Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
J Cancer ; 10(10): 2229-2236, 2019.
Article in English | MEDLINE | ID: mdl-31258726

ABSTRACT

Purpose: The study was conducted to investigate the relationship of serum pepsinogens PGI, PGII, gastrin-17, and Hp-IgG with colorectal cancer (CRC), aiming to explore the clinical significance of serum markers reflecting gastric function and H. pylori infection in CRC. Methods: A total of 569 CRC cases and 569 age and sex-matched controls were enrolled in this study between June 2012 and April 2016 from The First Hospital of China Medical University. The serum markers reflecting gastric function and H. pylori infection were detected using ELISA, including PGI, PGII, PGI/II ratio, G-17 and Hp-IgG. Information of clinicopathological parameters and tumor biomarkers was collected from the medical records of inpatients, including CEA, CA199, CA125, CA153 and AFP. Results: Serum PGII, G-17 levels and Hp-IgG were increased in CRC, while PGI and PGI/II ratio appeared no significant difference between CRC and controls. In subgroup analysis, PGII was more significant in males (P=0.014). Hp-IgG was demonstrated higher in age<60y (P=0.001). With respect to the association with serum tumor biomarkers, G-17 level was associated with the rise of CA125 (P=0.005, OR (95%CI): 4.89 (1.90-12.57)), Hp-IgG increasing was associated with the rise of CA125 (P=0.024, OR (95%CI): 4.10 (1.54-10.93)). Conclusions: Serum PGII, G-17 and Hp-IgG were associated with CRC risk. The serum levels of G-17 and Hp-IgG were associated with the rise of CA125 in patients with CRC.

2.
World J Gastrointest Oncol ; 11(12): 1141-1150, 2019 Dec 15.
Article in English | MEDLINE | ID: mdl-31908719

ABSTRACT

BACKGROUND: In recent years, the incidence of gastrointestinal (GI) cancer in China has increased annually. Early detection and appropriate therapy are considered to be the key to treat GI cancer. DNMT1 takes an active part in the advancement of GI cancer, which will change as the disease progresses. But its expression characteristics in the dynamic variations of GI carcinogenesis are still unclear. AIM: To investigate the expression characteristics of DNMT1 in different GI diseases. METHODS: We detected the expression of DNMT1 in 650 cases of different GI diseases by immunohistochemistry, including 90 cases of chronic superficial gastritis (CSG), 72 cases of atrophic gastritis with intestinal metaplasia (AG/GIM), 54 cases of low-grade intraepithelial neoplasia (GLIN), 66 cases of high-grade intraepithelial neoplasia (GHIN), 71 cases of early gastric cancer (EGC), 90 cases of normal intestinal mucosa (NIM), 54 cases of intestinal low-grade intraepithelial neoplasia (ILIN), 71 cases of intestinal high-grade intraepithelial neoplasia (IHIN), and 82 cases of early colorectal cancer (ECRC). RESULTS: In the CSG group, all cases showed weakly positive or negative expression of DNMT1. However, in other four groups (AG/GIM, GLIN, GHIN, and EGC), the positive expression rate gradually increased with the severity of the diseases; the negative or weakly positive cases accounted for 55.56% (40/72), 38.89% (21/54), 1.52% (1/66), and 1.41% (1/71), respectively. Besides, the moderately positive cases were 44.44% (32/72), 57.41% (31/54), 80.30% (53/66), and 43.66% (31/71), respectively. The strongly positive cases only existed in the GLIN (3.70%, 2/54), GHIN (18.18%, 12/66), and EGC (54.93%, 39/71) groups. The differences between any two groups were statistically significant (P < 0.05). Similarly, in the NIM group, cases with weakly positive expression of DNMT1 were predominant (91.11%, 82/90), and the rest were moderately positive cases (8.89%, 8/90). In the ILIN, IHIN, and ECRC groups, the rates of cases with weak or negative expression of DNMT1 were 46.30% (25/54), 12.68% (9/71), and 4.88% (4/82), respectively; with moderately positive expression were 53.70% (29/54), 71.83% (51/71), and 34.15% (28/82), respectively; and with strongly positive expression were 0.00% (0/54), 15.49% (11/71), and 60.98% (50/82), respectively. The differences between any two groups were also statistically significant (P < 0.05). CONCLUSION: The overexpression of DNMT1 protein could effectively predict early GI cancers and severe precancerous lesions, which may have potential clinical application value.

3.
Clin Chim Acta ; 483: 69-75, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29649455

ABSTRACT

BACKGROUND: Immune inflammation appears to play a role in aortic aneurysm (AA) pathology. Toll-like receptor 4 (TLR4) has been proved to involve in immune inflammatory diseases. However, the relationship between serum TLR4 and AA is still unclear. METHODS: The study included 282 AA patients and 287 controls. The clinical test related information was collected in medical records. The levels of serum TLR4 were measured by enzyme-linked immunosorbent assay. RESULTS: Serum TLR4 levels were significantly higher in case groups and could be influenced by age, smoking, hypertension, diabetes and hyperlipidemia. Serum TLR4 was positively correlated with circulating CRP, Hcy, D-dimer, Fg and Cys-c in AA patients, even after adjusting the possible influencing factors. The optimal cut-off value of TLR4 was 13.64 ng/ml for discriminating AA, and the screening accuracy was higher for those who were males (sensitivity of 63.5% and specificity of 68.6%), smokers (sensitivity of 63.5% and specificity of 82.7%) and hyperlipidemia (sensitivity of 59.1% and specificity of 81.2%). Multiple logistic analyses showed that serum TLR4 was significantly correlated with AA risk (OR = 1.119, 95% CI = 1.077-1.162, p < 0.001) and subjects with high TLR4 levels (>13.64 ng/ml) were more likely to have AA (OR = 4.225, 95% CI = 2.477-7.206, p < 0.001). CONCLUSIONS: Serum TLR4 was closely related to AA and associated with some AA-related circulating markers. Serum TLR4 could be a novel and promising biomarker with important diagnostic and predictive value in the identification of aortic aneurysmal diseases.


Subject(s)
Aortic Aneurysm/blood , Toll-Like Receptor 4/blood , Biomarkers/blood , Female , Humans , Male , Middle Aged , Predictive Value of Tests
4.
Zhongguo Dang Dai Er Ke Za Zhi ; 20(4): 295-297, 2018 Apr.
Article in Chinese | MEDLINE | ID: mdl-29658454

ABSTRACT

OBJECTIVE: To investigate the clinical features and prognosis of malignancy-associated hemophagocytic lymphohistiocytosis (MAHS) in children. METHODS: A retrospective analysis was performed for the primary diseases, clinical features, and prognosis of 24 children with MAHS. RESULTS: Among the 24 children, 11 (46%) had MAHS induced by tumor and 13 (54%) had chemotherapy-associated MAHS. As for primary diseases, 17 children had acute leukemia, 6 had lymphoma, and 1 had neuroblastoma. The most common clinical manifestations were pyrexia, respiratory symptoms, and hepatosplenomegaly. The most common laboratory abnormalities were hemocytopenia, elevated serum ferritin, and elevated lactate dehydrogenase. Of the 24 children, 22 were treated according to the HLH-2004 protocol and 2 gave up treatment; 18 children died, 1 was lost to follow-up, and 5 survived. The survival time ranged from 3 days to 2 years and 4 months (median 28 days). CONCLUSIONS: Children with MAHS have various clinical features and extremely poor treatment outcomes.


Subject(s)
Lymphohistiocytosis, Hemophagocytic/mortality , Neoplasms/complications , Adolescent , Child , Child, Preschool , Female , Humans , Infant , Lymphohistiocytosis, Hemophagocytic/therapy , Male , Prognosis , Retrospective Studies , Treatment Outcome
5.
Oncotarget ; 8(26): 43140-43152, 2017 Jun 27.
Article in English | MEDLINE | ID: mdl-28562347

ABSTRACT

Excision repair cross-complementing group 6 and 8 (ERCC6 and ERCC8) are two indispensable genes for the initiation of transcription-coupled nucleotide excision repair pathway. This study aimed to evaluate the interactions between single nucleotide polymorphisms of ERCC6 (rs1917799) and ERCC8 (rs158572 and rs158916) in gastric cancer and its precancerous diseases. Besides, protein level analysis were performed to compare ERCC6 and ERCC8 expression in different stages of gastric diseases, and to correlate SNPs jointly with gene expression. Sequenom MassARRAY platform method was used to detect polymorphisms of ERCC6 and ERCC8 in 1916 subjects. In situ ERCC6 and ERCC8 protein expression were detected by immunohistochemistry in 109 chronic superficial gastritis, 109 chronic atrophic gastritis and 109 gastric cancer cases. Our results demonstrated pairwise epistatic interactions between ERCC6 and ERCC8 SNPs that ERCC6 rs1917799-ERCC8 rs158572 combination was associated with decreased risk of chronic atrophic gastritis and increased risk of gastric cancer. ERCC6 rs1917799 also showed a significant interaction with ERCC8 rs158916 to reduce gastric cancer risk. The expressions of ERCC6, ERCC8 and ERCC6-ERCC8 combination have similarities that higher positivity was observed in chronic superficial gastritis compared with chronic atrophic gastritis and gastric cancer. As for the effects of ERCC6 and ERCC8 SNPs on the protein expression, single SNP had no correlation with corresponding gene expression, whereas the ERCC6 rs1917799-ERCC8 rs158572 pair had significant influence on ERCC6 and ERCC6-ERCC8 expression. In conclusion, ERCC6 rs1917799, ERCC8 rs158572 and rs158916 demonstrated pairwise epistatic interactions to associate with chronic atrophic gastritis and gastric cancer risk. The ERCC6 rs1917799-ERCC8 rs158572 pair significantly influence ERCC6 and ERCC6-ERCC8 expression.


Subject(s)
DNA Helicases/genetics , DNA Repair Enzymes/genetics , Gastritis, Atrophic/genetics , Poly-ADP-Ribose Binding Proteins/genetics , Stomach Neoplasms/genetics , Transcription Factors/genetics , Case-Control Studies , DNA Helicases/biosynthesis , DNA Repair Enzymes/biosynthesis , Female , Gastritis, Atrophic/enzymology , Gastritis, Atrophic/pathology , Genetic Predisposition to Disease , Humans , Immunohistochemistry , Male , Middle Aged , Poly-ADP-Ribose Binding Proteins/biosynthesis , Polymorphism, Single Nucleotide , Risk Factors , Stomach Neoplasms/enzymology , Stomach Neoplasms/pathology , Transcription Factors/biosynthesis
6.
PLoS One ; 9(9): e108704, 2014.
Article in English | MEDLINE | ID: mdl-25268735

ABSTRACT

BACKGROUND: Xeroderma pigmentosum group G (XPG) plays a critical role in preventing cells from oxidative DNA damage. This study aimed to investigate XPG protein expression in different gastric tissues and in patients with diverse prognoses, thus providing insights into its role in the development, progression and prognosis of gastric cancer (GC). METHODS: A total of 176 GC, 131 adjacent non-tumour tissues, 53 atrophic gastritis (AG) and 49 superficial gastritis (SG) samples were included. Immunohistochemical staining was used to detect XPG protein expression. RESULTS: XPG expression was significantly higher in GC tissues compared with adjacent non-tumour tissues. In the progressive disease sequence SG→AG→GC, XPG expression was significantly higher in AG and GC compared with SG. Analysis of clinicopathological parameters and survival in GC patients demonstrated a significant association between XPG expression level and depth of tumour invasion, macroscopic type, Lauren's classification, smoking, Helicobacter pylori infection and family history. Cox multivariate survival analysis indicated that patients with positive XPG expression had significantly longer overall survival (P = 0.020, HR = 0.394, 95%CI 0.179-0.866), especially in aged younger than 60 years (P = 0.027, HR = 0.361, 95%CI 0.147-0.888) and male patients (P = 0.002, HR = 0.209, 95%CI 0.077-0.571). CONCLUSIONS: This study demonstrated that XPG protein expression was related to the development, progression and prognosis of GC, and might thus serve as a potential biomarker for its diagnosis and prognosis.


Subject(s)
Stomach Neoplasms/diagnosis , Xeroderma Pigmentosum/metabolism , Age Factors , Aged , Disease Progression , Female , Gastric Mucosa/metabolism , Gastritis/metabolism , Gastritis/pathology , Gastritis, Atrophic/metabolism , Gastritis, Atrophic/pathology , Helicobacter Infections/complications , Humans , Immunohistochemistry , Male , Middle Aged , Neoplasm Metastasis , Neoplasm Staging , Prognosis , Sex Factors , Smoking , Stomach Neoplasms/metabolism , Stomach Neoplasms/mortality , Survival Analysis
7.
PLoS One ; 9(12): e115955, 2014.
Article in English | MEDLINE | ID: mdl-25551587

ABSTRACT

BACKGROUND: Pepsinogen C (PGC) plays an important role in sustaining the cellular differentiation during the process of gastric carcinogenesis. This study aimed to assess the role of PGC tagSNPs and their interactions with Helicobacter pylori (H. pylori) in the development of gastric cancer and its precursor, atrophic gastritis. METHODS: Four PGC tagSNPs (rs6941539, rs6912200, rs3789210 and rs6939861) were genotyped by Sequenom MassARRAY platform in a total of 2311 subjects consisting of 642 gastric cancer, 774 atrophic gastritis, and 895 healthy control subjects. The mRNA and protein expression levels of PGC in gastric tissues and in serum were respectively measured by quantitative reverse transcriptase-polymerase chain reaction (qRT-PCR), immunohistochemistry, and Eenzyme-linked immunoabsorbent assay (ELISA). RESULTS: We found associations between PGC rs3789210 CG/GG genotypes and reduced gastric cancer risk and between PGC rs6939861 A variant allele and increased risks of both gastric cancer and atrophic gastritis. As for the haplotypes of PGC rs6941539-rs6912200-rs3789210-rs6939861 loci, the TTCA and TTGG haplotypes were respectively associated with increased and reduced risks of both gastric cancer and atrophic gastritis; additionally, the CTCA haplotype was associated with increased atrophic gastritis risk. Very interestingly, rs6912200 CT/TT genotypes had a positive interaction with H. pylori, synergistically elevating the gastric cancer risk. Moreover, healthy subjects who carried rs6912200 CT, TT and CT/TT variant genotypes had lower histological and serum expression levels of PGC protein. CONCLUSIONS: Our findings highlight an important role of PGC rs3789210 and rs6939861 in altering susceptibility to atrophic gastritis and/or gastric cancer. Moreover, people who carry rs6912200 variant genotypes exhibit higher gastric cancer risk in case of getting H. pylori infection, which strongly suggest a necessity of preventing and/or eliminating H. pylori infection in those individuals.


Subject(s)
Gastritis, Atrophic/genetics , Helicobacter Infections/pathology , Helicobacter pylori/pathogenicity , Pepsinogen C/genetics , Stomach Neoplasms/genetics , Cell Transformation, Neoplastic/genetics , Gastric Mucosa/metabolism , Genetic Association Studies , Genetic Predisposition to Disease , Genotype , Helicobacter Infections/microbiology , Helicobacter pylori/isolation & purification , Humans , Pepsinogen C/blood , Polymorphism, Single Nucleotide , RNA, Messenger/biosynthesis , Stomach/microbiology , Stomach/pathology , Stomach Neoplasms/blood
8.
J Gastroenterol Hepatol ; 26(6): 1039-46, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21303408

ABSTRACT

BACKGROUND AND AIM: Serum pepsinogen II (sPGII) is underutilized and considered an inconspicuous biomarker in clinical practice. We refocused on this neglected but novel biomarker and conducted the present study, aiming to elucidate the normal level of sPGII in healthy Chinese patients and to investigate the clinical utility of sPGII for gastric disease screening. METHODS: In 2008-2009, a total of 2022 participants from northern China were selected and enrolled in the study. sPGII and Helicobacter pylori (H. pylori)-immunoglobulin G were measured with ELISA. RESULTS: sPGII showed a normal value of 6.6 microg/L in a total of 466 patients with endoscopically- and histologically-normal stomachs. A small sex difference was observed: the average value of sPGII was 7 microg/L and 6 microg/L in males and females, respectively (P < 0.001). In the differentiation between healthy and diseased (endoscopically-diseased stomach or gastritis/atrophic gastritis in endoscopic biopsies) stomach mucosae, the best sPGII cut-off value was 8.25 microg/L (sensitivity 70.6%, specificity 70.8%). In screening the H. pylori seropositivity, the optimum cut-off sPGII value was 10.25 microg/L (sensitivity 71.6%, specificity 70.1%). CONCLUSIONS: We demonstrated that the mean values of sPGII in a healthy Chinese population are 7 microg/L and 6 microg/L for males and females, respectively. sPGII significantly increases in diseased and H. pylori-infected stomach, and the best sPGII cut-off value is 8.25 microg/L in the differentiation between patients with healthy and diseased stomach mucosae. Furthermore, Chinese patients with sPGII greater than 10.25 microg/L are at greater risk of various H. pylori-related gastropathies, and are therefore prior candidates for gastro-protection therapy.


Subject(s)
Clinical Enzyme Tests , Gastric Mucosa/enzymology , Gastritis/diagnosis , Pepsinogen C/blood , Stomach Neoplasms/diagnosis , Adult , Aged , Aged, 80 and over , Antibodies, Bacterial/blood , Biomarkers/blood , Case-Control Studies , China , Enzyme-Linked Immunosorbent Assay , Female , Gastric Mucosa/microbiology , Gastric Mucosa/pathology , Gastritis/blood , Gastritis/microbiology , Gastritis/pathology , Gastroscopy , Helicobacter pylori/immunology , Humans , Immunoglobulin G/blood , Male , Metaplasia , Middle Aged , Predictive Value of Tests , Prognosis , ROC Curve , Stomach Neoplasms/blood , Stomach Neoplasms/microbiology , Stomach Neoplasms/pathology
9.
Int J Oncol ; 35(6): 1313-20, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19885554

ABSTRACT

Identifying the genetic variants that alter MUC1 protein expression may further our understanding of the risk for development of gastric cancer (GC). We used PCR-SSPs to identify the genotype of MUC1 A/G polymorphism at its 568 site of exon 2 and immunohistochemistry to detect MUC1 protein expression in GC patients and non-cancer subjects and analyzed the association between this polymorphism and MUC1 protein expression. We found that the frequency of AA genotype was significantly high in the GC patients and the risk for GC in AA genotype carriers increased 1.81-fold. Moreover, we found a significant underexpression of MUC1 protein in GC as compared to non-cancer subjects, which was negatively correlated to AA genotype of MUC1 (r=-0.1790, P=0.004). Furthermore, this study provides a possible mechanistic insight that the MUC1 A/G polymorphism at its 568 site disrupts the physiological functions of MUC1 which is important to the physiological protection of gastric mucosa. Thus we have provided evidence that may identify the MUC1 A/G polymorphism at 568 site, as a potential genetic factor which leads to an increase in susceptibility for GC through alteration of MUC1 gene and MUC1 expression in the population that carry the A allele.


Subject(s)
Carcinoma/genetics , Genetic Predisposition to Disease , Mucin-1/genetics , Stomach Neoplasms/genetics , Adult , Aged , Aged, 80 and over , Base Sequence , Female , Genotype , Humans , Immunohistochemistry , Male , Middle Aged , Molecular Sequence Data , Polymerase Chain Reaction , Polymorphism, Single Nucleotide , Risk Factors , Tissue Array Analysis
10.
Ai Zheng ; 28(5): 487-92, 2009 May.
Article in Chinese | MEDLINE | ID: mdl-19624876

ABSTRACT

BACKGROUND AND OBJECTIVE: Human pepsinogen C (PGC) is an aspartic protease synthesized in gastric mucosa. PGC gene insertion/deletion polymorphism, which is located between exon 7 and 8, has been found to associate with gastric cancer (GC) susceptibility. This study was to investigate the relationship between PGC polymorphism with protein expression of PGC in gastric mucosa and serum. METHODS: PGC insertion/deletion polymorphism was evaluated by PCR, followed by direct DNA sequencing in 493 cases of GC, atrophic gastritis (AG), gastric erosion ulcer (GEU) and superficial gastritis (SG). PGC protein expression in gastric mucosa was measured by immunohistochemistry. The serum PGC level was determined by enzyme-linked immunosorbent assay (ELISA). RESULTS: In accordance with the following order SG-->GEU-->GA-->GC, the frequency of PGC homozygous allele 1 was gradually increased, which was higher in GC than in SG (P=0.018); while the protein expression of PGC in gastric mucosa was gradually decreased (P<0.01), along with a gradual decrease in the strong positive rate of PGC (P<0.05) except for SG vs. GEU. The serum level of PGC was significantly lower in SG than in GU(P=0.000) and GC(P=0.000). The frequency of PGC homozygous allele 1 was negatively correlated to PGC protein expression in gastric mucosa (r=-0.1085, P=0.023). From homozygous allele 1 to heterozygous allele 1, and to other genotypes, the PGC positive rate was gradually increased in gastric mucosa, with significant differences between homozygous allele 1 and other genotypes (P=0.009); while the strong-positive rate of PGC was gradually decreased only in SG group (P=0.047). CONCLUSION: PGC gene insertion/deletion polymorphism is negatively related to PGC protein expression in gastric mucosa, but is not related to the serum PGC level.


Subject(s)
Gastric Mucosa/metabolism , INDEL Mutation , Pepsinogen C/genetics , Polymorphism, Genetic , Stomach Neoplasms , Adult , Aged , Aged, 80 and over , Alleles , Female , Gastritis/blood , Gastritis/genetics , Gastritis/metabolism , Gastritis, Atrophic/blood , Gastritis, Atrophic/genetics , Gastritis, Atrophic/metabolism , Genetic Predisposition to Disease , Genotype , Humans , Male , Middle Aged , Pepsinogen C/blood , Pepsinogen C/metabolism , Sequence Analysis, DNA , Stomach Neoplasms/blood , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Stomach Ulcer/blood , Stomach Ulcer/genetics , Stomach Ulcer/metabolism
11.
Yi Chuan ; 30(9): 1163-8, 2008 Sep.
Article in Chinese | MEDLINE | ID: mdl-18779174

ABSTRACT

Analyzed the relationship between 568 site A/G polymorphism in mucin 1(MUC1) gene in a population from Liaoning Province and susceptibility to gastric cancer. Sequence specific primers-polymerase chain reaction(PCR-SSPs) were performed to analyze the genotype of the A/G polymorphism in its 568 site of exon 2 for 138 gastric cancer cases and 131 normal ones tested, and ELISA was performed to test IgG antibody of H. pylori. We found that the distribution frequency of AA, AG, GG three genotypes were 73.3%, 22.1%, 4.6%, respectively. The frequency of AA genotype was statistically higher in the gastric cancer (GC) group compared to the control group(P=0.03), moreover, individuals with the MUC1 AA genotype increased 1.92 fold risk for gastric cancer. And compared with subjects carrying both AG+GG genotype and H. pylori-IgG negative, there was an obviously increased risk of developing gastric cancer for those who carried both AA genotype and H. pylori-IgG negative, both AG+GG genotype and H. pylori-IgG positive, and both AG+GG genotype and H. pylori-IgG negative, but there were no significantly differences between the every two of the latter three. Our results indicated that there was a relationship between MUC1 A/G polymorphism and susceptibility to gastric cancer. And there were probably no statistically interaction between the 568 site A/G polymorphism in MUC1 and H. pylori infection in the occurrence and development process of gastric cancer.


Subject(s)
Helicobacter Infections/genetics , Mucin-1/genetics , Polymorphism, Genetic , Stomach Neoplasms/genetics , Adult , Aged , Aged, 80 and over , Databases, Genetic , Disease Susceptibility , Female , Genetic Predisposition to Disease , Genotype , Humans , Male , Middle Aged , Sequence Analysis, DNA
12.
Zhonghua Yi Xue Za Zhi ; 88(4): 230-2, 2008 Jan 22.
Article in Chinese | MEDLINE | ID: mdl-18361831

ABSTRACT

OBJECTIVE: To investigate the condition of Helicobacter pylori (Hp) colonization in different gastric diseases and the application of immunostaining technique in Hp density measurement. METHODS: Biopsy specimens of gastric mucosa were obtained from 174 patients with different gastric diseases confirmed by pathological examination, 99 males and 75 females, aged 53.37 (19-87), 135 from Zhuanghe, a high risk area of gastric cancer, and 39 from Shenyang, a low risk area. Hematoxylin and eosin (HE) staining and immunohistochemistry were used to detect the Hp density in different parts of gastric mucosa. RESULTS: When the grade of Hp density in the gastric mucosa epithelium cell surface and lumen of gland < 1 there was a significant difference between HE stain and immunohistochemistry (chi(2) = 9.446, P < 0.01), however, when the grade of Hp density in the gastric mucosa epithelium cell surface and lumen of gland > 1 there was no significant difference between them (P > 0.05). The grade of Hp density of gastric mucosa epithelium cell surface and lumen of gland in superficial gastritis was > 1, significantly higher than those of atrophic gastritis group (23.8%) and gastric cancer group (16.7%) (chi(2) = 2.780, P > 0.05; chi(2) = 4.876, P > 0.05). The Hp colonization density in the gastric mucosa epithelium cell surface and lumen of the patients from Zhuanghe region was significantly lower than that of the patients from Shenyang region (chi(2) = 5.915, P = 0.015); and the Hp density in the gastric mucus in the patients from Zhuanghe was significantly higher than that of the patients from Shenyang region (chi(2) = 8.879, P = 0.003, chi(2) = 22.036, P = 0.000). The grades of Hp density within the surface mucous gel layer in atrophic gastritis and gastric cancer was significantly higher than that of the superficial gastritis group (53.7%, chi(2) = 11.002, P < 0.05; chi(2) = 19.401, P < 0.05). CONCLUSION: The density of Hp colonization in gastric mucosa epithelium cell surface and lumen of gland gradually decreases in the order of superficial gastritis-atrophic gastritis-gastric cancer, while the density of Hp within the surface mucous gel layer increases in the order of superficial gastritis- atrophic gastritis-gastric cancer.


Subject(s)
Gastric Mucosa/microbiology , Helicobacter pylori/isolation & purification , Stomach Diseases/microbiology , Adult , Aged , Aged, 80 and over , Colony Count, Microbial , Female , Gastric Mucosa/pathology , Gastritis/microbiology , Gastritis/pathology , Gastritis, Atrophic/microbiology , Gastritis, Atrophic/pathology , Helicobacter Infections/microbiology , Helicobacter Infections/pathology , Helicobacter pylori/growth & development , Humans , Immunohistochemistry , Male , Middle Aged , Stomach Diseases/pathology , Stomach Neoplasms/microbiology , Stomach Neoplasms/pathology , Stomach Ulcer/microbiology , Stomach Ulcer/pathology , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...