Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Blood Adv ; 7(18): 5382-5395, 2023 09 26.
Article in English | MEDLINE | ID: mdl-37505194

ABSTRACT

Acute myeloid leukemia (AML) is initiated and propagated by leukemia stem cells (LSCs), a self-renewing population of leukemia cells responsible for therapy resistance. Hence, there is an urgent need to identify new therapeutic opportunities targeting LSCs. Here, we performed an in vivo CRISPR knockout screen to identify potential therapeutic targets by interrogating cell surface dependencies of LSCs. The facilitated glucose transporter type 1 (GLUT1) emerged as a critical in vivo metabolic dependency for LSCs in a murine MLL::AF9-driven model of AML. GLUT1 disruption by genetic ablation or pharmacological inhibition led to suppression of leukemia progression and improved survival of mice that received transplantation with LSCs. Metabolic profiling revealed that Glut1 inhibition suppressed glycolysis, decreased levels of tricarboxylic acid cycle intermediates and increased the levels of amino acids. This metabolic reprogramming was accompanied by an increase in autophagic activity and apoptosis. Moreover, Glut1 disruption caused transcriptional, morphological, and immunophenotypic changes, consistent with differentiation of AML cells. Notably, dual inhibition of GLUT1 and oxidative phosphorylation (OXPHOS) exhibited synergistic antileukemic effects in the majority of tested primary AML patient samples through restraining of their metabolic plasticity. In particular, RUNX1-mutated primary leukemia cells displayed striking sensitivity to the combination treatment compared with normal CD34+ bone marrow and cord blood cells. Collectively, our study reveals a GLUT1 dependency of murine LSCs in the bone marrow microenvironment and demonstrates that dual inhibition of GLUT1 and OXPHOS is a promising therapeutic approach for AML.


Subject(s)
Leukemia, Myeloid, Acute , Oxidative Phosphorylation , Animals , Mice , Apoptosis , Bone Marrow/metabolism , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/metabolism , Leukemia, Myeloid, Acute/genetics , Tumor Microenvironment
2.
Anticancer Res ; 39(4): 1719-1728, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30952711

ABSTRACT

BACKGROUND: The wingless-type mammary tumour virus integration site 5A (WNT5A) agonist Foxy5 was shown in vitro to affect intracellular signalling implicated in the regulation of colonic cancer stem cells (CSCs). MATERIALS AND METHODS: In order to study whether Foxy5 can modulate CSCs, either HT-29 or Caco-2 human colonic cancer cells, both lacking endogenous WNT5A expression, were inoculated subcutaneously into nude mice. RESULTS: Foxy5 reduced the expression of the stem-cell marker aldehyde dehydrogenase and, interestingly, the specific colon CSC marker double cortin-like kinase 1. Foxy5 also reduced active ß-catenin and the expression of its downstream target Achaete Scute complex homolog 2, a CSC-preserving transcription factor. Foxy5 also reduced cyclo-oxygenase 2 expression, responsible for the formation of the CSC-promoting prostaglandin E2 (PGE2), but increased that of 15-hydroxyprostaglandin dehydrogenase expression, a PGE2-degrading enzyme. Accordingly, Foxy5 impairs both ß-catenin and PGE2 signalling, both of which have been implicated in promoting the niche of colonic CSCs. CONCLUSION: Our data suggest that Foxy5 can complement the traditional adjuvant chemotherapeutic treatment to which CSCs are resistant.


Subject(s)
Antineoplastic Agents/pharmacology , Colonic Neoplasms/drug therapy , Neoplastic Stem Cells/drug effects , Oligopeptides/pharmacology , Wnt-5a Protein/agonists , Aldehyde Dehydrogenase/genetics , Aldehyde Dehydrogenase/metabolism , Aldehyde Dehydrogenase 1 Family , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Caco-2 Cells , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , Doublecortin-Like Kinases , Female , HT29 Cells , Humans , Hydroxyprostaglandin Dehydrogenases/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Mice, Inbred BALB C , Mice, Nude , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Retinal Dehydrogenase , Tumor Burden/drug effects , Tumor Microenvironment , Wnt Signaling Pathway/drug effects , Wnt-5a Protein/metabolism , Xenograft Model Antitumor Assays , beta Catenin/metabolism
3.
Neoplasia ; 21(3): 269-281, 2019 03.
Article in English | MEDLINE | ID: mdl-30738331

ABSTRACT

Dysfunctional inflammatory pathways are associated with an increased risk of cancer, including colorectal cancer. We have previously identified and enriched for a self-renewing, colon cancer stem cell (CCSC) subpopulation in primary sporadic colorectal cancers (CRC) and a related subpopulation in ulcerative colitis (UC) patients defined by the stem cell marker, aldehyde dehydrogenase (ALDH). Subsequent work demonstrated that CCSC-initiated tumors are dependent on the inflammatory chemokine, CXCL8, a known inducer of tumor proliferation, angiogenesis and invasion. Here, we use RNA interference to target CXCL8 and its receptor, CXCR1, to establish the existence of a functional signaling pathway promoting tumor growth initiated by sporadic and colitis CCSCs. Knocking down either CXCL8 or CXCR1 had a dramatic effect on inhibiting both in vitro proliferation and angiogenesis. Likewise, tumorigenicity was significantly inhibited due to reduced levels of proliferation and angiogenesis. Decreased expression of cycle cell regulators cyclins D1 and B1 along with increased p21 levels suggested that the reduction in tumor growth is due to dysregulation of cell cycle progression. Therapeutically targeting the CXCL8-CXCR1 signaling pathway has the potential to block sustained tumorigenesis by inhibiting both CCSC- and pCCSC-induced proliferation and angiogenesis.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Colonic Neoplasms/etiology , Colonic Neoplasms/metabolism , Inflammation/metabolism , Interleukin-8/metabolism , Neoplastic Stem Cells/metabolism , Receptors, Interleukin-8A/metabolism , Signal Transduction , Animals , Biomarkers , Cell Line, Tumor , Cell Proliferation , Colitis/complications , Colitis/genetics , Colitis/metabolism , Colonic Neoplasms/pathology , Disease Models, Animal , Gene Dosage , Gene Expression , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Heterografts , Humans , Immunophenotyping , Inflammation/complications , Inflammation/genetics , Interleukin-8/genetics , Mice , Models, Biological , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Receptors, Interleukin-8A/genetics
4.
Cancer Lett ; 437: 13-24, 2018 11 28.
Article in English | MEDLINE | ID: mdl-30144515

ABSTRACT

Inflammation is implicated in the etiology of sporadic colon cancer (CC), which is one of the leading causes of cancer-related deaths worldwide. Here, we report that inhibition of the inflammatory receptor CysLT1 through its antagonist, montelukast, is beneficial in minimizing stemness in CC and thereby minimizing tumor growth in a mouse xenograft model of human colon cancer. Upon treatment with montelukast, colonospheres derived from HT-29 and SW-480 human colon cancer cells exhibited a significant phenotypic change coupled with the downregulation of mRNA and protein expression of cancer stem cell (CSC) markers ALDH1 and DCLK1. Moreover, montelukast reduced the size of HT-29 cell-derived tumors in mice. The reduction in tumor size was associated with decreased levels of ALDH1A1, DCLK1, BCL2 mRNA and macrophage infiltration into the tumor tissue. Interestingly, this treatment elevated levels of the tumor suppressor 15-PGDH while reducing COX-2 expression. Our data highlight the association of CysLT1R with CSCs and demonstrate that inhibition of CysLT1R could prove beneficial in minimizing CSC-induced tumor growth. This work advances the notion that targeting CSCs is a promising approach to improve outcomes in those afflicted with colon cancer.


Subject(s)
Acetates/pharmacology , Colonic Neoplasms/drug therapy , Neoplastic Stem Cells/drug effects , Quinolines/pharmacology , Receptors, Leukotriene/metabolism , Tumor Burden/drug effects , Xenograft Model Antitumor Assays/methods , Animals , Cell Line, Tumor , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Cyclopropanes , Female , Gene Expression Regulation, Neoplastic/drug effects , HT29 Cells , Humans , Leukotriene Antagonists/pharmacology , Mice, Inbred BALB C , Mice, Nude , Neoplastic Stem Cells/metabolism , Receptors, Leukotriene/genetics , Sulfides
5.
Oncotarget ; 8(21): 34773-34786, 2017 May 23.
Article in English | MEDLINE | ID: mdl-28410235

ABSTRACT

Cysteinyl leukotriene receptor 1 (CysLT1R) has been shown to be up-regulated in the adenocarcinomas of colorectal cancer patients, which is associated with a poor prognosis. In a spontaneous model of colon cancer, CysLT1R disruption was associated with a reduced tumor burden in double-mutant female mice (ApcMin/+/Cysltr1-/-) compared to ApcMin/+ littermates. In the current study, we utilized a genetic approach to investigate the effect of CysLT1R in the induced azoxymethane/dextran sulfate sodium (AOM/DSS) model of colitis-associated colon cancer. We found that AOM/DSS female mice with a global disruption of the Cysltr1 gene (Cysltr1-/-) had a higher relative body weight, a more normal weight/length colon ratio and smaller-sized colonic polyps compared to AOM/DSS wild-type counterparts. The Cysltr1-/- colonic polyps exhibited low-grade dysplasia, while wild-type polyps had an adenoma-like phenotype. The Cysltr1-/- colonic polyps exhibited significant decreases in nuclear ß-catenin and COX-2 protein expression, while the normal crypts surrounding the polyps exhibited increased Mucin 2 expression. Furthermore, Cysltr1-/- mice exhibited an overall reduction in inflammation, with a significant decrease in proinflammatory cytokines, polyp 5-LOX expression and infiltration of CD45 leukocytes and F4/80 macrophages. In conclusion, the present genetic approach in an AOM/DSS model further supports an important role for CysLT1R in colon tumorigenesis.


Subject(s)
Colitis/chemically induced , Colonic Neoplasms/chemically induced , Colonic Neoplasms/pathology , Receptors, Leukotriene/genetics , Animals , Azoxymethane , Body Weight , Colitis/complications , Colitis/genetics , Colonic Neoplasms/genetics , Cyclooxygenase 2/metabolism , Dextran Sulfate , Disease Models, Animal , Mice , beta Catenin/metabolism
6.
BMC Cancer ; 16: 425, 2016 07 07.
Article in English | MEDLINE | ID: mdl-27388564

ABSTRACT

BACKGROUND: Colorectal cancer is one of the most common types of cancers worldwide. Recent studies have identified cancer-initiating cells (CICs) as a subgroup of replication-competent cells in the development of colorectal cancer. Although it is understood that an inflammation-rich tumor microenvironment presumably supports CIC functions, the contributory factors are not very well defined. The present study advances our understanding of the role of the eicosanoids leukotriene D4 (LTD4) and prostaglandin E2 (PGE2) in the tumorigenic ability of CICs and investigates the consequential changes occurring in the tumor environment that might support tumor growth. METHODS: In this study we used human HCT-116 colon cancer ALDH(+) cells in a nude mouse xenograft model. Protein expression and immune cell was determined in tumor-dispersed cells by flow cytometry and in tumor sections by immunohistochemistry. mRNA expressions were quantified using RT-q-PCR and plasma cytokine levels by Multiplex ELISA. RESULTS: We observed that LTD4 and PGE2 treatment augmented CIC-induced tumor growth. LTD4-and PGE2-treated xenograft tumors revealed a robust increase in ALDH and Dclk1 protein expression, coupled with activated ß-catenin signaling and COX-2 up-regulation. Furthermore, LTD4 or PGE2 accentuated the accumulation of CD45 expressing cells within xenograft tumors. Further analysis revealed that these infiltrating immune cells consisted of neutrophils (LY6G) and M2 type macrophages (CD206(+)). In addition, LTD4 and PGE2 treatment significantly elevated the plasma levels of cysteinyl leukotrienes and PGE2, as well as levels of IL-1ß, IL-2, IL-6, TNF-α and CXCL1/KC/GRO. In addition, increased mRNA expression of IL-1ß, IL-6 and IL-10 were detected in tumors from mice that had been treated with LTD4 or PGE2. CONCLUSION: Our data suggest that both LTD4 and PGE2 promote CICs in initiating tumor growth by allowing modifications in the tumor environment. Our data indicate that new therapeutic strategies targeting eicosanoids, specifically LTD4 and PGE2, could be tested for better therapeutic management of colon cancer.


Subject(s)
Colonic Neoplasms/metabolism , Cytokines/genetics , Dinoprostone/toxicity , Leukotriene D4/toxicity , Neoplastic Stem Cells/metabolism , Animals , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Cytokines/metabolism , Doublecortin-Like Kinases , Female , HCT116 Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Nude , Neoplasms, Experimental , Neoplastic Stem Cells/pathology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Tumor Microenvironment
7.
Carcinogenesis ; 37(5): 491-9, 2016 05.
Article in English | MEDLINE | ID: mdl-26979937

ABSTRACT

There is emerging literature emphasizing the role of inflammatory eicosanoids, including prostaglandins and leukotrienes, in cancer development. Increased expression of both the cysteinyl leukotriene receptor 1 (CysLTR1) and the enzyme responsible for the production of leukotrienes, 5-lipoxygenase, is associated with poor prognosis in patients with colorectal adenocarcinomas. Apc mutation is an early event in the development of sporadic and hereditary (familial adenomatous polyposis) colorectal cancer. We utilized the Apc(Min/+) mouse model of familial adenomatous polyposis/sporadic colorectal cancer to investigate the role of CysLTR1 in intestinal tumorigenesis by crossing Apc(Min/+) mice with mice lacking the Cysltr1 gene. We could observe a reduced tumor burden in the small intestine of double-mutant female (Cysltr1 (-/-) Apc (Min/+) ) but not double-mutant male mice, compared with gender-matched single-mutant (Cysltr1 (+/+) Apc (Min/+) ) mice. This reduction was in a Cysltr1-dependent manner, female double-mutant mice having significantly reduced tumor formation compared with control littermates. The female double-mutant phenotype was accompanied with decreased systemic inflammation, as evidenced by significantly reduced serum levels of prostaglandin E2 and CysLTs, as well as increased CD3(+)CD8(+) T-cell tumor infiltration. Furthermore, the reduced formation of polyps in double-mutant (Cysltr1 (-/-) Apc (Min/+) ) female mice could in part be explained by the cytotoxic action of CD3(+)CD8(+) T cells in the polyp and reduced nuclear accumulation of ß-catenin in the epithelium of small intestinal polyps. Our results stress the important role that CysLTR1 plays in colorectal cancer and its potential as a therapeutic target in cancer therapy.


Subject(s)
Intestinal Polyps/genetics , Adenomatous Polyposis Coli Protein/genetics , Animals , Colorectal Neoplasms/genetics , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Dinoprostone/blood , Dinoprostone/genetics , Female , Gene Expression Regulation, Neoplastic , Intestinal Polyps/epidemiology , Intestinal Polyps/pathology , Intestine, Small/metabolism , Intestine, Small/pathology , Leukocyte Common Antigens/genetics , Leukocyte Common Antigens/metabolism , Lymphocytes, Tumor-Infiltrating/pathology , Male , Mice, Inbred C57BL , Mice, Mutant Strains , Mucin-2/genetics , Mucin-2/metabolism , Neoplasms, Experimental/genetics , Receptors, Leukotriene/genetics , beta Catenin/metabolism
8.
Mol Carcinog ; 54(11): 1315-27, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25154976

ABSTRACT

The role of inflammatory lipid-mediators in tumor progression is well recognized in colorectal cancer; however, if this includes promotion of cancer-initiating cells remains unclear. We show that the inflammatory lipid-mediators leukotriene D4 and prostaglandin E2 increased the Aldehyde dehydrogenase (ALDH(+) ) population, the colony formation capacity, and tumor growth in a xenograft model of colon cancer. The ALDH(+) cells showed significant resistance to irradiation and 5-fluorouracil treatment that could be further augmented by these lipid-mediators, occurring in parallel with increased target gene expression. Our data emphasize a role for tumor microenvironment derived inflammatory lipid-mediators to favor cancer stem cells-like characteristics and thus promote tumor progression.


Subject(s)
Colonic Neoplasms/genetics , Dinoprostone/genetics , Inflammation/genetics , Leukotriene D4/genetics , Lipids/genetics , Neoplastic Stem Cells/metabolism , Animals , Caco-2 Cells , Cell Line, Tumor , Colonic Neoplasms/pathology , Disease Progression , Drug Resistance, Neoplasm/genetics , Female , Fluorouracil/pharmacology , HCT116 Cells , HT29 Cells , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplastic Stem Cells/pathology , Tumor Microenvironment/drug effects , Tumor Microenvironment/genetics , Xenograft Model Antitumor Assays
9.
Mol Biol Cell ; 21(9): 1546-55, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20237161

ABSTRACT

During spermiogenesis, Drosophila melanogaster spermatids coordinate their elongation in interconnected cysts that become highly polarized, with nuclei localizing to one end and sperm tail growth occurring at the other. Remarkably little is known about the signals that drive spermatid polarity and elongation. Here we identify phosphoinositides as critical regulators of these processes. Reduction of plasma membrane phosphatidylinositol 4,5-bisphosphate (PIP(2)) by low-level expression of the PIP(2) phosphatase SigD or mutation of the PIP(2) biosynthetic enzyme Skittles (Sktl) results in dramatic defects in spermatid cysts, which become bipolar and fail to fully elongate. Defects in polarity are evident from the earliest stages of elongation, indicating that phosphoinositides are required for establishment of polarity. Sktl and PIP(2) localize to the growing end of the cysts together with the exocyst complex. Strikingly, the exocyst becomes completely delocalized when PIP(2) levels are reduced, and overexpression of Sktl restores exocyst localization and spermatid cyst polarity. Moreover, the exocyst is required for polarity, as partial loss of function of the exocyst subunit Sec8 results in bipolar cysts. Our data are consistent with a mechanism in which localized synthesis of PIP(2) recruits the exocyst to promote targeted membrane delivery and polarization of the elongating cysts.


Subject(s)
Cell Polarity , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Phosphatidylinositol Phosphates/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Spermatids/metabolism , Animals , Animals, Genetically Modified , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Membrane/metabolism , Cells, Cultured , Drosophila Proteins/genetics , Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Exocytosis , Immunoblotting , Infertility, Male/genetics , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , Microscopy, Electron , Microscopy, Fluorescence , Mutation , Phosphatidylinositol 4,5-Diphosphate , Phosphotransferases (Alcohol Group Acceptor)/genetics , Spermatids/cytology , Testis/cytology , Testis/metabolism , Testis/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...