Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
ACS Appl Mater Interfaces ; 16(24): 30703-30714, 2024 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-38848451

RESUMEN

In this paper, we propose a model that connects two standard inflammatory responses to viral infection, namely, elevation of fibrinogen and the lipid drop shower, to the initiation of non-thrombin-generated clot formation. In order to understand the molecular basis for the formation of non-thrombin-generated clots following viral infection, human epithelial and Madin-Darby Canine Kidney (MDCK, epithelial) cells were infected with H1N1, OC43, and adenovirus, and conditioned media was collected, which was later used to treat human umbilical vein endothelial cells and human lung microvascular endothelial cells. After direct infection or after exposure to conditioned media from infected cells, tissue surfaces of both epithelial and endothelial cells, exposed to 8 mg/mL fibrinogen, were observed to initiate fibrillogenesis in the absence of thrombin. No fibers were observed after direct viral exposure of the endothelium or when the epithelium cells were exposed to SARS-CoV-2 isolated spike proteins. Heating the conditioned media to 60 °C had no effect on fibrillogenesis, indicating that the effect was not enzymatic but rather associated with relatively thermally stable inflammatory factors released soon after viral infection. Spontaneous fibrillogenesis had previously been reported and interpreted as being due to the release of the alpha C domains due to strong interactions of the interior of the fibrinogen molecule in contact with hydrophobic material surfaces rather than cleavage of the fibrinopeptides. Contact angle goniometry and immunohistochemistry were used to demonstrate that the lipids produced within the epithelium and released in the conditioned media, probably after the death of infected epithelial cells, formed a hydrophobic residue responsible for fibrillogenesis. Hence, the standard inflammatory response constitutes the ideal conditions for surface-initiated clot formation.


Asunto(s)
Fibrinógeno , Humanos , Perros , Animales , Fibrinógeno/química , Fibrinógeno/metabolismo , Trombina/metabolismo , Trombina/farmacología , Células de Riñón Canino Madin Darby , Células Endoteliales de la Vena Umbilical Humana , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus/metabolismo , Glicoproteína de la Espiga del Coronavirus/química , Coagulación Sanguínea , COVID-19/virología , COVID-19/metabolismo , Medios de Cultivo Condicionados/farmacología , Medios de Cultivo Condicionados/química , Células Endoteliales/metabolismo , Células Endoteliales/virología , Células Epiteliales/virología , Células Epiteliales/metabolismo
2.
Nano Lett ; 24(7): 2218-2225, 2024 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-38277614

RESUMEN

Significant untapped energy exists within low-grade heat sources and salinity gradients. Traditional nanofluidic membranes exhibit inherent limitations, including low ion selectivity, high internal resistance, reliance on nonrenewable resources, and instability in aqueous solutions, invariably constraining their practical application. Here, an innovative composite membrane-based nanofluidic system is reported, involving the strategy of integrating tailor-modified bacterial nanofibers with boron nitride nanosheets, enabling high surface charge densities while maintaining a delicate balance between ion selectivity and permeability, ultimately facilitating effective thermo-osmotic energy harvesting. The device exhibits an impressive output power density of 10 W m-2 with artificial seawater and river water at a 50 K temperature gradient. Furthermore, it demonstrates robust power density stability under prolonged exposure to salinity gradients or even at elevated temperatures. This work opens new avenues for the development of nanofluidic systems utilizing composite materials and presents promising solutions for low-grade heat recovery and osmotic energy harvesting.

3.
Acta Pharmacol Sin ; 45(3): 517-530, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37880339

RESUMEN

Malignant ventricular arrhythmia (VA) after myocardial infarction (MI) is mainly caused by myocardial electrophysiological remodeling. Brahma-related gene 1 (BRG1) is an ATPase catalytic subunit that belongs to a family of chromatin remodeling complexes called Switch/Sucrose Non-Fermentable Chromatin (SWI/SNF). BRG1 has been reported as a molecular chaperone, interacting with various transcription factors or proteins to regulate transcription in cardiac diseases. In this study, we investigated the potential role of BRG1 in ion channel remodeling and VA after ischemic infarction. Myocardial infarction (MI) mice were established by ligating the left anterior descending (LAD) coronary artery, and electrocardiogram (ECG) was monitored. Epicardial conduction of MI mouse heart was characterized in Langendorff-perfused hearts using epicardial optical voltage mapping. Patch-clamping analysis was conducted in single ventricular cardiomyocytes isolated from the mice. We showed that BRG1 expression in the border zone was progressively increased in the first week following MI. Cardiac-specific deletion of BRG1 by tail vein injection of AAV9-BRG1-shRNA significantly ameliorated susceptibility to electrical-induced VA and shortened QTc intervals in MI mice. BRG1 knockdown significantly enhanced conduction velocity (CV) and reversed the prolonged action potential duration in MI mouse heart. Moreover, BRG1 knockdown improved the decreased densities of Na+ current (INa) and transient outward potassium current (Ito), as well as the expression of Nav1.5 and Kv4.3 in the border zone of MI mouse hearts and in hypoxia-treated neonatal mouse ventricular cardiomyocytes. We revealed that MI increased the binding among BRG1, T-cell factor 4 (TCF4) and ß-catenin, forming a transcription complex, which suppressed the transcription activity of SCN5A and KCND3, thereby influencing the incidence of VA post-MI.


Asunto(s)
Infarto del Miocardio , Ratones , Animales , Infarto del Miocardio/metabolismo , Arritmias Cardíacas/genética , Miocardio/patología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Miocitos Cardíacos/metabolismo
4.
PeerJ ; 11: e15407, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37250720

RESUMEN

Background: PFI-3 is a small-molecule inhibitor that targets the bromodomains (BRDs) of Brahma-related gene 1 (BRG1). This monomeric compound, which has high selectivity and potent cellular effects, has recently been developed. Although PFI-3 has been reported as a potential therapeutic agent targeting thrombomodulin, its role in the regulation of vascular function remains unknown. Therefore, we aimed to investigate the impact of PFI-3 on arterial vessel tone. Methods: A microvascular tension measurement device (DMT) was utilized to identify alterations in vascular tension within the mesenteric artery. To detect variations in cytosolic [Ca2+]i, a Fluo-3/AM fluorescent probe and fluorescence microscope were employed. Additionally, whole-cell patch clamp techniques were utilized to evaluate the activity of L-type voltage-dependent calcium channels (VDCCs) in cultured arterial smooth muscle cells (A10 cells). Results: PFI-3 exerted a dose-dependent relaxation effect on rat mesenteric arteries with both intact and denuded endothelium after phenylephrine (PE)- and high-K+-induced constriction. PFI-3-induced vasorelaxation was not affected by the presence of L-NAME/ODQ or K+ channel blockers (Gli/TEA). PFI-3 abolished Ca2+-induced contraction on endothelium-denuded mesenteric arteries preincubated by PE in Ca2+-free solution. Incubation with TG had no impact on PFI-3-induced vasorelaxation pre-contracted by PE. PFI-3 reduced Ca2+-induced contraction on endothelium-denuded mesenteric arteries pre-incubated by KCl (60 mM) in Ca2+-free solution. PFI-3 declined extracellular calcium influx in A10 cells detected by Fluo-3/AM fluorescent probe and fluorescence microscope. Furthermore, we observed that PFI-3 decreased the current densities of L-type VDCC by whole-cell patch clamp techniques. Conclusions: PFI-3 blunted PE and high K+-induced vasoconstriction independent of endothelium on rat mesenteric artery. The vasodilatory effect of PFI-3 may be attributed to its inhibition of VDCCs and receptor-operated calcium channels (ROCCs) on vascular smooth muscle cells (VSMCs).


Asunto(s)
Calcio , Colorantes Fluorescentes , Animales , Ratas , Calcio/metabolismo , Canales de Calcio Tipo L/farmacología , Colorantes Fluorescentes/farmacología , Arterias Mesentéricas
5.
Front Pharmacol ; 14: 1340247, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38269270

RESUMEN

Background: Vascular endothelial injury is a contributing factor to the development of atherosclerosis and the resulting cardiovascular diseases. One particular factor involved in endothelial cell apoptosis and atherosclerosis is palmitic acid (PA), which is a long-chain saturated fatty acid. In addition, transient receptor potential melastatin 4 (TRPM4), a non-selective cation channel, plays a significant role in endothelial dysfunction caused by various factors related to cardiovascular diseases. Despite this, the specific role and mechanisms of TRPM4 in atherosclerosis have not been fully understood. Methods: The protein and mRNA expressions of TRPM4, apoptosis - and inflammation-related factors were measured after PA treatment. The effect of TRPM4 knockout on the protein and mRNA expression of apoptosis and inflammation-related factors was detected. The changes of intracellular Ca2+, mitochondrial membrane potential, and reactive oxygen species were detected by Fluo-4 AM, JC-1, and DCFH-DA probes, respectively. To confirm the binding of miR-133a-3p to TRPM4, a dual luciferase reporter gene assay was conducted. Finally, the effects of miR-133a-3p and TRPM4 on intracellular Ca2+, mitochondrial membrane potential, and reactive oxygen species were examined. Results: Following PA treatment, the expression of TRPM4 increases, leading to calcium overload in endothelial cells. This calcium influx causes the assemblage of Bcl-2, resulting in the opening of mitochondrial calcium channels and mitochondrial damage, ultimately triggering apoptosis. Throughout this process, the mRNA and protein levels of IL-1ß, ICAM-1, and VCAM1 significantly increase. Database screenings and luciferase assays have shown that miR-133a-3p preferentially binds to the 3'UTR region of TRPM4 mRNA, suppressing TRPM4 expression. During PA-induced endothelial injury, miR-133a-3p is significantly decreased, but overexpression of miR-133a-3p can attenuate the progression of endothelial injury. On the other hand, overexpression of TRPM4 counteracts the aforementioned changes. Conclusion: TRPM4 participates in vascular endothelial injury caused by PA. Therefore, targeting TRPM4 or miR-133a-3p may offer a novel pharmacological approach to preventing endothelial injury.

6.
Development ; 149(21)2022 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-36281807

RESUMEN

Plants develop throughout their lives: seeds become seedlings that mature and form fruits and seeds. Although the underlying mechanisms that drive these developmental phase transitions have been well elucidated for shoots, the extent to which they affect the root is less clear. However, root anatomy does change as some plants mature; meristems enlarge and radial thickening occurs. Here, in Arabidopsis thaliana, we show that overexpressing miR156A, a gene that promotes the juvenile phase, increased the density of the root system, even in grafted plants in which only the rootstock had the overexpression genotype. In the root, overexpression of miR156A resulted in lower levels of PLETHORA 2, a protein that affects formation of the meristem and elongation zone. Crossing in an extra copy of PLETHORA 2 partially rescued the effects of miR156A overexpression on traits affecting root architecture, including meristem length and the rate of lateral root emergence. Consistent with this, PLETHORA 2 also inhibited the root-tip expression of another miR156 gene, miR156C. We conclude that the system driving phase change in the shoot affects developmental progression in the root, and that PLETHORA 2 participates in this network.


Asunto(s)
Proteínas de Arabidopsis , Arabidopsis , MicroARNs , Meristema/metabolismo , Proteínas de Arabidopsis/metabolismo , Regulación de la Expresión Génica de las Plantas/genética , Arabidopsis/metabolismo , Plantones/genética , MicroARNs/genética , MicroARNs/metabolismo
7.
J Surg Res ; 195(2): 396-405, 2015 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-25758338

RESUMEN

BACKGROUND: Vascular smooth muscle cell (VSMC) migration in response to urokinase is dependent on binding of the urokinase molecule to the urokinase plasminogen receptor (uPAR) and cleavage of the receptor. The aim of this study was to examine the role of the soluble uPAR (suPAR) in VSMC migration. METHODS: Human VSMCs were cultured in vitro. Linear wound and Boyden microchemotaxis assays of migration were performed in the presence of suPAR. Inhibitors to G-protein signaling and kinase activation were used to study these pathways. Assays were performed for mitogen-activated protein kinase and epidermal growth factor receptor activation. RESULTS: suPAR induced concentration-dependent migration of VSMC, which was G protein-dependent and was blocked by Gαi and Gßγ inhibitors. Removal of the full uPAR molecule by incubation of the cells with a phospholipase did not interfere with this response. suPAR induced ERK1/2, p38(MAPK), and c-Jun N-terminal kinase [JNK] activation in a Gαi/Gßγ-dependent manner, and interruption of these signaling pathways prevented suPAR-mediated migration. suPAR activity was independent of plasmin activity. suPAR did not activate epidermal growth factor receptor. Interruption of the low affinity N-formyl-Met-Leu-Phe receptor (FPRL1) but not high affinity N-formyl-Met-Leu-Phe receptor (FPR) prevented cell migration and activation in response to suPAR. suPAR increased matrix metalloproteinase-2 expression and activity, and this was dependent on the low affinity N-formyl-Met-Leu-Phe receptor (FPRL1) and ERK1/2. CONCLUSIONS: suPAR induces human smooth muscle cell activation and migration independent of the full uPAR through activation of the G protein-coupled receptor FPRL1, which is not linked to the plasminogen activation cascade.


Asunto(s)
Músculo Liso Vascular/citología , Miocitos del Músculo Liso/fisiología , Receptores de Formil Péptido/fisiología , Receptores del Activador de Plasminógeno Tipo Uroquinasa/fisiología , Movimiento Celular , Receptores ErbB/fisiología , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Humanos , Sistema de Señalización de MAP Quinasas , Metaloproteinasa 2 de la Matriz/fisiología
8.
J Surg Res ; 192(2): 254-62, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25082749

RESUMEN

BACKGROUND: Urokinase (uPA) modulates cellular and extracellular matrix responses within the microenvironment of the vessel wall and has been shown to activate the epidermal growth factor receptor (EGFR). This study examines the role of the protease domain of uPA during EGFR activation in human vascular smooth muscle cells (VSMC). METHODS: Human coronary VSMC were cultured in vitro. Assays of cell proliferation and EGFR phosphorylation were examined in response to the carboxyterminal fragment of uPA (CTF) in the presence and absence of the plasmin, metalloprotease and a disintegrin and metalloproteinase (ADAM) inhibitors, heparin-bound epidermal growth factor (HB-EGF), and EGFR inhibitors, and small interfering RNA to EGFR and ADAMs. RESULTS: CTF produced a dose-dependent increase in DNA synthesis and cell proliferation in human VSMC, which was blocked in a dose-dependent manner by both plasmin inhibitors and the EGFR inhibitor, AG1478. CTF induced time-dependent EGFR phosphorylation, which was blocked by inhibitors of plasmin and metalloproteinases activity. The presence of urokinase plasminogen activator receptor was not required. Inhibition of ADAM-10 and -12, and of HB-EGF blocked EGFR activation in response to CTF. CTF-mediated activation of EGFR was mediated through Gßγ, src, and NAD(P)H oxidase. CONCLUSIONS: In human coronary VSMC, uPA induces uPAR-independent, domain-dependent smooth muscle cell proliferation through transactivation of EGFR by a plasmin-mediated, ADAM-induced, and HB-EGF-dependent process, which is mediated by the intracellular pathways involving Gαi, Gßγ, src, and NAD(P)H oxidase.


Asunto(s)
Vasos Coronarios/citología , Receptores ErbB/metabolismo , Músculo Liso Vascular/citología , Músculo Liso Vascular/enzimología , Transducción de Señal/fisiología , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , Proteínas ADAM/metabolismo , Proteína ADAM10 , Proteína ADAM12 , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Inhibidores Enzimáticos/farmacología , Receptores ErbB/genética , Humanos , Proteínas de la Membrana/metabolismo , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/enzimología , Fosforilación/efectos de los fármacos , Fosforilación/fisiología , Estructura Terciaria de Proteína , Quinazolinas/farmacología , ARN Interferente Pequeño/genética , Receptores del Activador de Plasminógeno Tipo Uroquinasa/metabolismo , Transducción de Señal/efectos de los fármacos , Tirfostinos/farmacología , Activador de Plasminógeno de Tipo Uroquinasa/química
9.
J Surg Res ; 191(1): 33-41, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24972735

RESUMEN

BACKGROUND: Metabolic syndrome is now an epidemic in the United States population. Intimal hyperplasia remains the principal lesion in the development of restenosis after vessel wall injury. The aim of this study is to characterize the changes induced in wall morphology in the developing intimal hyperplasia within a murine model in the presence of diabetes (type 1) and metabolic syndrome. METHODS: Control (wild type B6), Non Obese Diabetic, and metabolic syndrome (RCS-10) mice were used. The murine femoral wire injury model was used in which a micro wire is passed through a branch of the femoral and used to denude the common femoral and iliac arteries. Specimens were perfusion fixed and sections were stained with hematoxylin and eosin and Movat stains such that dimensional and compositional morphometry could be performed using an ImagePro system. Additional stains for proliferation and apoptosis were used. RESULTS: In control mice, the injured femoral arteries develop intimal hyperplasia, which is maximal at 28 d and remains stable to day 56. Sham-operated vessels do not produce such a response. In diabetic mice, the intimal response increased 5-fold with a 2-fold increase in proteoglycan deposition, whereas in the metabolic syndrome mice there was a 6-fold increase in the intimal response and a similar increase in proteoglycan deposition. Collagen deposition was different with a 22-fold increase over control in collagen deposition in diabetes and a 100-fold increase over control in collagen deposition in metabolic syndrome as compared with the control injury mice. Maximal vascular smooth muscle cell (VSMC) proliferation was decreased in both diabetes and metabolic syndrome compared with controls, whereas early cell apoptosis in both diabetes and metabolic syndrome was sustained over a longer period of time compared with wild-type mice. CONCLUSIONS: These data demonstrate that development of intimal hyperplasia is markedly different in diabetes and metabolic syndrome compared with controls, with an increase in collagen deposition, a reduction in VSMC proliferation, and an increase in early VSMC apoptosis. These findings suggest that preventative strategies against restenosis must be tailored for the diabetic and metabolic syndrome patients.


Asunto(s)
Arteria Femoral/lesiones , Arteria Ilíaca/lesiones , Síndrome Metabólico/complicaciones , Síndrome Metabólico/metabolismo , Lesiones del Sistema Vascular/complicaciones , Lesiones del Sistema Vascular/metabolismo , Animales , Apoptosis , Glucemia/metabolismo , Proliferación Celular , Matriz Extracelular/metabolismo , Matriz Extracelular/patología , Femenino , Arteria Femoral/metabolismo , Arteria Femoral/patología , Hiperplasia/metabolismo , Hiperplasia/patología , Arteria Ilíaca/metabolismo , Arteria Ilíaca/patología , Lípidos/sangre , Masculino , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Túnica Íntima/lesiones , Túnica Íntima/metabolismo , Túnica Íntima/patología , Lesiones del Sistema Vascular/patología , Vasculitis/metabolismo , Vasculitis/patología
10.
J Surg Res ; 181(1): 32-40, 2013 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-22595017

RESUMEN

BACKGROUND: Vessels heal after injury and G protein-coupled receptors are involved in the vascular smooth muscle cell proliferation required to form intimal hyperplasia. We have previously identified the role of Gαq in vascular smooth muscle cell proliferation in vitro. This study now examines the role of Gαq in the developing intimal hyperplasia in a murine model and the impact of disruption of Gαq signaling on intimal hyperplasia development. METHODS: We employed a murine femoral wire injury model in which a micro-wire is passed through a branch of the femoral artery and used to denude the common femoral artery. We perfusion-fixed specimens and stained sections with hematoxylin-eosin and Movat's stains such that morphometric analysis could be performed using an Image-Pro system. We also harvested additional specimens of femoral artery and snap-froze them for Western blotting or zymography, to allow for the study of G protein expression and both protease expression and activity. We used contralateral vessels as controls. We immersed additional vessels in pluronic gel containing the chemical Gαq G protein inhibitors GP-2A, siRNA to Gαq or adenovirus containing mutant inactive Gαq. RESULTS: Gαq expression increased in a time-dependent manner after femoral artery injury. Sham-operated vessels did not produce such a response. Inhibition of Gαq reduced cell proliferation without affecting cell migration. Interruption of Gαq signaling also inhibited the development of intimal hyperplasia. Inhibition of Gαq did not alter peak urinary-type plasminogen activator activity and expression, but did increase early plasminogen activator inhibitor-1 activity and expression. Inhibition of Gαq reduced peak metalloproteinase (MMP)-9 activity at Day 3 but did not influence peak MMP-2 activity at Day 7. Protein expression for MMP-9 was also decreased, but that of MMP-2 was not affected. There were no changes in the expression or the activity of the respective inhibitors for MMP-9 and MMP-2, and tissue inhibitor of metalloproteinases-1 and -2. CONCLUSIONS: These data demonstrate that femoral wire injury in the mouse is associated with a time-dependent increase in Gαq expression. Inhibition of Gαq alters cell proliferation and is associated with decreased MMP-9 expression and activity.


Asunto(s)
Arteria Femoral/patología , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/fisiología , Metaloproteinasa 9 de la Matriz/metabolismo , Animales , Hiperplasia , Metaloproteinasa 2 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Inhibidor 1 de Activador Plasminogénico/análisis , Túnica Íntima/patología , Activador de Plasminógeno de Tipo Uroquinasa/análisis
11.
Surgery ; 152(5): 879-85, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22575880

RESUMEN

BACKGROUND: Cell migration is an integral part of the development of intimal hyperplasia, and proteases are pivotal components in the process. Cell migration in response to urokinase is mediated through the aminoterminal fragment (ATF) of the protein. This study examines the role of NAD(P)H oxidase during epidermal growth factor receptor (EGFR) transactivation by ATF in human vascular smooth muscle cells (VSMC). METHODS: Human VSMCs were cultured in vitro. Linear wound and Boyden microchemotaxis assays of migration in response to ATF were performed in the presence and absence of NAD(P)H oxidase inhibitors (diphenyleneiodonium [DPI] and apocynin) and small interfering RNA (siRNA) to Nox1. Additional assays were performed to examine the upstream pathways that lead to NAD(P)H oxidase activity. Assays were also performed for EGFR activation. RESULTS: ATF produced concentration-dependent VSMC migration, which was inhibited by increasing concentrations of DPI and apocynin. ATF was shown to induce time-dependent EGFR phosphorylation, which peaked at 4-fold greater than control. This response was inhibited by DPI and apocynin in a concentration-dependent manner. ATF induced a concentration-dependent increase in intracellular oxygen free radical species, which was mitigated by the presence of DPI and apocynin. Inhibition of Gßγ by ßARK(CT) reduced both NAD(P)H oxidase activity and EGFR activation. Inhibition of rac, which allows the NAD(P)H complex to assemble on the membrane, and inhibition of src, which induces assembly of the complex, both reduced ATF-dependent NAD(P)H oxidase activity and EGFR phosphorylation. siRNA to Nox1 prevented ATF-mediated EGFR activation and cell migration. CONCLUSION: ATF requires NAD(P)H oxidase activity through a Gßγ-, rac-, and src-mediated pathway to facilitate transactivation of EGFR and VSMC migration.


Asunto(s)
Receptores ErbB/metabolismo , Músculo Liso Vascular/enzimología , Miocitos del Músculo Liso/enzimología , NADPH Oxidasas/metabolismo , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , Movimiento Celular , Células Cultivadas , Humanos , Especies Reactivas de Oxígeno/metabolismo
12.
J Surg Res ; 177(2): e75-82, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22480845

RESUMEN

BACKGROUND: Sphingosine-1-phosphate (S-1-P) is a bioactive sphingolipid released from activated platelets that stimulates migration of vascular smooth muscle cells (VSMC) in vitro. S-1-P is associated with oxidized low-density lipoprotein (oxLDL) and is important in vessel remodeling. S-1-P will activate multiple G protein-coupled receptors (S-1-PR 1 to 5), which can regulate multiple cellular functions, including cell migration. The aim of this study is to examine the role of S-1-PR signaling during smooth muscle cell migration in response to S-1-P. METHODS: Human VSMCs were cultured in vitro. Expression of S-1-PR 1 to 5 was determined in conditions mirroring diabetes (40 mM glucose) and metabolic syndrome (25 mM glucose with 20 µM linoleic acid and 20 µM oleic acid). Linear wound and Boyden microchemotaxis assays of migration were performed in the presence of S-1-P with and without siRNA against S-1-PR 1 to 5. Assays were performed for activation of ERK1/2, p38(MAPK) and JNK. RESULTS: Human VSMCs express S-1-PR1, S-1-PR2, and S-1-PR3. There was no significant expression of S-1-PR4 and S-1-PR5. The expression of S-1-PR1 and S-1-PR3 is enhanced under high glucose conditions and metabolic syndrome conditions. Migration of VSMC in response to S-1-P is enhanced 2-fold by diabetes and 4-fold by metabolic syndrome. In diabetes, S-1-PR1 expression is enhanced, while S-1-PR2 and S-1-PR3 expression are both maintained. In metabolic syndrome, S-1-PR1 and 3 expressions are enhanced and that of S-1-PR2 is reduced. siRNA to S-1-PR1 results in a 2-fold reduction in S-1-P-mediated cell migration under all conditions. siRNA to S-1-PR2 enhanced cell migration only under normal conditions, while siRNA S-1-PR3 decreased migration in metabolic syndrome only. Down-regulation of S-1-PR1 reduced ERK1/2 activation in response to S-1-P, while that of S-1-PR2 had no effect under normal conditions. In diabetes, down-regulation of S-1-PR1 reduced activation of all three MAPKs. In metabolic syndrome, down-regulation of S-1-PR1 and S-1-PR3 reduced activation of all three MAPKs. CONCLUSION: S-1-PR 1, 2, and 3 regulate human VSMC migration and their expression level and function are modulated by conditions simulating diabetes and metabolic syndrome.


Asunto(s)
Diabetes Mellitus/metabolismo , Lisofosfolípidos/metabolismo , Síndrome Metabólico/metabolismo , Miocitos del Músculo Liso/fisiología , Receptores de Lisoesfingolípidos/metabolismo , Esfingosina/análogos & derivados , Ensayos de Migración Celular , Células Cultivadas , Humanos , Sistema de Señalización de MAP Quinasas , Músculo Liso Vascular/citología , Esfingosina/metabolismo
13.
Clin Immunol ; 143(1): 59-72, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22341910

RESUMEN

Catheters are implanted into the peritoneal cavity during the process of peritoneal dialysis. Though these catheters may be effective and beneficial, the impact of catheters on the immune system is poorly understood. Catheters and other devices implanted in the peritoneal cavity elicit a foreign body reaction. However, the immunological consequences of this remain uncharacterized. To model this, catheters were implanted into the peritoneal cavity of healthy mice. Catheter implantation induced rapid cellular changes within the peritoneal cavity. Whereas B-cells and T-cells were reduced, catheter implantation was associated with the rapid expansion of F4/80-low-positive, CD11b-positive macrophages that elaborated IL-10, and suppressed T-cell division and Th1 skewing in co-culture assays. Peritoneal catheter elicited macrophages had increased Jmjd3 but reduced NF-κB activation, and their emergence was MyD88-dependent. Collectively, these studies indicate that foreign body implantation into the peritoneal cavity is associated with the expansion of suppressor macrophages. Whether peritoneal cavity catheter implantation may have systemic immunoregulatory roles remains to be explored.


Asunto(s)
Interleucina-10/inmunología , Macrófagos Peritoneales/inmunología , Factor 88 de Diferenciación Mieloide/inmunología , Transducción de Señal/inmunología , Animales , Antígenos de Diferenciación/inmunología , Antígenos de Diferenciación/metabolismo , Linfocitos B/citología , Linfocitos B/inmunología , Linfocitos B/metabolismo , Antígeno CD11b/inmunología , Antígeno CD11b/metabolismo , Antígenos CD5/inmunología , Antígenos CD5/metabolismo , Catéteres de Permanencia , Recuento de Células , Citometría de Flujo , Reacción a Cuerpo Extraño/inmunología , Humanos , Interleucina-10/metabolismo , Histona Demetilasas con Dominio de Jumonji/inmunología , Histona Demetilasas con Dominio de Jumonji/metabolismo , Activación de Macrófagos/inmunología , Macrófagos Peritoneales/citología , Macrófagos Peritoneales/metabolismo , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Factor 88 de Diferenciación Mieloide/metabolismo , Cavidad Peritoneal/citología , Diálisis Peritoneal , Linfocitos T/citología , Linfocitos T/inmunología , Linfocitos T/metabolismo
14.
J Surg Res ; 178(1): 40-7, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21962816

RESUMEN

BACKGROUND: Intimal hyperplasia remains the principal lesion in the development of restenosis after vessel wall injury. G-protein coupled receptors are involved in smooth muscle cell proliferation but the role of Gßγ in arterial intimal hyperplasia has not been well defined. The aim of this study is to characterize the expression of Gßγ G-proteins in the developing intimal hyperplasia in a murine model and the impact of disruption of Gßγ signaling on intimal hyperplasia development. METHODS: The murine femoral wire injury model was employed. Specimens were perfusion-fixed and sections were stained with H&E and Movat's stains such that morphometry could be performed using an Image-Pro system. Additional specimens of femoral artery were also harvested and snap frozen for Western blotting for the Gßγ expression and for Western blotting and zymography to allow for the study of gelatinase and plasminogen activator expression and activation. Contralateral vessels were used as controls. Additional vessels were immersed in pluronic gel containing an adenovirus with the Gßγ inhibitor ßARK(CT). RESULTS: The injured femoral arteries developed intimal hyperplasia, while sham vessels did not produce such a response. Cell proliferation peaked at 3-5 d and cell migration at 7 d after injury. There was a marked time-dependent increase in Gßγ over the 28 d following injury. Inhibition of Gßγ with ßARK(CT) inhibited cell proliferation, cell migration and the development of intimal hyperplasia. Inhibition of Gßγ decreased peak uPA activity and expression without increasing early PAI-1 activity and expression. Inhibition of Gßγ reduced peak MMP-2 activity at d 1 but not at d 7 and also reduced peak MMP-9 activity at d 3. Protein expression for both MMP-2 and MMP-9 was also transiently decreased. There were no changes in TIMP-1 and TIMP-2 expression and activity. CONCLUSIONS: These data demonstrate a time-dependent increase in Gßγ G-protein expression following wire injury in the mouse. Inhibition of Gßγ alters cell proliferation and migration with associated changes in MMP-2, MMP-9, and uPA expression and activity.


Asunto(s)
Arteria Femoral/enzimología , Arteria Femoral/lesiones , Subunidades beta de la Proteína de Unión al GTP/metabolismo , Subunidades gamma de la Proteína de Unión al GTP/metabolismo , Péptido Hidrolasas/metabolismo , Animales , Apoptosis/fisiología , Modelos Animales de Enfermedad , Arteria Femoral/patología , Subunidades beta de la Proteína de Unión al GTP/antagonistas & inhibidores , Subunidades gamma de la Proteína de Unión al GTP/antagonistas & inhibidores , Hiperplasia/patología , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Músculo Liso Vascular/enzimología , Músculo Liso Vascular/lesiones , Músculo Liso Vascular/patología , Péptidos/farmacología , Proteínas Recombinantes/farmacología , Transducción de Señal/fisiología , Inhibidor Tisular de Metaloproteinasa-1/metabolismo , Inhibidor Tisular de Metaloproteinasa-2/metabolismo , Túnica Íntima/enzimología , Túnica Íntima/lesiones , Túnica Íntima/patología
15.
J Surg Res ; 175(1): 30-4, 2012 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-21920544

RESUMEN

BACKGROUND: Sphingosine-1-phosphate (S-1-P) is a bioactive sphingolipid released from activated platelets at sites of arterial injury that stimulates migration of smooth muscle cells (SMC). The kinase src is a significant focal point in transmembrane signaling. This study examines the role of src during smooth muscle cell migration in response to S-1-P. METHODS: Human coronary arterial SMCs were cultured in vitro. Boyden microchemotaxis assays of migration were performed in response to S-1-P in the presence and absence the src inhibitor (PP2, 10 µM) and a dominant negative src construct (DNsrc). siRNA to S-1-P receptors was used to down-regulate the S-1-P receptors. Western blotting was performed for src and MAPK phosphorylation. RESULTS: Inhibition of src with PP2 but not PP3 partially blocked S-1-P-mediated cell migration. S-1-P induced time-dependent activation of src, which was inhibited by PP2 and adenoviral DNsrc. PP3 or an empty vector had no effect. Activation of src by S-1-P was inhibited by siRNA to S-1-PR1 and S-1-PR3 but not by S-1-PR2. When the VSMC were transfected with adenovirus containing ßARK(CT), an inhibitor to Gßγ, src activation was significantly attenuated. Src inhibition with PP2 reduced p38(MAPK) and JNK activation but did not alter ERK1/2 activation. CONCLUSION: S-1-P mediated VSMC migration is modulated by a G-protein-coupled src pathway partially through src-mediated p38(MAPK) and JNK signaling and requires S-1-PR1 and S-1-PR3 receptors.


Asunto(s)
Movimiento Celular/fisiología , Lisofosfolípidos/metabolismo , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/metabolismo , Esfingosina/análogos & derivados , Familia-src Quinasas/metabolismo , Células Cultivadas , Vasos Coronarios/citología , Regulación hacia Abajo , Humanos , Transducción de Señal , Esfingosina/metabolismo
16.
J Clin Immunol ; 31(1): 128-35, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20886281

RESUMEN

Experimental autoimmune nephritis in mice and spontaneous lupus nephritis are both associated with elevated expression of several chemokines in the kidneys. Nevertheless, the role that different chemokines play in mediating renal inflammation is far from complete. This study focuses on elucidating the functional role of RANTES, a chemokine that has been noted to be hyper-expressed within the kidneys, both in experimental renal disease as well as in spontaneous lupus nephritis. To elucidate if RANTES was essential for immune-mediated glomerulonephritis, DBA/1 mice that are highly sensitive to nephrotoxic serum nephritis were rendered RANTES-deficient and then tested for disease susceptibility. Nephritis-sensitive DBA/1 mice expressed more RANTES within the diseased kidneys. Compared to wild-type DBA/1 mice, RANTES-deficient DBA/1 mice developed significantly less proteinuria, azotemia, and renal inflammation, with reduced crescent formation and tubulo-interstitial nephritis. These findings indicate that RANTES ablation attenuates immune-mediated nephritis and suggest that this chemokine could be a potential therapeutic target in these diseases.


Asunto(s)
Autoanticuerpos/inmunología , Quimiocina CCL5/deficiencia , Glomerulonefritis/inmunología , Glomerulonefritis/patología , Animales , Quimiocina CCL5/metabolismo , Femenino , Riñón/patología , Nefritis Lúpica/inmunología , Nefritis Lúpica/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos DBA , Proteinuria , Conejos
17.
J Immunol ; 184(4): 2183-93, 2010 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-20065116

RESUMEN

To identify potential biomarkers in immune-mediated nephritis, urine from mice subjected to an augmented passive model of anti-glomerular basement membrane (GBM)-induced experimental nephritis was resolved using two-dimensional gels. The urinary proteome in these diseased mice was comprised of at least 71 different proteins. Using orthogonal assays, several of these molecules, including serum amyloid P (SAP), PG D synthase, superoxide dismutase, renin, and total protease were validated to be elevated in the urine and kidneys of mice during anti-GBM disease, as well as in mice with spontaneously arising lupus nephritis. Among these, urinary protease was the only marker that appeared to be exclusively renal in origin, whereas the others were partly serum-derived. Longitudinal studies in murine lupus demonstrated that total urinary protease had better predictive value for histologically active nephritis (r = 0.78) compared with proteinuria (r = -0.04), azotemia (r = 0.28), or the other markers examined, whereas urine SAP emerged as the single most predictive marker of histological glomerulonephritis. Collectively, these studies uncover total urinary protease, PG D synthase, SAP, and superoxide dismutase as novel biomarkers of anti-GBM disease and lupus nephritis, with stronger correlation to renal disease compared with currently employed biomarkers. These findings could have important diagnostic and prognostic ramifications in the management of these renal diatheses.


Asunto(s)
Modelos Animales de Enfermedad , Oxidorreductasas Intramoleculares/orina , Lipocalinas/orina , Nefritis Lúpica/enzimología , Nefritis Lúpica/orina , Péptido Hidrolasas/orina , Proteoma/análisis , Componente Amiloide P Sérico/orina , Superóxido Dismutasa/orina , Secuencia de Aminoácidos , Animales , Enfermedad por Anticuerpos Antimembrana Basal Glomerular/enzimología , Enfermedad por Anticuerpos Antimembrana Basal Glomerular/inmunología , Enfermedad por Anticuerpos Antimembrana Basal Glomerular/orina , Autoanticuerpos/fisiología , Biomarcadores/orina , Femenino , Humanos , Oxidorreductasas Intramoleculares/inmunología , Riñón/enzimología , Riñón/inmunología , Riñón/patología , Lipocalinas/inmunología , Estudios Longitudinales , Nefritis Lúpica/inmunología , Ratones , Ratones Congénicos , Ratones Endogámicos C57BL , Ratones Endogámicos MRL lpr , Datos de Secuencia Molecular , Péptido Hidrolasas/inmunología , Valor Predictivo de las Pruebas , Proteoma/inmunología , Componente Amiloide P Sérico/inmunología , Superóxido Dismutasa/inmunología , Regulación hacia Arriba/inmunología
18.
J Immunol ; 183(10): 6831-8, 2009 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-19864599

RESUMEN

Increased Type I IFNs or IFN-I have been associated with human systemic lupus erythematosus. Interestingly augmenting or negating IFN-I activity in murine lupus not only modulates systemic autoimmunity, but also impacts lupus nephritis, suggesting that IFN-I may be acting at the level of the end-organ. We find resident renal cells to be a dominant source of IFN-I in an experimental model of autoantibody-induced nephritis. In this model, augmenting IFN-I amplified antibody-triggered nephritis, whereas ablating IFN-I activity ameliorated disease. One mechanism through which increased IFN-I drives immune-mediated nephritis might be operative through increased recruitment of inflammatory monocytes and neutrophils, though this hypothesis needs further validation. Collectively, these studies indicate that an important contribution of IFN-I toward the disease pathology seen in systemic autoimmunity may be exercised at the level of the end-organ.


Asunto(s)
Citocinas/inmunología , Glomerulonefritis/inmunología , Interferón Tipo I/inmunología , Fallo Renal Crónico/inmunología , Receptor de Interferón alfa y beta/inmunología , Animales , Antivirales/inmunología , Antivirales/farmacología , Autoanticuerpos/sangre , Autoanticuerpos/efectos de los fármacos , Citocinas/efectos de los fármacos , Citocinas/metabolismo , Modelos Animales de Enfermedad , Glomerulonefritis/metabolismo , Humanos , Interferón Tipo I/farmacología , Riñón/efectos de los fármacos , Riñón/inmunología , Riñón/patología , Fallo Renal Crónico/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/metabolismo , Proteínas Recombinantes
19.
Arch Immunol Ther Exp (Warsz) ; 56(1): 31-40, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18250969

RESUMEN

Systemic lupus erythematosus (SLE) is an autoimmune disease that results in immune-mediated damage to multiple organs. Among these, kidney involvement is the most common and fatal. Spontaneous lupus nephritis (SLN) in mouse models has provided valuable insights into the underlying mechanisms of human lupus nephritis. However, SLN in mouse models takes 6-12 months to manifest; hence there is clearly the need for a mouse model that can be used to unveil the pathogenic processes that lead to immune nephritis over a shorter time frame. In this article more than 25 different molecules are reviewed that have been studied both in the anti-glomerular basement membrane (anti-GBM) model and in SLN and it was found that these molecules influence both diseases in a parallel fashion, suggesting that the two disease settings share common molecular mechanisms. Based on these observations, the authors believe the experimental anti-GBM disease model might be one of the best tools currently available for uncovering the downstream molecular mechanisms leading to SLN.


Asunto(s)
Enfermedad por Anticuerpos Antimembrana Basal Glomerular , Modelos Animales de Enfermedad , Inmunidad Celular , Mediadores de Inflamación/metabolismo , Riñón/inmunología , Riñón/metabolismo , Riñón/patología , Nefritis Lúpica , Animales , Enfermedad por Anticuerpos Antimembrana Basal Glomerular/etiología , Enfermedad por Anticuerpos Antimembrana Basal Glomerular/metabolismo , Enfermedad por Anticuerpos Antimembrana Basal Glomerular/patología , Autoanticuerpos/biosíntesis , Autoanticuerpos/inmunología , Autoanticuerpos/metabolismo , Autoantígenos/metabolismo , Humanos , Riñón/citología , Nefritis Lúpica/etiología , Nefritis Lúpica/metabolismo , Nefritis Lúpica/patología , Ratones , Ratones Endogámicos MRL lpr , Ratones Endogámicos NZB , Ratas , Ratas Endogámicas WKY
20.
Clin Immunol ; 124(2): 109-18, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17640604

RESUMEN

Lupus nephritis is an immune-mediated disease, where antibodies and T cells both play pathogenic roles. Since spontaneous lupus nephritis in mouse models takes 6-12 months to manifest, there is an urgent need for a mouse model that can be used to delineate the pathogenic processes that lead to immune nephritis, over a quicker time frame. We propose that the experimental anti-glomerular basement membrane (GBM) disease model might be a suitable tool for uncovering some of the molecular steps underlying lupus nephritis. This article reviews the current evidence that supports the use of the experimental anti-GBM nephritis model for studying spontaneous lupus nephritis. Importantly, out of about 25 different molecules that have been specifically examined in the experimental anti-GBM model and also spontaneous lupus nephritis, all influence both diseases concordantly, suggesting that the experimental model might be a useful tool for unraveling the molecular basis of spontaneous lupus nephritis. This has important clinical implications, both from the perspective of genetic susceptibility as well as clinical therapeutics.


Asunto(s)
Enfermedad por Anticuerpos Antimembrana Basal Glomerular/inmunología , Nefritis Lúpica/inmunología , Animales , Enfermedad por Anticuerpos Antimembrana Basal Glomerular/genética , Modelos Animales de Enfermedad , Humanos , Nefritis Lúpica/genética , Ratones , Ratas , Linfocitos T/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA