Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nature ; 627(8003): 416-423, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38418872

RESUMEN

Permanent epigenetic silencing using programmable editors equipped with transcriptional repressors holds great promise for the treatment of human diseases1-3. However, to unlock its full therapeutic potential, an experimental confirmation of durable epigenetic silencing after the delivery of transient delivery of editors in vivo is needed. To this end, here we targeted Pcsk9, a gene expressed in hepatocytes that is involved in cholesterol homeostasis. In vitro screening of different editor designs indicated that zinc-finger proteins were the best-performing DNA-binding platform for efficient silencing of mouse Pcsk9. A single administration of lipid nanoparticles loaded with the editors' mRNAs almost halved the circulating levels of PCSK9 for nearly one year in mice. Notably, Pcsk9 silencing and accompanying epigenetic repressive marks also persisted after forced liver regeneration, further corroborating the heritability of the newly installed epigenetic state. Improvements in construct design resulted in the development of an all-in-one configuration that we term evolved engineered transcriptional repressor (EvoETR). This design, which is characterized by a high specificity profile, further reduced the circulating levels of PCSK9 in mice with an efficiency comparable with that obtained through conventional gene editing, but without causing DNA breaks. Our study lays the foundation for the development of in vivo therapeutics that are based on epigenetic silencing.


Asunto(s)
Epigénesis Genética , Epigenoma , Edición Génica , Silenciador del Gen , Animales , Ratones , Colesterol/metabolismo , Epigénesis Genética/genética , Epigenoma/genética , Edición Génica/métodos , Hepatocitos/metabolismo , Hígado/metabolismo , Regeneración Hepática , Nanopartículas , Proproteína Convertasa 9/sangre , Proproteína Convertasa 9/deficiencia , Proproteína Convertasa 9/genética , Proteínas Represoras/administración & dosificación , Proteínas Represoras/metabolismo , Dedos de Zinc
2.
Stem Cell Reports ; 16(12): 2958-2972, 2021 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-34739847

RESUMEN

Proprotein convertase subtilisin kexin type 9 (PCSK9) is a key regulator of low-density lipoprotein (LDL) cholesterol metabolism and the target of lipid-lowering drugs. PCSK9 is mainly expressed in hepatocytes. Here, we show that PCSK9 is highly expressed in undifferentiated human induced pluripotent stem cells (hiPSCs). PCSK9 inhibition in hiPSCs with the use of short hairpin RNA (shRNA), CRISPR/cas9-mediated knockout, or endogenous PCSK9 loss-of-function mutation R104C/V114A unveiled its new role as a potential cell cycle regulator through the NODAL signaling pathway. In fact, PCSK9 inhibition leads to a decrease of SMAD2 phosphorylation and hiPSCs proliferation. Conversely, PCSK9 overexpression stimulates hiPSCs proliferation. PCSK9 can interfere with the NODAL pathway by regulating the expression of its endogenous inhibitor DACT2, which is involved in transforming growth factor (TGF) ß-R1 lysosomal degradation. Using different PCSK9 constructs, we show that PCSK9 interacts with DACT2 through its Cys-His-rich domain (CHRD) domain. Altogether these data highlight a new role of PCSK9 in cellular proliferation and development.


Asunto(s)
Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Proteína Nodal/metabolismo , Proproteína Convertasa 9/metabolismo , Transducción de Señal , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Diferenciación Celular , Línea Celular , Membrana Celular/metabolismo , Proliferación Celular , Regulación de la Expresión Génica , Humanos , Mutación con Pérdida de Función , Proteína Nodal/genética , Fosforilación , Proproteína Convertasa 9/química , Proproteína Convertasa 9/deficiencia , Proproteína Convertasa 9/genética , Unión Proteica , Dominios Proteicos , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Proteína Smad2/metabolismo , Regulación hacia Arriba
3.
J Lipid Res ; 62: 100096, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34280453

RESUMEN

Proprotein convertase subtilisin/kexin type 9 (PCSK9) promotes lysosomal degradation of the LDL receptor and is a key regulator of cholesterol metabolism. After the liver, the small intestine is the second organ that highly expresses PCSK9. However, the small intestine's ability to secrete PCSK9 remains a matter of debate. While liver-specific PCSK9-deficient mice present no PCSK9 in systemic blood, human intestinal Caco-2 cells can actively secrete PCSK9. This raises the possibility for active intestinal secretion via the portal blood. Here, we aimed to determine whether enterocytes can secrete PCSK9 using in vitro, ex vivo, and in vivo approaches. We first observed that PCSK9 secretion from Caco-2 cells was biphasic and dependent on Caco-2 maturation status. Transcriptional analysis suggested that this transient reduction in PCSK9 secretion might be due to loss of SREBP2-mediated transcription of PCSK9. Consistently, PCSK9 secretion was not detected ex vivo in human or mouse intestinal biopsies mounted in Ussing chambers. Finally, direct comparison of systemic versus portal blood PCSK9 concentrations in WT or liver-specific PCSK9-deficient mice confirmed the inability of the small intestine to secrete PCSK9 into the portal compartment. Altogether, our data demonstrate that mature enterocytes do not secrete PCSK9 and reinforce the central role of the liver in the regulation of the concentration of circulating PCSK9 and consequently of cellular LDL receptors.


Asunto(s)
Proproteína Convertasa 9/metabolismo , Animales , Células CACO-2 , Diferenciación Celular , Células Cultivadas , Humanos , Intestino Delgado/citología , Intestino Delgado/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Proproteína Convertasa 9/sangre , Proproteína Convertasa 9/deficiencia
4.
J Lipid Res ; 62: 100091, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34118252

RESUMEN

Plasma LDL is produced from catabolism of VLDL and cleared from circulation mainly via the hepatic LDL receptor (LDLR). Proprotein convertase subtilisin/kexin type 9 (PCSK9) promotes LDLR degradation, increasing plasma LDL-C levels. Circulating PCSK9 is mainly secreted by the liver, whereas VLDL is exclusively secreted by hepatocytes. However, the mechanism regulating their secretion is not completely understood. Surfeit 4 (Surf4) is a cargo receptor localized in the ER membrane. It recruits cargos into coat protein complex II vesicles to facilitate their secretion. Here, we investigated the role of Surf4 in VLDL and PCSK9 secretion. We generated Surf4 liver-specific knockout mice and found that knockout of Surf4 did not affect PCSK9 secretion, whereas it significantly reduced plasma levels of cholesterol, triglyceride, and lipid-binding protein apolipoprotein B (apoB). In cultured human hepatocytes, Surf4 coimmunoprecipitated and colocalized with apolipoprotein B100, and Surf4 silencing reduced secretion of apolipoprotein B100. Furthermore, knockdown of Surf4 in LDLR knockout (Ldlr-/-) mice significantly reduced triglyceride secretion, plasma levels of apoB and non-HDL-C, and the development of atherosclerosis. However, Surf4 liver-specific knockout mice and Surf4 knockdown in Ldlr-/- mice displayed similar levels of liver lipids and plasma alanine aminotransferase activity as control mice, indicating that inhibition of Surf4 does not cause notable liver damage. Expression of stearoyl-CoA desaturase-1 was also reduced in the liver of these mice, suggesting a reduction in de novo lipogenesis. In summary, hepatic deficiency of Surf4 reduced VLDL secretion and the development of atherosclerosis but did not cause significant hepatic lipid accumulation or liver damage.


Asunto(s)
Aterosclerosis/metabolismo , Lipoproteínas VLDL/metabolismo , Proteínas de la Membrana/metabolismo , Animales , Células Cultivadas , Proteínas de la Membrana/deficiencia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proproteína Convertasa 9/deficiencia , Proproteína Convertasa 9/metabolismo , Receptores de LDL/deficiencia , Receptores de LDL/metabolismo
5.
Artículo en Inglés | MEDLINE | ID: mdl-33992809

RESUMEN

Proprotein convertase subtilisin/kexin type 9 (PCSK9) plays an important role in cholesterol homeostasis by promoting the degradation of the LDL receptor (LDLR). PCSK9 loss-of-function mutations are associated with increased fasting plasma glucose levels and slightly elevated risk of type 2-diabetes. Considering the known detrimental effects of cholesterol accumulation in ß-cell, and the widespread use of PCSK9 inhibitors to treat hypercholesterolemia, it is important to gain insight into the role of pancreatic PCSK9 in glucose homeostasis and ß-cell function. We generated the first ß-cell-specific KO of PCSK9 (ßKO). PCSK9 mRNA and protein expression were reduced by 48% and 78% in ßKO islets, respectively, indicating that ß-cells constitute a major site of PCSK9 expression. In islets, loss of ß-cell PCSK9 resulted in unchanged LDLR protein levels, but reduced LDLR mRNA, indicating that cholesterol internalization is enhanced and that ß-cell PCSK9 promotes LDLR degradation. In contrast, whole body PCSK9 KO mice exhibited 2-fold higher LDLR protein levels in islets and a stable expression of cholesterogenic genes. Whole body KO and ßKO mice presented normal glucose tolerance, insulin release in response to glucose load and insulin sensitivity. Ex vivo glucose-stimulated insulin secretion in presence or absence of fatty acids was similar in WT and KO islets. Like KO mice, individuals carrying loss-of-function PCSK9 variants may be protected from cholesterol-induced toxicity due to reduced circulating cholesterol levels. Using both whole body KO or ßKO models, our data demonstrate that PCSK9 deletion in mouse does not have any toxic effect on ß-cell function and glucose homeostasis.


Asunto(s)
Glucosa/metabolismo , Homeostasis , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Proproteína Convertasa 9/deficiencia , Proproteína Convertasa 9/genética , Animales , Activación Enzimática , Técnicas de Inactivación de Genes , Ratones
6.
Nature ; 588(7839): 693-698, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33177715

RESUMEN

Despite its success in achieving the long-term survival of 10-30% of treated individuals, immune therapy is still ineffective for most patients with cancer1,2. Many efforts are therefore underway to identify new approaches that enhance such immune 'checkpoint' therapy3-5 (so called because its aim is to block proteins that inhibit checkpoint signalling pathways in T cells, thereby freeing those immune cells to target cancer cells). Here we show that inhibiting PCSK9-a key protein in the regulation of cholesterol metabolism6-8-can boost the response of tumours to immune checkpoint therapy, through a mechanism that is independent of PCSK9's cholesterol-regulating functions. Deleting the PCSK9 gene in mouse cancer cells substantially attenuates or prevents their growth in mice in a manner that depends on cytotoxic T cells. It also enhances the efficacy of immune therapy that is targeted at the checkpoint protein PD1. Furthermore, clinically approved PCSK9-neutralizing antibodies synergize with anti-PD1 therapy in suppressing tumour growth in mouse models of cancer. Inhibiting PCSK9-either through genetic deletion or using PCSK9 antibodies-increases the expression of major histocompatibility protein class I (MHC I) proteins on the tumour cell surface, promoting robust intratumoral infiltration of cytotoxic T cells. Mechanistically, we find that PCSK9 can disrupt the recycling of MHC I to the cell surface by associating with it physically and promoting its relocation and degradation in the lysosome. Together, these results suggest that inhibiting PCSK9 is a promising way to enhance immune checkpoint therapy for cancer.


Asunto(s)
Inhibidores de Puntos de Control Inmunológico/farmacología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inmunoterapia/métodos , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Inhibidores de PCSK9 , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Neutralizantes/farmacología , Anticuerpos Neutralizantes/uso terapéutico , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Antígenos de Histocompatibilidad Clase I/inmunología , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Lisosomas/metabolismo , Ratones , Neoplasias/metabolismo , Neoplasias/patología , Proproteína Convertasa 9/deficiencia , Proproteína Convertasa 9/genética , Proproteína Convertasa 9/inmunología , Linfocitos T Citotóxicos/citología , Linfocitos T Citotóxicos/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Arterioscler Thromb Vasc Biol ; 40(9): 2084-2094, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32673528

RESUMEN

OBJECTIVE: Increased postprandial lipemia (PPL) is an independent risk factor for atherosclerotic cardiovascular diseases. PCSK9 (Proprotein convertase subtilisin kexin type 9) is an endogenous inhibitor of the LDLR (low-density lipoprotein receptor) pathway. We previously showed that PCSK9 inhibition in mice reduces PPL. However, the relative contribution of intracellular intestinal PCSK9 or liver-derived circulating PCSK9 to this effect is still unclear. Approach and Results: To address this issue, we generated the first intestine-specific Pcsk9-deficient (i-Pcsk9-/-) mouse model. PPL was measured in i-Pcsk9-/- as well as in wild-type and streptozotocin-induced diabetic mice following treatment with a PCSK9 monoclonal antibody (alirocumab). Blocking the circulating form of PCSK9 with alirocumab significantly reduced PPL, while overexpressing human PCSK9 in the liver of full Pcsk9-/- mice had the opposite effect. Alirocumab regulated PPL in a LDLR-dependent manner as this effect was abolished in Ldlr-/- mice. In contrast, i-Pcsk9-/- mice did not exhibit alterations in plasma lipid parameters nor in PPL. Finally, PPL was highly exacerbated by streptozotocin-induced diabetes mellitus in Pcsk9+/+ but not in Pcsk9-/- mice, an effect that was mimicked by the use of alirocumab in streptozotocin-treated Pcsk9+/+ mice. CONCLUSIONS: Taken together, our data demonstrate that PPL is significantly altered by full but not intestinal PCSK9 deficiency. Treatment with a PCSK9 monoclonal antibody mimics the effect of PCSK9 deficiency on PPL suggesting that circulating PCSK9 rather than intestinal PCSK9 is a critical regulator of PPL. These data validate the clinical relevance of PCSK9 inhibitors to reduce PPL, especially in patients with type 2 diabetes mellitus.


Asunto(s)
Diabetes Mellitus Experimental/sangre , Diabetes Mellitus Tipo 2/sangre , Hiperlipidemias/sangre , Intestinos/enzimología , Lípidos/sangre , Proproteína Convertasa 9/sangre , Animales , Anticuerpos Monoclonales Humanizados/farmacología , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/enzimología , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/enzimología , Hiperlipidemias/enzimología , Hiperlipidemias/genética , Hiperlipidemias/prevención & control , Hipolipemiantes/farmacología , Intestinos/efectos de los fármacos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Inhibidores de PCSK9 , Periodo Posprandial , Proproteína Convertasa 9/deficiencia , Proproteína Convertasa 9/genética , Receptores de LDL/genética , Receptores de LDL/metabolismo
8.
Curr Opin Lipidol ; 31(4): 182-186, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32487819

RESUMEN

PURPOSE OF REVIEW: Proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibition is a new strategy to reduce LDL cholesterol (LDL-C), that is currently pursued by mAbs. A promising novel approach to target PCSK9 is using small interfering RNAs to inhibit hepatic PCSK9 synthesis. The first small interfering RNA developed for this purpose is inclisiran. Here, we review its clinical trial data and potential impact on patient management. RECENT FINDINGS: Inclisiran achieves sustained, additional 50% LDL-C reduction in patients receiving background statin therapy. Resulting LDL-C changes can be maintained by an infrequent dosing regimen with twice per year injections, that appear safe and well tolerated. Thus far, inclisiran has been studied in patients with established cardiovascular disease, high-risk primary prevention and in patients with familial hypercholesterolemia. SUMMARY: High and very high-risk individuals may benefit from the additional LDL-C-lowering effect of inclisiran when added to current lipid-lowering therapies. Furthermore, the simple dosing regimen may improve the convenience for users and facilitate patient adherence to therapy. The safety and convenience of inclisiran may offer new opportunities for population health.


Asunto(s)
LDL-Colesterol/sangre , Proproteína Convertasa 9/genética , ARN Interferente Pequeño/genética , Animales , Humanos , Hiperlipoproteinemia Tipo II/sangre , Hiperlipoproteinemia Tipo II/genética , Hiperlipoproteinemia Tipo II/terapia , Proproteína Convertasa 9/biosíntesis , Proproteína Convertasa 9/deficiencia , Riesgo
9.
Bioorg Med Chem ; 28(6): 115344, 2020 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-32051094

RESUMEN

Proprotein convertase (PC) subtilisin kexin type 9 (PCSK9) inhibits the clearance of low density lipoprotein (LDL) cholesterol from plasma by directly interacting with the LDL receptor (LDLR). As the interaction promotes elevated plasma LDL cholesterol levels and a predisposition to cardiovascular disease (CVD), it has attracted much interest as a therapeutic target. While anti-PCSK9 monoclonal antibodies have been successful in the treatment of hypercholesteremia by decreasing CVD risk, their high cost and a requirement for injection have prohibited widespread use. The advent of an orally bioavailable small molecule inhibitor of the PCSK9-LDLR interaction is an attractive alternative, however efforts have been tempered as the binding interface is unfavourable for binding by small organic molecules. Despite its challenging nature, we report herein the discovery of compound 3f as a small molecule inhibitor of PCSK9. The kinase inhibitor nilotinib emerged from a computational screen that was applied to identify compounds that may bind to a cryptic groove within PCSK9 and proximal to the LDLR-binding interface. A subsequent in vitro PCSK9-LDLR binding assay established that nilotinib was a bona fide but modest inhibitor of the interaction (IC50 = 9.8 µM). Through multiple rounds of medicinal chemistry, 3f emerged as a lead-like molecule by demonstrating disruption of the PCSK9-LDLR interaction at nanomolar levels in vitro (IC50 = 537 nM) with no inhibitory activity (IC50 > 10 µM) against a small panel of kinases. Compound 3f restored LDL uptake by liver cells at sub-micromolar levels and demonstrated excellent bioavailability when delivered subcutaneously in mice. Most significantly, compound 3f lowered total cholesterol levels in the plasma of wild-type mice, thereby providing proof-of-concept that the notion of a small molecule inhibitor against PCSK9 is therapeutically viable.


Asunto(s)
Inhibidores de PCSK9 , Receptores de LDL/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Sitios de Unión/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos , Femenino , Células Hep G2 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Estructura Molecular , Proproteína Convertasa 9/deficiencia , Proproteína Convertasa 9/metabolismo , Receptores de LDL/metabolismo , Bibliotecas de Moléculas Pequeñas/síntesis química , Bibliotecas de Moléculas Pequeñas/química , Relación Estructura-Actividad
10.
J Lipid Res ; 60(11): 1946-1952, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31511398

RESUMEN

The pharmacologic inhibition of proprotein convertase subtilisin-kexin type 9 (PCSK9) lowers lipoprotein (a) [Lp(a)] concentrations. However, the impact of genetic PCSK9 loss-of-function variants (LOFVs) on Lp(a) is uncertain. We determined the association of PCSK9 LOFVs with Lp(a) measures among black adults. Genotyping for PCSK9 LOFVs was conducted in 10,196 black Reasons for Geographic and Racial Differences in Stroke study participants. Among 241 participants with and 723 randomly selected participants without PCSK9 LOFVs, Lp(a) concentations, apo(a) kringle IV (KIV) repeats (a proxy for isoform size), and oxidized phospholipid (OxPL) apoB levels were measured using validated methods. Median Lp(a) concentrations among participants with and without PCSK9 LOFVs were 63.2 and 80.4 nmol/l, respectively (P = 0.016). After adjusting for age, sex, estimated glomerular filtration rate, LDL cholesterol, and statin use, participants with versus without a PCSK9 LOFV had a lower median Lp(a) concentration [Δ = -18.8 nmol/l (95% CI: -34.2, -3.3)]. Median apo(a) isoform sizes were 24 and 23 KIV repeats (P = 0.12) among participants with and without PCSK9 LOFVs, respectively [Δ = 1.1 (95% CI: 0.2, 2.0) after adjustment]. Median OxPL-apoB levels among participants with and without PCSK9 LOFVs were 3.4 and 4.1 nM (P = 0.20), respectively [Δ = -1.2 nM (95% CI -2.4, -0.04) after adjustment]. Among black adults, PCSK9 LOFVs were associated with lower Lp(a) concentration and OxPL-apoB levels.


Asunto(s)
Negro o Afroamericano/genética , Lipoproteína(a)/genética , Mutación con Pérdida de Función/genética , Proproteína Convertasa 9/genética , Femenino , Humanos , Masculino , Persona de Mediana Edad , Fenotipo , Proproteína Convertasa 9/deficiencia , Proproteína Convertasa 9/metabolismo , Estados Unidos
11.
Circ Res ; 124(1): 38-51, 2019 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-30582457

RESUMEN

RATIONALE: Glucagon is a key hormone that regulates the adaptive metabolic responses to fasting. In addition to maintaining glucose homeostasis, glucagon participates in the regulation of cholesterol metabolism; however, the molecular pathways underlying this effect are incompletely understood. OBJECTIVE: We sought to determine the role of hepatic Gcgr (glucagon receptor) signaling in plasma cholesterol regulation and identify its underlying molecular mechanisms. METHODS AND RESULTS: We show that Gcgr signaling plays an essential role in LDL-C (low-density lipoprotein cholesterol) homeostasis through regulating the PCSK9 (proprotein convertase subtilisin/kexin type 9) levels. Silencing of hepatic Gcgr or inhibition of glucagon action increased hepatic and plasma PCSK9 and resulted in lower LDLR (LDL receptor) protein and increased plasma LDL-C. Conversely, treatment of wild-type (WT) mice with glucagon lowered LDL-C levels, whereas this response was abrogated in Pcsk9-/- and Ldlr-/- mice. Our gain- and loss-of-function studies identified Epac2 (exchange protein activated by cAMP-2) and Rap1 (Ras-related protein-1) as the downstream mediators of glucagon's action on PCSK9 homeostasis. Moreover, mechanistic studies revealed that glucagon affected the half-life of PCSK9 protein without changing the level of its mRNA, indicating that Gcgr signaling regulates PCSK9 degradation. CONCLUSIONS: These findings provide novel insights into the molecular interplay between hepatic glucagon signaling and lipid metabolism and describe a new posttranscriptional mechanism of PCSK9 regulation.


Asunto(s)
LDL-Colesterol/sangre , Metabolismo Energético , Glucagón/metabolismo , Hígado/metabolismo , Proproteína Convertasa 9/metabolismo , Animales , Línea Celular , Estabilidad de Enzimas , Glucagón/deficiencia , Glucagón/genética , Factores de Intercambio de Guanina Nucleótido/metabolismo , Semivida , Ratones Endogámicos C57BL , Ratones Noqueados , Proproteína Convertasa 9/deficiencia , Proproteína Convertasa 9/genética , Proteolisis , Receptores de Glucagón/metabolismo , Receptores de LDL/genética , Receptores de LDL/metabolismo , Transducción de Señal , Proteínas de Unión al GTP rap1/metabolismo
12.
Biol Chem ; 399(12): 1363-1374, 2018 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-30044755

RESUMEN

Proprotein convertase subtilisin/kexin type 9 (PCSK9) is a secreted protein that binds and escorts the low density lipoprotein receptor (LDLR) into the lysosomal degradation pathway. Prescribed monoclonal antibodies (mAbs) against PCSK9 prevent its binding to the LDLR, and result in ~60% lower LDL cholesterol (LDLc) levels. Although efficient, mAbs are expensive. Hence other PCSK9 inhibitors are needed. For screening purpose, we developed C57BL/6J mice expressing the human PCSK9 gene under the control of its own promoter, but lacking endogenous mouse PCSK9. All lines recapitulate the endogenous PCSK9 expression pattern. The Tg2 line that expresses physiological levels of human PCSK9 (hPCSK9) was selected to characterize the inhibitory properties of a previously reported single domain antibody (sdAb), PKF8-mFc, which binds the C-terminal domain of PCSK9. Upon intraveinous injection of 10 mg/kg, PKF8-mFc and the mAb evolocumab neutralized ~50% and 100% of the hPCSK9 impact on total cholesterol (TC) levels, respectively, but PKF8-mFc had a more sustained effect. PKF8-mFc barely affected hPCSK9 levels, whereas evolocumab promoted a 4-fold increase 3 days post-injection, suggesting very different inhibitory mechanisms. The present study also shows that the new transgenic mice are well suited to screen a variety of hPCSK9 inhibitors.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Cisteína/antagonistas & inhibidores , Histidina/antagonistas & inhibidores , Inhibidores de PCSK9 , Animales , Anticuerpos Monoclonales Humanizados , Cisteína/metabolismo , Genotipo , Histidina/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Proproteína Convertasa 9/deficiencia , Proproteína Convertasa 9/metabolismo
13.
Cardiovasc Res ; 114(13): 1738-1751, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-29800228

RESUMEN

Aims: Inhibition of proprotein convertase subtilisin/kexin type 9 (PCSK9) has emerged as a novel therapy to treat hypercholesterolaemia and related cardiovascular diseases. This study determined if PCSK9 can regulate infarct size, cardiac function, and autophagy during ischaemia. Methods and results: Mice hearts were subjected to left coronary artery (LCA) occlusion. There was intense expression of PCSK9 in the zone bordering the infarct area in association with marked cardiac contractile dysfunction in the wild-type mice. This region also revealed intense autophagy. To assess the role of PCSK9 in the evolution of infarct size and function and development of autophagy, we used wild-type mice pre-treated with two different PCSK9 inhibitors (Pep2-8 and EGF-A) or mice lacking PCSK9 gene. Both strategies resulted in smaller infarcts and improved cardiac function following LCA ligation. PCSK9 inhibition also markedly reduced autophagy. Relationship between myocardial ischaemia and PCSK9 expression and autophagy was examined in cultured mouse cardiomyocytes. Exposure of cardiomyocytes to hypoxia resulted in prompt PCSK9 expression and autophagy signals; both were blocked by HIF-1α siRNA. Further, treatment of cardiomyocytes with recombinant PCSK9 during hypoxia induced, and treatment with PCSK9 siRNA reduced, autophagy suggesting a possible role of PCSK9 in the determination of autophagy. Other studies revealed activation of ROS-ATM-LKB1-AMPK axis as a possible mechanism of PCSK-induced autophagy. Hearts of humans with recent infarcts also showed expression of PCSK9 and autophagy in the border zone-similar to that in the infarcted mouse heart. Conclusion: PCSK9 is up-regulated in the ischaemic hearts and determines development of infarct size, cardiac function, and autophagy.


Asunto(s)
Autofagia , Contracción Miocárdica , Infarto del Miocardio/enzimología , Miocardio/enzimología , Proproteína Convertasa 9/metabolismo , Función Ventricular Izquierda , Anciano , Animales , Autofagia/efectos de los fármacos , Proteínas Relacionadas con la Autofagia/metabolismo , Hipoxia de la Célula , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Mediadores de Inflamación/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Contracción Miocárdica/efectos de los fármacos , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Infarto del Miocardio/prevención & control , Miocardio/patología , Inhibidores de PCSK9 , Proproteína Convertasa 9/deficiencia , Proproteína Convertasa 9/genética , Interferencia de ARN , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptores de LDL/genética , Receptores de LDL/metabolismo , Inhibidores de Serina Proteinasa/farmacología , Transducción de Señal , Función Ventricular Izquierda/efectos de los fármacos
14.
Cardiovasc Res ; 114(8): 1145-1153, 2018 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-29617722

RESUMEN

Aims: Proprotein convertase subtilisin/kexin type 9 (PCSK9) has been shown to influence macrophage biology and modulate atherogenesis. We conducted this study to examine the regulation of scavenger receptors (SRs) (LOX-1, SRA, and CD36) and oxidized liporoptein cholesterol (ox-LDL) uptake in macrophages by PCSK9. Methods and results: Treatment of mouse peritoneal macrophages with tumour necrosis factor alpha (TNF-α) resulted in concentration-dependent modest, but significant, increase in PCSK9 expression. Importantly, treatment of TNF-α primed macrophages with recombinant murine PCSK9 increased the expression of LOX-1, SRA, and CD36 2-5 fold, and enhanced ox-LDL uptake by ≈five-fold. The increase in LOX-1 was much greater than in SRA or CD36. PCSK9 inhibition (by siRNA transfection or use of macrophages from PCSK9-/- mice) reduced the expression of SRs (LOX-1 ≫ SRA or CD36). Ox-LDL uptake in response to PCSK9 was also inhibited in macrophages from LOX-1-/- mice (P < 0.05 vs. macrophages from SRA-/- and CD36-/- mice). Upregulation of PCSK9 by cDNA transfection induced intense ox-LDL uptake which was inhibited by co-transfection of cells with siRNA LOX-1 (P < 0.05 vs. siRNA SRA or siRNA CD36). Further, TNF-α-mediated PCSK9 upregulation and subsequent expression of SRs and ox-LDL uptake were reduced in macrophages from gp91phox-/-, p47phox-/- and p22phox-/- mice (vs. macrophages from wild-type mice). Conclusions: This study shows that in an inflammatory milieu, elevated levels of PCSK9 potently stimulate the expression of SRs (principally LOX-1) and ox-LDL uptake in macrophages, and thus contribute to the process of atherogenesis.


Asunto(s)
Aterosclerosis/enzimología , Lipoproteínas LDL/metabolismo , Macrófagos Peritoneales/enzimología , Proproteína Convertasa 9/metabolismo , Receptores Depuradores de Clase E/metabolismo , Animales , Aterosclerosis/genética , Aterosclerosis/patología , Transporte Biológico , Antígenos CD36/genética , Antígenos CD36/metabolismo , Células Cultivadas , Grupo Citocromo b/genética , Grupo Citocromo b/metabolismo , Macrófagos Peritoneales/efectos de los fármacos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , NADPH Oxidasas/genética , NADPH Oxidasas/metabolismo , Proproteína Convertasa 9/deficiencia , Proproteína Convertasa 9/genética , Especies Reactivas de Oxígeno/metabolismo , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo , Receptores Depuradores de Clase A/genética , Receptores Depuradores de Clase A/metabolismo , Receptores Depuradores de Clase E/deficiencia , Receptores Depuradores de Clase E/genética , Factor de Necrosis Tumoral alfa/farmacología
15.
J Infect Dis ; 217(4): 667-680, 2018 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-29294034

RESUMEN

Periodontitis is a highly prevalent infectious disease associated genetically with coronary heart disease (CHD). The effects of proprotein convertase subtilisin/kexin type 9 (PCSK9), a critical regulator of CHD, on periodontitis have not been studied to date. Here, we found that PCSK9 expression was increased in periodontitis patients and Porphyromonas gingivalis (Pg)-infected mice. Loss of PCSK9 attenuated Pg-induced periodontal bone loss in mice. First, PCSK9 deficiency reduced the release of inflammation-associated cytokines, such as tumor necrosis factor alpha (TNF-α) and interleukin 1ß, in vitro and in vivo. Second, its deficiency enhanced Pg and endotoxin clearance during Pg invasion in part by upregulating CD36 and low-density lipoprotein receptor (LDLR), respectively. However, after berberine treatment, periodontal bone regeneration in the PCSK9 knockout group was significantly lower than that in wild-type. This was because PCSK9 overexpression promoted osteogenic differentiation of periodontal ligament stem cells (PDLCs) prechallenged by TNF-α. Furthermore, PCSK9 could rescue PDLC osteogenesis by repressing the NF-κB signaling pathway by interacting with TRAF2. These results suggest that PCSK9 may be a potent drug target for treating periodontitis.


Asunto(s)
Infecciones por Bacteroidaceae/patología , Periodontitis/patología , Proproteína Convertasa 9/sangre , Adulto , Cuidados Posteriores , Animales , Berberina/administración & dosificación , Resorción Ósea/patología , Citocinas/sangre , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Porphyromonas gingivalis/crecimiento & desarrollo , Proproteína Convertasa 9/deficiencia , Adulto Joven
16.
J Lipid Res ; 59(2): 207-223, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29180444

RESUMEN

Proprotein convertase subtilisin/kexin type 9 (PCSK9) interacts directly with cytoplasmic apoB and prevents its degradation via the autophagosome/lysosome pathway. This process affects VLDL and LDL production and influences atherogenesis. Here, we investigated the molecular machinery by which PCSK9 modulates autophagy and affects atherogenesis. We backcrossed Pcsk9-/- mice with atherosclerosis-prone Ldlr-/-Apobec1-/- (LDb) mice to generate Ldlr-/-Apobec1-/-Pcsk9-/- (LTp) mice. Deletion of PCSK9 resulted in decreased hepatic apoB secretion, increased autophagic flux, and decreased plasma levels of IDL and LDL particles. The LDLs from LTp mice (LTp-LDLs) were less atherogenic and contained less cholesteryl ester and phospholipids than LDb-LDLs. Moreover LTp-LDLs induced lower endothelial expression of the genes encoding TLR2, Lox-1, ICAM-1, CCL2, CCL7, IL-6, IL-1ß, Beclin-1, p62, and TRAF6 Collectively, these effects were associated with substantially less atherosclerosis development (>4-fold) in LTp mice. The absence of PCSK9 in LDb mice results in decreased lipid and apoB levels, fewer atherogenic LDLs, and marked reduction of atherosclerosis. The effect on atherogenesis may be mediated in part by the effects of modified LDLs on endothelial cell receptors and proinflammatory and autophagy molecules. These findings suggest that there may be clinical benefits of PCSK9 inhibition due to mechanisms unrelated to increased LDL receptor activity.


Asunto(s)
Apolipoproteínas B/metabolismo , Aterosclerosis/metabolismo , Endotelio Vascular/metabolismo , Proproteína Convertasa 9/metabolismo , Animales , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proproteína Convertasa 9/deficiencia
17.
Sci Rep ; 7(1): 14360, 2017 10 30.
Artículo en Inglés | MEDLINE | ID: mdl-29084995

RESUMEN

The effect of lipid lowering on the incidence of deep venous thrombosis (DVT) is controversial. The purpose of this study was to determine the effect of proprotein convertase subtilisin/kexin type 9 (PCSK9) deficiency on development of DVT in mice. Pcsk9 deficient (pcsk9 -/-) and wild-type (WT) littermates underwent partial inferior vena cava (IVC) ligation to induce venous thrombosis. 48 hours following IVC ligation, IVC thrombosis was evident in 60% of WT mice and 25% of pcsk9 -/- mice (p < 0.05). Analysis of IVC thrombosis revealed greater thrombus weight, length, myeloid cell recruitment, and more neutrophil extracellular trap formation (NETs) in WT compared to pcsk9 -/- mice. Intravital microscopy performed two hours following partial IVC ligation revealed that leukocyte firm attachment was increased in WT mice compared to mice undergoing a sham operation, however leukocyte attachment was reduced in pcsk9 -/- mice compared to WT mice. In conclusion, deficiency of PCSK9 is associated with protection from venous thrombosis. This protection is associated with reduced leukocyte recruitment and NET formation at the site of thrombosis.


Asunto(s)
Proproteína Convertasa 9/fisiología , Trombosis de la Vena/prevención & control , Animales , HDL-Colesterol/sangre , HDL-Colesterol/efectos de los fármacos , LDL-Colesterol/sangre , LDL-Colesterol/efectos de los fármacos , Metabolismo de los Lípidos/fisiología , Lípidos/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proproteína Convertasa 9/deficiencia , Proproteína Convertasa 9/metabolismo , Proproteína Convertasas , Serina Endopeptidasas , Subtilisinas , Trombosis de la Vena/metabolismo
18.
Eur J Prev Cardiol ; 24(17): 1870-1877, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28758421

RESUMEN

Background Proprotein convertase subtilisin kexin type 9 (PCSK9) regulates low-density lipoprotein and very low-density lipoprotein receptor expression in several tissues. Here we evaluated whether PCSK9 may modulate the handling of triglycerides in the liver and peripheral tissues. Methods Subjects from the PLIC cohort were genotyped for the loss-of-function PCSK9 R46L variant and characterized for clinical and biochemical parameters, total and android fat mass, hepatic steatosis and epicardial fat thickness. Visceral adipose tissue and subcutaneous adipose tissue in PCSK9 KO and wild type mice were quantified by nuclear magnetic resonance imaging. Results Carriers of the R46L variant ( n = 13) had lower low-density lipoprotein cholesterol levels, higher body mass index and increased percentage of total and android fat masses compared with non-carriers ( n = 521). R46L variant associated with a two-fold increase prevalence of hepatic steatosis and higher epicardial fat thickness. These observations were replicated in PCSK9 KO mice, which showed increased visceral adipose tissue (but not subcutaneous adipose tissue) when fed chow or high-fat diet for 20 weeks, compared with wild type mice. Conclusions These data suggest that genetically determined PCSK9 deficiency might be associated with ectopic fat accumulation.


Asunto(s)
Tejido Adiposo/fisiopatología , Adiposidad , Proproteína Convertasa 9/deficiencia , Tejido Adiposo/diagnóstico por imagen , Tejido Adiposo/metabolismo , Adiposidad/genética , Animales , Índice de Masa Corporal , LDL-Colesterol/sangre , Genotipo , Humanos , Italia , Mutación con Pérdida de Función , Masculino , Ratones Noqueados , Fenotipo , Proproteína Convertasa 9/genética , Factores de Tiempo
19.
Circulation ; 134(1): 61-72, 2016 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-27358438

RESUMEN

BACKGROUND: In nephrotic syndrome, damage to the podocytes of the kidney produces severe hypercholesterolemia for which novel treatments are urgently needed. PCSK9 (proprotein convertase subtilisin/kexin type 9) has emerged as an important regulator of plasma cholesterol levels and therapeutic target. Here, we tested the role of PCSK9 in mediating the hypercholesterolemia of nephrotic syndrome. METHODS: PCSK9 and plasma lipids were studied in nephrotic syndrome patients before and after remission of disease, mice with genetic ablation of the podocyte (Podocyte Apoptosis Through Targeted Activation of Caspase-8, Pod-ATTAC mice) and mice treated with nephrotoxic serum (NTS), which triggers immune-mediated podocyte damage. In addition, mice with hepatic deletion of Pcsk9 were treated with NTS to determine the contribution of PCSK9 to the dyslipidemia of nephrotic syndrome. RESULTS: Patients with nephrotic syndrome showed a decrease in plasma cholesterol and plasma PCSK9 on remission of their disease (P<0.05, n=47-50). Conversely, Pod-ATTAC mice and NTS-treated mice showed hypercholesterolemia and a 7- to 24-fold induction in plasma PCSK9. The induction of plasma PCSK9 appeared to be attributable to increased secretion of PCSK9 from the hepatocyte coupled with decreased clearance. Interestingly, knockout of Pcsk9ameliorated the effects of NTS on plasma lipids. Thus, in the presence of NTS, mice lacking hepatic Pcsk9 showed a 40% to 50% decrease in plasma cholesterol and triglycerides. Moreover, the ability of NTS treatment to increase the percentage of low-density lipoprotein-associated cholesterol (from 9% in vehicle-treated Flox mice to 47% after NTS treatment), was lost in mice with hepatic deletion of Pcsk9 (5% in both the presence and absence of NTS). CONCLUSIONS: Podocyte damage triggers marked inductions in plasma PCSK9, and knockout of Pcsk9 ameliorates dyslipidemia in a mouse model of nephrotic syndrome. These data suggest that PCSK9 inhibitors may be beneficial in patients with nephrotic syndrome-associated hypercholesterolemia.


Asunto(s)
Hipercolesterolemia/etiología , Síndrome Nefrótico/complicaciones , Proproteína Convertasa 9/fisiología , Animales , Humanos , Hipercolesterolemia/enzimología , Lípidos/sangre , Hígado/enzimología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Síndrome Nefrótico/sangre , Síndrome Nefrótico/enzimología , Podocitos/patología , Proproteína Convertasa 9/deficiencia , Proproteína Convertasa 9/genética , Proproteína Convertasa 9/uso terapéutico , Proteínas Recombinantes/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...