Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 10(1): 2085, 2020 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-32034241

RESUMEN

The purpose of this study is to evaluate the efficiency of local platelet-rich plasma (PRP) injection as an adjuvant treatment after carpal ligament release. We conducted a prospective randomized, triple-blinded, controlled trial. Fifty participants with mild to extreme carpal tunnel syndrome (CTS) were randomly assigned either to the PRP (n = 25) or the platelet-poor plasma (PPP, n = 25) group. After performing open surgical release of the carpal ligament, the inside of the carpal tunnel was irrigated with 3 mL of PRP or PPP according to each participant's group allocation. The primary outcome was hand grip strength (HGS). Secondary outcomes were the time taken off work after surgery (in days) and scores on the Wong-Baker Faces Scale, Boston Carpal Tunnel Questionnaire, and Southampton Wound Assessment Scale. We evaluated patients before treatment and at 6-weeks. As expected, the pain levels, symptom severity, and functional status improved in all the patients after surgery. However, intragroup analysis revealed that only the participants in the PRP group had regained their pre-operative HGS levels at 6-weeks follow-up. These findings indicate that PRP is an effective adjuvant treatment in patients with mild to severe CTS who require surgery.


Asunto(s)
Síndrome del Túnel Carpiano/cirugía , Ligamentos Articulares/cirugía , Plasma Rico en Plaquetas , Síndrome del Túnel Carpiano/terapia , Método Doble Ciego , Femenino , Fuerza de la Mano , Humanos , Masculino , Persona de Mediana Edad , Plasma Rico en Plaquetas/fisiología , Reinserción al Trabajo , Resultado del Tratamiento
2.
J Mol Cell Cardiol ; 132: 154-163, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31121182

RESUMEN

The CC chemokine 1 (CCL1, also called I-309 or TCA3) is a potent chemoattractant for leukocytes that plays an important role in inflammatory processes and diseases through binding to its receptor CCR8. Here, we investigated the role of the CCL1-CCR8 axis in atherosclerosis. We found increased expression of CCL1 in the aortas of atherosclerosis-prone fat-fed apolipoprotein E (Apoe)-null mice; moreover, in vitro flow chamber assays and in vivo intravital microscopy demonstrated an essential role for CCL1 in leukocyte recruitment. Mice doubly deficient for CCL1 and Apoe exhibited enhanced atherosclerosis in aorta, which was associated with reduced plasma levels of the anti-inflammatory interleukin 10, an increased splenocyte Th1/Th2 ratio, and a reduced regulatory T cell (Treg) content in aorta and spleen. Reduced Treg recruitment and aggravated atherosclerosis were also detected in the aortas of fat-fed low-density lipoprotein receptor-null mice treated with CCR8 blocking antibodies. These findings demonstrate that disruption of the CCL1-CCR8 axis promotes atherosclerosis by inhibiting interleukin 10 production and Treg recruitment and function.


Asunto(s)
Aterosclerosis/inmunología , Quimiocina CCL1/inmunología , Receptores CCR8/inmunología , Linfocitos T Reguladores/inmunología , Animales , Apolipoproteínas E/inmunología , Citocinas/inmunología , Inflamación/inmunología , Interleucina-10/inmunología , Leucocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células TH1/inmunología , Células Th2/inmunología
3.
Proc Natl Acad Sci U S A ; 113(47): 13516-13521, 2016 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-27834218

RESUMEN

The circadian clock and cell cycle networks are interlocked on the molecular level, with the core clock loop exerting a multilevel regulatory role over cell cycle components. This is particularly relevant to the circadian factor Period 2 (Per2), which modulates the stability of the tumor suppressor p53 in unstressed cells and transcriptional activity in response to genotoxic stress. Per2 binding prevents Mdm2-mediated ubiquitination of p53 and, therefore, its degradation, and oscillations in the peaks of Per2 and p53 were expected to correspond. However, our findings showed that Per2 and p53 rhythms were significantly out-of-phase relative to each other in cell lysates and in purified cytoplasmic fractions. These seemingly conflicting experimental data motivated the use of a combined theoretical and experimental approach focusing on the role played by Per2 in dictating the phase of p53 oscillations. Systematic modeling of all possible regulatory scenarios predicted that the observed phase relationship between Per2 and p53 could be simulated if (i) p53 was more stable in the nucleus than in the cytoplasm, (ii) Per2 associates to various ubiquitinated forms of p53, and (iii) Per2 mediated p53 nuclear import. These predictions were supported by a sevenfold increase in p53's half-life in the nucleus and by in vitro binding of Per2 to the various ubiquitinated forms of p53. Last, p53's nuclear shuttling was significantly favored by ectopic expression of Per2 and reduced because of Per2 down-regulation. Our combined theoretical/mathematical approach reveals how clock regulatory nodes can be inferred from oscillating time course data.


Asunto(s)
Relojes Circadianos , Modelos Biológicos , Proteínas Circadianas Period/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Núcleo Celular/metabolismo , Relojes Circadianos/genética , Simulación por Computador , Regulación de la Expresión Génica , Células HCT116 , Semivida , Humanos , Cinética , Transporte de Proteínas , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Fracciones Subcelulares/metabolismo , Factores de Tiempo , Proteínas Ubiquitinadas/metabolismo , Ubiquitinación
4.
J Control Release ; 238: 31-42, 2016 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-27444816

RESUMEN

Stealth pH-responsive liposomes for the delivery of therapeutic proteins to the bladder epithelium were prepared using methoxy-poly(ethylene glycol)5kDa-1,2-distearoyl-sn-glycero-3-phosphoethanolamine (mPEG5kDa-DSPE) and stearoyl-poly(ethylene glycol)-poly(methacryloyl sulfadimethoxine) copolymer (stearoyl-PEG-polySDM), which possesses an apparent pKa of 7.2. Liposomes of 0.2:0.6:100, 0.5:1.5:100 and 1:3:100 mPEG5kDa-DSPE/stearoyl-PEG-polySDM/(soybean phosphatidylcholine+cholesterol) molar ratios were loaded with bovine serum albumin (BSA) as a protein model. The loading capacity was 1.3% w/w BSA/lipid. At pH7.4, all liposome formulations displayed a negative zeta-potential and were stable for several days. By pH decrease or addition to mouse urine, the zeta potential strongly decreased, and the liposomes underwent a rapid size increase and aggregation. Photon correlation spectroscopy (PCS) and transmission electron microscopy (TEM) analyses showed that the extent of the aggregation depended on the stearoyl-PEG-polySDM/lipid molar ratio. Cytofluorimetric analysis and confocal microscopy showed that at pH6.5, the incubation of MB49 mouse bladder cancer cells and macrophages with fluorescein isothiocyanate-labelled-BSA (FITC-BSA) loaded and N-(Lissamine Rhodamine B sulfonyl)-1, 2-dihexadecanoyl-sn-glycero-3-phosphoethanolamine triethylammonium salt (rhodamine-DHPE) labelled 1:3:100 mPEG5kDa-DSPE/stearoyl-PEG-polySDM/lipid molar ratio liposomes resulted in a time-dependent liposome association with the cells. At pH7.4, the association of BSA-loaded liposomes with the MB49 cells and macrophages was remarkably lower than at pH6.5. Confocal images of bladder sections revealed that 2h after the instillation, liposomes at pH7.4 and control non-responsive liposomes at pH7.4 or 6.5 did not associate nor delivered FITC-BSA to the bladder epithelium. On the contrary, the pH-responsive liposome formulation set at pH6.5 and soon administered to mice by bladder instillation showed that, 2h after administration, the pH-responsive liposomes efficiently delivered the loaded FITC-BSA to the bladder epithelium.


Asunto(s)
Antineoplásicos/administración & dosificación , Preparaciones de Acción Retardada/metabolismo , Fluoresceína-5-Isotiocianato/análogos & derivados , Liposomas/metabolismo , Polietilenglicoles/metabolismo , Ácidos Polimetacrílicos/metabolismo , Albúmina Sérica Bovina/administración & dosificación , Sulfonamidas/metabolismo , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Animales , Línea Celular Tumoral , Epitelio/metabolismo , Femenino , Fluoresceína-5-Isotiocianato/administración & dosificación , Concentración de Iones de Hidrógeno , Ratones , Ratones Endogámicos C57BL , Vejiga Urinaria/metabolismo , Neoplasias de la Vejiga Urinaria/metabolismo
5.
Mol Biol Cell ; 26(2): 359-72, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25411341

RESUMEN

Circadian period proteins influence cell division and death by associating with checkpoint components, although their mode of regulation has not been firmly established. hPer2 forms a trimeric complex with hp53 and its negative regulator Mdm2. In unstressed cells, this association leads to increased hp53 stability by blocking Mdm2-dependent ubiquitination and transcription of hp53 target genes. Because of the relevance of hp53 in checkpoint signaling, we hypothesize that hPer2 association with hp53 acts as a regulatory module that influences hp53's downstream response to genotoxic stress. Unlike the trimeric complex, whose distribution was confined to the nuclear compartment, hPer2/hp53 was identified in both cytosol and nucleus. At the transcriptional level, a reporter containing the hp21(WAF1/CIP1) promoter, a target of hp53, remained inactive in cells expressing a stable form of the hPer2/hp53 complex even when treated with γ-radiation. Finally, we established that hPer2 directly acts on the hp53 node, as checkpoint components upstream of hp53 remained active in response to DNA damage. Quantitative transcriptional analyses of hp53 target genes demonstrated that unbound hp53 was absolutely required for activation of the DNA-damage response. Our results provide evidence of the mode by which the circadian tumor suppressor hPer2 modulates hp53 signaling in response to genotoxic stress.


Asunto(s)
Daño del ADN , Regulación Neoplásica de la Expresión Génica , Proteínas Circadianas Period/genética , Transducción de Señal/genética , Proteína p53 Supresora de Tumor/genética , Línea Celular Tumoral , Núcleo Celular/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Citosol/metabolismo , Células HCT116 , Humanos , Immunoblotting , Microscopía Fluorescente , Modelos Genéticos , Proteínas Circadianas Period/metabolismo , Unión Proteica , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína p53 Supresora de Tumor/metabolismo
6.
J Immunol ; 193(11): 5584-94, 2014 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-25339679

RESUMEN

BAFF is a crucial cytokine that affects the activity of both innate and adaptive immune cells. It promotes the expansion of Th17 cells in autoimmune disorders. With this study, we investigated the BAFF/Th17 responses in Helicobacter pylori-induced gastritis in humans. Our results show that the mucosa from Helicobacter(+) patients with chronic gastritis is enriched in IL-17 and BAFF, whereas the two cytokines are weakly expressed in Helicobacter(-) patients with chronic gastritis; moreover, the expression of both BAFF and IL-17 decreases after bacteria eradication. We demonstrate that BAFF accumulates in macrophages in vivo and that it is produced by monocyte-derived macrophages in vitro, after Helicobacter stimulation. Application of BAFF on monocytes triggers the accumulation of reactive oxygen species that are crucial for the release of pro-Th17 cytokines, such as IL-23, IL-1ß, and TGF-ß. Moreover, BAFF directly promotes the differentiation of Th17 cells. In conclusion, our results support the notion that an axis BAFF/Th17 exists in chronic gastritis of Helicobacter(+) patients and that its presence strictly depends on the bacterium. Moreover, we demonstrated that BAFF is able to drive Th17 responses both indirectly, by creating a pro-Th17 cytokine milieu through the involvement of innate immune cells, and directly, via the differentiation of T cells toward the specific profile. The results obtained in this study are of great interest for Helicobacter-related diseases and the development of novel therapeutic strategies based on the inhibition of the BAFF/IL-17 response.


Asunto(s)
Factor Activador de Células B/metabolismo , Gastritis/inmunología , Infecciones por Helicobacter/inmunología , Helicobacter pylori/inmunología , Macrófagos/inmunología , Membrana Mucosa/inmunología , Células Th17/inmunología , Inmunidad Adaptativa , Diferenciación Celular , Células Cultivadas , Enfermedad Crónica , Gastritis/etiología , Infecciones por Helicobacter/complicaciones , Humanos , Inmunidad Innata , Interleucina-17/metabolismo , Macrófagos/microbiología , Membrana Mucosa/microbiología , Especies Reactivas de Oxígeno/metabolismo
7.
Eur J Pharm Biopharm ; 88(3): 670-82, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25157908

RESUMEN

Novel, acid-sensitive liposomes that respond to physiopathological pH for tumour targeting applications were obtained by surface decoration with 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[methoxy(polyethylene glycol)] (mPEG-DSPE) and stearoyl-poly(ethylene glycol)-poly(methacryloyl sulfadimethoxine) copolymer (stearoyl-PEG-polySDM). The pH-sensitive stearoyl-PEG-polySDM copolymer contained an average of seven methacryloyl sulfadimethoxines per molecule and was found to possess an apparent pKa of 7.2. Preliminary cloud point studies showed that the hydrophilic/hydrophobic copolymer conversion occurred at pH 7.0. The copolymer was soluble above pH 7.0 and underwent aggregation at lower pH. Liposome formulations were prepared with 0.2:0.6:100, 0.5:1.5:100 and 1:3:100 mPEG-DSPE/stearoyl-PEG-polySDM/lipids molar ratios. All of the liposome formulations were stable at pH 7.4, even in the presence of foetal bovine serum, but they underwent rapid size increase at pH 6.5. TEM analysis showed that, at pH 6.5, the formulations coated with a stearoyl-PEG-polySDM/lipids molar ratio greater than 1:100 underwent aggregation. At pH 7.4, the liposomes showed negative zeta potential that significantly decreased after incubation at pH 6.5. Cell-culture studies indicated that the liposomes were not toxic up to 10mg/mL. Fluorescence spectroscopy, cytofluorimetry and confocal microscopy showed that at pH 6.5, the incubation of MCF-7 tumour cells with fluorescein-labelled 1:3:100 mPEG-DSPE/stearoyl-PEG-polySDM/lipids molar ratio liposomes resulted in time-dependent cell association, while at pH 7.4 the cell interaction was significantly lower. The same pH-responsive liposome formulation loaded with gemcitabine (98.2±4.7nmol gemcitabine/lipid µmol loading capacity) was stable at pH 7.4 for several hours, while at pH 6.5 it rapidly aggregated. At pH 6.5, these liposomes displayed higher cytotoxicity than at pH 7.4 or compared to non-responsive control liposomes at both incubation pH. Notably, treatment with free gemcitabine did not yield cytotoxic effects, indicating that the carrier can efficiently deliver the anticancer drug to the cytosolic compartment.


Asunto(s)
Antimetabolitos Antineoplásicos/administración & dosificación , Desoxicitidina/análogos & derivados , Portadores de Fármacos/química , Polietilenglicoles/química , Ácidos Polimetacrílicos/química , Sulfonamidas/química , Antimetabolitos Antineoplásicos/farmacología , Técnicas de Cultivo de Célula , Supervivencia Celular/efectos de los fármacos , Desoxicitidina/administración & dosificación , Desoxicitidina/farmacología , Portadores de Fármacos/síntesis química , Estabilidad de Medicamentos , Humanos , Concentración de Iones de Hidrógeno , Liposomas , Células MCF-7 , Microscopía Confocal , Microscopía Electrónica de Transmisión , Estructura Molecular , Tamaño de la Partícula , Polietilenglicoles/síntesis química , Ácidos Polimetacrílicos/síntesis química , Espectrometría de Fluorescencia , Sulfonamidas/síntesis química , Propiedades de Superficie , Gemcitabina
8.
Mol Biol Cell ; 25(19): 3081-93, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25103245

RESUMEN

Human Period 2 (hPer2) is a transcriptional regulator at the core of the circadian clock mechanism that is responsible for generating the negative feedback loop that sustains the clock. Its relevance to human disease is underlined by alterations in its function that affect numerous biochemical and physiological processes. When absent, it results in the development of various cancers and an increase in the cell's susceptibility to genotoxic stress. Thus we sought to define a yet-uncharacterized checkpoint node in which circadian components integrate environmental stress signals to the DNA-damage response. We found that hPer2 binds the C-terminal half of human p53 (hp53) and forms a stable trimeric complex with hp53's negative regulator, Mdm2. We determined that hPer2 binding to hp53 prevents Mdm2 from being ubiquitinated and targeting hp53 by the proteasome. Down-regulation of hPer2 expression directly affects hp53 levels, whereas its overexpression influences both hp53 protein stability and transcription of targeted genes. Overall our findings place hPer2 directly at the heart of the hp53-mediated response by ensuring that basal levels of hp53 are available to precondition the cell when a rapid, hp53-mediated, transcriptional response is needed.


Asunto(s)
Relojes Circadianos/genética , Proteínas Circadianas Period/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Activación Transcripcional/genética , Proteína p53 Supresora de Tumor/metabolismo , Proteínas 14-3-3/genética , Biomarcadores de Tumor/genética , Proteínas de Ciclo Celular/genética , Ritmo Circadiano/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Daño del ADN , Reparación del ADN , Exorribonucleasas/genética , Regulación de la Expresión Génica , Células HCT116 , Humanos , Complejos Multiproteicos/metabolismo , Proteínas Nucleares/genética , Proteínas Circadianas Period/biosíntesis , Unión Proteica , Transcripción Genética , Proteína p53 Supresora de Tumor/biosíntesis , Proteína p53 Supresora de Tumor/genética , Ubiquitinación
9.
Cardiovasc Res ; 94(1): 38-47, 2012 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-22253285

RESUMEN

AIMS: Mitofusin-2 (Mfn2) expression is dysregulated in vascular proliferative disorders and its overexpression attenuates the proliferation of vascular smooth muscle cells (VSMCs) and neointimal lesion development after balloon angioplasty. We sought to gain insight into the mechanisms that control Mfn2 expression in VSMCs. METHODS AND RESULTS: We cloned and characterized 2 kb of the 5'-flanking region of the human Mfn2 gene. Its TATA-less promoter contains a CpG island. In keeping with this, 5'-rapid amplification of cDNA ends revealed six transcriptional start sites (TSSs), of which TSS2 and TSS5 were the most frequently used. The strong CpG island was found to be non-methylated under conditions characterized by large differences in Mfn2 gene expression. The proximal Mfn2 promoter contains six putative Sp1 motifs. Sp1 binds to the Mfn2 promoter and its overexpression activates the Mfn2 promoter in VSMCs. Chemical inhibition of Sp1 reduced Mfn2 expression, and Sp1 silencing reduced transcriptional activity of the Mfn2 promoter. In keeping with this view, Sp1 and Mfn2 mRNA levels were down-regulated in the aorta early after an atherogenic diet in apolipoprotein E-knockout mice or in VSMCs cultured in the presence of low serum. CONCLUSION: Sp1 is a key factor in maintaining basal Mfn2 transcription in VSMCs. Given the anti-proliferative actions of Mfn2, Sp1-induced Mfn2 transcription may represent a mechanism for prevention of VSMC proliferation and neointimal lesion and development.


Asunto(s)
GTP Fosfohidrolasas/genética , Proteínas Mitocondriales/genética , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Regiones Promotoras Genéticas , Factor de Transcripción Sp1/metabolismo , Activación Transcripcional , Animales , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Aterosclerosis/metabolismo , Aterosclerosis/patología , Aterosclerosis/prevención & control , Secuencia de Bases , Sitios de Unión , Línea Celular , Islas de CpG , Metilación de ADN , Modelos Animales de Enfermedad , GTP Fosfohidrolasas/metabolismo , Regulación de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Mitocondriales/metabolismo , Datos de Secuencia Molecular , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/patología , Neointima/metabolismo , Neointima/patología , Neointima/prevención & control , Interferencia de ARN , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Factor de Transcripción Sp1/antagonistas & inhibidores , Factor de Transcripción Sp1/genética , Sitio de Iniciación de la Transcripción , Activación Transcripcional/efectos de los fármacos , Transfección
10.
Arterioscler Thromb Vasc Biol ; 28(12): 2187-94, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18802016

RESUMEN

OBJECTIVE: The mechanisms underlying accelerated atherosclerosis in metabolic syndrome (MetS) patients remain poorly defined. In the mouse, complete disruption of insulin receptor substrate-2 (Irs2) causes insulin resistance, MetS-like manifestations, and accelerates atherosclerosis. Here, we performed human, mouse, and cell culture studies to gain insight into the contribution of defective Irs2 signaling to MetS-associated alterations. METHODS AND RESULTS: In circulating leukocytes from insulin-resistant MetS patients, Irs2 and Akt2 mRNA levels inversely correlate with plasma insulin levels and HOMA index and are reduced compared to insulin-sensitive MetS patients. Notably, a moderate reduction in Irs2 expression in fat-fed apolipoprotein E-null mice lacking one allele of Irs2 (apoE(-/-)Irs2(+/-)) accelerates atherosclerosis compared to apoE-null controls, without affecting plaque composition. Partial Irs2 inactivation also increases CD36 and SRA scavenger receptor expression and modified LDL uptake in macrophages, diminishes Akt2 and Ras expression in aorta, and enhances expression of the proatherogenic cytokine MCP1 in aorta and primary vascular smooth muscle cells (VSMCs) and macrophages. Inhibition of AKT or ERK1/2, a downstream target of RAS, upregulates Mcp1 in VSMCs. CONCLUSIONS: Enhanced levels of MCP1 resulting from reduced IRS2 expression and accompanying defects in AKT2 and Ras/ERK1/2 signaling pathways may contribute to accelerated atherosclerosis in MetS states.


Asunto(s)
Aterosclerosis/etiología , Aterosclerosis/metabolismo , Proteínas Sustrato del Receptor de Insulina/deficiencia , Proteínas Sustrato del Receptor de Insulina/genética , Síndrome Metabólico/complicaciones , Síndrome Metabólico/metabolismo , Animales , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Aterosclerosis/genética , Femenino , Humanos , Insulina/sangre , Resistencia a la Insulina/genética , Leucocitos/metabolismo , Sistema de Señalización de MAP Quinasas , Masculino , Síndrome Metabólico/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Proto-Oncogénicas c-akt/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal
11.
Cardiovasc Res ; 76(2): 340-50, 2007 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-17673191

RESUMEN

OBJECTIVE: Mounting evidence suggests that activation of complement, an important constituent of innate immunity, contributes to atherosclerosis. Here we investigated the expression of complement components (CCs) in the setting of experimental and clinical hypercholesterolemia, a major risk factor for atherosclerosis, their effects on vascular smooth muscle cell (VSMC) and macrophage proliferation, and the underlying molecular mechanisms. METHODS: For this study we analyzed the mRNA and protein expression of several CCs in plasma and aorta of hypercholesterolemic atherosclerosis-prone apolipoprotein E-null mice (apoE-KO) and in plasma of normocholesterolemic subjects and familial hypercholesterolemia (FH) patients. We also carried out in vitro molecular studies to assess the role of CCs on the control of macrophage and VSMC proliferation. RESULTS: Fat-fed apoE-KO mice experiencing severe hypercholesterolemia (approximately 400 mg/dL), but not fat-fed wild-type controls with plasma cholesterol level<110 mg/dL, displayed in aortic tissue upregulation of several CC mRNAs, including C3, C4, C1s, and C1q. In apoE-KO mice, induction of C3 mRNA was already apparent two days after fat feeding when hypercholesterolemia was manifested yet atherosclerotic lesions were absent or incipient. Rapid C3 and C4 protein upregulation was also observed in the plasma of fat-fed apoE-KO mice, and FH patients exhibited higher plasmatic C3a, C4 gamma chain, C1s and C3c alpha chain protein levels than normocholesterolemic subjects. In vitro, C3 and C3a, but not C3a-desArg, C4 and C1q, promoted macrophage and VSMC proliferation through Gi protein-dependent activation of extracellular signal-regulated kinase 1/2 (ERK1/2). We also found that C3-enriched FH plasma evoked a stronger mitogenic response in macrophages than normocholesterolemic plasma, and treatment with anti-C3 antibodies eliminated this difference. CONCLUSIONS: Both experimental and clinical hypercholesterolemia coincides with a concerted activation of several CCs. However, only C3 and C3a elicited a mitogenic response in cultured VSMCs and macrophages through Gi protein-dependent ERK1/2 activation. Thus, excess of C3/C3a in hypercholesterolemic apoE-KO mice and FH patients may contribute to atheroma growth by promoting neointimal cell proliferation.


Asunto(s)
Proteínas del Sistema Complemento/fisiología , Hipercolesterolemia/etiología , Macrófagos/citología , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/fisiología , Secuencia de Aminoácidos , Animales , Apolipoproteínas E/fisiología , Proliferación Celular , Complemento C3/fisiología , Complemento C3a/fisiología , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/fisiología , Humanos , Masculino , Ratones , Ratones Noqueados , Datos de Secuencia Molecular
12.
Front Biosci ; 12: 2291-8, 2007 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-17127239

RESUMEN

Atherosclerosis is increased in type 2 diabetic patients but the precise mechanisms underlying this predisposition remain vague. Mice deficient for insulin receptor substrate 2 (IRS2) develop type 2-like diabetes and thus, provide a model to explore the molecular connection between deranged carbohydrate metabolism and atherosclerosis. To explore the relationship between defective insulin signalling and atherosclerosis, we have examined the development of atherosclerosis in the following groups of fat-fed mice: wild-type, diabetic Irs2-null (Irs2-/-), atherosclerosis-prone apolipoprotein E-null (apoE-/-), and doubly-deficient apoE-/- Irs2-/-. Surprisingly, glucose levels of apoE-/- Irs2-/- mice were comparable to those seen in wild-type and apoE-/- and significantly lower than in Irs2-/- mice. Irs2-/- and apoE-/- Irs2-/- were hyperinsulinemic compared to wild-type and apoE-/- mice. Atherosclerotic lesions were barely detectable in wild-type and Irs2-/- mice, which displayed moderate hypercholesterolemia (approximately 280 mg/dL). Notably, atherosclerosis was significantly enhanced in apoE-/- Irs2-/- compared with apoE-/- mice, although both models displayed similar levels of severe hypercholesterolemia (>600 mg/dL). Circulating insulin levels predicted atherosclerotic lesion burden in apoE-/- Irs2-/- mice. Our results suggest that hyperinsulinemia as a result of Irs2 genetic ablation contributes to increased atherosclerosis when combined with severe hypercholesterolemia in the absence of hyperglycaemia (apoE-/- Irs2-/- mice), thus implicating IRS2 as an important modulator of murine hypercholesterolemia-dependent atherosclerosis. Future studies are necessary to determine whether IRS2 dysfunction may promote atherosclerosis in normoglycemic, pre-diabetic patients with clinical manifestations of hyperinsulinemia and insulin resistance.


Asunto(s)
Aterosclerosis/etiología , Diabetes Mellitus Tipo 2/complicaciones , Angiopatías Diabéticas/etiología , Hipercolesterolemia/complicaciones , Insulina/sangre , Péptidos y Proteínas de Señalización Intracelular/genética , Fosfoproteínas/genética , Animales , Apolipoproteínas E/genética , Aterosclerosis/patología , Glucemia/análisis , Angiopatías Diabéticas/sangre , Femenino , Hipercolesterolemia/sangre , Proteínas Sustrato del Receptor de Insulina , Péptidos y Proteínas de Señalización Intracelular/deficiencia , Lípidos/sangre , Macrófagos/citología , Masculino , Ratones , Ratones Noqueados , Músculo Liso Vascular/citología , Fosfoproteínas/deficiencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...