Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Cancer Immunol Immunother ; 72(12): 4001-4014, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37973660

ABSTRACT

BACKGROUND: Regulation of alternative splicing is a new therapeutic approach in cancer. The programmed cell death receptor 1 (PD-1) is an immunoinhibitory receptor expressed on immune cells that binds to its ligands, PD-L1 and PD-L2 expressed by cancer cells forming a dominant immune checkpoint pathway in the tumour microenvironment. Targeting this pathway using blocking antibodies (nivolumab and pembrolizumab) is the mainstay of anti-cancer immunotherapies, restoring the function of exhausted T cells. PD-1 is alternatively spliced to form isoforms that are either transmembrane signalling receptors (flPD1) that mediate T cell death by binding to the ligand, PD-L1 or an alternatively spliced, soluble, variant that lacks the transmembrane domain. METHODS: We used PCR and western blotting on primary peripheral blood mononuclear cells (PBMCs) and Jurkat T cells, IL-2 ELISA, flow cytometry, co-culture of melanoma and cholangiocarcinoma cells, and bioinformatics analysis and molecular cloning to examine the mechanism of splicing of PD1 and its consequence. RESULTS: The soluble form of PD-1, generated by skipping exon 3 (∆Ex3PD1), was endogenously expressed in PBMCs and T cells and prevents cancer cell-mediated T cell repression. Multiple binding sites of SRSF1 are adjacent to PD-1 exon 3 splicing sites. Overexpression of phosphomimic SRSF1 resulted in preferential expression of flPD1. Inhibition of SRSF1 phosphorylation both by SRPK1 shRNA knockdown and by a selective inhibitor, SPHINX31, resulted in a switch in splicing to ∆Ex3PD1. Cholangiocarcinoma cell-mediated repression of T cell IL-2 expression was reversed by SPHINX31 (equivalent to pembrolizumab). CONCLUSIONS: These results indicate that switching of the splicing decision from flPD1 to ∆Ex3PD1 by targeting SRPK1 could represent a potential novel mechanism of immune checkpoint inhibition in cancer.


Subject(s)
Alternative Splicing , Cholangiocarcinoma , Humans , Phosphorylation , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , Protein Serine-Threonine Kinases/genetics , RNA Splicing Factors/genetics , RNA Splicing Factors/metabolism , Arginine/genetics , Arginine/metabolism , Serine/chemistry , Serine/genetics , Serine/metabolism , T-Cell Exhaustion , Interleukin-2/genetics , Leukocytes, Mononuclear/metabolism , Programmed Cell Death 1 Receptor/metabolism , Serine-Arginine Splicing Factors/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , Immunotherapy
2.
Oncogenesis ; 11(1): 4, 2022 Jan 19.
Article in English | MEDLINE | ID: mdl-35046388

ABSTRACT

Little is known about the role of microRNAs (miRNAs) in rewiring the metabolism within tumours and adjacent non-tumour bearing normal tissue and their potential in cancer therapy. This study aimed to investigate the relationship between deregulated miRNAs and metabolic components in murine duodenal polyps and non-polyp-derived organoids (mPOs and mNPOs) from a double-mutant ApcMinFbxw7∆G mouse model of intestinal/colorectal cancer (CRC). We analysed the expression of 373 miRNAs and 12 deregulated metabolic genes in mPOs and mNPOs. Our findings revealed miR-135b might target Spock1. Upregulation of SPOCK1 correlated with advanced stages of CRCs. Knockdown of miR-135b decreased the expression level of SPOCK1, glucose consumption and lactic secretion in CRC patient-derived tumours organoids (CRC tPDOs). Increased SPOCK1 induced by miR-135b overexpression promoted the Warburg effect and consequently antitumour effect of 5-fluorouracil. Thus, combination with miR-135b antisense nucleotides may represent a novel strategy to sensitise CRC to the chemo-reagent based treatment.

3.
Am J Physiol Lung Cell Mol Physiol ; 322(2): L283-L293, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34936509

ABSTRACT

Lymphangioleiomyomatosis (LAM) is a female-specific cystic lung disease in which tuberous sclerosis complex 2 (TSC2)-deficient LAM cells, LAM-associated fibroblasts (LAFs), and other cell types infiltrate the lungs. LAM lesions can be associated with type II alveolar epithelial (AT2) cells. We hypothesized that the behavior of AT2 cells in LAM is influenced locally by LAFs. We tested this hypothesis in the patient samples and in vitro. In human LAM lung, nodular AT2 cells show enhanced proliferation when compared with parenchymal AT2 cells, demonstrated by increased Ki67 expression. Furthermore, nodular AT2 cells express proteins associated with epithelial activation in other disease states including matrix metalloproteinase 7, and fibroblast growth factor 7 (FGF7). In vitro, LAF-conditioned medium is mitogenic and positively chemotactic for epithelial cells, increases the rate of epithelial repair, and protects against apoptosis. In vitro, LAM patient-derived TSC2 null cells cocultured with LAFs upregulate LAF expression of the epithelial chemokine and mitogen FGF7, a potential mediator of fibroblast-epithelial cross talk, in a mechanistic target of rapamycin (mTOR)-dependent manner. In a novel in vitro model of LAM, ex vivo cultured LAM lung-derived microtissues promote both epithelial migration and adhesion. Our findings suggest that AT2 cells in LAM display a proliferative, activated phenotype and fibroblast accumulation following LAM cell infiltration into the parenchyma contributes to this change in AT2 cell behavior. Fibroblast-derived FGF7 may contribute to the cross talk between LAFs and hyperplastic epithelium in vivo, but does not appear to be the main driver of the effects of LAFs on epithelial cells in vitro.


Subject(s)
Lung Neoplasms , Lymphangioleiomyomatosis , Female , Humans , Alveolar Epithelial Cells/metabolism , Fibroblasts/metabolism , Lung Neoplasms/pathology , Lymphangioleiomyomatosis/metabolism , Tuberous Sclerosis , Tuberous Sclerosis Complex 2 Protein/metabolism , Tumor Suppressor Proteins/metabolism
4.
Cells ; 10(4)2021 04 13.
Article in English | MEDLINE | ID: mdl-33924684

ABSTRACT

Over recent years, several Cys2-His2 (C2H2) domain-containing proteins have emerged as critical players in repairing DNA-double strand breaks. Human FLYWCH1 is a newly characterised nuclear transcription factor with (C2H2)-type zinc-finger DNA-binding domains. Yet, our knowledge about FLYWCH1 is still in its infancy. This study explores the expression, role and regulation of FLYWCH1 in the context of DNA damage and repair. We provide evidence suggesting a potential contribution of FLYWCH1 in facilitating the recruitment of DNA-damage response proteins (DDRPs). We found that FLYWCH1 colocalises with γH2AX in normal fibroblasts and colorectal cancer (CRC) cell lines. Importantly, our results showed that enforced expression of FLYWCH1 induces the expression of γH2AX, ATM and P53 proteins. Using an ATM-knockout (ATMKO) model, we indicated that FLYWCH1 mediates the phosphorylation of H2AX (Ser139) independently to ATM expression. On the other hand, the induction of DNA damage using UV-light induces the endogenous expression of FLYWCH1. Conversely, cisplatin treatment reduces the endogenous level of FLYWCH1 in CRC cell lines. Together, our findings uncover a novel FLYWCH1/H2AX phosphorylation axis in steady-state conditions and during the induction of the DNA-damage response (DDR). Although the role of FLYWCH1 within the DDR machinery remains largely uncharacterised and poorly understood, we here report for the first-time findings that implicate FLYWCH1 as a potential participant in the DNA damage response signaling pathways.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/genetics , DNA Repair , DNA-Binding Proteins/genetics , DNA/genetics , Histones/genetics , Ataxia Telangiectasia Mutated Proteins/deficiency , Cell Line , Cell Line, Tumor , Cisplatin/pharmacology , DNA/metabolism , DNA Damage , DNA-Binding Proteins/metabolism , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression Regulation , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HCT116 Cells , Histones/metabolism , Humans , Phosphorylation , Ribonuclease III/genetics , Ribonuclease III/metabolism , Signal Transduction , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
5.
Am J Respir Crit Care Med ; 204(4): 431-444, 2021 08 15.
Article in English | MEDLINE | ID: mdl-33882264

ABSTRACT

Rationale: Lymphangioleiomyomatosis (LAM) is a multisystem disease that causes lung cysts and respiratory failure. Loss of TSC (tuberous sclerosis complex) gene function results in a clone of "LAM cells" with dysregulated mTOR (mechanistic target of rapamycin) activity. LAM cells and fibroblasts form lung nodules that also contain mast cells, although their significance is unknown. Objectives: To understand the mechanism of mast-cell accumulation and the role of mast cells in the pathogenesis of LAM. Methods: Gene expression was examined using transcriptional profiling and qRT-PCR. Mast cell/LAM nodule interactions were examined in vitro using spheroid TSC2-null cell/fibroblast cocultures and in vivo using an immunocompetent Tsc2-null murine homograft model. Measurements and Main Results: LAM-derived cell/fibroblast cocultures induced multiple CXC chemokines in fibroblasts. LAM lungs had increased tryptase-positive mast cells expressing CXCRs (CXC chemokine receptors) (P < 0.05). Mast cells located around the periphery of LAM nodules were positively associated with the rate of lung function loss (P = 0.016). LAM spheroids attracted mast cells, and this process was inhibited by pharmacologic and CRISPR/cas9 inhibition of CXCR1 and CXCR2. LAM spheroids caused mast-cell tryptase release, which induced fibroblast proliferation and increased LAM-spheroid size (1.36 ± 0.24-fold; P = 0.0019). The tryptase inhibitor APC366 and sodium cromoglycate (SCG) inhibited mast cell-induced spheroid growth. In vivo, SCG reduced mast-cell activation and Tsc2-null lung tumor burden (vehicle: 32.5.3% ± 23.6%; SCG: 5.5% ± 4.3%; P = 0.0035). Conclusions: LAM-cell/fibroblast interactions attract mast cells where tryptase release contributes to disease progression. Repurposing SCG for use in LAM should be studied as an alternative or adjunct to mTOR inhibitor therapy.


Subject(s)
Biomarkers, Tumor/metabolism , Fibroblasts/metabolism , Lung Neoplasms/metabolism , Lymphangioleiomyomatosis/metabolism , Mast Cells/metabolism , Tryptases/metabolism , Adult , Animals , Biomarkers, Tumor/genetics , Chemokines/metabolism , Disease Progression , Fibroblasts/pathology , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lymphangioleiomyomatosis/genetics , Lymphangioleiomyomatosis/pathology , Mast Cells/pathology , Mice , Mice, Inbred C57BL , Middle Aged , Spheroids, Cellular , Tumor Cells, Cultured
6.
J Pathol Clin Res ; 6(3): 215-226, 2020 07.
Article in English | MEDLINE | ID: mdl-32352655

ABSTRACT

Lymphangioleiomyomatosis (LAM) is a rare multisystem disease with a variable clinical course. The lungs are infiltrated by nodules of LAM cells, stromal cells and inflammatory cells, causing lung cysts and respiratory failure. We used immunohistochemical markers in lung biopsy and transplant samples from a national cohort of women with LAM with linked clinical data to understand how LAM nodule cell populations changed with disease progression. Marker distribution was examined qualitatively by dual immunohistochemistry, and markers for LAM cells, fibroblasts, lymphatics, mast cells, proliferation, cathepsin K and mTOR pathway activity were quantitated in LAM nodules and compared with clinical features and prospective lung function loss. The LAM cell marker PNL2 was more extensively expressed in those with higher forced expiratory volume in one second (FEV1 ), higher diffusion in the lung for carbon monoxide (DLCO ) and less extensive disease involvement whilst the converse was true for the protease cathepsin K. Each percentage increase in cathepsin K reactivity was associated with a 0.65% decrease in FEV1 (95% CI -1.11 to -0.18) and a 0.50% decrease in DLCO (95% CI -0.96 to -0.05). Higher reactivity to the mTOR complex 1 activation marker, phospho-ribosomal protein S6, was associated with a better lung function response to rapamycin (p = 0.0001). We conclude that LAM nodules evolve with disease progression, with LAM cells becoming outnumbered by fibroblasts. Increasing cathepsin K expression is associated with more severe disease and lung function loss. Markers of mTOR activation predict the response to rapamycin, suggesting that more advanced LAM may be less mTOR responsive and treatments specifically targeted towards LAM associated fibroblasts may have roles as adjuncts to mTOR inhibition.


Subject(s)
Disease Progression , Lung/pathology , Lymphangioleiomyomatosis , Perivascular Epithelioid Cell Neoplasms , Adult , Antigens, Neoplasm/metabolism , Biomarkers, Tumor/metabolism , Cathepsin K/metabolism , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Immunohistochemistry , Lung/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lymphangioleiomyomatosis/metabolism , Lymphangioleiomyomatosis/pathology , Middle Aged , Perivascular Epithelioid Cell Neoplasms/metabolism , Perivascular Epithelioid Cell Neoplasms/pathology , Prospective Studies , TOR Serine-Threonine Kinases/metabolism
7.
Cells ; 8(10)2019 10 18.
Article in English | MEDLINE | ID: mdl-31635307

ABSTRACT

Ataxia-telegiectasia mutated (ATM), phosphatase and tensin homolog (PTEN), and p85α are key tumour suppressors. Whether ATM regulates PTEN expression and influence platinum sensitivity is unknown. We generated ATM knockdowns (KD) and CRISPR knock outs (KO) in glioblastoma (LN18, LN229) and ovarian cancer cells (OVCAR3, OVCAR4). Doxycycline inducible PTEN expression was generated in LN18 and LN229 cells. Transient KD of p85α, CK2, and XIAP was accomplished using siRNAs. Stable p85α knock-in was isolated in LN18 cells. Molecular biology assays included proteasome activity assays, PCR, flow cytometry analysis (cell cycle, double strand break accumulation, apoptosis), immunofluorescence, co-immunoprecipitation, clonogenic, invasion, migration, and 3D neurosphere assays. The clinicopathological significance of ATM, PTEN, p85α, and XIAP (X-linked inhibitor of apoptosis protein) was evaluated in 525 human ovarian cancers using immunohistochemistry. ATM regulated PTEN is p85α dependant. ATM also controls CK2α level which in turn phosphorylates and stabilizes PTEN. In addition, p85α physically interacts with CK2α and protects CK2α from ATM regulated degradation. ATM deficiency resulted in accumulation of XIAP/p-XIAP levels which ubiquitinated PTEN and CK2α thereby directing them to degradation. ATM depletion in the context of p85α deficiency impaired cancer cell migration and invasion reduced 3D-neurosphere formation and increased toxicity to cisplatin chemotherapy. Increased sensitivity to platinum was associated with DNA double strand breaks accumulation, cell cycle arrest, and induction of autophagy. In ovarian cancer patients, ATM, PTEN, p85α, and XIAP protein levels predicted better progression free survival after platinum therapy. We unravel a previously unknown function of ATM in the regulation of PTEN throµgh XIAP mediated proteasome degradation.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/metabolism , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , PTEN Phosphohydrolase/metabolism , Platinum/therapeutic use , Ubiquitin-Protein Ligases/metabolism , X-Linked Inhibitor of Apoptosis Protein/metabolism , Ataxia Telangiectasia Mutated Proteins/genetics , Blotting, Western , Caspase 3/genetics , Caspase 3/metabolism , Caspase 7/genetics , Caspase 7/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cisplatin/therapeutic use , Doxycycline/therapeutic use , Female , Flow Cytometry , HeLa Cells , Humans , Ovarian Neoplasms/genetics , PTEN Phosphohydrolase/genetics , Plasmids/genetics , Protein Stability/drug effects , Real-Time Polymerase Chain Reaction , Tissue Array Analysis , Ubiquitin-Protein Ligases/genetics , X-Linked Inhibitor of Apoptosis Protein/genetics
8.
Int J Mol Sci ; 20(11)2019 Jun 04.
Article in English | MEDLINE | ID: mdl-31167387

ABSTRACT

Acute myeloid leukaemia (AML) is a heterogeneous clonal malignancy of hematopoietic progenitor cells. The Wnt pathway and its downstream targets are tightly regulated by ß-catenin. We recently discovered a new protein, FLYWCH1, which can directly bind nuclear ß-catenin. Herein, we studied the FLYWCH1/ß-catenin pathway in AML cells using qRT-PCR, Western blot, and immunofluorescence assays. In addition, the stemness activity and cell cycle were analysed by the colony-forming unit (CFU) using methylcellulose-based and Propidium iodide/flow cytometry assays. We found that FLYWCH1 mRNA and protein were differentially expressed in the AML cell lines. C-Myc, cyclin D1, and c-Jun expression decreased in the presence of higher FLYWCH1 expression, and vice versa. There appeared to be the loss of FLYWCH1 expression in dividing cells. The sub-G0 phase was prolonged and shortened in the low and high FLYWCH1 expression cell lines, respectively. The G0/G1 arrest correlated with FLYWCH1-expression, and these cell lines also formed colonies, whereas the low FLYWCH1 expression cell lines could not. Thus, FLYWCH1 functions as a negative regulator of the Wnt/ß-catenin pathway in AML.


Subject(s)
DNA-Binding Proteins/genetics , Gene Expression Regulation, Leukemic , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Wnt Proteins/metabolism , beta Catenin/metabolism , Cell Cycle/genetics , Cell Line, Tumor , DNA-Binding Proteins/metabolism , Fluorescent Antibody Technique , Humans , Neoplastic Stem Cells/metabolism , RNA, Messenger/genetics , Wnt Signaling Pathway
9.
Oncogenesis ; 8(3): 13, 2019 Feb 19.
Article in English | MEDLINE | ID: mdl-30783098

ABSTRACT

Colorectal cancer (CRC) patients develop recurrence after chemotherapy owing to the survival of stem cell-like cells referred to as cancer stem-like cells (CSCs). The origin of CSCs is linked to the epithelial-mesenchymal transition (EMT) process. Currently, it remains poorly understood how EMT programmes enable CSCs residing in the tumour microenvironment to escape the effects of chemotherapy. This study identifies a key molecular pathway that is responsible for the formation of drug-resistant CSC populations. Using a modified yeast-2-hybrid system and 2D gel-based proteomics methods, we show that the E3-ubiquitin ligase FBXW7 directly binds and degrades the EMT-inducing transcription factor ZEB2 in a phosphorylation-dependent manner. Loss of FBXW7 induces an EMT that can be effectively reversed by knockdown of ZEB2. The FBXW7-ZEB2 axis regulates such important cancer cell features, as stemness/dedifferentiation, chemoresistance and cell migration in vitro, ex vivo and in animal models of metastasis. High expression of ZEB2 in cancer tissues defines the reduced ZEB2 expression in the cancer-associated stroma in patients and in murine intestinal organoids, demonstrating a tumour-stromal crosstalk that modulates a niche and EMT activation. Our study thus uncovers a new molecular mechanism, by which the CRC cells display differences in resistance to chemotherapy and metastatic potential.

10.
Cancers (Basel) ; 12(1)2019 Dec 31.
Article in English | MEDLINE | ID: mdl-31906201

ABSTRACT

Tumour-promoting inflammation is involved in colorectal cancer (CRC) development and therapeutic resistance. However, the antibiotics and antibacterial drugs and signalling that regulate the potency of anticancer treatment upon forced differentiation of cancer stem-like cell (CSC) are not fully defined yet. We screened an NIH-clinical collection of the small-molecule compound library of antibacterial/anti-inflammatory agents that identified potential candidate drugs targeting CRC-SC for differentiation. Selected compounds were validated in both in vitro organoids and ex vivo colon explant models for their differentiation induction, impediment on neoplastic cell growth, and to elucidate the mechanism of their anticancer activity. We initially focused on AM404, an anandamide uptake inhibitor. AM404 is a metabolite of acetaminophen with antibacterial activity, which showed high potential in preventing CRC-SC features, such as stemness/de-differentiation, migration and drug-resistance. Furthermore, AM404 suppressed the expression of FBXL5 E3-ligase, where AM404 sensitivity was mimicked by FBXL5-knockout. This study uncovers a new molecular mechanism for AM404-altering FBXL5 oncogene which mediates chemo-resistance and CRC invasion, thereby proposes to repurpose antibacterial AM404 as an anticancer agent.

11.
Nat Commun ; 9(1): 5378, 2018 12 19.
Article in English | MEDLINE | ID: mdl-30568163

ABSTRACT

We recently identified the splicing kinase gene SRPK1 as a genetic vulnerability of acute myeloid leukemia (AML). Here, we show that genetic or pharmacological inhibition of SRPK1 leads to cell cycle arrest, leukemic cell differentiation and prolonged survival of mice transplanted with MLL-rearranged AML. RNA-seq analysis demonstrates that SRPK1 inhibition leads to altered isoform levels of many genes including several with established roles in leukemogenesis such as MYB, BRD4 and MED24. We focus on BRD4 as its main isoforms have distinct molecular properties and find that SRPK1 inhibition produces a significant switch from the short to the long isoform at the mRNA and protein levels. This was associated with BRD4 eviction from genomic loci involved in leukemogenesis including BCL2 and MYC. We go on to show that this switch mediates at least part of the anti-leukemic effects of SRPK1 inhibition. Our findings reveal that SRPK1 represents a plausible new therapeutic target against AML.


Subject(s)
Leukemia, Myeloid, Acute/metabolism , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Transcription Factors/metabolism , Cell Cycle Checkpoints , Cell Cycle Proteins , Cell Differentiation , Chromatin/metabolism , Epigenesis, Genetic , HL-60 Cells , Hematopoiesis , Humans , K562 Cells , Protein Isoforms/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , RNA Splicing
12.
Mol Cancer Res ; 16(12): 1977-1990, 2018 12.
Article in English | MEDLINE | ID: mdl-30097457

ABSTRACT

Wnt/ß-catenin signaling plays a critical role during development of both normal and malignant colorectal cancer tissues. Phosphorylation of ß-catenin protein alters its trafficking and function. Such conventional allosteric regulation usually involves a highly specialized set of molecular interactions, which may specifically turn on a particular cell phenotype. This study identifies a novel transcription modulator with an FLYWCH/Zn-finger DNA-binding domain, called "FLYWCH1." Using a modified yeast-2-hybrid based Ras-Recruitment system, it is demonstrated that FLYWCH1 directly binds to unphosphorylated (nuclear) ß-catenin efficiently suppressing the transcriptional activity of Wnt/ß-catenin signaling that cannot be rescued by TCF4. FLYWCH1 rearranges the transcriptional activity of ß-catenin/TCF4 to selectively block the expression of specific downstream genes associated with colorectal cancer cell migration and morphology, including ZEB1, EPHA4, and E-cadherin. Accordingly, overexpression of FLYWCH1 reduces cell motility and increases cell attachment. The expression of FLYWCH1 negatively correlates with the expression level of ZEB1 and EPHA4 in normal versus primary and metastatic colorectal cancer tissues in patients. Thus, FLYWCH1 antagonizes ß-catenin/TCF4 signaling during cell polarity/migration in colorectal cancer. IMPLICATIONS: This study uncovers a new molecular mechanism by which FLYWCH1 with a possible tumor suppressive role represses ß-catenin-induced ZEB1 and increases cadherin-mediated cell attachment preventing colorectal cancer metastasis.


Subject(s)
Colorectal Neoplasms/metabolism , DNA-Binding Proteins/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism , Antigens, CD/metabolism , Cadherins/metabolism , Cell Line, Tumor , Cell Movement , DNA-Binding Proteins/chemistry , Down-Regulation , Gene Expression Regulation, Neoplastic , HCT116 Cells , Humans , Tissue Array Analysis , Zinc Finger E-box-Binding Homeobox 1/metabolism , Zinc Fingers
13.
Stem Cells ; 35(4): 839-850, 2017 04.
Article in English | MEDLINE | ID: mdl-28142204

ABSTRACT

Increasing evidence suggests that cancer cell populations contain a small proportion of cells that display stem-like cell properties and which may be responsible for overall tumor maintenance. These cancer stem-like cells (CSCs) appear to have unique tumor-initiating ability and innate survival mechanisms that allow them to resist cancer therapies, consequently promoting relapses. Selective targeting of CSCs may provide therapeutic benefit and several recent reports have indicated this may be possible. In this article, we review drugs targeting CSCs, in selected epithelial cell-derived cancers. Stem Cells 2017;35:839-850.


Subject(s)
Antineoplastic Agents/therapeutic use , Epithelial Cells/pathology , Neoplasms/drug therapy , Neoplasms/pathology , Neoplastic Stem Cells/pathology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Epithelial Cells/drug effects , Humans , Neoplastic Stem Cells/drug effects
14.
ACS Chem Biol ; 12(3): 825-832, 2017 03 17.
Article in English | MEDLINE | ID: mdl-28135068

ABSTRACT

Serine/arginine-protein kinase 1 (SRPK1) regulates alternative splicing of VEGF-A to pro-angiogenic isoforms and SRPK1 inhibition can restore the balance of pro/antiangiogenic isoforms to normal physiological levels. The lack of potency and selectivity of available compounds has limited development of SRPK1 inhibitors, with the control of alternative splicing by splicing factor-specific kinases yet to be translated. We present here compounds that occupy a binding pocket created by the unique helical insert of SRPK1, and trigger a backbone flip in the hinge region, that results in potent (<10 nM) and selective inhibition of SRPK1 kinase activity. Treatment with these inhibitors inhibited SRPK1 activity and phosphorylation of serine/arginine splicing factor 1 (SRSF1), resulting in alternative splicing of VEGF-A from pro-angiogenic to antiangiogenic isoforms. This property resulted in potent inhibition of blood vessel growth in models of choroidal angiogenesis in vivo. This work identifies tool compounds for splice isoform selective targeting of pro-angiogenic VEGF, which may lead to new therapeutic strategies for a diversity of diseases where dysfunctional splicing drives disease development.


Subject(s)
Choroidal Neovascularization/drug therapy , Enzyme Inhibitors/therapeutic use , Protein Serine-Threonine Kinases/antagonists & inhibitors , Administration, Ophthalmic , Humans
15.
Mol Ther Methods Clin Dev ; 3: 16024, 2016.
Article in English | MEDLINE | ID: mdl-27110583

ABSTRACT

Colorectal cancer (CRC) is one of the top three cancer-related causes of death worldwide. FBXW7 is a known tumor-suppressor gene, commonly mutated in CRC and in a variety of other epithelial tumors. Low expression of FBXW7 is also associated with poor prognosis. Loss of FBXW7 sensitizes cancer cells to certain drugs, while making them more resistant to other types of chemotherapies. However, is not fully understood how epithelial cells within normal gut and primary tumors respond to potential cancer therapeutics. We have studied genetically engineered mice in which the fbxw7 gene is conditionally knocked-out in the intestine (fbxw7(∆G)). To further investigate the mechanism of Fbxw7-action, we grew intestinal crypts from floxed-fbxw7 (fbxw7(fl/fl)) and fbxw7(ΔG) mice, in a Matrigel-based organoid (mini-gut) culture. The fbxw7(ΔG) organoids exhibited rapid budding events in the crypt region. Furthermore, to test organoids for drug response, we exposed day 3 intestinal organoids from fbxw7(fl/fl) and fbxw7(∆G) mice, to various concentrations of 5-fluorouracil (5-FU) for 72 hours. 5-FU triggers phenotypic differences in organoids including changing shape, survival, resistance, and death. 5-FU however, rescues the drug-resistance phenotype of fbxw7(ΔG) through the induction of terminal differentiation. Our results support the hypothesis that a differentiating therapy successfully targets FBXW7-mutated CRC cells.

16.
Oncotarget ; 6(11): 9240-56, 2015 Apr 20.
Article in English | MEDLINE | ID: mdl-25860929

ABSTRACT

FBXW7 mutations occur in a variety of human cancers including colorectal cancer (CRC). Elucidating its mechanism of action has become crucial for cancer therapy; however, it is also complicated by the fact that FBXW7 can influence many pathways due to its role as an E3-ubiquitin ligase in proteasome degradation. FBXW7 and TP53 are tumour suppressors intensively implicated in colorectal carcinogenesis. Deletion mutations in these two genes in animal models mark the progression from adenoma to carcinoma. Although still largely unknown, the last defense mechanism against CRC at the molecular level could be through a synergistic effect of the two genes. The underlying mechanism requires further investigation. In our laboratory, we have used a phospho-kinase profiler array to illustrate a potential molecular link between FBXW7 and p53 in CRC cells. In vitro and in vivo assessments demonstrated aberrant induction of phosphorylated p53 at Serine 15 [phospho-p53(Ser15)] in human FBXW7-deficient CRC cells as compared to their FBXW7-wild-type counterparts. FBXW7 loss in HCT116 cells promoted resistance to oxaliplatin. Immunoblotting data further confirmed that reduction of phospho-p53(Ser15) may contribute to the decreased efficacy of therapy in FBXW7-mutated CRC cells. The findings may suggest the applicability of phospho-p53(Ser15) as an indicative marker of FBXW7-mutations. Phospho-p53(Ser15) regulation by FBXW7 E3-ligase activity could provide important clues for understanding FBXW7 behavior in tumour progression and grounds for its clinical applicability thereafter.


Subject(s)
Cell Cycle Proteins/genetics , Colorectal Neoplasms/drug therapy , Drug Resistance, Neoplasm/genetics , F-Box Proteins/genetics , Organoplatinum Compounds/pharmacology , Tumor Suppressor Protein p53/genetics , Ubiquitin-Protein Ligases/genetics , Antineoplastic Agents/pharmacology , Biomarkers, Tumor/genetics , Cell Line, Tumor , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , F-Box-WD Repeat-Containing Protein 7 , Gene Expression Regulation, Neoplastic , HCT116 Cells , Humans , Oxaliplatin , Phosphorylation , Tumor Suppressor Protein p53/biosynthesis , Ubiquitin-Protein Ligases/metabolism
17.
Curr Protoc Stem Cell Biol ; Chapter 1: Unit 1B.10, 2013 May.
Article in English | MEDLINE | ID: mdl-23661244

ABSTRACT

Chromatin immunoprecipitation (ChIP) is a widely used and pre-eminent technique for detecting the association of an individual protein or a particular protein complex with its specific DNA sequence(s) in vivo. Herein we introduce a novel and simple biotinylated-oligonucleotide-mediated ChIP method for testing specific binding of the c-JUN protein to the M1-DNA-regulatory element in the NANOG promoter. We prepared a 260-bp DNA PCR amplicon containing -300 bp to -59 bp, relative to the transcriptional start site of the human NANOG gene, which was transfected into mouse embryonic fibroblasts (MEF) containing wild-type (c-jun(+/+)) or knockout c-jun (c-jun(-/-)) alleles. Whole cells that were cross-linked using formaldehyde and protein-DNA interactions were immunoprecipitated using streptavidin-coupled Dynabeads. Protein-DNA cross-links were reversed during incubation at 95°C, and protein samples were visualized using SDS-PAGE electrophoresis and western blotting. This streptavidin/biotinylated DNA/protein-bound complex protocol can be used for detecting the interactions between multiple transcription factors and their DNA binding sites.


Subject(s)
Chromatin Immunoprecipitation/methods , DNA-Binding Proteins/metabolism , DNA/metabolism , Homeodomain Proteins/genetics , Promoter Regions, Genetic , Proto-Oncogene Proteins c-jun/metabolism , Streptavidin/metabolism , Animals , Biotinylation/drug effects , Blotting, Western , Cross-Linking Reagents/pharmacology , Embryo, Mammalian/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Mice , Nanog Homeobox Protein , Polymerase Chain Reaction , Protein Binding/drug effects
18.
Stem Cells ; 30(10): 2076-87, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22851508

ABSTRACT

Embryonic NANOG (NANOG1) is considered as an important regulator of pluripotency while NANOGP8 (NANOG-pseudogene) plays a role in tumorigenesis. Herein, we show NANOG is expressed from both NANOG1 and NANOGP8 in human colorectal cancers (CRC). Enforced NANOG1-expression increases clonogenic potential and tumor formation in xenograft models, although it is expressed only in a small subpopulation of tumor cells and is colocalized with endogenous nuclear ß-catenin(High) . Moreover, single NANOG1-CRCs form spherical aggregates, similar to the embryoid body of embryonic stem cells (ESCs), and express higher levels of stem-like Wnt-associated target genes. Furthermore, we show that NANOG1-expression is positively regulated by c-JUN and ß-catenin/TCF4. Ectopic expression of c-Jun in murine Apc(Min/+) -ESCs results in the development of larger xenograft tumors with higher cell density compared to controls. Chromatin immunoprecipitation assays demonstrate that c-JUN binds to the NANOG1-promoter via the octamer M1 DNA element. Collectively, our data suggest that ß-Catenin/TCF4 and c-JUN together drive a subpopulation of CRC tumor cells that adopt a stem-like phenotype via the NANOG1-promoter.


Subject(s)
Adaptor Protein Complex 1/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Colorectal Neoplasms/metabolism , Homeodomain Proteins/metabolism , Neoplastic Stem Cells/metabolism , Transcription Factors/metabolism , Adaptor Protein Complex 1/genetics , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Binding Sites , Cell Transformation, Neoplastic , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Female , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/genetics , Humans , JNK Mitogen-Activated Protein Kinases/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Mice , Nanog Homeobox Protein , Neoplasm Transplantation , Neoplastic Stem Cells/pathology , Promoter Regions, Genetic , Protein Binding , Pseudogenes , Signal Transduction/genetics , Transcription Factor 4 , Transcription Factors/genetics , Wnt Proteins/genetics , Wnt Proteins/metabolism , beta Catenin/genetics
19.
J Exp Med ; 208(2): 295-312, 2011 Feb 14.
Article in English | MEDLINE | ID: mdl-21282377

ABSTRACT

The Fbxw7 (F-box/WD repeat-containing protein 7; also called CDC4, Sel10, Ago, and Fbw7) component of the SCF (Skp1/Cullin/F-box protein) E3 ubiquitin ligase complex acts as a tumor suppressor in several tissues and targets multiple transcriptional activators and protooncogenes for ubiquitin-mediated degradation. To understand Fbxw7 function in the murine intestine, in this study, we specifically deleted Fbxw7 in the murine gut using Villin-Cre (Fbxw7(ΔG)). In wild-type mice, loss of Fbxw7 in the gut altered homeostasis of the intestinal epithelium, resulted in elevated Notch and c-Jun expression, and induced development of adenomas at 9-10 mo of age. In the context of APC (adenomatous polyposis coli) deficiency (Apc(Min/+) mice), loss of Fbxw7 accelerated intestinal tumorigenesis and death and promoted accumulation of ß-catenin in adenomas at late but not early time points. At early time points, Fbxw7 mutant tumors showed accumulation of the DEK protooncogene. DEK expression promoted cell division and altered splicing of tropomyosin (TPM) RNA, which may also influence cell proliferation. DEK accumulation and altered TPM RNA splicing were also detected in FBXW7 mutant human colorectal tumor tissues. Given their reduced lifespan and increased incidence of intestinal tumors, Apc(Min/+)Fbxw7(ΔG) mice may be used for testing carcinogenicity and drug screening.


Subject(s)
Adenoma/metabolism , F-Box Proteins/pharmacology , Homeostasis/physiology , Intestinal Neoplasms/metabolism , Intestines/physiology , Tumor Suppressor Proteins/pharmacology , Ubiquitin-Protein Ligases/pharmacology , Animals , Cell Line , Cell Movement/physiology , Cell Proliferation , Colony-Forming Units Assay , DNA-Binding Proteins/metabolism , F-Box Proteins/genetics , F-Box Proteins/metabolism , F-Box-WD Repeat-Containing Protein 7 , Gene Deletion , Histological Techniques , Homeostasis/drug effects , Homeostasis/genetics , Humans , In Situ Hybridization , Intestinal Mucosa/metabolism , Mice , Oncogene Protein p65(gag-jun)/metabolism , Oncogene Proteins/metabolism , Poly-ADP-Ribose Binding Proteins , Receptors, Notch/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Survival Analysis , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
20.
Mol Cancer Res ; 7(8): 1189-96, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19671687

ABSTRACT

Interactions between transcription and signaling are fundamentally important for understanding both the structure and function of genetic pathways and their role in diseases such as cancer. The finding that beta-catenin/TCF4 and JNK/c-Jun cooperate has important implications in carcinogenesis. Previously, we found that binding of c-Jun and beta-catenin/TCF4 to the c-jun promoter is dependent upon JNK activity, thus one role for this complex is to contribute to the repression and/or activation of genes that may mediate cell maintenance, proliferation, differentiation, and death, whereas deregulation of these signals may contribute to carcinogenesis. Here we address the functional links reported between activated beta-catenin/JNK signaling pathways, their component genes, and their common targets, and discuss how alterations in the properties of these genes lead to the development of cancer.


Subject(s)
JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Signaling System , Neoplasms/enzymology , Neoplasms/genetics , Transcription, Genetic , beta Catenin/metabolism , Animals , Genes, Neoplasm , Humans , Neoplasms/metabolism , Wnt Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...