Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 103
Filter
Add more filters

Country/Region as subject
Affiliation country
Publication year range
1.
Cell ; 187(7): 1651-1665.e21, 2024 Mar 28.
Article in English | MEDLINE | ID: mdl-38490195

ABSTRACT

The immune checkpoint blockade (ICB) response in human cancers is closely linked to the gut microbiota. Here, we report that the abundance of commensal Lactobacillus johnsonii is positively correlated with the responsiveness of ICB. Supplementation with Lactobacillus johnsonii or tryptophan-derived metabolite indole-3-propionic acid (IPA) enhances the efficacy of CD8+ T cell-mediated αPD-1 immunotherapy. Mechanistically, Lactobacillus johnsonii collaborates with Clostridium sporogenes to produce IPA. IPA modulates the stemness program of CD8+ T cells and facilitates the generation of progenitor exhausted CD8+ T cells (Tpex) by increasing H3K27 acetylation at the super-enhancer region of Tcf7. IPA improves ICB responsiveness at the pan-cancer level, including melanoma, breast cancer, and colorectal cancer. Collectively, our findings identify a microbial metabolite-immune regulatory pathway and suggest a potential microbial-based adjuvant approach to improve the responsiveness of immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes , Immunotherapy , Lactobacillus , Neoplasms , Humans , Lactobacillus/metabolism , Neoplasms/immunology , Neoplasms/therapy , Indoles/metabolism , Immune Checkpoint Inhibitors/therapeutic use
2.
Gastroenterology ; 165(1): 88-103, 2023 Jul.
Article in English | MEDLINE | ID: mdl-36921674

ABSTRACT

BACKGROUND & AIMS: Gastric cancer (GC) is a major cancer type characterized by high heterogeneity in both tumor cells and the tumor immune microenvironment (TIME). One intractable GC subtype is gastric signet-ring cell carcinoma (GSRCC), which is associated with poor prognosis. However, it remains unclear what the GSRCC TIME characteristics are and how these characteristics may contribute to clinical outcomes. METHODS: We enrolled 32 patients with advanced GC of diverse subtypes and profiled their TIME using an immune-targeted single-cell profiling strategy, including (1) immune-targeted single-cell RNA sequencing (n = 20 patients) and (2) protein expression profiling by a targeted antibody panel for mass cytometry (n = 12 patients). We also generated matched V(D)J (variable, diversity, and joining gene segments) sequencing of T and B cells along CD45+ immunocytes. RESULTS: We found that compared to non-GSRCC, the GSRCC TIME appears to be quiescent, where both CD4+ and CD8+ T cells are difficult to be mobilized, which further impairs the proper functions of B cells. CXCL13, mainly produced by follicular helper T cells, T helper type 17, and exhausted CD8+ T cells, is a central coordinator of this transformation. We show that CXCL13 expression can predict the response to immune checkpoint blockade in GC patients, which may be related to its effects on tertiary lymphoid structures. CONCLUSIONS: Our study provides a comprehensive molecular portrait of immune cell compositions and cell states in advanced GC patients, highlighting adaptive immune irresponsiveness in GSRCC and a mediator role of CXCL13 in TIME. Our targeted single-cell transcriptomic and proteomic profiling represents a powerful approach for TIME-oriented translational research.


Subject(s)
Carcinoma, Signet Ring Cell , Stomach Neoplasms , Humans , Stomach Neoplasms/genetics , CD8-Positive T-Lymphocytes , Proteomics , Carcinoma, Signet Ring Cell/genetics , Tumor Microenvironment
3.
J Clin Gastroenterol ; 58(1): 53-56, 2024 01 01.
Article in English | MEDLINE | ID: mdl-36507929

ABSTRACT

OBJECTIVES: This study aimed to confirm whether premedication with pronase before endoscopy improves mucosal visualization and increases precancerous lesion and cancer lesion detection rates. MATERIALS AND METHODS: From June 2018 to April 2019, out-patients scheduled for endoscopy from 13 hospitals were screened to be randomly allocated in a 2:1 ratio to premedication with pronase (group A) and water (group B). The primary endpoint was mucosal visibility scores, and the secondary endpoint was precancerous and cancer lesion detection rates. This trial was registered at Chinese Clinical Trial Registry, and the registration number was ChiCTR1800016853. RESULTS: Group A showed significantly lower mucosal visibility scores (better mucosal visibility) of esophagus, stomach, and duodenum than group B, with all P -values <0.001. The overall cancer detection rates between group A and group B were 0.83 and 1.08%, and overall detection rates of precancerous and cancer lesion were 4.4 and 4.9%, both without significant difference ( P =1.000 and 0.824). In addition, the flushing volume (milliliter) of group A (10.52±23.41) was less than group B (36.30±52.11) ( P <0.001), and the flushing frequency of group A (0.46±1.01) was fewer than group B (1.62±2.12) ( P <0.001). CONCLUSIONS: Premedication with pronase could achieve better mucosal visibility and decrease flushing frequency and volume, but may not increase lesion detection rates.


Subject(s)
Endoscopy, Gastrointestinal , Precancerous Conditions , Humans , Pronase/therapeutic use , Prospective Studies , Premedication
4.
Int J Mol Sci ; 24(6)2023 Mar 19.
Article in English | MEDLINE | ID: mdl-36982915

ABSTRACT

Resmetirom, a liver-directed, orally active agonist of THR-ß, could play a favorable role in treating NASH, but little is known about the underlying mechanism. A NASH cell model was established to test the preventive effect of resmetirom on this disease in vitro. RNA-seq was used for screening, and rescue experiments were performed to validate the target gene of the drug. A NASH mouse model was used to further elucidate the role and the underlying mechanism of resmetirom. Resmetirom effectively eliminated lipid accumulation and decreased triglyceride (TG) levels. In addition, repressed RGS5 in the NASH model could be recovered by resmetirom treatment. The silencing of RGS5 effectively impaired the role of resmetirom. In the NASH mouse model, obvious gray hepatization, liver fibrosis and inflammation, and increased macrophage infiltration were observed in liver tissues, while resmetirom almost returned them to normal conditions as observed in the control group. Pathological experimental data also confirmed that resmetirom has great potential in NASH treatment. Finally, RGS5 expression was suppressed in the NASH mouse model, but it was upregulated by resmetirom treatment, while the STAT3 and NF-κB signaling pathways were activated in NASH but inhibited by the agent. Resmetirom could improve NASH by recovering RGS5 expression and subsequently inactivating the STAT3 and NF-κB signaling pathways.


Subject(s)
Non-alcoholic Fatty Liver Disease , RGS Proteins , Mice , Animals , NF-kappa B/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Signal Transduction , Liver/metabolism , Mice, Inbred C57BL , RGS Proteins/genetics , RGS Proteins/metabolism
5.
Genomics ; 113(4): 2683-2694, 2021 07.
Article in English | MEDLINE | ID: mdl-34129933

ABSTRACT

The AJCC staging system is considered as the golden standard in clinical practice. However, it remains some pitfalls in assessing the prognosis of gastric cancer (GC) patients with similar clinicopathological characteristics. We aim to develop a new clinic and genetic risk score (CGRS) to improve the prognosis prediction of GC patients. We established genetic risk score (GRS) based on nine-gene signature including APOD, CCDC92, CYS1, GSDME, ST8SIA5, STARD3NL, TIMEM245, TSPYL5, and VAT1 based on the gene expression profiles of the training set from the Asian Cancer Research Group (ACRG) cohort by LASSO-Cox regression algorithms. CGRS was established by integrating GRS with clinical risk score (CRS) derived from Surveillance, Epidemiology, and End Results (SEER) database. GRS and CGRS dichotomized GC patients into high and low risk groups with significantly different prognosis in four independent cohorts with different data types, such as microarray, RNA sequencing and qRT-PCR (all HR > 1, all P < 0.001). Both GRS and CGRS were prognostic signatures independent of the AJCC staging system. Receiver operating characteristic (ROC) analysis showed that area under ROC curve of CGRS was larger than that of the AJCC staging system in most cohorts we studied. Nomogram and web tool (http://39.100.117.92/CGRS/) based on CGRS were developed for clinicians to conveniently assess GC prognosis in clinical practice. CGRS integrating genetic signature with clinical features shows strong robustness in predicting GC prognosis, and can be easily applied in clinical practice through the web application.


Subject(s)
Stomach Neoplasms , Transcriptome , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Humans , Nomograms , Nuclear Proteins/genetics , Prognosis , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology
6.
FASEB J ; 34(2): 2161-2172, 2020 02.
Article in English | MEDLINE | ID: mdl-31909528

ABSTRACT

Gastric cancer (GC) patients with metastasis had limited treatment options and dismal outcome. We have previously reported the aberrant expression of Zic family member 1 (Zic1) in GC. However, the functional roles and underlying mechanism of Zic1 in GC metastasis remain unknown. Here, we demonstrate that lower expression of Zic1 was correlated with more lymph node metastasis and poor outcome of GC patients. Ectopic expression of Zic1 suppressed both lung metastasis and peritoneal tumor dissemination of GC in mice. The metastatic suppressing ability of Zic1 was mediated by regulating the process of cell invasion, adhesion and epithelial-mesenchymal transition (EMT). Mechanistically, Zic1 could downregulate Wnt targets including c-Myc and Cyclin D1 by inhibiting LEF transcriptional activity in GC cells. Notably, Zic1 was inversely related to the expression of Cyclin D1 in GC tissues tested. In addition, Zic1 could physically interact with ß-catenin/transcription factor 4 (TCF4) and disrupt their complex formation, while not affecting ß-catenin nuclear localization. Collectively, our study indicated that Zic1 suppressed GC metastasis through attenuating Wnt/ß-catenin signaling and the EMT process. Our work may provide novel therapeutic strategies for the metastasis of GC.


Subject(s)
Epithelial-Mesenchymal Transition , Neoplasm Proteins/metabolism , Stomach Neoplasms/metabolism , Transcription Factors/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism , Animals , Cell Line, Tumor , Female , HEK293 Cells , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Metastasis , Neoplasm Proteins/genetics , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Transcription Factors/genetics , beta Catenin/genetics
7.
FASEB J ; 34(6): 8544-8557, 2020 06.
Article in English | MEDLINE | ID: mdl-32356314

ABSTRACT

MicroRNAs (miRNAs) play important roles in posttranscriptional regulation and may serve as targets for the diagnosis and treatment of cancers. Nevertheless, a comprehensive understanding of miRNAs profiles in gastric cancer progression is still lacking. Here, we report that miR-129-5p is downregulated in gastric cancer by analyzing TCGA database (n = 41) and clinical tumor samples (n = 60). MiR-129-5p transfection suppressed gastric cancer cell proliferation through inducing G1 phase arrest in vitro and inhibit xenograft tumor growth in vivo. MiR-129-5p directly targeted the 3' untranslated regions (3' UTR) of HOXC10 mRNA and downregulated its expression. Importantly, miR-129-5p could reverse the oncogenic effect induced by HOXC10. We systemically screened the downstream target of HOXC10 by ChIP sequencing, and found that HOXC10 could transcriptionally regulate the expression of Cyclin D1 and facilitate G1/S cell cycle transition. Notably, high levels of HOXC10 and Cyclin D1 were related with poor prognosis of gastric cancer patients (n = 90). These findings reveal a novel role of miR-129-5p/HOXC10/Cyclin D1 axis in modulating cell cycle and gastric tumorigenesis, which might provide potential prognostic biomarkers and therapeutic targets for gastric cancer patients.


Subject(s)
Cell Cycle Checkpoints/genetics , Cyclin D1/genetics , Homeodomain Proteins/genetics , MicroRNAs/genetics , Stomach Neoplasms/genetics , 3' Untranslated Regions/genetics , Animals , Cell Line , Cell Line, Tumor , Cell Proliferation/genetics , Disease Progression , Down-Regulation/genetics , Female , G1 Phase/genetics , Gene Expression Regulation, Neoplastic/genetics , HEK293 Cells , Humans , Mice , Mice, Inbred BALB C , Oncogenes/genetics , S Phase/genetics , Stomach/pathology
8.
J Nanobiotechnology ; 19(1): 381, 2021 Nov 21.
Article in English | MEDLINE | ID: mdl-34802453

ABSTRACT

BACKGROUND: The prognosis of patients with advanced gastric cancer (GC) remains unsatisfactory owing to distant metastasis and resistance to concurrent systemic therapy. Cancer-associated fibroblasts (CAFs), as essential participators in the tumor microenvironment (TME), play a vital role in tumor progression. Thus, CAFs-targeting therapy is appealing for remodeling TME and sensitizing GC to conventional systemic therapy. METHODS: Amphiphilic SN38 prodrug polymeric micelles (PSN38) and encapsulated the hydrophobic esterase-responsive prodrug of Triptolide (TPL), triptolide-naphthalene sulfonamide (TPL-nsa), were synthesized to form PSN38@TPL-nsa nanoparticles. Then, CAFs were isolated from fresh GC tissues and immortalized. TPL at low dose concentration was used to investigate its effect on CAFs and CAFs-induced GC cells proliferation and migration. The synergistic mechanism and antitumor efficiency of SN38 and TPL co-delivery nanoparticle were investigated both in vitro and in vivo. RESULTS: Fibroblast activation protein (FAP), a marker of CAFs, was highly expressed in GC tissues and indicated poorer prognosis. TPL significantly reduced CAFs activity and inhibited CAFs-induced proliferation, migration and chemotherapy resistance of GC cells. In addition, TPL sensitized GC cells to SN38 treatment through attenuated NF-κB activation in both CAFs and GC cells. PSN38@TPL-nsa treatment reduced the expression of collagen, FAP, and α-smooth muscle actin (α-SMA) in tumors. Potent inhibition of primary tumor growth and vigorous anti-metastasis effect were observed after systemic administration of PSN38@TPL-nsa to CAFs-rich peritoneal disseminated tumor and patient-derived xenograft (PDX) model of GC. CONCLUSION: TPL suppressed CAFs activity and CAFs-induced cell proliferation, migration and chemotherapy resistance to SN38 of GC. CAFs-targeted TPL and SN38 co-delivery nanoparticles exhibited potent efficacy of antitumor and reshaping TME, which was a promising strategy to treat advanced GC.


Subject(s)
Antineoplastic Agents , Micelles , Prodrugs , Stomach Neoplasms/metabolism , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Cancer-Associated Fibroblasts/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Diterpenes/chemistry , Diterpenes/pharmacokinetics , Diterpenes/pharmacology , Drug Synergism , Epoxy Compounds/chemistry , Epoxy Compounds/pharmacokinetics , Epoxy Compounds/pharmacology , Female , Humans , Mice , Mice, Inbred BALB C , Phenanthrenes/chemistry , Phenanthrenes/pharmacokinetics , Phenanthrenes/pharmacology , Prodrugs/chemistry , Prodrugs/pharmacokinetics , Prodrugs/pharmacology , Tumor Microenvironment/drug effects
9.
Environ Toxicol ; 36(4): 520-529, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33166055

ABSTRACT

Colorectal cancer stem cell (CSC) has been regarded to be the root of colorectal cancer progression. However, there is still no effective therapeutic method targeting colorectal CSC in clinical application. Here, we investigated the effects of dichloroacetate (DCA) on colorectal cancer cell stemness. We showed that DCA could reduce colorectal cancer cell stemness in a dose-dependent manner, which is evident by the decreased expression of stemness markers, tumor cell sphere-formation and cell migration ability. In addition, it was found that DCA trigerred the ferroptosis of colorectal CSC, which is characterized as the upregulation of iron concentration, lipid peroxides, and glutathione level, and decreased cell viability. Mechanistic studies demonstrated that DCA could sequester iron in lysosome and thus trigger ferroptosis, which is necessary for DCA-mediated attenuation on colorectal cancer cell stemness. Taken together, this work suggests that DCA might be a colorectal CSC-killer.


Subject(s)
Colorectal Neoplasms/pathology , Dichloroacetic Acid/pharmacology , Ferroptosis/drug effects , Iron/metabolism , Lysosomes/drug effects , Neoplastic Stem Cells/drug effects , Sequestering Agents/pharmacology , Cell Movement/drug effects , Cell Survival/drug effects , Colorectal Neoplasms/metabolism , Dose-Response Relationship, Drug , HCT116 Cells , HT29 Cells , Humans , Lysosomes/metabolism , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology
10.
Nanomedicine ; 29: 102250, 2020 10.
Article in English | MEDLINE | ID: mdl-32619706

ABSTRACT

Accurate identification of tumor tissues and their margins are still challenging for conventional clinical imaging methods during liver cancer surgery. In this study, dual-band confocal laser endomicroscopy (CLE) combined with image mosaic was used to guide liver cancer surgery. In the experiments with mice bearing orthotropic liver tumor, CLE can accurately detect the tumors and identify their margins with two excitation wavelengths of 488 nm and 660 nm by clinically available dyes fluorescein sodium (FS) or indocyanine green (ICG). The mosaic CLE images enlarged the imaging field and detected the liver tumor margins more accurately. Normal liver tissues fluorescence intensity of CLE images was significantly higher than that of tumor tissues in the same tumor-bearing mice (P < 0.0001). Overall, dual-band CLE imaging demonstrates to be a promising method to identify liver tumor tissues and margins, which has the prospect of clinical application and helps to achieve intraoperative radical resection.


Subject(s)
Image Processing, Computer-Assisted , Liver Neoplasms/diagnostic imaging , Liver/ultrastructure , Microscopy, Confocal , Animals , Cell Line, Tumor , Heterografts , Humans , Indocyanine Green/chemistry , Liver/pathology , Liver Neoplasms/pathology , Mice
11.
Neuroendocrinology ; 107(3): 209-217, 2018.
Article in English | MEDLINE | ID: mdl-29920478

ABSTRACT

BACKGROUND: The effectiveness of the current Tumor, Lymph node, Metastases (TNM) staging system in small intestinal neuroendocrine tumors (SiNETs) is unsatisfactory. Current N classification only distinguishes between node-negative and node-positive status. We aim to refine the N classification for updated TNM stage. METHODS: During the period from 1988 to 2012, patients with non-metastatic -SiNETs were enrolled in the Surveillance, Epidemiology, and End Results database. Using the X-tile program, we calculated an optimal cutoff value for lymph node ratio (LNR) and proposed a novel Nr category. Survival outcomes were estimated using the Kaplan-Meier method and Cox regression model. Adjusted hazard ratio (HR) and cluster analysis were performed to differentiate TNrM stages. RESULTS: Patients with existing TNM stage I and II had equivalent survival prognosis (p = 0.214). Current N classification was not a significant predictor of patient survival (p = 0.372). Multivariate analyses identified the revised Nr classification, based on LNR of 0.6 optimal cutoff value, as an independent prognostic factor (p = 0.020). By incorporating the Nr classification, a revised TNrM, which categorized patients into 3 new stages was proposed: stage I (T1-2Nr0-1), stage II (T3Nr0-1), and stage III (TxNr2 or T4Nrx). TNrM stage had better stratification according to the survival outcome (primary cohort: stage I: reference, II: HR 3.852, 95% CI 1.731-8.575; III: HR 7.169, 95% CI 3.220-15.963, p < 0.001; validation cohort: stage I: reference, II: HR 2.034; III: HR 3.815; p < 0.001). CONCLUSIONS: The Nr classification more accurately stratifies SiNET patients than current N classification. The new TNrM staging system could improve the ability to predict survival outcome of SiNET patients.


Subject(s)
Intestinal Neoplasms/pathology , Intestine, Small/pathology , Lymph Nodes/pathology , Lymphatic Metastasis/pathology , Neuroendocrine Tumors/pathology , Adolescent , Adult , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Neoplasm Staging , SEER Program , Young Adult
12.
Gastric Cancer ; 21(5): 756-764, 2018 09.
Article in English | MEDLINE | ID: mdl-29417297

ABSTRACT

BACKGROUND: Long non-coding RNAs (LncRNAs) exert their functions mainly by binding to their corresponding proteins. Runt-related transcription factor 3 (Runx3) is an important transcription factor that functions as a tumor suppressor in gastric cancer. Whether there is an interplay between LncRNAs and Runx3 remains unclear. METHODS: RPISeq was applied to screen the LncRNAs that potentially bind to Runx3. The interaction between LncRNA HOX antisense intergenic RNA (HOTAIR) and Runx3 was validated by RNA Immunoprecipitation and RNA pull-down assays. The role of Mex3b in the ubiquitination of Runx3 induced by HOTAIR was assessed by immunoprecipitation. Pearson's correlation between HOTAIR mRNA expression and Runx3 protein expression was analyzed. Cell migration and invasion were explored by transwell assays. RESULTS: We found that HOTAIR was bound to Runx3 protein and identified the fragment of HOTAIR spanning 1951-2100 bp as the specific binding site. In addition, mex-3 RNA binding family member B (Mex3b) was an E3 ligase involved in HOTAIR-induced ubiquitous degradation of Runx3. Silencing the expression of HOTAIR or Mex3b attenuated the degradation of Runx3. In human gastric cancer tissues, HOTAIR was negatively associated with the expression level of Runx3 protein (Pearson coefficient - 0.501, p = 0.025). Inhibition of HOTAIR significantly suppressed gastric cancer cell migration and invasion through upregulating claudin1, which could be reversed by co-deficiency of Runx3. CONCLUSIONS: These results uncovered the novel interaction between HOTAIR and Runx3, and provided potential therapeutic targets on the metastasis of gastric cancer.


Subject(s)
Core Binding Factor Alpha 3 Subunit/metabolism , RNA, Long Noncoding/metabolism , RNA-Binding Proteins/metabolism , Stomach Neoplasms/genetics , Binding Sites , Cell Line, Tumor , Cell Movement/genetics , Core Binding Factor Alpha 3 Subunit/genetics , Gene Expression Regulation, Neoplastic , Humans , RNA, Long Noncoding/genetics , RNA-Binding Proteins/genetics , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Ubiquitination
13.
Cancer ; 123(13): 2432-2443, 2017 Jul 01.
Article in English | MEDLINE | ID: mdl-28267199

ABSTRACT

BACKGROUND: The real-world occurrence rate of non-breast cancer-specific death (non-BCSD) and its impact on patients with breast cancer are poorly recognized. METHODS: Women with resectable breast cancer from 1990 to 2007 in the Surveillance, Epidemiology, and End Results database (n = 199,963) were analyzed. The outcome events of breast cancer were classified as breast cancer-specific death (BCSD), non-BCSD, or survival. Binary logistics was used to estimate the occurrence rates of non-BCSD and BCSD with different clinicopathological factors. The Gray method was used to measure the cumulative incidence of non-BCSD and BCSD. The ratio of non-BCSDs to all causes of death and stacked cumulative incidence function plots were used to present the impact of non-BCSD on overall survival (OS). Models of Cox proportional hazards regression and competing risk regression were compared to highlight the suitable model. RESULTS: There were 12,879 non-BCSDs (6.44%) and 28,784 BCSDs (14.39%). The oldest age group (>62 years), black race, and a single or divorced marital status were associated with more non-BCSDs. With adjustments for age, a hormone receptor-positive (HoR+) status was no longer related to increased non-BCSDs. In patients with grade 1, stage I disease and an HoR+ status as well as the oldest subgroup, a great dilution of non-BCSD on all causes of death could be observed, and this led to incorrect interpretations. The inaccuracy, caused by the commonly used Cox proportional hazards model, could be corrected by a competing risk model. CONCLUSIONS: OS was largely impaired by non-BCSD during early breast cancer. For some future clinical trial planning, especially for the oldest patients and those with HoR+ breast cancer, non-BCSD should be considered a competing risk event. Cancer 2017;123:2432-43. © 2017 American Cancer Society.


Subject(s)
Breast Neoplasms/mortality , Carcinoma, Ductal, Breast/mortality , Carcinoma, Lobular/mortality , Survival Rate , Adult , Black or African American/statistics & numerical data , Age Factors , Aged , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Breast Neoplasms/therapy , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Ductal, Breast/pathology , Carcinoma, Ductal, Breast/therapy , Carcinoma, Lobular/metabolism , Carcinoma, Lobular/pathology , Carcinoma, Lobular/therapy , Cause of Death , Ethnicity/statistics & numerical data , Female , Humans , Incidence , Kaplan-Meier Estimate , Logistic Models , Marital Status , Mastectomy , Middle Aged , Neoplasm Staging , Proportional Hazards Models , Radiotherapy, Adjuvant , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Retrospective Studies , SEER Program , United States
15.
Gastrointest Endosc ; 81(1): 186-93, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25012561

ABSTRACT

BACKGROUND: Patients frequently consult primary care physicians and gastroenterologists when experiencing chronic abdominal pain. Although its diagnostic efficacy in these settings is uncertain, small-bowel capsule endoscopy (SBCE) has been used to evaluate the unexplained reasons for abdominal pain. OBJECTIVE: To evaluate the diagnostic yield of SBCE in patients with unexplained chronic abdominal pain. DESIGN: We performed a retrospective review of publications reporting the diagnostic yield of SBCE in patients with unexplained chronic abdominal pain and calculated the overall diagnostic yield. SETTING: Two investigators independently searched studies from databases and analyzed the results. PATIENTS: A total of 1520 patients from 21 studies were included. INTERVENTIONS: Small-bowel capsule endoscopy. MAIN OUTCOME MEASUREMENTS: Per-patient diagnostic yield, with 95% confidence intervals (CI), was evaluated by a random-effect model. Clear categorical analysis also was performed. RESULTS: The pooled diagnostic yield of SBCE in patients with unexplained chronic abdominal pain was 20.9% (95% CI, 15.9%-25.9%), with high heterogeneity (I(2) = 80.0%; P < .001). Inflammatory lesions were the most common (78.3%) positive findings, followed by tumors (9.0%). LIMITATIONS: Heterogeneity among studies, retrospective design, variable chronicity of abdominal pain, and different previous examinations before SBCE. CONCLUSION: SBCE provides a noninvasive diagnostic tool for patients with unexplained chronic abdominal pain, but the diagnostic yield is limited (20.9%). Among patients with positive findings, inflammatory lesions are the most common.


Subject(s)
Abdominal Pain/diagnosis , Capsule Endoscopy/methods , Crohn Disease/diagnosis , Intestinal Neoplasms/diagnosis , Intestine, Small/pathology , Celiac Disease/diagnosis , Chronic Disease , Enteritis/diagnosis , Humans , Intestinal Diseases/diagnosis
16.
Int J Cancer ; 134(9): 2041-50, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24519534

ABSTRACT

The KRAS oncogene influences angiogenesis, metastasis and chemoresistance in colorectal cancers (CRCs), and these processes are all enhanced in hypoxic conditions. To define functional activities of mutant KRAS in a hypoxic microenvironment, we first performed cDNA microarray experiments in isogenic DKs5 and DKO3 colon cancer cell lines that differ only by their expression of mutant KRAS (K-ras(D13)). Adrenomedullin (ADM) was identified as one of the most significantly upregulated genes in DKs5 cells that express the KRAS oncogene in hypoxia (3.2-fold, p = 1.47 × 10(-5)). Ectopic expression of mutant KRAS (K-ras(V12)) in Caco-2 cells (K-ras(WT)) induced ADM, whereas selective knockdown of mutant KRAS alleles (K-ras(D13) or K-ras(V12)) in HCT116, DLD1 and SW480 colon cancer cells suppressed the expression of ADM in hypoxia. Knockdown of ADM in colon tumor xenografts blocked angiogenesis and stimulated apoptosis, resulting in tumor suppression. Furthermore, ADM also regulated colon cancer cell invasion in vitro. Among 56 patients with CRC, significantly higher expression levels of ADM were observed in samples harboring a KRAS mutation. Collectively, ADM is a new target of oncogenic KRAS in the setting of hypoxia. This observation suggests that therapeutic targets may differ depending upon the specific tumor microenvironment.


Subject(s)
Adrenomedullin/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Proto-Oncogene Proteins/genetics , ras Proteins/genetics , Animals , Cell Hypoxia/physiology , Cell Line, Tumor , Female , Humans , Immunohistochemistry , Mice, Nude , Oligonucleotide Array Sequence Analysis , Proto-Oncogene Proteins p21(ras) , Tumor Microenvironment/physiology , Xenograft Model Antitumor Assays
17.
Front Med (Lausanne) ; 11: 1344644, 2024.
Article in English | MEDLINE | ID: mdl-38716417

ABSTRACT

Objective: This study aimed to systematically evaluate the efficacy and safety of the double-guidewire technique along with other methods (persistent standard cannulation techniques, transpancreatic sphincterotomy, and pancreatic stent-assisted technique) for difficult biliary cannulation. Methods: Two researchers searched for literature on the efficacy and safety of the double-guidewire technique and other techniques in difficult biliary cannulation in databases, including PubMed, Embase, Cochrane, China National Knowledge Infrastructure, and Wanfang Data, based on the inclusion and exclusion criteria. The success rate of cannulation, duration of cannulation, post-ERCP pancreatitis, and overall postoperative complications were also analyzed using RevMan 5.4 software. Results: In total, 20 randomized controlled trial (RCT) studies involving 2008 participants were identified. The success rate of cannulation in the double-guidewire technique was much higher than that in persistent standard cannulation techniques [RR = 1.37, 95%CI (1.05, 1.79), p = 0.02]. However, it was lower than the success rate observed with transpancreatic sphincterotomy [RR = 0.89, 95%CI (0.81, 0.97), p = 0.01]. There was no significance in post-ERCP pancreatitis [RR = 1.09, 95% CI (0.85, 1.40), p = 0.49], overall postoperative complications [RR = 0.90, 95% CI (0.56, 1.45), p = 0.66], and duration of cannulation [SMD = -0.14, 95%C I (-1.43, 1.15), p = 0.83] between the double-guidewire technique and other techniques. Conclusion: This study demonstrated that the success rate of cannulation ranged from transpancreatic sphincterotomy to the double-guidewire technique and then to persistent standard cannulation techniques.

18.
Sci China Life Sci ; 67(6): 1199-1211, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38240906

ABSTRACT

Metastasis accounts for the major cause of colorectal cancer (CRC) related mortality due to the lack of effective treatments. In this study, we integrated the single-cell RNA-seq (scRNA-seq) and bulk RNA-seq data and identified the transcriptional coactivator SUB1 homolog (Sac-Saccharomyces cerevisiae)/PC4 (positive cofactor 4) associated with CRC metastasis. Elevated SUB1 expression was correlated with advanced tumor stage and poor survival in CRC. In vivo and vitro assays showed that SUB1 depletion could inhibit the invasive and metastatic abilities of CRC cells. SUB1 activated NF-κB signaling and its transcriptional target genes CXCL1 and CXCL3 to drive CRC metastasis. Mechanistically, SUB1 integrated with the E3 ubiquitin-protein ligase UBR5 and increased its protein level in CRC cells. Subsequently, the increased UBR5 mainly mediated Lys11-linked polyubiquitination and degradation of NF-κB negative regulator UBXN1, thus to activate the NF-κB signaling. Overall, our study demonstrated that SUB1 promoted CRC progression by modulating UBR5/UBXN1 and activating NF-κB signaling, providing a new therapeutic strategy for treating metastatic CRC through targeting SUB1.


Subject(s)
Colorectal Neoplasms , NF-kappa B , Signal Transduction , Ubiquitin-Protein Ligases , Ubiquitination , Humans , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Colorectal Neoplasms/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , NF-kappa B/metabolism , Animals , Mice , Neoplasm Metastasis , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Mice, Nude , Transcription Factors/metabolism , Transcription Factors/genetics , Female , Male , Adaptor Proteins, Signal Transducing
19.
Aging Cell ; 23(4): e14081, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38236004

ABSTRACT

Aging-induced cognitive impairment is associated with a loss of metabolic homeostasis and plasticity. An emerging idea is that targeting key metabolites is sufficient to impact the function of other organisms. Therefore, more metabolism-targeted therapeutic intervention is needed to improve cognitive impairment. We first conducted untargeted metabolomic analyses and 16S rRNA to identify the aging-associated metabolic adaption and intestinal microbiome change. Untargeted metabolomic analyses of plasma revealed L-arginine metabolic homeostasis was altered during the aging process. Impaired L-arginine metabolic homeostasis was associated with low abundance of intestinal Akkermansia muciniphila (AKK) colonization in mice. Long-term supplementation of AKK outer membranes protein-Amuc_1100, rescued the L-arginine level and restored cognitive impairment in aging mice. Mechanically, Amuc_1100 acted directly as a source of L-arginine and enriched the L-arginine-producing bacteria. In aged brain, Amuc_1100 promoted the superoxide dismutase to alleviated oxidation stress, and increased nitric oxide, derivatives of L-arginine, to improve synaptic plasticity. Meanwhile, L-arginine repaired lipopolysaccharide-induced intestinal barrier damage and promoted growth of colon organoid. Our findings indicated that aging-related cognitive impairment was closely associated with the disorders of L-arginine metabolism. AKK-derived Amuc_1100, as a potential postbiotic, targeting the L-arginine metabolism, might provide a promising therapeutic strategy to maintain the intestinal homeostasis and cognitive function in aging.


Subject(s)
Cognitive Dysfunction , Verrucomicrobia , Mice , Animals , RNA, Ribosomal, 16S , Homeostasis , Arginine
20.
World J Gastroenterol ; 30(9): 1108-1120, 2024 Mar 07.
Article in English | MEDLINE | ID: mdl-38577179

ABSTRACT

BACKGROUND: Although chronic erosive gastritis (CEG) is common, its clinical characteristics have not been fully elucidated. The lack of consensus regarding its treatment has resulted in varied treatment regimens. AIM: To explore the clinical characteristics, treatment patterns, and short-term outcomes in CEG patients in China. METHODS: We recruited patients with chronic non-atrophic or mild-to-moderate atrophic gastritis with erosion based on endoscopy and pathology. Patients and treating physicians completed a questionnaire regarding history, endoscopic findings, and treatment plans as well as a follow-up questionnaire to investigate changes in symptoms after 4 wk of treatment. RESULTS: Three thousand five hundred sixty-three patients from 42 centers across 24 cities in China were included. Epigastric pain (68.0%), abdominal distension (62.6%), and postprandial fullness (47.5%) were the most common presenting symptoms. Gastritis was classified as chronic non-atrophic in 69.9% of patients. Among those with erosive lesions, 72.1% of patients had lesions in the antrum, 51.0% had multiple lesions, and 67.3% had superficial flat lesions. In patients with epigastric pain, the combination of a mucosal protective agent (MPA) and proton pump inhibitor was more effective. For those with postprandial fullness, acid regurgitation, early satiety, or nausea, a MPA appeared more promising. CONCLUSION: CEG is a multifactorial disease which is common in Asian patients and has non-specific symptoms. Gastroscopy may play a major role in its detection and diagnosis. Treatment should be individualized based on symptom profile.


Subject(s)
Gastritis, Atrophic , Gastritis , Helicobacter Infections , Helicobacter pylori , Stomach Ulcer , Humans , Gastric Mucosa/pathology , Gastritis/diagnosis , Gastritis/drug therapy , Gastritis/epidemiology , Gastritis, Atrophic/diagnosis , Gastritis, Atrophic/epidemiology , Gastritis, Atrophic/pathology , Gastroscopy , Helicobacter Infections/pathology , Life Style , Pain , Stomach Ulcer/pathology
SELECTION OF CITATIONS
SEARCH DETAIL