Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
Add more filters










Publication year range
1.
Cancer Res ; 81(10): 2745-2759, 2021 05 15.
Article in English | MEDLINE | ID: mdl-34003774

ABSTRACT

Porphyromonas gingivalis (P. gingivalis) is a keystone periodontal pathogen associated with various digestive cancers. However, whether P. gingivalis can promote colorectal cancer and the underlying mechanism associated with such promotion remains unclear. In this study, we found that P. gingivalis was enriched in human feces and tissue samples from patients with colorectal cancer compared with those from patients with colorectal adenoma or healthy subjects. Cohort studies demonstrated that P. gingivalis infection was associated with poor prognosis in colorectal cancer. P. gingivalis increased tumor counts and tumor volume in the ApcMin/+ mouse model and increased tumor growth in orthotopic rectal and subcutaneous carcinoma models. Furthermore, orthotopic tumors from mice exposed to P. gingivalis exhibited tumor-infiltrating myeloid cell recruitment and a proinflammatory signature. P. gingivalis promoted colorectal cancer via NLRP3 inflammasome activation in vitro and in vivo. NLRP3 chimeric mice harboring orthotopic tumors showed that the effect of NLRP3 on P. gingivalis pathogenesis was mediated by hematopoietic sources. Collectively, these data suggest that P. gingivalis contributes to colorectal cancer neoplasia progression by activating the hematopoietic NLRP3 inflammasome. SIGNIFICANCE: This study demonstrates that the periodontal pathogen P. gingivalis can promote colorectal tumorigenesis by recruiting myeloid cells and creating a proinflammatory tumor microenvironment. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/81/10/2745/F1.large.jpg.


Subject(s)
Carcinogenesis/pathology , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Inflammasomes/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/physiology , Neoplastic Stem Cells/pathology , Porphyromonas gingivalis/pathogenicity , Animals , Apoptosis , Bacteroidaceae Infections/complications , Bacteroidaceae Infections/immunology , Bacteroidaceae Infections/microbiology , Bacteroidaceae Infections/pathology , Carcinogenesis/immunology , Carcinogenesis/metabolism , Cell Proliferation , Colorectal Neoplasms/immunology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/microbiology , Humans , Inflammasomes/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Myeloid Cells/immunology , Myeloid Cells/metabolism , Myeloid Cells/microbiology , Myeloid Cells/pathology , Neoplastic Stem Cells/immunology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/microbiology , Prognosis , Survival Rate , Tumor Cells, Cultured , Tumor Microenvironment/immunology , Xenograft Model Antitumor Assays
2.
Gastric Cancer ; 24(1): 133-144, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32940810

ABSTRACT

BACKGROUND: The main cause of gastric cancer is the infection by the bacterium Helicobacter pylori which induces a chronic inflammation and an epithelial-to-mesenchymal transition (EMT) leading to the emergence of cells with cancer stem cell (CSC) properties. However, the underlying mechanisms have not been fully characterized. Moreover, H. pylori modulates the host cell autophagic process, but a few studies have investigated the role of this process in tumoral transformation. The aim of this study was to determine whether H. pylori-induced autophagy has a role in CSC emergence. METHODS: Autophagic flux in response to H. pylori infection was characterized in AGS cell line expressing the tandem-tagged mCherry-GFP-LC3 protein and using a ratiometric flow cytometry analysis. Then, AGS and MKN45 cell lines were treated with bafilomycin or chloroquine, two pharmaceutical well-known inhibitors of autophagy, and different EMT and CSC characteristics were analyzed. RESULTS: First, a co-expression of the gastric CSC marker CD44 and the autophagic marker LC3 in mice and human stomach tissues infected with H. pylori was observed. Then, we demonstrated in vitro that H. pylori was able to activate the autophagy process with a reduced autophagic flux. Finally, infected cells were treated with autophagy inhibitors, which reduced (i) appearance of mesenchymal phenotypes and migration ability related to EMT and (ii) CD44 expression as well as tumorsphere formation capacities reflecting CSC properties. CONCLUSION: In conclusion, all these data show that H. pylori-induced autophagy is implicated in gastric CSC emergence and could represent an interesting therapeutic target.


Subject(s)
Autophagy/physiology , Helicobacter Infections/microbiology , Helicobacter pylori/physiology , Neoplastic Stem Cells/microbiology , Stomach Neoplasms/microbiology , Animals , Cell Line, Tumor , Epithelial-Mesenchymal Transition , Helicobacter Infections/complications , Humans , Hyaluronan Receptors/metabolism , Mice , Microtubule-Associated Proteins/metabolism , Stomach/cytology , Stomach/microbiology
3.
Aging (Albany NY) ; 12(22): 23337-23350, 2020 11 14.
Article in English | MEDLINE | ID: mdl-33197886

ABSTRACT

Growing evidence suggests that microbes can influence the onset of cancer and its consequent development. By researching samples from patients afflicted by cervical cancer, we aimed to explore the associated dynamics and prognostic value of intratumoral levels of F. nucleatum. We used qPCR to analyze tumor tissues obtained from 112 cervical cancer patients in order to characterize the levels and influences of intratumoral levels of the F. nucleatum. Especially for recurrent tissues, there was a distinct observation of higher levels of F. nucleatum in cervical cancer. Patients with high burdens of F. nucleatum intratumoral infiltration exhibited correspondingly poor rates of both overall survival and progression-free survival. Measures of the levels of F. nucleatum were found to have been reliable independent prognostic factors that could predict rates of PFS for afflicted patients (HR = 4.8, 95%CI = 1.2-18.6, P = 0.024). Notably, the levels ofF. nucleatum were positively correlated with tumor differentiation. Cancer cells from patients with relatively high levels of F. nucleatum were observed to possess the characteristics of cancer stem cells (CSCs). We propose that F. nucleatum might be one potential cervical cancer diagnostic and prognostic biomarker, and these findings will help to provide a sound rationale and merit for further study of this bacterium.


Subject(s)
Fusobacterium nucleatum/isolation & purification , Uterine Cervical Neoplasms/diagnosis , Uterine Cervical Neoplasms/microbiology , Adult , Female , Humans , Neoplastic Stem Cells/microbiology , Prognosis , Survival Analysis , Uterine Cervical Neoplasms/mortality
4.
Sci Rep ; 10(1): 18024, 2020 10 22.
Article in English | MEDLINE | ID: mdl-33093503

ABSTRACT

Bacteria is recognized as opportunistic tumor inhabitant, giving rise to an environmental stress that may alter tumor microenvironment, which directs cancer behavior. In vitro infection of the T24 cell line with E. coli was performed to study the bacterial impact on bladder cancer cells. EMT markers were assessed using immunohistochemistry, western blot and RT-PCR. Stemness characteristics were monitored using RT-PCR. Furthermore, the metabolic reprograming was investigated by detection of ROS and metabolic markers. A significant (p ≤ 0.001) upregulation of vimentin as well as downregulation of CK19 transcription and protein levels was reported. A significant increase (p ≤ 0.001) in the expression level of stemness markers (CD44, NANOG, SOX2 and OCT4) was reported. ROS level was elevated, that led to a significant increase (p ≤ 0.001) in UCP2. This enhanced a significant increase (p ≤ 0.001) in PDK1 to significantly downregulate PDH (p ≤ 0.001) in order to block oxidative phosphorylation in favor of glycolysis. This resulted in a significant decrease (p ≤ 0.001) of AMPK, and a significant elevation (p ≤ 0.001) of MCT1 to export the produced lactate to extracellular matrix. Thus, bacteria may induce alteration to the heterogonous tumor cell population through EMT, CSCs and metabolic reprogramming, which may improve cancer cell ability to migrate and self-renew.


Subject(s)
Cellular Reprogramming , Escherichia coli Infections/complications , Escherichia coli/pathogenicity , Neoplastic Stem Cells/pathology , Urinary Bladder Neoplasms/pathology , Apoptosis , Cell Proliferation , Disease Progression , Epithelial-Mesenchymal Transition , Escherichia coli Infections/microbiology , Humans , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/microbiology , Tumor Cells, Cultured , Tumor Microenvironment , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/microbiology
5.
Gut Microbes ; 12(1): 1788900, 2020 11 09.
Article in English | MEDLINE | ID: mdl-32684087

ABSTRACT

The enrichment of Enterotoxigenic Bacteroides fragilis (ETBF) has been identified in CRC patients and associated with worse prognosis. Cancer stem cells (CSCs) play essential roles in CRC development. However, whether ETBF is involved in CSCs regulation is unknown. To clarify the role of ETBF in CSCs properties, we performed extreme limited dilution assays (ELDA) in nude mice injected with ETBF-treated or untreated CRC cells subcutaneously, tumor organoids culture in azoxymethane (AOM) mouse model after gavaging with or without ETBF, and cell sphere formation assay after incubating CRC cell lines with or without ETBF. The results indicated that ETBF increased the stemness of CRC cells in vivo and in vitro. Furthermore, ETBF enhanced the expression of core stemness transcription factors Nanog homeobox (NANOG) and sex determining region Y-box 2 (SOX2). Histone H3 Lysine 9 trimethylation (H3K9me3) is critical in regulating CSCs properties. As an epigenetic and transcriptional regulator, JmjC-domain containing histone demethylase 2B (JMJD2B) is essential for embryonic stem cell (ESC) transformation and H3K9me3 demethylation. Mechanistically, ETBF infection significantly upregulated JMJD2B levels in CRC cell lines and nude mice xenograft model. JMJD2B epigenetically upregulated NANOG expression via demethylating its promoter H3K9me3, to mediate ETBF-induced stemness of CRC cells. Subsequently, we found that the Toll-like receptor 4 (TLR4) pathway, activated by ETBF, contributed to the enhanced expression of JMJD2B via nuclear transcription factor nuclear factor of activated T cells 5 (NFAT5). Finally, in human CRC samples, the amount of ETBF positively correlated with nuclear NFAT5, JMJD2B, and NANOG expression levels. In summary, ETBF upregulated JMJD2B levels in a TLR4-NFAT5-dependent pathway, and played an important role in stemness regulation, which promoted colorectal carcinogenesis.


Subject(s)
Bacteroides fragilis/pathogenicity , Colorectal Neoplasms/microbiology , Colorectal Neoplasms/pathology , Jumonji Domain-Containing Histone Demethylases/metabolism , Animals , Bacteroides fragilis/metabolism , Carcinogenesis/metabolism , Carcinogenesis/pathology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Nanog Homeobox Protein/genetics , Nanog Homeobox Protein/metabolism , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/microbiology , Neoplastic Stem Cells/pathology , Prognosis , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Toll-Like Receptor 4/metabolism , Transcription Factors/metabolism
6.
Mol Carcinog ; 56(9): 2035-2047, 2017 09.
Article in English | MEDLINE | ID: mdl-28383782

ABSTRACT

Accumulating evidence suggests that human hepatocellular carcinoma (HCC) can be derived from cancer stem cells (CSCs), which contribute to tumor initiation, metastasis, chemoresistance, and recurrence. A great variety of HCC CSCs resulting in diverse clinical manifestations have been reported. However, how CSC diversity is regulated and generated remains unclear. Here we report that the miR-200b-ZEB1 circuit is closely involved with the induction and maintenance of a diverse group of CSCs. We found that miR-200b downregulation occurred in early HCC and associated with poor prognosis. The downregulation was attributable to genome deletion and promoter methylation of the miR-200a/b/429 gene. Ectopic expression of miR-200b or silencing of ZEB1 led to a decrease in CD13+ and CD24+ HCC CSCs and an increase in EpCAM+ HCC CSCs. Mechanistically, miR-200b directly suppressed BMI1 and ZEB1 expressions. ZEB1 recognized promoters of CD13, CD24, and EpCAM genes resulting in CD13 and CD24 upregulation and EpCAM downregulation. Neither miR-200b nor ZEB1 had obvious effects on CD133 or CD90 expression. Silencing CD13 or CD24 expression suppressed tumorigenicity of HCC cells. Ectopic expression of CD24 reversed the suppression of tumorigenicity by ectopic expression of miR-200b. Clinically, miR-200b downregulation was coupled with ZEB1 upregulation in approximately two-thirds of HCC patients. ZEB1 expression was positively correlated with CD13 and CD24 expressions in HCCs, while miR-200b expression was positively correlated with EpCAM. Our findings suggest that the miR-200b-ZEB1 circuit is a master regulator of diverse stemness of HCC, which differentiates HCCs into those containing CD13+ /CD24+ CSCs from those containing EpCAM+ CSCs.


Subject(s)
Carcinoma, Hepatocellular/metabolism , MicroRNAs/metabolism , Neoplastic Stem Cells/metabolism , Zinc Finger E-box-Binding Homeobox 1/metabolism , Antigens, Differentiation/metabolism , CD13 Antigens/metabolism , CD24 Antigen/metabolism , Carcinoma, Hepatocellular/genetics , Cell Differentiation , Cell Line, Tumor , Cell Transformation, Neoplastic , DNA Methylation , DNA, Neoplasm/metabolism , Down-Regulation , Gene Deletion , Gene Expression Regulation, Neoplastic , Humans , Neoplastic Stem Cells/microbiology , Promoter Regions, Genetic
7.
Cell Death Dis ; 8(3): e2667, 2017 03 16.
Article in English | MEDLINE | ID: mdl-28300841

ABSTRACT

Commensal Escherichia coli has been identified as a major protagonist of microbe-induced colorectal oncogenesis. Its tumour-promoting attribute is linked to the expression of DNA-damaging genotoxins. Using a constitutively invasive variant of non-pathogenic E. coli, we demonstrate that chronic presence of internalized E. coli leads to enhanced oncogenicity in colon cancer cells. Instead of genomic damage, the tumorigenic effect is mediated through an expansion of the cancer stem cell (CSC) population, likely through dedifferentiation of lineage-committed intestinal epithelial cells. Stemness-linked intestinal tumorigenicity is directly correlated to absence of microbial virulence factor expression and is specific for intestinal cells. The enriched CSC fraction remains stable in the absence of the instigating bacteria and can foster stemness traits in unexposed cells through secreted factors. Mechanistically, aberrant host invasion leads to realignment of multiple host signal transduction cascades, notably mutually re-enforcing NF-κB and ß-catenin activation, through reciprocal modulation of microbe sensing pathways Nod1/Rip2 and TLR/MyD88. The expanded tumorigenic CSC population is marked by enhanced malignancy traits, long-term self-renewal capacity and robust tumorigenic ability, both in vitro and in vivo. Our study shows that microbe-induced oncogenicity is not a strict correlate of commensal virulence and can be invoked by even non-pathogenic E. coli by engendering tumorigenic stemness in host cells.


Subject(s)
Carcinogenesis/metabolism , Escherichia coli/pathogenicity , Intestines/microbiology , Intestines/pathology , Neoplastic Stem Cells/microbiology , Neoplastic Stem Cells/pathology , Animals , Caco-2 Cells , Carcinogenesis/pathology , Cell Line, Tumor , Colon/metabolism , Colon/microbiology , Colon/pathology , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Epithelial Cells/pathology , HCT116 Cells , HT29 Cells , Hep G2 Cells , Humans , Intestinal Mucosa/metabolism , Male , Mice , Mice, Nude , Myeloid Differentiation Factor 88/metabolism , NF-kappa B/metabolism , Neoplastic Stem Cells/metabolism , Nod1 Signaling Adaptor Protein/metabolism , Receptor-Interacting Protein Serine-Threonine Kinase 2/metabolism , Signal Transduction/physiology , beta Catenin/metabolism
8.
Free Radic Biol Med ; 105: 3-15, 2017 04.
Article in English | MEDLINE | ID: mdl-27810411

ABSTRACT

Colorectal cancer (CRC) is a leading cause of cancer death and archetype for cancer as a genetic disease. However, the mechanisms for genetic change and their interactions with environmental risk factors have been difficult to unravel. New hypotheses, models, and methods are being used to investigate a complex web of risk factors that includes the intestinal microbiome. Recent research has clarified how the microbiome can generate genomic change in CRC. Several phenotypes among a small group of selected commensals have helped us better understand how mutations and chromosomal instability (CIN) are induced in CRC (e.g., toxin production, metabolite formation, radical generation, and immune modulation leading to a bystander effect). This review discusses recent hypotheses, models, and mechanisms by which the intestinal microbiome contributes to the initiation and progression of sporadic and colitis-associated forms of CRC. Overall, it appears the microbiome can initiate and/or promote CRC at all stages of tumorigenesis by acting as an inducer of DNA damage and CIN, regulating cell growth and death, generating epigenetic changes, and modulating host immune responses. Understanding how the microbiome interacts with other risk factors to define colorectal carcinogenesis will ultimately lead to more accurate risk prediction. A deeper understanding of CRC etiology will also help identify new targets for prevention and treatment and help accelerate the decline in mortality for this common cancer.


Subject(s)
Colorectal Neoplasms/microbiology , Gastrointestinal Microbiome/immunology , Animals , Cell Transformation, Neoplastic/immunology , Chromosomal Instability , Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , DNA Damage , Enterobacteriaceae/immunology , Enterobacteriaceae/physiology , Epithelial-Mesenchymal Transition , Humans , Neoplastic Stem Cells/microbiology , Risk Factors
9.
Oncotarget ; 7(49): 80688-80699, 2016 12 06.
Article in English | MEDLINE | ID: mdl-27729612

ABSTRACT

Helicobacter pylori infection is responsible for gastric carcinogenesis but host factors are also implicated. IQGAP1, a scaffolding protein of the adherens junctions interacting with E-cadherin, regulates cellular plasticity and proliferation. In mice, IQGAP1 deficiency leads to gastric hyperplasia. The aim of this study was to elucidate the consequences of IQGAP1 deletion on H. pylori-induced gastric carcinogenesis.Transgenic mice deleted for iqgap1 and WT littermates were infected with Helicobacter sp., and histopathological analyses of the gastric mucosa were performed. IQGAP1 and E-cadherin expression was evaluated in gastric tissues and in gastric epithelial cell lines in response to H. pylori infection. The consequences of IQGAP1 deletion on gastric epithelial cell behaviour and on the acquisition of cancer stem cell (CSC)-like properties were evaluated. After one year of infection, iqgap1+/- mice developed more preneoplastic lesions and up to 8 times more gastro-intestinal neoplasia (GIN) than WT littermates. H. pylori infection induced IQGAP1 and E-cadherin delocalization from cell-cell junctions. In vitro, knock-down of IQGAP1 favoured the acquisition of a mesenchymal phenotype and CSC-like properties induced by H. pylori infection.Our results indicate that alterations in IQGAP1 signalling promote the emergence of CSCs and gastric adenocarcinoma development in the context of an H. pylori infection.


Subject(s)
Adenocarcinoma/microbiology , Gastric Mucosa/microbiology , Helicobacter Infections/microbiology , Helicobacter pylori/pathogenicity , Neoplastic Stem Cells/microbiology , Precancerous Conditions/microbiology , Stomach Neoplasms/microbiology , ras GTPase-Activating Proteins/deficiency , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Cadherins/metabolism , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Disease Models, Animal , Epithelial-Mesenchymal Transition , Female , Gastric Mucosa/metabolism , Gastric Mucosa/pathology , Genetic Predisposition to Disease , Helicobacter Infections/genetics , Helicobacter Infections/metabolism , Helicobacter Infections/pathology , Host-Pathogen Interactions , Hyaluronan Receptors/metabolism , Hyperplasia , Mice, 129 Strain , Mice, Knockout , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Phenotype , Precancerous Conditions/genetics , Precancerous Conditions/metabolism , Precancerous Conditions/pathology , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Time Factors , ras GTPase-Activating Proteins/genetics
10.
Cancer Lett ; 374(2): 292-303, 2016 May 01.
Article in English | MEDLINE | ID: mdl-26940070

ABSTRACT

Helicobacter pylori (H. pylori) infection is considered a major risk factor for gastric cancer. CagA behaves as a major bacterial oncoprotein playing a key role in H. pylori-induced tumorigenesis. Cancer stem cells (CSCs) are believed to possess the ability to initiate tumorigenesis and promote progression. Although studies have suggested that cancer cells can exhibit CSC-like properties in the tumor microenvironment, it remains unclear whether H. pylori infection could induce the emergence of CSC-like properties in gastric cancer cells and, the underlying mechanism. Here, gastric cancer cells were co-cultured with a CagA-positive H. pylori strain or a CagA isogenic mutant strain. We found that H. pylori-infected gastric cancer cells exhibited CSC-like properties, including an increased expression of CSC specific surface markers CD44 and Lgr5, as well as that of Nanog, Oct4 and c-myc, which are known pluripotency genes, and an increased capacity for self-renewal, whereas these properties were not observed in the CagA isogenic mutant strain-infected cells. Further studies revealed that H. pylori activated Wnt/ß-catenin signaling pathway in a CagA-dependent manner and that the activation of this pathway was dependent upon CagA-positive H. pylori-mediated phosphorylation of ß-catenin at the C-terminal Ser675 and Ser552 residues in a c-met- and/or Akt-dependent manner. We further demonstrated that this activation was responsible for H. pylori-induced CSC-like properties. Moreover, we found the promoter activity of Nanog and Oct4 were upregulated, and ß-catenin was observed to bind to these promoters during H. pylori infection, while a Wnt/ß-catenin inhibitor suppressed promoter activity and binding. Taken together, these results suggest that H. pylori upregulates Nanog and Oct4 via Wnt/ß-catenin signaling pathway to promote CSC-like properties in gastric cancer cells.


Subject(s)
Helicobacter Infections/metabolism , Helicobacter pylori/metabolism , Homeodomain Proteins/metabolism , Neoplastic Stem Cells/microbiology , Octamer Transcription Factor-3/metabolism , Stomach Neoplasms/microbiology , Wnt Signaling Pathway , Antigens, Bacterial/metabolism , Bacterial Proteins/metabolism , Cell Line, Tumor , Cell Nucleus/genetics , Cell Nucleus/metabolism , Helicobacter Infections/genetics , Helicobacter Infections/pathology , Humans , Nanog Homeobox Protein , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Promoter Regions, Genetic , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Transcriptional Activation , Up-Regulation , beta Catenin/metabolism
11.
Stem Cell Rev Rep ; 12(1): 156-61, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26514153

ABSTRACT

Cancer cell lines can be useful to model cancer stem cells. Infection with Mycoplasma species is an insidious problem in mammalian cell culture. While investigating stem-like properties in early passage melanoma cell lines, we noted poorly reproducible results from an aliquot of a cell line that was later found to be infected with Mycoplasma hyorhinis. Deliberate infection of other early passage melanoma cell lines aliquots induced variable and unpredictable effects on expression of putative cancer stem cell markers, clonogenicity, proliferation and global gene expression. Cell lines established in stem cell media (SCM) were equally susceptible. Mycoplasma status is rarely reported in publications using cultured cells to study the cancer stem cell hypothesis. Our work highlights the importance of surveillance for Mycoplasma infection while using any cultured cells to interrogate tumor heterogeneity.


Subject(s)
Gene Expression Regulation, Neoplastic , Mycoplasma hyorhinis/isolation & purification , Neoplasm Proteins/genetics , Neoplastic Stem Cells/microbiology , RNA, Ribosomal, 16S/isolation & purification , Bacterial Typing Techniques , Cell Culture Techniques/standards , Cell Line, Tumor , Host-Pathogen Interactions , Humans , Melanoma/genetics , Melanoma/metabolism , Melanoma/microbiology , Melanoma/pathology , Mycoplasma hyorhinis/genetics , Neoplasm Proteins/metabolism , Neoplastic Stem Cells/metabolism , Polymerase Chain Reaction , Primary Cell Culture , Quality Control , RNA, Ribosomal, 16S/genetics , Reproducibility of Results , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Skin Neoplasms/microbiology , Skin Neoplasms/pathology
12.
Int J Mol Sci ; 16(8): 19153-69, 2015 Aug 14.
Article in English | MEDLINE | ID: mdl-26287172

ABSTRACT

Gastric cancer is still a leading cause of cancer-related mortality worldwide in spite of declining incidence. Gastric cancers are, essentially, adenocarcinomas and one of the strongest risk factors is still infection with Helicobacter pylori. Within the last years, it became clear that gastric self-renewal and carcinogenesis are intimately linked, particularly during chronic inflammatory conditions. Generally, gastric cancer is now regarded as a disease resulting from dysregulated differentiation of stem and progenitor cells, mainly due to an inflammatory environment. However, the situation in the stomach is rather complex, consisting of two types of gastric units which show bidirectional self-renewal from an unexpectedly large variety of progenitor/stem cell populations. As in many other tumors, cancer stem cells have also been characterized for gastric cancer. This review focuses on the various gastric epithelial stem cells, how they contribute to self-renewal and which routes are known to gastric adenocarcinomas, including their stem cells.


Subject(s)
Adenocarcinoma/pathology , Gastric Mucosa/pathology , Neoplastic Stem Cells/pathology , Stomach Neoplasms/pathology , Stomach/pathology , Adenocarcinoma/microbiology , Animals , Gastric Mucosa/microbiology , Helicobacter Infections/complications , Helicobacter Infections/microbiology , Helicobacter Infections/pathology , Helicobacter pylori/isolation & purification , Humans , Neoplastic Stem Cells/microbiology , Stomach/microbiology , Stomach Neoplasms/microbiology
13.
Tumour Biol ; 36(12): 9947-60, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26178482

ABSTRACT

Periodontitis is the most common chronic inflammatory condition occurring in the human oral cavity, but our knowledge on its contribution to oral cancer is rather limited. To define crosstalk between chronic periodontitis and oral cancer, we investigated whether Porphyromonas gingivalis, a major pathogen of chronic periodontitis, plays a role in oral cancer progression. To mimic chronic irritation by P. gingivalis in the oral cavity, oral squamous cell carcinoma (OSCC) cells were infected with P. gingivalis twice a week for 5 weeks. Repeated infection of oral cancer cells by P. gingivalis resulted in morphological changes of host cancer cells into an elongated shape, along with the decreased expression of epithelial cell markers, suggesting acquisition of an epithelial-to-mesenchymal transition (EMT) phenotype. The prolonged exposure to P. gingivalis also promoted migratory and invasive properties of OSCC cells and provided resistance against a chemotherapeutic agent, all of which are described as cellular characteristics undergoing EMT. Importantly, long-term infection by P. gingivalis induced an increase in the expression level of CD44 and CD133, well-known cancer stem cell markers, and promoted the tumorigenic properties of infected cancer cells compared to non-infected controls. Furthermore, increased invasiveness of P. gingivalis-infected OSCC cells was correlated with enhanced production of matrix metalloproteinase (MMP)-1 and MMP-10 that was stimulated by interleukin-8 (IL-8) release. This is the first report demonstrating that P. gingivalis can increase the aggressiveness of oral cancer cells via epithelial-mesenchymal transition-like changes and the acquisition of stemness, implicating P. gingivalis as a potential bacterial risk modifier.


Subject(s)
Mouth Neoplasms/pathology , Neoplastic Stem Cells/pathology , Periodontitis/pathology , Porphyromonas gingivalis/pathogenicity , Carcinoma, Squamous Cell , Cell Line, Tumor , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Humans , Interleukin-8/metabolism , Matrix Metalloproteinase 1/biosynthesis , Mouth Neoplasms/complications , Mouth Neoplasms/microbiology , Neoplastic Stem Cells/microbiology , Periodontitis/complications , Periodontitis/microbiology
14.
World J Gastroenterol ; 21(1): 112-23, 2015 Jan 07.
Article in English | MEDLINE | ID: mdl-25574084

ABSTRACT

Gastric cancer (GC) is one of the leading causes of cancer-related mortality worldwide. Cancer stem cells (CSCs), which were first identified in acute myeloid leukemia and subsequently in a large array of solid tumors, play important roles in cancer initiation, dissemination and recurrence. CSCs are often transformed tissue-specific stem cells or de-differentiated transit amplifying progenitor cells. Several populations of multipotent gastric stem cells (GSCs) that reside in the stomach have been determined to regulate physiological tissue renewal and injury repair. These populations include the Villin+ and Lgr5+ GSCs in the antrum, the Troy+ chief cells in the corpus, and the Sox2+ GSCs that are found in both the antrum and the corpus. The disruption of tumor suppressors in Villin+ or Lgr5+ GSCs leads to GC in mouse models. In addition to residing GSCs, bone marrow-derived cells can initiate GC in a mouse model of chronic Helicobacter infection. Furthermore, expression of the cell surface markers CD133 or CD44 defines gastric CSCs in mouse models and in human primary GC tissues and cell lines. Targeted elimination of CSCs effectively reduces tumor size and grade in mouse models. In summary, the recent identification of normal GSCs and gastric CSCs has greatly improved our understanding of the molecular and cellular etiology of GC and will aid in the development of effective therapies to treat patients.


Subject(s)
Multipotent Stem Cells/pathology , Neoplastic Stem Cells/pathology , Stomach Neoplasms/pathology , Animals , Antigens, CD , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cadherins/genetics , Cadherins/metabolism , Cell Differentiation , Cell Proliferation , Cell Survival , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Epithelial-Mesenchymal Transition , Helicobacter Infections/complications , Helicobacter Infections/microbiology , Helicobacter pylori/pathogenicity , Humans , Multipotent Stem Cells/metabolism , Multipotent Stem Cells/microbiology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/microbiology , Phenotype , Risk Factors , Signal Transduction , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Stomach Neoplasms/microbiology , Stomach Neoplasms/therapy
15.
Methods Mol Biol ; 946: 181-204, 2013.
Article in English | MEDLINE | ID: mdl-23179833

ABSTRACT

The cancer stem cell hypothesis is an appealing concept to account for intratumoral heterogeneity and the observation that systemic metastasis and treatment failure are often associated with the survival of a small number of cancer cells. Whilst in vivo evidence forms the foundation of this concept, in vitro methods and reagents are attractive as they offer opportunities to perform experiments that are not possible in an animal model. While there is abundant evidence that existing cancer cell lines are not reliable models of tumor heterogeneity, recent advances based on well validated novel cancer cell lines established de novo in defined serum-free media are encouraging, particularly in the study of glioblastoma multiforme. In this chapter we wish to broadly outline the process of establishing, characterizing, and managing novel cancer cell lines in defined serum-free media, and discuss the limitations and potential opportunities that may arise from these model systems.


Subject(s)
Cell Separation/methods , Neoplastic Stem Cells/pathology , Cell Culture Techniques , Cell Line, Tumor , Humans , Mycoplasma/isolation & purification , Neoplastic Stem Cells/microbiology , Specimen Handling
16.
Exp Biol Med (Maywood) ; 237(10): 1189-96, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23045719

ABSTRACT

Systemic administration of Salmonella to tumor-bearing mice leads to the preferential accumulation within tumor sites and retardation of tumor growth. The cancer stem-like cell (CSC) hypothesis suggests that CSCs are the root of cancer and induce metastasis and recurrence. The objective of this study was to examine if Salmonella could inhibit the growth of CSCs derived from mouse breast cancer. Systemically injected Salmonella preferentially accumulated within tumors for at least three weeks and the bacteria accumulated preferentially not only in subcutaneous but also in orthotopic tumors over livers and spleens at ratios ranging from 1000:1 to 10,000:1. Salmonella were capable of delaying tumor growth and enhancing survival in both subcutaneous and orthotopic tumor models. More strikingly, Salmonella acted to retard tumor growth and extensively prolong the survival time of the mice bearing CSC-induced tumors. Our results also found that Salmonella predominantly, although not exclusively, resided in the CSC regions of the tumor. These data suggest that Salmonella can inhibit the growth of breast cancer by targeting the CSC niche. In conclusion, Salmonella can be used for the management of breast cancer.


Subject(s)
Breast Neoplasms/pathology , Neoplastic Stem Cells/microbiology , Neoplastic Stem Cells/pathology , Salmonella , Animals , Cell Line, Tumor , Female , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Stem Cell Niche , Transplantation, Heterologous
18.
J Theor Biol ; 264(2): 570-7, 2010 May 21.
Article in English | MEDLINE | ID: mdl-20219476

ABSTRACT

Epigenetic changes correspond to heritable modifications of the chromatin structure, which do not involve any alteration of the DNA sequence but nonetheless affect gene expression. These mechanisms play an important role in cell differentiation, but aberrant occurrences are also associated with a number of diseases, including cancer and neural development disorders. In particular, aberrant DNA methylation induced by H. Pylori has been found to be a significant risk factor in gastric cancer. To investigate the sensitivity of different genes and cell types to this infection, a computational model of methylation in gastric crypts is developed. In this article, we review existing results from physical experiments and outline their limitations, before presenting the computational model and investigating the influence of its parameters.


Subject(s)
DNA Methylation , Helicobacter Infections/complications , Models, Genetic , Stomach Neoplasms/genetics , Computer Simulation , Gastric Mucosa/metabolism , Gastric Mucosa/microbiology , Gastric Mucosa/pathology , Helicobacter pylori/physiology , Host-Pathogen Interactions , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/microbiology , Stomach Neoplasms/complications , Stomach Neoplasms/pathology
19.
Cancer Lett ; 278(1): 3-8, 2009 Jun 08.
Article in English | MEDLINE | ID: mdl-18922632

ABSTRACT

Until the last century, infectious diseases were the leading cause of human mortality. Therefore, our current medical reasoning is profoundly influenced by views that originated from medical microbiology. The notion that cancer growth is sustained by a sub-population of particular cells, the cancer stem cells, is highly reminiscent of the germ theory of disease as exemplified by Koch's postulates in the XIXth century. However, accumulating data underscore the importance of cell-cell interactions and tumor environment. Hence it is essential to critically review the basic tenets of the cancer stem cell concept on the light of their relationships with Koch's postulates. Shifting the pathogenic element from a special cellular entity (cancer stem cell or microorganism) to a "pathogenic field" could be critical for curing both cancer and drug-resistant infectious diseases.


Subject(s)
Neoplasms/pathology , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/pathology , Cell Differentiation , Cell Division , Humans , Infections/complications , Models, Biological , Neoplasms/surgery , Neoplasms/therapy , Neoplastic Stem Cells/microbiology
20.
Cancer Cell ; 14(1): 7-9, 2008 Jul 08.
Article in English | MEDLINE | ID: mdl-18598939

ABSTRACT

The etiology of gastrointestinal tumors implicates a role for chronic inflammation in response to pathogenic microflora as a promoting force for full neoplastic progression. Recently, Oguma and coworkers (2008) demonstrated that TNFalpha, derived from recruited macrophages, potentiates Wnt/beta-catenin signaling and gastric carcinogenesis by activating Akt signaling and GSK3beta phosphorylation independent of the NF-kappaB pathway in initiated epithelial cells. These observations provide a missing link in the mechanism whereby chronic inflammation, in response to Helicobacter, regulates the "penetrance" of initiating oncogenic mutations in the gastrointestinal tract leading to gastrointestinal tumorigenesis.


Subject(s)
Gastric Mucosa/immunology , Gastritis/immunology , Helicobacter Infections/immunology , Stomach Neoplasms/immunology , Animals , Cell Transformation, Neoplastic/immunology , Gastric Mucosa/microbiology , Gastritis/complications , Gastritis/genetics , Gastritis/microbiology , Gene Expression Regulation, Neoplastic , Genes, APC , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Helicobacter Infections/complications , Helicobacter Infections/microbiology , Humans , Macrophage Activation , Macrophages/immunology , Mutation , Neoplastic Stem Cells/immunology , Neoplastic Stem Cells/microbiology , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Stomach Neoplasms/genetics , Stomach Neoplasms/microbiology , Tumor Necrosis Factor-alpha/metabolism , Wnt Proteins/metabolism , beta Catenin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...