Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters

Country/Region as subject
Affiliation country
Publication year range
1.
FASEB J ; 31(4): 1482-1493, 2017 04.
Article in English | MEDLINE | ID: mdl-28003340

ABSTRACT

ADAM10 (a disintegrin and metalloproteinase domain-containing protein 10) is the α-secretase that is involved in APP (ß-amyloid precursor protein) processing. Enhancement of the nonamyloidogenic APP pathway by ADAM10 provides therapeutic potential for Alzheimer's disease (AD). By using high-throughput screening that targeted ADAM10, we determined that apicidin-an inhibitor of HDACs (histone deacetylases)-significantly increased mRNA and protein levels of ADAM10 in SH-SY5Y cells. A luciferase assay revealed that the nucleotides -444 to -300 in the ADAM10 promoter were sufficient to mediate this effect. In addition, knockdown of USF1 (upstream transcription factor 1) and HDAC2/3 prevented apicidin regulation of ADAM10. Moreover, USF1 acetylation was increased by apicidin, which enhanced the association of USF1 with HDAC2/3 and with the ADAM10 promoter. We further found that apicidin did not affect the phosphorylation of ERK or USF1; however, ERK inhibitor U0126 blocked the effect of apicidin on ADAM10. Finally, apicidin increased the level of α-site C-terminal fragment from APP and reduced the production of ß-amyloid peptide 1-42. Collectively, our study provides evidence that ADAM10 expression can be regulated by HDAC2/3 inhibitor apicidin via USF1-dependent mechanisms in which ERK signaling plays an important role. Thus, HDAC regulation of ADAM10 might shed new light on the understanding of AD pathology.-Hu, X.-T., Zhu, B.-L., Zhao, L.-G., Wang, J.-W., Liu, L., Lai, Y.-J., He, L., Deng, X.-J., Chen, G.-J. Histone deacetylase inhibitor apicidin increases expression of the α-secretase ADAM10 through transcription factor USF1-mediated mechanisms.


Subject(s)
ADAM10 Protein/genetics , Amyloid Precursor Protein Secretases/genetics , Histone Deacetylase Inhibitors/pharmacology , Membrane Proteins/genetics , Peptides, Cyclic/pharmacology , Upstream Stimulatory Factors/metabolism , ADAM10 Protein/metabolism , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Cell Line, Tumor , Humans , Membrane Proteins/metabolism , Mice , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcriptional Activation/drug effects , Up-Regulation/drug effects , Upstream Stimulatory Factors/genetics
2.
Biochem Biophys Res Commun ; 486(2): 492-498, 2017 04 29.
Article in English | MEDLINE | ID: mdl-28320515

ABSTRACT

HMGCS2 (mitochondrial 3-hydroxy-3-methylglutaryl-COA synthase 2) is a control enzyme in ketogenesis. The mitochondrial localization and interaction with APP (ß-amyloid precursor protein) suggest that HMGCS2 may play a role in the pathophysiology of AD (Alzheimer's disease). Here we report that overexpression of HMGCS2 decreased levels of APP and related CTFs (carboxy-terminal fragments), which was largely prevented by an autophagic inhibitor chloroquine. In addition, HMGCS2 enhancement of autophagic marker LC3II was diminished by rapamycin, an inhibitor of mechanistic target of rapamycin. Moreover, deprivation of EBSS (Earle's Balanced Salt Solution) significantly augmented the effect of HMGCS2 on LC3II, while acetoacetate reversed the reduction of LC3II, APP and CTFs which was induced by HMGCS2 knockdown. In the presence of acetoacetate, rapamycin failed to induce further increase of LC3II, which mimicked the effect of HMGCS2 overexpression. Finally, HMGCS2 enhanced the antioxidant response. Collectively, HMGCS2 shares with ketone bodies common features in autophagic clearance of APP and CTFs, suggesting that ketone bodies play an important role in HMGCS2 regulation of the autophagy.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Autophagy/genetics , Hydroxymethylglutaryl-CoA Synthase/genetics , Ketone Bodies/metabolism , Microtubule-Associated Proteins/genetics , TOR Serine-Threonine Kinases/genetics , Acetoacetates/pharmacology , Animals , Cell Line , Chloroquine/pharmacology , Gene Expression Regulation , HEK293 Cells , Hippocampus/cytology , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Hydroxymethylglutaryl-CoA Synthase/antagonists & inhibitors , Hydroxymethylglutaryl-CoA Synthase/metabolism , Mice , Microtubule-Associated Proteins/metabolism , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Proteolysis/drug effects , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Transgenes
3.
Cell Mol Neurobiol ; 35(7): 1027-37, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25971983

ABSTRACT

Chronic inflammation appears to play a critical role in sickness behavior caused by diabetes mellitus. Astaxanthin has been used in treating diabetes mellitus and diabetic complications because of its neuroprotective and anti-inflammatory actions. However, whether astaxanthin can improve sickness behavior induced by diabetes and its potential mechanisms are still unknown. The aim of this study was to investigate the effects of astaxanthin on diabetes-elicited abnormal behavior in mice and its corresponding mechanisms. An experimental diabetic model was induced by streptozotocin (150 mg/kg) and astaxanthin (25 mg/kg/day) was provided orally for 10 weeks. Body weight and water consumption were measured, and the sickness behavior was evaluated by the open field test (OFT) and closed field test (CFT). The expression of glial fibrillary acidic protein (GFAP) was measured, and the frontal cortical cleaved caspase-3 positive cells, interleukin-6 (IL-6), and interleukin-1ß (IL-1ß) expression levels were also investigated. Furthermore, cystathionine ß-synthase (CBS) in the frontal cortex was detected to determine whether the protective effect of astaxanthin on sickness behavior in diabetic mice is closely related to CBS. As expected, we observed that astaxanthin improved general symptoms and significantly increase horizontal distance and the number of crossings in the OFT and CFT. Furthermore, data showed that astaxanthin could decrease GFAP-positive cells in the brain and down-regulate the cleaved caspase-3, IL-6, and IL-1ß, and up-regulate CBS in the frontal cortex. These results suggest that astaxanthin provides neuroprotection against diabetes-induced sickness behavior through inhibiting inflammation, and the protective effects may involve CBS expression in the brain.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Diabetes Mellitus, Experimental/drug therapy , Illness Behavior/drug effects , Animals , Anti-Inflammatory Agents/pharmacology , Body Weight/drug effects , Body Weight/physiology , Brain/drug effects , Brain/metabolism , Brain/pathology , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Drinking/drug effects , Drinking/physiology , Illness Behavior/physiology , Male , Mice , Mice, Inbred ICR , Treatment Outcome , Xanthophylls/pharmacology , Xanthophylls/therapeutic use
4.
Adv Sci (Weinh) ; 11(11): e2305260, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38183387

ABSTRACT

It is long been suggested that one-carbon metabolism (OCM) is associated with Alzheimer's disease (AD), whereas the potential mechanisms remain poorly understood. Taking advantage of chemical biology, that mitochondrial serine hydroxymethyltransferase (SHMT2) directly regulated the translation of ADAM metallopeptidase domain 10 (ADAM10), a therapeutic target for AD is reported. That the small-molecule kenpaullone (KEN) promoted ADAM10 translation via the 5' untranslated region (5'UTR) and improved cognitive functions in APP/PS1 mice is found. SHMT2, which is identified as a target gene of KEN and the 5'UTR-interacting RNA binding protein (RBP), mediated KEN-induced ADAM10 translation in vitro and in vivo. SHMT2 controls AD signaling pathways through binding to a large number of RNAs and enhances the 5'UTR activity of ADAM10 by direct interaction with GAGGG motif, whereas this motif affected ribosomal scanning of eukaryotic initiation factor 2 (eIF2) in the 5'UTR. Together, KEN exhibits therapeutic potential for AD by linking OCM with RNA processing, in which the metabolic enzyme SHMT2 "moonlighted" as RBP by binding to GAGGG motif and promoting the 5'UTR-dependent ADAM10 translation initiation.


Subject(s)
Alzheimer Disease , Glycine Hydroxymethyltransferase , Animals , Mice , 5' Untranslated Regions , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Glycine Hydroxymethyltransferase/genetics , RNA, Messenger/genetics
5.
Chin J Cancer ; 32(1): 31-41, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22059906

ABSTRACT

Gastric cancer is one of the most common malignancies and a leading cause of cancer mortality worldwide. The pathogenesis mechanisms of gastric cancer are still not fully clear. Inactivation of tumor suppressor genes and activation of oncogenes caused by genetic and epigenetic alterations are known to play significant roles in carcinogenesis. Accumulating evidence has shown that epigenetic silencing of the tumor suppressor genes, particularly caused by hypermethylation of CpG islands in promoters, is critical to carcinogenesis and metastasis. Here, we review the recent progress in the study of methylations of tumor suppressor genes involved in the pathogenesis of gastric cancer. We also briefly describe the mechanisms that induce tumor suppressor gene methylation and the status of translating these molecular mechanisms into clinical applications.


Subject(s)
DNA Methylation , Epigenesis, Genetic , Genes, Tumor Suppressor , Stomach Neoplasms/genetics , Tumor Suppressor Proteins/genetics , Apoptosis , Cell Adhesion , Cell Cycle , CpG Islands/genetics , DNA Repair , Helicobacter Infections/genetics , Helicobacter pylori , Humans , Neoplasm Invasiveness , Promoter Regions, Genetic/genetics , Stomach Neoplasms/metabolism , Stomach Neoplasms/microbiology , Stomach Neoplasms/pathology , Tumor Suppressor Proteins/metabolism
6.
Asian J Psychiatr ; 86: 103680, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37352754

ABSTRACT

Helicobacter pylori (H. pylori) infection confers risk for Alzheimer's Disease (AD), with the mechanisms unknown. Infections are linked to the etiology of AD partly through modulating the humoral immunity post-infection. This study found increased plasma levels of tTau and pTau181 in H. Pylori infected individuals with intact cognition. Plasma antibodies to H. pylori were positively associated with Aß40, Aß42, tTau, and pTau181, adjusting for age, sex, education level, BMI, ApoE ε4 genotype, hypertension, diabetes mellitus, and hypercholesteremia. This study presents novel insights into the relationship between H. pylori infection and AD from an autoimmune perspective.


Subject(s)
Alzheimer Disease , Diabetes Mellitus , Helicobacter Infections , Helicobacter pylori , Humans , Adult , Biomarkers , Helicobacter Infections/complications
7.
CNS Neurosci Ther ; 29(5): 1300-1311, 2023 05.
Article in English | MEDLINE | ID: mdl-36708130

ABSTRACT

AIMS: Amyloid beta (Aß) is an important pathological feature of Alzheimer's disease (AD). A disintegrin and metalloproteinase 10 (ADAM10) can reduce the production of toxic Aß by activating the nonamyloidogenic pathway of amyloid precursor protein (APP). We previously found that apicidin, which is a histone deacetylase (HDAC) inhibitor, can promote the expression of ADAM10 and reduce the production of Aß in vitro. This study was designed to determine the potential of apicidin treatment to reverse learning and memory impairments in an AD mouse model and the possible correlation of these effects with ADAM10. METHODS: Nine-month-old APP/PS1 mice and C57 mice received intraperitoneal injections of apicidin or vehicle for 2 months. At 11 months of age, we evaluated the memory performance of mice with Morris water maze (MWM) and context fear conditioning tests. The Aß levels were assessed in mouse brain using the immunohistochemical method and ELISA. The expression of corresponding protein involved in proteolytic processing of APP and the phosphorylation of tau were assessed by Western blotting. RESULTS: Apicidin reversed the deficits of spatial reference memory and contextual fear memory, attenuated the formation of Aß-enriched plaques, and decreased the levels of soluble and insoluble Aß40/42 in APP/PS1 mice. Moreover, apicidin significantly increased the expression of ADAM10, improved the level of sAPPα, and reduced the production of sAPPß, but did not affect the levels of phosphorylated tau in APP/PS1 mice. CONCLUSION: Apicidin significantly improves the AD symptoms of APP/PS1 mice by regulating the expression of ADAM10, which may contribute to decreasing the levels of Aß rather than decreasing the phosphorylation of tau.


Subject(s)
Alzheimer Disease , Amyloid beta-Protein Precursor , Mice , Animals , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Amyloid beta-Peptides/metabolism , Mice, Transgenic , Alzheimer Disease/metabolism , Memory Disorders/drug therapy , Memory Disorders/etiology , Memory Disorders/metabolism , Spatial Memory , Disease Models, Animal , Presenilin-1/genetics , Presenilin-1/metabolism
8.
Blood ; 115(12): 2458-61, 2010 Mar 25.
Article in English | MEDLINE | ID: mdl-20093404

ABSTRACT

Using inverse polymerase chain reaction, we identified CD44, located on chromosome 11p13, as a novel translocation partner of IGH in 9 of 114 cases of gastric, nongastric extranodal, follicular, and nodal diffuse large B-cell lymphoma (DLBCL). Notably, these translocations involving IGHSmu were detected in follicular lymphomas and exclusively in germinal center B cell-ike (GCB)-DLBCLs. CD44 is not expressed in reactive GC B cells. The IGHSmu/CD44 translocations substitute Smu for the CD44 promoter and remove exon 1 of CD44, resulting in the overexpression of Imu-CD44 hybrid mRNA transcripts activated from derivative 11 that encode a new CD44 variant lacking the leader peptide and with a unique C-terminus (CD44DeltaEx1). When overexpressed in vitro in the CD44(-) GCB-DLBCL cell line BJAB, CD44DeltaEx1-green fluorescent protein localized to the cytoplasm and nucleus, whereas CD44s-green fluorescent protein (standard form) localized to the plasma membrane. The ectopic expression of CD44DeltaEx1 in BJAB cells enhanced their proliferation rate and clonogenic ability, indicating a possible pathogenic role of the translocation.


Subject(s)
Hyaluronan Receptors/genetics , Immunoglobulin Heavy Chains/genetics , Lymphoma, Follicular/genetics , Lymphoma, Large B-Cell, Diffuse/genetics , Stomach Neoplasms/genetics , Translocation, Genetic , Cell Line, Tumor , Chromosome Breakpoints , Chromosomes, Human, Pair 11 , Chromosomes, Human, Pair 14 , Gene Expression Regulation, Neoplastic , Green Fluorescent Proteins/genetics , Humans , Hyaluronan Receptors/metabolism , Lymphoma, Follicular/pathology , Lymphoma, Large B-Cell, Diffuse/pathology , Polymerase Chain Reaction , Stomach Neoplasms/pathology
9.
Eur J Med Chem ; 225: 113762, 2021 Dec 05.
Article in English | MEDLINE | ID: mdl-34411893

ABSTRACT

Neuraminidase (NA) is an important target for the treatment of influenza. In this study, a new lead NA inhibitor, 4 (ZINC01121127), was discovered by pharmacophore-based virtual screening and molecular dynamic (MD) simulation. Some novel NA inhibitors containing thiophene ring were synthesized by optimizing the skeleton of the lead compound 4. Compound 4b had the most potent inhibitory activity against NA (IC50 = 0.03 µM), which was better than the positive control oseltamivir carboxylate (IC50 = 0.06 µM). 4b (EC50 = 1.59 µM) also exhibits excellent antiviral activity against A/chicken/Hubei/327/2004 (H5N1-DW), which is superior to the reference drug OSC (EC50 = 5.97 µM). Molecular docking study shows that the thiophene moiety plays an essential role in compound 4b, which can bind well to the active site of NA. The good activity of 4b may be also ascribed to the extending of quinoline ring into the 150-cavity. The results of this study may provide an insightful help for the development of new NA inhibitors.


Subject(s)
Antiviral Agents/pharmacology , Drug Discovery , Enzyme Inhibitors/pharmacology , Influenza A Virus, H5N1 Subtype/drug effects , Neuraminidase/antagonists & inhibitors , Thiophenes/pharmacology , Animals , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Dogs , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Influenza A Virus, H5N1 Subtype/enzymology , Madin Darby Canine Kidney Cells/drug effects , Madin Darby Canine Kidney Cells/virology , Microbial Sensitivity Tests , Molecular Docking Simulation , Molecular Structure , Neuraminidase/metabolism , Structure-Activity Relationship , Thiophenes/chemical synthesis , Thiophenes/chemistry
10.
Genome Biol ; 21(1): 99, 2020 04 28.
Article in English | MEDLINE | ID: mdl-32345342

ABSTRACT

BACKGROUND: Influenza is a severe respiratory illness that continually threatens global health. It has been widely known that gut microbiota modulates the host response to protect against influenza infection, but mechanistic details remain largely unknown. Here, we took advantage of the phenomenon of lethal dose 50 (LD50) and metagenomic sequencing analysis to identify specific anti-influenza gut microbes and analyze the underlying mechanism. RESULTS: Transferring fecal microbes from mice that survive virulent influenza H7N9 infection into antibiotic-treated mice confers resistance to infection. Some gut microbes exhibit differential features to lethal influenza infection depending on the infection outcome. Bifidobacterium pseudolongum and Bifidobacterium animalis levels are significantly elevated in surviving mice when compared to dead or mock-infected mice. Oral administration of B. animalis alone or the combination of both significantly reduces the severity of H7N9 infection in both antibiotic-treated and germ-free mice. Functional metagenomic analysis suggests that B. animalis mediates the anti-influenza effect via several specific metabolic molecules. In vivo tests confirm valine and coenzyme A produce an anti-influenza effect. CONCLUSIONS: These findings show that the severity of influenza infection is closely related to the heterogeneous responses of the gut microbiota. We demonstrate the anti-influenza effect of B. animalis, and also find that the gut population of endogenous B. animalis can expand to enhance host influenza resistance when lethal influenza infection occurs, representing a novel interaction between host and gut microbiota. Further, our data suggest the potential utility of Bifidobacterium in the prevention and as a prognostic predictor of influenza.


Subject(s)
Bifidobacterium animalis , Gastrointestinal Microbiome , Orthomyxoviridae Infections/prevention & control , Animals , Bifidobacterium/isolation & purification , Bifidobacterium animalis/isolation & purification , Bifidobacterium animalis/physiology , Coenzyme A/therapeutic use , Feces/microbiology , Influenza A Virus, H7N9 Subtype , Lethal Dose 50 , Lung/pathology , Mice , Mice, Inbred C57BL , Orthomyxoviridae Infections/microbiology , Orthomyxoviridae Infections/pathology , Valine/therapeutic use
11.
Oncol Rep ; 21(2): 531-7, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19148532

ABSTRACT

The novel proteasome subunit Adrm1 located on the 20q13 amplicon was differentially expressed in colorectal cancer by semiquantitative RT-PCR. Adrm1 mRNA was overexpressed in 46.2% (18/39) colorectal cancer tissues compared to their matched normal mucosa and significantly correlated with lymph node metastasis of colorectal cancer (P=0.037). Knockdown of Adrm1 by shRNA in human colon carcinoma RKO cells inhibited their anchorage-independent growth, cell migration as well as cell proliferation through inducing apoptosis and cell cycle arrest at the G1 phase. In addition, stable RNA interference of Adrm1 gene synergistic with 5-Fu treatment suppressed RKO cell growth in vitro. Collectively, these data suggested that Adrm1 is potentially oncogenic and may play an important role in colon tumorigenesis. Regiment with combined application of Adrm1 RNA interference and chemotherapy may emerge as a novel therapeutic strategy for Adrm1 overexpressed colorectal cancer.


Subject(s)
Cell Movement/genetics , Chromosomes, Human, Pair 20/genetics , Colorectal Neoplasms/pathology , Membrane Glycoproteins/metabolism , RNA Interference , Blotting, Western , Cell Line, Tumor , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Gene Expression , Humans , In Situ Nick-End Labeling , Intracellular Signaling Peptides and Proteins , Membrane Glycoproteins/genetics , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , RNA, Messenger/analysis , RNA, Small Interfering , Reverse Transcriptase Polymerase Chain Reaction , Transfection
12.
Oncol Rep ; 22(5): 1247-52, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19787246

ABSTRACT

Colorectal cancer is one of the most common causes of cancer-related deaths throughout the world. Recently, we reported that proteasome subunit PSMA7 located on 20q13 amplicon was overexpressed and associated with liver metastasis of colorectal cancer. The results indicate that PSMA7 may play an important role in the colorectal cancer progression and provide a unique target site for the development of therapeutic drugs. However, it is unknown how aberrant PSMA7 activation critically regulates the metastatic behavior of colorectal cancer cells. To investigate the role of PSMA7 in the progression of colorectal cancer, we employed the RNA interference technology to knock down the PSMA7 gene in human colon cancer cell line RKO and analyzed its effect and explored the involved mechanisms. Depletion of PSMA7 by shRNA in RKO cells inhibited their anchorage-independent growth and cell invasion and migration. Moreover, PSMA7 depletion was able to strongly suppress the in vivo tumorigenic ability of RKO cells. These effects may be induced by inhibiting CD44 expression directly or indirectly. Genetic or pharmacological inhibition of PSMA7 may therefore be a beneficial strategy in the treatment of colorectal cancer patients.


Subject(s)
Cell Movement , Colorectal Neoplasms/prevention & control , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors , RNA, Small Interfering/pharmacology , Animals , Apoptosis , Blotting, Western , Cell Adhesion , Cell Cycle , Cell Proliferation , Colony-Forming Units Assay , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Female , Genetic Therapy , Humans , Immunoenzyme Techniques , Mice , Mice, Inbred BALB C , Mice, Nude , Proteasome Endopeptidase Complex/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
13.
Zhonghua Zhong Liu Za Zhi ; 31(11): 815-9, 2009 Nov.
Article in Zh | MEDLINE | ID: mdl-20137344

ABSTRACT

OBJECTIVE: To investigate the effects of the novel proteasome subunit Adrm1 knockdown by RNA interference on proliferation of colorectal cancer cells. METHODS: The shRNA eukaryotic expression vector against Adrm1 was constructed and transfected into colon cancer RKO cells. The Adrm1-shRNA stable transfected clones were selected. Experimental cells were divided into 3 groups: the experimental group containing stable Adrm1-shRNA transfected cells, the control group containing only RKO colon cancer cells and stable empty vector transfected control group. The Adrm1 protein expression level was analyzed by Western blot. The colony-forming ability of the three groups was assessed by soft agar assay. The cell proliferation and apoptosis were analyzed by methyl thiazolyl tetrazolium (MTT) method and in situ end labeling (TUNEL) assay. Cell cycle changes were assayed by flow cytometry. RESULTS: Adrm1-shRNA effectively suppressed Adrm1 expression in the experimental group. Silencing of Adrm1 in RKO cells significantly inhibited their anchorage-independent growth, only occasional individual colonies were formed. The apoptosis rate of experimental group was (12.4 +/- 1.1)%, significantly higher than that of the stable empty vector transfected control group. The proportion of G(0)/G(1) and S/G(2) phase cells in the experimental group was (41.2 +/- 1.1)% and (58.8 +/- 1.1)%, respectively. The cells were arrested at G(1) phase. In addition, Adrm1 RNA interference combined with 5-Fu treatment significantly suppressed colorectal cancer cell growth in vitro. CONCLUSION: Silencing of Adrm1 by RNA interference can significantly suppress proliferation of RKO cells through inducing apoptosis and arresting the cell cycle. The combined application of Adrm1 RNA interference and chemotherapy may become as a novel therapeutic strategy for Adrm1 overexpressed colorectal cancer.


Subject(s)
Cell Proliferation , Colorectal Neoplasms/pathology , Membrane Glycoproteins/genetics , RNA Interference , RNA, Small Interfering/genetics , Antimetabolites, Antineoplastic/pharmacology , Apoptosis , Cell Cycle , Cell Line, Tumor , Colorectal Neoplasms/metabolism , Drug Resistance, Neoplasm , Fluorouracil/pharmacology , Genetic Vectors , Humans , Intracellular Signaling Peptides and Proteins , Membrane Glycoproteins/metabolism , Plasmids , Transfection
14.
Aging Cell ; 18(4): e12961, 2019 08.
Article in English | MEDLINE | ID: mdl-31012223

ABSTRACT

Cav1.2 is the pore-forming subunit of L-type voltage-gated calcium channel (LTCC) that plays an important role in calcium overload and cell death in Alzheimer's disease. LTCC activity can be regulated by estrogen, a sex steroid hormone that is neuroprotective. Here, we investigated the potential mechanisms in estrogen-mediated regulation of Cav1.2 protein. We found that in cultured primary neurons, 17ß-estradiol (E2) reduced Cav1.2 protein through estrogen receptor α (ERα). This effect was offset by a proteasomal inhibitor MG132, indicating that ubiquitin-proteasome system was involved. Consistently, the ubiquitin (UB) mutant at lysine 29 (K29R) or the K29-deubiquitinating enzyme TRAF-binding protein domain (TRABID) attenuated the effect of ERα on Cav1.2. We further identified that the E3 ligase Mdm2 (double minute 2 protein) and the PEST sequence in Cav1.2 protein played a role, as Mdm2 overexpression and the membrane-permeable PEST peptides prevented ERα-mediated Cav1.2 reduction, and Mdm2 overexpression led to the reduced Cav1.2 protein and the increased colocalization of Cav1.2 with ubiquitin in cortical neurons in vivo. In ovariectomized (OVX) APP/PS1 mice, administration of ERα agonist PPT reduced cerebral Cav1.2 protein, increased Cav1.2 ubiquitination, and improved cognitive performances. Taken together, ERα-induced Cav1.2 degradation involved K29-linked UB chains and the E3 ligase Mdm2, which might play a role in cognitive improvement in OVX APP/PS1 mice.


Subject(s)
Amyloid beta-Protein Precursor/genetics , Calcium Channels, L-Type/metabolism , Estrogen Receptor alpha/metabolism , Neurons/metabolism , Oligopeptides/genetics , Proteolysis/drug effects , Ubiquitination/drug effects , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Animals , Cell Line, Tumor , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/metabolism , Disease Models, Animal , Estradiol/pharmacology , Estrogen Receptor alpha/agonists , Female , Gene Knockdown Techniques , Humans , Leupeptins/pharmacology , Mice/embryology , Mice, Inbred C57BL , Mice, Transgenic , Phenols/pharmacology , Phenols/therapeutic use , Proteasome Inhibitors/pharmacology , Proto-Oncogene Proteins c-mdm2/genetics , Pyrazoles/pharmacology , Pyrazoles/therapeutic use , Transfection , Ubiquitin/metabolism
15.
Oncol Rep ; 19(2): 441-6, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18202793

ABSTRACT

The proteasome subunit PSMA7 located on the 20q13 amplicon was found to be differentially expressed in colorectal cancer by semiquantitative RT-PCR. PSMA7 mRNA was overexpressed in 37.5% (12/32) colorectal cancer tissues while it was either of a low level or not expressed in matched normal mucosa. The aim of this study was to examine the PSMA7 protein expression in 62 colorectal cancer primary sites, 34 lymph node metastatic sites and 13 liver metastatic sites by immunohistochemistry and clarify the correlations of this expression with the clinicopathological parameters. PSMA7 high expression was detected in 38.8% (24/62) colorectal cancer primary sites, 52.9% (18/34) lymph node metastatic sites and 100% (13/13) liver metastatic sites but not in the normal colorectal tissues. The PSMA7 high expression was significantly correlated with liver metastasis (P=0.028). Survival was significantly lower in patients with a PSMA7 high expression than in those with a PSMA7 low expression (P=0.0012). Moreover, in multivariate analysis, the PSMA7 expression demonstrated an independent prognostic factor (P=0.004, relative risk 5.057; 95% confidence interval, 1.682-15.201). These results indicated that PSMA7 may play an important role in colorectal cancer progression and provide a unique target site for the development of therapeutic drugs. The evaluation of PSMA7 expression in primary colorectal cancer at the time of surgery may be a valuable tool for defining patients with a high risk of developing liver metastasis.


Subject(s)
Biomarkers, Tumor/metabolism , Colorectal Neoplasms/pathology , Liver Neoplasms/diagnosis , Liver Neoplasms/secondary , Proteasome Endopeptidase Complex/metabolism , Adult , Aged , Biomarkers, Tumor/analysis , Biomarkers, Tumor/genetics , Chromosomes, Human, Pair 20/genetics , Colorectal Neoplasms/enzymology , Female , Gene Expression , Humans , Male , Middle Aged , Proteasome Endopeptidase Complex/analysis , Proteasome Endopeptidase Complex/genetics , RNA, Messenger/analysis , RNA, Messenger/metabolism , Up-Regulation
16.
Zhonghua Zhong Liu Za Zhi ; 30(7): 515-8, 2008 Jul.
Article in Zh | MEDLINE | ID: mdl-19062718

ABSTRACT

OBJECTIVE: To investigate the correlation between the proteasome subunit PSMA7 expression in colorectal cancer and its role in liver metastasis. METHODS: To identify the PSMA7 protein expression in 62 primary site colorectal cancers, 34 lymph node metastatic sites and 13 liver metastatic sites by immunohistochemistry and clarify the correlation of its expression with the clinicopathological parameters. RESULTS: High expression of PSMA7 was detected in 38.7% (24/62) of primary site colorectal cancer, 52.9% (18/34) of lymph node metastatic sites and 100% (13/13) liver metastatic sites but not in the normal colorectal tissue. High expression of PSMA7 was significantly correlated with liver metastasis (P = 0.028). The survival rate was significantly lower in patients with high expression of PSMA7 than in those with low expression of PSMA7 (P = 0.0008). As well, in multivariate analysis, PSMA7 expression demonstrated to be an independent prognostic factor (P = 0.004, relative risk 5.057; 95% confidence interval, 1.682-15.201). CONCLUSION: PSMA7 may play an important role in the colorectal cancer progression. Evaluation of PSMA7 expression in primary colorectal cancer at the time of surgery might be a valuable test in defining patients with a high risk of developing liver metastasis.


Subject(s)
Colonic Neoplasms/enzymology , Liver Neoplasms/secondary , Proteasome Endopeptidase Complex/metabolism , Rectal Neoplasms/enzymology , Adult , Aged , Colonic Neoplasms/pathology , Colonic Neoplasms/surgery , Female , Follow-Up Studies , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/enzymology , Lymphatic Metastasis , Male , Middle Aged , Multivariate Analysis , Neoplasm Staging , Rectal Neoplasms/pathology , Rectal Neoplasms/surgery , Survival Rate , Young Adult
17.
Front Mol Neurosci ; 11: 198, 2018.
Article in English | MEDLINE | ID: mdl-29942252

ABSTRACT

The α-secretase "a disintegrin and metalloproteinase domain-containing protein" (ADAM10) is involved in the processing of amyloid precursor protein (APP). Upregulation of ADAM10 precludes the generation of neurotoxic ß-amyloid protein (Aß) and represents a plausible therapeutic strategy for Alzheimer's disease (AD). In this study, we explored compounds that can potentially promote the expression of ADAM10. Therefore, we performed high-throughput small-molecule screening in SH-SY5Y (human neuroblastoma) cells that stably express a luciferase reporter gene driven by the ADAM10 promoter, including a portion of its 5'-untranslated region (5'UTR). This has led to the discovery of cosmosiin (apigenin 7-O-ß-glucoside). Here, we report that in human cell lines (SH-SY5Y and HEK293), cosmosiin proportionally increased the levels of the immature and mature forms of the ADAM10 protein without altering its mRNA level. This effect was attenuated by translation inhibitors or by deleting the 5'UTR of ADAM10, suggesting that a translational mechanism was responsible for the increased levels of ADAM10. Luciferase deletion assays revealed that the first 144 nucleotides of the 5'UTR were necessary for mediating the cosmosiin-induced enhancement of ADAM10 expression in SH-SY5Y cells. Cosmosiin failed to increase the levels of the ADAM10 protein in murine cells, which lack native expression of the ADAM10 transcript containing the identified 5'UTR element. The potential signaling pathway may involve phosphatidylinositide 3-kinase (PI3K) because pharmacological inhibition of PI3K attenuated the effect of cosmosiin on the expression of the ADAM10 protein. Finally, cosmosiin attenuated Aß generation because the levels of Aß40/42 in HEK-APP cells were significantly reduced after cosmosiin treatment. Collectively, we found that the first 144 nucleotides of the ADAM10 5'UTR, and PI3K signaling, are involved in cosmosiin-induced enhancement of the expression of ADAM10 protein. These results suggest that cosmosiin may be a potential therapeutic agent in the treatment of AD.

18.
J Mol Neurosci ; 61(3): 436-448, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28150190

ABSTRACT

Estrogen exerts multiple actions in the brain and is an important neuroprotective factor in a number of neuronal disorders. However, the underlying mechanism remains unknown. Studies demonstrate that ubiquitin-conjugating enzyme 9 (ubc9) has an integral role in synaptic plasticity and may contribute to the pathology of neuronal disorders. We aimed to investigate the effects of estrogen on ubc9 and in the Alzheimer's disease brain. Ubc9 protein and mRNA were significantly increased in the cortex and hippocampus of APP/PS1 mice with enhanced SUMOylation. Systemic estrogen administration led to reduced ubc9 expression in ovariectomized APP/PS1 mice and reduced SUMOylation. The inhibition of ubc9 expression by estrogen was found to be dose-dependent in cultured neurons. However, estrogen receptor (ER) antagonist ICI182780 did not block the inhibition of ubc9 expression by estrogen. Furthermore, the reduced expression of ubc9 was not mediated by ERα or ERß agonists alone or in combination, but by the membrane-impermeable ER agonist E2-bovine serum albumin. The activation of the G protein-coupled ER mediated the inhibition of ubc9 expression of estrogen. A phosphoinositide 3-kinase (PI3K) inhibitor, rather than an extracellular signal-regulated kinase inhibitor, blocked the inhibition of ubc9 by estrogen. Estrogen treatment significantly increased the phosphorylation of PI3K, which suggests that activation of the PI3K pathway by estrogen is required for ubc9 regulation. Further, ubc9 interacted with the synaptic proteins post-synaptic density protein 95 (PSD95) and synaptophysin. Estrogen decreased the interaction of ubc9 with post-synaptic PSD95, but increased the interaction of ubc9 with pre-synaptic synaptophysin. These results suggest that a membrane-bound ER might mediate the estrogen inhibition of ubc9 in cortical neurons, where PI3K plays an important role. We also show that ubc9 can interact with synaptic proteins, which are subject to estrogen regulation.


Subject(s)
Alzheimer Disease/metabolism , Cerebral Cortex/cytology , Estrogens/pharmacology , Neurons/metabolism , Synapses/metabolism , Ubiquitin-Conjugating Enzymes/metabolism , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Animals , Cells, Cultured , Cerebral Cortex/metabolism , Disks Large Homolog 4 Protein , Female , Guanylate Kinases/metabolism , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Neurons/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Presenilin-1/genetics , Protein Binding , Protein Transport , Receptors, Estrogen/metabolism , Synapses/drug effects , Synaptophysin/metabolism , Ubiquitin-Conjugating Enzymes/genetics
19.
Oncotarget ; 8(36): 60159-60172, 2017 Sep 01.
Article in English | MEDLINE | ID: mdl-28947961

ABSTRACT

Using high-throughput small molecule screening targeting furin gene, we identified that phorbol esters dPPA (12-Deoxyphorbol 13-phenylacetate 20-acetate) and dPA (12-Deoxyphorbol 13-acetate) significantly increased furin protein and mRNA expression in SH-SY5Y cells. This effect was prevented by PKC (protein kinase C) inhibitor calphostin C but not Ro318220, suggesting that the C1 domain, rather than the catalytic domain of PKC plays an important role. Luciferase assay revealed that nucleotides -7925 to -7426 were sufficient to mediate dPPA/dPA enhancement of furin P1 promoter activity. RNA interference of transcriptional factors CEBPß (CCAAT/enhancer-binding protein ß) and GATA1 revealed that knockdown of CEBPß significantly attenuated the effect of dPPA on furin expression. Pharmacological inhibition of ERK and PI3K but not TGFß receptor diminished the up-regulation of furin by dPPA. These results suggested that in neuronal cells, transcriptional activation of furin by dPPA/dPA may be initiated by C1 domain containing proteins including PKC; the intracellular signaling involves ERK and PI3K and transcription factor CEBPß.

20.
Brain Res ; 1657: 262-268, 2017 02 15.
Article in English | MEDLINE | ID: mdl-28017669

ABSTRACT

The critical factor considered in a depression induced by diabetes is the inflammation eliciting hippocampal, amygdala and thalamic neuronal injury. Therefore, inhibiting inflammatory reactions in the brain and reducing neuronal injury can alleviate depression in rodents suffering from diabetes mellitus. The oral administration of astaxanthin has been employed in emotional disorders and diabetic complications due to its anti-depressant, anti-inflammatory and anti-apoptotic functions. However, it has not been reported whether astaxanthin can improve diabetes-related depression-like behavior, and its potential mechanisms have not been elucidated. The objective of the present study is to elucidate the effect of astaxanthin on depression in diabetic mice and to understand the underlying molecular mechanisms. In this study, experimental diabetic mice were given a single intraperitoneal injection of streptozotocin (STZ, 150mg/kg, dissolved in citrate buffer) after fasting for 12h. The diabetic model was assessed 72h after STZ injection, and mice with a fasting blood glucose level more than or equal to 16.7mmol/L were used in this study, and oral astaxanthin (25mg/kg) was provided uninterrupted for ten weeks. Depression-like behavior was evaluated by the tail suspension test (TST) and forced swimming test (FST). The glial fibrillary acidic protein (GFAP) and cleaved caspase-3-positive cells were measured by immunohistochemistry, and the western blotting was used to test the protein levels of interleukin-6 (IL-6), interleukin-1ß (IL-1ß) and cyclooxygenase (COX-2). The results showed that astaxanthin had an anti-depressant effect on diabetic mice. Furthermore, we observed that astaxanthin significantly reduced the number of GFAP-positive cells in the hippocampus and hypothalamus, and also the expression of cleaved caspase-3 in the hippocampus, amygdala and hypothalamus was decreased as well. Moreover, astaxanthin could down-regulate the expression of IL-6, IL-1ß and COX-2 in the hippocampus. These findings suggest that the mechanism of astaxanthin in preventing depression in diabetic mice involves the inhibition of inflammation/inflammation inhibition, thereby protecting neurons in hippocampus, amygdala and hypothalamus against hyperglycemic damage.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Depression/prevention & control , Diabetes Mellitus, Experimental/drug therapy , Hippocampus/drug effects , Inflammation/drug therapy , Amygdala/drug effects , Amygdala/immunology , Amygdala/pathology , Animals , Antidepressive Agents/pharmacology , Caspase 3/metabolism , Cyclooxygenase 2/metabolism , Depression/immunology , Depression/pathology , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/psychology , Drug Evaluation, Preclinical , Glial Fibrillary Acidic Protein/metabolism , Hippocampus/immunology , Hippocampus/pathology , Hypothalamus/drug effects , Hypothalamus/immunology , Hypothalamus/pathology , Inflammation/metabolism , Inflammation/pathology , Inflammation/psychology , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Male , Mice, Inbred ICR , Random Allocation , Xanthophylls/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL