Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 682
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Cell ; 175(1): 133-145.e15, 2018 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-30220454

RESUMEN

Nonalcoholic fatty liver disease (NAFLD) progresses to nonalcoholic steatohepatitis (NASH) in response to elevated endoplasmic reticulum (ER) stress. Whereas the onset of simple steatosis requires elevated de novo lipogenesis, progression to NASH is triggered by accumulation of hepatocyte-free cholesterol. We now show that caspase-2, whose expression is ER-stress inducible and elevated in human and mouse NASH, controls the buildup of hepatic-free cholesterol and triglycerides by activating sterol regulatory element-binding proteins (SREBP) in a manner refractory to feedback inhibition. Caspase-2 colocalizes with site 1 protease (S1P) and cleaves it to generate a soluble active fragment that initiates SCAP-independent SREBP1/2 activation in the ER. Caspase-2 ablation or pharmacological inhibition prevents diet-induced steatosis and NASH progression in ER-stress-prone mice. Caspase-2 inhibition offers a specific and effective strategy for preventing or treating stress-driven fatty liver diseases, whereas caspase-2-generated S1P proteolytic fragments, which enter the secretory pathway, are potential NASH biomarkers.


Asunto(s)
Caspasa 2/fisiología , Lipogénesis/fisiología , Proproteína Convertasas/fisiología , Serina Endopeptidasas/fisiología , Animales , Colesterol/metabolismo , Retículo Endoplásmico/fisiología , Estrés del Retículo Endoplásmico/fisiología , Hígado Graso/fisiopatología , Células HEK293 , Hepatocitos/metabolismo , Humanos , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Proproteína Convertasas/metabolismo , Serina Endopeptidasas/metabolismo , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Triglicéridos/metabolismo
2.
Development ; 150(23)2023 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-38032088

RESUMEN

Heart development is a complex process that requires asymmetric positioning of the heart, cardiac growth and valve morphogenesis. The mechanisms controlling heart morphogenesis and valve formation are not fully understood. The pro-convertase FurinA functions in heart development across vertebrates. How FurinA activity is regulated during heart development is unknown. Through computational analysis of the zebrafish transcriptome, we identified an RNA motif in a variant FurinA transcript harbouring a long 3' untranslated region (3'UTR). The alternative 3'UTR furina isoform is expressed prior to organ positioning. Somatic deletions in the furina 3'UTR lead to embryonic left-right patterning defects. Reporter localisation and RNA-binding assays show that the furina 3'UTR forms complexes with the conserved RNA-binding translational repressor, Ybx1. Conditional ybx1 mutant embryos show premature and increased Furin reporter expression, abnormal cardiac morphogenesis and looping defects. Mutant ybx1 hearts have an expanded atrioventricular canal, abnormal sino-atrial valves and retrograde blood flow from the ventricle to the atrium. This is similar to observations in humans with heart valve regurgitation. Thus, the furina 3'UTR element/Ybx1 regulon is important for translational repression of FurinA and regulation of heart development.


Asunto(s)
Regulón , Pez Cebra , Animales , Humanos , Regiones no Traducidas 3' , Regulón/genética , Morfogénesis/genética , Válvulas Cardíacas , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo , Proproteína Convertasas/genética , Proproteína Convertasas/metabolismo
3.
EMBO Rep ; 25(3): 951-970, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38287192

RESUMEN

The exquisite specificity of antibodies can be harnessed to effect targeted degradation of membrane proteins. Here, we demonstrate targeted protein removal utilising a protein degradation domain derived from the endogenous human protein Proprotein Convertase Subtilisin/Kexin type 9 (PCSK9). Recombinant antibodies genetically fused to this domain drive the degradation of membrane proteins that undergo constitutive internalisation and recycling, including the transferrin receptor and the human cytomegalovirus latency-associated protein US28. We term this approach PACTAC (PCSK9-Antibody Clearance-Targeting Chimeras).


Asunto(s)
Proproteína Convertasa 9 , Serina Endopeptidasas , Humanos , Proproteína Convertasa 9/metabolismo , Proproteína Convertasas/metabolismo , Proteínas de la Membrana , Receptores de LDL/metabolismo
4.
PLoS Genet ; 19(9): e1010944, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37721936

RESUMEN

Some types of collagens, including transmembrane MACIT collagens and C. elegans cuticle collagens, are N-terminally cleaved at a dibasic site that resembles the consensus for furin or other proprotein convertases of the subtilisin/kexin (PCSK) family. Such cleavage may release transmembrane collagens from the plasma membrane and affect extracellular matrix assembly or structure. However, the functional consequences of such cleavage are unclear and evidence for the role of specific PCSKs is lacking. Here, we used endogenous collagen fusions to fluorescent proteins to visualize the secretion and assembly of the first collagen-based cuticle in C. elegans and then tested the role of the PCSK BLI-4 in these processes. Unexpectedly, we found that cuticle collagens SQT-3 and DPY-17 are secreted into the extraembryonic space several hours before cuticle matrix assembly. Furthermore, this early secretion depends on BLI-4/PCSK; in bli-4 and cleavage-site mutants, SQT-3 and DPY-17 are not efficiently secreted and instead form large intracellular puncta. Their later assembly into cuticle matrix is reduced but not entirely blocked. These data reveal a role for collagen N-terminal processing in intracellular trafficking and the control of matrix assembly in vivo. Our observations also prompt a revision of the classic model for C. elegans cuticle matrix assembly and the pre-cuticle-to-cuticle transition, suggesting that cuticle layer assembly proceeds via a series of regulated steps and not simply by sequential secretion and deposition.


Asunto(s)
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Subtilisina , Animales , Secuencia de Aminoácidos , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Colágeno/genética , Colágeno/metabolismo , Proproteína Convertasas/genética , Proproteína Convertasas/metabolismo , Subtilisina/genética , Subtilisina/metabolismo
5.
Arterioscler Thromb Vasc Biol ; 44(2): 328-333, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-38059350

RESUMEN

The tremendous burden of lipid metabolism diseases, coupled with recent developments in human somatic gene editing, has motivated researchers to propose population-wide somatic gene editing of PCSK9 (proprotein convertase subtilisin/kexin type 9) within the livers of otherwise healthy humans. The best-characterized molecular function of PCSK9 is its ability to regulate plasma LDL (low-density lipoprotein) levels through promoting LDL receptor degradation. Individuals with loss-of-function PCSK9 variants have lower levels of plasma LDL and reduced cardiovascular disease. Gain-of-function variants of PCSK9 are strongly associated with familial hypercholesterolemia. A new therapeutic strategy delivers CRISPR/Cas9 (clustered regularly interspaced short palindromic repeats; CRISPR-associated protein 9) specifically to liver cells to edit the wild-type alleles of PCSK9 with the goal of producing a loss-of-function allele. This direct somatic gene editing approach is being pursued despite the availability of US Food and Drug Administration-approved PCSK9 inhibitors that lower plasma LDL levels. Here, we discuss other characterized functions of PCSK9 including its role in infection and host immunity. We explore important factors that may have contributed to the evolutionary selection of PCSK9 in several vertebrates, including humans. Until such time that more fully understand the multiple biological roles of PCSK9, the ethics of permanently editing the gene locus in healthy, wild-type populations remains highly questionable.


Asunto(s)
Proproteína Convertasa 9 , Proproteína Convertasas , Animales , Humanos , Proproteína Convertasa 9/genética , Proproteína Convertasa 9/metabolismo , Proproteína Convertasas/genética , Proproteína Convertasas/metabolismo , Serina Endopeptidasas/genética , Alelos , Receptores de LDL/genética
6.
Arterioscler Thromb Vasc Biol ; 43(2): 352-358, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36475702

RESUMEN

BACKGROUND: PCSK9 (proprotein convertase subtilisin-kexin type 9) chaperones the hepatic LDLR (low-density lipoprotein receptor) for lysosomal degradation, elevating serum LDL (low-density lipoprotein) cholesterol and promoting atherosclerotic heart disease. Though the major effect on the hepatic LDLR comes from secreted PCSK9, the details of PCSK9 reuptake into the hepatocyte remain unclear. In both tissue culture and animal models, HSPGs (heparan sulfate proteoglycans) on hepatocytes act as co-receptors to promote PCSK9 reuptake. We hypothesized that if this PCSK9:HSPG interaction is important in humans, disrupting it with unfractionated heparin (UFH) would acutely displace PCSK9 from the liver and increase plasma PCSK9. METHODS: We obtained remnant plasma samples from 160 subjects undergoing cardiac catheterization before and after administration of intravenous UFH. PCSK9 levels were determined using a commercial enzyme-linked immunosorbent assay. RESULTS: Median plasma PCSK9 was 113 ng/mL prior to UFH and 119 ng/mL afterward. This difference was not significant (P=0.83 [95% CI, -6.23 to 6.31 ng/mL]). Equivalence testing provided 95% confidence that UFH would not raise plasma PCSK9 by > 4.7%. Among all subgroups, only subjects with the lowest baseline PCSK9 concentrations exhibited a response to UFH (8.8% increase, adj. P=0.044). A modest correlation was observed between baseline plasma PCSK9 and the change in plasma PCSK9 due to UFH (RS=-0.3634; P<0.0001). CONCLUSIONS: Administration of UFH does not result in a clinically meaningful effect on circulating PCSK9 among an unselected population of humans. The results cast doubt on the clinical utility of disrupting the PCSK9:HSPG interaction as a general therapeutic strategy for PCSK9 inhibition. However, the observations suggest that in selected populations, disrupting the PCSK9:HSPG interaction could still affect PCSK9 reuptake and offer a therapeutic benefit.


Asunto(s)
Heparina , Proproteína Convertasa 9 , Animales , Humanos , Proproteína Convertasa 9/metabolismo , Serina Endopeptidasas , Proproteína Convertasas/metabolismo , Proteoglicanos de Heparán Sulfato , Receptores de LDL/metabolismo , LDL-Colesterol , Subtilisinas
7.
Arterioscler Thromb Vasc Biol ; 43(7): 1219-1233, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37165876

RESUMEN

BACKGROUND: Lower plasma levels of LDL (low-density lipoprotein) cholesterol (LDL-C) can reduce the risk of atherosclerotic cardiovascular disease. The loss-of-function mutations in PCSK9 (proprotein convertase subtilisin/kexin type 9) have been known to associate with low LDL-C in many human populations. PCSK9 genetic variants in Chinese Uyghurs who are at high risk of atherosclerotic cardiovascular disease due to their dietary habits have not been reported. METHODS: The study involved the whole-exome and target sequencing of college students from Uyghur and other ethnic groups in Xinjiang, China, for the association of PCSK9 loss-of-function mutations with low plasma levels of LDL-C. The mechanisms by which the identified mutations affect the function of PCSK9 were investigated in cultured cells using biochemical and cell assays. The causal effects of the identified PCSK9 mutations on LDL-C levels were verified in mice injected with adeno-associated virus expressing different forms of PCSK9 and fed a high-cholesterol diet. RESULTS: We identified 2 PCSK9 mutations-E144K and C378W-in Chinese Uyghurs with low plasma levels of LDL-C. The E144K and C378W mutations impaired the maturation and secretion of the PCSK9 protein, respectively. Adeno-associated virus-mediated expression of E144K and C378W mutants in Pcsk9 KO (knockout) mice fed a high-cholesterol diet also hampered PCSK9 secretion into the serum, resulting in elevated levels of LDL receptor in the liver and reduced levels of LDL-C in the serum. CONCLUSIONS: Our study shows that E144K and C378W are PCSK9 loss-of-function mutations causing low LDL-C levels in mice and probably in humans as well.


Asunto(s)
Aterosclerosis , Enfermedades Cardiovasculares , Hipercolesterolemia , Humanos , Ratones , Animales , Proproteína Convertasa 9/genética , LDL-Colesterol , Serina Endopeptidasas/genética , Proproteína Convertasas/genética , Proproteína Convertasas/metabolismo , Receptores de LDL/genética , Receptores de LDL/metabolismo , Ratones Noqueados , Aterosclerosis/genética , Aterosclerosis/prevención & control , Aterosclerosis/metabolismo , Mutación
8.
Gastric Cancer ; 27(2): 292-307, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38280128

RESUMEN

BACKGROUND: Gastric cancer (GC), the fourth leading cause of cancer-related death worldwide, with most deaths caused by advanced and metastatic disease, has limited curative options. Here, we revealed the importance of proprotein convertases (PCs) in the malignant and metastatic potential of GC cells through the regulation of the YAP/TAZ/TEAD pathway and epithelial-to-mesenchymal transition (EMT) in cancer stem cells (CSC). METHODS: The general PCs inhibitor, decanoyl-RVKR-chloromethyl-ketone (CMK), was used to repress PCs activity in CSCs of various GC cell lines. Their tumorigenic properties, drug resistance, YAP/TAZ/TEAD pathway activity, and invasive properties were then investigated in vitro, and their metastatic properties were explored in a mouse xenograft model. The prognostic value of PCs in GC patients was also explored in molecular databases of GC. RESULTS: Inhibition of PCs activity in CSCs in all GC cell lines reduced tumorsphere formation and growth, drug efflux, EMT phenotype, and invasive properties that are associated with repressed YAP/TAZ/TEAD pathway activity in vitro. In vivo, PCs' inhibition in GC cells reduced their metastatic spread. Molecular analysis of tumors from GC patients has highlighted the prognostic value of PCs. CONCLUSIONS: PCs are overexpressed in GC and associated with poor prognosis. PCs are involved in the malignant and metastatic potential of CSCs via the regulation of EMT, the YAP/TAZ/TEAD oncogenic pathway, and their stemness and invasive properties. Their repression represents a new strategy to target CSCs and impair metastatic spreading in GC.


Asunto(s)
Neoplasias Gástricas , Factores de Transcripción , Humanos , Animales , Ratones , Factores de Transcripción/genética , Proteínas Señalizadoras YAP , Neoplasias Gástricas/patología , Modelos Animales de Enfermedad , Proproteína Convertasas/metabolismo , Células Madre Neoplásicas/metabolismo
9.
Proc Natl Acad Sci U S A ; 118(32)2021 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-34349020

RESUMEN

Family with sequence similarity 20C (Fam20C), the major protein kinase in the secretory pathway, generates the vast majority of the secreted phosphoproteome. However, the regulatory mechanisms of Fam20C transport, secretion, and function remain largely unexplored. Here, we show that Fam20C exists as a type II transmembrane protein within the secretory compartments, with its N-terminal signal peptide-like region serving as a membrane anchor for Golgi retention. The secretion and kinase activity of Fam20C are governed by site-1 protease (S1P), a key regulator of cholesterol homeostasis. We find that only mature Fam20C processed by S1P functions in osteoblast differentiation and mineralization. Together, our findings reveal a unique mechanism for Fam20C secretion and activation via proteolytic regulation, providing a molecular link between biomineralization and lipid metabolism.


Asunto(s)
Quinasa de la Caseína I/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Proproteína Convertasas/metabolismo , Serina Endopeptidasas/metabolismo , Secuencias de Aminoácidos , Animales , Vesículas Cubiertas por Proteínas de Revestimiento/metabolismo , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Quinasa de la Caseína I/genética , Diferenciación Celular/efectos de los fármacos , Proteínas de la Matriz Extracelular/genética , Aparato de Golgi/metabolismo , Células HeLa , Humanos , Ratones , Mutación , Osteoblastos/citología , Osteoblastos/metabolismo , Proproteína Convertasas/antagonistas & inhibidores , Proproteína Convertasas/genética , Dominios Proteicos , Transporte de Proteínas , Pirrolidinas/farmacología , Vías Secretoras , Serina Endopeptidasas/genética
10.
J Allergy Clin Immunol ; 151(5): 1379-1390.e11, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36623776

RESUMEN

BACKGROUND: Oncostatin M (OSM) may promote type 2 inflammation in chronic rhinosinusitis with nasal polyps (CRSwNP) by inducing thymic stromal lymphopoietin (TSLP). OBJECTIVE: We sought to study the impact of OSM on TSLP synthesis and release from nasal epithelial cells (NECs). METHODS: OSM receptors, IL-4 receptors (IL-4R), and TSLP were evaluated in mucosal tissue and primary NECs from patients with CRSwNP by quantitative PCR and immunofluorescence. Air-liquid interface-cultured NECs were stimulated with cytokines, including OSM, and quantitative PCR, ELISA, Western blot, and flow cytometry were used to assess the expression of OSM receptors, IL-4R, and TSLP. RESULTS: Increased levels of OSM receptor ß chain (OSMRß), IL-4Rα, and TSLP were observed in nasal polyp tissues and primary epithelial cells from nasal polyps of patients with CRSwNP compared with control tissues or cells from control subjects. The level of expression of OSMRß in tissue was correlated with levels of both IL-4Rα and TSLP. OSM stimulation of NECs increased the expression of OSMRß and IL-4Rα. Stimulation with IL-4 plus OSM augmented the production of TSLP; the response was suppressed by a signal transducer and activator of transcription 6 inhibitor. Stimulation of NECs with IL-4 plus OSM increased the expression of proprotein convertase subtilisin/kexin 3, an enzyme that truncates and activates TSLP. CONCLUSIONS: OSM increases the expression of IL-4Rα and synergizes with IL-4 to induce the synthesis and release of TSLP in NECs. Because the combination of IL-4 and OSM also augmented the expression of proprotein convertase subtilisin/kexin 3, these results suggest that OSM can induce both synthesis and posttranslational processing/activation of TSLP, promoting type 2 inflammation.


Asunto(s)
Interleucina-4 , Pólipos Nasales , Oncostatina M , Rinitis , Sinusitis , Humanos , Enfermedad Crónica , Citocinas/metabolismo , Inflamación/metabolismo , Interleucina-4/metabolismo , Mucosa Nasal/metabolismo , Pólipos Nasales/metabolismo , Oncostatina M/metabolismo , Proproteína Convertasas/metabolismo , Rinitis/metabolismo , Sinusitis/metabolismo , Subtilisinas/metabolismo , Linfopoyetina del Estroma Tímico
11.
Mar Drugs ; 21(4)2023 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-37103355

RESUMEN

Metastatic castration-resistant prostate cancer (mCRPC) cells can de novo biosynthesize their own cholesterol and overexpress proprotein convertase subtilisin/kexin type 9 (PCSK9). PCSK9 proved to contribute to mCRPC cell motility since PCSK9 knockdown (KD) in mCRPC CWR-R1ca cells led to notable reductions in cell migration and colony formation. Human tissue microarray results proved a higher immunohistoscore in patients ≥ 65 years old, and PCSK9 proved to be expressed higher at an early Gleason score of ≤7. The fermentation product pseurotin A (PS) suppressed PCSK9 expression, protein-protein interactions with LDLR, and breast and prostate cancer recurrences. PS suppressed migration and colony formation of the CWR-R1ca cells. The progression and metastasis of the CWR-R1ca-Luc cells subcutaneously (sc) xenografted into male nude mice fed a high-fat diet (HFD, 11% fat content) showed nearly 2-fold tumor volume, metastasis, serum cholesterol, low-density lipoprotein cholesterol (LDL-C), prostate-specific antigen (PSA), and PCSK9 levels versus mice fed a regular chow diet. Daily oral PS 10 mg/kg treatments prevented the locoregional and distant tumor recurrence of CWR-R1ca-Luc engrafted into nude mice after primary tumor surgical excision. PS-treated mice showed a significant reduction in serum cholesterol, LDL-C, PCSK9, and PSA levels. These results comprehensively validate PS as an mCRPC recurrence-suppressive lead by modulating the PCSK9-LDLR axis.


Asunto(s)
Proproteína Convertasa 9 , Neoplasias de la Próstata Resistentes a la Castración , Humanos , Masculino , Ratones , Animales , Anciano , Proproteína Convertasa 9/genética , Proproteína Convertasa 9/metabolismo , Ratones Desnudos , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Serina Endopeptidasas/metabolismo , Proproteína Convertasas/metabolismo , Antígeno Prostático Específico , Receptores de LDL/genética , Receptores de LDL/metabolismo , Colesterol
12.
Molecules ; 28(3)2023 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-36771126

RESUMEN

The proprotein convertase subtilisin kexin type 9 (PCSK9) emerged as a molecular target of great interest for the management of cardiovascular disorders due to its ability to reduce low density lipoprotein (LDL) cholesterol by binding and targeting at LDLR for lysosomal degradation in cells. Preliminary studies revealed that pseurotin A (PsA), a spiro-heterocyclic γ-lactam alkaloid from several marine and terrestrial Aspergillus and Penicillium species, has the ability to dually suppress the PCSK9 expression and protein-protein interaction (PPI) with LDLR, resulting in an anti-hypercholesterolemic effect and modulating the oncogenic role of PCSK9 axis in breast and prostate cancers progression and recurrence. Thus, a preliminary assessment of the PsA acute toxicity represents the steppingstone to develop PsA as a novel orally active PCSK9 axis modulating cancer recurrence inhibitor. PsA studies for in vitro toxicity on RWPE-1 and CCD 841 CoN human non-tumorigenic prostate and colon cells, respectively, indicated a cellular death shown at a 10-fold level of its reported anticancer activity. Moreover, a Western blot analysis revealed a significant downregulation of the pro-survival marker Bcl-2, along with the upregulation of the proapoptotic Bax and caspases 3/7, suggesting PsA-mediated induction of cell apoptosis at very high concentrations. The Up-and-Down methodology determined the PsA LD50 value of >550 mg/kg in male and female Swiss albino mice. Animals were orally administered single doses of PsA at 10, 250, and 500 mg/kg by oral gavage versus vehicle control. Mice were observed daily for 14 days with special care over the first 24 h after dosing to monitor any abnormalities in their behavioral, neuromuscular, and autonomic responses. After 14 days, the mice were euthanized, and their body and organ weights were recorded and collected. Mice plasma samples were subjected to comprehensive hematological and biochemical analyses. Collected mouse organs were histopathologically examined. No morbidity was detected following the PsA oral dosing. The 500 mg/kg female dosing group showed a 45% decrease in the body weight after 14 days but displayed no other signs of toxicity. The 250 mg/kg female dosing group had significantly increased serum levels of liver transaminases AST and ALT versus vehicle control. Moreover, a modest upregulation of apoptotic markers was observed in liver tissues of both animal sexes at 500 mg/kg dose level. However, a histopathological examination revealed no damage to the liver, kidneys, heart, brain, or lungs. While these findings suggest a possible sex-related toxicity at higher doses, the lack of histopathological injury implies that single oral doses of PsA, up to 50-fold the therapeutic dose, do not cause acute organ toxicity in mice though further studies are warranted.


Asunto(s)
Artritis Psoriásica , Neoplasias de la Próstata , Masculino , Ratones , Humanos , Animales , Proproteína Convertasa 9 , Serina Endopeptidasas/metabolismo , Proproteína Convertasas/metabolismo , Próstata/metabolismo , Receptores de LDL/metabolismo , Neoplasias de la Próstata/tratamiento farmacológico
13.
J Lipid Res ; 63(10): 100272, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36067830

RESUMEN

Proprotein Convertase Subtilisin/Kexin type 9 (PCSK9) regulates the cell-surface localization of LDL receptors in hepatocytes and is associated with LDL and lipoprotein(a) [Lp(a)] uptake, reducing blood concentrations. However, the connection between PCSK9 and HDL is unclear. Here, we investigated the association of plasma PCSK9 with HDL subpopulations and examined the effects of PCSK9 on the atheroprotective function of HDL. We examined the association of PCSK9 with HDL in apoB-depleted plasma by ELISA, native PAGE, and immunoblotting. Our analyses showed that upon apoB-depletion, total circulating PCSK9 levels were 32% of those observed in normolipidemic plasma, and only 6% of PCSK9 in the apoB-depleted plasma, including both the mature and furin-cleaved forms, was associated with HDL. We also show human recombinant PCSK9 abolished the capacity of reconstituted HDL to reduce the formation of ROS in endothelial cells, while a PCSK9-blocking antibody enhanced the capacity of human HDL (in apoB-depleted plasma) to reduce ROS formation in endothelial cells and promote endothelial cell migration. Overall, our findings suggest that PCSK9 is only minimally associated with HDL particles, but PCSK9 in apoB-depleted plasma can affect the atheroprotective properties of HDL related to preservation of endothelial function. This study contributes to the elucidation of the pathophysiological role of plasma PCSK9 and highlights further the anti-atherosclerotic effect of PCSK9 inhibition.


Asunto(s)
Proproteína Convertasa 9 , Proproteína Convertasas , Humanos , Apolipoproteínas B , Células Endoteliales/metabolismo , Furina , Lipoproteína(a) , Proproteína Convertasas/metabolismo , Especies Reactivas de Oxígeno , Receptores de LDL/metabolismo , Serina Endopeptidasas/metabolismo , Subtilisinas
14.
Biochem Cell Biol ; 100(3): 189-198, 2022 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-35263196

RESUMEN

Proprotein Convertase Subtilisin/Kexin-type 9 (PCSK9) is a circulating negative regulator of hepatic low-density lipoprotein receptor (LDLR), which clears cholesterol from blood. Gain-of-function genetic mutations that amplify PCSK9 activity have been found to cause potentially lethal familial hypercholesterolemia. Inversely, reduction of its activity through loss-of-function genetics or with pharmaceuticals was shown to increase hepatic LDLR, to lower blood cholesterol, and to protect against cardiovascular diseases. New epidemiological and experimental evidence suggests that this reduction could also attenuate inflammation, reinforce cancer immunity, provide resistance to infections, and protect against liver pathologies. In this review, we question the relevance of this protein under normal physiology. We propose that PCSK9 is an important, but nonessential, modulator of cholesterol metabolism and immunity, and that its pathogenicity results from its chronic overexpression.


Asunto(s)
Proproteína Convertasa 9 , Proproteína Convertasas , Colesterol , Proproteína Convertasa 9/genética , Proproteína Convertasa 9/metabolismo , Proproteína Convertasas/genética , Proproteína Convertasas/metabolismo , Serina Endopeptidasas/genética
15.
Immunity ; 39(4): 711-21, 2013 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-24138882

RESUMEN

Toll-like receptor 7 (TLR7) triggers antiviral immune responses by recognizing viral single-stranded RNA in endosomes, but the biosynthetic pathway of human TLR7 (hTLR7) remains unclear. Here, we show that hTLR7 is proteolytically processed and that the C-terminal fragment selectively accumulates in endocytic compartments. hTLR7 processing occurred at neutral pH and was dependent on furin-like proprotein convertases (PCs). Furthermore, TLR7 processing was required for its functional response to TLR7 agonists such as R837 or influenza virus. Notably, proinflammatory and differentiation stimuli increased the expression of furin-like PCs in immune cells, suggesting a positive feedback mechanism for TLR7 processing during infection. Because self-RNA can under certain conditions activate TLR7 and trigger autoimmunity, our results identify furin-like PCs as a possible target to attenuate TLR7-dependent autoimmunity and other immune pathologies.


Asunto(s)
Furina/metabolismo , Macrófagos/metabolismo , Proproteína Convertasas/metabolismo , Procesamiento Proteico-Postraduccional , Receptor Toll-Like 7/metabolismo , Secuencia de Aminoácidos , Autoinmunidad , Línea Celular , Endosomas/efectos de los fármacos , Endosomas/inmunología , Retroalimentación Fisiológica , Furina/genética , Furina/inmunología , Regulación de la Expresión Génica , Vectores Genéticos , Humanos , Lentivirus/genética , Macrófagos/citología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Datos de Secuencia Molecular , Orthomyxoviridae/inmunología , Proproteína Convertasas/genética , Proproteína Convertasas/inmunología , Estructura Terciaria de Proteína , Quinolinas/farmacología , Transducción de Señal , Receptor Toll-Like 7/genética , Receptor Toll-Like 7/inmunología
16.
Circ Res ; 126(5): 571-585, 2020 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-31893970

RESUMEN

RATIONALE: PCSKs (Proprotein convertase subtilisins/kexins) are a protease family with unknown functions in vasculature. Previously, we demonstrated PCSK6 upregulation in human atherosclerotic plaques associated with smooth muscle cells (SMCs), inflammation, extracellular matrix remodeling, and mitogens. OBJECTIVE: Here, we applied a systems biology approach to gain deeper insights into the PCSK6 role in normal and diseased vessel wall. METHODS AND RESULTS: Genetic analyses revealed association of intronic PCSK6 variant rs1531817 with maximum internal carotid intima-media thickness progression in high-cardiovascular risk subjects. This variant was linked with PCSK6 mRNA expression in healthy aortas and plaques but also with overall plaque SMA+ cell content and pericyte fraction. Increased PCSK6 expression was found in several independent human cohorts comparing atherosclerotic lesions versus healthy arteries, using transcriptomic and proteomic datasets. By immunohistochemistry, PCSK6 was localized to fibrous cap SMA+ cells and neovessels in plaques. In human, rat, and mouse intimal hyperplasia, PCSK6 was expressed by proliferating SMA+ cells and upregulated after 5 days in rat carotid balloon injury model, with positive correlation to PDGFB (platelet-derived growth factor subunit B) and MMP (matrix metalloprotease) 2/MMP14. Here, PCSK6 was shown to colocalize and cointeract with MMP2/MMP14 by in situ proximity ligation assay. Microarrays of carotid arteries from Pcsk6-/- versus control mice revealed suppression of contractile SMC markers, extracellular matrix remodeling enzymes, and cytokines/receptors. Pcsk6-/- mice showed reduced intimal hyperplasia response upon carotid ligation in vivo, accompanied by decreased MMP14 activation and impaired SMC outgrowth from aortic rings ex vivo. PCSK6 silencing in human SMCs in vitro leads to downregulation of contractile markers and increase in MMP2 expression. Conversely, PCSK6 overexpression increased PDGFBB (platelet-derived growth factor BB)-induced cell proliferation and particularly migration. CONCLUSIONS: PCSK6 is a novel protease that induces SMC migration in response to PDGFB, mechanistically via modulation of contractile markers and MMP14 activation. This study establishes PCSK6 as a key regulator of SMC function in vascular remodeling. Visual Overview: An online visual overview is available for this article.


Asunto(s)
Miocitos del Músculo Liso/metabolismo , Proproteína Convertasas/genética , Serina Endopeptidasas/genética , Remodelación Vascular , Animales , Arterias Carótidas/metabolismo , Arterias Carótidas/patología , Movimiento Celular , Proliferación Celular , Células Cultivadas , Masculino , Metaloproteinasas de la Matriz/genética , Metaloproteinasas de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Miocitos del Músculo Liso/fisiología , Polimorfismo de Nucleótido Simple , Proproteína Convertasas/metabolismo , Proteínas Proto-Oncogénicas c-sis/metabolismo , Ratas , Ratas Sprague-Dawley , Serina Endopeptidasas/metabolismo , Transcriptoma
17.
J Bone Miner Metab ; 40(2): 240-250, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35066669

RESUMEN

INTRODUCTION: The diagnosis and treatment of osteoporosis, a frequent age-related metabolic bone disorder, remain incomprehensive and challenging. The potential regulatory role of lncRNA XIST and sphingosine kinase 1 (SPHK1) pathway need experimental investigations. MATERIALS AND METHODS: RAW264.7 cells and BMMs were obtained for in vitro studies and 30 ng/mL RANKL was implemented for induction of osteoclast differentiation. The suppressing of lncRNA XIST, SPHK1 and fused in sarcoma (FUS) was achieved using small hairpin RNA, while overexpression of XIST and FUS was constructed by pcDNA3.1 vector system. Tartrate-resistant acid phosphatase (TRAP) staining was used for observation of formation of osteoclasts. RNA-pulldown analysis and RNA binding protein immunoprecipitation (RIP) was implemented for measuring mRNA and protein interactions. RT-qPCR was conducted to determining mRNA expression, whereas ELISA and Western blotting assay was performed for monitoring protein expression. RESULTS: RANKL induced osteoclast differentiation and upregulated expression of osteoclastogenesis-related genes that included NFATc1, CTSK, TRAP and SPHK1 and the level of lncRNA XIST in both RAW264.7 cells and BMMs. However, knockdown of lncRNA XIST or suppressing SPHK1 significantly reserved the effects of RANKL. LncRNA XIST was further demonstrated to be interacted with FUS and increased the stability of SPHK1, indicating its ability in promoting osteoclast differentiation through SPHK1/S1P/ERK signaling pathway. CONCLUSION: LncRNA XIST promoted osteoclast differentiation via interacting with FUS and upregulating SPHK1/S1P/ERK pathway.


Asunto(s)
Resorción Ósea , Osteoclastos , Proproteína Convertasas/metabolismo , ARN Largo no Codificante , Proteína FUS de Unión a ARN/metabolismo , Serina Endopeptidasas/metabolismo , Animales , Resorción Ósea/metabolismo , Catepsina K/metabolismo , Diferenciación Celular , Hematopoyesis , Ratones , Factores de Transcripción NFATC/metabolismo , Osteoclastos/citología , Osteogénesis , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Ligando RANK/metabolismo , Células RAW 264.7 , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Fosfatasa Ácida Tartratorresistente/metabolismo
18.
Appl Microbiol Biotechnol ; 106(21): 7063-7072, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36195703

RESUMEN

Protein expression with a fusion partner followed by the removal of the fusion partner via in vitro processing with a specific endoprotease is a favored method for the efficient production of intact recombinant proteins. Due to the high cost of commercial endoproteases, this process is restricted to laboratories. Kex2p is a membrane-bound serine protease that cleaves after dibasic residues of substrates in the late Golgi network. Although Kex2p is a very efficient endoprotease with exceptional specificity, it has not yet been used for the in vitro processing of fusion proteins due to its autolysis and high production cost. In this study, we developed an alternative endoprotease, autolysis-proof Kex2p, via site-directed mutagenesis of truncated KEX2 from Candida albicans (CaKEX2). Secretory production of manipulated CaKex2p was improved by employing target protein-specific translational fusion partner in Saccharomyces cerevisiae. The mass production of autolysis-proof Kex2p could facilitate the use of Kex2p for the large-scale production of recombinant proteins. KEY POINTS: • A soluble and active CaKex2p variant was produced by autocatalytic cleavage of the pro-peptide after truncation of C-terminus • Autolysis-proof CaKex2p was developed by site-directed mutagenesis • Secretion of autolysis-proof CaKex2p was improved by employing optimal translational fusion partner in Saccharomyces cerevisiae.


Asunto(s)
Proteínas Fúngicas , Proproteína Convertasas , Saccharomyces cerevisiae , Candida albicans/enzimología , Candida albicans/genética , Péptido Hidrolasas/metabolismo , Péptidos/metabolismo , Proproteína Convertasas/metabolismo , Proteínas Recombinantes de Fusión/biosíntesis , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Serina Endopeptidasas/metabolismo , Subtilisinas/metabolismo , Proteínas Fúngicas/biosíntesis
19.
Proc Natl Acad Sci U S A ; 116(4): 1279-1288, 2019 01 22.
Artículo en Inglés | MEDLINE | ID: mdl-30610172

RESUMEN

The protein acyl transferase ZDHHC5 was recently proposed to regulate trafficking in the endocytic pathway. Therefore, we explored the function of this enzyme in controlling the action of bacterial toxins. We found that ZDHHC5 activity is required for two very different toxins: the anthrax lethal toxin and the pore-forming toxin aerolysin. Both of these toxins have precursor forms, the protoxins, which can use the proprotein convertases Furin and PC7 for activation. We show that ZDHHC5 indeed affects the processing of the protoxins to their active forms. We found that Furin and PC7 can both be S-palmitoylated and are substrates of ZDHHC5. The impact of ZDHHC5 on Furin/PC7-mediated anthrax toxin cleavage is dual, having an indirect and a direct component. First, ZDHHC5 affects the homeostasis and trafficking of a subset of cellular proteins, including Furin and PC7, presumably by affecting the endocytic/recycling pathway. Second, while not inhibiting the protease activity per se, ZDHHC5-mediated Furin/PC7 palmitoylation is required for the cleavage of the anthrax toxin. Finally, we show that palmitoylation of Furin and PC7 promotes their association with plasma membrane microdomains. Both the receptor-bound toxin and the convertases are of very low abundance at the cell surface. Their encounter is unlikely on reasonable time scales. This work indicates that palmitoylation drives their encounter in specific domains, allowing processing and thereby intoxication of the cell.


Asunto(s)
Acetiltransferasas/metabolismo , Antígenos Bacterianos/metabolismo , Bacillus anthracis/metabolismo , Toxinas Bacterianas/metabolismo , Interacciones Huésped-Patógeno/fisiología , Transporte de Proteínas/fisiología , Línea Celular Tumoral , Membrana Celular/metabolismo , Endocitosis/fisiología , Furina/metabolismo , Células HeLa , Humanos , Microdominios de Membrana/metabolismo , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Proproteína Convertasas/metabolismo , Subtilisinas/metabolismo
20.
Clin Exp Ophthalmol ; 50(6): 632-652, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35322530

RESUMEN

BACKGROUND: Furin converts inactive proproteins into bioactive forms. By activating proinflammatory and proangiogenic factors, furin might play a role in pathophysiology of proliferative diabetic retinopathy (PDR). METHODS: We studied vitreous samples from PDR and nondiabetic patients, epiretinal membranes from PDR patients, retinal microvascular endothelial cells (HRMECs), retinal Müller cells and rat retinas by ELISA, Western blot analysis, immunohistochemistry and immunofluorescence microscopy. We performed in vitro angiogenesis assays and assessed adherence of monocytes to HRMECs. RESULTS: Furin levels were significantly increased in PDR vitreous samples. In epiretinal membranes, immunohistochemistry analysis revealed furin expression in monocytes/macrophages, vascular endothelial cells and myofibroblasts. Furin was significantly upregulated in diabetic rat retinas. Hypoxia and TNF-α induced significant upregulation of furin in Müller cells and HRMECs. Furin induced upregulation of phospho-ERK1/2, p65 subunit of NF-κB, ADAM17 and MCP-1 in cultured Müller cells and phospho-ERK1/2 in cultured HRMECs and induced HRMECs migration. Treatment of monocytes with furin significantly increased their adhesion to HRMECs. Intravitreal administration of furin in normal rats induced significant upregulation of p65 subunit of NF-κB, phospho-ERK1/2 and ICAM-1 in the retina. Inhibition of furin with dec-CMK significantly decreased levels of MCP-1 in culture medium of Müller cells and HRMECs and significantly attenuated TNF-α-induced upregulation of p65 subunit of NF-κB, ICAM-1 and VCAM-1 in HRMECs. Dec-CMK significantly decreased adherence of monocytes to HRMECs and TNF-α-induced upregulation of adherence of monocytes to HRMECs. Treatment of HRMECs with dec-CMK significantly attenuated migration of HRMECs. CONCLUSIONS: Furin is a potential driver molecule of PDR-associated inflammation and angiogenesis.


Asunto(s)
Diabetes Mellitus Experimental , Retinopatía Diabética , Membrana Epirretinal , Furina , Animales , Diabetes Mellitus Experimental/metabolismo , Retinopatía Diabética/metabolismo , Células Endoteliales/metabolismo , Furina/metabolismo , Humanos , Molécula 1 de Adhesión Intercelular/metabolismo , FN-kappa B/metabolismo , Neovascularización Patológica/metabolismo , Proproteína Convertasas/metabolismo , Ratas , Factor de Necrosis Tumoral alfa/metabolismo , Cuerpo Vítreo/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA