Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
1.
FASEB J ; 34(1): 1231-1246, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31914695

RESUMEN

Endothelial progenitor cells (EPCs) promote neovascularization and tissue repair by migrating to vascular injury sites; therefore, factors that enhance EPC homing to damaged tissues are of interest. Here, we provide evidence of the prominent role of the Netrin-4 (NTN4)-Unc-5 Netrin receptor B (UNC5B) axis in EPC-specific promotion of ischemic neovascularization. Our results showed that NTN4 promoted the proliferation, chemotactic migration, and paracrine effects of small EPCs (SEPCs) and significantly increased the incorporation of large EPCs (LEPCs) into tubule networks. Additionally, NTN4 prominently augmented neovascularization in mice with hindlimb ischemia by increasing the homing of exogenously transplanted EPCs to the ischemic limb and incorporating EPCs into vessels. Moreover, silencing of UNC5B, an NTN4 receptor, abrogated the NTN4-induced cellular activities of SEPCs in vitro and blood-flow recovery and neovascularization in vivo in ischemic muscle by reducing EPC homing and incorporation. These findings suggest NTN4 as an EPC-based therapy for treating angiogenesis-dependent diseases.


Asunto(s)
Células Progenitoras Endoteliales/metabolismo , Isquemia/metabolismo , Músculo Esquelético/metabolismo , Neovascularización Patológica/metabolismo , Receptores de Netrina/metabolismo , Netrinas/metabolismo , Animales , Células Progenitoras Endoteliales/patología , Células Progenitoras Endoteliales/trasplante , Silenciador del Gen , Xenoinjertos , Miembro Posterior/irrigación sanguínea , Humanos , Isquemia/genética , Isquemia/patología , Isquemia/terapia , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Músculo Esquelético/irrigación sanguínea , Músculo Esquelético/patología , Neovascularización Patológica/genética , Neovascularización Patológica/patología , Neovascularización Patológica/terapia , Receptores de Netrina/genética , Netrinas/genética
2.
J Environ Manage ; 269: 110739, 2020 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-32560984

RESUMEN

Since the Chernobyl and the Fukushima Daiichi disasters, contamination caused by radioactive accidents has attracted increasing attention. The present study evaluated immediate cleanup of 137Cs dissolved in surface water reservoir using an illite adsorbent, simulating an event of 137Cs contamination at Lake Paldang, South Korea. The study was conducted in two parts: (1) calculation of the residence time (tr) of illite adsorbent, and (2) evaluation of the adsorption time (ta) of illite adsorbent. tr was calculated based on physical properties (e.g., density, diameter, shape, and roughness) of the illite adsorbent at designated depth of surface water. Subsequently, ta was measured for 4 illite adsorbents (Korea01-Illite, Korea02-Illite, USA-Illite, and China-Illite) at 100 and 100,000 µg/L Cs, via kinetic adsorption experiment. Upon spraying of illite adsorbents with 50-150 µm diameter to locations where lake depth was between 6.5 m and 25.5 m, tr ranged from 0.132 to 3.300 h ta of 4 illite adsorbents was shorter than 0.6 and 2.5 h, for respective tests using 100 and 100,000 µg/L Cs. Based on the two characteristic times (tr and ta), the optimal particle diameter for the 4 illite adsorbents were evaluated at available lake depths in Lake Paldang. The study revealed that the USA-Illite is the efficient adsorbent at 100 µg/L Cs; in contrast, China-Illite could serve as the effective adsorbent at 100,000 µg/L Cs. Also, it was suggested that adsorbent efficiency had seasonal variations; tr was 2 h longer in winter than summer. In general, the study suggests that in the event of 137Cs contamination at Lake Paldang, Korea01-Illite is likely the best adsorbent to remove 137Cs due to its removal efficiency and accessibility from the illite deposit in Korea.


Asunto(s)
Lagos , Adsorción , Radioisótopos de Cesio , China , Minerales , República de Corea
3.
BMC Genomics ; 20(1): 199, 2019 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-30866796

RESUMEN

BACKGROUND: Peroxisome Proliferator-Activated receptor α (PPARα) and cAMP-Responsive Element Binding Protein 3-Like 3 (CREB3L3) are transcription factors involved in the regulation of lipid metabolism in the liver. The aim of the present study was to characterize the interrelationship between PPARα and CREB3L3 in regulating hepatic gene expression. Male wild-type, PPARα-/-, CREB3L3-/- and combined PPARα/CREB3L3-/- mice were subjected to a 16-h fast or 4 days of ketogenic diet. Whole genome expression analysis was performed on liver samples. RESULTS: Under conditions of overnight fasting, the effects of PPARα ablation and CREB3L3 ablation on plasma triglyceride, plasma ß-hydroxybutyrate, and hepatic gene expression were largely disparate, and showed only limited interdependence. Gene and pathway analysis underscored the importance of CREB3L3 in regulating (apo)lipoprotein metabolism, and of PPARα as master regulator of intracellular lipid metabolism. A small number of genes, including Fgf21 and Mfsd2a, were under dual control of PPARα and CREB3L3. By contrast, a strong interaction between PPARα and CREB3L3 ablation was observed during ketogenic diet feeding. Specifically, the pronounced effects of CREB3L3 ablation on liver damage and hepatic gene expression during ketogenic diet were almost completely abolished by the simultaneous ablation of PPARα. Loss of CREB3L3 influenced PPARα signalling in two major ways. Firstly, it reduced expression of PPARα and its target genes involved in fatty acid oxidation and ketogenesis. In stark contrast, the hepatoproliferative function of PPARα was markedly activated by loss of CREB3L3. CONCLUSIONS: These data indicate that CREB3L3 ablation uncouples the hepatoproliferative and lipid metabolic effects of PPARα. Overall, except for the shared regulation of a very limited number of genes, the roles of PPARα and CREB3L3 in hepatic lipid metabolism are clearly distinct and are highly dependent on dietary status.


Asunto(s)
Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Perfilación de la Expresión Génica/métodos , Hígado/crecimiento & desarrollo , PPAR alfa/genética , Ácido 3-Hidroxibutírico/sangre , Animales , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Dieta Cetogénica , Factores de Crecimiento de Fibroblastos/genética , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Metabolismo de los Lípidos , Hígado/química , Masculino , Ratones , PPAR alfa/metabolismo , Transducción de Señal , Simportadores , Triglicéridos/sangre , Proteínas Supresoras de Tumor/genética , Secuenciación Completa del Genoma
4.
Pharmacol Res ; 139: 325-336, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30472463

RESUMEN

Adipogenesis involved in hypertrophy and hyperplasia of adipocytes is responsible for expanding the mass of adipose tissues in obese individuals. Peroxisome proliferator-activated receptor γ (PPARγ) and CCAAT/enhancer-binding protein α (C/EBPα) are two principal transcription factors induced by delicate signaling pathways, including signal transducer and activator of transcription 5 (STAT5), in adipogenesis. Here, we demonstrated a novel role of ginkgetin, a biflavone from Ginkgo biloba leaves, as a STAT5 inhibitor that blocks the differentiation of preadipocytes into adipocytes. During the differentiation of 3T3-L1 cells, ginkgetin treatment during the first 2 days markedly inhibited the formation of lipid-bearing adipocytes. PPARγ and C/EBPα expression was decreased in 3T3-L1 cells during adipogenesis following ginkgetin treatment, whereas no change was observed in C/EBPß or C/EBPδ expression. Inhibition of PPARγ and C/EBPα expression by ginkgetin occurred through the prevention of STAT5 activation during the initiation phase of adipogenesis. In addition, ginkgetin-mediated the inhibition of adipogenesis was recapitulated in the differentiation of primary preadipocytes. Lastly, we confirmed the inhibitory effects of ginkgetin on the hypertrophy of white adipose tissues from high-fat diet-fed mice. These results indicate that ginkgetin is a potential anti-adipogenesis and anti-obesity drug.


Asunto(s)
Adipogénesis/efectos de los fármacos , Fármacos Antiobesidad/farmacología , Fármacos Antiobesidad/uso terapéutico , Biflavonoides/farmacología , Biflavonoides/uso terapéutico , Células 3T3-L1 , Animales , Proteína alfa Potenciadora de Unión a CCAAT/genética , Proteína alfa Potenciadora de Unión a CCAAT/metabolismo , Dieta Alta en Grasa , Ginkgo biloba , Masculino , Ratones , Ratones Endogámicos C57BL , PPAR gamma/genética , PPAR gamma/metabolismo , Hojas de la Planta , Transducción de Señal/efectos de los fármacos
5.
J Nanobiotechnology ; 17(1): 24, 2019 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-30722792

RESUMEN

BACKGROUND: Silica nanoparticles (SiNPs) are widely used for biosensing and diagnostics, and for the targeted delivery of therapeutic agents. Safety concerns about the biomedical and clinical applications of SiNPs have been raised, necessitating analysis of the effects of their intrinsic properties, such as sizes, shapes, and surface physicochemical characteristics, on human health to minimize risk in biomedical applications. In particular, SiNP size-associated toxicological effects, and the underlying molecular mechanisms in the vascular endothelium remain unclear. This study aimed to elucidate the detailed mechanisms underlying the cellular response to exposure to trace amounts of SiNPs and to determine applicable size criteria for biomedical application. METHODS: To clarify whether these SiNP-mediated cytotoxicity due to induction of apoptosis or necrosis, human ECs were treated with SiNPs of four different non-overlapping sizes under low serum-containing condition, stained with annexin V and propidium iodide (PI), and subjected to flow cytometric analysis (FACS). Two types of cell death mechanisms were assessed in terms of production of reactive oxygen species (ROS), endoplasmic reticulum (ER) stress induction, and autophagy activity. RESULTS: Spherical SiNPs had a diameter of 21.8 nm; this was further increased to 31.4, 42.9, and 56.7 nm. Hence, we investigated these effects in human endothelial cells (ECs) treated with these nanoparticles under overlap- or agglomerate-free conditions. The 20-nm SiNPs, but not SiNPs of other sizes, significantly induced apoptosis and necrosis. Surprisingly, the two types of cell death occurred independently and through different mechanisms. Apoptotic cell death resulted from ROS-mediated ER stress. Furthermore, autophagy-mediated necrotic cell death was induced through the PI3K/AKT/eNOS signaling axis. Together, the present results indicate that SiNPs within a diameter of < 20-nm pose greater risks to cells in terms of cytotoxic effects. CONCLUSION: These data provide novel insights into the size-dependence of the cytotoxic effects of silica nanoparticles and the underlying molecular mechanisms. The findings are expected to inform the applicable size range of SiNPs to ensure their safety in biomedical and clinical applications.


Asunto(s)
Apoptosis/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Nanopartículas/toxicidad , Necrosis/patología , Transducción de Señal/efectos de los fármacos , Dióxido de Silicio , Autofagia/efectos de los fármacos , Células Cultivadas , Medios de Cultivo , Estrés del Retículo Endoplásmico/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Nanopartículas/química , Necrosis/metabolismo , Tamaño de la Partícula , Fosfatidilinositol 3-Quinasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Dióxido de Silicio/química , Dióxido de Silicio/toxicidad
6.
Arterioscler Thromb Vasc Biol ; 36(9): 1772-81, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27417587

RESUMEN

OBJECTIVE: Liver-enriched transcription factor cAMP-responsive element-binding protein H (CREBH) regulates plasma triglyceride clearance by inducing lipoprotein lipase cofactors, such as apolipoprotein A-IV (apoA-IV), apoA-V, and apoC-II. CREBH also regulates apoA-I transcription. This study aims to determine whether CREBH has a role in lipoprotein metabolism and development of atherosclerosis. APPROACH AND RESULTS: CREBH-deficient Creb3l3(-/-) mice were bred with Ldlr(-/-) mice creating Ldlr(-/-) Creb3l3(-/-) double knockout mice. Mice were fed on a high-fat and high-sucrose Western diet for 20 weeks. We showed that CREBH deletion in Ldlr(-/-) mice increased very low-density lipoprotein-associated triglyceride and cholesterol levels, consistent with the impairment of lipoprotein lipase-mediated triglyceride clearance in these mice. In contrast, high-density lipoprotein cholesterol levels were decreased in CREBH-deficient mice, which was associated with decreased production of apoA-I from the liver. The results indicate that CREBH directly activated Apoa1 gene transcription. Accompanied by the worsened atherogenic lipid profile, Ldlr(-/-) Creb3l3(-/-) mice developed significantly more atherosclerotic lesions in the aortas than Ldlr(-/-) mice. CONCLUSIONS: We identified CREBH as an important regulator of lipoprotein metabolism and suggest that increasing hepatic CREBH activity may be a novel strategy for prevention and treatment of atherosclerosis.


Asunto(s)
Aorta/metabolismo , Enfermedades de la Aorta/metabolismo , Aterosclerosis/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/deficiencia , Receptores de LDL/deficiencia , Animales , Aorta/patología , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/patología , Apolipoproteína A-I/sangre , Apolipoproteína A-I/genética , Apolipoproteína B-100 , Apolipoproteínas B/sangre , Aterosclerosis/genética , Aterosclerosis/patología , Biomarcadores/sangre , Línea Celular Tumoral , HDL-Colesterol/sangre , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Dieta Alta en Grasa , Sacarosa en la Dieta , Modelos Animales de Enfermedad , Predisposición Genética a la Enfermedad , Humanos , Hiperlipidemias/genética , Hiperlipidemias/metabolismo , Lipoproteína Lipasa/metabolismo , Lipoproteínas VLDL/sangre , Hígado/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Placa Aterosclerótica , Receptores de LDL/genética , Índice de Severidad de la Enfermedad , Transcripción Genética , Activación Transcripcional , Triglicéridos/sangre
7.
Hepatology ; 61(3): 857-69, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25125366

RESUMEN

UNLABELLED: Fat-specific protein 27 (Fsp27) is a lipid droplet-associated protein that promotes lipid droplet (LD) growth and triglyceride (TG) storage in white adipocytes. Fsp27 is also highly expressed in the steatotic liver and contributes to TG accumulation. In this study we discovered that the liver produces Fsp27ß, an alternative Fsp27 isoform, which contains 10 additional amino acids at the N-terminus of the original Fsp27 (Fsp27α). White adipose tissue (WAT) and the liver specifically expressed Fsp27α and Fsp27ß transcripts, respectively, which were driven by distinct promoters. The Fsp27ß promoter was activated by the liver-enriched transcription factor cyclic-AMP-responsive-element-binding protein H (CREBH) but not by peroxisome proliferator-activated receptor gamma (PPARγ), which activated the Fsp27α promoter. Enforced expression of the constitutively active CREBH strongly induced Fsp27ß and the human ortholog CIDEC2 in mouse hepatocytes and HepG2 cells, respectively. In contrast, loss of CREBH decreased hepatic Fsp27ß in fasted mice, suggesting that CREBH plays a critical role in Fsp27ß expression in the liver. Similar to Fsp27α, Fsp27ß localized on the surface of lipid droplets and suppressed lipolysis. Consequently, enforced expression of Fsp27ß or CREBH promoted lipid droplet enlargement and TG accumulation in the liver. CONCLUSION: The CREBH-Fsp27ß axis is important for regulating lipid droplet dynamics and TG storage in the liver.


Asunto(s)
Proteína de Unión a Elemento de Respuesta al AMP Cíclico/fisiología , Hígado Graso/etiología , Gotas Lipídicas/fisiología , Hígado/metabolismo , Proteínas/genética , Activación Transcripcional , Animales , Ratones , Ratones Endogámicos C57BL , Especificidad de Órganos , PPAR gamma/fisiología , Proteínas/fisiología , Triglicéridos/metabolismo
8.
J Lipid Res ; 55(5): 850-9, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24598141

RESUMEN

cAMP responsive element-binding protein H (CREBH) is an endoplasmic reticulum (ER) anchored transcription factor that is highly expressed in the liver and small intestine and implicated in nutrient metabolism and proinflammatory response. ApoA-IV is a glycoprotein secreted primarily by the intestine and to a lesser degree by the liver. ApoA-IV expression is suppressed in CREBH-deficient mice and strongly induced by enforced expression of the constitutively active form of CREBH, indicating that CREBH is the major transcription factor regulating Apoa4 gene expression. Here, we show that CREBH directly controls Apoa4 expression through two tandem CREBH binding sites (5'-CCACGTTG-3') located on the promoter, which are conserved between human and mouse. Chromatin immunoprecipitation and electrophoretic mobility-shift assays demonstrated specific association of CREBH with the CREBH binding sites. We also demonstrated that a substantial amount of CREBH protein was basally processed to the active nuclear form in normal mouse liver, which was further increased in steatosis induced by high-fat diet or fasting, increasing apoA-IV expression. However, we failed to find significant activation of CREBH in response to ER stress, arguing against the critical role of CREBH in ER stress response.


Asunto(s)
Apolipoproteínas A/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Regulación de la Expresión Génica , Transcripción Genética , Animales , Secuencia de Bases , Sitios de Unión , Línea Celular , Hígado Graso/genética , Hepatocitos/metabolismo , Humanos , Ratones , Regiones Promotoras Genéticas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo
9.
Semin Liver Dis ; 33(4): 301-11, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24222088

RESUMEN

The liver is a central organ that controls systemic energy homeostasis and nutrient metabolism. Dietary carbohydrates and lipids, and fatty acids derived from adipose tissue are delivered to the liver, and utilized for gluconeogenesis, lipogenesis, and ketogenesis, which are tightly regulated by hormonal and neural signals. Hepatic lipogenesis is activated primarily by insulin that is secreted from the pancreas after a high-carbohydrate meal. Sterol regulatory element binding protein-1c (SREBP-1c) and carbohydrate-responsive element-binding protein (ChREBP) are major transcriptional regulators that induce key lipogenic enzymes to promote lipogenesis in the liver. Sterol regulatory element binding protein-1c is activated by insulin through complex signaling cascades that control SREBP-1c at both transcriptional and posttranslational levels. Carbohydrate-responsive element-binding protein is activated by glucose independently of insulin. Here, the authors attempt to summarize the current understanding of the molecular mechanism for the transcriptional regulation of hepatic lipogenesis, focusing on recent studies that explore the signaling pathways controlling SREBPs and ChREBP.


Asunto(s)
Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Hígado Graso/metabolismo , Lipogénesis , Hígado/metabolismo , Proteínas de Unión a los Elementos Reguladores de Esteroles/metabolismo , Transcripción Genética , Transporte Activo de Núcleo Celular , Animales , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Hígado Graso/genética , Regulación de la Expresión Génica , Humanos , Lipogénesis/genética , MicroARNs/metabolismo , Estabilidad Proteica , Transducción de Señal , Proteínas de Unión a los Elementos Reguladores de Esteroles/genética
10.
Circ Res ; 109(7): 739-49, 2011 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-21835911

RESUMEN

RATIONALE: Peroxiredoxin 2 (Prdx2), a thiol-specific peroxidase, has been reported to regulate proinflammatory responses, vascular remodeling, and global oxidative stress. OBJECTIVE: Although Prdx2 has been proposed to retard atherosclerosis development, no direct evidence and mechanisms have been reported. METHODS AND RESULTS: We show that Prdx2 is highly expressed in endothelial and immune cells in atherosclerotic lesions and blocked the increase of endogenous H(2)O(2) by atherogenic stimulation. Deficiency of Prdx2 in apolipoprotein E-deficient (ApoE(-/-)) mice accelerated plaque formation with enhanced activation of p65, c-Jun, JNKs, and p38 mitogen-activated protein kinase; and these proatherogenic effects of Prdx2 deficiency were rescued by administration of the antioxidant ebselen. In bone marrow transplantation experiments, we found that Prdx2 has a major role in inhibiting atherogenic responses in both vascular and immune cells. Prdx2 deficiency resulted in increased expression of vascular adhesion molecule-1, intercellular adhesion molecule-1, and monocyte chemotactic protein-1, which led to increased immune cell adhesion and infiltration into the aortic intima. Compared with deficiency of glutathione peroxidase 1 or catalase, Prdx2 deficiency showed a severe predisposition to develop atherosclerosis. CONCLUSIONS: Prdx2 is a specific peroxidase that inhibits atherogenic responses in vascular and inflammatory cells, and specific activation of Prdx2 may be an effective means of antiatherogenic therapy.


Asunto(s)
Aorta/enzimología , Apolipoproteínas E/deficiencia , Aterosclerosis/enzimología , Peroxirredoxinas/deficiencia , Animales , Antioxidantes/farmacología , Aorta/efectos de los fármacos , Aorta/inmunología , Aorta/patología , Apolipoproteínas E/genética , Aterosclerosis/genética , Aterosclerosis/inmunología , Aterosclerosis/patología , Aterosclerosis/prevención & control , Azoles/farmacología , Células de la Médula Ósea/enzimología , Trasplante de Médula Ósea , Catalasa/genética , Catalasa/metabolismo , Quimiocina CCL2/metabolismo , Modelos Animales de Enfermedad , Células Endoteliales/enzimología , Glutatión Peroxidasa/deficiencia , Glutatión Peroxidasa/genética , Peróxido de Hidrógeno/metabolismo , Molécula 1 de Adhesión Intercelular/metabolismo , Isoindoles , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Compuestos de Organoselenio/farmacología , Peroxirredoxinas/genética , Índice de Severidad de la Enfermedad , Transducción de Señal , Factores de Tiempo , Factor de Transcripción ReIA/metabolismo , Molécula 1 de Adhesión Celular Vascular/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Glutatión Peroxidasa GPX1
11.
Exp Mol Med ; 55(12): 2519-2530, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-38036736

RESUMEN

Aortic aneurysm is a chronic disease characterized by localized expansion of the aorta, including the ascending aorta, arch, descending aorta, and abdominal aorta. Although aortic aneurysms are generally asymptomatic, they can threaten human health by sudden death due to aortic rupture. Aortic aneurysms are estimated to lead to 150,000 ~ 200,000 deaths per year worldwide. Currently, there are no effective drugs to prevent the growth or rupture of aortic aneurysms; surgical repair or endovascular repair is the only option for treating this condition. The pathogenic mechanisms and therapeutic targets for aortic aneurysms have been examined over the past decade; however, there are unknown pathogenic mechanisms involved in cellular heterogeneity and plasticity, the complexity of the transforming growth factor-ß signaling pathway, inflammation, cell death, intramural neovascularization, and intercellular communication. This review summarizes the latest research findings and current pathogenic mechanisms of aortic aneurysms, which may enhance our understanding of aortic aneurysms.


Asunto(s)
Aneurisma de la Aorta Torácica , Rotura de la Aorta , Humanos , Enfermedad Crónica , Rotura de la Aorta/etiología , Rotura de la Aorta/cirugía , Aorta
12.
Theranostics ; 13(3): 1076-1090, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36793871

RESUMEN

Precise regulation of kinases and phosphatases is crucial for human metabolic homeostasis. This study aimed to investigate the roles and molecular mechanisms of protein tyrosine phosphatase type IVA1 (PTP4A1) in regulating hepatosteatosis and glucose homeostasis. Method: Ptp4a1-/- mice, adeno-associated virus encoding Ptp4a1 under liver-specific promoter, adenovirus encoding Fgf21, and primary hepatocytes were used to evaluate PTP4A1-mediated regulation in the hepatosteatosis and glucose homeostasis. Glucose tolerance test, insulin tolerance test, 2-deoxyglucose uptake assay, and hyperinsulinemic-euglycemic clamp were performed to estimate glucose homeostasis in mice. The staining, including oil red O, hematoxylin & eosin, and BODIPY, and biochemical analysis for hepatic triglycerides were performed to assess hepatic lipids. Luciferase reporter assays, immunoprecipitation, immunoblots, quantitative real-time polymerase chain reaction, and immunohistochemistry staining were conducted to explore the underlying mechanism. Results: Here, we found that deficiency of PTP4A1 aggravated glucose homeostasis and hepatosteatosis in mice fed a high-fat (HF) diet. Increased lipid accumulation in hepatocytes of Ptp4a1-/- mice reduced the level of glucose transporter 2 on the plasma membrane of hepatocytes leading to a diminution of glucose uptake. PTP4A1 prevented hepatosteatosis by activating the transcription factor cyclic adenosine monophosphate-responsive element-binding protein H (CREBH)/fibroblast growth factor 21 (FGF21) axis. Liver-specific PTP4A1 or systemic FGF21 overexpression in Ptp4a1-/- mice fed an HF diet restored the disorder of hepatosteatosis and glucose homeostasis. Finally, liver-specific PTP4A1 expression ameliorated an HF diet-induced hepatosteatosis and hyperglycemia in wild-type mice. Conclusions: Hepatic PTP4A1 is critical for regulating hepatosteatosis and glucose homeostasis by activating the CREBH/FGF21 axis. Our current study provides a novel function of PTP4A1 in metabolic disorders; hence, modulating PTP4A1 may be a potential therapeutic strategy against hepatosteatosis-related diseases.


Asunto(s)
Dieta Alta en Grasa , Hiperglucemia , Humanos , Animales , Ratones , Dieta Alta en Grasa/efectos adversos , Hígado/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Hiperglucemia/metabolismo , Proteínas Tirosina Fosfatasas/metabolismo , Glucosa/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de Ciclo Celular/metabolismo
13.
Front Cell Infect Microbiol ; 13: 1294801, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38089817

RESUMEN

Introduction: The binary PirA/PirB toxin expressed by Vibrio parahaemolyticus (PirABVp) is a virulent complex that causes acute hepatopancreatic necrosis disease (AHPND) in shrimps, affecting the global shrimp farming industry. AHPND is currently diagnosed by detecting pirA and pirB genes by PCR; however, several V. parahaemolyticus strains do not produce the two toxins as proteins. Thus, an immunoassay using antibodies may be the most effective tool for detecting toxin molecules. In this study, we report a sandwich ELISA-based immunoassay for the detection of PirABVp. Methods: We utilized a single-chain variable fragment (scFv) antibody library to select scFvs against the PirA or PirB subunits. Phage display panning rounds were conducted to screen and identify scFv antibodies directed against each recombinant toxin subunit. Selected scFvs were converted into IgGs to develop a sandwich immunoassay to detect recombinant and bacterial PirABVp. Results: Antibodies produced as IgG forms showed sub-nanomolar to nanomolar affinities (KD), and a pair of anti-PirA antibody as a capture and anti-PirB antibody as a detector showed a limit of detection of 201.7 ng/mL for recombinant PirABVp. The developed immunoassay detected PirABVp in the protein lysates of AHPND-causing V. parahaemolyticus (VpAHPND) and showed a significant detectability in moribund or dead shrimp infected with a VpAHPND virulent strain compared to that in non-infected shrimp. Discussion: These results indicate that the developed immunoassay is a reliable method for diagnosing AHPND by detecting PirABVp at the protein level and could be further utilized to accurately determine the virulence of extant or newly identified VpAHPND in the global shrimp culture industry.


Asunto(s)
Penaeidae , Toxinas Biológicas , Vibrio parahaemolyticus , Animales , Vibrio parahaemolyticus/genética , Penaeidae/microbiología , Ensayo de Inmunoadsorción Enzimática , Enfermedad Aguda , Necrosis
14.
Bioengineering (Basel) ; 10(10)2023 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-37892862

RESUMEN

Blood-brain barrier (BBB) models are important tools for studying CNS drug delivery, brain development, and brain disease. In vitro BBB models have been obtained from animals and immortalized cell lines; however, brain microvascular endothelial cells (BMECs) derived from them have several limitations. Furthermore, obtaining mature brain microvascular endothelial-like cells (BME-like cells) from human pluripotent stem cells (hPSCs) with desirable properties for establishing BBB models has been challenging. Here, we developed an efficient method for differentiating hPSCs into BMECs that are amenable to the development and application of human BBB models. The established conditions provided an environment similar to that occurring during BBB differentiation in the presence of the co-differentiating neural cell population by the modulation of TGF-ß and SHH signaling. The developed BME-like cells showed well-organized tight junctions, appropriate expression of nutrient transporters, and polarized efflux transporter activity. In addition, BME-like cells responded to astrocytes, acquiring substantial barrier properties as measured by transendothelial electrical resistance. Moreover, the BME-like cells exhibited an immune quiescent property of BBB endothelial cells by decreasing the expression of adhesion molecules. Therefore, our novel cellular platform could be useful for drug screening and the development of brain-permeable pharmaceuticals.

15.
Cardiovasc Res ; 119(5): 1265-1278, 2023 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-36534975

RESUMEN

AIMS: The nuclear factor-κB (NF-κB) signalling pathway plays a critical role in the pathogenesis of multiple vascular diseases. However, in endothelial cells (ECs), the molecular mechanisms responsible for the negative regulation of the NF-κB pathway are poorly understood. In this study, we investigated a novel role for protein tyrosine phosphatase type IVA1 (PTP4A1) in NF-κB signalling in ECs. METHODS AND RESULTS: In human tissues, human umbilical artery ECs, and mouse models for loss of function and gain of function of PTP4A1, we conducted histological analysis, immunostaining, laser-captured microdissection assay, lentiviral infection, small interfering RNA transfection, quantitative real-time PCR and reverse transcription-PCR, as well as luciferase reporter gene and chromatin immunoprecipitation assays. Short hairpin RNA-mediated knockdown of PTP4A1 and overexpression of PTP4A1 in ECs indicated that PTP4A1 is critical for inhibiting the expression of cell adhesion molecules (CAMs). PTP4A1 increased the transcriptional activity of upstream stimulatory factor 1 (USF1) by dephosphorylating its S309 residue and subsequently inducing the transcription of tumour necrosis factor-alpha-induced protein 3 (TNFAIP3/A20) and the inhibition of NF-κB activity. Studies on Ptp4a1 knockout or transgenic mice demonstrated that PTP4A1 potently regulates the interleukin 1ß-induced expression of CAMs in vivo. In addition, we verified that PTP4A1 deficiency in apolipoprotein E knockout mice exacerbated high-fat high-cholesterol diet-induced atherogenesis with upregulated expression of CAMs. CONCLUSION: Our data indicate that PTP4A1 is a novel negative regulator of vascular inflammation by inducing USF1/A20 axis-mediated NF-κB inactivation. Therefore, the expression and/or activation of PTP4A1 in ECs might be useful for the treatment of vascular inflammatory diseases.


Asunto(s)
Células Endoteliales , FN-kappa B , Vasculitis , Animales , Humanos , Ratones , Proteínas de Ciclo Celular/metabolismo , Células Endoteliales/metabolismo , Inflamación/genética , Inflamación/metabolismo , Proteínas de la Membrana/metabolismo , FN-kappa B/metabolismo , Proteínas Tirosina Fosfatasas/metabolismo , Transducción de Señal , Factores Estimuladores hacia 5'/metabolismo , Vasculitis/genética , Vasculitis/metabolismo
16.
BMB Rep ; 55(3): 142-147, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-34674794

RESUMEN

Human pluripotent stem cells (PSCs) have been utilized as a promising source in regenerative medicine. However, the risk of teratoma formation that comes with residual undifferentiated PSCs in differentiated cell populations is most concerning in the clinical use of PSC derivatives. Here, we report that a monoclonal antibody (mAb) targeting PSCs could distinguish undifferentiated PSCs, with potential teratoma-forming activity, from differentiated PSC progeny. A panel of hybridomas generated from mouse immunization with H9 human embryonic stem cells (hESCs) was screened for ESC-specific binding using flow cytometry. A novel mAb, K312, was selected considering its high stem cell-binding activity, and this mAb could bind to several human induced pluripotent stem cells and PSC lines. Cell-binding activity of K312 was markedly decreased as hESCs were differentiated into embryoid bodies or by retinoic acid treatment. In addition, a cell population negatively isolated from undifferentiated or differentiated H9 hESCs via K312 targeting showed a significantly reduced expression of pluripotency markers, including Oct4 and Nanog. Furthermore, K312-based depletion of pluripotent cells from differentiated PSC progeny completely prevented teratoma formation. Therefore, our findings suggest that K312 is utilizable in improving stem cell transplantation safety by specifically distinguishing residual undifferentiated PSCs. [BMB Reports 2022; 55(3): 142-147].


Asunto(s)
Células Madre Pluripotentes Inducidas , Células Madre Pluripotentes , Teratoma , Animales , Anticuerpos Monoclonales/metabolismo , Diferenciación Celular , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Células Madre Pluripotentes/metabolismo
17.
Biosensors (Basel) ; 12(10)2022 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-36290925

RESUMEN

Staphylococcal enterotoxin B (SEB) is a potent bacterial toxin that causes inflammatory stimulation and toxic shock, thus it is necessary to detect SEB in food and environmental samples. Here, we developed a sensitive immunodetection system using monoclonal antibodies (mAbs). Our study is the first to employ a baculovirus expression vector system (BEVS) to produce recombinant wild-type SEB. BEVS facilitated high-quantity and pure SEB production from suspension-cultured insect cells, and the SEB produced was characterized by mass spectrometry analysis. The SEB was stable at 4 °C for at least 2 years, maintaining its purity, and was further utilized for mouse immunization to generate mAbs. An optimal pair of mAbs non-competitive to SEB was selected for sandwich enzyme-linked immunosorbent assay-based immunodetection. The limit of detection of the immunodetection method was 0.38 ng/mL. Moreover, it displayed higher sensitivity in detecting SEB than commercially available immunodetection kits and retained detectability in various matrices and S. aureus culture supernatants. Thus, the results indicate that BEVS is useful for producing pure recombinant SEB with its natural immunogenic property in high yield, and that the developed immunodetection assay is reliable and sensitive for routine identification of SEB in various samples, including foods.


Asunto(s)
Toxinas Bacterianas , Staphylococcus aureus , Ratones , Animales , Baculoviridae , Enterotoxinas/análisis , Ensayo de Inmunoadsorción Enzimática/métodos , Anticuerpos Monoclonales
18.
Exp Mol Med ; 54(8): 1250-1261, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-36028759

RESUMEN

Obesity is a growing global epidemic that can cause serious adverse health consequences, including insulin resistance (IR) and nonalcoholic fatty liver disease (NAFLD). Obesity development can be attributed to energy imbalance and metabolic inflexibility. Here, we demonstrated that lack of Kelch-like protein 3 (KLHL3) mitigated the development of obesity, IR, and NAFLD by increasing energy expenditure. KLHL3 mutations in humans cause Gordon's hypertension syndrome; however, the role of KLHL3 in obesity was previously unknown. We examined differences in obesity-related parameters between control and Klhl3-/- mice. A significant decrease in body weight concomitant with fat mass loss and improved IR and NAFLD were observed in Klhl3-/- mice fed a high-fat (HF) diet and aged. KLHL3 deficiency inhibited obesity, IR, and NAFLD by increasing energy expenditure with augmentation of O2 consumption and CO2 production. Delivering dominant-negative (DN) Klhl3 using adeno-associated virus into mice, thereby dominantly expressing DN-KLHL3 in the liver, ameliorated diet-induced obesity, IR, and NAFLD. Finally, adenoviral overexpression of DN-KLHL3, but not wild-type KLHL3, in hepatocytes revealed an energetic phenotype with an increase in the oxygen consumption rate. The present findings demonstrate a novel function of KLHL3 mutation in extrarenal tissues, such as the liver, and may provide a therapeutic target against obesity and obesity-related diseases.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Metabolismo Energético , Resistencia a la Insulina , Proteínas de Microfilamentos , Enfermedad del Hígado Graso no Alcohólico , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Dieta Alta en Grasa/efectos adversos , Metabolismo Energético/genética , Humanos , Resistencia a la Insulina/genética , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Enfermedad del Hígado Graso no Alcohólico/genética , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Obesidad/genética , Obesidad/metabolismo
19.
Circulation ; 121(9): 1124-33, 2010 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-20176988

RESUMEN

BACKGROUND: The tumor necrosis factor receptor superfamily, which includes CD40, LIGHT, and OX40, plays important roles in atherosclerosis. CD137 (4-1BB), a member of the tumor necrosis factor receptor superfamily, has been reported to be expressed in human atherosclerotic lesions. However, limited information is available on the precise role of CD137 in atherosclerosis and the effects of blocking CD137/CD137 ligand signaling on lesion formation. METHODS AND RESULTS: We generated CD137-deficient apolipoprotein E-knockout mice (ApoE(-/-) CD137(-/-)) and LDL-receptor-knockout mice (Ldlr(-/-)CD137(-/-)) to investigate the role of CD137 in atherogenesis. The deficiency of CD137 induced a reduction in atherosclerotic plaque lesions in both atherosclerosis mouse models, which was attributed to the downregulation of cytokines such as interferon-gamma, monocyte chemoattractant protein-1, and tumor necrosis factor-alpha. CD137 signaling promoted the production of inflammatory molecules, including monocyte chemoattractant protein-1, interleukin-6, vascular cell adhesion molecule-1, and intercellular adhesion molecule-1, in endothelial cells. Stimulation of CD137 ligand signaling activated monocytes/macrophages and augmented the production of proinflammatory cytokines in atherosclerotic vessels. CONCLUSIONS: CD137/CD137 ligand signaling plays multiple roles in the progression of atherosclerosis, and thus, blockade of this pathway is a promising therapeutic target for the disease.


Asunto(s)
Ligando 4-1BB/fisiología , Aterosclerosis/prevención & control , Hipercolesterolemia/complicaciones , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/fisiología , Animales , Animales Congénicos , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Aterosclerosis/etiología , Aterosclerosis/inmunología , Cruzamientos Genéticos , Citocinas/biosíntesis , Citocinas/genética , Dieta Aterogénica , Modelos Animales de Enfermedad , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Retroalimentación Fisiológica , Femenino , Hipercolesterolemia/genética , Mediadores de Inflamación/metabolismo , Interferón gamma/inmunología , Activación de Linfocitos , Activación de Macrófagos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de LDL/deficiencia , Receptores de LDL/genética , Transducción de Señal , Linfocitos T/inmunología , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/deficiencia , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/genética
20.
Antioxidants (Basel) ; 10(8)2021 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-34439492

RESUMEN

Increased oxidative stress (OS) is considered a common etiology in the pathogenesis of cardiovascular disease (CVD). Therefore, the precise regulation of reactive oxygen species (ROS) in cardiovascular cells is essential to maintain normal physiological functions. Numerous regulators of cellular homeostasis are reportedly influenced by ROS. Hydrogen peroxide (H2O2), as an endogenous ROS in aerobic cells, is a toxic substance that can induce OS. However, many studies conducted over the past two decades have provided substantial evidence that H2O2 acts as a diffusible intracellular signaling messenger. Antioxidant enzymes, including superoxide dismutases, catalase, glutathione peroxidases, and peroxiredoxins (Prdxs), maintain the balance of ROS levels against augmentation of ROS production during the pathogenesis of CVD. Especially, Prdxs are regulatory sensors of transduced intracellular signals. The intracellular abundance of Prdxs that specifically react with H2O2 act as regulatory proteins. In this review, we focus on the role of Prdxs in the regulation of ROS-induced pathological changes in the development of CVD.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA