Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Cancers (Basel) ; 14(19)2022 Sep 30.
Article in English | MEDLINE | ID: mdl-36230720

ABSTRACT

Histone deacetylases 1 (HDAC1), an enzyme that functions to remove acetyl molecules from ε-NH3 groups of lysine in histones, eliminates the histone acetylation at the promoter regions of tumor suppressor genes to block their expression during tumorigenesis. However, it remains unclear why HDAC1 fails to impair oncogene expression. Here we report that HDAC1 is unable to occupy at the promoters of oncogenes but maintains its occupancy with the tumor suppressors due to its interaction with CREPT (cell cycle-related and expression-elevated protein in tumor, also named RPRD1B), an oncoprotein highly expressed in tumors. We observed that CREPT competed with HDAC1 for binding to oncogene (such as CCND1, CLDN1, VEGFA, PPARD and BMP4) promoters but not the tumor suppressor gene (such as p21 and p27) promoters by a chromatin immunoprecipitation (ChIP) qPCR experiment. Using immunoprecipitation experiments, we deciphered that CREPT specifically occupied at the oncogene promoter via TCF4, a transcription factor activated by Wnt signaling. In addition, we performed a real-time quantitative PCR (qRT-PCR) analysis on cells that stably over-expressed CREPT and/or HDAC1, and we propose that HDAC1 inhibits CREPT to activate oncogene expression under Wnt signaling activation. Our findings revealed that HDAC1 functions differentially on tumor suppressors and oncogenes due to its interaction with the oncoprotein CREPT.

2.
Nat Commun ; 12(1): 270, 2021 01 11.
Article in English | MEDLINE | ID: mdl-33431892

ABSTRACT

Intestinal stem cells (ISCs) residing in the crypts are critical for the continual self-renewal and rapid recovery of the intestinal epithelium. The regulatory mechanism of ISCs is not fully understood. Here we report that CREPT, a recently identified tumor-promoting protein, is required for the maintenance of murine ISCs. CREPT is preferably expressed in the crypts but not in the villi. Deletion of CREPT in the intestinal epithelium of mice (Vil-CREPTKO) results in lower body weight and slow migration of epithelial cells in the intestine. Vil-CREPTKO intestine fails to regenerate after X-ray irradiation and dextran sulfate sodium (DSS) treatment. Accordingly, the deletion of CREPT decreases the expression of genes related to the proliferation and differentiation of ISCs and reduces Lgr5+ cell numbers at homeostasis. We identify that CREPT deficiency downregulates Wnt signaling by impairing ß-catenin accumulation in the nucleus of the crypt cells during regeneration. Our study provides a previously undefined regulator of ISCs.


Subject(s)
Cell Cycle Proteins/metabolism , Intestines/physiology , Neoplasm Proteins/metabolism , Regeneration/physiology , Stem Cells/metabolism , Animals , Cell Count , Cell Cycle Proteins/deficiency , Cell Differentiation , Cell Proliferation , Epithelium/metabolism , Gene Deletion , Humans , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Neoplasm Proteins/deficiency , Organoids/metabolism , Stem Cells/cytology , Wnt Signaling Pathway , X-Rays , beta Catenin/metabolism
3.
Theranostics ; 10(8): 3708-3721, 2020.
Article in English | MEDLINE | ID: mdl-32206117

ABSTRACT

Cancers remain a threat to human health due to the lack of effective therapeutic strategies. Great effort has been devoted to the discovery of drug targets to treat cancers, but novel oncoproteins still need to be unveiled for efficient therapy. Methods: We show that CREPT is highly expressed in pancreatic cancer and is associated with poor disease-free survival. CREPT overexpression promotes but CREPT deletion blocks colony formation and proliferation of pancreatic cancer cells. To provide a proof of concept for CREPT as a new target for the inhibition of pancreatic cancer, we designed a cell-permeable peptide-based proteolysis targeting chimera (PROTAC), named PRTC, based on the homodimerized leucine-zipper-like motif in the C-terminus domain of CREPT to induce its degradation in vivo. Results: PRTC has high affinity for CREPT, with Kd = 0.34 +/- 0.11 µM and is able to permeate into cells because of the attached membrane-transportable peptide RRRRK. PRTC effectively induces CREPT degradation in a proteasome-dependent manner. Intriguingly, PRTC inhibits colony formation, cell proliferation, and motility in pancreatic cancer cells and ultimately impairs xenograft tumor growth, comparable to the effect of CREPT deletion. Conclusions: PRTC-induced degradation of CREPT leads to inhibition of tumor growth, which is promising for the development of new drugs against pancreatic cancer. In addition, using an interacting motif based on the dimerized structure of proteins may be a new way to design a PROTAC aiming at degrading any protein without known interacting small molecules or peptides.


Subject(s)
Cell Cycle Proteins/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Neoplasm Proteins/metabolism , Pancreatic Neoplasms/drug therapy , Peptides/therapeutic use , Proteolysis , Animals , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude
4.
Cell Biol Int ; 43(12): 1483-1491, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31286589

ABSTRACT

Fibrinogen-like protein 2 (FGL2) has been reported to play a key role in the development of human cancers. However, it is still unmasked whether FGL2 plays a potential role in colorectal carcinogenesis. In this study, the messenger RNA and protein expression levels were measured by quantitative real-time polymerase chain reaction and western blot. Cell counting kit-8 assay, transwell migration, and invasion assay were carried out to evaluate the proliferation, migration, and invasion of LOVO and SW620 cells. FGL2 was upregulated in colorectal cancer (CRC) tissues, as well as cell lines. Mitogen-activated protein kinase (MAPK) signaling was activated in CRC tissues and cell lines. FGL2 was confirmed to be downregulated by MAPK signaling inhibitor U0126. Further, we determined that knockdown of FGL2 caused a reduction of proliferation, migration, and invasion in LOVO and SW620 cells. Consistently, treatment of LOVO and SW620 cells with U0126 led to a decrease in cell proliferation, migration, and invasion. However, these changes initiated by U0126 were abolished by FGL2 overexpression. To conclude, MAPK-mediated upregulation of FGL2 promotes the proliferation, migration, and invasion of CRC cells.

6.
Cancer Sci ; 109(2): 297-307, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29215765

ABSTRACT

Dysregulation of microRNAs (miRNAs) has been linked to virulence factors of Helicobacter pylori. The role of H. pylori in esophageal disease has not been clearly defined. We previously reported that H. pylori esophageal colonization promotes the incidence of Barrett's esophagus and esophageal adenocarcinoma in vivo. Here, we studied the direct effects of H. pylori on the transformation of esophageal epithelial cells, with particular focus on whether H. pylori exerts its effects by modulating miRNAs and their downstream target genes. The normal human esophageal cell line HET-1A was chronically exposed to H. pylori extract and/or acidified deoxycholic acid for up to 36 weeks. The miRNA profiles of the esophageal epithelial cells associated with H. pylori infection were determined by microarray analysis. We found that chronic H. pylori exposure promoted acidified deoxycholic acid-induced morphological changes in HET-1A cells, along with aberrant overexpression of intestinal metaplasia markers and tumorigenic factors, including caudal-type homeobox protein 2 (CDX2), mucin 2, and cyclooxygenase 2 (COX2). Helicobacter pylori modified the miRNA profiles of esophageal epithelial cells, particularly aberrant silencing of miR-212-3p and miR-361-3p. Moreover, in biopsies from Barrett's esophagus patients, esophageal H. pylori colonization was associated with a significant decrease in miR-212-3p and miR-361-3p expression. Furthermore, we identified COX2 as a target of miR-212-3p, and CDX2 as a target of miR-361-3p. Helicobacter pylori infection of esophageal epithelial cells was associated with miRNA-mediated upregulation of oncoprotein CDX2 and COX2. Our observations provide new evidence about the molecular mechanisms underlying the association between H. pylori infection and esophageal carcinogenesis.


Subject(s)
Barrett Esophagus/pathology , CDX2 Transcription Factor/metabolism , Cyclooxygenase 2/metabolism , Esophagus/microbiology , Helicobacter Infections/genetics , Helicobacter pylori/pathogenicity , MicroRNAs/genetics , Aged , Barrett Esophagus/genetics , Barrett Esophagus/microbiology , Biopsy , CDX2 Transcription Factor/genetics , Cell Line , Cyclooxygenase 2/genetics , Epithelial Cells/cytology , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Esophagus/cytology , Esophagus/metabolism , Female , Humans , Male , Middle Aged , Up-Regulation
8.
Biochem Biophys Res Commun ; 486(3): 788-795, 2017 05 06.
Article in English | MEDLINE | ID: mdl-28347815

ABSTRACT

The sigma-2 receptor is overexpressed in proliferating cells compared to quiescent cells and has been used as a target for imaging solid tumors by positron emission tomography. Recent work has suggested that the sigma-2 receptor may also be an effective therapeutic target for cancer therapy. Poly (ADP-ribose) polymerase (PARP) is a family of enzymes involved in DNA damage response. In this study, we looked for potential synergy of cytotoxicity between PARP inhibitors and sigma-2 receptor ligands in breast cancer cell lines. We showed that the PARP inhibitor, YUN3-6, sensitized mouse breast cancer cell line, EMT6, to sigma-2 receptor ligand (SV119, WC-26, and RHM-138) induced cell death determined by cell viability assay and colony forming assay. The PARP inhibitor, olaparib, sensitized tumor cells to a different sigma-2 receptor ligand SW43-induced apoptosis and cell death in human triple negative cell line, MDA-MB-231. Olaparib inhibited PARP activity and cell proliferation, and arrested cells in G2/M phase of the cell cycle in MDA-MB-231 cells. Subsequently cells became sensitized to SW43 induced cell death. In conclusion, the combination of sigma-2 receptor ligands and PARP inhibitors appears to hold promise for synergistically triggering cell death in certain types of breast cancer cells and merits further investigation.


Subject(s)
Antineoplastic Agents/pharmacology , Azabicyclo Compounds/pharmacology , Carbamates/pharmacology , Phthalazines/pharmacology , Piperazines/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerases/genetics , Receptors, sigma/antagonists & inhibitors , Animals , Antineoplastic Combined Chemotherapy Protocols , Cell Death/drug effects , Cell Line, Tumor , Drug Synergism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , G2 Phase Cell Cycle Checkpoints/drug effects , Gene Expression , Humans , Inhibitory Concentration 50 , Mice , Poly(ADP-ribose) Polymerases/metabolism , Receptors, sigma/genetics , Receptors, sigma/metabolism
11.
World J Gastroenterol ; 20(42): 15715-26, 2014 Nov 14.
Article in English | MEDLINE | ID: mdl-25400455

ABSTRACT

AIM: To investigate esophageal Helicobacter pylori (H. pylori) colonization on esophageal injury caused by reflux and the related mechanisms. METHODS: An esophagitis model, with acid and bile reflux, was surgically produced in male rats. The rats were randomly divided into either: (1) an esophagogastroduodenal anastomosis (EGDA) group; (2) an EGDA with H. pylori infection group; (3) a pseudo-operation with H. pylori infection group; or (4) a pseudo-operation group. All rats were kept for 36 wk. Based on the location of H. pylori colonization, the EGDA rats with H. pylori infection were subdivided into those with concomitant esophageal H. pylori colonization or those with only gastric H. pylori colonization. The esophageal injuries were evaluated grossly and microscopically. The expressions of CDX2 and MUC2 were determined by real-time polymerase chain reaction (RT-PCR) and immunohistochemistry. Ki-67 antigen expression was determined by immunohistochemistry. The mRNA levels of cyclin D1, c-Myc, Bax and Bcl-2 were determined by RT-PCR. Cell apoptosis was evaluated using the TdT-mediated dUTP nick-end labeling method. RESULTS: Esophagitis, Barrett's esophagus (BE), and esophageal adenocarcinoma (EAC) developed in rats that underwent EGDA. When comparing rats with EGDA and concomitant esophageal H. pylori colonization to EGDA-only rats, the severity of injury (87.9 ± 5.2 vs 77.2 ± 8.6, macroscopically, 92.5 ± 8.0 vs 83.8 ± 5.5, microscopically, both P < 0.05) and the incidences of BE (80.0% vs 33.3%, P = 0.055) and EAC (60.0% vs 11.1%, P < 0.05) were increased. These increases were associated with upregulation of CDX2 and MUC2 mRNA (10.1 ± 5.4 vs 3.0 ± 2.9, 8.4 ± 4.6 vs 2.0 ± 3.2, respectively, Ps < 0.01) and protein (8.1 ± 2.3 vs 3.3 ± 3.1, 7.3 ± 4.0 vs 1.8 ± 2.7, respectively, all P < 0.05). The expression of Ki-67 (8.9 ± 0.7 vs 6.0 ± 1.7, P < 0.01) and the presence of apoptotic cells (8.3 ± 1.1 vs 5.3 ± 1.7, P < 0.01) were also increased significantly in rats with EGDA and concomitant esophageal H. pylori colonization compared with rats with EGDA only. The mRNA levels of cyclin D1 (5.8 ± 1.9 vs 3.4 ± 1.3, P < 0.01), c-Myc (6.4 ± 1.7 vs 3.7 ± 1.2, P < 0.01), and Bax (8.6 ± 1.6 vs 5.1 ± 1.3, P < 0.01) were significantly increased, whereas the mRNA level of Bcl-2 (0.6 ± 0.3 vs 0.8 ± 0.3, P < 0.01) was significantly reduced in rats with EGDA and concomitant esophageal H. pylori colonization compared with rats with EGDA only. CONCLUSION: Esophageal H. pylori colonization increases esophagitis severity, and facilitates the development of BE and EAC with the augmentation of cell proliferation and apoptosis in esophageal mucosa.


Subject(s)
Adenocarcinoma/microbiology , Barrett Esophagus/microbiology , Esophageal Neoplasms/microbiology , Esophagitis, Peptic/microbiology , Esophagus/microbiology , Gastroesophageal Reflux/complications , Helicobacter Infections/microbiology , Helicobacter pylori/pathogenicity , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Apoptosis , Barrett Esophagus/genetics , Barrett Esophagus/metabolism , Barrett Esophagus/pathology , CDX2 Transcription Factor , Cell Proliferation , Cyclin D1/genetics , Cyclin D1/metabolism , Disease Models, Animal , Esophageal Neoplasms/genetics , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Esophagitis, Peptic/genetics , Esophagitis, Peptic/metabolism , Esophagitis, Peptic/pathology , Esophagus/metabolism , Esophagus/pathology , Gene Expression Regulation , Helicobacter Infections/complications , Helicobacter Infections/genetics , Helicobacter Infections/metabolism , Helicobacter Infections/pathology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Ki-67 Antigen/metabolism , Male , Mucin-2/genetics , Mucin-2/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , RNA, Messenger/metabolism , Rats, Sprague-Dawley , Severity of Illness Index , Transcription Factors/genetics , Transcription Factors/metabolism , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
12.
Elife ; 3: e03011, 2014 Aug 05.
Article in English | MEDLINE | ID: mdl-25097249

ABSTRACT

In presynaptic boutons, calcium (Ca(2+)) triggers both neurotransmitter release and short-term synaptic plasticity. Whereas synaptotagmins are known to mediate vesicle fusion through binding of high local Ca(2+) to their C2 domains, the proteins that sense smaller global Ca(2+) increases to produce short-term plasticity have remained elusive. Here, we identify a Ca(2+) sensor for post-tetanic potentiation (PTP), a form of plasticity thought to underlie short-term memory. We find that at the functionally mature calyx of Held synapse the Ca(2+)-dependent protein kinase C isoforms α and ß are necessary for PTP, and the expression of PKCß in PKCαß double knockout mice rescues PTP. Disruption of Ca(2+) binding to the PKCß C2 domain specifically prevents PTP without impairing other PKCß-dependent forms of synaptic enhancement. We conclude that different C2-domain-containing presynaptic proteins are engaged by different Ca(2+) signals, and that Ca(2+) increases evoked by tetanic stimulation are sensed by PKCß to produce PTP.DOI: http://dx.doi.org/10.7554/eLife.03011.001.


Subject(s)
Calcium Signaling/physiology , Neuronal Plasticity/physiology , Protein Kinase C beta/metabolism , Protein Kinase C-alpha/metabolism , Animals , Brain Stem/metabolism , Female , Male , Memory, Short-Term/physiology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Models, Neurological , Presynaptic Terminals/metabolism , Protein Kinase C beta/deficiency , Protein Kinase C beta/genetics , Protein Kinase C-alpha/deficiency , Protein Kinase C-alpha/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
13.
Neuron ; 82(4): 859-71, 2014 May 21.
Article in English | MEDLINE | ID: mdl-24794094

ABSTRACT

Posttetanic potentiation (PTP) is a widely observed form of short-term plasticity lasting for tens of seconds after high-frequency stimulation. Here we show that although protein kinase C (PKC) mediates PTP at the calyx of Held synapse in the auditory brainstem before and after hearing onset, PTP is produced primarily by an increased probability of release (p) before hearing onset, and by an increased readily releasable pool of vesicles (RRP) thereafter. We find that these mechanistic differences, which have distinct functional consequences, reflect unexpected differential actions of closely related calcium-dependent PKC isoforms. Prior to hearing onset, when PKCγ and PKCß are both present, PKCγ mediates PTP by increasing p and partially suppressing PKCß actions. After hearing onset, PKCγ is absent and PKCß produces PTP by increasing RRP. In hearing animals, virally expressed PKCγ overrides PKCß to produce PTP by increasing p. Thus, two similar PKC isoforms mediate PTP in distinctly different ways.


Subject(s)
Neurons/physiology , Pons/cytology , Protein Isoforms/metabolism , Protein Kinase C/metabolism , Synapses/physiology , Age Factors , Animals , Animals, Newborn , Benzothiadiazines/pharmacology , Calcium/metabolism , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Gene Expression Regulation, Developmental/genetics , In Vitro Techniques , Kynurenic Acid/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuronal Plasticity/genetics , Neuronal Plasticity/physiology , Presynaptic Terminals/metabolism , Protein Isoforms/deficiency , Protein Kinase C/deficiency , Vesicular Glutamate Transport Protein 1/metabolism
14.
Zhonghua Yi Xue Za Zhi ; 94(8): 576-9, 2014 Mar 04.
Article in Chinese | MEDLINE | ID: mdl-24762684

ABSTRACT

OBJECTIVE: To compare the efficacies of non-bismuth quadruple therapy for 7 days versus standard triple therapy for 7 or 10 days in initial treatment of Helicobacter pylori (H.pylori) . METHODS: A randomized, open-labeled, controlled trial comparing non-bismuth quadruple therapy with standard triple therapy was performed at Peking University First Hospital from August 2010 to July 2012. A total of 246 patients with a diagnosis of H.pylori infection by (13)C-urea breath test and receiving no eradication therapy were randomly divided into non-bismuth quadruple therapy and standard triple therapy for 7 or 10 days. There were 110 males and 136 females with an age range of 18-75 years. Among them, 81 patients received non-bismuth quadruple therapy (esomeprazole 20 mg, amoxicillin 1 000 mg, clarithromycin 500 mg and tinidazole 500 mg given twice daily for 7 days); 82 standard triple therapy (esomeprazole 20 mg, amoxicillin 1 000 mg and clarithromycin 500 mg given twice daily) for 7 days and 83 standard triple therapy for 10 days. The efficacies were examined at Week 4 post-therapy by (13)C-urea breath test. The incidence of adverse drug reactions was recorded. RESULTS: Among them, 242 patients completed the follow-up. The eradication rates for non-bismuth quadruple therapy and standard triple therapy for 7 or 10 days were 91.4% (74/81), 79.3% (65/82) and 79.5% (66/83) as determined by intention-to-treat analysis (ITT). The eradication rates were 92.5% (74/80), 81.3% (65/80) and 80.5% (66/82) respectively as determined by per-protocol analysis (PP).Non-bismuth quadruple therapy was superior to standard triple therapy for 7 days (ITT analysis P = 0.029, PP analysis P = 0.035) and 10 days (ITT analysis P = 0.032, PP analysis P = 0.026). The differences for the eradication rates between standard triple therapy for 7 days and for 10 days were insignificant (ITT analysis P = 0.968, PP analysis P = 0.902): Adverse reaction rates for non-bismuth quadruple therapy (8.8%, 7/80) and standard triple therapy for 7 days (7.5%, 6/80) and 10 days (9.8%, 8/82) were not significantly different (P = 0.872). CONCLUSION: Non-bismuth quadruple therapy for 7 days is both effective and safe for the first-line eradication of H.pylori.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/therapeutic use , Bismuth/administration & dosage , Bismuth/therapeutic use , Helicobacter Infections/drug therapy , Adolescent , Adult , Aged , Drug Therapy, Combination , Female , Helicobacter pylori , Humans , Male , Middle Aged , Treatment Outcome , Young Adult
15.
J Neurosci ; 33(14): 5895-902, 2013 Apr 03.
Article in English | MEDLINE | ID: mdl-23554471

ABSTRACT

Golgi cells (GoCs) are inhibitory interneurons that influence the cerebellar cortical response to sensory input by regulating the excitability of the granule cell layer. While GoC inhibition is essential for normal motor coordination, little is known about the circuit dynamics that govern the activity of these cells. In particular, although GoC spontaneous spiking influences the extent of inhibition and gain throughout the granule cell layer, it is not known whether this spontaneous activity can be modulated in a long-term manner. Here we describe a form of long-term plasticity that regulates the spontaneous firing rate of GoCs in the rat cerebellar cortex. We find that membrane hyperpolarization, either by mGluR2 activation of potassium channels, or by somatic current injection, induces a long-lasting increase in GoC spontaneous firing. This spike rate plasticity appears to result from a strong reduction in the spike after hyperpolarization. Pharmacological manipulations suggest the involvement of calcium-calmodulin-dependent kinase II and calcium-activated potassium channels in mediating these firing rate increases. As a consequence of this plasticity, GoC spontaneous spiking is selectively enhanced, but the gain of evoked spiking is unaffected. Hence, this plasticity is well suited for selectively regulating the tonic output of GoCs rather than their sensory-evoked responses.


Subject(s)
Action Potentials/physiology , Cerebellum/cytology , Interneurons/physiology , Action Potentials/drug effects , Animals , Animals, Newborn , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Electric Stimulation , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Female , GABA Antagonists/pharmacology , In Vitro Techniques , Inhibitory Postsynaptic Potentials/drug effects , Interneurons/drug effects , Male , Patch-Clamp Techniques , Phosphinic Acids/pharmacology , Potassium Channels, Calcium-Activated/metabolism , Propanolamines/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Metabotropic Glutamate/metabolism , Time Factors
16.
PLoS One ; 8(2): e56709, 2013.
Article in English | MEDLINE | ID: mdl-23437218

ABSTRACT

OBJECTIVES: Toll-like receptors (TLRs) are important initiators in native immune responses to microbial infections. TLR4 is up-regulated in response to H.pylori infection in gastric epithelial cells. However, the regulatory mechanisms for the expression of TLR4 in H.pylori infection have not been clearly defined. The aims of this study are to present the evidence that microRNA let-7b directly regulates TLR4 expression in human gastric epithelial cells, and subsequently influences the activation of NF-κB and the expression of the downstream genes in H.pylori infection. METHODS: The expression of let-7b was determined in gastric mucosa specimens and in two gastric epithelial cell lines using quantitative RT-PCR. The expression of TLR4 was determined by immunohistochemistry staining and RT-PCR. The potential target of let-7b was identified by luciferase reporter assay and Western blot. Let-7b mimics and inhibitors were used to examine the effects of let-7b on NF-κB activity. The expression of the downstream genes of NF-κB was also determined in cells infected with H.pylori 26695. RESULTS: Let-7b was significantly decreased in gastric mucosa specimens and in gastric epithelial cell lines (AGS, GES-1) infected with H.pylori 26695 (cagA+). Let-7b was complementary to the 3'-UTR of TLR4 mRNA and regulated TLR4 expression via post-transcriptional suppression in gastric epithelium. Infection of H.pylori induced the expression of TLR4 and activated NF-κB in AGS and GES-1 cells. Overexpression of let-7b by mimics downregulated TLR4, and subsequently attenuated NF-κB, MyD88, NF-κB1/p50, RelA/p65. The expression of IL-8, COX-2 and CyclinD1 was inhibited in H.pylori infected cells with let-7b overexpression. Both TAK-242 (TLR4 inhibitor) and SN50 (NF-κB inhibitor) significantly inhibited the H.pylori induced downregulation of let-7b. CONCLUSIONS: Let-7b targets at TLR4 mRNA, and regulates the activation of NF-κB and the expression of the downstream genes related to the inflammation and immune responses in H.pylori infection.


Subject(s)
Helicobacter Infections/genetics , Helicobacter pylori/genetics , MicroRNAs/genetics , NF-kappa B/metabolism , Toll-Like Receptor 4/genetics , Down-Regulation , Epithelial Cells/cytology , Epithelial Cells/metabolism , Gastric Mucosa/metabolism , Gene Expression Regulation , HEK293 Cells , Helicobacter Infections/microbiology , Helicobacter pylori/pathogenicity , Humans , Immunity, Innate/genetics , Inflammation/genetics , Inflammation/metabolism , Inflammation/microbiology , MicroRNAs/metabolism , NF-kappa B/antagonists & inhibitors , Peptides/administration & dosage , Sulfonamides/administration & dosage , Toll-Like Receptor 4/metabolism
17.
J Neurosci ; 32(40): 13796-804, 2012 Oct 03.
Article in English | MEDLINE | ID: mdl-23035091

ABSTRACT

Depolarization of presynaptic terminals that arises from activation of presynaptic ionotropic receptors, or somatic depolarization, can enhance neurotransmitter release; however, the molecular mechanisms mediating this plasticity are not known. Here we investigate the mechanism of this enhancement at the calyx of Held synapse, in which presynaptic glycine receptors depolarize presynaptic terminals, elevate resting calcium levels, and potentiate release. Using knock-out mice of the calcium-sensitive PKC isoforms (PKC(Ca)), we find that enhancement of evoked but not spontaneous synaptic transmission by glycine is mediated primarily by PKC(Ca). Measurements of calcium at the calyx of Held indicate that deficits in synaptic modulation in PKC(Ca) knock-out mice occur downstream of presynaptic calcium increases. Glycine enhances synaptic transmission primarily by increasing the effective size of the pool of readily releasable vesicles. Our results reveal that PKC(Ca) can enhance evoked neurotransmitter release in response to calcium increases caused by small presynaptic depolarizations.


Subject(s)
Calcium Signaling/physiology , Cochlear Nucleus/enzymology , Glycine/pharmacology , Long-Term Potentiation/drug effects , Nerve Tissue Proteins/physiology , Protein Kinase C-alpha/physiology , Protein Kinase C/physiology , Synapses/enzymology , Animals , Calcium Signaling/drug effects , Cochlear Nucleus/physiology , Cochlear Nucleus/ultrastructure , Electric Stimulation , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Female , Male , Mice , Mice, Knockout , Presynaptic Terminals/drug effects , Presynaptic Terminals/physiology , Protein Kinase C/deficiency , Protein Kinase C/genetics , Protein Kinase C beta , Protein Kinase C-alpha/deficiency , Protein Kinase C-alpha/genetics , Strychnine/pharmacology , Synapses/drug effects , Synapses/physiology
18.
Nature ; 488(7413): 647-51, 2012 Aug 30.
Article in English | MEDLINE | ID: mdl-22763451

ABSTRACT

Autism spectrum disorders (ASDs) are highly prevalent neurodevelopmental disorders, but the underlying pathogenesis remains poorly understood. Recent studies have implicated the cerebellum in these disorders, with post-mortem studies in ASD patients showing cerebellar Purkinje cell (PC) loss, and isolated cerebellar injury has been associated with a higher incidence of ASDs. However, the extent of cerebellar contribution to the pathogenesis of ASDs remains unclear. Tuberous sclerosis complex (TSC) is a genetic disorder with high rates of comorbid ASDs that result from mutation of either TSC1 or TSC2, whose protein products dimerize and negatively regulate mammalian target of rapamycin (mTOR) signalling. TSC is an intriguing model to investigate the cerebellar contribution to the underlying pathogenesis of ASDs, as recent studies in TSC patients demonstrate cerebellar pathology and correlate cerebellar pathology with increased ASD symptomatology. Functional imaging also shows that TSC patients with ASDs display hypermetabolism in deep cerebellar structures, compared to TSC patients without ASDs. However, the roles of Tsc1 and the sequelae of Tsc1 dysfunction in the cerebellum have not been investigated so far. Here we show that both heterozygous and homozygous loss of Tsc1 in mouse cerebellar PCs results in autistic-like behaviours, including abnormal social interaction, repetitive behaviour and vocalizations, in addition to decreased PC excitability. Treatment of mutant mice with the mTOR inhibitor, rapamycin, prevented the pathological and behavioural deficits. These findings demonstrate new roles for Tsc1 in PC function and define a molecular basis for a cerebellar contribution to cognitive disorders such as autism.


Subject(s)
Autistic Disorder/physiopathology , Cerebellum/physiopathology , Purkinje Cells/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Animals , Autistic Disorder/complications , Autistic Disorder/genetics , Autistic Disorder/pathology , Behavior, Animal/drug effects , Cell Count , Cell Shape/drug effects , Cerebellum/drug effects , Cerebellum/pathology , Grooming/drug effects , Grooming/physiology , Heterozygote , Maze Learning/drug effects , Maze Learning/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mutation/genetics , Purkinje Cells/drug effects , Rotarod Performance Test , Sirolimus/pharmacology , Synapses/metabolism , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Tuberous Sclerosis/complications , Tuberous Sclerosis/genetics , Tuberous Sclerosis Complex 1 Protein , Tumor Suppressor Proteins/deficiency , Vocalization, Animal/drug effects , Vocalization, Animal/physiology
19.
Neuron ; 70(5): 1005-19, 2011 Jun 09.
Article in English | MEDLINE | ID: mdl-21658591

ABSTRACT

High-frequency stimulation leads to a transient increase in the amplitude of evoked synaptic transmission that is known as posttetanic potentiation (PTP). Here we examine the roles of the calcium-dependent protein kinase C isoforms PKCα and PKCß in PTP at the calyx of Held synapse. In PKCα/ß double knockouts, 80% of PTP is eliminated, whereas basal synaptic properties are unaffected. PKCα and PKCß produce PTP by increasing the size of the readily releasable pool of vesicles evoked by high-frequency stimulation and by increasing the fraction of this pool released by the first stimulus. PKCα and PKCß do not facilitate presynaptic calcium currents. The small PTP remaining in double knockouts is mediated partly by an increase in miniature excitatory postsynaptic current amplitude and partly by a mechanism involving myosin light chain kinase. These experiments establish that PKCα and PKCß are crucial for PTP and suggest that long-lasting presynaptic calcium increases produced by tetanic stimulation may activate these isoforms to produce PTP.


Subject(s)
Calcium/metabolism , Excitatory Postsynaptic Potentials/physiology , Protein Kinase C-alpha/metabolism , Protein Kinase C/metabolism , Synapses/physiology , Tectum Mesencephali/cytology , Animals , Azepines/pharmacology , Biophysics , Electric Stimulation , Excitatory Postsynaptic Potentials/genetics , In Vitro Techniques , Mice , Mice, Inbred C57BL , Mice, Knockout , Patch-Clamp Techniques , Phorbol Esters/pharmacology , Presynaptic Terminals/physiology , Protein Kinase C/deficiency , Protein Kinase C beta , Protein Kinase C-alpha/deficiency , Synaptic Transmission/drug effects , Synaptic Transmission/genetics , Teprotide/pharmacology , Vesicular Glutamate Transport Protein 1/metabolism
20.
Proc Natl Acad Sci U S A ; 107(17): 7975-80, 2010 Apr 27.
Article in English | MEDLINE | ID: mdl-20375278

ABSTRACT

Excessive CNS synapses are eliminated during development to establish mature patterns of neuronal connectivity. A complement cascade protein, C1q, is involved in this process. Mice deficient in C1q fail to refine retinogeniculate connections resulting in excessive retinal innervation of lateral geniculate neurons. We hypothesized that C1q knockout (KO) mice would exhibit defects in neocortical synapse elimination resulting in enhanced excitatory synaptic connectivity and epileptiform activity. We recorded spontaneous and evoked field potential activity in neocortical slices and obtained video-EEG recordings from implanted C1q KO and wild-type (WT) mice. We also used laser scanning photostimulation of caged glutamate and whole cell recordings to map excitatory and inhibitory synaptic connectivity. Spontaneous and evoked epileptiform field potentials occurred at multiple sites in neocortical slices from C1q KO, but not WT mice. Laser mapping experiments in C1q KO slices showed that the proportion of glutamate uncaging sites from which excitatory postsynaptic currents (EPSCs) could be evoked ("hotspot ratio") increased significantly in layer IV and layer V, although EPSC amplitudes were unaltered. Density of axonal boutons was significantly increased in layer V pyramidal neurons of C1q KO mice. Implanted KO mice had frequent behavioral seizures consisting of behavioral arrest associated with bihemispheric spikes and slow wave activity lasting from 5 to 30 s. Results indicate that epileptogenesis in C1q KO mice is related to a genetically determined failure to prune excessive excitatory synapses during development.


Subject(s)
Complement C1q/deficiency , Epilepsy/physiopathology , Neocortex/physiopathology , Synapses/physiology , Analysis of Variance , Animals , Complement C1q/genetics , Complement C1q/metabolism , Electroencephalography , Evoked Potentials , Glutamates/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Video , Patch-Clamp Techniques , Photic Stimulation , Synapses/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...