Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 442
Filtrar
1.
Exp Dermatol ; 30(1): 112-120, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32813921

RESUMEN

Hypertrophic scars are a common complication of burn injuries and represent a major challenge in terms of prevention and treatment. These scars are characterized by a supraphysiological vascular density and by the presence of pathological myofibroblasts (Hmyos) displaying a low apoptosis propensity. However, the nature of the association between these two hallmarks of hypertrophic scarring remains largely unexplored. Here, we show that Hmyos produce signalling entities known as microvesicles that significantly increase the three cellular processes underlying blood vessel formation: endothelial cell proliferation, migration and assembly into capillary-like structures. The release of microvesicles from Hmyos was dose-dependently induced by the serum protein α-2-macroglobulin. Using flow cytometry, we revealed the presence of the α-2-macroglobulin receptor-low-density lipoprotein receptor-related protein 1-on the surface of Hmyos. The inhibition of the binding of α-2-macroglobulin to its receptor abolished the shedding of proangiogenic microvesicles from Hmyos. These findings suggest that the production of microvesicles by Hmyos contributes to the excessive vascularization of hypertrophic scars. α-2-Macroglobulin modulates the release of these microvesicles through interaction with low-density lipoprotein receptor-related protein 1.


Asunto(s)
Micropartículas Derivadas de Células/metabolismo , Cicatriz Hipertrófica/metabolismo , Miofibroblastos , Neovascularización Patológica/metabolismo , alfa-Macroglobulinas/metabolismo , Adulto , Movimiento Celular , Proliferación Celular , Células Cultivadas , Cicatriz Hipertrófica/patología , Células Endoteliales/fisiología , Humanos , Proteína Asociada a Proteínas Relacionadas con Receptor de LDL/metabolismo , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Miofibroblastos/metabolismo , Neovascularización Patológica/patología , Adulto Joven , alfa-Macroglobulinas/farmacología
2.
J Cell Biochem ; 118(7): 1810-1818, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28012205

RESUMEN

Distinct modes of cell migration contribute to diverse types of cell movements. The mesenchymal mode is characterized by a multistep cycle of membrane protrusion, the formation of focal adhesion, and the stabilization at the leading edge associated with the degradation of extracellular matrix (ECM) components and with regulated extracellular proteolysis. Both α2 -Macroglobulin (α2 M) and its receptor, low density lipoprotein receptor-related protein 1 (LRP1), play important roles in inflammatory processes, by controlling the extracellular activity of several proteases. The binding of the active form of α2 M (α2 M*) to LRP1 can also activate different signaling pathways in macrophages, thus inducing extracellular matrix metalloproteinase-9 (MMP-9) activation and cellular proliferation. In the present study, we investigated whether the α2 M*/LRP1 interaction induces cellular migration of the macrophage-derived cell line, Raw264.7. By using the wound-scratch migration assay and confocal microscopy, we demonstrate that α2 M* induces LRP1-mediated mesenchymal cellular migration. This migration exhibits the production of enlarged cellular protrusions, MT1-MMP distribution to these leading edge protrusions, actin polymerization, focal adhesion formation, and increased intracellular LRP1/ß1-integrin colocalization. Moreover, the presence of calphostin-C blocked the α2 M*-stimulated cellular protrusions, suggesting that the PKC activation is involved in the cellular motility of Raw264.7 cells. These findings could constitute a therapeutic target for inflammatory processes with deleterious consequences for human health, such as rheumatoid arthritis, atherosclerosis and cancer. J. Cell. Biochem. 118: 1810-1818, 2017. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Macrófagos/citología , Macrófagos/efectos de los fármacos , alfa-Macroglobulinas/farmacología , Animales , Movimiento Celular/efectos de los fármacos , Citoprotección/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Matriz Extracelular/metabolismo , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Microscopía Confocal , Naftalenos/farmacología , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/metabolismo , Células RAW 264.7 , Transducción de Señal/efectos de los fármacos
3.
Biol Chem ; 398(9): 975-994, 2017 08 28.
Artículo en Inglés | MEDLINE | ID: mdl-28253193

RESUMEN

Peptidases must be exquisitely regulated to prevent erroneous cleavage and one control is provided by protein inhibitors. These are usually specific for particular peptidases or families and sterically block the active-site cleft of target enzymes using lock-and-key mechanisms. In contrast, members of the +1400-residue multi-domain α2-macroglobulin inhibitor family (α2Ms) are directed against a broad spectrum of endopeptidases of disparate specificities and catalytic types, and they inhibit their targets without disturbing their active sites. This is achieved by irreversible trap mechanisms resulting from large conformational rearrangement upon cleavage in a promiscuous bait region through the prey endopeptidase. After decades of research, high-resolution structural details of these mechanisms have begun to emerge for tetrameric and monomeric α2Ms, which use 'Venus-flytrap' and 'snap-trap' mechanisms, respectively. In the former, represented by archetypal human α2M, inhibition is exerted through physical entrapment in a large cage, in which preys are still active against small substrates and inhibitors that can enter the cage through several apertures. In the latter, represented by a bacterial α2M from Escherichia coli, covalent linkage and steric hindrance of the prey inhibit activity, but only against very large substrates.


Asunto(s)
Endopeptidasas/metabolismo , Inhibidores de Proteasas/química , Inhibidores de Proteasas/farmacología , alfa-Macroglobulinas/química , alfa-Macroglobulinas/farmacología , Animales , Endopeptidasas/química , Humanos , Multimerización de Proteína , Estructura Cuaternaria de Proteína
4.
J Biol Chem ; 290(15): 9571-87, 2015 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-25720493

RESUMEN

Ligation of cell surface GRP78 by activated α2-macroglobulin (α2M*) promotes cell proliferation and suppresses apoptosis. α2M*-treated human prostate cancer cells exhibit a 2-3-fold increase in glucose uptake and lactate secretion, an effect similar to insulin treatment. In both α2M* and insulin-treated cells, the mRNA levels of SREBP1-c, SREBP2, fatty-acid synthase, acetyl-CoA carboxylase, ATP citrate lyase, and Glut-1 were significantly increased together with their protein levels, except for SREBP2. Pretreatment of cells with α2M* antagonist antibody directed against the carboxyl-terminal domain of GRP78 blocks these α2M*-mediated effects, and silencing GRP78 expression by RNAi inhibits up-regulation of ATP citrate lyase and fatty-acid synthase. α2M* induces a 2-3-fold increase in lipogenesis as determined by 6-[(14)C]glucose or 1-[(14)C]acetate incorporation into free cholesterol, cholesterol esters, triglycerides, free fatty acids, and phosphatidylcholine, which is blocked by inhibitors of fatty-acid synthase, PI 3-kinase, mTORC, or an antibody against the carboxyl-terminal domain of GRP78. We also assessed the incorporation of [(14)CH3]choline into phosphatidylcholine and observed similar effects. Lipogenesis is significantly affected by pretreatment of prostate cancer cells with fatostatin A, which blocks sterol regulatory element-binding protein proteolytic cleavage and activation. This study demonstrates that α2M* functions as a growth factor, leading to proliferation of prostate cancer cells by promoting insulin-like responses. An antibody against the carboxyl-terminal domain of GRP78 may have important applications in prostate cancer therapy.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Glucosa/metabolismo , Insulina/farmacología , alfa-Macroglobulinas/farmacología , Anticuerpos Antineoplásicos/inmunología , Anticuerpos Antineoplásicos/farmacología , Western Blotting , Línea Celular Tumoral , Colesterol/metabolismo , Chaperón BiP del Retículo Endoplásmico , Acido Graso Sintasa Tipo I/genética , Acido Graso Sintasa Tipo I/metabolismo , Expresión Génica/efectos de los fármacos , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/inmunología , Proteínas de Choque Térmico/metabolismo , Humanos , Hipoglucemiantes/farmacología , Lactatos/metabolismo , Metabolismo de los Lípidos/efectos de los fármacos , Metabolismo de los Lípidos/genética , Lipogénesis/efectos de los fármacos , Lipogénesis/genética , Masculino , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Unión Proteica , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , alfa-Macroglobulinas/metabolismo
5.
J Sci Food Agric ; 96(9): 3188-94, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26467490

RESUMEN

BACKGROUND: Hen egg white ovomacroglobulin (OVM) possesses a variety of bioactivities and could potentially be used as a pharmaceutical agent. It has been reported that OVM is involved in wound healing and cancer pathological processes, and previous results suggest that OVM plays a potential role in cell proliferation and migration; however, this has not yet been proven. In the present study, the effects of OVM on fibroblast proliferation and migration were evaluated. RESULTS: Results of cell counting, cell viability, and cell cycle indicated that proliferation of fibroblasts was not altered by OVM treatment. However, scratch assays showed that OVM could promote the migration of 3 T6 mouse embryonic fibroblasts and human skin fibroblasts (HSF). Also, the adhesion of HSF to the collagen matrix was also enhanced by OVM treatment. RT-qPCR and western blot analysis showed that ß1 -integrin, ß-tubulin, and ß-actin were up-regulated while E-cadherin was down-regulated in OVM-treated HSF cells. The effect of OVM was silenced after forming a complex with trypsin, suggesting that the protease inhibitory ability of OVM is important for its effect on cell migration. CONCLUSION: These results suggested that promotion of OVM on cell migration was achieved by enhancing cell adhesion to extracellular matrix, reducing intercellular aggregation, and strengthening cytoskeleton. The finding of the promotion effect of OVM on cell migration is important for understanding its role in wound healing and cancer pathological processes. © 2015 Society of Chemical Industry.


Asunto(s)
Adhesión Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Citoesqueleto/efectos de los fármacos , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , alfa-Macroglobulinas/farmacología , Animales , Ciclo Celular/efectos de los fármacos , Línea Celular , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Pollos , Clara de Huevo , Matriz Extracelular/efectos de los fármacos , Femenino , Fibroblastos/metabolismo , Humanos , Ratones , Microtúbulos/efectos de los fármacos , Piel/citología , Piel/efectos de los fármacos , Piel/metabolismo , Tubulina (Proteína)/metabolismo , Cicatrización de Heridas/efectos de los fármacos
6.
FASEB J ; 27(8): 3181-97, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23640058

RESUMEN

In retinal proliferative diseases, Müller glial cells (MGCs) acquire migratory abilities. However, the mechanisms that regulate this migration remain poorly understood. In addition, proliferative disorders associated with enhanced activities of matrix metalloprotease 2 (MMP-2) and MMP-9 also present increased levels of the protease inhibitor α2-macroglobulin (α2M) and its receptor, the low-density lipoprotein receptor-related protein 1 (LRP1). In the present work, we investigated whether the protease activated form of α2M, α2M*, and LRP1 are involved with the MGC migratory process. By performing wound-scratch migration and zymography assays, we demonstrated that α2M* induced cell migration and proMMP-2 activation in the human Müller glial cell line, MIO-M1. This induction was blocked when LRP1 and MT1-MMP were knocked down with siRNA techniques. Using fluorescence microscopy and biochemical procedures, we found that α2M* induced an increase in LRP1 and MT1-MMP accumulation in early endosomes, followed by endocytic recycling and intracellular distribution of MT1-MMP toward cellular protrusions. Moreover, Rab11-dominant negative mutant abrogated MT1-MMP recycling pathway, cell migration, and proMMP-2 activation induced by α2M*. In conclusion, α2M*, through its receptor LRP1, induces cellular migration of Müller glial cells by a mechanism that involves MT1-MMP intracellular traffic to the plasma membrane by a Rab11-dependent recycling pathway.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Metaloproteinasa 14 de la Matriz/metabolismo , alfa-Macroglobulinas/farmacología , Proteínas Portadoras/farmacología , Línea Celular , Membrana Celular/metabolismo , Endocitosis/efectos de los fármacos , Endosomas/metabolismo , Glutatión Transferasa/farmacología , Humanos , Immunoblotting , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Metaloproteinasa 14 de la Matriz/genética , Microscopía Confocal , Modelos Biológicos , Mutación , Neuroglía/citología , Unión Proteica/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Interferencia de ARN , Proteínas Recombinantes de Fusión/farmacología , Transducción de Señal/efectos de los fármacos , Proteínas de Unión al GTP rab/genética , Proteínas de Unión al GTP rab/metabolismo
7.
Biol Chem ; 393(10): 1193-200, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23089609

RESUMEN

ASP is a serine protease secreted by Aeromonas sobria. ASP cleaves various plasma proteins, which is associated with onset of sepsis complications, such as shock and blood coagulation disorder. To investigate a host defense mechanism against this virulence factor, we examined the plasma for ASP inhibitor(s). Human plasma inhibited ASP activity for azocasein, which was almost completely abolished by treating plasma with methylamine, which inactivates α2-macroglobulin (α2-MG). The ASP-inhibitor complex in ASP-added plasma was not detected by immunoblotting using anti-ASP antibody; however, using gel filtration of the plasma ASP activity for an oligopeptide, the ASP substrate was eluted in the void fraction (Mw>200 000), suggesting ASP trapping by α2-MG. Indeed, human α2-MG inhibited ASP azocaseinolytic activity in a dose-dependent manner, rapidly forming a complex with the ASP. Fibrinogen degradation by ASP was completely inhibited in the presence of α2-MG. α1-Protease inhibitor, antithrombin, and α2-plasmin inhibitor neither inhibited ASP activity nor formed a complex with ASP. Surprisingly, ASP degraded these plasma serine protease inhibitors. Thus, α2-MG is the major ASP inhibitor in the human plasma and can limit ASP virulence activities in A. sobria infection sites. However, as shown by fluorescence correlation spectroscopy, slow ASP inhibition by α2-MG in plasma may indicate insufficient ASP control in vivo.


Asunto(s)
Aeromonas/enzimología , Serina Proteasas/metabolismo , Inhibidores de Serina Proteinasa/farmacología , alfa-Macroglobulinas/farmacología , Fibrinógeno/metabolismo , Humanos , Proteolisis/efectos de los fármacos , Inhibidores de Serina Proteinasa/sangre
8.
J Cell Biochem ; 112(6): 1685-95, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21503958

RESUMEN

Receptor-recognized forms of α2 -macroglobulin (α2 M) bind to cell surface-associated GRP78 and initiate pro-proliferative and anti-apoptotic signaling. Ligation of GRP78 with α2 M also upregulates TFII-I, which binds to the GRP78 promoter and enhances GRP78 synthesis. In addition to its transcriptional functions, cytosolic TFII-I regulates agonist-induced Ca(2+) entry. In this study we show that down regulation of TFII-I gene expression by RNAi profoundly impairs its cell surface expression and anti-apoptotic signaling as measured by significant reduction of GRP78, Bcl-2, and cyclin D1 in 1-Ln and DU-145 human prostate cancer cells stimulated with α2 M. In contrast, this treatment significantly increases levels of the pro-apoptotic proteins p53, p27, Bax, and Bak and causes DNA fragmentation. Furthermore, down regulation of TFII-I expression activates agonist-induced Ca(2+) entry. In plasma membrane lysates p-PLCγ1, TRPC3, GRP78, MTJ1, and caveolin co-immunoprecipitate with TFII-I suggesting multimeric complexes of these proteins. Consistent with this hypothesis, down regulating TFII-I, MTJ1, or GRP78 expression by RNAi greatly attenuates cell surface expression of TFII-I. In conclusion, we demonstrate that not only does cell surface GRP78 regulate apoptosis, but it also regulates Ca(2+) homeostasis by controlling cell surface localization of TFII-I.


Asunto(s)
Apoptosis/efectos de los fármacos , Neoplasias de la Próstata/metabolismo , Factores de Transcripción TFII/metabolismo , alfa-Macroglobulinas/farmacología , Apoptosis/genética , Western Blotting , Caveolina 1/genética , Caveolina 1/metabolismo , Línea Celular Tumoral , Membrana Celular/metabolismo , Chaperón BiP del Retículo Endoplásmico , Citometría de Flujo , Proteínas del Choque Térmico HSP40/genética , Proteínas del Choque Térmico HSP40/metabolismo , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Humanos , Inmunoprecipitación , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Neoplasias de la Próstata/genética , Unión Proteica , Interferencia de ARN , Canales Catiónicos TRPC/genética , Canales Catiónicos TRPC/metabolismo , Factores de Transcripción TFII/genética
9.
Radiat Environ Biophys ; 50(1): 167-79, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20848291

RESUMEN

Previously, we showed that administration of the acute-phase protein α(2)-macroglobulin (α(2)M) to rats before total-body irradiation with 6.7 Gy (LD(50/30)) of X-rays provides the same level of radioprotection as amifostine. Here, we compare the cytoprotective effects of α(2)M and amifostine on rat liver. The potential of the liver to replenish cells destroyed by ionizing radiation was assessed by immunoblot analysis with antibody to proliferating cell nuclear antigen (PCNA). After irradiation, in unprotected rats PCNA decreased 6-fold from the basal level. In rats pretreated with either α(2)M or amifostine, PCNA was increased throughout a 4 week follow-up period, indicating that hepatocyte proliferation was unaffected. Since PCNA is an important component of the repair machinery, its increased expression was accompanied by significantly lower DNA damage in α(2)M- and amifostine-treated rats. At 2 weeks after irradiation, the Comet assay revealed a 15-fold increase in DNA damage in unprotected rats, while in α(2)M- and amifostine-treated rats we observed 3- and 4-fold rise in damage, respectively. The improved protection to DNA damage was supported by elevated activity of the antioxidant systems. Compared to untreated rats, pretreatments with α(2)M and amifostine led to similar increases in levels of the inflammatory cytokine IL-6 and the redox-sensitive transcription factor NFκB, promoting upregulation of MnSOD, the major component of the cell's antioxidant axis, and subsequent increases in Mn/CuZnSOD and catalase enzymatic activities. The results show that α(2)M induces protein factors whose interplay underlies radioprotection and support the idea that α(2)M is the central effector of natural radioprotection in the rat.


Asunto(s)
Citoprotección/efectos de los fármacos , Hígado/efectos de los fármacos , Hígado/efectos de la radiación , Irradiación Corporal Total , alfa-Macroglobulinas/administración & dosificación , alfa-Macroglobulinas/farmacología , Amifostina/farmacología , Animales , Antioxidantes/metabolismo , Catalasa/metabolismo , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Reparación del ADN/efectos de los fármacos , Reparación del ADN/efectos de la radiación , Hepatocitos/citología , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Hepatocitos/efectos de la radiación , Interleucina-6/sangre , Hígado/citología , Hígado/metabolismo , Dosis de Radiación , Ratas , Ratas Wistar , Especies Reactivas de Oxígeno/metabolismo , Superóxido Dismutasa/metabolismo , Factor de Transcripción ReIA/metabolismo
10.
Sci Rep ; 11(1): 7953, 2021 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-33846468

RESUMEN

The molecular mechanism for acute kidney injury (AKI) and its progression to chronic kidney disease (CKD) continues to be unclear. In this study, we investigated the pathophysiological role of the acute phase protein α1-acid glycoprotein (AGP) in AKI and its progression to CKD using AGP KO mice. Plasma AGP levels in WT mice were increased by about 3.5-fold on day 1-2 after renal ischemia-reperfusion (IR), and these values then gradually decreased to the level before renal IR on day 7-14. On day 1 after renal IR, the AGP KO showed higher renal dysfunction, tubular injury and renal inflammation as compared with WT. On day 14, renal function, tubular injury and renal inflammation in WT had recovered, but the recovery was delayed, and renal fibrosis continued to progress in AGP KO. These results obtained from AGP KO were rescued by the administration of human-derived AGP (hAGP) simultaneously with renal IR. In vitro experiments using RAW264.7 cells showed hAGP treatment suppressed the LPS-induced macrophage inflammatory response. These data suggest that endogenously induced AGP in early renal IR functions as a renoprotective molecule via its anti-inflammatory action. Thus, AGP represents a potential target molecule for therapeutic development in AKI and its progression CKD.


Asunto(s)
Lesión Renal Aguda/tratamiento farmacológico , Lesión Renal Aguda/patología , Antiinflamatorios/uso terapéutico , Progresión de la Enfermedad , Insuficiencia Renal Crónica/tratamiento farmacológico , alfa-Macroglobulinas/uso terapéutico , Lesión Renal Aguda/sangre , Lesión Renal Aguda/complicaciones , Animales , Antiinflamatorios/farmacología , Humanos , Inflamación/sangre , Inflamación/complicaciones , Inflamación/tratamiento farmacológico , Riñón/efectos de los fármacos , Riñón/patología , Riñón/fisiopatología , Pruebas de Función Renal , Lipopolisacáridos , Macrófagos/efectos de los fármacos , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células RAW 264.7 , Insuficiencia Renal Crónica/sangre , Insuficiencia Renal Crónica/complicaciones , Insuficiencia Renal Crónica/patología , Daño por Reperfusión/sangre , alfa-Macroglobulinas/administración & dosificación , alfa-Macroglobulinas/farmacología
11.
J Exp Med ; 143(2): 462-7, 1976 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-55456

RESUMEN

The following observations indicate that cultured human WI-38 fibroblasts synthesize and secrete alpha2-macroglobulin into serum-free medium: (a) after incubation of cultures with [35S]L-methionine, a labeled protein appeared in the medium which was precipitated by antiserum directed against alpha2-macroglobulin; (b) after incubation of cultures with [35S]L-methionine, a major band of radioactivity detected by polyacrylamide gel electrophoresis of the proteins in medium co-migrated with alpha2-macroglobulin; and (c) the amount of alpha2-macroblobulin in the medium, estimated both functionally and immunologically, increased with time in normal but not not puromycin-treated cultures.


Asunto(s)
Fibroblastos/metabolismo , alfa-Macroglobulinas/metabolismo , Células Cultivadas , Humanos , Sueros Inmunes , Técnicas In Vitro , Inhibidores de Proteasas , alfa-Macroglobulinas/farmacología
12.
Biochem Biophys Res Commun ; 392(2): 135-9, 2010 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-20056109

RESUMEN

Omega-3 (n-3) fatty acids are emerging as bioactive agents protective against cardiovascular disease. However, their cellular delivery pathways are poorly defined. Here we questioned whether the uptake of n-3 triglyceride-rich particles (TGRP) is mediated by cell surface proteoglycans (PG) using LDL receptor (LDLR)+/+ and LDLR-/- cell models. LDLR+/+ but not LDLR-/- cells showed higher n-6 over n-3 TGRP uptake. Removal of cell surface proteins and receptors by pronase markedly enhanced the uptake of n-3 but not n-6 TGRP. Lactoferrin blockage of apoE-mediated pathways decreased the uptake of n-6 TGRP by up to 85% (p<0.05) but had insignificant effect on n-3 TGRP uptake. PG removal by sodium chlorate in LDLR+/+ cells substantially reduced n-3 TGRP uptake but had little effect on n-6 TGRP uptake. Thus, while n-6 TGRP uptake is preferentially mediated by LDLR-dependent pathways, the uptake of n-3 TGRP depends more on PG and non-LDLR cell surface anchoring.


Asunto(s)
Membrana Celular/metabolismo , Ácidos Grasos Omega-3/metabolismo , Ácidos Grasos Omega-6/metabolismo , Receptores de LDL/metabolismo , Triglicéridos/metabolismo , Línea Celular , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Humanos , Lactoferrina/farmacología , Proteoglicanos/metabolismo , Receptores de LDL/genética , alfa-Macroglobulinas/farmacología
13.
J Shoulder Elbow Surg ; 19(3): 384-91, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19800260

RESUMEN

HYPOTHESIS: Recent studies have demonstrated a potentially critical role of matrix metalloproteinases (MMPs) and tissue inhibitors of matrix metalloproteinases (TIMPs) in the pathophysiology of rotator cuff tears. We hypothesize that local delivery of a MMP inhibitor after surgical repair of the rotator cuff will improve healing at the tendon-to-bone surface interface. MATERIALS AND METHODS: Sixty-two male Sprague-Dawley rats underwent acute supraspinatus detachment and repair. In the control group (n=31), the supraspinatus was repaired to its anatomic footprint. In the experimental group (n=31), recombinant alpha-2-macroglobulin (A2M) protein, a universal MMP inhibitor, was applied at the tendon-bone interface with an identical surgical repair. Animals were sacrificed at 2 and 4 weeks for histomorphometry, immunohistochemistry, and biomechanical testing. Statistical comparisons were performed using unpaired t tests. Significance was set at P < .05. RESULTS: Significantly greater fibrocartilage was seen at the healing enthesis in the A2M-treated specimens compared with controls at 2 weeks (P < .05). Significantly greater collagen organization was observed in the A2M-treated animals compared with controls at 4 weeks (P < .01). A significant reduction in collagen degradation was observed at both 2 and 4 weeks in the experimental group (P < .05). Biomechanical testing revealed no significant differences in stiffness or ultimate load-to-failure. CONCLUSION: Local delivery of an MMP inhibitor is associated with distinct histologic differences at the tendon-to-bone interface after rotator cuff repair. Modulation of MMP activity after rotator cuff repair may offer a novel biologic pathway to augment tendon-to-bone healing after rotator cuff repair.


Asunto(s)
Huesos/efectos de los fármacos , Inhibidores de la Metaloproteinasa de la Matriz , Manguito de los Rotadores/fisiopatología , Tendones/efectos de los fármacos , Cicatrización de Heridas/efectos de los fármacos , Animales , Fenómenos Biomecánicos , Huesos/fisiopatología , Colágeno/biosíntesis , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Fibrocartílago/fisiología , Masculino , Ratas , Ratas Sprague-Dawley , Manguito de los Rotadores/cirugía , Tendones/fisiopatología , alfa-Macroglobulinas/farmacología
14.
J Cell Biochem ; 106(3): 381-9, 2009 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-19097122

RESUMEN

Receptor-recognized forms of alpha(2)-macroglobulin (alpha(2)M*) bind to cell surface-associated GRP78 and induce proliferative and survival signaling in prostate cancer cells. As part of the cellular response to alpha(2)M*, GRP78 expression is itself upregulated. In response to other stimuli, the transcription factor TFII-I upregulates GRP78 by binding to its gene promoter. We have, therefore, studied the role of TFII-I in transcriptional upregulation of GRP78 in 1-LN human prostate cancer cells stimulated with alpha(2)M*. This treatment caused a two- to threefold increase in TFII-I and GRP78 synthesis from [(35)S]-labeled precursor amino acids. Synthesis of both TFII-I and GRP78 were significantly reduced by silencing TFII-I gene expression or pretreatment of cells with genistein or actinomycin D. Confocal microscopy was employed to demonstrate relocation of TFII-I to the nucleus. In alpha(2)M*-stimulated cells, moreover, TFII-I bound to the GRP78 promoter as determined by CHIP assay. We also demonstrate binding of TFII-I to the c-fos promoter, consistent with its role in upregulating c-fos gene expression. In non-lymphoid cells, phosphorylated c-Src is an activator of TFII-I. Ligation of GRP78 on 1-LN cells with alpha(2)M* was followed by tyrosine phosphorylation of c-Src as well as TFII-I. We conclude that alpha(2)M*-induced increase in GRP78 synthesis is caused by transcriptional upregulation of TFII-I which binds to the GRP78 promoter and thus potentiates its cell survival and antipoptotic functions in 1-LN prostate cancer cells.


Asunto(s)
Regulación Neoplásica de la Expresión Génica/genética , Proteínas de Choque Térmico/genética , Chaperonas Moleculares/genética , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Factores de Transcripción TFII/metabolismo , Transcripción Genética/genética , Regulación hacia Arriba/genética , Proteína Tirosina Quinasa CSK , Línea Celular Tumoral , Chaperón BiP del Retículo Endoplásmico , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Proteínas de Choque Térmico/biosíntesis , Humanos , Masculino , Chaperonas Moleculares/biosíntesis , Fosforilación , Fosfotirosina/metabolismo , Neoplasias de la Próstata/metabolismo , Unión Proteica , Transporte de Proteínas , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Interferencia de ARN , Factores de Transcripción TFII/genética , Transcripción Genética/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , alfa-Macroglobulinas/farmacología , Familia-src Quinasas
15.
Am J Physiol Lung Cell Mol Physiol ; 297(4): L568-77, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19666776

RESUMEN

The proenzyme single-chain urokinase plasminogen activator (scuPA) more effectively resolved intrapleural loculations in rabbits with tetracycline (TCN)-induced loculation than a range of clinical doses of two-chain uPA (Abbokinase) and demonstrated a trend toward greater efficacy than single-chain tPA (Activase) (Idell S et al., Exp Lung Res 33: 419, 2007.). scuPA more slowly generates durable intrapleural fibrinolytic activity than Abbokinase or Activase, but the interactions of these agents with inhibitors in pleural fluids (PFs) have been poorly understood. PFs from rabbits with TCN-induced pleural injury treated with intrapleural scuPA, its inactive Ser195Ala mutant, Abbokinase, Activase, or vehicle, were analyzed to define the mechanism by which scuPA induces durable fibrinolysis. uPA activity was elevated in PFs of animals treated with scuPA, correlated with the ability to clear pleural loculations, and resisted (70-80%) inhibition by PAI-1. Alpha-macroglobulin (alphaM) but not urokinase receptor complexes immunoprecipitated from PFs of scuPA-treated rabbits retained uPA activity that resists PAI-1 and activates plasminogen. Conversely, little plasminogen activating or enzymatic activity resistant to PAI-1 was detectable in PFs of rabbits treated with Abbokinase or Activase. Consistent with these findings, PAI-1 interacts with scuPA much slower than with Activase or Abbokinase in vitro. An equilibrium between active and inactive scuPA (k(on) = 4.3 h(-1)) limits the rate of its inactivation by PAI-1, favoring formation of complexes with alphaM. These observations define a newly recognized mechanism that promotes durable intrapleural fibrinolysis via formation of alphaM/uPA complexes. These complexes promote uPA-mediated plasminogen activation in scuPA-treated rabbits with TCN-induced pleural injury.


Asunto(s)
Fibrinólisis/efectos de los fármacos , Inhibidor 1 de Activador Plasminogénico/farmacología , Pleura/efectos de los fármacos , Tetraciclina/toxicidad , Activador de Plasminógeno de Tipo Uroquinasa/farmacología , alfa-Macroglobulinas/farmacología , Animales , Western Blotting , Femenino , Inmunoprecipitación , Pleura/metabolismo , Inhibidores de la Síntesis de la Proteína/toxicidad , Conejos , Receptores del Activador de Plasminógeno Tipo Uroquinasa , Proteínas Recombinantes/farmacología , Activador de Tejido Plasminógeno/metabolismo , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo
16.
Int J Immunopathol Pharmacol ; 22(1): 183-93, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19309566

RESUMEN

Internalin A (InlA), a cell wall-bound protein of Listeria monocytogenes, is among the major components involved in the adhesion to and invasion of host cells expressing specific forms of E-cadherin. Some L. monocytogenes strains secrete truncated non-functional forms of InlA. The purpose of this study is to compare the biofilm-forming abilities of L. monocytogenes strains from clinical sources expressing InlA proteins in the different forms. A total of 70 L. monocytogenes strains were examined using SDS-PAGE, Western blot, DNA sequencing, and microtitre plate biofilm formation assays. We found that 8 of the 70 strains expressed truncated InlA, and that this group of strains exhibited significantly enhanced biofilm-forming ability compared to the group expressing full-length InlA. Further experiments showed that: (i) L. monocytogenes biofilms were detached by treatment with protease K; (ii) protein fragments resulting from proteolysis, rather than intact proteins, are responsible for biofilm enhancement, because biofilm formation was impaired by the protease inhibitor alpha2-macroglobulin; (iii) truncated and/or proteolytically cleaved InlA are likely involved in the biofilm enhancement, based on the effects that anti-InlA monoclonal antibodies produced on the biofilm formation of L. monocytogenes strains expressing either truncated or full-length InlA. These data provide a basis for further investigation of the molecular structure and composition of L. monocytogenes biofilms.


Asunto(s)
Proteínas Bacterianas/fisiología , Listeria monocytogenes/fisiología , Anticuerpos Monoclonales/inmunología , Proteínas Bacterianas/análisis , Proteínas Bacterianas/genética , Biopelículas , Western Blotting , Humanos , Análisis de Secuencia de ADN , alfa-Macroglobulinas/farmacología
17.
Med Sci Monit ; 15(7): BR188-93, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19564818

RESUMEN

BACKGROUND: Administration of the acute-phase protein alpha2-macroglobulin (MG) prior to total-body irradiation of rats with a 6.7 Gy (LD50) dose of X-rays exerts a radioprotective effect. MATERIAL/METHODS: MG was administered 30 min before irradiation with a 6.7 Gy (LD50) dose of X-rays. Its radioprotective efficacy was compared with that of the synthetic agent amifostine (WR-2721), a sulfhydryl compound which is currently the most effective radioprotector in clinical use. After administration of either MG or amifostine, changes in body and liver weight were recorded and histological liver sections were examined during a four-week follow-up period. RESULTS: As observed in the experimental group administered amifostine, rats that received MG prior to irradiation exhibited 100% survival and restoration of the body and liver weight to the control values. The morphological damage seen in the liver after irradiation of untreated rats was absent in both the MG- and amifostine-pretreated rats. Also, hepatocytes and granulated cells had prominent nuclei and did not exhibit major changes in volume. Dilation of the central vein was not observed. CONCLUSIONS: Administration of MG before irradiation, similar to pretreatment with amifostine, provided complete survival of experimental rats and recovery of liver weight and preserved major histological parameters of the liver.


Asunto(s)
Hígado/efectos de los fármacos , Hígado/patología , Protectores contra Radiación/farmacología , alfa-Macroglobulinas/farmacología , Amifostina/administración & dosificación , Amifostina/farmacología , Animales , Peso Corporal/efectos de los fármacos , Tamaño de los Órganos/efectos de los fármacos , Ratas , Análisis de Supervivencia , alfa-Macroglobulinas/administración & dosificación
18.
Cardiovasc Res ; 75(1): 118-28, 2007 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-17412314

RESUMEN

OBJECTIVE: Alpha2-macroglobulin (alpha2M) is an acute phase protein released to the serum upon challenges such as cardiac hypertrophy and infarction. Here we report on the role of alpha2M in the induction of hypertrophic cell growth, contractile responsiveness of rat ventricular cardiomyocytes, and on the underlying extracellular regulated kinase 1,2 (ERK1,2) and phosphoinositide 3-kinase (PI3-kinase)/Akt pathways. METHODS: Cell volume and cross-sectional areas were assessed as parameters of hypertrophic growth, and real time RT-PCR for the analysis of hypertrophy-related genes was performed. Protein synthesis was analyzed by 14C-phenylalanine incorporation. Activation of ERK1,2, PI3-kinase and Akt was assessed by immunohistochemical analysis of phosphorylated proteins. Contractile responsiveness was investigated by determination of cell shortening following electrical field stimulation. Intracellular calcium concentration [Ca2+]i was determined by fluo-3 microfluorometry. RESULTS: Treatment of ventricular cardiomyocytes for 24 h with alpha2M significantly increased cell volume and protein synthesis as well as expression of hypertrophy-associated genes [brain natriuretic protein (BNP), beta-myosin heavy chain (beta-MHC), myosin light chain-2 (MLC-2), atrial natriuretic factor (ANF), and skeletal alpha-actin]. Comparable effects were achieved by treatment of cells with an antibody directed against the alpha2M-receptor LDL receptor-related protein-1 (LRP-1) and counteracted upon coincubation with receptor-associated protein (RAP), suggesting an involvement of alpha2M-LRP-1 signalling. Furthermore, alpha2M treatment increased sarcoplasmic reticulum Ca2+-ATPase (SERCA-2a) expression, diastolic and systolic [Ca2+]i, and contractile responsiveness after electrical stimulation. Shortly after alpha2M stimulation, activation of ERK1,2, Akt, and PI3-kinase pathways was observed. Consequently, alpha2M-induced protein synthesis was inhibited upon treatment with the ERK1,2 inhibitor UO126 as well as by LY294002 and wortmannin, which inhibit PI3-kinase, and by rapamycin, which inhibits mammalian target of rapamycin (mTOR) downstream of Akt. CONCLUSIONS: Our data show that alpha2M induces hypertrophic cell growth in rat ventricular cardiomyocytes via ERK1,2 and PI3-kinase/Akt and improves cardiac cell function.


Asunto(s)
Cardiomegalia/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/fisiología , alfa-Macroglobulinas/metabolismo , Androstadienos/farmacología , Animales , Butadienos/farmacología , Calcio/metabolismo , Células Cultivadas , Cromonas/farmacología , Activación Enzimática , Inmunohistoquímica , Masculino , Morfolinas/farmacología , Nitrilos/farmacología , Fenilalanina/metabolismo , Ratas , Ratas Wistar , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Sirolimus/farmacología , Wortmanina , alfa-Macroglobulinas/antagonistas & inhibidores , alfa-Macroglobulinas/farmacología
19.
J Neurochem ; 103(6): 2482-90, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17908238

RESUMEN

Soluble amyloid-beta peptide (Abeta) exists in the form of monomers and oligomers, and as complexes with Abeta-binding molecules, such as low-density lipoprotein receptor-related protein-1 (LRP-1) ligands. The present study investigated the effect of self-aggregation and LRP-1 ligands on the elimination of human Abeta(1-40) [hAbeta(1-40)] from the rat brain across the blood-brain barrier. Incubation of [(125)I]hAbeta(1-40) monomer resulted in time-dependent and temperature-dependent dimer formation, and the apparent elimination rate of [(125)I]hAbeta(1-40) dimer was significantly decreased by 92.7% compared with that of [(125)I]hAbeta(1-40) monomer. Pre-incubation with LRP-1 ligands, such as activated alpha2-macroglobulin (alpha2M), apolipoprotein E2 (apoE2), apoE3, apoE4, and lactoferrin, reduced the elimination of [(125)I]hAbeta(1-40). By contrast, pre-administration of the same concentration of these molecules in the rat brain did not significantly inhibit [(125)I]hAbeta(1-40) monomer elimination. Purified [(125)I]hAbeta(1-40)/activated alpha2M complex and [(125)I]activated alpha2M were not significantly eliminated from the rat brain up to 60 min. MEF-1 cells, which have LRP-1-mediated endocytosis, exhibited uptake of [(125)I]activated alpha2M, and enhancement of [(125)I]hAbeta(1-40) uptake upon pre-incubation with apoE, suggesting that [(125)I]activated alpha2M and [(125)I]hAbeta(1-40)/apoE complex function as LRP-1 ligands. These findings indicate that dimerization and LRP-1-ligand complex formation prevent the elimination of hAbeta(1-40) from the brain across the blood-brain barrier.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Barrera Hematoencefálica/metabolismo , Corteza Cerebral/metabolismo , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Fragmentos de Péptidos/metabolismo , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/fisiopatología , Animales , Apolipoproteínas E/metabolismo , Apolipoproteínas E/farmacología , Barrera Hematoencefálica/efectos de los fármacos , Línea Celular , Corteza Cerebral/efectos de los fármacos , Dimerización , Endocitosis/fisiología , Humanos , Lactoferrina/metabolismo , Ligandos , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/efectos de los fármacos , Sustancias Macromoleculares/metabolismo , Masculino , Tasa de Depuración Metabólica/fisiología , Ratones , Unión Proteica/fisiología , Ratas , Ratas Sprague-Dawley , alfa-Macroglobulinas/metabolismo , alfa-Macroglobulinas/farmacología
20.
J Clin Invest ; 77(5): 1565-73, 1986 May.
Artículo en Inglés | MEDLINE | ID: mdl-2422211

RESUMEN

Recent studies from our laboratory indicate that a high concentration of platelet-derived calcium-activated cysteine protease (calpain) can cleave high molecular weight kininogen (HMWK). On immunodiffusion and immunoblot, antiserum directed to the heavy chain of HMWK showed immunochemical identity with alpha-cysteine protease inhibitor--a major plasma inhibitor of tissue calpains. Studies were then initiated to determine whether purified or plasma HMWK was also an inhibitor of platelet calpain. Purified alpha-cysteine protease inhibitor, alpha-2-macroglobulin, as well as purified heavy chain of HMWK or HMWK itself inhibited purified platelet calpain. Kinetic analysis revealed that HMWK inhibited platelet calpain noncompetitively (Ki approximately equal to 5 nM). Incubation of platelet calpain with HMWK, alpha-2-macroglobulin, purified heavy chain of HMWK, or purified alpha-cysteine protease inhibitor under similar conditions resulted in an IC50 of 36, 500, 700, and 1,700 nM, respectively. The contribution of these proteins in plasma towards the inhibition of platelet calpain was investigated next. Normal plasma contained a protein that conferred a five to sixfold greater IC50 of purified platelet calpain than plasma deficient in either HMWK or total kininogen. Reconstitution of total kininogen deficient plasma with purified HMWK to normal levels (0.67 microM) completely corrected the subnormal inhibitory activity. However, reconstitution of HMWK deficient plasma to normal levels of low molecular weight kininogen (2.4 microM) did not fully correct the subnormal calpain inhibitory capacity of this plasma. These studies indicate that HMWK is a potent inhibitor as well as a substrate of platelet calpain and that the plasma and cellular kininogens may function as regulators of cytosolic, calcium-activated cysteine proteases.


Asunto(s)
Plaquetas/enzimología , Glicoproteínas , Quininógenos/farmacología , Fenómenos Fisiológicos Sanguíneos , Proteínas Sanguíneas/farmacología , Calcio/fisiología , Calpaína/aislamiento & purificación , Humanos , Radioisótopos de Yodo , Cinética , Quininógenos/metabolismo , Peso Molecular , alfa-Macroglobulinas/farmacología
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda