Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
1.
Cell Rep ; 43(9): 114691, 2024 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-39235944

RESUMEN

The strategy of lowering cholesterol levels by promoting cholesterol excretion is still lacking, and few molecular targets act on multiple cholesterol metabolic processes. In this study, we find that Nogo-B deficiency/inhibition simultaneously promotes hepatic uptake of cholesterol and cholesterol excretion. Nogo-B deficiency decreases cholesterol levels by activating ATP-binding cassette transporters (ABCs), apolipoprotein E (ApoE), and low-density lipoprotein receptor (LDLR) expression. We discover that Nogo-B interacts with liver X receptor α (LXRα), and Nogo-B deficiency inhibits ubiquitination degradation of LXRα, thereby enhancing its function on cholesterol excretion. Decreased cellular cholesterol levels further activate SREBP2 and LDLR expression, thereby promoting hepatic uptake of cholesterol. Nogo-B inhibition decreases atherosclerotic plaques and cholesterol levels in mice, and Nogo-B levels are correlated to cholesterol levels in human plasma. In this study, Nogo-B deficiency/inhibition not only promotes hepatic uptake of blood cholesterol but also facilitates cholesterol excretion. This study reports a strategy to lower cholesterol levels by inhibiting Nogo-B expression to promote hepatic cholesterol uptake and cholesterol excretion.


Asunto(s)
Colesterol , Hipercolesterolemia , Proteínas Nogo , Receptores de LDL , Animales , Humanos , Masculino , Ratones , Apolipoproteínas E/metabolismo , Colesterol/metabolismo , Hipercolesterolemia/metabolismo , Hipercolesterolemia/patología , Hígado/metabolismo , Receptores X del Hígado/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Nogo/antagonistas & inhibidores , Proteínas Nogo/metabolismo , Receptores de LDL/metabolismo , Proteína 2 de Unión a Elementos Reguladores de Esteroles/metabolismo , Ubiquitinación
2.
Genes Nutr ; 19(1): 17, 2024 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-39182019

RESUMEN

BACKGROUND: Nonalcoholic fatty liver disease (NAFLD) is a prevalent chronic liver ailment that can lead to serious conditions such as cirrhosis and hepatocellular carcinoma. Hepatic Nogo-B regulates glucose and lipid metabolism, and its inhibition has been shown to be protective against metabolic syndrome. Increasing evidence suggests that imbalances in the gut microbiota (GM) and lipid metabolism disorders are significant contributors to NAFLD progression. Nevertheless, it is not yet known whether Nogo-B can affect NAFLD by influencing the gut microbiota and metabolites. Hence, the aim of the present study was to characterize this process and explore its possible underlying mechanisms. METHODS: A NAFLD model was constructed by administering a high-fat diet (HFD) to Nogo-B-/- and WT mice from the same litter, and body weight was measured weekly in each group. The glucose tolerance test (GTT) and insulin tolerance test (ITT) were performed to assess blood glucose levels. At the end of the 12-week period, samples of serum, liver, and intestinal contents were collected and used for serum biochemical marker and inflammatory factor detection; pathology evaluation; and gut microbiome and metabolomics analysis. Spearman's correlation analysis was performed to determine possible correlations between differential gut microbiota and differential serum metabolites between groups. RESULTS: Nogo-B deficiency attenuated the effects of the HFD, including weight gain, liver weight gain, impaired glucose tolerance, hepatic steatosis, elevated serum lipid biochemicals levels, and liver function. Nogo-B deficiency suppressed M1 polarization and promoted M2 polarization, thus inhibiting inflammatory responses. Furthermore, Nogo-B-/--HFD-fed mice presented increased gut microbiota richness and diversity, decreased Firmicutes/Bacteroidota (F/B) ratios, and altered serum metabolites compared with those of WT-HFD-fed mice. During analysis, several differential gut microbiota, including Lachnoclostridium, Harryflintia, Odoribacter, UCG-009, and unclassified_f_Butyricoccaceae, were screened between groups. These microbiota were found to be positively correlated with upregulated purine metabolism and bile acid metabolites in Nogo-B deficiency, while they were negatively correlated with downregulated corticosterone and tricarboxylic acid cyclic metabolites in Nogo-B deficiency. CONCLUSION: Nogo-B deficiency delayed NAFLD progression, as demonstrated by reduced hepatocellular lipid accumulation, attenuated inflammation and liver injury, and ameliorated gut microbiota dysbiosis and metabolic disorders. Importantly, Odoribacter was strongly positively correlated with ALB and taurodeoxycholic acid, suggesting that it played a considerable role in the influence of Nogo-B on the progression of NAFLD, a specific feature of NAFLD in Nogo-B-/- mice. The regulation of bile acid metabolism by the gut microbiota may be a potential target for Nogo-B deficiency to ameliorate NAFLD.

3.
Int Immunopharmacol ; 140: 112763, 2024 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-39083925

RESUMEN

Nogo-B, a ubiquitously expressed member of the reticulon family, plays an important role in maintaining endoplasmic reticulum (ER) structure, regulating protein folding, and calcium homeostasis. In this study, we demonstrate that Nogo-B expression and secretion are upregulated in lung cancer and correlate to overall survival. Nogo-B is secreted by various cells, particularly lung cancer cells. ER stress and phosphorylation at serine 107 can induce Nogo-B secretion. Secretory Nogo-B suppresses the differentiation of Th2 cells and the release of type 2 cytokines, thus influencing the anti-tumor effects of Th2-related immune cells, including IgE+B cell class switching and eosinophil activation.


Asunto(s)
Diferenciación Celular , Neoplasias Pulmonares , Proteínas Nogo , Células Th2 , Microambiente Tumoral , Humanos , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Microambiente Tumoral/inmunología , Células Th2/inmunología , Proteínas Nogo/metabolismo , Proteínas Nogo/genética , Estrés del Retículo Endoplásmico/inmunología , Línea Celular Tumoral , Citocinas/metabolismo , Masculino , Femenino , Fosforilación
4.
Mol Nutr Food Res ; 68(14): e2300915, 2024 07.
Artículo en Inglés | MEDLINE | ID: mdl-38862276

RESUMEN

SCOPE: Polycystic ovary syndrome (PCOS) is closely related to non-alcoholic fatty liver disease (NAFLD), and sex hormone-binding globulin (SHBG) is a glycoprotein produced by the liver. Hepatic lipogenesis inhibits hepatic SHBG synthesis, which leads to hyperandrogenemia and ovarian dysfunction in PCOS. Therefore, this study aims to characterize the mechanism whereby liver lipogenesis inhibits SHBG synthesis. METHODS AND RESULTS: This study establishes a rat model of PCOS complicated by NAFLD using a high-fat diet in combination with letrozole and performs transcriptomic analysis of the liver. Transcriptomic analysis of the liver shows that the expression of neurite growth inhibitor-B receptor (NgBR), hepatocyte nuclear factor 4α (HNF4α), and SHBG is low. Meantime, HepG2 cells are treated with palmitic acid (PA) to model NAFLD in vitro, which causes decreases in the expression of NgBR, HNF4α, and SHBG. However, the expression of HNF4α and SHBG is restored by treatment with the AMP-activated protein kinase (AMPK) agonist AICAR. CONCLUSIONS: NgBR regulates the expression of HNF4α by activating the AMPK signaling pathway, thereby affecting the synthesis of SHBG in the liver. Further mechanistic studies regarding the effect of liver fat on NGBR expression are warranted.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Dieta Alta en Grasa , Factor Nuclear 4 del Hepatocito , Hiperglucemia , Letrozol , Hígado , Síndrome del Ovario Poliquístico , Globulina de Unión a Hormona Sexual , Animales , Letrozol/farmacología , Factor Nuclear 4 del Hepatocito/metabolismo , Factor Nuclear 4 del Hepatocito/genética , Femenino , Síndrome del Ovario Poliquístico/metabolismo , Dieta Alta en Grasa/efectos adversos , Hígado/metabolismo , Hígado/efectos de los fármacos , Globulina de Unión a Hormona Sexual/metabolismo , Globulina de Unión a Hormona Sexual/genética , Células Hep G2 , Humanos , Proteínas Quinasas Activadas por AMP/metabolismo , Ratas Sprague-Dawley , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/etiología , Ratas , Transducción de Señal/efectos de los fármacos , Lipogénesis/efectos de los fármacos
5.
Mol Biotechnol ; 2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38727882

RESUMEN

Liver fibrosis is a severe liver pathology in response to chronic or iterative liver injury. Senescence has emerged as a protective mechanism against liver fibrosis. Nogo-B has been well established as a significant contributor to liver fibrosis. Nonetheless, researches regarding the role of Nogo-B in cell senescence during liver fibrosis are few. In platelet-derived growth factor-BB (PDGF-BB)-treated human hepatic stellate cell line LX-2, cell proliferation was assayed by CCK-8 method. Western blotting estimated the expression of Nogo-B and fibrosis markers. After Nogo-B was silenced in LX-2 cells pretreated by an autophagy activator Rapamycin and PDGF-BB, CCK-8 method was used to assess cell proliferation. Fibrosis was measured by western blotting and immunofluorescence. Cell cycle was subjected to flow cytometry analysis and cell senescence was evaluated by SA-ß-gal staining. Immunofluorescence staining assessed autophagy. Nogo-B was elevated in PDGF-BB-exposed LX-2 cells. Nogo-B silencing suppressed the proliferation, fibrosis, and autophagy while induced cell cycle arrest and senescence of LX-2 cells. Additionally, pretreatment with Rapamycin partially restored the effects of Nogo-B knockdown on the autophagy, proliferation, fibrosis, cell cycle, and senescence of LX-2 cells upon exposure to PDGF-BB. Collectively, inactivation of autophagy mediated by Nogo-B deficiency might elicit protective activities against the development of liver fibrosis.

6.
Tissue Eng Regen Med ; 21(5): 777-789, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38630369

RESUMEN

BACKGROUND: Hepatic fibrosis (HF) is a histopathological change in the process of long-term liver injury caused by cytokine secretion and internal environment disturbance, resulting in excessive liver repair and fiber scar. Nogo-B protein is widely distributed in peripheral tissues and organs and can regulate the migration of endothelial cells by activating TGF-ß1 in vascular remodeling after injury. Nogo-B has been shown to promote organ fibrosis. This study was to determine the role of Nogo-B in HF. METHODS: An HF model was built by intraperitoneal injections with 20% carbon tetrachloride. Localization of Nogo-B was detected by FISH. The interaction between Nogo-B and BACE1 was confirmed by Co-IP. Autophagy flux was analyzed using tandem mRFP-GFP-LC3 fluorescence microscopy, electron microscopy, and western blotting. Detection of serum AST and ALT and H&E staining were utilized to detect the degree of liver injury. The HF was evaluated by Masson trichromatic staining. RT-qPCR, western blotting, and immunofluorescence were employed to detect relevant indicators. RESULTS: Reducing Nogo-B suppressed AST and ALT levels, the accumulation of collagen I and α-SMA, and expressions of pro-fibrotic genes in mouse liver. BACE1 was a potential downstream target of Nogo-B. Nogo-B was upregulated in TGF-ß1-activated hepatic stellate cells (HSCs). Knocking down Nogo-B caused the downregulation of pro-fibrotic genes and inhibited viability of HSCs. Nogo-B knockdown prevented CCL4-induced fibrosis, accompanied by downregulation of extracellular matrix. Nogo-B inhibited HSC autophagy and increased lipid accumulation. BACE1 knockdown inhibited HSC autophagy and activation in LX-2 cells. CONCLUSION: Nogo-B knockdown prevents HF by directly inhibiting BACe1-mediated autophagy.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide , Ácido Aspártico Endopeptidasas , Autofagia , Cirrosis Hepática , Proteínas Nogo , Animales , Humanos , Masculino , Ratones , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Ácido Aspártico Endopeptidasas/metabolismo , Tetracloruro de Carbono , Células Estrelladas Hepáticas/metabolismo , Hígado/metabolismo , Hígado/patología , Cirrosis Hepática/metabolismo , Cirrosis Hepática/patología , Ratones Endogámicos C57BL , Proteínas Nogo/metabolismo
7.
Artículo en Inglés | MEDLINE | ID: mdl-38497748

RESUMEN

Aims: Endothelial cells are the critical targets of injury in diabetic nephropathy (DN), and endothelial cell lesions contribute to the disease progression. Neurite outgrowth inhibitor B (Nogo-B), an endoplasmic reticulum (ER)-resident protein, plays a pivotal role in vascular remodeling after injury, and maintains the structure and function of the ER. Yet, the role of Nogo-B in the regulation of ER stress and endothelial cell injury remains largely unknown. Herein, we tested the hypothesis that Nogo-B activates ER stress-mediated autophagy and protects endothelial cells in DN. Results: The level of Nogo-B was decreased in glomerular endothelial cells in biopsy specimens from DN patients. In vivo and in vitro studies have shown that silencing Nogo-B activated ER stress signaling, and affected the expression of autophagy-related marker early growth response 1 and microtubule-associated protein light chain 3 (LC3) in endothelial cells in hyperglycemic condition. Conclusion and Innovation: These results denote that Nogo-B contributes to ER stress-mediated autophagy and protects endothelial cells in DN, providing new evidence for understanding the role of ER stress-mediated autophagy in endothelial cells of DN.

8.
Mov Disord Clin Pract ; 11(1): 76-85, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38291835

RESUMEN

BACKGROUND: Variants in dehydrodolichol diphosphate synthetase (DHDDS) and nuclear undecaprenyl pyrophosphate synthase 1 (NUS1) cause a neurodevelopmental disorder, classically with prominent epilepsy. Recent reports suggest a complex movement disorder and an overlapping phenotype has been postulated due to their combined role in dolichol synthesis. CASES: We describe three patients with heterozygous variants in DHDDS and five with variants affecting NUS1. They bear a remarkably similar phenotype of a movement disorder dominated by multifocal myoclonus. Diagnostic clues include myoclonus exacerbated by action and facial involvement, and slowly progressive or stable, gait ataxia with disproportionately impaired tandem gait. Myoclonus is confirmed with neurophysiology, including EMG of facial muscles. LITERATURE REVIEW: Ninety-eight reports of heterozygous variants in DHDDS, NUS1 and chromosome 6q22.1 structural alterations spanning NUS1, confirm the convergent phenotype of hypotonia at birth, developmental delay, multifocal myoclonus, ataxia, dystonia and later parkinsonism with or without generalized epilepsy. Other features include periodic exacerbations, stereotypies, anxiety, and dysmorphisms. Although their gene products contribute to dolichol biosynthesis, a key step in N-glycosylation, transferrin isoform profiles are typically normal. Imaging is normal or non-specific. CONCLUSIONS: Recognition of their shared phenotype may expedite diagnosis through chromosomal microarray and by including DHDDS/NUS1 in movement disorder gene panels.


Asunto(s)
Trastornos del Movimiento , Mioclonía , Recién Nacido , Humanos , Difosfatos , Fenotipo , Ataxia , Dolicoles/metabolismo , Receptores de Superficie Celular
9.
Redox Biol ; 68: 102944, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37890359

RESUMEN

AIMS: Endothelial dysfunction plays a pivotal role in atherosclerosis, but the detailed mechanism remains incomplete understood. Nogo-B is an endoplasmic reticulum (ER)-localized protein mediating ER-mitochondrial morphology. We previously showed endothelial Nogo-B as a key regulator of endothelial function in the setting of hypertension. Here, we aim to further assess the role of Nogo-B in coronary atherosclerosis in ApoE-/- mice with pressure overload. METHODS AND RESULTS: We generated double knockout (DKO) mouse models of systemically or endothelium-specifically excising Nogo-A/B gene on an ApoE-/- background. After 7 weeks of transverse aortic constriction (TAC) surgery, compared to ApoE-/- mice DKO mice were resistant to the development of coronary atherosclerotic lesions and plaque rapture. Sustained elevation of Nogo-B and adhesion molecules (VCAM-1/ICAM-1), early markers of atherosclerosis, was identified in heart tissues and endothelial cells (ECs) isolated from TAC ApoE-/- mice, changes that were significantly repressed by Nogo-B deficiency. In cultured human umbilical vein endothelial cells (HUVECs) exposure to inflammatory cytokines (TNF-α, IL-1ß), Nogo-B was upregulated and activated reactive oxide species (ROS)-p38-p65 signaling axis. Mitofusin 2 (Mfn2) is a key protein tethering ER to mitochondria in ECs, and we showed that Nogo-B expression positively correlated with Mfn2 protein level. And Nogo-B deletion in ECs or in ApoE-/- mice reduced Mfn2 protein content and increased ER-mitochondria distance, reduced ER-mitochondrial Ca2+ transport and mitochondrial ROS generation, and prevented VCAM-1/ICAM-1 upregulation and EC dysfunction, eventually restrained atherosclerotic lesions development. CONCLUSION: Our study revealed that Nogo-B is a critical modulator in promoting endothelial dysfunction and consequent pathogenesis of coronary atherosclerosis in pressure overloaded hearts of ApoE-/- mice. Nogo-B may hold the promise to be a common therapeutic target in the setting of hypertension.


Asunto(s)
Aterosclerosis , Enfermedad de la Arteria Coronaria , Hipertensión , Placa Aterosclerótica , Humanos , Animales , Ratones , Enfermedad de la Arteria Coronaria/genética , Enfermedad de la Arteria Coronaria/metabolismo , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Molécula 1 de Adhesión Celular Vascular/genética , Molécula 1 de Adhesión Celular Vascular/metabolismo , Proteínas Nogo/genética , Proteínas Nogo/metabolismo , Aterosclerosis/genética , Aterosclerosis/metabolismo , Placa Aterosclerótica/metabolismo , Estrés Oxidativo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Inflamación/metabolismo , Endotelio/metabolismo , Hipertensión/metabolismo , Apolipoproteínas E/genética , Ratones Noqueados , Ratones Endogámicos C57BL
10.
Sheng Wu Gong Cheng Xue Bao ; 39(10): 4168-4188, 2023 Oct 25.
Artículo en Chino | MEDLINE | ID: mdl-37877398

RESUMEN

Cluster of differentiation 36 (CD36) is a membrane glycoprotein receptor capable of binding and transporting fatty acid. Nogo-B regulates the metabolism of fatty acids in the liver and affects the development of liver cancer. To date, it remains unclear whether the interaction between CD36 and Nogo-B affects the proliferation and migration of breast cancer cells. In the current study, we aimed to determine whether the interference of CD36 and Nogo-B affects the proliferation and migration of triple-negative breast cancer (TNBC) cells. The results showed that inhibition of CD36 or Nogo-B alone can inhibit the proliferation and migration of TNBC cells, and the inhibitory effect was more pronounced when CD36 and Nogo-B were inhibited simultaneously. Meanwhile, it was found that inhibition of CD36 and Nogo-B expression can inhibit the expression of Vimentin, B-cell lympoma-2 (BCL2) and proliferating cell nuclear antigen (PCNA). In vivo, knockdown of CD36 or Nogo-B in E0771 cells reduced its tumorigenic ability, which was further enhanced by knockdown of CD36 and Nogo-B simultaneously. Mechanistically, inhibition of CD36 and Nogo-B expression can decrease fatty acid binding protein 4 (FABP4) and fatty acid transport protein 4 (FATP4) expression. Moreover, overexpression of CD36 and Nogo-B-induced cell proliferation was attenuated by FABP4 siRNA, indicating that inhibition of CD36 and Nogo-B expression could inhibit the absorption and transport of fatty acids, thereby inhibiting the proliferation and migration of TNBC. Furthermore, inhibition of CD36 and Nogo-B expression activated the P53-P21-Rb signaling pathway which contributed to the CD36 and Nogo-B-inhibited proliferation and migration of TNBC. Taken together, the results suggest that inhibition of CD36 and Nogo-B can reduce the proliferation and migration of TNBC, which provides new targets for the development of drugs against TNBC.


Asunto(s)
Neoplasias de la Mama Triple Negativas , Humanos , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/metabolismo , Movimiento Celular , Proliferación Celular , Línea Celular Tumoral , Ácidos Grasos
11.
Acta Pharmacol Sin ; 44(11): 2216-2229, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37402997

RESUMEN

Excessive fructose consumption increases hepatic de novo lipogenesis, resulting in cellular stress, inflammation and liver injury. Nogo-B is a resident protein of the endoplasmic reticulum that regulates its structure and function. Hepatic Nogo-B is a key protein in glycolipid metabolism, and inhibition of Nogo-B has protective effects against metabolic syndrome, thus small molecules that inhibit Nogo-B have therapeutic benefits for glycolipid metabolism disorders. In this study we tested 14 flavones/isoflavones in hepatocytes using dual luciferase reporter system based on the Nogo-B transcriptional response system, and found that 6-methyl flavone (6-MF) exerted the strongest inhibition on Nogo-B expression in hepatocytes with an IC50 value of 15.85 µM. Administration of 6-MF (50 mg· kg-1 ·d-1, i.g. for 3 weeks) significantly improved insulin resistance along with ameliorated liver injury and hypertriglyceridemia in high fructose diet-fed mice. In HepG2 cells cultured in a media containing an FA-fructose mixture, 6-MF (15 µM) significantly inhibited lipid synthesis, oxidative stress and inflammatory responses. Furthermore, we revealed that 6-MF inhibited Nogo-B/ChREBP-mediated fatty acid synthesis and reduced lipid accumulation in hepatocytes by restoring cellular autophagy and promoting fatty acid oxidation via the AMPKα-mTOR pathway. Thus, 6-MF may serve as a potential Nogo-B inhibitor to treat metabolic syndrome caused by glycolipid metabolism dysregulation.


Asunto(s)
Enfermedad Hepática Crónica Inducida por Sustancias y Drogas , Flavonas , Síndrome Metabólico , Enfermedad del Hígado Graso no Alcohólico , Ratones , Animales , Fructosa/efectos adversos , Fructosa/metabolismo , Síndrome Metabólico/metabolismo , Hígado/metabolismo , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Metabolismo de los Lípidos , Dieta , Lipogénesis , Flavonas/farmacología , Flavonas/uso terapéutico , Flavonas/metabolismo , Ácidos Grasos/metabolismo , Glucolípidos , Lípidos
12.
Neurochem Int ; 167: 105553, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37230196

RESUMEN

Many studies have shown a close association between Nogo-B and inflammation-related diseases. However, uncertainty does exist, regarding Nogo-B function in the pathological progression of cerebral ischemia/reperfusion (I/R) injury. Middle cerebral artery occlusion/reperfusion (MCAO/R) model was utilized in C57BL/6L mice to mimic ischemic stroke in vivo. Using oxygen-glucose deprivation and reoxygenation (ODG/R) model in microglia cells (BV-2) to establish cerebral I/R injury in vitro. Various methods, including Nogo-B siRNA transfection, mNSS and the rotarod test, TTC, HE and Nissl staining, immunofluorescence staining, immunohistochemistry, Western blot, ELISA, TUNEL and qRT-PCR were employed to probe into the effect of Nogo-B downregulation on cerebral I/R injury and the potential mechanisms. A small amount of Nogo-B expression (protein and mRNA) was observed in cortex and hippocampus before ischemia, then Nogo-B expression increased significantly on day 1, reaching the maximum on day 3, remaining stable on day 14 after I/R, and decreasing gradually after 21 days, but it still rose significantly compared with that observed preischemia. Nogo-B down-regulation could markedly reduce the neurological score and infarct volume, improve the histopathological changes and neuronal apoptosis, lower the number of CD86+/Iba1+ cells and the levels of IL-1ß, IL-6, and TNF-α, and raise the density of NeuN fluorescence, the number of CD206+/Iba1+ cells, and the level of IL-4, IL-10 and TGF-ß in brain of MCAO/R mice. Treatment with Nogo-B siRNA or TAK-242 in BV-2 cells could obviously decrease the CD86 fluorescence density and the mRNA expression of IL-1ß, IL-6 and TNF-α, increase CD206 fluorescence density and the mRNA expression of IL-10 after OGD/R injury. In addition, the expression of TLR4, p-IκBα and p-p65 proteins significantly increased in the brain after MCAO/R and BV-2 cells exposed to OGD/R. Treatment with Nogo-B siRNA or TAK-242 prominently reduced the expression of TLR4, p-IκBα and p-p65. Our findings suggest that the down-regulation of Nogo-B exerts protective effect on cerebral I/R injury by modulating the microglia polarization through inhibiting TLR4/NF-κB signaling pathway. Nogo-B may be a potential therapeutic target for ischemic stroke.


Asunto(s)
Isquemia Encefálica , Accidente Cerebrovascular Isquémico , Daño por Reperfusión , Animales , Ratones , Isquemia Encefálica/metabolismo , Regulación hacia Abajo , Infarto de la Arteria Cerebral Media/metabolismo , Interleucina-10/genética , Interleucina-10/metabolismo , Interleucina-10/farmacología , Interleucina-6/metabolismo , Accidente Cerebrovascular Isquémico/metabolismo , Ratones Endogámicos C57BL , Microglía/metabolismo , FN-kappa B/metabolismo , Inhibidor NF-kappaB alfa/genética , Inhibidor NF-kappaB alfa/metabolismo , Inhibidor NF-kappaB alfa/farmacología , Daño por Reperfusión/metabolismo , ARN Mensajero/metabolismo , ARN Interferente Pequeño/farmacología , Receptor Toll-Like 4/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
13.
Int Immunopharmacol ; 120: 110378, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37244119

RESUMEN

BACKGROUND & AIMS: Ulcerative colitis (UC) is a main type of inflammatory bowel diseases which spreads globally during the westernization of lifestyle over the past few decades. However, the cause of UC is still not fully understood. We aimed to disclose the role of Nogo-B in the development of UC. METHODS: Nogo-deficiency (Nogo-/-) and wild-type male mice were treated with dextran sodium sulfate (DSS) to conduct a UC model, followed by determination of colon and serum inflammatory cytokines level. RAW264.7, THP1 and NCM460 cells were used to determine macrophage inflammation as well as proliferation and migration of NCM460 cells under Nogo-B or miR-155 intervention. RESULTS: Nogo deficiency significantly reduced DSS-induced weight loss, colon length and weight reduction, and inflammatory cells accumulation in the intestinal villus, while increased the expression of tight junctions (TJs) proteins (Zonula occludens-1, Occludin) and adherent junctions (AJs) proteins (E-cadherin, α-catenin), implying that Nogo deficiency attenuated DSS-induced UC. Mechanistically, Nogo-B deficiency reduced TNFα, IL-1ß and IL-6 levels in the colon, serum, RAW264.7 cells and THP1-derived macrophages. Furthermore, we identified that Nogo-B inhibition can reduce the maturation of miR-155, which is essential for Nogo-B-affected inflammatory cytokines expression. Interestingly, we determined that Nogo-B and p68 can interact with each other to promote the expression and activation of Nogo-B and p68, thus facilitating miR-155 maturation to induce macrophage inflammation. Blocking p68 inhibited Nogo-B, miR-155, TNFα, IL-1ß and IL-6 expression. Moreover, the culture medium collected from Nogo-B overexpressed macrophages can inhibit enterocytes NCM460 cells proliferation and migration. CONCLUSION: We disclose that Nogo deficiency reduced DSS-induced UC via inhibiting p68-miR-155-activated inflammation. Our results indicate that Nogo-B inhibition serves as a new potential therapeutic candidate for the prevention and treatment of UC.


Asunto(s)
Colitis Ulcerosa , MicroARNs , Masculino , Animales , Ratones , Colitis Ulcerosa/inducido químicamente , Colitis Ulcerosa/tratamiento farmacológico , Colitis Ulcerosa/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Interleucina-6/metabolismo , Colon/metabolismo , Inflamación/tratamiento farmacológico , Transducción de Señal , Citocinas/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Sulfato de Dextran , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL
14.
Life Sci ; 321: 121571, 2023 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-36931495

RESUMEN

AIMS: Obesity is a global epidemic around the world. Reticulon-4B (Nogo-B) is an endoplasmic reticulum-resident protein. Our previous work demonstrated that Nogo-B deficiency inhibited obesity and decreased the size of white adipocytes. However, the underlying molecular mechanism of Nogo-B in white adipogenesis remains poorly understood. This study aims to explore the effect of Nogo-B in white adipogenesis, as well as its underlying molecular mechanisms. MAIN METHODS AND FINDINGS: The study adopted mouse embryonic fibroblasts (MEFs) and 3T3-L1 preadipocytes to induce white adipogenesis and investigate the effect of Nogo-B on adipogenesis using qRT-PCR, Western blotting, immunofluorescence, lipid quantification, and Oil Red O staining. During white adipogenesis, Nogo-B expression was increased accompanied by upregulation of adipogenic markers. In contrast, Nogo-B deficiency inhibited white adipocyte markers expression and lipid accumulation. Furthermore, the mechanism study showed that Nogo-B deficiency decreased the destruction complex [AXIN1-APC-glycogen synthase kinase 3ß (GSK3ß)] levels through activating protein kinase B 2 (AKT2), resulting in ß-catenin translocating into the nucleus and inhibiting the expression of adipogenic markers. Moreover, Nogo-B deficiency promoted the expression of brown/beige adipocytes markers while improving mitochondrial thermogenesis by activating ß-catenin pathway. In addition, Nogo-B deficiency reduced the levels of inflammatory molecules during white adipogenic differentiation. SIGNIFICANCE: This study revealed that Nogo-B deficiency inhibited white adipogenesis through AKT2/GSK3ß/ß-catenin pathway. Meanwhile, Nogo-B deficiency increased the expression of brown/beige adipocyte markers and promoted mitochondrial thermogenesis. In addition, Nogo-B deficiency reduced inflammatory cytokine levels caused by adipogenesis. Collectively, blocking Nogo-B expression may be a potential strategy to suppress white adipogenesis.


Asunto(s)
Adipogénesis , beta Catenina , Animales , Ratones , Células 3T3-L1 , Adipocitos/metabolismo , beta Catenina/metabolismo , Diferenciación Celular , Fibroblastos/metabolismo , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Lípidos/farmacología , Obesidad/metabolismo
15.
Chinese Journal of Biotechnology ; (12): 4168-4188, 2023.
Artículo en Chino | WPRIM (Pacífico Occidental) | ID: wpr-1008019

RESUMEN

Cluster of differentiation 36 (CD36) is a membrane glycoprotein receptor capable of binding and transporting fatty acid. Nogo-B regulates the metabolism of fatty acids in the liver and affects the development of liver cancer. To date, it remains unclear whether the interaction between CD36 and Nogo-B affects the proliferation and migration of breast cancer cells. In the current study, we aimed to determine whether the interference of CD36 and Nogo-B affects the proliferation and migration of triple-negative breast cancer (TNBC) cells. The results showed that inhibition of CD36 or Nogo-B alone can inhibit the proliferation and migration of TNBC cells, and the inhibitory effect was more pronounced when CD36 and Nogo-B were inhibited simultaneously. Meanwhile, it was found that inhibition of CD36 and Nogo-B expression can inhibit the expression of Vimentin, B-cell lympoma-2 (BCL2) and proliferating cell nuclear antigen (PCNA). In vivo, knockdown of CD36 or Nogo-B in E0771 cells reduced its tumorigenic ability, which was further enhanced by knockdown of CD36 and Nogo-B simultaneously. Mechanistically, inhibition of CD36 and Nogo-B expression can decrease fatty acid binding protein 4 (FABP4) and fatty acid transport protein 4 (FATP4) expression. Moreover, overexpression of CD36 and Nogo-B-induced cell proliferation was attenuated by FABP4 siRNA, indicating that inhibition of CD36 and Nogo-B expression could inhibit the absorption and transport of fatty acids, thereby inhibiting the proliferation and migration of TNBC. Furthermore, inhibition of CD36 and Nogo-B expression activated the P53-P21-Rb signaling pathway which contributed to the CD36 and Nogo-B-inhibited proliferation and migration of TNBC. Taken together, the results suggest that inhibition of CD36 and Nogo-B can reduce the proliferation and migration of TNBC, which provides new targets for the development of drugs against TNBC.


Asunto(s)
Humanos , Neoplasias de la Mama Triple Negativas/metabolismo , Movimiento Celular , Proliferación Celular , Línea Celular Tumoral , Ácidos Grasos
16.
Int J Mol Sci ; 23(21)2022 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-36362109

RESUMEN

Retinitis pigmentosa-59 (RP59) is a rare, recessive form of RP, caused by mutations in the gene encoding DHDDS (dehydrodolichyl diphosphate synthase). DHDDS forms a heterotetrameric complex with Nogo-B receptor (NgBR; gene NUS1) to form a cis-prenyltransferase (CPT) enzyme complex, which is required for the synthesis of dolichol, which in turn is required for protein N-glycosylation as well as other glycosylation reactions in eukaryotic cells. Herein, we review the published phenotypic characteristics of RP59 models extant, with an emphasis on their ocular phenotypes, based primarily upon knock-in of known RP59-associated DHDDS mutations as well as cell type- and tissue-specific knockout of DHDDS alleles in mice. We also briefly review findings in RP59 patients with retinal disease and other patients with DHDDS mutations causing epilepsy and other neurologic disease. We discuss these findings in the context of addressing "knowledge gaps" in our current understanding of the underlying pathobiology mechanism of RP59, as well as their potential utility for developing therapeutic interventions to block the onset or to dampen the severity or progression of RP59.


Asunto(s)
Transferasas Alquil y Aril , Animales , Ratones , Dolicoles/metabolismo , Mutación , Vertebrados/metabolismo , Modelos Animales
17.
Cells ; 11(19)2022 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-36231046

RESUMEN

Nogo-B has been reported to play a critical role in angiogenesis and the repair of damaged blood vessels; however, its role in the tumor microenvironment remains unclear. Here, we observed the differential expression of Nogo-B in endothelial cells from hepatocellular carcinoma (HCC) and glioma samples. Downregulation of Nogo-B expression correlated with the malignant phenotype of cancer and a poor prognosis for patients. In subsequent studies, endothelial Nogo-B inhibition robustly promoted the growth of HCC or glioma xenografts in nude mice. Intriguingly, endothelial Nogo-B silencing dramatically suppressed endothelial cell expansion and tumor angiogenesis, but potently enhanced the proliferation of neighboring HCC and glioma cells. Based on the results of the ELISA assay, Nogo-B silencing reduced TGF-ß production in endothelial cells, which attenuated the phosphorylation and nuclear translocation of Smad in neighboring cancer cells. The endothelial Nogo-B silencing-mediated increase in cancer cell proliferation was abolished by either a TGF-ß neutralizing antibody or TGF-ß receptor inhibitor, indicating the essential role for TGF-ß in endothelial Nogo-B-mediated suppression of cancer growth. These findings not only broaden our understanding of the crosstalk between cancer cells and endothelial cells but also provide a novel prognostic biomarker and a therapeutic target for cancer treatments.


Asunto(s)
Carcinoma Hepatocelular , Glioma , Neoplasias Hepáticas , Proteínas Nogo/metabolismo , Animales , Anticuerpos Neutralizantes , Biomarcadores , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Proliferación Celular , Células Endoteliales/metabolismo , Humanos , Neoplasias Hepáticas/patología , Ratones , Ratones Desnudos , Neovascularización Patológica , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Proteínas Smad/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Microambiente Tumoral
18.
Biomed Pharmacother ; 153: 113444, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-36076559

RESUMEN

Liver fibrosis is an important pathologic process in response to chronic or repetitive liver injury. It can advance to liver cirrhosis. Both peroxisome proliferator-activated receptor gamma (PPARγ) and Nogo-B play critical roles in fibrogenesis, while PPARγ is essential for the development. However, the effect of Nogo-B deficiency on the development of liver fibrosis in cell-specific PPARγ deficient mice remains unknown. In this study, hepatocyte or macrophage PPARγ deficient (hPPARγ KO or mPPARγ KO) mice, Nogo-B deficient mice, and cell-specific PPARγ plus Nogo-B double deficient (hPPARγ/Nogo-B DKO or mPPARγ/Nogo-B DKO) mice were induced liver fibrosis by CCl4 injection. We found hPPARγ KO mice showed enhanced liver fibrotic signatures compared to mPPARγ KO mice after CCl4 administration. Hepatocyte or macrophage PPARγ deficiency further enhanced CCl4-induced severe inflammation infiltration, apoptosis and M1 macrophage polarization in the liver. In contrast, Nogo-B deficiency effectively ameliorated PPARγ deficiency-aggravated liver injury and fibrosis. It ameliorated PPARγ deficiency-aggravated liver inflammation and fibrosis by suppressing hepatic stellate cell activation, TLR4-NF-κB/TNF-α signaling and M1 macrophage polarization. In conclusion, our study demonstrates that PPARγ deficiency increases susceptibility of mice to develop CCl4-induced liver injury/fibrosis, which is potently reduced by Nogo-B deficiency, indicating Nogo-B inhibition might be a therapeutic approach for liver fibrosis treatment.


Asunto(s)
FN-kappa B , PPAR gamma , Animales , Fibrosis , Células Estrelladas Hepáticas/metabolismo , Hígado/metabolismo , Cirrosis Hepática/metabolismo , Macrófagos/metabolismo , Ratones , FN-kappa B/metabolismo , Proteínas Nogo , PPAR gamma/metabolismo , Receptor Toll-Like 4/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
19.
Front Neurosci ; 16: 894454, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35958994

RESUMEN

Background: Recently, the neurite outgrowth inhibitor-B (Nogo-B) receptor has been reported as a novel candidate gene for Parkinson's disease (PD). Nogo-B receptors need to combine with soluble Nogo-B to exert their physiological function. However, little is known about the relationship between serum soluble Nogo-B and PD. Methods: Serum levels of sNogo-B and α-Synuclein (α-Syn) were measured in a cohort of 53 patients with PD and 49 healthy controls with the ELISA kit method. Results: Serum sNogo-B level is significantly lower in the PD group than that in healthy controls and is negatively correlated with UPDRS-III score (p = 0.049), H&Y stage (p = 0.0108) as well as serum α-Syn level (p = 0.0001). The area under the curve (AUC) of serum sNogo-B in differentiating patients with PD from controls was 0.801 while the AUC of serum α-Syn was 0.93. Combining serum sNogo-B and α-Syn in differentiating patients with PD from HC presented higher discriminatory potential (AUC = 0.9534). Conclusion: Decreased serum sNogo-B may be a potential biomarker for PD. Lower Nogo-B level reflects worse motor function and disease progression of PD. Serum sNogo-B is of added value to serum α-Syn panel in distinguishing PD from controls. Future studies are needed to confirm in larger samples and different populations.

20.
BMC Cardiovasc Disord ; 22(1): 248, 2022 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-35655131

RESUMEN

BACKGROUND: Cellular and animal studies have shown that endoplasmic reticulum protein B (Nogo-B) is associated with hypertension, but that association has not been fully studied in humans. Therefore, the expression levels of Nogo-B were investigated in hypertensive patients. METHODS: The plasma Nogo-B levels of 74 patients with hypertension and 67 non-hypertensive patients were measured by enzyme-linked immunosorbent assay. RESULTS: The plasma Nogo-B levels in the hypertensive group [523.43(411.41-746.79)] were higher than in the non-hypertensive group [380.29(281.57-462.13)] (P < 0.01). Pearson's correlation analysis indicated that systolic blood pressure and diastolic blood pressure were linearly and positively correlated with plasma Nogo-B levels. Multivariable logistic regression analysis was performed based on sex, age, BMI, smoking history, drinking history, and levels of TC, TG, LDL, and HDL. The results indicated that the plasma Nogo-B levels were independently associated with hypertension (OR = 1.007, 95%CI: 1.004-1.010, P < 0.01). CONCLUSIONS: The present study suggests that hypertensive participants exhibited higher plasma Nogo-B levels than those without hypertension. Plasma Nogo-B levels are independently associated with hypertension.


Asunto(s)
Hipertensión , Animales , Pueblo Asiatico , China/epidemiología , Humanos , Hipertensión/diagnóstico , Plasma , Fumar
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA