Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Bioorg Chem ; 143: 107094, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38199139

ABSTRACT

Microtubule dynamics are critical for spindle assembly and chromosome segregation during cell division. Pharmacological inhibition of microtubule dynamics in cells causes prolonged mitotic arrest, resulting in apoptosis, an approach extensively employed in treating different types of cancers. The present study reports the synthesis of thirty-two novel bis-amides (SSE1901-SSE1932) and the evaluation of their antiproliferative activities. N-(1-oxo-3-phenyl-1-(phenylamino)propan-2-yl)benzamide (SSE1917) exhibited the most potent activity with GI50 values of 0.331 ± 0.01 µM in HCT116 colorectal and 0.48 ± 0.27 µM in BT-549 breast cancer cells. SSE1917 stabilized microtubules in biochemical and cellular assays, bound to taxol site in docking studies, and caused aberrant mitosis and G2/M arrest in cells. Prolonged treatment of cells with the compound increased p53 expression and triggered apoptotic cell death. Furthermore, SSE1917 suppressed the growth of both mouse and patient-derived human colon cancer organoids, highlighting its potential therapeutic value as an anticancer agent.


Subject(s)
Antineoplastic Agents , Tubulin Modulators , Tubulin , Animals , Humans , Mice , Amides/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/metabolism , Apoptosis , Cell Line, Tumor , Cell Proliferation , Microtubules/metabolism , Mitosis , Tubulin/drug effects , Tubulin/metabolism , Tubulin Modulators/chemistry , Tubulin Modulators/pharmacology
2.
ACS Med Chem Lett ; 14(10): 1369-1377, 2023 Oct 12.
Article in English | MEDLINE | ID: mdl-37849542

ABSTRACT

Microtubules are dynamic structures that form spindle fibers during cell division; pharmacological inhibition of microtubule dynamics arrests cells in mitosis, leading to apoptosis, and they have been extensively used to treat various cancers. However, the efficacy of such drugs is often limited by multidrug resistance. This study synthesized and evaluated 30 novel derivatives of podophyllotoxin, a natural antimitotic compound, for their antiproliferative activities. Compound SSE1806 exhibited the most potent antiproliferative activity with GI50 values ranging from 1.29 ± 0.01 to 21.15 ± 2.1 µM in cancer cell lines of different origins; it directly inhibited microtubule polymerization, causing aberrant mitosis and G2/M arrest. Prolonged treatment with SSE1806 increased p53 expression, induced cell death in monolayer cultures, and reduced the growth of mouse- and patient-derived human colon cancer organoids. Importantly, SSE1806 overcame multidrug resistance in a cell line overexpressing MDR-1. Thus, SSE1806 represents a potential anticancer agent that can overcome multidrug resistance.

3.
Front Cell Dev Biol ; 11: 1272730, 2023.
Article in English | MEDLINE | ID: mdl-37886398

ABSTRACT

Cellular plasticity defines the capacity of cells to adopt distinct identities during development, tissue homeostasis and regeneration. Dynamic fluctuations between different states, within or across lineages, are regulated by changes in chromatin accessibility and in gene expression. When deregulated, cellular plasticity can contribute to cancer initiation and progression. Cancer cells are remarkably plastic which contributes to phenotypic and functional heterogeneity within tumours as well as resistance to targeted therapies. It is for these reasons that the scientific community has become increasingly interested in understanding the molecular mechanisms governing cancer cell plasticity. The purpose of this mini-review is to discuss different examples of cellular plasticity associated with metaplasia and epithelial-mesenchymal transition with a focus on therapy resistance.

4.
Nat Commun ; 13(1): 2791, 2022 05 19.
Article in English | MEDLINE | ID: mdl-35589755

ABSTRACT

Tumour cell plasticity is a major barrier to the efficacy of targeted cancer therapies but the mechanisms that mediate it are poorly understood. Here, we identify dysregulated RNA splicing as a key driver of tumour cell dedifferentiation in colorectal cancer (CRC). We find that Apc-deficient CRC cells have dysregulated RNA splicing machinery and exhibit global rewiring of RNA splicing. We show that the splicing factor SRSF1 controls the plasticity of tumour cells by controlling Kras splicing and is required for CRC invasion in a mouse model of carcinogenesis. SRSF1 expression maintains stemness in human CRC organoids and correlates with cancer stem cell marker expression in human tumours. Crucially, partial genetic downregulation of Srsf1 does not detrimentally affect normal tissue homeostasis, demonstrating that tumour cell plasticity can be differentially targeted. Thus, our findings link dysregulation of the RNA splicing machinery and control of tumour cell plasticity.


Subject(s)
Cell Plasticity , Colorectal Neoplasms , Animals , Carcinogenesis/genetics , Carcinogenesis/metabolism , Cell Plasticity/genetics , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Mice , RNA Splicing/genetics , Serine-Arginine Splicing Factors/genetics , Serine-Arginine Splicing Factors/metabolism
5.
Dis Model Mech ; 15(1)2022 01 01.
Article in English | MEDLINE | ID: mdl-35014671

ABSTRACT

Alternative splicing is a process by which a single gene is able to encode multiple different protein isoforms. It is regulated by the inclusion or exclusion of introns and exons that are joined in different patterns prior to protein translation, thus enabling transcriptomic and proteomic diversity. It is now widely accepted that alternative splicing is dysregulated across nearly all cancer types. This widespread dysregulation means that nearly all cellular processes are affected - these include processes synonymous with the hallmarks of cancer - evasion of apoptosis, tissue invasion and metastasis, altered cellular metabolism, genome instability and drug resistance. Emerging evidence indicates that the dysregulation of alternative splicing also promotes a permissive environment for increased tumour heterogeneity and cellular plasticity. These are fundamental regulators of a patient's response to therapy. In this Review, we introduce the mechanisms of alternative splicing and the role of aberrant splicing in cancer, with particular focus on newfound evidence of alternative splicing promoting tumour heterogeneity, cellular plasticity and altered metabolism. We discuss recent in vivo models generated to study alternative splicing and the importance of these for understanding complex tumourigenic processes. Finally, we review the effects of alternative splicing on immune evasion, cell death and genome instability, and how targeting these might enhance therapeutic efficacy.


Subject(s)
Alternative Splicing , Neoplasms , Alternative Splicing/genetics , Carcinogenesis/genetics , Humans , Introns , Neoplasms/genetics , Neoplasms/pathology , Neoplasms/therapy , Proteomics , RNA Splicing
6.
Cell Death Dis ; 12(10): 873, 2021 09 25.
Article in English | MEDLINE | ID: mdl-34564693

ABSTRACT

RAC1B is a tumour-related alternative splice isoform of the small GTPase RAC1, found overexpressed in a large number of tumour types. Building evidence suggests it promotes tumour progression but compelling in vivo evidence, demonstrating a role in driving tumour invasion, is currently lacking. In the present study, we have overexpressed RAC1B in a colorectal cancer mouse model with potential invasive properties. Interestingly, RAC1B overexpression did not trigger tumour invasion, rather it led to an acceleration of tumour initiation and reduced mouse survival. By modelling early stages of adenoma initiation we observed a reduced apoptotic rate in RAC1B overexpressing tumours, suggesting protection from apoptosis as a mediator of this phenotype. RAC1B overexpressing tumours displayed attenuated TGFß signalling and functional analysis in ex vivo organoid cultures demonstrated that RAC1B negatively modulates TGFß signalling and confers resistance to TGFß-driven cell death. This work defines a novel mechanism by which early adenoma cells can overcome the cytostatic and cytotoxic effects of TGFß signalling and characterises a new oncogenic function of RAC1B in vivo.


Subject(s)
Apoptosis , Carcinogenesis/metabolism , Carcinogenesis/pathology , Intestines/pathology , Transforming Growth Factor beta/metabolism , rac1 GTP-Binding Protein/metabolism , Adenoma/pathology , Adenomatous Polyposis Coli/metabolism , Animals , Carcinogenesis/genetics , Disease Models, Animal , Down-Regulation , Gene Expression Regulation, Neoplastic , Mice , Models, Biological , Signal Transduction , Survival Analysis , Tumor Suppressor Protein p53/metabolism
7.
Nat Commun ; 12(1): 2335, 2021 04 20.
Article in English | MEDLINE | ID: mdl-33879799

ABSTRACT

Current therapeutic options for treating colorectal cancer have little clinical efficacy and acquired resistance during treatment is common, even following patient stratification. Understanding the mechanisms that promote therapy resistance may lead to the development of novel therapeutic options that complement existing treatments and improve patient outcome. Here, we identify RAC1B as an important mediator of colorectal tumourigenesis and a potential target for enhancing the efficacy of EGFR inhibitor treatment. We find that high RAC1B expression in human colorectal cancer is associated with aggressive disease and poor prognosis and deletion of Rac1b in a mouse colorectal cancer model reduces tumourigenesis. We demonstrate that RAC1B interacts with, and is required for efficient activation of the EGFR signalling pathway. Moreover, RAC1B inhibition sensitises cetuximab resistant human tumour organoids to the effects of EGFR inhibition, outlining a potential therapeutic target for improving the clinical efficacy of EGFR inhibitors in colorectal cancer.


Subject(s)
Colorectal Neoplasms/etiology , Colorectal Neoplasms/metabolism , rac1 GTP-Binding Protein/metabolism , Animals , Antineoplastic Agents, Immunological/pharmacology , Carcinogenesis , Cell Line, Tumor , Cetuximab/pharmacology , Colorectal Neoplasms/genetics , Drug Resistance, Neoplasm , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuropeptides/deficiency , Neuropeptides/genetics , Neuropeptides/metabolism , Signal Transduction , Up-Regulation , Wnt Signaling Pathway , rac1 GTP-Binding Protein/deficiency , rac1 GTP-Binding Protein/genetics
8.
Cell Mol Gastroenterol Hepatol ; 11(2): 465-489, 2021.
Article in English | MEDLINE | ID: mdl-32971322

ABSTRACT

BACKGROUND & AIMS: Aspirin reduces colorectal cancer (CRC) incidence and mortality. Understanding the biology responsible for this protective effect is key to developing biomarker-led approaches for rational clinical use. Wnt signaling drives CRC development from initiation to progression through regulation of epithelial-mesenchymal transition (EMT) and cancer stem cell populations. Here, we investigated whether aspirin can rescue these proinvasive phenotypes associated with CRC progression in Wnt-driven human and mouse intestinal organoids. METHODS: We evaluated aspirin-mediated effects on phenotype and stem cell markers in intestinal organoids derived from mouse (ApcMin/+ and Apcflox/flox) and human familial adenomatous polyposis patients. CRC cell lines (HCT116 and Colo205) were used to study effects on motility, invasion, Wnt signaling, and EMT. RESULTS: Aspirin rescues the Wnt-driven cystic organoid phenotype by promoting budding in mouse and human Apc deficient organoids, which is paralleled by decreased stem cell marker expression. Aspirin-mediated Wnt inhibition in ApcMin/+ mice is associated with EMT inhibition and decreased cell migration, invasion, and motility in CRC cell lines. Chemical Wnt activation induces EMT and stem-like alterations in CRC cells, which are rescued by aspirin. Aspirin increases expression of the Wnt antagonist Dickkopf-1 in CRC cells and organoids derived from familial adenomatous polyposis patients, which contributes to EMT and cancer stem cell inhibition. CONCLUSIONS: We provide evidence of phenotypic biomarkers of response to aspirin with an increased epithelial and reduced stem-like state mediated by an increase in Dickkopf-1. This highlights a novel mechanism of aspirin-mediated Wnt inhibition and potential phenotypic and molecular biomarkers for trials.


Subject(s)
Adenomatous Polyposis Coli/drug therapy , Aspirin/pharmacology , Intercellular Signaling Peptides and Proteins/agonists , Intestinal Mucosa/drug effects , Wnt Signaling Pathway/drug effects , Adenomatous Polyposis Coli/genetics , Adenomatous Polyposis Coli/pathology , Adenomatous Polyposis Coli Protein/genetics , Animals , Aspirin/therapeutic use , Disease Models, Animal , Female , Gene Expression Regulation, Neoplastic/drug effects , HCT116 Cells , Humans , Intercellular Signaling Peptides and Proteins/analysis , Intercellular Signaling Peptides and Proteins/metabolism , Intestinal Mucosa/cytology , Intestinal Mucosa/pathology , Intravital Microscopy , Male , Mice , Mice, Transgenic , Organoids/drug effects , Organoids/pathology , Primary Cell Culture
9.
BMC Biol ; 16(1): 3, 2018 01 10.
Article in English | MEDLINE | ID: mdl-29329541

ABSTRACT

BACKGROUND: Nitric oxide (NO) has been highlighted as an important agent in cancer-related events. Although the inducible nitric oxide synthase (iNOS) isoform has received most attention, recent studies in the literature indicate that the endothelial isoenzyme (eNOS) can also modulate different tumor processes including resistance, angiogenesis, invasion, and metastasis. However, the role of eNOS in cancer stem cell (CSC) biology and mesenchymal tumors is unknown. RESULTS: Here, we show that eNOS was significantly upregulated in VilCre ERT2 Apc fl/+ and VilCre ERT2 Apc fl/fl mouse intestinal tissue, with intense immunostaining in hyperproliferative crypts. Similarly, the more invasive VilCre ERT2 Apc fl/+ Pten fl/+ mouse model showed an overexpression of eNOS in intestinal tumors whereas this isoform was not expressed in normal tissue. However, none of the three models showed iNOS expression. Notably, when 40 human colorectal tumors were classified into different clinically relevant molecular subtypes, high eNOS expression was found in the poor relapse-free and overall survival mesenchymal subtype, whereas iNOS was absent. Furthermore, Apc fl/fl organoids overexpressed eNOS compared with wild-type organoids and NO depletion with the scavenger carboxy-PTIO (c-PTIO) decreased the proliferation and the expression of stem-cell markers, such as Lgr5, Troy, Vav3, and Slc14a1, in these intestinal organoids. Moreover, specific NO depletion also decreased the expression of CSC-related proteins in human colorectal cancer cells such as ß-catenin and Bmi1, impairing the CSC phenotype. To rule out the contribution of iNOS in this effect, we established an iNOS-knockdown colorectal cancer cell line. NO-depleted cells showed a decreased capacity to form tumors and c-PTIO treatment in vivo showed an antitumoral effect in a xenograft mouse model. CONCLUSION: Our data support that eNOS upregulation occurs after Apc loss, emerging as an unexpected potential new target in poor-prognosis mesenchymal colorectal tumors, where NO scavenging could represent an interesting therapeutic alternative to targeting the CSC subpopulation.


Subject(s)
Biomarkers, Tumor/biosynthesis , Cell Proliferation/physiology , Colorectal Neoplasms/enzymology , Intestines/enzymology , Mesenchymal Stem Cells/enzymology , Nitric Oxide Synthase Type III/physiology , Animals , Caco-2 Cells , Colorectal Neoplasms/pathology , HCT116 Cells , Humans , Intestines/pathology , Male , Mesenchymal Stem Cells/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Xenograft Model Antitumor Assays/methods
10.
EMBO Mol Med ; 9(2): 181-197, 2017 02.
Article in English | MEDLINE | ID: mdl-28003334

ABSTRACT

Cancer genome sequencing projects have identified hundreds of genetic alterations, often at low frequencies, raising questions as to their functional relevance. One exemplar gene is HUWE1, which has been found to be mutated in numerous studies. However, due to the large size of this gene and a lack of functional analysis of identified mutations, their significance to carcinogenesis is unclear. To determine the importance of HUWE1, we chose to examine its function in colorectal cancer, where it is mutated in up to 15 per cent of tumours. Modelling of identified mutations showed that they inactivate the E3 ubiquitin ligase activity of HUWE1. Genetic deletion of Huwe1 rapidly accelerated tumourigenic in mice carrying loss of the intestinal tumour suppressor gene Apc, with a dramatic increase in tumour initiation. Mechanistically, this phenotype was driven by increased MYC and rapid DNA damage accumulation leading to loss of the second copy of Apc The increased levels of DNA damage sensitised Huwe1-deficient tumours to DNA-damaging agents and to deletion of the anti-apoptotic protein MCL1. Taken together, these data identify HUWE1 as a bona fide tumour suppressor gene in the intestinal epithelium and suggest a potential vulnerability of HUWE1-mutated tumours to DNA-damaging agents and inhibitors of anti-apoptotic proteins.


Subject(s)
Carcinogenesis , Colorectal Neoplasms/pathology , DNA Damage , Genes, Tumor Suppressor , Proto-Oncogene Proteins c-myc/metabolism , Signal Transduction , Ubiquitin-Protein Ligases/metabolism , Animals , Gene Deletion , Mice , Mutant Proteins/genetics , Mutant Proteins/metabolism , Tumor Suppressor Proteins , Ubiquitin-Protein Ligases/genetics
11.
Cell Rep ; 12(6): 1019-31, 2015 Aug 11.
Article in English | MEDLINE | ID: mdl-26235622

ABSTRACT

An understanding of the mechanisms determining MYC's transcriptional and proliferation-promoting activities in vivo could facilitate approaches for MYC targeting. However, post-translational mechanisms that control MYC function in vivo are poorly understood. Here, we demonstrate that MYC phosphorylation at serine 62 enhances MYC accumulation on Lamin A/C-associated nuclear structures and that the protein phosphatase 2A (PP2A) inhibitor protein CIP2A is required for this process. CIP2A is also critical for serum-induced MYC phosphorylation and for MYC-elicited proliferation induction in vitro. Complementary transgenic approaches and an intestinal regeneration model further demonstrated the in vivo importance of CIP2A and serine 62 phosphorylation for MYC activity upon DNA damage. However, targeting of CIP2A did not influence the normal function of intestinal crypt cells. These data underline the importance of nuclear organization in the regulation of MYC phosphorylation, leading to an in vivo demonstration of a strategy for inhibiting MYC activity without detrimental physiological effects.


Subject(s)
Autoantigens/metabolism , Cell Nucleus/metabolism , Lamin Type A/metabolism , Membrane Proteins/metabolism , Animals , Autoantigens/genetics , Cell Nucleus/genetics , Gene Expression Regulation, Neoplastic/genetics , Immunohistochemistry , Lamin Type A/genetics , Membrane Proteins/genetics , Mice , Models, Biological , Phosphorylation , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism
12.
Nature ; 517(7535): 497-500, 2015 Jan 22.
Article in English | MEDLINE | ID: mdl-25383520

ABSTRACT

Inactivation of APC is a strongly predisposing event in the development of colorectal cancer, prompting the search for vulnerabilities specific to cells that have lost APC function. Signalling through the mTOR pathway is known to be required for epithelial cell proliferation and tumour growth, and the current paradigm suggests that a critical function of mTOR activity is to upregulate translational initiation through phosphorylation of 4EBP1 (refs 6, 7). This model predicts that the mTOR inhibitor rapamycin, which does not efficiently inhibit 4EBP1 (ref. 8), would be ineffective in limiting cancer progression in APC-deficient lesions. Here we show in mice that mTOR complex 1 (mTORC1) activity is absolutely required for the proliferation of Apc-deficient (but not wild-type) enterocytes, revealing an unexpected opportunity for therapeutic intervention. Although APC-deficient cells show the expected increases in protein synthesis, our study reveals that it is translation elongation, and not initiation, which is the rate-limiting component. Mechanistically, mTORC1-mediated inhibition of eEF2 kinase is required for the proliferation of APC-deficient cells. Importantly, treatment of established APC-deficient adenomas with rapamycin (which can target eEF2 through the mTORC1-S6K-eEF2K axis) causes tumour cells to undergo growth arrest and differentiation. Taken together, our data suggest that inhibition of translation elongation using existing, clinically approved drugs, such as the rapalogs, would provide clear therapeutic benefit for patients at high risk of developing colorectal cancer.


Subject(s)
Cell Transformation, Neoplastic/pathology , Intestinal Neoplasms/metabolism , Intestinal Neoplasms/pathology , Multiprotein Complexes/metabolism , Peptide Chain Elongation, Translational , TOR Serine-Threonine Kinases/metabolism , Adenomatous Polyposis Coli Protein/deficiency , Adenomatous Polyposis Coli Protein/genetics , Animals , Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Elongation Factor 2 Kinase/deficiency , Elongation Factor 2 Kinase/genetics , Elongation Factor 2 Kinase/metabolism , Enzyme Activation , Genes, APC , Intestinal Neoplasms/genetics , Male , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Inbred C57BL , Oncogene Protein p55(v-myc)/metabolism , Peptide Elongation Factor 2/metabolism , Ribosomal Protein S6 Kinases/metabolism , Signal Transduction , Wnt Proteins/metabolism
13.
EMBO Mol Med ; 6(12): 1525-41, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25253726

ABSTRACT

Deregulated expression of MYC is a driver of colorectal carcinogenesis, necessitating novel strategies to inhibit MYC function. The ubiquitin ligase HUWE1 (HECTH9, ARF-BP1, MULE) associates with both MYC and the MYC-associated protein MIZ1. We show here that HUWE1 is required for growth of colorectal cancer cells in culture and in orthotopic xenograft models. Using high-throughput screening, we identify small molecule inhibitors of HUWE1, which inhibit MYC-dependent transactivation in colorectal cancer cells, but not in stem and normal colon epithelial cells. Inhibition of HUWE1 stabilizes MIZ1. MIZ1 globally accumulates on MYC target genes and contributes to repression of MYC-activated target genes upon HUWE1 inhibition. Our data show that transcriptional activation by MYC in colon cancer cells requires the continuous degradation of MIZ1 and identify a novel principle that allows for inhibition of MYC function in tumor cells.


Subject(s)
Colorectal Neoplasms/enzymology , Oncogene Protein p55(v-myc)/antagonists & inhibitors , Oncogene Protein p55(v-myc)/metabolism , Small Molecule Libraries/pharmacology , Ubiquitin-Protein Ligases/antagonists & inhibitors , Animals , Cell Line, Tumor , Cell Proliferation , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/physiopathology , Gene Expression Regulation, Neoplastic , Humans , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Mice , Mice, SCID , Oncogene Protein p55(v-myc)/genetics , Protein Binding , Small Molecule Libraries/administration & dosage , Transcriptional Activation , Tumor Suppressor Proteins , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
14.
Cell Rep ; 6(6): 1153-1164, 2014 Mar 27.
Article in English | MEDLINE | ID: mdl-24630994

ABSTRACT

The small G protein family Rac has numerous regulators that integrate extracellular signals into tight spatiotemporal maps of its activity to promote specific cell morphologies and responses. Here, we have generated a mouse strain, Rac-FRET, which ubiquitously expresses the Raichu-Rac biosensor. It enables FRET imaging and quantification of Rac activity in live tissues and primary cells without affecting cell properties and responses. We assessed Rac activity in chemotaxing Rac-FRET neutrophils and found enrichment in leading-edge protrusions and unexpected longitudinal shifts and oscillations during protruding and stalling phases of migration. We monitored Rac activity in normal or disease states of intestinal, liver, mammary, pancreatic, and skin tissue, in response to stimulation or inhibition and upon genetic manipulation of upstream regulators, revealing unexpected insights into Rac signaling during disease development. The Rac-FRET strain is a resource that promises to fundamentally advance our understanding of Rac-dependent responses in primary cells and native environments.


Subject(s)
Neutrophils/enzymology , rac GTP-Binding Proteins/metabolism , Animals , Enzyme Activation , Fluorescence Resonance Energy Transfer/methods , Mice , Neutrophils/cytology , Signal Transduction , Spatio-Temporal Analysis , rac GTP-Binding Proteins/chemistry
15.
Cell Cycle ; 12(18): 2973-7, 2013 Sep 15.
Article in English | MEDLINE | ID: mdl-23974108

ABSTRACT

Adult stem cells are responsible for maintaining the balance between cell proliferation and differentiation within self-renewing tissues. The molecular and cellular mechanisms mediating such balance are poorly understood. The production of reactive oxygen species (ROS) has emerged as an important mediator of stem cell homeostasis in various systems. Our recent work demonstrates that Rac1-dependent ROS production mediates intestinal stem cell (ISC) proliferation in mouse models of colorectal cancer (CRC). Here, we use the adult Drosophila midgut and the mouse small intestine to directly address the role of Rac1 in ISC proliferation and tissue regeneration in response to damage. Our results demonstrate that Rac1 is necessary and sufficient to drive ISC proliferation and regeneration in an ROS-dependent manner. Our data point to an evolutionarily conserved role of Rac1 in intestinal homeostasis and highlight the value of combining work in the mammalian and Drosophila intestine as paradigms to study stem cell biology.


Subject(s)
Drosophila Proteins/metabolism , Intestines/physiology , Regeneration , Stem Cells/cytology , rac1 GTP-Binding Protein/metabolism , Animals , Cell Proliferation , Drosophila , Hydrogen-Ion Concentration , Mice , Mice, Inbred C57BL , Oxidative Stress , RNA Interference , Reactive Oxygen Species/metabolism , Stem Cells/metabolism , rac1 GTP-Binding Protein/antagonists & inhibitors , rac1 GTP-Binding Protein/genetics
16.
Cell Stem Cell ; 12(6): 761-73, 2013 Jun 06.
Article in English | MEDLINE | ID: mdl-23665120

ABSTRACT

The Adenomatous Polyposis Coli (APC) gene is mutated in the majority of colorectal cancers (CRCs). Loss of APC leads to constitutively active WNT signaling, hyperproliferation, and tumorigenesis. Identification of pathways that facilitate tumorigenesis after APC loss is important for therapeutic development. Here, we show that RAC1 is a critical mediator of tumorigenesis after APC loss. We find that RAC1 is required for expansion of the LGR5 intestinal stem cell (ISC) signature, progenitor hyperproliferation, and transformation. Mechanistically, RAC1-driven ROS and NF-κB signaling mediate these processes. Together, these data highlight that ROS production and NF-κB activation triggered by RAC1 are critical events in CRC initiation.


Subject(s)
Colorectal Neoplasms/pathology , Intestine, Small/cytology , NF-kappa B/metabolism , Neuropeptides/metabolism , Reactive Oxygen Species/metabolism , Stem Cells/cytology , Wnt Proteins/metabolism , rac1 GTP-Binding Protein/metabolism , Animals , Cell Proliferation , Colorectal Neoplasms/metabolism , Intestine, Small/metabolism , Mice , Mice, Inbred C57BL , Signal Transduction , Stem Cells/metabolism
17.
Development ; 139(24): 4524-35, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23172913

ABSTRACT

Inactivating mutations within adenomatous polyposis coli (APC), a negative regulator of Wnt signaling, are responsible for most sporadic and hereditary forms of colorectal cancer (CRC). Here, we use the adult Drosophila midgut as a model system to investigate the molecular events that mediate intestinal hyperplasia following loss of Apc in the intestine. Our results indicate that the conserved Wnt target Myc and its binding partner Max are required for the initiation and maintenance of intestinal stem cell (ISC) hyperproliferation following Apc1 loss. Importantly, we find that loss of Apc1 leads to the production of the interleukin-like ligands Upd2/3 and the EGF-like Spitz in a Myc-dependent manner. Loss of Apc1 or high Wg in ISCs results in non-cell-autonomous upregulation of upd3 in enterocytes and subsequent activation of Jak/Stat signaling in ISCs. Crucially, knocking down Jak/Stat or Spitz/Egfr signaling suppresses Apc1-dependent ISC hyperproliferation. In summary, our results uncover a novel non-cell-autonomous interplay between Wnt/Myc, Egfr and Jak/Stat signaling in the regulation of intestinal hyperproliferation. Furthermore, we present evidence suggesting potential conservation in mouse models and human CRC. Therefore, the Drosophila adult midgut proves to be a powerful genetic system to identify novel mediators of APC phenotypes in the intestine.


Subject(s)
Drosophila Proteins/physiology , Drosophila , ErbB Receptors/physiology , Intestines/pathology , Janus Kinases/physiology , Receptors, Invertebrate Peptide/physiology , STAT Transcription Factors/physiology , Transcription Factors/physiology , Adult Stem Cells/metabolism , Adult Stem Cells/pathology , Adult Stem Cells/physiology , Age Factors , Animals , Animals, Genetically Modified , Apc1 Subunit, Anaphase-Promoting Complex-Cyclosome , DNA Replication/genetics , DNA Replication/physiology , Drosophila/genetics , Drosophila/growth & development , Drosophila/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Enterocytes/metabolism , Enterocytes/pathology , Enterocytes/physiology , ErbB Receptors/genetics , ErbB Receptors/metabolism , Hyperplasia/genetics , Intestinal Mucosa/metabolism , Janus Kinases/genetics , Janus Kinases/metabolism , Receptor Cross-Talk/physiology , Receptors, Invertebrate Peptide/genetics , Receptors, Invertebrate Peptide/metabolism , STAT Transcription Factors/genetics , STAT Transcription Factors/metabolism , Signal Transduction/genetics , Signal Transduction/physiology , Transcription Factors/genetics , Transcription Factors/metabolism
18.
J Cell Sci ; 125(Pt 22): 5524-34, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-22946062

ABSTRACT

LSH, a protein related to the SNF2 family of chromatin-remodelling ATPases, is essential for the correct establishment of DNA methylation levels and patterns in plants and mammalian cells. However, some of the phenotypes resulting from LSH deficiency cannot be explained easily by defects in DNA methylation. Here we show that LSH-deficient mouse and human fibroblasts show reduced viability after exposure to ionizing radiation and repair DNA double-strand breaks less efficiently than wild-type cells. A more detailed characterisation of this phenotype revealed that, in the absence of LSH, the histone variant H2AX is not efficiently phosphorylated in response to DNA damage. This results in impaired recruitment of MDC1 and 53BP1 proteins to DNA double-strand breaks and compromises phosphorylation of checkpoint kinase CHK2. Furthermore, we demonstrate that the ability of LSH to hydrolyse ATP is necessary for efficient phosphorylation of H2AX at DNA double-strand breaks and successful repair of DNA damage. Taken together, our data reveal a previously unsuspected role of LSH ATPase in the maintenance of genome stability in mammalian somatic cells, which is independent of its function in de novo DNA methylation during development.


Subject(s)
Adenosine Triphosphatases/metabolism , DNA Breaks, Double-Stranded , DNA Helicases/metabolism , DNA Repair , Histones/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Transcription Factors/metabolism , Adaptor Proteins, Signal Transducing , Animals , Cell Cycle Proteins , Checkpoint Kinase 2 , DNA Breaks, Double-Stranded/radiation effects , DNA Helicases/deficiency , DNA Methylation/radiation effects , DNA Repair/radiation effects , Enzyme Activation/radiation effects , Fibroblasts/metabolism , Fibroblasts/radiation effects , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Kinetics , Mammals/metabolism , Mice , Mutant Proteins/metabolism , Phosphorylation/radiation effects , Protein Serine-Threonine Kinases/metabolism , Radiation, Ionizing , Signal Transduction/radiation effects
19.
EMBO J ; 31(14): 3079-91, 2012 Jun 12.
Article in English | MEDLINE | ID: mdl-22692129

ABSTRACT

Two types of stem cells are currently defined in small intestinal crypts: cycling crypt base columnar (CBC) cells and quiescent '+4' cells. Here, we combine transcriptomics with proteomics to define a definitive molecular signature for Lgr5(+) CBC cells. Transcriptional profiling of FACS-sorted Lgr5(+) stem cells and their daughters using two microarray platforms revealed an mRNA stem cell signature of 384 unique genes. Quantitative mass spectrometry on the same cell populations identified 278 proteins enriched in intestinal stem cells. The mRNA and protein data sets showed a high level of correlation and a combined signature of 510 stem cell-enriched genes was defined. Spatial expression patterns were further characterized by mRNA in-situ hybridization, revealing that approximately half of the genes were expressed in a gradient with highest levels at the crypt bottom, while the other half was expressed uniquely in Lgr5(+)stem cells. Lineage tracing using a newly established knock-in mouse for one of the signature genes, Smoc2, confirmed its stem cell specificity. Using this resource, we find-and confirm by independent approaches-that the proposed quiescent/'+4' stem cell markers Bmi1, Tert, Hopx and Lrig1 are robustly expressed in CBC cells.


Subject(s)
Antigens, Differentiation/metabolism , Gene Expression Regulation , Intestinal Mucosa/metabolism , Receptors, G-Protein-Coupled/metabolism , Stem Cells/metabolism , Animals , Antigens, Differentiation/genetics , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Gene Expression Profiling , Intestines/cytology , Mice , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, G-Protein-Coupled/genetics , Stem Cells/cytology
20.
Exp Cell Res ; 317(19): 2725-31, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-21851818

ABSTRACT

Loss of the APC (adenomatous polyposis coli) gene in colorectal cancer leads to a rapid deregulation of TCF/LEF target genes. Of all these target genes, the transcription factor c-MYC appears the most critical. In this review we will discuss the interplay of Wnt and c-MYC signaling during intestinal homeostasis and transformation. Furthermore, we will discuss recent data showing that further deregulation of c-MYC levels during colorectal carcinogenesis may drive tumor progression. Moreover, understanding these additional control mechanisms may allow targeting of c-MYC during colorectal carcinogenesis.


Subject(s)
Carcinoma/genetics , Epistasis, Genetic/physiology , Genes, myc , Intestinal Neoplasms/genetics , Wnt Proteins/genetics , Animals , Carcinoma/metabolism , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Crime , Humans , Intestinal Neoplasms/metabolism , Models, Biological , Wit and Humor as Topic , Wnt Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...