Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Front Immunol ; 14: 1086898, 2023.
Article in English | MEDLINE | ID: mdl-36742294

ABSTRACT

Background: Inflammatory bowel diseases, including ulcerative colitis (UC) and Crohn's disease, are some of the most common inflammatory disorders of the gastrointestinal tract. The dysfunction of the immune system in the intestines is suggested to be the underlying cause of the pathogenesis of UC. However, the mechanisms regulating these dysfunctional immune cells and inflammatory phenotypes are still unclear. Methods: The differential expression analysis on microarray datasets were performed including GSE24287, GSE87466, GSE102133, and GSE107499, including 376 samples. "Gene Ontology" and "Kyoto Encyclopedia of Genes and Genomes" pathway enrichment analyses were conducted to identify the common differentially expressed genes (DEGs) in these datasets and explore their underlying biological mechanisms. Further algorithms like "Cell-type Identification by Estimating Relative Subsets of RNA Transcripts" were used to determine the infiltration status of immune cells in patients with UC. "Cytoscape" and "Gene Set Enrichment Analysis" were used to screen for hub genes and to investigate their biological mechanisms. The Tumor Immune Estimation Resource database was used to study the correlation between hub genes and infiltrating immune cells in patients with UC. A total of three hub genes, CCL3, MMP3, and TIMP1, were identified using Cytoscape. Results: A positive correlation was observed between these hub genes and patients with active UC. These genes served as a biomarker for active UC. Moreover, a decrease in CCL3, MMP3, and TIMP1 expression was observed in the mucosa of the intestine of patients with active UC who responded to Golimumab therapy. In addition, results show a significant positive correlation between CCL3, MMP3, and TIMP1 expression and different immune cell types including dendritic cells, macrophages, CD8+ T cells, and neutrophils in patients with colon cancer. Moreover, CCL3, MMP3, and TIMP1 expression were strongly correlated with different immune cell markers. Conclusion: Study results show the involvement of hub genes like CCL3, MMP3, and TIMP1 in the pathogenesis of UC. These genes could serve as a novel pharmacological regulator of UC. These could be used as a therapeutic target for treating patients with UC and may serve as biomarkers for immune cell infiltration in colon cancer.


Subject(s)
Colitis, Ulcerative , Colonic Neoplasms , Crohn Disease , Humans , Matrix Metalloproteinase 3 , Transcriptome , Crohn Disease/pathology , Colonic Neoplasms/genetics , Biomarkers
2.
Int J Oncol ; 56(2): 407-416, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31894326

ABSTRACT

MicroRNAs (miRNAs) are short non­coding RNAs that are known to regulate gene expression at the post­transcriptional level. miRNA expression is often deregulated in several human cancers, affecting the communication between tumor stroma and tumor cells, among other functions. Understanding the role of miRNAs in the tumor microenvironment is crucial for fully elucidating the molecular mechanisms underlying tumor progression and exploring novel diagnostic biomarkers and therapeutic targets. The present review focused on the role of miRNAs in remodeling the tumor microenvironment, with an emphasis on their impact on tumor growth, metastasis and resistance to treatment, as well as their potential clinical applications.


Subject(s)
Gene Expression Regulation, Neoplastic , MicroRNAs/metabolism , Neoplasms/genetics , Tumor Microenvironment/genetics , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Disease Progression , Drug Resistance, Neoplasm/genetics , Humans , Neoplasm Metastasis/genetics , Neoplasms/drug therapy , Neoplasms/pathology , Tumor Microenvironment/drug effects
3.
Stem Cells Int ; 2019: 8071842, 2019.
Article in English | MEDLINE | ID: mdl-31885627

ABSTRACT

Mesenchymal stem cells (MSCs) have been declared to not only participate in wound repair but also affect tumor progression. Tumor-associated MSCs, directly existing in the tumor microenvironment, play a critical role in tumor initiation, progression, and development. And different tumor-derived MSCs have their own unique characteristics. In this review, we mainly describe and discuss recent advances in our understanding of the emerging role of gastric cancer-derived MSC-like cells (GC-MSCs) in regulating gastric cancer progression and development, as well as the bidirectional influence between GC-MSCs and immune cells of the tumor microenvironment. Moreover, we also discuss the potential biomarker and therapeutic role of GC-MSCs. It is anticipated that new and deep insights into the functionality of GC-MSCs and the underlying mechanisms will promote the novel and promising therapeutic strategies against gastric cancer.

4.
Mol Cancer Res ; 17(9): 1777-1786, 2019 09.
Article in English | MEDLINE | ID: mdl-31308148

ABSTRACT

Yes-associated protein (YAP)/WW domain-containing transcription regulator 1 (TAZ) is an important transcriptional regulator and effector of the Hippo signaling pathway that has emerged as a critical determinant of malignancy in many human tumors. YAP/TAZ expression regulates the cross-talk between immune cells and tumor cells in the tumor microenvironment through its influence on T cells, myeloid-derived suppressor cells, and macrophages. However, the mechanisms underlying these effects are poorly understood. An improved understanding of the role of YAP/TAZ in tumor immunity is essential for exploring innovative tumor treatments and making further breakthroughs in antitumor immunotherapy. This review primarily focuses on the role of YAP/TAZ in immune cells, their interactions with tumor cells, and how this impacts on tumorigenesis, progression, and therapy resistance.


Subject(s)
Neoplasms/immunology , Trans-Activators/metabolism , Disease Progression , Gene Expression Regulation, Neoplastic , Humans , Macrophages/metabolism , Myeloid-Derived Suppressor Cells/metabolism , Neoplasms/metabolism , T-Lymphocytes/metabolism , Transcriptional Coactivator with PDZ-Binding Motif Proteins , Tumor Microenvironment
5.
Int J Oncol ; 51(4): 1055-1066, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28848999

ABSTRACT

Cancer-associated mesenchymal stem cells (MSCs) are critically involved in tumor development and progression. However, the mechanisms of action for MSCs in cancer remain largely unknown. Herein, we reported that the expression of Yes-associated protein 1 (YAP) was higher in gastric cancer derived mesenchymal stem cells (GC­MSCs) than that in bone marrow derived MSCs (BM­MSCs). YAP knockdown not only inhibited the growth, migration and invasion, and stemness of GC­MSCs, but also suppressed their promoting effect on gastric cancer growth in vitro and in vivo. In addition, the interference of YAP expression in GC­MSCs also attenuated the promoting role of gastric cancer cells in endothelial cell tube formation and migration. Mechanistically, YAP knockdown reduced the activation of ß-catenin and its target genes in gastric cancer cells by GC­MSCs. Taken together, these findings suggest that YAP activation in GC­MSCs plays an important role in promoting gastric cancer progression, which may represent a potential target for gastric cancer therapy.


Subject(s)
Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Nuclear Proteins/metabolism , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Transcription Factors/metabolism , Aged , Animals , Cell Cycle Proteins , Cell Proliferation/physiology , Disease Progression , Epithelial-Mesenchymal Transition , Female , Gene Knockdown Techniques , Heterografts , Humans , Male , Mice , Mice, Inbred BALB C , Middle Aged , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Nuclear Proteins/genetics , Signal Transduction , Stomach Neoplasms/blood supply , Transcription Factors/genetics
6.
Theranostics ; 7(6): 1674-1688, 2017.
Article in English | MEDLINE | ID: mdl-28529644

ABSTRACT

Human umbilical cord-derived mesenchymal stem cells (hucMSCs) are suggested as a promising therapeutic tool in regenerative medicine, however, their efficacy requires improvement. Small molecules and drugs come up to be a convenient strategy in regulating stem cells fate and function. Here, we evaluated 3,3'-diindolylmethane (DIM), a natural small-molecule compound involved in the repairing effects of hucMSCs on a deep second-degree burn injury rat model. HucMSCs primed with 50 µM of DIM exhibited desirable repairing effects compared with untreated hucMSCs. DIM enhanced the stemness of hucMSCs, which was related to the activation of Wnt/ß-catenin signaling. ß-catenin inhibition impaired the healing effects of DIM-primed hucMSCs (DIM-hucMSCs) in vivo. Moreover, we demonstrated that DIM upregulated Wnt11 expression in hucMSC-derived exosomes. Wnt11 knockdown inhibited ß-catenin activation and stemness induction in DIM-hucMSCs and abrogated their therapeutic effects in vivo. Thus, our findings indicate that DIM promotes the stemness of hucMSCs through increased exosomal Wnt11 autocrine signaling, which provides a novel strategy for improving the therapeutic effects of hucMSCs on wound healing.


Subject(s)
Indoles/metabolism , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/physiology , Signal Transduction , Umbilical Cord , Wnt Proteins/metabolism , Wound Healing , Animals , Burns/pathology , Cells, Cultured , Disease Models, Animal , Humans , Rats
7.
Biotechnol Lett ; 39(6): 929-938, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28258529

ABSTRACT

OBJECTIVE: To investigate the role of human umbilical cord mesenchymal stem cells (hucMSCs) in the treatment of dextran sulfate sodium (DSS)-induced inflammatory bowel disease (IBD). RESULTS: ICG-hucMSCs homed to colon tissues of IBD mice 12 h after injection. The injection of hucMSCs significantly relieved the IBD symptoms and inflammatory cell infiltration. The expression of IL-10 gene increased while those of 15-LOX-1, TNF-α, IL-6, IL-1ß, and IP-10 genes decreased in colon tissues and spleens of hucMSCs-treated mice. The activation of STAT3 was inhibited in colon tissues and spleens of IBD mice that were treated with hucMSCs. In addition, the percentage of macrophages decreased in colon tissues and spleens of hucMSCs-treated IBD mice. Moreover, we provided evidence that in vitro co-culture with hucMSCs inhibited the expression of 15-LOX-1, IL-6 and p-STAT3 in mouse enterocoelia macrophages. CONCLUSIONS: HucMSCs alleviate DSS-induced IBD through the modulation of 15-LOX-1 in macrophages.


Subject(s)
Arachidonate 15-Lipoxygenase/metabolism , Inflammatory Bowel Diseases/surgery , Macrophages/metabolism , Mesenchymal Stem Cell Transplantation , Animals , Colon/metabolism , Cytokines/analysis , Cytokines/metabolism , Dextran Sulfate/adverse effects , Human Umbilical Vein Endothelial Cells , Humans , Inflammatory Bowel Diseases/chemically induced , Male , Mesenchymal Stem Cells/physiology , Mice , STAT3 Transcription Factor/analysis , STAT3 Transcription Factor/metabolism , Signal Transduction , Spleen/metabolism
8.
Stem Cells ; 34(10): 2485-2500, 2016 10.
Article in English | MEDLINE | ID: mdl-27334574

ABSTRACT

Numerous studies showed that mesenchymal stem cells derived exosome (MSC-Ex) markedly enhanced tissue regeneration, however, the issue of whether MSC-Ex could control stem cells expansion after a regenerative response to prevent tissue from overcrowding and dysplasia remains to be established. Herein, we found that human umbilical cord MSC (hucMSC)-exosomal14-3-3ζ mediated the binding of YAP and p-LATS by forming a complex to promote the phosphorylation of YAP, which orchestrate exosomal Wnt4 signal in cutaneous regeneration. First, we assessed deep second-degree burn rats treated with hucMSC-Ex and discovered that hucMSC-Ex promoting self-regulation of Wnt/ß-catenin signaling at the remodeling phase of cutaneous regeneration. HucMSC-Ex restricted excessive skin cell expansion and collagen deposition at 4 weeks. Under high cell density conditions, hucMSC-Ex inhibited Wnt/ß-catenin signaling through induction of YAP phosphorylation. Second, hucMSC-Ex proteomic analysis revealed that 14-3-3 proteins could be transported by exosome. Using gain- and loss-of-function studies, our results showed that hucMSC-exosomal 14-3-3ζ controlled YAP activities and phosphorylation at Ser127 site, and were required for the binding of YAP and p-LATS. Further studies revealed that 14-3-3ζ recruited YAP and p-LATS to form a complex under high cells density status and 14-3-3ζ other than YAP or p-LATS was the key regulatory molecule of this complex. These findings collectively indicate that hucMSC-Ex functions not only as an "accelerator" of the Wnt/ß-catenin signal to repair damaged skin tissue but also as a "brake" of the signal by modulating YAP to orchestrate controlled cutaneous regeneration. Stem Cells 2016;34:2485-2500.


Subject(s)
14-3-3 Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Exosomes/metabolism , Mesenchymal Stem Cells/metabolism , Phosphoproteins/metabolism , Regeneration , Skin/metabolism , Umbilical Cord/cytology , Wnt Proteins/metabolism , Cell Count , Cell Line , Cell Proliferation/drug effects , Collagen/metabolism , Exosomes/drug effects , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Serine-Threonine Kinases/metabolism , Protein Transport/drug effects , Regeneration/drug effects , Serine/metabolism , Substrate Specificity/drug effects , Transcription Factors , Wnt Signaling Pathway/drug effects , YAP-Signaling Proteins , beta Catenin/metabolism
9.
Tumour Biol ; 37(9): 12169-12180, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27220495

ABSTRACT

Exosomes are nano-sized membrane vesicles secreted by both normal and cancer cells. Emerging evidence indicates that cancer cells derived exosomes contribute to cancer progression through the modulation of tumor microenvironment. However, the effects of exosomes derived from gastric cancer cells on macrophages are not well understood. In this study, we investigated the biological role of gastric cancer cells derived exosomes in the activation of macrophages. We demonstrated that gastric cancer cells derived exosomes activated macrophages to express increased levels of proinflammatory factors, which in turn promoted tumor cell proliferation and migration. In addition, gastric cancer cells derived exosomes remarkably upregulated the phosphorylation of NF-κB in macrophages. Inhibiting the activation of NF-κB reversed the upregulation of proinflammatory factors in macrophages and blocked their promoting effects on gastric cancer cells. Moreover, we found that gastric cancer cells derived exosomes could also activate macrophages from human peripheral blood monocytes through the activation of NF-κB. In conclusion, our results suggest that gastric cancer cells derived exosomes stimulate the activation of NF-κB pathway in macrophages to promote cancer progression, which provides a potential therapeutic approach for gastric cancer by interfering with the interaction between exosomes and macrophages in tumor microenvironment.


Subject(s)
Exosomes/physiology , Macrophages/physiology , NF-kappa B/physiology , Signal Transduction/physiology , Stomach Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Disease Progression , Humans , Neoplasm Invasiveness , Tumor Microenvironment
10.
Oncotarget ; 7(13): 16311-24, 2016 Mar 29.
Article in English | MEDLINE | ID: mdl-26918831

ABSTRACT

As a natural health supplement, 3,3'-diindolylmethane (DIM) is proposed as a preventive and chemotherapeutic agent for cancer by inhibiting cell proliferation and inducing cell apoptosis. However, we found that in contrary to high level of DIM (30 µM), low level of DIM (1 µM and 10 µM) obviously promoted gastric cancer cell growth and migration. In addition, we found that low level of DIM increased the expression of stemness factors and enhanced the pluripotency of gastric cancer cells. Low level of DIM promoted gastric cancer progression by inducing the PORCN-dependent secretion of Wnt4 and the activation of ß-catenin signaling. Wnt4 knockdown reversed the effects of low level of DIM on gastric cancer cells. The results of in vivo studies showed that gastric cancer cells treated with low level of DIM (1 µM) grew faster and expressed higher level of Wnt4 than control cells. Taken together, our findings indicate that low level of DIM activates autocrine Wnt4 signaling to enhance the progression of gastric cancer, which may suggest an adverse aspect of DIM in cancer therapy. Our findings will provide a new aspect for the safety of DIM in its clinical application.


Subject(s)
Antineoplastic Agents/adverse effects , Indoles/adverse effects , Neoplastic Stem Cells/drug effects , Stomach Neoplasms/pathology , Wnt4 Protein/metabolism , Animals , Carcinogenesis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Humans , Mice , Signal Transduction/drug effects , Wnt4 Protein/drug effects , Xenograft Model Antitumor Assays
11.
Stem Cells Int ; 2016: 1929536, 2016.
Article in English | MEDLINE | ID: mdl-28105054

ABSTRACT

Our and other groups have discovered that mesenchymal stem cells (MSCs) derived exosomes are a novel therapeutical modality for many diseases. In this study, we summarized a method to extract and purify hucMSCs-exosomes using ultrafiltration and gradient centrifugation in our laboratory and proved that hucMSCs-exosomes prepared according to our procedure were stable and bioactive. Results showed that exosomes derived from hucMSC were 40~100 nm and CD9 and CD81 positive. Functionally, hucMSCs-exosomes promoted cell proliferation and protected against oxidative stress-induced cell apoptosis in vitro by activation of ERK1/2 and p38. Interestingly, UV exposure abrogated the regulatory roles of exosomes under oxidative stress, indicating that hucMSCs-exosomes may regulate cell growth and apoptosis by exosomal shuttle of RNA. Furthermore, cytokine profile analysis revealed that hucMSCs-exosomes contained high dose of IL-6, IL-8, and other cytokines. The established method is practical and efficient, which provides a basis for further evaluating the potential of hucMSCs-exosomes as therapeutic agents.

12.
Stem Cells Transl Med ; 4(5): 513-22, 2015 May.
Article in English | MEDLINE | ID: mdl-25824139

ABSTRACT

Human umbilical cord mesenchymal stem cells (hucMSCs) and their exosomes have been considered as potential therapeutic tools for tissue regeneration; however, the underlying mechanisms are still not well understood. In this study, we isolated and characterized the exosomes from hucMSCs (hucMSC-Ex) and demonstrated that hucMSC-Ex promoted the proliferation, migration, and tube formation of endothelial cells in a dose-dependent manner. Furthermore, we demonstrated that hucMSC-Ex promoted wound healing and angiogenesis in vivo by using a rat skin burn model. We discovered that hucMSC-Ex promoted ß-catenin nuclear translocation and induced the increased expression of proliferating cell nuclear antigen, cyclin D3, N-cadherin, and ß-catenin and the decreased expression of E-cadherin. The activation of Wnt/ß-catenin is critical in the induction of angiogenesis by hucMSC-Ex, which could be reversed by ß-catenin inhibitor ICG-001. Wnt4 was delivered by hucMSC-Ex, and the knockdown of Wnt4 in hucMSC-Ex abrogated ß-catenin nuclear translocation in endothelial cells. The in vivo proangiogenic effects were also inhibited by interference of Wnt4 expression in hucMSC-Ex. Taken together, these results suggest that hucMSC-Ex-mediated Wnt4 induces ß-catenin activation in endothelial cells and exerts proangiogenic effects, which could be an important mechanism for cutaneous wound healing.


Subject(s)
Neovascularization, Physiologic , Regeneration , Wnt4 Protein/biosynthesis , Wound Healing , beta Catenin/biosynthesis , Animals , Bridged Bicyclo Compounds, Heterocyclic/administration & dosage , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/transplantation , Exosomes/metabolism , Exosomes/transplantation , Gene Expression Regulation/drug effects , Humans , Mesenchymal Stem Cells/cytology , Pyrimidinones/administration & dosage , Rats , Signal Transduction/drug effects , Umbilical Cord/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...