Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 82
Filter
1.
Nat Commun ; 15(1): 6845, 2024 Aug 10.
Article in English | MEDLINE | ID: mdl-39122737

ABSTRACT

Glucagon-like peptide 1 (GLP1), which is mainly processed and cleaved from proglucagon in enteroendocrine cells (EECs) of the intestinal tract, acts on the GLP1 receptor in pancreatic cells to stimulate insulin secretion and to inhibit glucagon secretion. However, GLP1 processing is not fully understood. Here, we show that reticulon 4B (Nogo-B), an endoplasmic reticulum (ER)-resident protein, interacts with the major proglucagon fragment of proglucagon to retain proglucagon on the ER, thereby inhibiting PCSK1-mediated cleavage of proglucagon in the Golgi. Intestinal Nogo-B knockout in male type 2 diabetes mellitus (T2DM) mice increases GLP1 and insulin levels and decreases glucagon levels, thereby alleviating pancreatic injury and insulin resistance. Finally, we identify aberrantly elevated Nogo-B expression and inhibited proglucagon cleavage in EECs from diabetic patients. Our study reveals the subcellular regulatory processes involving Nogo-B during GLP1 production and suggests intestinal Nogo-B as a potential therapeutic target for T2DM.


Subject(s)
Diabetes Mellitus, Type 2 , Endoplasmic Reticulum , Glucagon-Like Peptide 1 , Nogo Proteins , Proglucagon , Proprotein Convertase 1 , Animals , Humans , Male , Mice , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/genetics , Endoplasmic Reticulum/metabolism , Enteroendocrine Cells/metabolism , Glucagon-Like Peptide 1/metabolism , Golgi Apparatus/metabolism , HEK293 Cells , Insulin/metabolism , Insulin Resistance , Intestines/pathology , Mice, Inbred C57BL , Mice, Knockout , Nogo Proteins/metabolism , Nogo Proteins/genetics , Proglucagon/metabolism , Proglucagon/genetics , Proprotein Convertase 1/metabolism , Proprotein Convertase 1/genetics , Protein Binding , Proteolysis
2.
Redox Biol ; 68: 102944, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37890359

ABSTRACT

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.


Subject(s)
Atherosclerosis , Coronary Artery Disease , Hypertension , Plaque, Atherosclerotic , Humans , Animals , Mice , Coronary Artery Disease/genetics , Coronary Artery Disease/metabolism , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , Reactive Oxygen Species/metabolism , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/metabolism , Nogo Proteins/genetics , Nogo Proteins/metabolism , Atherosclerosis/genetics , Atherosclerosis/metabolism , Plaque, Atherosclerotic/metabolism , Oxidative Stress , Human Umbilical Vein Endothelial Cells/metabolism , Inflammation/metabolism , Endothelium/metabolism , Hypertension/metabolism , Apolipoproteins E/genetics , Mice, Knockout , Mice, Inbred C57BL
3.
J Biol Chem ; 299(10): 105232, 2023 10.
Article in English | MEDLINE | ID: mdl-37690690

ABSTRACT

After adult mammalian central nervous system injury, axon regeneration is extremely limited or absent, resulting in persistent neurological deficits. Axon regeneration failure is due in part to the presence of inhibitory proteins, including NogoA (Rtn4A), from which two inhibitory domains have been defined. When these inhibitory domains are deleted, but an amino-terminal domain is still expressed in a gene trap line, mice show axon regeneration and enhanced recovery from injury. In contrast, when there is no amino-terminal Nogo-A fragment in the setting of inhibitory domain deletion, then axon regeneration and recovery are indistinguishable from WT. These data indicated that an amino-terminal Nogo-A fragment derived from the gene trap might promote axon regeneration, but this had not been tested directly and production of this fragment without gene targeting was unclear. Here, we describe posttranslation production of an amino-terminal fragment of Nogo-A from the intact gene product. This fragment is created by proteolysis near amino acid G214-N215 and levels are enhanced by axotomy. Furthermore, this fragment promotes axon regeneration in vitro and acts cell autonomously in neurons, in contrast to the inhibitory extracellular action of other Nogo-A domains.Proteins interacting with the amino-terminal Nogo-A fragment by immunoprecipitation include HSPA8 (HSC70, HSP7C). Suppression of HSPA8 expression by shRNA decreases axon regeneration from cerebral cortical neurons and overexpression increases axon regeneration. Moreover, the amino-terminal Nogo-A fragment increases HSPA8 chaperone activity. These data provide an explanation for varied results in different gene-targeted Nogo-A mice, as well as revealing an axon regeneration promoting domain of Nogo-A.


Subject(s)
Axons , Myelin Proteins , Animals , Mice , Axons/metabolism , Growth Inhibitors/metabolism , Mammals/metabolism , Myelin Proteins/genetics , Myelin Proteins/metabolism , Nerve Regeneration/physiology , Nogo Proteins/genetics , Nogo Proteins/metabolism , Proteolysis , Female , Mice, Inbred C57BL
4.
Int J Mol Sci ; 24(17)2023 Aug 24.
Article in English | MEDLINE | ID: mdl-37685993

ABSTRACT

Endothelial Progenitor Cells (EPCs) can actively participate in revascularization in oxygen-induced retinopathy (OIR). Yet the mechanisms responsible for their dysfunction is unclear. Nogo-A, whose function is traditionally related to the inhibition of neurite function in the central nervous system, has recently been documented to display anti-angiogenic pro-repellent properties. Based on the significant impact of EPCs in retinal vascularization, we surmised that Nogo-A affects EPC function, and proceeded to investigate the role of Nogo-A on EPC function in OIR. The expression of Nogo-A and its specific receptor NgR1 was significantly increased in isolated EPCs exposed to hyperoxia, as well as in EPCs isolated from rats subjected to OIR compared with respective controls (EPCs exposed to normoxia). EPCs exposed to hyperoxia displayed reduced migratory and tubulogenic activity, associated with the suppressed expression of prominent EPC-recruitment factors SDF-1/CXCR4. The inhibition of Nogo-A (using a Nogo-66 neutralizing antagonist peptide) or siRNA-NGR1 in hyperoxia-exposed EPCs restored SDF-1/CXCR4 expression and, in turn, rescued the curtailed neovascular functions of EPCs in hyperoxia. The in vivo intraperitoneal injection of engineered EPCs (Nogo-A-inhibited or NgR1-suppressed) in OIR rats at P5 (prior to exposure to hyperoxia) prevented retinal and choroidal vaso-obliteration upon localization adjacent to vasculature; coherently, the inhibition of Nogo-A/NgR1 in EPCs enhanced the expression of key angiogenic factors VEGF, SDF-1, PDGF, and EPO in retina; CXCR4 knock-down abrogated suppressed NgR1 pro-angiogenic effects. The findings revealed that hyperoxia-induced EPC malfunction is mediated to a significant extent by Nogo-A/NgR1 signaling via CXCR4 suppression; the inhibition of Nogo-A in EPCs restores specific angiogenic growth factors in retina and the ensuing vascularization of the retina in an OIR model.


Subject(s)
Endothelial Progenitor Cells , Hyperoxia , Retinal Diseases , Animals , Rats , Oxygen/adverse effects , Nogo Proteins/genetics , Hyperoxia/complications
5.
J Cell Biol ; 222(7)2023 07 03.
Article in English | MEDLINE | ID: mdl-37318453

ABSTRACT

SARS-CoV-2 depends on host proteins for successful replication. In this issue, Williams et al. (2023. J. Cell Biol.https://doi.org/10.1083/jcb.202203060) report that the ER membrane-modulating proteins RTN3 and RTN4 are required for the formation of SARS-CoV-2 replication organelles via direct interaction with viral proteins NSP3 and NSP4.


Subject(s)
Carrier Proteins , Membrane Proteins , Nerve Tissue Proteins , Nogo Proteins , SARS-CoV-2 , Virus Replication , Humans , Carrier Proteins/genetics , COVID-19 , Endoplasmic Reticulum , Membrane Proteins/genetics , Nerve Tissue Proteins/genetics , Nogo Proteins/genetics , SARS-CoV-2/physiology , Viral Nonstructural Proteins , Coronavirus Papain-Like Proteases
6.
Cell Mol Biol (Noisy-le-grand) ; 69(2): 74-78, 2023 Feb 28.
Article in English | MEDLINE | ID: mdl-37224043

ABSTRACT

Although studies have reported the association of two insertion/deletion (indel) polymorphisms in the 3'-untranslated region (UTR) of the RTN4 gene with the risk of tumorigenesis, the findings are inconsistent and require further explanation. Comprehensive literature searches were undertaken in Pubmed, Embase, Web of Science, China National Knowledge Infrastructure, and WangFang database. The risk of tumorigenesis was determined using odds ratios (ORs) and 95% confidence intervals (CIs) based on STATA 12.0 software. A total of four case-control studies with 1214 patients and 1850 controls focused on the RTN4 gene TATC/- polymorphism and five case-control studies with 1625 patients and 2321 controls on the RTN4 gene CAA/- polymorphism. Pooled analysis showed that the TATC/- polymorphism was not associated with the risk of tumorigenesis under all genetic models and the CAA/- polymorphism was significantly associated with the risk of tumorigenesis under the homozygote genetic model (Del/Del vs. Ins/Ins: OR=1.32, 95%CI=1.04-1.68, P=0.02). In conclusion, the current findings suggested that the CAA/- polymorphism in the 3'-UTR of the RTN4 gene was significantly associated with the risk of tumorigenesis in the Chinese population and may serve as a valuable marker for predicting tumor risk.


Subject(s)
Carcinogenesis , East Asian People , Nogo Proteins , Humans , 3' Untranslated Regions/genetics , Case-Control Studies , East Asian People/genetics , Nogo Proteins/genetics
7.
Cardiovasc Res ; 119(2): 506-519, 2023 03 31.
Article in English | MEDLINE | ID: mdl-35815623

ABSTRACT

AIMS: Growing evidence correlate the accrual of the sphingolipid ceramide in plasma and cardiac tissue with heart failure (HF). Regulation of sphingolipid metabolism in the heart and the pathological impact of its derangement remain poorly understood. Recently, we discovered that Nogo-B, a membrane protein of endoplasmic reticulum, abundant in the vascular wall, down-regulates the sphingolipid de novo biosynthesis via serine palmitoyltransferase (SPT), first and rate liming enzyme, to impact vascular functions and blood pressure. Nogo-A, a splice isoform of Nogo, is transiently expressed in cardiomyocyte (CM) following pressure overload. Cardiac Nogo is up-regulated in dilated and ischaemic cardiomyopathies in animals and humans. However, its biological function in the heart remains unknown. METHODS AND RESULTS: We discovered that Nogo-A is a negative regulator of SPT activity and refrains ceramide de novo biosynthesis in CM exposed to haemodynamic stress, hence limiting ceramide accrual. At 7 days following transverse aortic constriction (TAC), SPT activity was significantly up-regulated in CM lacking Nogo-A and correlated with ceramide accrual, particularly very long-chain ceramides, which are the most abundant in CM, resulting in the suppression of 'beneficial' autophagy. At 3 months post-TAC, mice lacking Nogo-A in CM showed worse pathological cardiac hypertrophy and dysfunction, with ca. 50% mortality rate. CONCLUSION: Mechanistically, Nogo-A refrains ceramides from accrual, therefore preserves the 'beneficial' autophagy, mitochondrial function, and metabolic gene expression, limiting the progression to HF under sustained stress.


Subject(s)
Heart Failure , Sphingolipids , Humans , Mice , Animals , Nogo Proteins/genetics , Nogo Proteins/metabolism , Sphingolipids/metabolism , Ceramides/metabolism , Heart Failure/genetics , Myocytes, Cardiac/metabolism
8.
Neuro Oncol ; 25(6): 1031-1043, 2023 06 02.
Article in English | MEDLINE | ID: mdl-36215168

ABSTRACT

BACKGROUND: IDH mutant gliomas are grouped into astrocytomas or oligodendrogliomas depending on the codeletion of chromosome arms 1p and 19q. Although the genomic alterations of IDH mutant gliomas have been well described, transcriptional changes unique to either tumor type have not been fully understood. Here, we identify Tripartite Motif Containing 67 (TRIM67), an E3 ubiquitin ligase with essential roles during neuronal development, as an oncogene distinctly upregulated in oligodendrogliomas. METHODS: We used several cell lines, including patient-derived oligodendroglioma tumorspheres, to knock down or overexpress TRIM67. We coupled high-throughput assays, including RNA sequencing, total lysate-mass spectrometry (MS), and coimmunoprecipitation (co-IP)-MS with functional assays including immunofluorescence (IF) staining, co-IP, and western blotting (WB) to assess the in vitro phenotype associated with TRIM67. Patient-derived oligodendroglioma tumorspheres were orthotopically implanted in mice to determine the effect of TRIM67 on tumor growth and survival. RESULTS: TRIM67 overexpression alters the abundance of cytoskeletal proteins and induces membrane bleb formation. TRIM67-associated blebbing was reverted with the nonmuscle class II myosin inhibitor blebbistatin and selective ROCK inhibitor fasudil. NOGO-A/Rho GTPase/ROCK2 signaling is altered upon TRIM67 ectopic expression, pointing to the underlying mechanism for TRIM67-induced blebbing. Phenotypically, TRIM67 expression resulted in higher cell motility and reduced cell adherence. In orthotopic implantation models of patient-derived oligodendrogliomas, TRIM67 accelerated tumor growth, reduced overall survival, and led to increased vimentin expression at the tumor margin. CONCLUSIONS: Taken together, our results demonstrate that upregulated TRIM67 induces blebbing-based rounded cell morphology through Rho GTPase/ROCK-mediated signaling thereby contributing to glioma pathogenesis.


Subject(s)
Astrocytoma , Brain Neoplasms , Glioma , Oligodendroglioma , Animals , Mice , Humans , Oligodendroglioma/genetics , Nogo Proteins/genetics , Glioma/pathology , Astrocytoma/genetics , Cell Transformation, Neoplastic , Carcinogenesis , Chromosomes, Human, Pair 1 , Brain Neoplasms/pathology , Chromosomes, Human, Pair 19 , Isocitrate Dehydrogenase/genetics , Mutation , Tripartite Motif Proteins/genetics , Cytoskeletal Proteins/genetics
9.
Dev Neurosci ; 45(1): 8-18, 2023.
Article in English | MEDLINE | ID: mdl-36323241

ABSTRACT

Cerebral palsy (CP) is a motor and postural disorder syndrome caused by the nonprogressive dysfunction of the developing brain. Previous studies strongly indicated that the Nogo-A gene might be related to the pathogenesis of CP. The objective of this research was to explore the relationship between Nogo-A polymorphisms (rs1012603, rs12464595, and rs2864052) and CP in Southern China. The Hardy-Weinberg equilibrium (HWE) testing, allele and genotype frequencies analysis, and haplotype association analysis were applied to the genotyping of 592 CP children and 600 controls. The results showed that the allele and genotype frequencies of rs1012603 of CP group were significantly different from the control group. The haplotype "TTGGG" was significantly associated with an increased risk of CP. The allele frequencies of rs1012603 were significant differences between CP with spastic diplegia, female CP cases, and controls. Furthermore, significant differences in allele and genotype frequencies were also noticed between GMFCS I of CP and controls for rs1012603, and significant differences in allele and genotype frequencies were observed between the ADL (>9) of CP and controls for rs1012603 and rs12464595. This study showed that the SNPs rs1012603 of Nogo-A were significantly correlated with CP, and the correlations were also found in spastic diplegia, GMFCS I of CP, ADL (>9) of CP, and female subgroups, indicating that Nogo-A might mainly affect mild types of CP and there might be sex-related differences.


Subject(s)
Cerebral Palsy , Child , Female , Humans , Case-Control Studies , Cerebral Palsy/genetics , China , Nogo Proteins/genetics , Polymorphism, Single Nucleotide/genetics , Male
10.
Cell Rep ; 41(4): 111505, 2022 10 25.
Article in English | MEDLINE | ID: mdl-36288715

ABSTRACT

Gene-based therapeutic strategies to lower ataxin-2 levels are emerging for the neurodegenerative diseases amyotrophic lateral sclerosis (ALS) and spinocerebellar ataxia type 2 (SCA2). Additional strategies to lower levels of ataxin-2 could be beneficial. Here, we perform a genome-wide arrayed small interfering RNA (siRNA) screen in human cells and identify RTN4R, the gene encoding the RTN4/NoGo-Receptor, as a potent modifier of ataxin-2 levels. RTN4R knockdown, or treatment with a peptide inhibitor, is sufficient to lower ataxin-2 protein levels in mouse and human neurons in vitro, and Rtn4r knockout mice have reduced ataxin-2 levels in vivo. We provide evidence that ataxin-2 shares a role with the RTN4/NoGo-Receptor in limiting axonal regeneration. Reduction of either protein increases axonal regrowth following axotomy. These data define the RTN4/NoGo-Receptor as a novel therapeutic target for ALS and SCA2 and implicate the targeting of ataxin-2 as a potential treatment following nerve injury.


Subject(s)
Amyotrophic Lateral Sclerosis , Spinocerebellar Ataxias , Animals , Mice , Humans , Ataxin-2/genetics , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , RNA, Small Interfering , Nogo Receptors/metabolism , Spinocerebellar Ataxias/genetics , Mice, Knockout , Peptides/metabolism , Nogo Proteins/genetics , Nogo Proteins/metabolism
11.
PLoS One ; 17(6): e0269404, 2022.
Article in English | MEDLINE | ID: mdl-35771867

ABSTRACT

Micropeptides, encoded from small open reading frames of 300 nucleotides or less, are hidden throughout mammalian genomes, though few functional studies of micropeptides in the brain are published. Here, we describe a micropeptide known as the Plasticity-Associated Neural Transcript Short (Pants), located in the 22q11.2 region of the human genome, the microdeletion of which conveys a high risk for schizophrenia. Our data show that Pants is upregulated in early adulthood in the mossy fiber circuit of the hippocampus, where it exerts a powerful negative effect on long-term potentiation (LTP). Further, we find that Pants is secreted from neurons, where it associates with synapses but is rapidly degraded with stimulation. Pants dynamically interacts with Rtn4/Nogo-A, a well-studied regulator of adult plasticity. Pants interaction with Nogo-A augments its influence over postsynaptic AMPA receptor clustering, thus gating plasticity at adult synapses. This work shows that neural micropeptides can act as architectural modules that increase the functional diversity of the known proteome.


Subject(s)
Long-Term Potentiation , Neuronal Plasticity , Adult , Hippocampus/metabolism , Humans , Long-Term Potentiation/physiology , Neuronal Plasticity/physiology , Nogo Proteins/genetics , Nogo Proteins/metabolism , Peptides/metabolism , Synapses/metabolism
12.
Zhen Ci Yan Jiu ; 47(5): 401-8, 2022 May 25.
Article in Chinese | MEDLINE | ID: mdl-35616413

ABSTRACT

OBJECTIVE: To observe the effect of moxibustion treatment on the expression of Nogo-A, Nogo receptor (NgR), neurotrophin receptor p75 (p75NTR) and leucine rich repeat and Ig domain containing 1 (Lingo-1) in brain tissue of rats with cerebral ischemia/reperfusion injury (CI/RI), so as to analyze its mechanism underlying improvement of CI/RI. METHODS: Male SD rats were randomly divided into sham operation group (16 rats), model group (17 rats), NEP1-40 (extracellular peptide residues 1-40, a blocker targeting NgR) group (model+blocker, 17 rats) and moxibustion group (model+moxibustion, 17 rats). The CI/RI model was established by occlusion of the left middle cerebral artery (MCAO). Moxibustion was applied to "Baihui"(GV20), right "Quchi"(LI11) and "Zusanli"(ST36) for 20 min, once a day for 14 days, with 2 days' rest after the top 7 days' intervention. For rats of the NEP1-40 group, 30 µL PBS containing 18 µg NEP 1-40 was injected into the epidural inferior vena (L5-S1) via a polyvinyl chloride conduit. The neurological deficit state in each group was evaluated by Longa's 5-point scale and Feeney's 7-point scale of beam walking test (BWT). The cerebral infarct volume was assessed by 2,3,5-triphenyltetrazole chloride staining. The brain tissue between the central anterior and posterior sulcus was taken for observing the expression of NgR and Lingo-1 by fluorescence double-label method, and for determining the expression levels of Nogo-A, NgR, p75NTR and Lingo-1 mRNAs and proteins by real-time quantitative PCR and Western blot, respectively. RESULTS: After modeling, the Longa's score, infarct volu-me percent, expression levels of Nogo-A, NgR, Lingo-1 and p75NTR mRNAs and proteins were significantly increased (P<0.01) and BWT score was obviously decreased (P<0.01) in the model group relevant to the sham operation group. In comparison with the model group, the increase of Longa's score, infarct volume percentage, expression levels of Nogo-A, NgR, Lingo-1 and p75NTR mRNAs and proteins and decrease of BWT score in NEP1-40 and moxibustion groups were reversed (P<0.01) except Nogo-A protein in the NEP1-40 group. The effect of moxibustion was significantly superior to that of blocker NEP1-40 in redu-cing the infarct volume percentage, and down-regulating the expression of Nogo-A mRNA and protein, p75NTR mRNA and protein, NgR and Lingo-1 proteins (P<0.01, P<0.05). CONCLUSION: Moxibustion, similar to blocker NEP1-40 of NgR, can improve neurological dysfunction in CI/RI rats, which may be related to its functions in reducing cerebral infarction and down-regulating the activity of Nogo/neurotrophin receptor signaling pathway.


Subject(s)
Brain Ischemia , Electroacupuncture , Moxibustion , Reperfusion Injury , Animals , Brain Ischemia/genetics , Brain Ischemia/therapy , Cerebral Infarction , Male , Nogo Proteins/genetics , RNA, Messenger , Rats , Rats, Sprague-Dawley , Receptors, Nerve Growth Factor , Reperfusion Injury/genetics , Reperfusion Injury/therapy , Signal Transduction
13.
Sheng Li Xue Bao ; 74(2): 301-308, 2022 Apr 25.
Article in Chinese | MEDLINE | ID: mdl-35503078

ABSTRACT

Nogo-B receptor (NgBR) is a specific receptor of Nogo-B, a member of reticulon 4 protein family. It is widely expressed in many tissues and mainly located in cell membrane and endoplasmic reticulum. Previous studies have revealed that NgBR is involved in a variety of physiological and pathophysiological processes, such as dolichol synthesis, lipid metabolism, cholesterol trafficking, insulin resistance, vascular remodeling and angiogenesis, tumorigenesis and nervous system diseases. Further studies on the molecular characteristics and biological function of NgBR might be of great significance to understand its role in diverse diseases and provide possible clinical strategies for the treatment of diseases.


Subject(s)
Endoplasmic Reticulum , Receptors, Cell Surface , Carrier Proteins/metabolism , Endoplasmic Reticulum/metabolism , Lipid Metabolism , Nogo Proteins/genetics , Nogo Proteins/metabolism , Receptors, Cell Surface/metabolism
14.
Mol Cancer ; 21(1): 10, 2022 01 04.
Article in English | MEDLINE | ID: mdl-34983537

ABSTRACT

BACKGROUND: Circular RNAs (circRNAs) play important roles in many biological processes. However, the detailed mechanism underlying the critical roles of circRNAs in cancer remains largely unexplored. We aim to explore the molecular mechanisms of circRTN4 with critical roles in pancreatic ductal adenocarcinoma (PDAC). METHODS: CircRTN4 expression level was examined in PDAC primary tumors. The oncogenic roles of circRTN4 in PDAC tumor growth and metastasis were studied in mouse tumor models. Bioinformatics analysis, luciferase assay and miRNA pulldown assay were performed to study the novel circRTN4-miRNA-lncRNA pathway. To identify circRTN4-interacting proteins, we performed circRNA-pulldown and mass spectrometry in PDAC cells. Protein stability assay and 3-Dimensional structure modeling were performed to reveal the role of circRTN4 in stabilizing RAB11FIP1. RESULTS: CircRTN4 was significantly upregulated in primary tumors from PDAC patients. In vitro and in vivo functional studies revealed that circRTN4 promoted PDAC tumor growth and liver metastasis. Mechanistically, circRTN4 interacted with tumor suppressor miR-497-5p in PDAC cells. CircRTN4 knockdown upregulated miR-497-5p to inhibit the oncogenic lncRNA HOTTIP expression. Furthermore, we identified critical circRTN4-intercting proteins by circRNA-pulldown in PDAC cells. CircRTN4 interacted with important epithelial-mesenchymal transition (EMT)- driver RAB11FIP1 to block its ubiquitination site. We found that circRTN4 knockdown promoted the degradation of RAB11FIP1 by increasing its ubiquitination. Also, circRTN4 knockdown inhibited the expression of RAB11FIP1-regulating EMT-markers Slug, Snai1, Twist, Zeb1 and N-cadherin in PDAC. CONCLUSION: The upregulated circRTN4 promotes tumor growth and liver metastasis in PDAC through the novel circRTN4-miR-497-5p-HOTTIP pathway. Also, circRTN4 stabilizes RAB11FIP1 to contribute EMT.


Subject(s)
Epithelial-Mesenchymal Transition , MicroRNAs/genetics , Nogo Proteins/genetics , Pancreatic Neoplasms/etiology , Pancreatic Neoplasms/metabolism , RNA, Circular , RNA, Long Noncoding/genetics , Adult , Aged , Animals , Biomarkers, Tumor , Cell Line, Tumor , Disease Models, Animal , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Middle Aged , Neoplasm Grading , Neoplasm Staging , Pancreatic Neoplasms/pathology , RNA Interference
15.
Nat Commun ; 12(1): 6906, 2021 11 25.
Article in English | MEDLINE | ID: mdl-34824275

ABSTRACT

Astrocytes play critical roles after brain injury, but their precise function is poorly defined. Utilizing single-nuclei transcriptomics to characterize astrocytes after ischemic stroke in the visual cortex of the marmoset monkey, we observed nearly complete segregation between stroke and control astrocyte clusters. Screening for the top 30 differentially expressed genes that might limit stroke recovery, we discovered that a majority of astrocytes expressed RTN4A/ NogoA, a neurite-outgrowth inhibitory protein previously only associated with oligodendrocytes. NogoA upregulation on reactive astrocytes post-stroke was significant in both the marmoset and human brain, whereas only a marginal change was observed in mice. We determined that NogoA mediated an anti-inflammatory response which likely contributes to limiting the infiltration of peripheral macrophages into the surviving parenchyma.


Subject(s)
Astrocytes/metabolism , Brain Injuries/metabolism , Macrophages/metabolism , Nogo Proteins/metabolism , Animals , Callithrix , Female , GAP-43 Protein , Membrane Glycoproteins , Membrane Proteins , Mice , Mice, Inbred C57BL , Nogo Proteins/genetics , Oligodendroglia , Receptors, Immunologic , Solitary Nucleus , Stroke , Transcriptome , Up-Regulation , Visual Cortex
16.
Int J Mol Sci ; 22(19)2021 Sep 23.
Article in English | MEDLINE | ID: mdl-34638567

ABSTRACT

Dopaminergic treatment in combination with rehabilitative training enhances long-term recovery after stroke. However, the underlying mechanisms on structural plasticity are unknown. Here, we show an increased dopaminergic innervation of the ischemic territory during the first week after stroke induced in Wistar rats subjected to transient occlusion of the middle cerebral artery (tMCAO) for 120 min. This response was also found in rats subjected to permanent focal ischemia induced by photothrombosis (PT) and mice subjected to PT or tMCAO. Dopaminergic branches were detected in the infarct core of mice and rats in both stroke models. In addition, the Nogo A pathway was significantly downregulated in rats treated with levodopa (LD) compared to vehicle-treated animals subjected to tMCAO. Specifically, the number of Nogo A positive oligodendrocytes as well as the levels of Nogo A and the Nogo A receptor were significantly downregulated in the peri-infarct area of LD-treated animals, while the number of Oligodendrocyte transcription factor 2 positive cells increased in this region after treatment. In addition, we observed lower protein levels of Growth Associated Protein 43 in the peri-infarct area compared to sham-operated animals without treatment effect. The results provide the first evidence of the plasticity-promoting actions of dopaminergic treatment following stroke.


Subject(s)
Dopamine Agents/pharmacology , Dopamine Agents/therapeutic use , Levodopa/pharmacology , Levodopa/therapeutic use , Neuronal Plasticity/drug effects , Stroke/drug therapy , Animals , Brain Ischemia/etiology , Brain Ischemia/metabolism , Brain Ischemia/pathology , Disease Models, Animal , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Down-Regulation/drug effects , GAP-43 Protein/metabolism , Infarction, Middle Cerebral Artery/complications , Male , Mice , Nogo Proteins/genetics , Nogo Proteins/metabolism , Nogo Receptors/metabolism , Oligodendrocyte Transcription Factor 2/metabolism , Oligodendroglia/drug effects , Oligodendroglia/metabolism , Rats, Wistar , Stroke/etiology , Stroke/metabolism , Thrombosis/complications
17.
Int J Mol Sci ; 22(19)2021 Sep 26.
Article in English | MEDLINE | ID: mdl-34638704

ABSTRACT

The microtubule, a major constituent of cytoskeletons, was shown to bind and interact with transient receptor potential vanilloid subfamily member 1 (TRPV1), and serves a pivotal role to produce thermal hyperalgesia in inflammatory pain. Nogo-A is a modulator of microtubule assembly and plays a key role in maintaining the function of TRPV1 in inflammatory heat pain. However, whether the microtubule dynamics modulated by Nogo-A in dorsal root ganglion (DRG) neurons participate in the inflammatory pain is not elucidated. Here we reported that the polymerization of microtubules in the DRG neurons, as indicated by the acetylated α-tubulin, tubulin polymerization-promoting protein 3 (TPPP3), and microtubule numbers, was significantly elevated in the complete Freund's adjuvant (CFA) induced inflammatory pain. Consistent with our previous results, knock-out (KO) of Nogo-A protein significantly attenuated the heat hyperalgesia 72 h after CFA injection and decreased the microtubule polymerization via up-regulation of phosphorylation of collapsin response mediator protein 2 (CRMP2) in DRG. The colocalization of acetylated α-tubulin and TRPV1 in DRG neurons was also reduced dramatically in Nogo-A KO rats under inflammatory pain. Moreover, the down-regulation of TRPV1 in DRG of Nogo-A KO rats after injection of CFA was reversed by intrathecal injection of paclitaxel, a microtubule stabilizer. Furthermore, intrathecal injection of nocodazole (a microtubule disruptor) attenuated significantly the CFA-induced inflammatory heat hyperalgesia and the mechanical pain in a rat model of spared nerve injury (SNI). In these SNI cases, the Nogo-A and acetylated α-tubulin in DRG were also significantly up-regulated. We conclude that the polymerization of microtubules promoted by Nogo-A in DRG contributes to the development of inflammatory heat hyperalgesia mediated by TRPV1.


Subject(s)
Ganglia, Spinal/metabolism , Hyperalgesia/metabolism , Neurons/metabolism , Nogo Proteins/metabolism , Pain/metabolism , Animals , Gene Knockdown Techniques , Hyperalgesia/genetics , Inflammation/genetics , Inflammation/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Male , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nogo Proteins/genetics , Pain/genetics , Rats , Rats, Sprague-Dawley , Rats, Transgenic , TRPV Cation Channels/genetics , TRPV Cation Channels/metabolism
18.
Zhen Ci Yan Jiu ; 46(9): 721-7, 2021 Sep 25.
Article in Chinese | MEDLINE | ID: mdl-34558236

ABSTRACT

OBJECTIVE: To observe the effect of electroacupuncture(EA)on the expression of myelin basic protein (MBP), axon growth inhibitor Nogo-A and Nogo receptor (NgR) in corpus callosum of rats with focal cerebral ischemia, so as to explore the mechanism of EA underlying improving ischemic white matter injury. METHODS: Fourty male SD rats were randomly divided into normal, sham operation, model and EA groups, with 10 rats in each group. The focal cerebral ischemia rat model was established by occlusion of the middle cerebral artery (MCAO). EA was applied to "Baihui"(GV20) and "Zusanli"(ST36) on the left side for 30 min, once daily for 14 days. Neurological function score and the adhensive removal test were used to evaluate neurological deficit severity; Hematoxylin-esion staining was used to observe the pathological changes in myelin of corpus callosum and luxol fast blue(LFB) staining was used to observe the myelin of corpus callosum. Immunohistochemical staining and Western blot were used to detect the expressions of MBP、Nogo-A and NgR in the ischemic corpus callosum. RESULTS: After MCAO, the neurological function score was significantly increased (P<0.05), the time required for contact with tape and tape removal was longer (P<0.001), the intensity of LFB staining and the expression of MBP decreased, while the veside area and the expression of Nogo-A and its receptor NgR increased (P<0.01, P<0.05) in the model group relevant to the normal and sham operation groups. The fiber arrangement of the corpus callosum on the ischemic side was disordered and a large amount of myelin sheath was lost in the model group. Following the treatment, the neurological deficit score of EA group was gradually decreased and significantly decreased on the 3rd, 7th and 14th day (P<0.05), and the time to remove the adhesive tape was shortened at the 7th and 14th day (P<0.001). The shape of the corpus callosum in the EA group was close to normal, and the myelin structure was relatively complete. The intensity of LFB staining and the expression of MBP was increased (P<0.05, P<0.01) while the expression of Nogo-A and its receptor NgR were decreased in the EA group relevant to the model group (P<0.01). CONCLUSION: EA can play a protective role in myelin of the corpus callosum after cerebral ischemia, which may be related to down-regulating the expressions of Nogo-A and NgR.


Subject(s)
Brain Ischemia , Electroacupuncture , Animals , Brain Ischemia/genetics , Brain Ischemia/therapy , Corpus Callosum , Male , Myelin Sheath , Nogo Proteins/genetics , Rats , Rats, Sprague-Dawley
19.
J Ethnopharmacol ; 281: 114559, 2021 Dec 05.
Article in English | MEDLINE | ID: mdl-34461189

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Naoluoxintong (NLXT) is a traditional Chinese Medicine (TCM) prescription that is clinically used in the treatment of ischemic stroke (IS). However, its therapeutic mechanism remains unclear. AIM OF THE STUDY: To obtain the mechanism of NLXT by observing the protective effects of NLXT on the NogoA/RhoA/Rock pathway in a rat model of IS by regulating DNA methylation. MATERIALS AND METHODS: Rats were divided into five groups using a random number table: normal group, model group, NLXT group, blocker group I (NLXT + SGI-1027) and blocker group II (NLXT + Y27632). The right middle cerebral artery occlusion-reperfusion (MCAO/R) rat model was made, and the regional cerebral blood flow (rCBF) of each group was detected using laser Doppler. The methylation levels of CpG sites of neurite outgrowth inhibitor protein-A (Nogo-A), Nogo receptor (NgR), ras homolog gene family member A (RhoA) and rho-associated coiled-coil protein kinase 2 (ROCK2) genes in rat brain tissue were detected using the bisulfite method. Reverse transcription-polymerase chain reaction (RT-PCR) was used to detect NogoA, RhoA, NgR1, NgR2 and ROCK2 mRNA expression in rat brain tissue. NogoA, RhoA, NgR1, NgR2 and ROCK2 proteins were detected using immunoblotting in rat brain tissue. RESULTS: After the modeling of middle cerebral artery occlusion (MCAO), neurological deficit test was made to ensure the success of the modeling. At each time point after surgery, the rCBF of the other groups decreased compared with the normal group (P < 0.01 or P < 0.05). Meanwhile, the rCBF increased in blocker group I as well as blocker group II after 3 days (P < 0.05). There were differences in the DNA methylation sites of NogoA, RhoA, NgR and ROCK2 genes between the model group and the NLXT group (P < 0.05). Compared with the normal group, NogoA, NgR1, NgR2, RhoA and ROCK2 gene expression in the model group increased observably (P < 0.01). In comparison with the model group, NogoA and NgR1 gene expression in the blocker group II was prominently observed on the 1st day. NogoA, NgR1, NgR2, RhoA and ROCK2 gene expression remarkably reduced (P < 0.01) on the 3rd and 7th days. Compared with the normal group, NogoA, RhoA, NgR1, NgR2 and ROCK2 protein expression in the model group increased observably (P < 0.01). In comparison with the model group, NogoA, RhoA, NgR1, NgR2 and ROCK2 protein expression in the other groups declined prominently (P < 0.01). CONCLUSION: NLXT can reduce the DNA methylation level of NogoA pathway after IS, thus inhibit the expression of NogoA/RhoA/ROCK pathway from producing anti-cerebral ischemia pharmacological effect.


Subject(s)
Down-Regulation/drug effects , Drugs, Chinese Herbal/pharmacology , Nogo Proteins/metabolism , rho GTP-Binding Proteins/metabolism , rho-Associated Kinases/metabolism , Animals , DNA/metabolism , DNA Methylation , Gene Expression Regulation/drug effects , Infarction, Middle Cerebral Artery , Male , Neuroprotective Agents/pharmacology , Nogo Proteins/genetics , Random Allocation , Rats , Rats, Sprague-Dawley , rho GTP-Binding Proteins/genetics , rho-Associated Kinases/genetics
20.
J Mol Histol ; 52(5): 955-964, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34406553

ABSTRACT

Heart failure (HF) is a rising epidemic and public health burden in modern society. It is of great need to find new biomarkers to ensure a timely diagnosis and to improve treatment and prognosis of the disease. The mouse model of HF was established by thoracic aortic constriction. Color Doppler ultrasound was performed to detect left ventricular end-diastolic diameter. Hematoxylin and eosin staining was conducted to observe the pathological changes of mouse myocardium. The RT-qPCR analysis was performed to detect miR-590-5p and RTN4 expression levels. Western blot was conducted to detect protein levels of the indicated genes. We found that the expression of miR-590-5p was downregulated in cardiac tissues of HF mice. Injection of AAV-miR-590-5p attenuated myocardium hypertrophy and myocyte apoptosis. Additionally, miR-590-5p overexpression promoted viability, inhibited apoptosis, and decreased ANF, BNP and beta-MHC protein levels in H9c2 cell. Mechanistically, miR-590-5p binds to RTN4 3'-untranslated region, as predicted by starBase online database and evidenced by luciferase reporter assay. Furthermore, miR-590-5p negatively regulates RTN4 mRNA expression and suppresses its translation. The final rescue experiments revealed that miR-590-5p modulated cardiomyocyte phenotypes by binding to RTN4. In conclusion, miR-590-5p modulates myocardium hypertrophy and myocyte apoptosis in HF by downregulating RTN4.


Subject(s)
Cardiomegaly/complications , Cardiomegaly/genetics , Heart Failure/complications , Heart Failure/genetics , MicroRNAs/metabolism , Nogo Proteins/metabolism , Angiotensin II , Animals , Apoptosis/genetics , Base Sequence , Biomarkers/metabolism , Cardiomegaly/pathology , Cell Line , Disease Models, Animal , Down-Regulation/genetics , Mice, Inbred C57BL , MicroRNAs/genetics , Nogo Proteins/genetics , Protein Binding
SELECTION OF CITATIONS
SEARCH DETAIL