Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
World J Gastrointest Oncol ; 14(12): 2329-2339, 2022 Dec 15.
Article in English | MEDLINE | ID: mdl-36568940

ABSTRACT

BACKGROUND: Nucleus accumbens-1 (NAC-1) is highly expressed in a variety of tumors, including colon cancer, and is closely associated with tumor recurrence, metastasis, and invasion. AIM: To determine whether and how NAC-1 affects antitumor immunity in colon cancer. METHODS: NAC-1-siRNA was transfected into RKO colon cancer cells to knock down NAC expression; tumor cells with or without knockdown of NAC-1 were treated with CD8+ T cells to test their cytocidal effect. The level of the immune checkpoint programmed death receptor-1 ligand (PD-L1) in colon cancer cells with or without knockdown of NAC-1 was analyzed using Quantitative real-time polymerase chain reaction and Western blotting. A double luciferase reporter assay was used to examine the effects of NAC-1 on the transcription of PD-L1. Mice bearing MC-38-OVA colon cancer cells expressing NAC-shRNA or control-shRNA were treated with OT-I mouse CD8+ T cells to determine the tumor response to immunotherapy. Immune cells in the tumor tissues were analyzed using flow cytometry. NAC-1, PD-L1 and CD8+ T cells in colon cancer specimens from patients were examined using immunohistochemistry staining. RESULTS: Knockdown of NAC-1 expression in colon cancer cells significantly enhanced the cytocidal effect of CD8+ T cells in cell culture experiments. The sensitizing effect of NAC-1 knockdown on the antitumor action of cytotoxic CD8+ T cells was recapitulated in a colon cancer xenograft animal model. Furthermore, knockdown of NAC-1 in colon cancer cells decreased the expression of PD-L1 at both the mRNA and protein levels, and this effect could be rescued by transfection of an RNAi-resistant NAC-1 expression plasmid. In a reporter gene assay, transient expression of NAC-1 in colon cancer cells increased the promoter activity of PD-L1, indicating that NAC-1 regulates PD-L1 expression at the transcriptional level. In addition, depletion of tumoral NAC-1 increased the number of CD8+ T cells but decreased the number of suppressive myeloid-derived suppressor cells and regulatory T cells. CONCLUSION: Tumor expression of NAC-1 is a negative determinant of immunotherapy.

2.
Gastroenterol Res Pract ; 2019: 1484598, 2019.
Article in English | MEDLINE | ID: mdl-31885541

ABSTRACT

BACKGROUND AND AIMS: Probiotics was considered as a potential therapy for nonalcoholic fatty liver disease (NAFLD) without approval and comprehensive assessment in recent years, which call for a meta-analysis. METHODS: We performed electronic and manual searches including English and Chinese databases published before April 2019, with the use of mesh term and free text of "nonalcoholic fatty liver disease" and "probiotics." Clinical trials evaluating the efficacy of probiotic therapy in NAFLD patients were included according to the eligibility criteria. With the use of random effects models, clinical outcomes were presented as weighted mean difference (WMD) with 95% confidence interval (CI), while heterogeneity and meta-regression were also assessed. RESULTS: 28 clinical trials enrolling 1555 criterion proven NAFLD patients with the use of probiotics from 4 to 28 weeks were included. Overall, probiotic therapy had beneficial effects on body mass index (WMD: -1.46, 95% CI: [-2.44, -0.48]), alanine aminotransferase (WMD: -13.40, 95% CI: [-17.03, -9.77]), aspartate transaminase (WMD: -13.54, 95% CI: [-17.86, -9.22]), gamma-glutamyl transpeptidase (WMD: -9.88, 95% CI: [-17.77, -1.99]), insulin (WMD: -1.32, 95% CI: [-2.43, -0.21]), homeostasis model assessment-insulin resistance (WMD: -0.42, 95% CI: [-0.73, -0.12]), and total cholesterol (WMD: -15.38, 95% CI: [-26.50, -4.25]), but not in fasting blood sugar, lipid profiles, or tumor necrosis factor-alpha. CONCLUSION: The systematic review and meta-analysis support that probiotics are superior to placebo in NAFLD patients and could be utilized as a common complementary therapeutic approach.

3.
World J Gastroenterol ; 24(1): 5-14, 2018 Jan 07.
Article in English | MEDLINE | ID: mdl-29358877

ABSTRACT

Ulcerative colitis (UC) is an inflammatory disease that mainly affects the colon and rectum. It is believed that genetic factors, host immune system disorders, intestinal microbiota dysbiosis, and environmental factors contribute to the pathogenesis of UC. However, studies on the role of intestinal microbiota in the pathogenesis of UC have been inconclusive. Studies have shown that probiotics improve intestinal mucosa barrier function and immune system function and promote secretion of anti-inflammatory factors, thereby inhibiting the growth of harmful bacteria in the intestine. Fecal microbiota transplantation (FMT) can reduce bowel permeability and thus the severity of disease by increasing the production of short-chain fatty acids, especially butyrate, which help maintain the integrity of the epithelial barrier. FMT can also restore immune dysbiosis by inhibiting Th1 differentiation, activity of T cells, leukocyte adhesion, and production of inflammatory factors. Probiotics and FMT are being increasingly used to treat UC, but their use is controversial because of uncertain efficacy. Here, we briefly review the role of intestinal microbiota in the pathogenesis and treatment of UC.


Subject(s)
Bacteria/growth & development , Colitis, Ulcerative/therapy , Colon/microbiology , Fecal Microbiota Transplantation , Gastrointestinal Microbiome , Probiotics/therapeutic use , Animals , Bacteria/immunology , Colitis, Ulcerative/diagnosis , Colitis, Ulcerative/immunology , Colitis, Ulcerative/microbiology , Colon/immunology , Fecal Microbiota Transplantation/adverse effects , Host-Pathogen Interactions , Humans , Probiotics/adverse effects , Treatment Outcome
4.
Oncol Lett ; 12(5): 3278-3284, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27899994

ABSTRACT

The bone marrow microenvironment provides a relative sanctuary from cytotoxic drugs for leukemia cells. The present niche models concentrate on a two-dimensional (2D) co-culture system in vitro, which does not imitate the in vivo environment, while the 3D scaffolds are more reflective of this. Osteopontin (Opn) secreted by bone marrow osteoblasts, may participate in protecting leukemia cells from apoptosis by binding to its receptor αvß3, which can be expressed on the surface of the leukemia MV4-11 cell line. However, the association between the Opn/αvß3 axis and leukemia cells is unknown. In the present study, experiments were conducted on 3D polystyrene scaffolds coated with osteoblasts and leukemia cells. The cells were exposed to cyclo(Arg-Gly-Asp-d-Phe-Val) [c(RGDfV)] (35 nmol/ml), which blocks αvß3, for a period of 24 h. Cytarabine was applied 24 h later. The adhesion, migration and apoptosis rates, and the cell cycle of the leukemia cells were analyzed after incubation for 24 and 48 h. In contrast to the 2D culture system, the stromal cells in the scaffolds secreted significantly more alkaline phosphatase and Opn (P<0.05). c(RGDfV) disrupted the adhesion and migration between the tumor cells and the matrix, induced the leukemia cells to leave the protective microenvironment and increased their sensitivity to cell cycle-dependent agents (P<0.05). In summary, the data certified that the 3D scaffolds are suitable for the growth of cells, and that c(RGDfV) inhibits the adhesion and migration abilities of leukemia cells in the endosteal niche. Therefore, blocking the function of Opn may be beneficial in the treatment of acute myeloid leukemia.

5.
Oncotarget ; 7(37): 60230-60244, 2016 09 13.
Article in English | MEDLINE | ID: mdl-27517626

ABSTRACT

MicroRNA-126 (miR-126) suppresses the migration, proliferation and invasion of colon cancer cells. However, the underlying mechanisms of miR-126 in colon cancer have not been fully elucidated. In this study, in vivo experiments revealed that miR-126 inhibits colon cancer growth and metastasis. Furthermore, miR-126 was down-regulated in human colon cancer tissue, and its expression was inversely correlated with TNM stage and metastasis of patients. Low level of miR-126 identified patients with poor prognosis. And we found that miR-126 expression was negatively correlated with the expression levels of chemokine (C-X-C motif) receptor 4 (CXCR4) and components of signaling pathway of Ras homolog gene family, member A (RhoA) in vitro and in vivo. Moreover, we verified that miR-126 negatively regulated CXCR4 and RhoA signaling in vitro. In addition, either in miR-126-overexpressing or in miR- 126-silenced colon cancer cells, the restoration of CXCR4 could significantly reverse the proliferation and invasion, as well as abolish the effects of miR-126 on RhoA signaling pathway. Collectively, these results demonstrated that miR-126 acts as a tumor suppressor by inactivating RhoA signaling via CXCR4 in colon cancer. And miR-126 may serve as a prognostic marker for monitoring and treating colon cancer.


Subject(s)
Cell Proliferation/genetics , Colonic Neoplasms/genetics , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Receptors, CXCR4/genetics , rhoA GTP-Binding Protein/genetics , Animals , Cell Line, Tumor , Cell Movement/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Female , HCT116 Cells , Humans , Male , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Models, Genetic , Neoplasm Invasiveness , RNA Interference , Receptors, CXCR4/metabolism , Signal Transduction/genetics , rhoA GTP-Binding Protein/metabolism
6.
Blood Cells Mol Dis ; 59: 16-24, 2016 07.
Article in English | MEDLINE | ID: mdl-27282562

ABSTRACT

BACKGROUND: The multidrug resistance of leukemia cells is closely related to the microenvironment. The present leukemia microenvironment models focus on two-dimensional co-culture system in vitro which does not mimic the in vivo cell growth, while the 3D polystyrene (PS) scaffolds have the advantage. Stromal cell derived factor-1 may be involved in the shielding of endosteal niche from leukemia cells by binding to its receptor CXCR4, but the relationship between SDF-1/CXCR4 axis and leukemia cells is unclear. DESIGN AND METHODS: The experiments were built on the 3D PS scaffolds coated with osteoblasts. Stromal cells and MV4-11 cells were plated on the scaffolds. Then G-CSF, AMD3100 and cytarabine were added. Adhesive rate, SDF-1 level, migration state, apoptosis rate, and cell cycle of leukemia cells were observed after incubation at 24h and 48h. RESULTS: G-CSF decreased the level of SDF-1 and inhibited the expression of CXCR4 and promoted stationary phase leukemia cells to enter the mitotic phase and enhanced the killing effect of chemotherapeutic drugs. AMD3100 disrupted the interaction between tumors and matrix, mobilized the leukemia cells to keep away from the protective microenvironment and strengthened the cytotoxic effect of Ara-C. The combination of G-CSF and AMD3100 had stronger effects on killing the leukemia cells induced by Ara-C. CONCLUSION: It demonstrates that AMD3100 and G-CSF may inhibit adhesion and migration abilities of leukemia cells with the bone marrow niche. Both of them inhibit the role of SDF-1/CXCR4 directly or indirectly. Thus inhibiting SDF-1/CXCR4 axis may be helpful to the treatment of refractory AML.


Subject(s)
Biomimetic Materials , Granulocyte Colony-Stimulating Factor/pharmacology , Heterocyclic Compounds/pharmacology , Leukemia/pathology , Receptor Cross-Talk/drug effects , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Benzylamines , Cell Adhesion , Cell Movement , Cells, Cultured , Chemokine CXCL12/metabolism , Cyclams , Cytarabine/pharmacology , Humans , Leukemia, Myeloid, Acute/pathology , Polystyrenes , Receptors, CXCR4/metabolism , Stromal Cells/cytology
7.
Med Oncol ; 32(12): 262, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26526720

ABSTRACT

AML is a common life-threatening blood system malignancy. The treatment of AML continues to face greater challenges. An abnormal haematopoietic niche with high adhesion and proliferation might be the root cause of resistance and relapse. Most leukaemia cells are stored in the endosteal niche and recess in the G0 phase, and they are not sensitive to varieties of radiotherapies and chemotherapies. G-CSF and AMD3100 are increasingly used in priming chemotherapy. G-CSF can promote leukaemia cells to the cell cycle, which improves the complete remission rate of leukaemia patients. AMD3100, the novel CXCR4 antagonist, could also potentially promote leukaemia cells to cell cycle and improve the susceptibility of leukaemia cells to chemotherapeutic agents. The combination of them enhances anti-leukaemia effect. So in this review, we explore the function of G-CSF and/or AMD3100 in the priming chemotherapy of haematological malignants.


Subject(s)
Antineoplastic Agents , Granulocyte Colony-Stimulating Factor , Heterocyclic Compounds , Leukemia/drug therapy , Benzylamines , Cyclams , Hematopoietic Stem Cells , Humans , Stem Cell Niche
SELECTION OF CITATIONS
SEARCH DETAIL
...