Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
J Biol Chem ; 300(2): 105655, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38237682

RESUMEN

Endoplasmic reticulum stress is associated with insulin resistance and the development of nonalcoholic fatty liver disease. Deficiency of the endoplasmic reticulum stress response T-cell death-associated gene 51 (TDAG51) (TDAG51-/-) in mice promotes the development of high-fat diet (HFD)-induced obesity, fatty liver, and hepatic insulin resistance. However, whether this effect is due specifically to hepatic TDAG51 deficiency is unknown. Here, we report that hepatic TDAG51 protein levels are consistently reduced in multiple mouse models of liver steatosis and injury as well as in liver biopsies from patients with liver disease compared to normal controls. Delivery of a liver-specific adeno-associated virus (AAV) increased hepatic expression of a TDAG51-GFP fusion protein in WT, TDAG51-/-, and leptin-deficient (ob/ob) mice. Restoration of hepatic TDAG51 protein was sufficient to increase insulin sensitivity while reducing body weight and fatty liver in HFD fed TDAG51-/- mice and in ob/ob mice. TDAG51-/- mice expressing ectopic TDAG51 display improved Akt (Ser473) phosphorylation, post-insulin stimulation. HFD-fed TDAG51-/- mice treated with AAV-TDAG51-GFP displayed reduced lipogenic gene expression, increased beta-oxidation and lowered hepatic and serum triglycerides, findings consistent with reduced liver weight. Further, AAV-TDAG51-GFP-treated TDAG51-/- mice exhibited reduced hepatic precursor and cleaved sterol regulatory-element binding proteins (SREBP-1 and SREBP-2). In vitro studies confirmed the lipid-lowering effect of TDAG51 overexpression in oleic acid-treated Huh7 cells. These studies suggest that maintaining hepatic TDAG51 protein levels represents a viable therapeutic approach for the treatment of obesity and insulin resistance associated with nonalcoholic fatty liver disease.


Asunto(s)
Resistencia a la Insulina , Enfermedad del Hígado Graso no Alcohólico , Animales , Humanos , Ratones , Muerte Celular , Dieta Alta en Grasa/efectos adversos , Hepatocitos/metabolismo , Resistencia a la Insulina/fisiología , Hígado/metabolismo , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Obesidad/metabolismo , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Linfocitos T/metabolismo , Masculino
2.
Glycobiology ; 33(3): 182-187, 2023 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-36728702

RESUMEN

Sialidases are found in viruses, bacteria, fungi, avians, and mammals. Mammalian sialidases differ in their specificity, optimum pH, subcellular localization, and tissue expression. To date, four genes encoding mammalian sialidases (NEU1-4) have been cloned. This review examines the functional impact of NEU4 sialidase on complex physiological and cellular processes. The intracellular localization and trafficking of NEU4 and its potential target molecules are discussed along with its impact on cancer, lysosomal storage disease, and cellular differentiation. Modulation of NEU4 expression may be essential not only for the breakdown of sialylated glycoconjugates, but also in the activation or inactivation of functionally important cellular events.


Asunto(s)
Diferenciación Celular , Enfermedades por Almacenamiento Lisosomal , Neoplasias , Neuraminidasa , Animales , Humanos , Diferenciación Celular/genética , Enfermedades por Almacenamiento Lisosomal/genética , Enfermedades por Almacenamiento Lisosomal/metabolismo , Mamíferos/genética , Mamíferos/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Neuraminidasa/genética , Neuraminidasa/metabolismo
3.
Arterioscler Thromb Vasc Biol ; 40(7): 1664-1679, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32434409

RESUMEN

OBJECTIVE: Cardiovascular disease is the primary cause of mortality in patients with chronic kidney disease. Vascular calcification (VC) in the medial layer of the vessel wall is a unique and prominent feature in patients with advanced chronic kidney disease and is now recognized as an important predictor and independent risk factor for cardiovascular and all-cause mortality in these patients. VC in chronic kidney disease is triggered by the transformation of vascular smooth muscle cells (VSMCs) into osteoblasts as a consequence of elevated circulating inorganic phosphate (Pi) levels, due to poor kidney function. The objective of our study was to investigate the role of TDAG51 (T-cell death-associated gene 51) in the development of medial VC. METHODS AND RESULTS: Using primary mouse and human VSMCs, we found that TDAG51 is induced in VSMCs by Pi and is expressed in the medial layer of calcified human vessels. Furthermore, the transcriptional activity of RUNX2 (Runt-related transcription factor 2), a well-established driver of Pi-mediated VC, is reduced in TDAG51-/- VSMCs. To explain these observations, we identified that TDAG51-/- VSMCs express reduced levels of the type III sodium-dependent Pi transporter, Pit-1, a solute transporter, a solute transporter, a solute transporter responsible for cellular Pi uptake. Significantly, in response to hyperphosphatemia induced by vitamin D3, medial VC was attenuated in TDAG51-/- mice. CONCLUSIONS: Our studies highlight TDAG51 as an important mediator of Pi-induced VC in VSMCs through the downregulation of Pit-1. As such, TDAG51 may represent a therapeutic target for the prevention of VC and cardiovascular disease in patients with chronic kidney disease.


Asunto(s)
Transdiferenciación Celular , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Osteogénesis , Factores de Transcripción/metabolismo , Calcificación Vascular/metabolismo , Anciano , Animales , Células Cultivadas , Colecalciferol , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica , Humanos , Hiperfosfatemia/inducido químicamente , Hiperfosfatemia/metabolismo , Hiperfosfatemia/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/patología , Fosfatos/metabolismo , Transducción de Señal , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo III/genética , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo III/metabolismo , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Calcificación Vascular/genética , Calcificación Vascular/patología , Calcificación Vascular/prevención & control
4.
J Biol Chem ; 293(19): 7329-7343, 2018 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-29593095

RESUMEN

The proprotein convertase subtilisin/kexin type-9 (PCSK9) plays a central role in cardiovascular disease (CVD) by degrading hepatic low-density lipoprotein receptor (LDLR). As such, loss-of-function (LOF) PCSK9 variants that fail to exit the endoplasmic reticulum (ER) increase hepatic LDLR levels and lower the risk of developing CVD. The retention of misfolded protein in the ER can cause ER stress and activate the unfolded protein response (UPR). In this study, we investigated whether a variety of LOF PCSK9 variants that are retained in the ER can cause ER stress and hepatic cytotoxicity. Although overexpression of these PCSK9 variants caused an accumulation in the ER of hepatocytes, UPR activation or apoptosis was not observed. Furthermore, ER retention of endogenous PCSK9 via splice switching also failed to induce the UPR. Consistent with these in vitro studies, overexpression of PCSK9 in the livers of mice had no impact on UPR activation. To elucidate the cellular mechanism to explain these surprising findings, we observed that the 94-kDa glucose-regulated protein (GRP94) sequesters PCSK9 away from the 78-kDa glucose-regulated protein (GRP78), the major activator of the UPR. As a result, GRP94 knockdown increased the stability of GRP78-PCSK9 complex and resulted in UPR activation following overexpression of ER-retained PCSK9 variants relative to WT secreted controls. Given that overexpression of these LOF PCSK9 variants does not cause UPR activation under normal homeostatic conditions, therapeutic strategies aimed at blocking the autocatalytic cleavage of PCSK9 in the ER represent a viable strategy for reducing circulating PCSK9.


Asunto(s)
Estrés del Retículo Endoplásmico/genética , Proteínas de Choque Térmico/metabolismo , Mutación con Pérdida de Función , Proproteína Convertasa 9/genética , Respuesta de Proteína Desplegada/genética , Animales , Apoptosis , Dominio Catalítico , Línea Celular , Retículo Endoplásmico/metabolismo , Chaperón BiP del Retículo Endoplásmico , Técnicas de Silenciamiento del Gen , Hepatocitos/metabolismo , Humanos , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Ratones , Proproteína Convertasa 9/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Interferencia de ARN , Empalme del ARN
5.
J Biol Chem ; 293(38): 14689-14706, 2018 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-30097518

RESUMEN

Atherosclerosis is a complex disease that involves alterations in lipoprotein metabolism and inflammation. Protein and lipid glycosylation events, such as sialylation, contribute to the development of atherosclerosis and are regulated by specific glycosidases, including sialidases. To evaluate the effect of the sialidase neuraminidase 1 (NEU1) on atherogenesis, here we generated apolipoprotein E (ApoE)-deficient mice that express hypomorphic levels of NEU1 (Neu1hypoApoe-/-). We found that the hypomorphic NEU1 expression in male Apoe-/- mice reduces serum levels of very-low-density lipoprotein (VLDL) and LDL cholesterol, diminishes infiltration of inflammatory cells into lesions, and decreases aortic sinus atherosclerosis. Transplantation of Apoe-/- bone marrow (BM) into Neu1hypoApoe-/- mice significantly increased atherosclerotic lesion development and had no effect on serum lipoprotein levels. Moreover, Neu1hypoApoe-/- mice exhibited a reduction in circulating monocyte and neutrophil levels and had reduced hyaluronic acid and P-selectin adhesion capability on monocytes/neutrophils and T cells. Consistent with these findings, administration of a sialidase inhibitor, 2-deoxy-2,3-dehydro-N-acetylneuraminic acid, had a significant anti-atherogenic effect in the Apoe-/- mice. In summary, the reduction in NEU1 expression or function decreases atherosclerosis in mice via its significant effects on lipid metabolism and inflammatory processes. We conclude that NEU1 may represent a promising target for managing atherosclerosis.


Asunto(s)
Apolipoproteínas E/genética , Aterosclerosis/metabolismo , Quimiotaxis de Leucocito , LDL-Colesterol/sangre , VLDL-Colesterol/sangre , Regulación hacia Abajo , Neuraminidasa/metabolismo , Animales , Aorta/patología , LDL-Colesterol/metabolismo , VLDL-Colesterol/metabolismo , Ácido Hialurónico/metabolismo , Hígado/metabolismo , Macrófagos/citología , Masculino , Ratones , Ratones Noqueados para ApoE , Músculo Liso Vascular/citología , Selectina-P/metabolismo , Linfocitos T/citología , Triglicéridos/metabolismo
6.
Hum Mol Genet ; 26(4): 661-673, 2017 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-28007910

RESUMEN

GM2 gangliosidoses are a group of lysosomal storage disorders which include Sandhoff disease and Tay-Sachs disease. Dysregulation of glutamate receptors has been recently postulated in the pathology of Sandhoff disease. Glutamate receptor association with neuronal pentraxins 1 and 2, and the neuronal pentraxin receptor facilitates receptor potentiation and synaptic shaping. In this study, we have observed an upregulation of a novel form of neuronal pentraxin 1 (NP1-38) in the brains of a mouse model of Sandhoff disease and Tay-Sachs disease. In order to determine the impact of NP1 on the pathophysiology of Sandhoff disease mouse models, we have generated an Np1-/-Hexb-/- double knockout mouse, and observed extended lifespan, improved righting reflex and enhanced body condition relative to Hexb-/- mice, with no effect on gliosis or apoptotic markers in the CNS. Sandhoff mouse brain slices reveals a reduction in AMPA receptor-mediated currents, and increased variability in total glutamate currents in the CA1 region of the hippocampus; Np1-/-Hexb-/- mice show a correction of this phenotype, suggesting NP1-38 may be interfering with glutamate receptor function. Indeed, some of the psychiatric aspects of Sandhoff and Tay-Sachs disease (particularly late onset) may be attributed to a dysfunctional hippocampal glutamatergic system. Our work highlights a potential role for synaptic proteins, such as NP1 and glutamate receptors in lysosomal storage diseases.


Asunto(s)
Proteína C-Reactiva/biosíntesis , Región CA1 Hipocampal/metabolismo , Proteínas del Tejido Nervioso/biosíntesis , Enfermedad de Sandhoff/metabolismo , Regulación hacia Arriba , Cadena beta de beta-Hexosaminidasa/biosíntesis , Animales , Proteína C-Reactiva/genética , Región CA1 Hipocampal/patología , Humanos , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Enfermedad de Sandhoff/patología , Cadena beta de beta-Hexosaminidasa/genética
7.
Curr Opin Lipidol ; 28(2): 209-212, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28092276

RESUMEN

PURPOSE OF REVIEW: This review seeks to describe the role of the asialoglycoprotein receptor (ASGR) in modulating non-HDL lipoprotein levels, platelet numbers and atherosclerosis. RECENT FINDINGS: Genetics studies have revealed that ASGR haplodeficiency provides protection from atherosclerosis. The potential interactions of ASGR with LDL receptor may regulate the rate of LDL uptake and as a result may lower plasma non-HDL cholesterol. ASGR clears senescent platelets and induces the expression of hepatic thrombopoietin. Platelet activity promotes plaque formation and atherosclerosis. SUMMARY: ASGR is a multifunctional receptor specializing in clearance of desialylated glycoproteins and platelets. The impact of ASGR on the levels of plasma non-HDL cholesterol makes it a potential therapeutic target for the treatment of hypercholesterolemia and atherosclerosis.


Asunto(s)
Receptor de Asialoglicoproteína/metabolismo , Aterosclerosis/metabolismo , Lípidos/sangre , Animales , Aterosclerosis/sangre , Plaquetas/metabolismo , Humanos , Neuraminidasa/metabolismo
8.
Hum Mol Genet ; 22(19): 3960-75, 2013 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-23727835

RESUMEN

Sandhoff disease (SD) is a lysosomal storage disorder caused by a lack of a functional ß-subunit of the ß-hexosaminidase A and B enzymes, leading to the accumulation of gangliosides in the central nervous system (CNS). The Hexb-/- mouse model of SD shows a progressive neurodegenerative phenotype similar to the human equivalent. Previous studies have revealed that Hexb-/- mice suffer from chronic neuroinflammation characterized by microglial activation and expansion. Tumor necrosis factor-α (TNFα), a key modulator of the CNS immune response in models of neurodegeneration, is a hallmark of this activation. In this study, we explore the role of TNFα in the development and progression of SD in mice, by creating a Hexb-/- Tnfα-/- double-knockout mouse. Our results revealed that the double-knockout mice have an ameliorated disease course, with an extended lifespan, enhanced sensorimotor coordination and improved neurological function. TNFα-deficient SD mice also show decreased levels of astrogliosis and reduced neuronal cell death, with no alterations in neuronal storage of gangliosides. Interestingly, temporal microglia activation appears similar between the Hexb-/- Tnfα-/- and SD mice. Evidence is provided for the TNFα activation of the JAK2/STAT3 pathway as a mechanism for astrocyte activation in the disease. Bone marrow transplantation experiments reveal that both CNS-derived and bone marrow-derived TNFα have a pathological effect in SD mouse models, with CNS-derived TNFα playing a larger role. This study reveals TNFα as a neurodegenerative cytokine mediating astrogliosis and neuronal cell death in SD and points to TNFα as a potential therapeutic target to attenuate neuropathogenesis.


Asunto(s)
Enfermedad de Sandhoff/metabolismo , Enfermedad de Sandhoff/patología , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/fisiología , Animales , Trasplante de Médula Ósea , Encéfalo/metabolismo , Muerte Celular , Modelos Animales de Enfermedad , Femenino , Gangliósidos/metabolismo , Gliosis/genética , Gliosis/patología , Humanos , Ratones , Ratones Noqueados , Microglía/metabolismo , Enfermedad de Sandhoff/genética , Enfermedad de Sandhoff/terapia , Transducción de Señal , beta-N-Acetilhexosaminidasas/genética , beta-N-Acetilhexosaminidasas/metabolismo
9.
Arterioscler Thromb Vasc Biol ; 34(11): 2394-403, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25212235

RESUMEN

OBJECTIVE: Deficiency of the high-density lipoprotein receptor, scavenger receptor class B, type I (SR-BI), in apolipoprotein E knockout or hypomorphic mice, respectively, results in spontaneous or diet-inducible occlusive coronary artery (CA) atherosclerosis, myocardial infarction, and early death. Here, we examine effects of SR-BI deficiency on cardiovascular phenotypes in low-density lipoprotein receptor (LDLR) knockout mice fed different atherogenic diets. APPROACH AND RESULTS: SR-BI/LDLR double knockout and control LDLR knockout mice were fed atherogenic diets containing different amounts of fat, cholesterol, and sodium cholate. Double knockout mice fed atherogenic diets high in cholesterol exhibited significantly reduced survival compared with LDLR knockout mice fed the same diets. In addition to increased diet-accelerated aortic sinus atherosclerosis, we observed significant diet-induced CA atherosclerosis in double knockout mice and diet-dependent accumulation of platelets in CA atherosclerotic plaques. This was accompanied by substantial myocardial fibrosis in double knockout mice fed high cholesterol diets. Atherogenic diet fed double knockout mice also exhibited higher circulating cytokine levels, monocytosis with increased proportions of Ly6C(hi) and Ly6C(int) monocytes, and higher adhesion molecule expression in CA endothelial cells compared with control LDLR knockout mice. CONCLUSIONS: Diet-accelerated atherosclerosis and occlusive, platelet-rich CA disease in SR-BI/LDLR double knockout mice is affected by amounts of cholesterol and cholate in atherogenic diets and is accompanied by increased expression of vascular cell adhesion molecule-1 and intercellular adhesion molecule-1 in CAs and increased Ly6C(hi) and Ly6C(int) monocytes in circulation. The increased vascular cell adhesion molecule-1 and intercellular adhesion molecule-1 in CA endothelial cells in SR-BI-deficient mice likely explains their increased susceptibility to atherosclerosis in CAs.


Asunto(s)
Arteriopatías Oclusivas/epidemiología , Enfermedad de la Arteria Coronaria/epidemiología , Dieta Aterogénica/efectos adversos , Infarto del Miocardio/epidemiología , Receptores de LDL/deficiencia , Receptores Depuradores de Clase B/deficiencia , Animales , Arteriopatías Oclusivas/metabolismo , Colatos/efectos adversos , Colesterol en la Dieta/efectos adversos , Enfermedad de la Arteria Coronaria/metabolismo , Citocinas/sangre , Modelos Animales de Enfermedad , Endotelio Vascular/metabolismo , Incidencia , Molécula 1 de Adhesión Intercelular/metabolismo , Ratones , Ratones Noqueados , Infarto del Miocardio/metabolismo , Receptores de LDL/genética , Factores de Riesgo , Receptores Depuradores de Clase B/genética , Molécula 1 de Adhesión Celular Vascular/metabolismo
10.
J Med Genet ; 49(9): 591-7, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22892202

RESUMEN

A viable treatment for lysosomal storage disease has been very difficult to attain. One option is pharmacological inhibition of synthetic pathways to reduce substrate accumulations. Miglustat N-butyldeoxynojirimycin (NBDNJ), an inhibitor of glucosylceramide synthase, has shown much promise in clinical trials for the treatment of Type I Gaucher disease. The molecular events invoked by NBDNJ in cell culture and in animal models have not been so definitive. This review discusses the biochemical and molecular impact of NBDNJ as it relates to its potential as a therapeutic drug.


Asunto(s)
1-Desoxinojirimicina/análogos & derivados , Enfermedad de Gaucher/tratamiento farmacológico , 1-Desoxinojirimicina/química , 1-Desoxinojirimicina/uso terapéutico , Animales , Ensayos Clínicos como Asunto , Modelos Animales de Enfermedad , Gangliósidos/biosíntesis , Gangliósidos/química , Humanos
11.
J Lipid Res ; 53(12): 2573-85, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22984145

RESUMEN

Lipoprotein metabolism is an important contributing factor in the development and progression of atherosclerosis. Plasma lipoproteins and their receptors are heavily glycosylated and sialylated, and levels of sialic acids modulate their biological functions. Sialylation is controlled by the activities of sialyltranferases and sialidases. To address the impact of sialidase (neu1) activity on lipoprotein metabolism, we have generated a mouse model with a hypomorphic neu1 allele (B6.SM) that displays reduced sialidase expression and sialidase activity. The objectives of this study are to determine the impact of sialidase on the rate of hepatic lipoprotein secretion and lipoprotein uptake. Our results indicate that hepatic levels of cholesterol and triglycerides are significantly higher in B6.SM mice compared with C57Bl/6 mice; however, VLDL-triglyceride production rate is lower. In addition, B6.SM mice show significantly lower levels of hepatic microsomal triglyceride transfer protein (MTP) and active sterol-regulatory element binding protein (SREBP)-2 but higher levels of diglyceride acyltransferase (DGAT)2; these are all indicative of increased hepatic lipid storage. Rescue of sialidase activity in hypomorphic sialidase mice using helper-dependent adenovirus resulted in increased VLDL production and an increase in MTP levels. Furthermore, hypomorphic sialidase expression results in stabilization of hepatic LDL receptor (LDLR) protein expression, which enhances LDL uptake. These findings provide novel evidence for a central role of sialidase in the cross talk between the uptake and production of lipoproteins.


Asunto(s)
Colesterol/sangre , Lipoproteínas VLDL/biosíntesis , Neuraminidasa/genética , Adenoviridae/aislamiento & purificación , Animales , Línea Celular , Colesterol/metabolismo , Regulación hacia Abajo , Humanos , Ratones , Ratones Endogámicos , Neuraminidasa/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Triglicéridos/biosíntesis
12.
Mol Genet Metab ; 107(1-2): 173-85, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22898113

RESUMEN

Sialidosis is an autosomal recessive disorder caused by a dysfunctional Sialidase enzyme. Categorised into two phenotypes, Sialidosis type I and II, Sialidosis is a highly heterogeneous disorder with varying ages of onset and pathologies. Currently, there is no viable therapy for the treatment of Sialidosis patients. At the molecular level, cells from Sialidosis patients with compound heterozygous mutations show improper enzyme folding, loss of Sialidase enzyme activity and subsequent accumulation of sialylconjugates within lysosomes. One promising treatment option is the use of small pharmacological molecules to increase the enzymatic activities of mutant proteins. In this study, we examined the efficacy of the immuno-suppressant (Celastrol) as well as a proteosomal inhibitor (MG132) to rescue mutant enzymes with altered conformation. Our results reveal that MG132 enhances enzyme activity and its localisation in cells expressing defective Sialidase. We also found that MG132 reduces accumulation of ganglioside products, GT1b, GD3, and GM3 in pre-loaded Sialidosis cells. Alternatively, Celastrol appears to reduce Sialidase expression and activity revealing a potentially novel effect of Celastrol on Sialidase. Interestingly, the combination of Celastrol and MG132 appears to amplify the beneficial impact of MG132 on both the endogenous and recombinant expression of defective Sialidase. This study explores a novel biological criteria to assess the efficacy of small molecules through accumulation analysis and points to a potential therapeutic strategy for the treatment of Sialidosis.


Asunto(s)
Inhibidores de Cisteína Proteinasa/farmacología , Leupeptinas/farmacología , Mucolipidosis/genética , Mutación , Neuraminidasa/genética , Triterpenos/farmacología , Línea Celular , Activación Enzimática/efectos de los fármacos , Activación Enzimática/genética , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Gangliósidos/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Proteína 2 de la Membrana Asociada a los Lisosomas/metabolismo , Mucolipidosis/metabolismo , Neuraminidasa/metabolismo , Triterpenos Pentacíclicos , Unión Proteica , Transporte de Proteínas
13.
Front Physiol ; 13: 1023397, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36714321

RESUMEN

High levels of low density lipoprotein (LDL) cholesterol and low levels of high density lipoprotein (HDL) cholesterol are risk factors for cardiovascular disease. Mice that lack genes involved in the clearance of LDL from the bloodstream, such as the LDL receptor and apolipoprotein E, are widely used models of experimental atherosclerosis. Conversely, mice that lack the HDL receptor, scavenger receptor class B type I, and therefore have disrupted HDL functionality, also develop diet-inducible atherosclerosis but are a seldom-used disease model. In this study, we compared atherosclerosis and associated phenotypes in scavenger receptor class B type I knockout mice with those of wild type, LDL receptor knockout, and apolipoprotein E knockout mice after 20 weeks of being fed an atherogenic diet containing sodium cholate. We found that while scavenger receptor class B type I knockout mice had substantially lower plasma cholesterol than LDL receptor and apolipoprotein E knockout mice, they developed atherosclerotic plaques with similar sizes and compositions in their aortic sinuses, and more extensive atherosclerosis in their descending aortas and coronary arteries. This was associated with elevated tumor necrosis factor alpha levels in scavenger receptor class B type I knockout mice compared to wild type and LDL receptor knockout mice, and lymphocytosis, monocytosis, and elevated vascular cell adhesion molecule expression in coronary artery endothelial cells compared to the other mice examined. We conclude that extensive atherosclerosis in arteries that are not generally susceptible to atherosclerosis in scavenger receptor class B type I knockout mice is driven by factors in addition to hypercholesterolemia, including inflammation, dysregulation of the immune system and increased sensitivity of endothelial cells in arteries that are normally resistant to atherosclerosis. Scavenger receptor class B type I knockout mice fed a cholate containing atherogenic diet may prove to be a useful model to study mechanisms of atherosclerosis and evaluate treatments that rely on intact LDL clearance pathways.

14.
Nat Commun ; 13(1): 770, 2022 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-35140212

RESUMEN

Evidence suggests that caffeine (CF) reduces cardiovascular disease (CVD) risk. However, the mechanism by which this occurs has not yet been uncovered. Here, we investigated the effect of CF on the expression of two bona fide regulators of circulating low-density lipoprotein cholesterol (LDLc) levels; the proprotein convertase subtilisin/kexin type 9 (PCSK9) and the low-density lipoprotein receptor (LDLR). Following the observation that CF reduced circulating PCSK9 levels and increased hepatic LDLR expression, additional CF-derived analogs with increased potency for PCSK9 inhibition compared to CF itself were developed. The PCSK9-lowering effect of CF was subsequently confirmed in a cohort of healthy volunteers. Mechanistically, we demonstrate that CF increases hepatic endoplasmic reticulum (ER) Ca2+ levels to block transcriptional activation of the sterol regulatory element-binding protein 2 (SREBP2) responsible for the regulation of PCSK9, thereby increasing the expression of the LDLR and clearance of LDLc. Our findings highlight ER Ca2+ as a master regulator of cholesterol metabolism and identify a mechanism by which CF may protect against CVD.


Asunto(s)
Cafeína/farmacología , Colesterol/metabolismo , Hígado/metabolismo , Proproteína Convertasa 9/metabolismo , Proteína 2 de Unión a Elementos Reguladores de Esteroles/farmacología , Animales , LDL-Colesterol/metabolismo , Células Hep G2 , Hepatocitos , Humanos , Metabolismo de los Lípidos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Proproteína Convertasa 9/genética , Receptores de LDL/genética , Receptores de LDL/metabolismo
15.
Kidney360 ; 3(8): 1394-1410, 2022 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-36176646

RESUMEN

Background: PCSK9 modulates the uptake of circulating lipids through a range of receptors, including the low-density lipoprotein receptor (LDLR) and CD36. In the kidney, CD36 is known to contribute to renal injury through pro-inflammatory and -fibrotic pathways. In this study, we sought to investigate the role of PCSK9 in modulating renal lipid accumulation and injury through CD36 using a high fat diet (HFD)-induced murine model. Methods: The effect of PCSK9 on the expression of CD36 and intracellular accumulation of lipid was examined in cultured renal cells and in the kidneys of male C57BL/6J mice. The effect of these findings was subsequently explored in a model of HFD-induced renal injury in Pcsk9 -/- and Pcsk9 +/+ littermate control mice on a C57BL/6J background. Results: In the absence of PCSK9, we observed heightened CD36 expression levels, which increased free fatty acid (FFA) uptake in cultured renal tubular cells. As a result, PCSK9 deficiency was associated with an increase in long-chain saturated FFA-induced ER stress. Consistent with these observations, Pcsk9-/- mice fed a HFD displayed elevated ER stress, inflammation, fibrosis, and renal injury relative to HFD-fed control mice. In contrast to Pcsk9-/- mice, pretreatment of WT C57BL/6J mice with evolocumab, an anti-PCSK9 monoclonal antibody (mAb) that binds to and inhibits the function of circulating PCSK9, protected against HFD-induced renal injury in association with reducing cell surface CD36 expression on renal epithelia. Conclusions: We report that circulating PCSK9 modulates renal lipid uptake in a manner dependent on renal CD36. In the context of increased dietary fat consumption, the absence of circulating PCSK9 may promote renal lipid accumulation and subsequent renal injury. However, although the administration of evolocumab blocks the interaction of PCSK9 with the LDLR, this evolocumab/PCSK9 complex can still bind CD36, thereby protecting against HFD-induced renal lipotoxicity.


Asunto(s)
Antígenos CD36 , Ácidos Grasos no Esterificados , Animales , Anticuerpos Monoclonales/farmacología , Dieta Alta en Grasa/efectos adversos , Grasas de la Dieta , Fibrosis , Riñón/metabolismo , Lipoproteínas LDL/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Proproteína Convertasa 9/genética
16.
Mol Genet Metab ; 97(1): 43-52, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19217813

RESUMEN

SM/J is an inbred mouse strain with a complex phenotype including small body size, impaired immune response and a tissue-specific sialidase deficiency. We identified a regulatory mutation, (-519G-->A) within the neu1 promoter which in reporter assays resulted in significantly reduced transcription. This mutation generates a consensus binding site for Nkx3 family transcription repressors. Recombinant Nkx3.2 bound strongly to and preferentially repressed transcription of the mutant promoter. This tissue-specific deficiency results in a retarded immune response and modulates leukocyte recruitment. Examination of the hepatic microcirculation in mutant mice revealed increased rolling and decreased adhesion of leukocytes. Our findings support a significant role for lysosomal sialidase in inflammation and highlight the significance of repressor-recruitment in genetic disease.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Neuraminidasa/deficiencia , Neuraminidasa/genética , Mutación Puntual/genética , Regiones Promotoras Genéticas , Proteínas Represoras/metabolismo , Factores de Transcripción/metabolismo , Animales , Secuencia de Bases , Línea Celular , Movimiento Celular , ADN/metabolismo , Modelos Animales de Enfermedad , Ensayo de Cambio de Movilidad Electroforética , Inflamación/enzimología , Leucocitos/citología , Hígado/irrigación sanguínea , Hígado/citología , Ratones , Microcirculación , Datos de Secuencia Molecular , Especificidad de Órganos , Unión Proteica , Transcripción Genética
17.
JHEP Rep ; 1(6): 418-429, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32039393

RESUMEN

The fatty acid translocase, also known as CD36, is a well-established scavenger receptor for fatty acid (FA) uptake and is abundantly expressed in many metabolically active tissues. In the liver, CD36 is known to contribute to the progression of non-alcoholic fatty liver disease and to the more severe non-alcoholic steatohepatitis, by promoting triglyceride accumulation and subsequent lipid-induced endoplasmic reticulum (ER) stress. Given the recent discovery that the hepatocyte-secreted proprotein convertase subtilisin/kexin type 9 (PCSK9) blocks CD36 expression, we sought to investigate the role of PCSK9 in liver fat accumulation and injury in response to saturated FAs and in a mouse model of diet-induced hepatic steatosis. METHODS: In this study, we investigated the role of PCSK9 on the uptake and accumulation of FAs, as well as FA-induced toxicity, in a variety of cultured hepatocytes. Diet-induced hepatic steatosis and liver injury were also assessed in Pcsk9 -/- mice. RESULTS: Our results indicate that PCSK9 deficiency in cultured hepatocytes increased the uptake and accumulation of saturated and unsaturated FAs. In the presence of saturated FAs, PCSK9 also protected cultured hepatocytes from ER stress and cytotoxicity. In line with these findings, a metabolic challenge using a high-fat diet caused severe hepatic steatosis, ER stress inflammation and fibrosis in the livers of Pcsk9 -/- mice compared to controls. Given that inhibition of CD36 ablated the observed accumulation of lipid in vitro and in vivo, our findings also highlight CD36 as a strong contributor to steatosis and liver injury in the context of PCSK9 deficiency. CONCLUSIONS: Collectively, our findings demonstrate that PCSK9 regulates hepatic triglyceride content in a manner dependent on CD36. In the presence of excess dietary fats, PCSK9 can also protect against hepatic steatosis and liver injury. LAY SUMMARY: The proprotein convertase subtilisin/kexin type 9 (PCSK9) is a circulating protein known to reduce the abundance of receptors on the surface of liver cells charged with the task of lipid uptake from the circulation. Although PCSK9 deficiency is known to cause lipid accumulation in mice and in cultured cells, the toxicological implications of this observation have not yet been reported. In this study, we demonstrate that PCSK9 can protect against cytotoxicity in cultured liver cells treated with a saturated fatty acid and we also show that Pcsk9 knockout mice develop increased liver injury in response to a high-fat diet.

18.
J Neuroimmunol ; 306: 55-67, 2017 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-28385189

RESUMEN

Sandhoff disease is an inherited lysosomal storage disease, resulting from the deficiency of lysosomal ß-hexosaminidase A and B enzyme activity. The Hexb-/- mouse model recapitulates human disease and leads to fatal neurodegeneration and neuroinflammation. IL-15 is important for the proliferation of NK, NK T, and CD8+ cytotoxic/memory T cells. In order to determine how changes to IL-15-dependent immune cell populations would alter the course of Sandhoff disease in mice, we generated a Hexb-/-Il-15-/- double knockout mouse and used motor behaviour tests, analyzed peripheral blood and brain leukocyte immunophenotypes, cytokine secretion, as well as examined markers of microgliosis, astrogliosis and apoptosis. Hexb-/-Il-15-/- mice had an accelerated neurodegenerative phenotype, and reached the humane endpoint at 118±3.5d, compared to Hexb-/- mice (127±2.2d). The performance of Hexb-/-Il-15-/- mice declined earlier than Hexb-/- mice on the rotarod and righting reflex motor behaviour tests. Hexb-/- mice had a significantly higher prevalence of pro-inflammatory monocytes in the blood relative to C57BL/6 mice, but this was unaltered by IL-15 deficiency. The prevalence of NK cells and CD8+ T cells in Il-15-/- and Hexb-/-Il-15-/- mice was decreased compared to wild type and Hexb-/- mice. While Hexb-/- mice displayed an increase in the prevalence of CD4+ and CD8+ T cells in brain leukocytes compared to C57BL/6 mice, there was a decrease in CD8+ T cells in Hexb-/-Il-15-/- compared to Hexb-/- mice. In addition, circulating IL-17 and IL-10 levels were significantly higher in Hexb-/-Il-15-/- mice, suggesting heightened inflammation compared to Hexb-/- mice. Interestingly, astrogliosis levels were significantly reduced in the cerebellum of Hexb-/-Il-15-/- mice compared to Hexb-/- mice while microgliosis was not affected in brains of Hexb-/-Il-15-/- mice. Our study demonstrated that IL-15 depletion dramatically reduced numbers of NK and CD8+ T cells as well as astrocytes but accelerated disease progression in Sandhoff mice. These results pointed to interactions between NK/CD8+ T cells and astrogliosis and potentially a protective role for NK/CD8+ T cells and/or astrocytes during disease progression. This observation supports the notion that expanding the IL-15-dependent NK and CD8+ T cells populations with IL-15 therapy may have therapeutic benefits for Sandhoff disease.


Asunto(s)
Linfocitos T CD8-positivos/patología , Enfermedades Cerebelosas/etiología , Gliosis/terapia , Células Asesinas Naturales/patología , Enfermedad de Sandhoff/complicaciones , Enfermedad de Sandhoff/mortalidad , Animales , Antígenos CD/metabolismo , Apoptosis/genética , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Enfermedades Cerebelosas/genética , Enfermedades Cerebelosas/patología , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Hexosaminidasa B/genética , Hexosaminidasa B/metabolismo , Interleucina-15/genética , Interleucina-15/metabolismo , Células Asesinas Naturales/metabolismo , Locomoción/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Trastornos del Movimiento/etiología , Trastornos del Movimiento/genética , Enfermedad de Sandhoff/genética
19.
J Neuroimmunol ; 299: 19-27, 2016 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-27725117

RESUMEN

Microgliosis and astrogliosis are known to be exacerbating factors in the progression of the lysosomal storage disorder Sandhoff disease. We have also found evidence for excitotoxicity via glutamate receptors in Sandhoff disease. To view the interaction of these cascades, we measured cerebellar expression of markers for gliosis, apoptosis, and excitatory synapses over the disease course in a Sandhoff disease mouse model. We observe a 2-stage model, with initial activation of microgliosis as early as 60days of age, followed by a later onset of astrogliosis, caspase-mediated apoptosis, and reduction in GluR1 at approximately 100days of age. These results implicate immune cells as first responders in Sandhoff disease.


Asunto(s)
Cerebelo/patología , Modelos Animales de Enfermedad , Gliosis/patología , Enfermedad de Sandhoff/patología , Animales , Cerebelo/metabolismo , Femenino , Gliosis/genética , Gliosis/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Enfermedad de Sandhoff/genética , Enfermedad de Sandhoff/metabolismo , Cadena beta de beta-Hexosaminidasa/genética
20.
J Am Heart Assoc ; 5(8)2016 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-27528409

RESUMEN

BACKGROUND: Monocyte recruitment leads to accumulation of macrophage foam cells and contributes to atherosclerotic lesion growth. Recent studies have reported that lesion-resident macrophages can proliferate and represent a major cellular component during lesion development. This study was designed to assess whether the rate of macrophage proliferation changes during well-established stages of lesion growth and to characterize other populations of proliferating cells within these lesions. METHODS AND RESULTS: Using murine models of atherosclerosis (Apoe(-/-) and LDLr(-/-) mice) and human coronary artery lesions, in situ proliferation of lesion-resident cells at different stages of growth was assessed by staining for Ki67 and bromodeoxyuridine (BrdU). In early lesions, close to half of all actively growing macrophages were proliferating in situ. BrdU pulse labeling allowed for accurate identification of in situ proliferating macrophages compared to those derived from monocyte recruitment. Local macrophage proliferation declined as lesions advanced. Interestingly, intimal inflammatory cell infiltrates containing proliferating T lymphocytes were identified during the active phase of lesion growth and correlated with apoptotic cell death. Inflammatory cell infiltrates were completely resolved in advanced lesions and replaced with the necrotic core. CONCLUSIONS: Our findings indicate that atherosclerotic lesions contain locally proliferating macrophages primarily during early and intermediate stages of lesion growth. Furthermore, T-lymphocyte-enriched inflammatory cell infiltrates represent a novel subset of proliferating cells within the atherosclerotic lesion that correlate with apoptosis and precede the necrotic core. These findings have novel implications in understanding the pathogenesis of atherosclerosis and may implicate proliferating T lymphocytes as a contributing factor to lesion progression and stability.


Asunto(s)
Enfermedad de la Arteria Coronaria/patología , Macrófagos/patología , Animales , Aorta/metabolismo , Apoptosis , Proliferación Celular/fisiología , Trombosis Coronaria/patología , Vasos Coronarios/patología , Modelos Animales de Enfermedad , Femenino , Humanos , Antígeno Ki-67/metabolismo , Masculino , Ratones Noqueados para ApoE , Linfocitos T/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA