Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Oxid Med Cell Longev ; 2022: 2785113, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35116091

RESUMEN

OBJECTIVE: Myocardial ischemia/reperfusion (I/R) injury can aggravate myocardial injury. Programmed necrosis plays a crucial role in this injury. However, the role of exosomal miRNAs in myocardial I/R injury remains unclear. Therefore, this study is aimed at exploring the function and mechanism of exosomal miR-17-3p in myocardial I/R injury. METHODS: The myocardial I/R injury animal model was established in C57BL/6 mice. Exosomes were identified using transmission electron microscopy (TEM), nanoparticle tracking analysis (NTA), and Western blotting. Programmed necrosis was detected by PI staining. Heart function and myocardial infarct size were evaluated using echocardiography and triphenyl tetrazolium chloride (TTC) staining, respectively. Histopathological changes were visualized by hematoxylin and eosin (H&E) and Masson staining. The regulation of TIMP3 expression by miR-17-3p was verified using a dual-luciferase reporter assay. Lactate dehydrogenase (LDH) and tumor necrosis factor-α (TNF-α) levels were measured by enzyme-linked immunosorbent assays (ELISA). TIMP3 expression was measured by quantitative reverse transcription-polymerase chain reaction (qRT-PCR) and Western blotting. RESULTS: We demonstrated that miR-17-3p was significantly downregulated in peripheral blood exosomes after cardiac I/R injury. Further analysis indicated that exosomal miR-17-3p attenuated H2O2-induced programmed necrosis in cardiomyocytes in vitro. Moreover, TIMP3 was a target for miR-17-3p. TIMP3 affected H2O2-induced programmed necrosis in cardiomyocytes. This effect was modulated by miR-17-3p in vitro. Furthermore, exosomal miR-17-3p greatly alleviated cardiac I/R injury in vivo. CONCLUSIONS: The present study demonstrated that exosomal miR-17-3p alleviated the programmed necrosis associated with cardiac I/R injury by regulating TIMP3 expression. These findings could represent a potential treatment for I/R injury.


Asunto(s)
Exosomas/metabolismo , MicroARNs/metabolismo , Inhibidor Tisular de Metaloproteinasa-3/metabolismo , Regiones no Traducidas 3' , Animales , Antagomirs/metabolismo , Apoptosis/efectos de los fármacos , Sitios de Unión , Células Cultivadas , Modelos Animales de Enfermedad , Regulación hacia Abajo , Peróxido de Hidrógeno/farmacología , Ratones , Ratones Endogámicos C57BL , MicroARNs/antagonistas & inhibidores , MicroARNs/genética , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Inhibidor Tisular de Metaloproteinasa-3/química , Inhibidor Tisular de Metaloproteinasa-3/genética , Factor de Necrosis Tumoral alfa/análisis
2.
J Pharmacol Exp Ther ; 375(2): 296-307, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32958629

RESUMEN

Infarct expansion can occur after myocardial infarction (MI), which leads to adverse left ventricular (LV) remodeling and failure. An imbalance between matrix metalloproteinase (MMP) induction and tissue inhibitors of MMPs (TIMPs) can accelerate this process. Past studies have shown different biologic effects of TIMP-3, which may depend upon specific domains within the TIMP-3 molecule. This study tested the hypothesis that differential effects of direct myocardial injections of either a full-length recombinant TIMP-3 (F-TIMP-3) or a truncated form encompassing the N-terminal region (N-TIMP-3) could be identified post-MI. MI was induced in pigs that were randomized for MI injections (30 mg) and received targeted injections within the MI region of F-TIMP-3 (n = 8), N-TIMP-3 (n = 9), or saline injection (MI-only, n = 11). At 14 days post-MI, LV ejection fraction fell post-MI but remained higher in both TIMP-3 groups. Tumor necrosis factor and interleukin-10 mRNA increased by over 10-fold in the MI-only and N-TIMP-3 groups but were reduced with F-TIMP-3 at this post-MI time point. Direct MI injection of either a full-length or truncated form of TIMP-3 is sufficient to favorably alter the course of post-MI remodeling. The functional and differential relevance of TIMP-3 domains has been established in vivo since the TIMP-3 constructs demonstrated different MMP/cytokine expression profiles. These translational studies identify a unique and more specific therapeutic strategy to alter the course of LV remodeling and dysfunction after MI. SIGNIFICANCE STATEMENT: Using different formulations of tissue inhibitor of matrix metalloproteinase-3 (TIMP-3), when injected into the myocardial infarction (MI) region, slowed the progression of indices of left ventricular (LV) failure, suggesting that the N terminus of TIMP-3 is sufficient to attenuate early adverse functional events post-MI. Injections of full-length recombinant TIMP-3, but not of the N-terminal region of TIMP-3, reduced relative indices of inflammation at the mRNA level, suggesting that the C-terminal region affects other biological pathways. These unique proof-of-concept studies demonstrate the feasibility of using recombinant small molecules to selectively interrupt adverse LV remodeling post-MI.


Asunto(s)
Infarto del Miocardio/patología , Fragmentos de Péptidos/farmacología , Inhibidor Tisular de Metaloproteinasa-3/química , Remodelación Ventricular/efectos de los fármacos , Secuencia de Aminoácidos , Colágeno/genética , Citocinas/genética , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Inyecciones , Metaloproteinasas de la Matriz/genética , Fragmentos de Péptidos/química , Dominios Proteicos , ARN Mensajero/genética , Inhibidor Tisular de Metaloproteinasa-3/genética
3.
FEBS Lett ; 594(1): 94-103, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31359422

RESUMEN

Extracellular levels of soluble TIMP-3 are low, reflecting its binding by extracellular matrix (ECM) components including sulfated glycosaminoglycans (SGAGs) and endocytosis via low density lipoprotein receptor-related protein 1. Since TIMP-3 inhibits ECM degradation, the ability of SGAGs to elevate extracellular TIMP-3 is significant for osteoarthritis treatment. Previous studies of such interactions have utilized immobilized TIMP-3 or ligands. Here, we report the thermodynamics of the interactions of the sGAG-binding N-domain of TIMP-3 with chondroitin sulfate, pentosan polysulfate, and suramin in solution using isothermal titration calorimetry. All three interactions are driven by a favorable negative enthalpy change combined with an unfavorable decrease in entropy. The heat capacity changes (ΔCp ) for all of the interactions are zero, indicating an insignificant contribution from hydrophobic interactions.


Asunto(s)
Sulfatos de Condroitina/farmacología , Simulación del Acoplamiento Molecular , Poliéster Pentosan Sulfúrico/farmacología , Suramina/farmacología , Inhibidor Tisular de Metaloproteinasa-3/química , Sitios de Unión , Sulfatos de Condroitina/química , Humanos , Poliéster Pentosan Sulfúrico/química , Unión Proteica , Suramina/química , Inhibidor Tisular de Metaloproteinasa-3/metabolismo
4.
Hum Mutat ; 40(5): 539-551, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30668888

RESUMEN

Sorsby fundus dystrophy (SFD) is a macular degeneration caused by mutations in TIMP3, the majority of which introduce a novel cysteine. However, the exact molecular mechanisms underlying SFD remain unknown. We aimed to provide novel insights into the functional consequences of a distinct N-terminal mutation. Haplotype reconstruction in three SFD families revealed that the identified c.113C>G, p.(Ser38Cys) mutation is a founder in Belgian and northern French families with a late-onset SFD phenotype. Functional consequences of the p.(Ser38Cys) mutation were investigated by high-resolution Western blot analysis of wild type and mutant TIMP3 using patient fibroblasts and in vitro generated proteins, and by molecular modeling of TIMP3 and its interaction partners. We could not confirm a previous hypothesis on dimerization of mutant TIMP3 proteins. However, we identified aberrant intramolecular disulfide bonding. Our data provide evidence for disruption of the established Cys36-Cys143 disulfide bond and formation of a novel Cys36-Cys38 bond, possibly associated with increased glycosylation of the protein. In conclusion, we propose a novel pathogenetic mechanism underlying the p.(Ser38Cys) TIMP3 founder mutation involving intramolecular disulfide bonding. These results provide new insights into the pathogenesis of SFD and other retinopathies linked to mutations in TIMP3, such as age-related macular degeneration.


Asunto(s)
Efecto Fundador , Degeneración Macular/diagnóstico , Degeneración Macular/genética , Mutación , Dominios y Motivos de Interacción de Proteínas , Inhibidor Tisular de Metaloproteinasa-3/química , Inhibidor Tisular de Metaloproteinasa-3/genética , Anciano , Disulfuros , Femenino , Fibroblastos/metabolismo , Regulación de la Expresión Génica , Haplotipos , Humanos , Masculino , Metaloproteinasa 2 de la Matriz/química , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 2 de la Matriz/metabolismo , Peso Molecular , Linaje , Conformación Proteica , Relación Estructura-Actividad , Inhibidor Tisular de Metaloproteinasa-3/metabolismo
5.
Pharmacol Res Perspect ; 6(6): e00442, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30459952

RESUMEN

Tissue Inhibitor of Metalloproteinase 3 (TIMP3) is a secreted protein that has a great utility to inhibit elevated metalloproteinase (MMP) activity in injured tissues including infarcted cardiac tissue, inflamed vessels, and joint cartilages. An imbalance between TIMP3 and active MMP levels in the local tissue area may cause worsening of disease progression. To counter balance elevated MMP levels, exogenous administration of TIMP3 appeared to be beneficial in preclinical studies. However, the current form of WT-TIMP3 molecule has a limitation to be a therapeutic candidate due to low production yield, short plasma half-life, injection site retention, and difficulty in delivery, etc. We have engineered TIMP3 molecules by adding extra glycosylation sites or fusing with albumin, Fc, and antibody to improve pharmacokinetic properties. In general, the C-terminal fusion of TIMP3 improved expression and production in mammalian cells and extended half-lives dramatically 5-20 folds. Of note, a site-specific glycosylation at K22S/F34N resulted in a higher level of expression and better cardiac function compared to other fusion proteins in the context of left ventricle ejection fraction (LVEF) changes in a rat myocardial infarction model. It appeared that cardiac efficacy depends on a high ECM binding affinity, in which K22S/F34N and N-TIMP3 showed a higher binding to the ECM compared to other engineered molecules. In conclusion, we found that the ECM binding and sustained residence of injected TIMP3 molecules are important for cardiac tissue localization and inhibition of adverse remodeling activity.


Asunto(s)
Proteína ADAM17/antagonistas & inhibidores , Metaloproteinasas de la Matriz/metabolismo , Infarto del Miocardio/tratamiento farmacológico , Proteínas Recombinantes de Fusión/farmacología , Inhibidor Tisular de Metaloproteinasa-3/farmacología , Función Ventricular Izquierda/efectos de los fármacos , Proteína ADAM17/metabolismo , Animales , Línea Celular , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Fibroblastos , Glicosilación , Humanos , Infusiones Intravenosas , Inyecciones Intralesiones , Masculino , Mutación , Infarto del Miocardio/etiología , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/uso terapéutico , Inhibidor Tisular de Metaloproteinasa-3/química , Inhibidor Tisular de Metaloproteinasa-3/genética , Inhibidor Tisular de Metaloproteinasa-3/uso terapéutico , Resultado del Tratamiento
6.
Am J Physiol Heart Circ Physiol ; 315(4): H814-H825, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-29979624

RESUMEN

Although improvements in timing and approach for early reperfusion with acute coronary syndromes have occurred, myocardial injury culminating in a myocardial infarction (MI) remains a common event. Although a multifactorial process, an imbalance between the induction of proteolytic pathways, such as matrix metalloproteinases (MMPs) and endogenous tissue inhibitors of metalloproteinase (TIMPs), has been shown to contribute to this process. In the present study, a full-length TIMP-3 recombinant protein (rTIMP-3) was encapsulated in a specifically formulated hyaluronic acid (HA)-based hydrogel that contained MMP-cleavable peptide cross-links, which influenced the rate of rTIMP-3 release from the HA gel. The effects of localized delivery of this MMP-sensitive HA gel (HAMMPS) alone and containing rTIMP-3 (HAMMPS/rTIMP-3) were examined in terms of the natural history of post-MI remodeling. Pigs were randomized to one of the following three different groups: MI and saline injection (MI/saline group, 100-µl injection at nine injection sites, n = 7), MI and HAMMPS injection (MI/HAMMPS group; 100-µl injection at nine injection sites, n = 7), and MI and HAMMPS/rTIMP-3 injection (MI/HAMMPS/rTIMP-3 group; 20-µg/100-µl injection at nine injection sites, n = 7). Left ventricular (LV) echocardiography was serially performed up to 28 days post-MI. LV dilation, as measured by end-diastolic volume, and the degree of MI wall thinning were reduced by ~50% in the HAMMPS/rTIMP-3 group ( P < 0.05). Furthermore, indexes of heart failure progression post-MI, such as LV filling pressures and left atrial size, were also attenuated to the greatest degree in the HAMMPS/rTIMP-3 group. At 28 days post-MI, HAMMPS/rTIMP-3 caused a relative reduction in the transcriptional profile for myofibroblasts as well as profibrotic pathways, which was confirmed by subsequent histochemistry. In conclusion, these findings suggest that localized delivery of a MMP-sensitive biomaterial that releases a recombinant TIMP holds promise as a means to interrupt adverse post-MI remodeling. NEW & NOTEWORTHY The present study targeted a myocardial matrix proteolytic system, matrix metalloproteinases (MMPs), through the use of a recombinant tissue inhibitor of MMPs incorporated into a MMP-sensitive hydrogel, which was regionally injected using a large animal model of myocardial infarction. Left ventricular geometry and function and indexes of myocardial remodeling were improved with this approach and support the advancement of localized therapeutic strategies that specifically target the myocardial matrix.


Asunto(s)
Fármacos Cardiovasculares/administración & dosificación , Sulfato de Dextran/química , Portadores de Fármacos , Ventrículos Cardíacos/efectos de los fármacos , Ácido Hialurónico/química , Infarto del Miocardio/tratamiento farmacológico , Inhibidor Tisular de Metaloproteinasa-3/administración & dosificación , Función Ventricular Izquierda/efectos de los fármacos , Remodelación Ventricular/efectos de los fármacos , Animales , Fármacos Cardiovasculares/química , Preparaciones de Acción Retardada , Sulfato de Dextran/análogos & derivados , Modelos Animales de Enfermedad , Composición de Medicamentos , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Matriz Extracelular/patología , Perfilación de la Expresión Génica/métodos , Ventrículos Cardíacos/metabolismo , Ventrículos Cardíacos/patología , Ventrículos Cardíacos/fisiopatología , Ácido Hialurónico/análogos & derivados , Hidrogeles , Masculino , Infarto del Miocardio/genética , Infarto del Miocardio/metabolismo , Infarto del Miocardio/fisiopatología , Miofibroblastos/efectos de los fármacos , Miofibroblastos/metabolismo , Miofibroblastos/patología , Proteínas Recombinantes/administración & dosificación , Inhibidor Tisular de Metaloproteinasa-3/química , Transcripción Genética/efectos de los fármacos , Transcriptoma
7.
FEBS Lett ; 592(14): 2499-2511, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29933498

RESUMEN

Due in part to the needs of the biopharmaceutical industry, there has been an increased drive to generate high quality recombinant proteins in large amounts. However, achieving high yields can be a challenge as the novelty and increased complexity of new targets often makes them 'difficult-to-express'. This study aimed to define the molecular features that restrict the production of a model 'difficult-to-express' recombinant protein, Tissue Inhibitor Metalloproteinase-3 (TIMP-3). Building from experimental data, computational approaches were used to rationalize the redesign of this recombinant target to generate a chimera with enhanced secretion. The results highlight the importance of early identification of unfavourable sequence attributes, enabling the generation of engineered protein forms that bypass 'secretory' bottlenecks and result in efficient recombinant protein production.


Asunto(s)
Clonación Molecular/métodos , Ingeniería de Proteínas/métodos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Secuencia de Aminoácidos , Animales , Células CHO , Biología Computacional , Cricetinae , Cricetulus , Expresión Génica , Humanos , Ratones , Modelos Biológicos , Transporte de Proteínas/genética , Proteínas Recombinantes/química , Vías Secretoras/genética , Biología Sintética/métodos , Inhibidor Tisular de Metaloproteinasa-2/química , Inhibidor Tisular de Metaloproteinasa-2/genética , Inhibidor Tisular de Metaloproteinasa-2/metabolismo , Inhibidor Tisular de Metaloproteinasa-3/química , Inhibidor Tisular de Metaloproteinasa-3/genética , Inhibidor Tisular de Metaloproteinasa-3/metabolismo
8.
Bioorg Chem ; 76: 13-22, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29102725

RESUMEN

The ADAMTS4 (a disintegrin and metalloproteinase with thrombospondin motifs 4) enzyme is a matrix-associated zinc metalloendopeptidase that plays an essential role in the degradation of cartilage aggrecan in arthritic diseases and has been recognized as one of the most primary targets for therapeutic intervention in osteoarthritis (OA). Here, we reported computational modeling of the atomic-level complex structure of ADAMTS4 with its cognate inhibitory protein TIMP3 based on high-resolution crystal template. By systematically examining the modeled complex structure we successfully identified a short inhibitory loop (62EASESLC68) in TIMP3 N-terminal inhibitory domain (NID) that directly participates in blocking the enzyme's active site, which, and its extended versions, were then broken from the full-length protein to serve as the peptide inhibitor candidates of ADAMTS4. Atomistic molecular dynamics simulation, binding energetic analysis, and fluorescence-based assay revealed that the TIMP3-derived linear peptides can only bind weakly to the enzyme (Kd = 74 ±â€¯8 µM), which would incur a considerable entropy penalty due to the high conformational flexibility and intrinsic disorder of these linear peptides. In this respect, we proposed a cyclization strategy to improve enzyme-peptide binding affinity by, instead of traditionally maximizing enthalpy contribution, minimizing entropy cost of the binding, where a disulfide bond was added across the two terminal residues of linear peptides, resulting in a number of TIMP3-derived cyclic peptides. Our studies confirmed that the cyclization, as might be expected, can promote peptide binding capability against ADAMTS4 substantially, with affinity increase by 3-fold, 9-fold and 7-fold for cyclic peptides , and , respectively.


Asunto(s)
Proteína ADAMTS4/metabolismo , Fragmentos de Péptidos/metabolismo , Péptidos Cíclicos/metabolismo , Inhibidor Tisular de Metaloproteinasa-3/metabolismo , Proteína ADAMTS4/química , Sitios de Unión , Entropía , Humanos , Simulación de Dinámica Molecular , Fragmentos de Péptidos/síntesis química , Fragmentos de Péptidos/química , Péptidos Cíclicos/síntesis química , Péptidos Cíclicos/química , Unión Proteica , Dominios Proteicos , Inhibidor Tisular de Metaloproteinasa-3/síntesis química , Inhibidor Tisular de Metaloproteinasa-3/química
9.
Eur Rev Med Pharmacol Sci ; 21(20): 4566-4576, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29131259

RESUMEN

OBJECTIVE: Renal cell carcinoma (RCC) displays an increasing incidence and mortality rate worldwide in recent years. More and more evidence identified microRNAs function as positive or negative regulatory factors in many cancers, but the role of miR-21 in RCC remains unclear. PATIENTS AND METHODS: Relative expression levels of miR-21 in human RCC tissue samples and RCC-derived cell lines were measured using quantitative real-time Polymerase Chain Reaction (PCR). Clinical features were collected to further study the relationship between the miR-21 level and clinicopathologic variables. Loss- and gain- of miR-21 experiments were employed to measure the influence of miR-21 in cell proliferation, apoptosis, invasion and migration. Downstream target gene was confirmed by using luciferase and Western blotting assays. RESULTS: MiR-21 significantly over-expressed in RCC tissues and cell lines than normal groups. Higher miR-21 expression level indicated larger tumor sizes, more lymph metastasis and advanced tumor node metastasis (TNM) stage. Knocking down miR-21 inhibited the cell growth, invasion and migration abilities but promoted the cell apoptosis, while over-expressing miR-21 promoted cell growth and metastasis. Furthermore, TIMP3 was confirmed as a direct target of moR-21 and inhibition of TIMP3 reserved the effect of down-regulating miR-21 in RCC cells. CONCLUSIONS: Our study demonstrated miR-21 was significantly over-expressed and functioned as a tumor oncogene via TIMP3 in RCC, which could provide a potential target for RCC diagnosis and therapy.


Asunto(s)
Movimiento Celular/genética , Neoplasias Renales/patología , MicroARNs/metabolismo , Inhibidor Tisular de Metaloproteinasa-3/metabolismo , Regiones no Traducidas 3' , Anciano , Antagomirs/metabolismo , Apoptosis , Secuencia de Bases , Línea Celular Tumoral , Proliferación Celular , Regulación hacia Abajo , Femenino , Humanos , Riñón/metabolismo , Riñón/patología , Neoplasias Renales/genética , Neoplasias Renales/mortalidad , Metástasis Linfática , Masculino , MicroARNs/antagonistas & inhibidores , MicroARNs/genética , Persona de Mediana Edad , Alineación de Secuencia , Tasa de Supervivencia , Inhibidor Tisular de Metaloproteinasa-3/química , Inhibidor Tisular de Metaloproteinasa-3/genética
10.
Comb Chem High Throughput Screen ; 20(6): 539-546, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28215146

RESUMEN

BACKGROUND: Mammary tumors are the second most common tumors (after skin tumors) in female dogs (Canis lupus familiaris). Tissue Inhibitor of Metlloproteinases-3 (TIMP-3) is a matrix associated endogenous inhibitor of Matrix Metalloproteinases (MMPs). Cancer metastasis occurs as a result of imbalance between MMPs and TIMPs. TIMP-3 is involved significantly in regulation of MMPs as well as progression of canine mammary tumor. OBJECTIVE: The present study was conducted to identify the structural and functional relationship between TIMP-3 and MMP which can aid in identifying the role of these proteins in canine mammary tumor. METHODS: Molecular characterization of TIMP-3 protein was done by molecular biology techniques such as gene cloning and sequencing. The homology based model of TIMP-3 protein was created and verified with a variety of available computational techniques as well as molecular dynamics simulation. RESULTS: The results indicated that predicted TIMP-3 protein structure of Canis lupus familiaris was reliable and more stable. The docking of TIMP-3 protein with MMP-2 and MMP-9 represents conformational structure of these two proteins which interact with each other but if misled canresult in the progression of tumor in canine. CONCLUSIONS: The three dimensional structure of TIMP-3 was generated and its interactions with MMP-2 and MMP-9, demonstrates the role of key binding residues. Until now, no structural details were available for canine TIMP-3 proteins, hence this study will broaden the horizon towards understanding the structural and functional aspects of this proteins in canine.


Asunto(s)
Simulación por Computador , Neoplasias Mamarias Animales/enzimología , Inhibidor Tisular de Metaloproteinasa-3/genética , Inhibidor Tisular de Metaloproteinasa-3/metabolismo , Animales , Clonación Molecular , Perros , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento , Metaloproteinasas de la Matriz/metabolismo , Modelos Moleculares , Unión Proteica , Análisis de Secuencia de ADN , Inhibidor Tisular de Metaloproteinasa-3/química
11.
Biomacromolecules ; 17(10): 3252-3261, 2016 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-27610455

RESUMEN

Sulfated glycosaminoglycans (sGAGs) modulate cellular processes via their interaction with extracellular matrix (ECM) proteins. We revealed a direct binding of tissue inhibitor of metalloproteinase-3 (TIMP-3) to the endocytic receptor low-density lipoprotein receptor-related protein (LRP-1) clusters II and IV using surface plasmon resonance. Sulfated hyaluronan (sHA) and chondroitin sulfate (sCS) derivatives interfered with TIMP-3/LRP-1 complex formation in a sulfation-dependent manner stronger than heparin. Electrostatic potential calculations suggested a competition between negatively charged GAGs and highly negatively charged complement-like domains of LRP-1 for the binding to a positively charged area of TIMP-3 as an underlying mechanism. In vitro studies revealed increased amounts of pericellular TIMP-3 in the presence of sHA as a consequence of the blocked protein uptake. GAG derivatives as part of biomaterials might post-translationally modulate TIMP-3 levels stronger than native GAGs, thus exhibiting catabolic effects on the ECM, which could prevent extensive pathological matrix degradation and promote wound healing.


Asunto(s)
Glicosaminoglicanos/administración & dosificación , Ácido Hialurónico/administración & dosificación , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/biosíntesis , Inhibidor Tisular de Metaloproteinasa-3/biosíntesis , Sulfatos de Condroitina/administración & dosificación , Sulfatos de Condroitina/química , Endocitosis/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Glicosaminoglicanos/química , Humanos , Ácido Hialurónico/química , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/química , Células Madre Mesenquimatosas/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Resonancia por Plasmón de Superficie , Inhibidor Tisular de Metaloproteinasa-3/química , Cicatrización de Heridas/efectos de los fármacos
12.
Sci Rep ; 6: 32544, 2016 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-27601084

RESUMEN

To revisit the autosomal dominant Sorsby fundus dystrophy (SFD) as a syndromic condition including late-onset pulmonary disease. We report clinical and imaging data of ten affected individuals from 2 unrelated families with SFD and carrying heterozygous TIMP3 mutations (c.572A > G, p.Y191C, exon 5, in family 1 and c.113C > G, p.S38C, exon 1, in family 2). In family 1, all SFD patients older than 50 (two generations) had also a severe emphysema, despite no history of smoking or asthma. In the preceding generation, the mother died of pulmonary emphysema and she was blind after the age of 50. Her two great-grandsons (<20 years), had abnormal Bruch Membrane thickness, a sign of eye disease. In family 2, eye and lung diseases were also associated in two generations, both occurred later, and lung disease was moderate (bronchiectasis). This is the first report of a syndromic SFD in line with the mouse model uncovering the role of TIMP3 in human lung morphogenesis and functions. The TIMP3 gene should be screened in familial pulmonary diseases with bronchiectasis, associated with a medical history of visual loss. In addition, SFD patients should be advised to avoid tobacco consumption, to practice sports, and to undergo regular pulmonary examinations.


Asunto(s)
Predisposición Genética a la Enfermedad , Degeneración Macular/genética , Inhibidor Tisular de Metaloproteinasa-3/genética , Anciano , Secuencia de Bases , Familia , Femenino , Fondo de Ojo , Humanos , Pulmón/patología , Degeneración Macular/diagnóstico por imagen , Masculino , Persona de Mediana Edad , Mutación , Linaje , Estructura Secundaria de Proteína , Inhibidor Tisular de Metaloproteinasa-3/química , Tomografía Computarizada por Rayos X
13.
Matrix Biol ; 56: 57-73, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27084377

RESUMEN

Matrix metalloproteinase 13 (MMP-13) degrades collagenous extracellular matrix and its aberrant activity associates with diseases such as arthritis, cancer, atherosclerosis and fibrosis. The wide range of MMP-13 proteolytic capacity suggests that it is a powerful, potentially destructive proteinase and thus it has been believed that MMP-13 is not produced in most adult human tissues in the steady state. Present study has revealed that human chondrocytes isolated from healthy adults constitutively express and secrete MMP-13, but that it is rapidly endocytosed and degraded by chondrocytes. Both pro- and activated MMP-13 bind to clusters II and III of low-density lipoprotein (LDL) receptor-related protein 1 (LRP1). Domain deletion studies indicated that the hemopexin domain is responsible for this interaction. Binding competition between MMP-13 and ADAMTS-4, -5 or TIMP-3, which also bind to cluster II, further shown that the MMP-13 binding site within cluster II is different from those of ADAMTS-4, -5 or TIMP-3. MMP-13 is therefore co-endocytosed with ADAMTS-5 and TIMP-3 by human chondrocytes. These findings indicate that MMP-13 may play a role on physiological turnover of cartilage extracellular matrix and that LRP1 is a key modulator of extracellular levels of MMP-13 and its internalization is independent of the levels of ADAMTS-4, -5 and TIMP-3.


Asunto(s)
Proteína ADAMTS5/metabolismo , Condrocitos/enzimología , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Metaloproteinasa 13 de la Matriz/metabolismo , Inhibidor Tisular de Metaloproteinasa-3/metabolismo , Proteína ADAMTS5/química , Unión Competitiva , Endocitosis , Células HEK293 , Humanos , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/química , Metaloproteinasa 13 de la Matriz/química , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Transporte de Proteínas , Inhibidor Tisular de Metaloproteinasa-3/química
14.
Mol Cell Endocrinol ; 412: 104-15, 2015 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-26054746

RESUMEN

Tissue inhibitor of metalloproteinase 3 (TIMP3) played a key role in female reproduction. However, its expression and function in goat are still unclear. In the present study, the full-length cDNA of goat TIMP3 was cloned from adult goat ovary; meanwhile, we demonstrated that putative TIMP3 protein shared a highly conserved amino acid sequence with known mammalian homologs. Real-time PCR results showed that TIMP3 was widely expressed in the tissues of adult goat. In the ovary, increasing expression of TIMP3 mRNA was discovered during the growth process of follicle and corpus luteum. Immunohistochemistry results suggested that TIMP3 protein existed in oocytes of all types of follicles, corpus luteum and granulosa and theca cells of primary, secondary, and antral but not primordial follicles. In vitro, human chorionic gonadotropin (hCG) stimulated the expression of TIMP3 in goat granulosa cells. hCG-induced TIMP3 mRNA expression was reduced by the inhibitors of protein kinase A, protein kinase C, MAPK kinase, or p38 kinase. Functionally, over-expression of TIMP3 significantly increased apoptosis and decreased the viability of cultured granulosa cells. Knockdown of TIMP3 could decrease hCG-induced progesterone secretion and the mRNA abundance of key steroidogenic enzymes (StAR, p450scc and HSD3B) as well as ECM proteins (DCN and FN). These findings provided evidence that the hCG induced expression of TIMP3 may play an important role in regulating goat granulosa cell survival and steroidogenesis.


Asunto(s)
Expresión Génica , Cabras/metabolismo , Células de la Granulosa/metabolismo , Inhibidor Tisular de Metaloproteinasa-3/fisiología , Secuencia de Aminoácidos , Animales , Apoptosis , Vías Biosintéticas , Proliferación Celular , Supervivencia Celular , Células Cultivadas , Gonadotropina Coriónica/farmacología , Secuencia Conservada , Femenino , Células de la Granulosa/efectos de los fármacos , Datos de Secuencia Molecular , Ovario/citología , Ovario/metabolismo , Progesterona/biosíntesis , Transducción de Señal , Inhibidor Tisular de Metaloproteinasa-3/química , Activación Transcripcional
15.
Nat Mater ; 13(6): 653-61, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24681647

RESUMEN

Inhibitors of matrix metalloproteinases (MMPs) have been extensively explored to treat pathologies where excessive MMP activity contributes to adverse tissue remodelling. Although MMP inhibition remains a relevant therapeutic target, MMP inhibitors have not translated to clinical application owing to the dose-limiting side effects following systemic administration of the drugs. Here, we describe the synthesis of a polysaccharide-based hydrogel that can be locally injected into tissues and releases a recombinant tissue inhibitor of MMPs (rTIMP-3) in response to MMP activity. Specifically, rTIMP-3 is sequestered in the hydrogels through electrostatic interactions and is released as crosslinks are degraded by active MMPs. Targeted delivery of the hydrogel/rTIMP-3 construct to regions of MMP overexpression following a myocardial infarction significantly reduced MMP activity and attenuated adverse left ventricular remodelling in a porcine model of myocardial infarction. Our findings demonstrate that local, on-demand MMP inhibition is achievable through the use of an injectable and bioresponsive hydrogel.


Asunto(s)
Hidrogeles/farmacología , Inhibidores de la Metaloproteinasa de la Matriz/farmacología , Infarto del Miocardio/tratamiento farmacológico , Inhibidor Tisular de Metaloproteinasa-3/farmacología , Remodelación Ventricular/efectos de los fármacos , Animales , Modelos Animales de Enfermedad , Humanos , Hidrogeles/química , Inhibidores de la Metaloproteinasa de la Matriz/química , Metaloproteinasas de la Matriz/metabolismo , Infarto del Miocardio/enzimología , Infarto del Miocardio/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/farmacología , Porcinos , Inhibidor Tisular de Metaloproteinasa-3/química
16.
Angiogenesis ; 17(1): 207-19, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24129822

RESUMEN

The binding of vascular endothelial growth factor (VEGF) to VEGF receptor-2 (VEGFR-2) on the surface of vascular endothelial cells stimulates many steps in the angiogenic pathway. Inhibition of this interaction is proving of value in moderating the neovascularization accompanying age-related macular degeneration and in the treatment of cancer. Tissue inhibitor of metalloproteinases-3 (TIMP-3) has been shown to be a natural VEGFR-2 specific antagonist-an activity that is independent of its ability to inhibit metalloproteinases. In this investigation we localize this activity to the C-terminal domain of the TIMP-3 molecule and characterize a short peptide, corresponding to part of this domain, that not only inhibits all three VEGF-family receptors, but also fibroblast growth factor and platelet-derived growth factor receptors. This multiple-receptor inhibition may explain why the peptide was also seen to be a powerful inhibitor of tumour growth and also a partial inhibitor of arthritic joint inflammation in vivo.


Asunto(s)
Artritis/tratamiento farmacológico , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Péptidos/farmacología , Inhibidor Tisular de Metaloproteinasa-3/farmacología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Animales , Artritis/metabolismo , Artritis/patología , Femenino , Células Endoteliales de la Vena Umbilical Humana , Humanos , Degeneración Macular/tratamiento farmacológico , Degeneración Macular/metabolismo , Degeneración Macular/patología , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Endogámicos BALB C , Péptidos/química , Inhibidor Tisular de Metaloproteinasa-3/química , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
17.
PLoS One ; 8(3): e55667, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23469166

RESUMEN

Tissue inhibitors of metalloproteinases (TIMPs) while originally characterized as inhibitors of matrix metalloproteinases (MMPs) have recently been shown to have a wide range of functions that are independent of their MMP inhibitory properties. Tissue inhibitor of metalloproteinases-3 (TIMP-3) is a potent inhibitor of VEGF-mediated angiogenesis and neovascularization through its ability to block the binding of VEGF to its receptor VEGFR-2. To identify and characterize the anti-angiogenic domain of TIMP-3, structure function analyses and synthetic peptide studies were performed using VEGF-mediated receptor binding, signaling, migration and proliferation. In addition, the ability of TIMP-3 peptides to inhibit CNV in a mouse model was evaluated. We demonstrate that the anti-angiogenic property resides in the COOH-terminal domain of TIMP-3 protein which can block the binding of VEGF specifically to its receptor VEGFR-2, but not to VEGFR-1 similar to the full-length wild-type protein. Synthetic peptides corresponding to putative loop 6 and tail region of TIMP-3 have anti-angiogenic properties as determined by inhibition of VEGF binding to VEGFR-2, VEGF-induced phosphorylation of VEGFR-2 and downstream signaling pathways as well as endothelial cell proliferation and migration in response to VEGF. In addition, we show that intravitreal administration of TIMP-3 peptide could inhibit the size of laser-induced choroidal neovascularization lesions in mice. Thus, we have identified TIMP-3 peptides to be efficient inhibitors of angiogenesis and have a potential to be used therapeutically in diseases with increased neovascularization.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Lámina Basal de la Coroides/irrigación sanguínea , Lámina Basal de la Coroides/efectos de los fármacos , Neovascularización Coroidal/prevención & control , Péptidos/farmacología , Inhibidor Tisular de Metaloproteinasa-3/antagonistas & inhibidores , Inhibidor Tisular de Metaloproteinasa-3/química , Inhibidores de la Angiogénesis/síntesis química , Animales , Lámina Basal de la Coroides/lesiones , Células Cultivadas , Neovascularización Coroidal/etiología , Neovascularización Coroidal/genética , Neovascularización Coroidal/metabolismo , Endotelio Vascular , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Coagulación con Láser/efectos adversos , Ratones , Ratones Endogámicos C57BL , Péptidos/síntesis química , Fosforilación , Transducción de Señal/efectos de los fármacos , Técnicas de Síntesis en Fase Sólida , Inhibidor Tisular de Metaloproteinasa-3/genética , Inhibidor Tisular de Metaloproteinasa-3/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
18.
PLoS One ; 8(3): e59157, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23527119

RESUMEN

The poor survival of adenocarcinomas of the gastroesophageal junction (GEJ) makes them clinically important. Discovery of host genetic factors that affect outcome may guide more individualized treatment. This study tests whether constitutional genetic variants in matrix metalloproteinases (MMP) and tissue inhibitors of metalloproteinases (TIMP) genes are associated with outcome of GEJ adenocarcinoma. Single nucleotide polymorphisms (SNPs) at four TIMP (TIMP1-4) and three MMP genes (MMP2, MMP7 and MMP9) were genotyped in DNA samples from a prospective cohort of patients with primary adenocarcinoma of the GEJ admitted to the British Columbia Cancer Agency. Cox proportional hazards regression, with adjustment for patient, disease and treatment variables, was used to estimate the association of SNPs with survival. Genotypes for 85 samples and 48 SNPs were analyzed. Four SNPs across TIMP3, (rs130274, rs715572, rs1962223 and rs5754312) were associated with survival. Interaction analyses revealed that the survival associations with rs715572 and rs5754312 are specific and significant for 5FU+cisplatin treated patients. Sanger sequencing of the TIMP3 coding and promoter regions revealed an additional SNP, rs9862, also associated with survival. TIMP3 genetic variants are associated with survival and may be potentially useful in optimizing treatment strategies for individual patients.


Asunto(s)
Adenocarcinoma/genética , Adenocarcinoma/mortalidad , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/mortalidad , Unión Esofagogástrica/patología , Polimorfismo Genético , Neoplasias Gástricas/genética , Neoplasias Gástricas/mortalidad , Neoplasias Gástricas/patología , Inhibidor Tisular de Metaloproteinasa-3/genética , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/patología , Anciano , Anciano de 80 o más Años , Secuencia de Bases , Neoplasias Esofágicas/tratamiento farmacológico , Neoplasias Esofágicas/patología , Femenino , Orden Génico , Genotipo , Humanos , Masculino , Persona de Mediana Edad , Datos de Secuencia Molecular , Estadificación de Neoplasias , Polimorfismo de Nucleótido Simple , Neoplasias Gástricas/tratamiento farmacológico , Inhibidor Tisular de Metaloproteinasa-3/química , Resultado del Tratamiento
19.
Biochem J ; 443(1): 307-15, 2012 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-22299597

RESUMEN

The semi-synthetic sulfated polysaccharide PPS (pentosan polysulfate) increases affinity between the aggrecan-degrading ADAMTSs (adamalysins with thrombospondin motifs) and their endogenous inhibitor, TIMP (tissue inhibitor of metalloproteinases)-3. In the present study we demonstrate that PPS mediates the formation of a high-affinity trimolecular complex with ADAMTS-5 and TIMP-3. A TIMP-3 mutant that lacks extracellular-matrix-binding ability was insensitive to this affinity increase, and truncated forms of ADAMTS-5 that lack the Sp (spacer) domain had reduced PPS-binding ability and sensitivity to the affinity increase. PPS molecules composed of 11 or more saccharide units were 100-fold more effective than those of eight saccharide units, indicating the involvement of extended or multiple protein-interaction sites. The formation of a high-affinity trimolecular complex was completely abolished in the presence of 0.4 M NaCl. These results suggest that PPS enhances the affinity between ADAMTS-5 and TIMP-3 by forming electrostatically driven trimolecular complexes under physiological conditions.


Asunto(s)
Proteínas ADAM/química , Poliéster Pentosan Sulfúrico/farmacología , Inhibidor Tisular de Metaloproteinasa-3/química , Proteínas ADAM/biosíntesis , Proteínas ADAM/genética , Proteína ADAMTS5 , Sustitución de Aminoácidos , Cromatografía en Gel , Células HEK293 , Humanos , Poliéster Pentosan Sulfúrico/aislamiento & purificación , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Eliminación de Secuencia , Cloruro de Sodio/química , Inhibidor Tisular de Metaloproteinasa-3/biosíntesis , Inhibidor Tisular de Metaloproteinasa-3/genética
20.
Biochem J ; 431(1): 113-22, 2010 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-20645923

RESUMEN

We have reported previously that reactive-site mutants of N-TIMP-3 [N-terminal inhibitory domain of TIMP-3 (tissue inhibitor of metalloproteinases 3)] modified at the N-terminus, selectively inhibited ADAM17 (a disintegrin and metalloproteinase 17) over the MMPs (matrix metalloproteinases). The primary aggrecanases ADAMTS (ADAM with thrombospondin motifs) -4 and -5 are ADAM17-related metalloproteinases which are similarly inhibited by TIMP-3, but are poorly inhibited by other TIMPs. Using a newly developed recombinant protein substrate based on the IGD (interglobular domain) of aggrecan, gst-IGD-flag, these reactive-site mutants were found to similarly inhibit ADAMTS-4 and ADAMTS-5. Further mutations of N-TIMP-3 indicated that up to two extra alanine residues can be attached to the N-terminus before the Ki (app) for ADAMTS-4 and ADAMTS-5 increased to over 100 nM. No other residues tested at the [-1] position produced inhibitors as potent as the alanine mutant. The mutants N-TIMP-3(T2G), [-1A]N-TIMP-3 and [-2A]N-TIMP-3 were effective inhibitors of aggrecan degradation, but not of collagen degradation in both IL-1α (interleukin-1α)-stimulated porcine articular cartilage explants and IL-1α with oncostatin M-stimulated human cartilage explants. Molecular modelling studies indicated that the [-1A]N-TIMP-3 mutant has additional stabilizing interactions with the catalytic domains of ADAM17, ADAMTS-4 and ADAMTS-5 that are absent from complexes with MMPs. These observations suggest that further mutation of the residues of N-TIMP-3 which make unique contacts with these metalloproteinases may allow discrimination between them.


Asunto(s)
Proteínas ADAM/antagonistas & inhibidores , Procolágeno N-Endopeptidasa/antagonistas & inhibidores , Procolágeno N-Endopeptidasa/química , Inhibidor Tisular de Metaloproteinasa-3/química , Inhibidor Tisular de Metaloproteinasa-3/genética , Proteínas ADAM/química , Proteínas ADAM/genética , Proteína ADAMTS4 , Agrecanos/metabolismo , Animales , Cartílago Articular/metabolismo , Dominio Catalítico , Células Cultivadas , Humanos , Mutación , Procolágeno N-Endopeptidasa/genética , Porcinos , Inhibidor Tisular de Metaloproteinasa-3/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...