , Psoriasis/terapia , Receptores del Factor de Necrosis Tumoral/inmunología , Receptores del Factor de Necrosis Tumoral/metabolismo , Transducción de Señal/inmunología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/fisiología , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/inmunología
19.
Arthritis Res Ther ; 19(1): 280, 2017 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-29258611

RESUMEN

BACKGROUND: Atsttrin, an engineered protein composed of three tumor necrosis factor receptor (TNFR)-binding fragments of progranulin (PGRN), shows therapeutic effect in multiple murine models of inflammatory arthritis . Additionally, intra-articular delivery of PGRN protects against osteoarthritis (OA) progression. The purpose of this study is to determine whether Atsttrin also has therapeutic effects in OA and the molecular mechanisms involved. METHODS: Surgically induced and noninvasive rupture OA models were established in mouse and rat, respectively. Cartilage degradation and OA were evaluated using Safranin O staining, immunohistochemistry, and ELISA. Additionally, expressions of pain-related markers, degenerative factors, and anabolic and catabolic markers known to be involved in OA were analyzed. Furthermore, the anabolic and anti-catabolic effects and underlying mechanisms of Atsttrin were determined using in-vitro assays with primary chondrocytes. RESULTS: Herein, we found Atsttrin effectively prevented the accelerated OA phenotype associated with PGRN deficiency. Additionally, Atsttrin exhibited a preventative effect in OA by protecting articular cartilage and reducing OA-associated pain in both nonsurgically induced rat and surgically induced murine OA models. Mechanistic studies revealed that Atsttrin stimulated TNFR2-Akt-Erk1/2-dependent chondrocyte anabolism, while inhibiting TNFα/TNFR1-mediated inflammatory catabolism. CONCLUSIONS: These findings not only provide new insights into the role of PGRN and its derived engineered protein Atsttrin in cartilage homeostasis as well as OA in vivo, but may also lead to new therapeutic alternatives for OA as well as other relative degenerative joint diseases.


Asunto(s)
Osteoartritis/patología , Proteínas Recombinantes de Fusión/metabolismo , Animales , Cartílago Articular/metabolismo , Células Cultivadas , Condrocitos/metabolismo , Modelos Animales de Enfermedad , Granulinas , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Osteoartritis/metabolismo , Progranulinas , Ratas , Ratas Sprague-Dawley
20.
Ann N Y Acad Sci ; 1383(1): 5-20, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27505256

RESUMEN

Progranulin (PGRN) is a growth factor with a unique beads-on-a-string structure that is involved in multiple pathophysiological processes, including anti-inflammation, tissue repair, wound healing, neurodegenerative diseases, and tumorigenesis. This review presents up-to-date information concerning recent studies on the role of PGRN in inflammatory arthritis and osteoarthritis, with a special focus on the involvement of the interactions and interplay between PGRN and tumor necrosis factor receptor (TNFR) family members in regulating such musculoskeletal diseases. In addition, this paper highlights the applications of atsttrin, an engineered protein comprising three TNFR-binding fragments of PGRN, as a promising intervention in treating arthritis.


Asunto(s)
Artritis/tratamiento farmacológico , Artritis/metabolismo , Péptidos y Proteínas de Señalización Intercelular/fisiología , Receptores del Factor de Necrosis Tumoral/metabolismo , Animales , Humanos , Péptidos y Proteínas de Señalización Intercelular/uso terapéutico , Progranulinas , Unión Proteica/fisiología , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes de Fusión/uso terapéutico , Transducción de Señal/fisiología , Factor de Necrosis Tumoral alfa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA