Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters











Publication year range
1.
Sci Rep ; 14(1): 7078, 2024 03 25.
Article in English | MEDLINE | ID: mdl-38528192

ABSTRACT

Mouse auditory cortex is composed of six sub-fields: primary auditory field (AI), secondary auditory field (AII), anterior auditory field (AAF), insular auditory field (IAF), ultrasonic field (UF) and dorsoposterior field (DP). Previous studies have examined thalamo-cortical connections in the mice auditory system and learned that AI, AAF, and IAF receive inputs from the ventral division of the medial geniculate body (MGB). However, the functional and thalamo-cortical connections between nonprimary auditory cortex (AII, UF, and DP) is unclear. In this study, we examined the locations of neurons projecting to these three cortical sub-fields in the MGB, and addressed the question whether these cortical sub-fields receive inputs from different subsets of MGB neurons or common. To examine the distributions of projecting neurons in the MGB, retrograde tracers were injected into the AII, UF, DP, after identifying these areas by the method of Optical Imaging. Our results indicated that neuron cells which in ventral part of dorsal MGB (MGd) and that of ventral MGB (MGv) projecting to UF and AII with less overlap. And DP only received neuron projecting from MGd. Interestingly, these three cortical areas received input from distinct part of MGd and MGv in an independent manner. Based on our foundings these three auditory cortical sub-fields in mice may independently process auditory information.


Subject(s)
Auditory Cortex , Geniculate Bodies , Mice , Animals , Geniculate Bodies/physiology , Auditory Cortex/physiology , Neurons , Neurites , Auditory Pathways/physiology , Thalamus/physiology
2.
Dig Dis ; 41(2): 325-334, 2023.
Article in English | MEDLINE | ID: mdl-36063790

ABSTRACT

BACKGROUND AND AIMS: Choledochoscopic gallbladder-preserving surgery (CGPS) has the advantage of treating benign gallbladder diseases on the premise of gallbladder preservation. However, it has no reliable preoperative diagnosis if the gallbladder is benign. Probe-based confocal laser endomicroscopy (pCLE) can obtain real-time and clear endoscopic images at the cell level in vivo. It is widely used in the diagnosis of digestive system diseases, but not in gallbladder diseases yet. We applied these two technologies in a complementary way into the diagnosis of gallbladder diseases and thereby lifted the reliability of CGPS. METHODS: We retrospectively analyzed the total 28 patients with the indication of CGPS with intraoperative pCLE scan referred to the Second Affiliated Hospital of Baotou Medical College between October 2019 and July 2020. The intraoperative pCLE results were compared with the postoperative pathology in various gallbladder diseases. RESULTS: We compared the intraoperative pCLE diagnosis with the postoperative pathological diagnosis and found a complete match without exception in both sensitivity and specificity. CONCLUSIONS: Based on our investigation, pCLE can provide the same accuracy as the traditional pathology in the diagnosis of gallbladder diseases with the additional advantages like noninvasive, real time, and instancy. This study serves to validate the correlation between CLE and histology. It holds a broad prospect in the application of pCLE as an intraoperative diagnosis in CGPS.


Subject(s)
Gallbladder Diseases , Laparoscopy , Humans , Reproducibility of Results , Retrospective Studies , Microscopy, Confocal/methods , Lasers
3.
Cell Signal ; 96: 110374, 2022 08.
Article in English | MEDLINE | ID: mdl-35654297

ABSTRACT

Esophageal cancer is commonly seen as either squamous cell carcinoma (ESCC) or adenocarcinoma (EAC), two very different cancers. CCN1 is a matricellular protein that induces apoptosis in EAC cells through upregulation of DR5, a death receptor, while its role in ESCC is unclear. DR6 is another death receptor, which has been reported to induce apoptosis, necroptosis, or pyroptosis in various cell systems with or without the engagement of its putative ligand amyloid precursor protein (APP). In this study, we found that CCN1 and DR6 were both highly expressed in ESCC but downregulated in EAC. Overexpression of CCN1 in ESCC cells inhibited cell proliferation through upregulation of APP and its association with p53 without DR6 involvement. Overexpression of APP stopped cell growth, but overexpression of DR6 did not affect cell growth or cell death whatsoever.


Subject(s)
Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Amyloid beta-Protein Precursor/metabolism , Apoptosis , Cell Line, Tumor , Cell Proliferation , Esophageal Neoplasms/metabolism , Humans , Receptors, Death Domain
5.
Arch Biochem Biophys ; 722: 109192, 2022 06 15.
Article in English | MEDLINE | ID: mdl-35367194

ABSTRACT

TNF, CCN1, and peptidoglycan recognition protein 1 (PGLYRP1) are often found together in the inflammatory tissue. While TNF and CCN1 promote tissue regeneration, PGLYRP1 protects it from bacterial infection. In fibroblasts, CCN1 was reported to support TNF in apoptosis induction while PGLYRP1 was found to compete with TNF for binding to TNFR1. When PGLYRP1 binds to TNFR1 by itself, it silences the receptor, but if HSP70 joins them, it leads to cell death. In cancer cells, however, CCN1 was found to antagonize TNF signaling by increasing the extracellular pool of TNFR1. In this study, we assessed their relationship in the esophageal cancer cells and found a more complex liaison among them. At first, TNF highly upregulated PGLYRP1 expression but downregulated CCN1. Secondly, PGLYRP1 bound TNFR1 and HSP70 both intracellularly and extracellularly, but TNF only promoted their extracellular interaction. Lastly, the knockdown of PGLYRP1 impaired TNF signaling. Taken together, this study shows that CCN1 interrupts TNF signaling by increasing the extracellular TNFR1 species while TNF fights back by upregulating PGLYRP1 to absorb them.


Subject(s)
Cytokines/metabolism , Esophageal Neoplasms , Receptors, Tumor Necrosis Factor, Type I , Carrier Proteins , Friends , HSP70 Heat-Shock Proteins/metabolism , Humans , Receptors, Tumor Necrosis Factor, Type I/metabolism
6.
Cancer Control ; 29: 10732748221074734, 2022.
Article in English | MEDLINE | ID: mdl-35291889

ABSTRACT

Background: Esophageal cancer is one of the most common and deadliest cancers in the world, particularly esophageal adenocarcinoma. There has never been a special drug to treat it.Purpose: This article summarizes the work that we have done in our laboratory about the role of CCN1 in esophageal cancer and gives a new perspective of CCN1 biology.Research Design: This is a review article. Study Sample: The work was done using validated cell lines and fixed human tissue slides.Data Collection and Analysis: This is a review article, therefore, no data collection or analysis was involved.Results: CCN1 is a matricellular protein supporting adhesion, migration, and survival in normal cells, but in the esophageal cancer cells, it induces TRAIL-mediated apoptosis. CCN1 promotes TRAIL and its death receptor expression but downregulates the decoy receptors and survivin in a p53-dependant manner. It was thought that CCN1 relies on TNF to induce apoptosis, but our study found that these two molecules antagonize each other. CCN1 promotes TNFR1 cleavage and uses the soluble product to block TNF signaling, while TNF upregulates PGLYRP1 to overcome this obstacle because PGLYRP1 is a secreted protein that competes with TNF for TNFR1 binding. As a result, when CCN1 and TNF are present together in the vicinity of esophageal tumors, they cancel each other out.Conclusions: Based on our laboratory study, CCN1 has much potential to be a candidate for the treatment of esophageal cancer.


Subject(s)
Adenocarcinoma , Esophageal Neoplasms , Adenocarcinoma/pathology , Apoptosis/physiology , Cysteine-Rich Protein 61/metabolism , Esophageal Neoplasms/pathology , Humans , Signal Transduction
7.
Cytokine ; 149: 155728, 2022 01.
Article in English | MEDLINE | ID: mdl-34634651

ABSTRACT

TNF signaling mostly supports cell growth by activating NFκB and only induces cell death when NFκB activation fails. CCN1 is a matricellular protein that has been reported capable to convert TNF from a pro-survival factor into a stimulus for cell death without interfering with NFκB signaling. In this study, we examined the relationship between CCN1 and TNF in the context of esophageal adenocarcinoma and found that CCN1 did not help TNF to induce cell death when they were together, instead, it inhibited TNF expression, as well as TNF-induced JNK activation and apoptosis. CCN1 induced apoptosis in the cancer cells by itself through upregulation of TRAIL and its death receptors. The presence of TNF significantly lowered CCN1 expression and its capability in apoptosis induction. Furthermore, we found that CCN1 boosted ADAM17-mediated cleavage of TNF receptors through ITGA11 and the soluble decoy receptors generated by this action neutralized TNF activity. Taken together, CCN1 and TNF antagonize each other in esophageal cancer cells.


Subject(s)
Adenocarcinoma/genetics , Apoptosis/genetics , Cysteine-Rich Protein 61/genetics , Esophageal Neoplasms/genetics , Tumor Necrosis Factors/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Gene Expression Regulation/genetics , Humans , NF-kappa B/genetics , Signal Transduction/genetics , Up-Regulation/genetics
8.
Cancer Control ; 28: 10732748211066311, 2021.
Article in English | MEDLINE | ID: mdl-34913371

ABSTRACT

DNA mutation is a common event in the human body, but in most situations, it is fixed right away by the DNA damage response program. In case the damage is too severe to repair, the programmed cell death system will be activated to get rid of the cell. However, if the damage affects some critical components of this system, the genetic scars are kept and multiply through mitosis, possibly leading to cancer someday. There are many forms of programmed cell death, but apoptosis and necroptosis represent the default and backup strategy, respectively, in the maintenance of optimal cell population as well as in cancer prevention. For the same reason, the ideal approach for cancer treatment is to induce apoptosis in the cancer cells because it proceeds 20 times faster than tumor cell proliferation and leaves no mess behind. Induction of necroptosis can be the second choice in case apoptosis becomes hard to achieve, however, necroptosis finishes the job at a cost-inflammation.


Subject(s)
Apoptosis/physiology , Necroptosis/physiology , Neoplasms/physiopathology , Cell Proliferation/physiology , DNA Repair/physiology , Humans , Inflammation
9.
Exp Cell Res ; 396(1): 112214, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32768499

ABSTRACT

Accumulating evidence shows the involvement of long non-coding RNAs (lncRNAs) in tumorigenesis of many types of human cancers. However, the role of LINC00858 in colon cancer has not been fully elucidated. Therefore, we investigated the involvement of LINC00858 in the progression of colon cancer and identified its downstream targets. After examining the expression of LINC00858 in colon cancer tissues and cell lines, we then identified the possible interaction between LINC00858 and WNK lysine deficient protein kinase 2 (WNK2) by fluorescence in situ hybridization, RNA immunoprecipitation, chromatin immunoprecipitation, and RNA pull-down assays. Next, the role of the LINC00858/WNK2 axis was explored by evaluating the apoptosis, autophagy, and senescence of colon cancer cells in vitro after ectopic expression and depletion experiments in HCT116 cells. Moreover, a mouse xenograft model of HCT116 cells was established to verify the function of the LINC00858/WNK2 axis in vivo. There was high expression of LINC00858 and low expression of WNK2 in colon cancer tissues and cell lines. Silencing of LINC00858 promoted apoptosis, senescence, and autophagy in colon cancer cells. Additionally, the enrichment of WNK2 was promoted when LINC00858 bound to DNA methyltransferases. Furthermore, in vivo assays demonstrated that silencing of LINC00858 resulted in inhibited tumor growth by upregulating WNK2. In summary, LINC00858 acts as a tumor-promoting lncRNA in colon cancer by downregulating WNK2. Our results may provide novel targets for the treatment for colon cancer.


Subject(s)
Carcinogenesis/genetics , Colonic Neoplasms/genetics , Colorectal Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Protein Serine-Threonine Kinases/genetics , RNA, Long Noncoding/genetics , Adult , Aged , Animals , Apoptosis/genetics , Autophagy/genetics , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cellular Senescence , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Female , HCT116 Cells , Humans , Lymphatic Metastasis , Male , Mice , Mice, Nude , Middle Aged , Neoplasm Staging , Protein Serine-Threonine Kinases/metabolism , RNA, Long Noncoding/antagonists & inhibitors , RNA, Long Noncoding/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Tumor Burden , Xenograft Model Antitumor Assays
10.
Curr Cancer Drug Targets ; 20(10): 789-801, 2020.
Article in English | MEDLINE | ID: mdl-32691711

ABSTRACT

Esophageal adenocarcinoma (EAC) is one of the fastest-growing cancers in the world. It occurs primarily due to the chronic gastroesophageal reflux disease (GERD), during which the esophageal epithelium is frequently exposed to the acidic fluid coming up from the stomach. This triggers gene mutations in the esophageal cells, which may lead to EAC development. While p53 is activated to get rid of the mutated cells, NFκB orchestrates the remaining cells to heal the wound. However, if the mutations happen to TP53 (a common occasion), the mutant product turns to support tumorigenesis. In this case, NFκB goes along with the mutant p53 to facilitate cancer progression. TRAIL is one of the cytokines produced in response to GERD episodes and it can kill cancer cells selectively, but its clinical use has not been as successful as expected, because some highly sophisticated defense mechanisms against TRAIL have developed during the malignancy. To clear the obstacles for TRAIL action, using a second agent to disarm the cancer cells is required. CCN1 appears to be such a molecule. While supporting normal esophageal cell growth, CCN1 suppresses malignant transformation by inhibiting NFκB and kills the EAC cell through TRAIL-mediated apoptosis.


Subject(s)
Adenocarcinoma/pathology , Cysteine-Rich Protein 61/metabolism , Esophageal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , NF-kappa B/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , Tumor Suppressor Protein p53/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Animals , Apoptosis , Cell Proliferation , Cysteine-Rich Protein 61/genetics , Esophageal Neoplasms/genetics , Esophageal Neoplasms/metabolism , Humans , NF-kappa B/genetics , TNF-Related Apoptosis-Inducing Ligand/genetics , Tumor Suppressor Protein p53/genetics
11.
Dig Liver Dis ; 52(10): 1195-1200, 2020 10.
Article in English | MEDLINE | ID: mdl-32505571

ABSTRACT

BACKGROUND: TRAIL is best known for killing cancer cells selectively, however, some cancers resist TRAIL treatment for various reasons. Esophageal adenocarcinoma is such an example. Previously, we reported that the tumor cells interrupted TRAIL-mediated apoptosis by overexpressing the decoy receptors and survivin. AIMS: To investigate TRAIL resistance in esophageal adenocarcinoma during GERD. METHODS: We simulated GERD episodes in vitro by exposing cancer cells to the acid/bile conditions acutely as well as chronically. TRAIL and its receptors were examined for expression, interaction, and induction of cell death. RESULTS: We found that acid/bile exposure drove the tumor cells to express TRAIL and TRAILR2 robustly, but did not lead to apoptosis, because the tumor cells overexpressed TRADD to replace FADD as the adaptor molecule to trigger NFκB activation instead of caspases, and thereby convert a death signal from TRAIL into a stimulus for survival. The tumor cells also overexpressed c-FLIP to keep caspases away from TRAILR2 in case FADD finds a way back to the death receptor. CONCLUSION: Multiple reasons contribute to TRAIL resistance in esophageal adenocarcinoma, including overexpression of the decoy receptors to block the death receptors, using TRADD to replace FADD, and using c-FLIP to replace caspase-8.


Subject(s)
Adenocarcinoma/pathology , Apoptosis , Esophageal Neoplasms/pathology , Gastroesophageal Reflux/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Bile Acids and Salts , Cell Line, Tumor , Gastroesophageal Reflux/chemically induced , Gastroesophageal Reflux/pathology , Humans
12.
Exp Cell Res ; 393(2): 112025, 2020 08 15.
Article in English | MEDLINE | ID: mdl-32325080

ABSTRACT

Colon cancer shows characteristics of metastasis, which is associated with angiogenesis. Increasing evidence highlights long non-coding RNAs (lncRNAs) as important participants in angiogenesis of cancers, including colon cancer. Hence, this study investigated the role of HNF1A-AS1 in angiogenesis of colon cancer. RT-qPCR and Western blot analysis were applied to detect HNF1A-AS1 and OTX1 expression in colon cancer tissues and cell lines. Then the interactions among HNF1A-AS1, PBX3, OTX1 and ERK/MAPK pathway were evaluated with RNA pull-down, RIP, ChIP and dual-luciferase reporter gene assays. Next, HCT116 and SW620 cells were treated with si-HNF1A-AS1 and/or oe-OTX1 plasmids to assess the effects of HNF1A-AS1 and OTX1 on angiogenesis, which was further evaluated in nude mice injected with SW620 cells transfected with sh-HNF1A-AS1 or sh-OTX1 lentivirus. HNF1A-AS1 and OTX1 were highly expressed in colon cancer. Silencing of HNF1A-AS1 inhibited angiogenesis of colon cancer in vivo and in vitro. HNF1A-AS1 increased the OTX1 expression by binding to transcription factor PBX3 to promote angiogenesis in colon cancer. Further, HNF1A-AS1 upregulated OTX1 to activate the ERK/MAPK pathway. Altogether, our findings identified HNF1A-AS1 as a tumor-promoting RNA in colon cancer, which could serve as a potential therapeutic target for colon cancer treatment.


Subject(s)
Colonic Neoplasms/genetics , Otx Transcription Factors/genetics , RNA, Long Noncoding/genetics , Animals , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic/genetics , Hepatocyte Nuclear Factor 1-alpha/genetics , Homeodomain Proteins/genetics , Humans , Mice , Neovascularization, Pathologic/genetics , Proto-Oncogene Proteins/genetics
13.
Int J Biochem Cell Biol ; 122: 105736, 2020 05.
Article in English | MEDLINE | ID: mdl-32135301

ABSTRACT

Esophageal adenocarcinoma essentially develops from esophageal inflammation caused by chronic GERD. During GERD episodes, the lower esophageal epithelium is repeatedly exposed to stomach acid, which often contains duodenal bile salts that prompt malignant transformation. TRAIL is one of the cytokines produced in response to such insults and targets the transformed cells exclusively. In this study, we simulated GERD episodes in vitro by exposing the cancer cells to acid or acid/bile combination and found that the cancer cells lived through acid attacks by expression of the decoy receptors and c-FLIPR but died of TRAIL-mediated apoptosis when bile salts were present. Further investigation revealed that acid/bile exposure downregulated the decoy receptors and thereby facilitated TRAIL signaling; meantime, it inhibited protein kinase C activity and thus expedited c-FLIPR degradation, allowing apoptosis to take place.


Subject(s)
Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Bile Acids and Salts/pharmacology , CASP8 and FADD-Like Apoptosis Regulating Protein/antagonists & inhibitors , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , TNF-Related Apoptosis-Inducing Ligand/metabolism , Tumor Necrosis Factor Decoy Receptors/antagonists & inhibitors , Apoptosis/drug effects , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Cell Line, Tumor , Gastroesophageal Reflux/chemically induced , Gastroesophageal Reflux/metabolism , Gastroesophageal Reflux/pathology , Humans , Hydrochloric Acid/pharmacology , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Transfection , Tumor Necrosis Factor Decoy Receptors/metabolism
14.
Cytokine ; 116: 61-69, 2019 04.
Article in English | MEDLINE | ID: mdl-30685604

ABSTRACT

GERD is the most common gastrointestinal diagnosis given during office visit. People who suffer from a long history of GERD eventually develop Barrett's esophagus, a premalignant intestinal metaplasia due to NFκB activation. Previous studies focused on the contribution of TNF-triggered canonical NFκB pathway to this event. In this study, we demonstrated in vitro that it was LTA, rather than TNF, initiated canonical NFκB activation at the beginning of acid/bile attacks, but later it switched to CD40-activated non-canonical pathway, which played a bigger part in esophageal metaplasia. CCN1 attenuated this cellular transformation by suppressing CD40 and its associated proteins involved in non-canonical signaling.


Subject(s)
Barrett Esophagus/pathology , CD40 Antigens/metabolism , Cysteine-Rich Protein 61/metabolism , Gastroesophageal Reflux/pathology , Lymphotoxin-alpha/metabolism , Bile/metabolism , Cell Line , Gene Expression Regulation , Humans , NF-kappa B/metabolism , Signal Transduction/physiology , Tumor Necrosis Factor-alpha/metabolism
15.
J Cell Biochem ; 120(2): 2070-2077, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30318638

ABSTRACT

Many cancer drugs have been developed to control tumor growth by inducing cancer cell apoptosis. However, several intracellular barriers could fail this attempt. One of these barrier is high expression of survivin. Survivin can interfere caspase activation and thereby abort apoptosis. In this study, we found that CCN1 suppressed the survivin expression in tumor cells of esophageal adenocarcinoma (EAC) and thus allowed apoptosis to finish. Furthermore, we demonstrated that this downregulation was dependent on p53 phosphorylation at Ser20, and CCN1 induced EAC cell apoptosis through the activation of p53.

16.
Article in English | MEDLINE | ID: mdl-29259640

ABSTRACT

OBJECTIVE: The aim of this study is to systematically evaluate the clinical efficacy and safety of the traditional Chinese medicine prescription Jade Screen combined with desloratadine in the treatment of chronic urticaria. METHODS: Two researchers independently conducted literature searches. The extracted data were analyzed using Rev Man 5.2.3 software. The established retrieval time range of the various databases was up to 15 March, 2017. RESULTS: Sixteen randomized controlled trials were included in this study. The results of the meta-analysis showed that the total effective rate of using Jade Screen and desloratadine in combination to treat chronic urticaria was higher than that with desloratadine alone (P < 0.00001), while its recurrence rate (P < 0.00001) and symptom score (P = 0.006) were both significantly lower than the latter. The rate of adverse reaction in the combination group was lower than that when orally taking desloratadine alone (P = 0.74), and the serum level of total IgE in the combination group was lower than that when orally taking desloratadine alone (P = 0.82); however, the results of the rate of adverse reaction and the serum level of total IgE were insignificant. CONCLUSION: Using Jade Screen and desloratadine together to treat chronic urticaria gains a better clinical effect than using desloratadine alone.

17.
Exp Cell Res ; 361(1): 163-169, 2017 12 01.
Article in English | MEDLINE | ID: mdl-29055676

ABSTRACT

TRAIL is one of the best anti-cancer molecules in our body. It kills a variety of cancer cells that are resistant to conventional chemotherapy, without causing much negative impact on normal cells, because its death receptors are almost exclusively found on cancer cells. However, some cancer cells are not sensitive to TRAIL treatment, even though they express its death receptors. A second molecule is needed to help TRAIL to complete its mission. Finding such molecules now becomes a top priority in cancer research. Our study shows that CCN1 is such a molecule. CCN1 was highly expressed in the esophageal epithelium of the patients suffering from gastroesophageal reflux disease, but faded away as the situation worsened towards adenocarcinoma. Treating the tumor cells with CCN1 resulted in apoptosis, while the same treatment to the normal cells only nourished cell growth. It was TRAIL that mediated this process. Apparently, CCN1 altered the expression profile of TRAIL and its receptors in tumor cells, namely, activating TRAIL and its death receptors and shutting down its decoy receptors. CCN1 and TRAIL worked as a team to put the cancer cells to death, as elimination of either one failed apoptosis.


Subject(s)
Adenocarcinoma/pathology , Apoptosis , Cysteine-Rich Protein 61/metabolism , Esophageal Neoplasms/pathology , TNF-Related Apoptosis-Inducing Ligand/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Cell Proliferation , Cysteine-Rich Protein 61/genetics , Esophageal Neoplasms/genetics , Esophageal Neoplasms/metabolism , Humans , TNF-Related Apoptosis-Inducing Ligand/genetics , Tumor Cells, Cultured
18.
World J Gastroenterol ; 18(45): 6521-6, 2012 Dec 07.
Article in English | MEDLINE | ID: mdl-23236223

ABSTRACT

Esophageal cancer is mainly found in Asia and east Africa and is one of the deadliest cancers in the world. However, it has not garnered much attention in the Western world due to its low incidence rate. An increasing amount of data indicate that esophageal cancer, particularly esophageal adenocarcinoma, has been rising by 6-fold annually and is now becoming the fastest growing cancer in the United States. This rise has been associated with the increase of the obese population, as abdominal fat puts extra pressure on the stomach and causes gastroesophageal reflux disease (GERD). Long standing GERD can induce esophagitis and metaplasia and, ultimately, leads to adenocarcinoma. Acid suppression has been the main strategy to treat GERD; however, it has not been proven to control esophageal malignancy effectively. In fact, its side effects have triggered multiple warnings from regulatory agencies. The high mortality and fast growth of esophageal cancer demand more vigorous efforts to look into its deeper mechanisms and come up with better therapeutic options.


Subject(s)
Adenocarcinoma/diagnosis , Adenocarcinoma/epidemiology , Esophageal Neoplasms/diagnosis , Esophageal Neoplasms/epidemiology , Animals , Bile Acids and Salts/chemistry , China , Esophagus/pathology , Gastroesophageal Reflux/diagnosis , Gastroesophageal Reflux/epidemiology , Humans , Inflammation , Obesity/complications , Obesity/diagnosis , Rats , United States
19.
World J Gastroenterol ; 18(30): 3931-5, 2012 Aug 14.
Article in English | MEDLINE | ID: mdl-22912541

ABSTRACT

Metastasis is the main reason for cancer-related death. S100A4 is one of the key molecules involved in this event. Several studies have shown that overexpression of S100A4 in non-metastatic cancer cells can make them become metastatic, and knockdown of S100A4 in metastatic cancer cells can curtail their invasive nature. A study by Chen et al([2]) published in the World J Gastroenterol 18(9): 915-922, 2012 is a typical example. This study showed in vitro and in vivo evidence that S100A4 expression level determines the invasiveness of esophageal squamous carcinoma. Considering the fact that more than half of the cancer-related deaths are caused by malignancies derived from the digestive system and esophageal cancer is the 4th top contributor to this fraction, this study warrants more attention.


Subject(s)
Neoplasms/metabolism , Neoplasms/pathology , S100 Proteins/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Cell Movement , Esophageal Neoplasms/genetics , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Humans , Neoplasm Metastasis , Neoplasms/genetics , S100 Calcium-Binding Protein A4 , S100 Proteins/genetics
20.
ISRN Gastroenterol ; 2012: 207235, 2012.
Article in English | MEDLINE | ID: mdl-22701179

ABSTRACT

Aims. Nuclear translocation of ß-catenin is common in many cancers including esophageal squamous cell carcinoma (ESCC). As a mediator of Wnt signaling pathway, nuclear ß-catenin can activate many growth-related genes including CCN1, which in turn can induce ß-catenin translocation. CCN1, a matricellular protein, signals through various integrin receptors in a cell-dependent manner to regulate cell adhesion, proliferation, and survival. Its elevation has been reported in ESCC as well as other esophageal abnormalities such as Barrett's esophagus. The aim of this study is to examine the relationship between CCN1 and ß-catenin in ESCC. Methods and Results. The expression and correlation between CCN1 and ß-catenin in ESCC tissue were examined through immunohistochemistry and further analyzed in both normal esophageal epithelial cells and ESCC cells through microarray, functional blocking and in situ protein ligation. We found that nuclear translocation of ß-catenin in ESCC cells required high level of CCN1 as knockdown of CCN1 in ESCC cells reduced ß-catenin expression and translocation. Furthermore, we found that integrin α(11) was highly expressed in ESCC tumor tissue and functional blocking integrin α(11) diminished CCN1-induced ß-catenin elevation and translocation. Conclusions. Integrin α(11) mediated the effect of CCN1 on ß-catenin in esophageal epithelial cells.

SELECTION OF CITATIONS
SEARCH DETAIL