Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 9 de 9
1.
Nat Genet ; 54(12): 1827-1838, 2022 12.
Article En | MEDLINE | ID: mdl-36175792

We identify the sodium leak channel non-selective protein (NALCN) as a key regulator of cancer metastasis and nonmalignant cell dissemination. Among 10,022 human cancers, NALCN loss-of-function mutations were enriched in gastric and colorectal cancers. Deletion of Nalcn from gastric, intestinal or pancreatic adenocarcinomas in mice did not alter tumor incidence, but markedly increased the number of circulating tumor cells (CTCs) and metastases. Treatment of these mice with gadolinium-a NALCN channel blocker-similarly increased CTCs and metastases. Deletion of Nalcn from mice that lacked oncogenic mutations and never developed cancer caused shedding of epithelial cells into the blood at levels equivalent to those seen in tumor-bearing animals. These cells trafficked to distant organs to form normal structures including lung epithelium, and kidney glomeruli and tubules. Thus, NALCN regulates cell shedding from solid tissues independent of cancer, divorcing this process from tumorigenesis and unmasking a potential new target for antimetastatic therapies.


Neoplasms , Humans , Mice , Animals , Ion Channels/genetics , Membrane Proteins/genetics
2.
Dis Model Mech ; 14(1)2021 01 01.
Article En | MEDLINE | ID: mdl-33093165

Somatic models of tissue pathology commonly use induction of gene-specific mutations in mice mediated by spatiotemporal regulation of Cre recombinase. Subsequent investigation of the onset and development of disease can be limited by the inability to track changing cellular behaviours over time. Here, a lineage-tracing approach based on ligand-dependent activation of Dre recombinase that can be employed independently of Cre is described. The clonal biology of the intestinal epithelium following Cre-mediated stabilisation of ß-catenin reveals that, within tumours, many new clones rapidly become extinct. Surviving clones show accelerated population of tumour glands compared to normal intestinal crypts but in a non-uniform manner, indicating that intra-tumour glands follow heterogeneous dynamics. In tumour-adjacent epithelia, clone sizes are smaller than in the background epithelia, as a whole. This suggests a zone of ∼seven crypt diameters within which clone expansion is inhibited by tumours and that may facilitate their growth.


Intestinal Neoplasms/genetics , Intestinal Neoplasms/metabolism , Mutation , Animals , Antibodies, Monoclonal/chemistry , Cell Lineage , Colon/metabolism , Epithelial Cells/metabolism , Epithelium/metabolism , Escherichia coli Proteins/metabolism , Female , Integrases/metabolism , Intestinal Mucosa/metabolism , Intestine, Small/metabolism , Intestines/metabolism , Male , Mice , Mice, Inbred C57BL , Neoplasms/metabolism , Probability , Recombinases/metabolism , Stem Cells/cytology , beta Catenin/metabolism
3.
Genetics ; 212(3): 655-665, 2019 07.
Article En | MEDLINE | ID: mdl-31126976

Microsatellite sequences have an enhanced susceptibility to mutation, and can act as sentinels indicating elevated mutation rates and increased risk of cancer. The probability of mutant fixation within the intestinal epithelium is dictated by a combination of stem cell dynamics and mutation rate. Here, we exploit this relationship to infer microsatellite mutation rates. First a sensitive, multiplexed, and quantitative method for detecting somatic changes in microsatellite length was developed that allowed the parallel detection of mutant [CA]n sequences from hundreds of low-input tissue samples at up to 14 loci. The method was applied to colonic crypts in Mus musculus, and enabled detection of mutant subclones down to 20% of the cellularity of the crypt (∼50 of 250 cells). By quantifying age-related increases in clone frequencies for multiple loci, microsatellite mutation rates in wild-type and Msh2-deficient epithelium were established. An average 388-fold increase in mutation per mitosis rate was observed in Msh2-deficient epithelium (2.4 × 10-2) compared to wild-type epithelium (6.2 × 10-5).


Adult Stem Cells/metabolism , Intestinal Mucosa/cytology , Microsatellite Repeats , MutS Homolog 2 Protein/genetics , Mutation Rate , Adult Stem Cells/cytology , Animals , Female , Intestinal Mucosa/metabolism , Male , Mice , Mice, Inbred C57BL , Mitosis , MutS Homolog 2 Protein/deficiency
4.
Nat Cell Biol ; 20(10): 1193-1202, 2018 10.
Article En | MEDLINE | ID: mdl-30177776

Solid malignancies have been speculated to depend on cancer stem cells (CSCs) for expansion and relapse after therapy. Here we report on quantitative analyses of lineage tracing data from primary colon cancer xenograft tissue to assess CSC functionality in a human solid malignancy. The temporally obtained clone size distribution data support a model in which stem cell function in established cancers is not intrinsically, but is entirely spatiotemporally orchestrated. Functional stem cells that drive tumour expansion predominantly reside at the tumour edge, close to cancer-associated fibroblasts. Hence, stem cell properties change in time depending on the cell location. Furthermore, although chemotherapy enriches for cells with a CSC phenotype, in this context functional stem cell properties are also fully defined by the microenvironment. To conclude, we identified osteopontin as a key cancer-associated fibroblast-produced factor that drives in situ clonogenicity in colon cancer.


Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Proliferation/drug effects , Colonic Neoplasms/drug therapy , Neoplastic Stem Cells/drug effects , Tumor Microenvironment/drug effects , Xenograft Model Antitumor Assays , Animals , Cell Proliferation/genetics , Cells, Cultured , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Humans , Mice, Nude , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Oxaliplatin/administration & dosage , Tamoxifen/administration & dosage , Tumor Microenvironment/genetics
5.
J Exp Med ; 215(3): 725-727, 2018 03 05.
Article En | MEDLINE | ID: mdl-29440363

In this issue of JEM, Balbinot et al. (https://doi.org/10.1084/jem.20170934) describe an original mechanism where Cdx2 inactivation regulates intestinal metaplastic to neoplastic transition in a paracrine fashion. Surprisingly, the target cells are neighboring "normal" Cdx2-positive cells.


Genes, Homeobox , Homeodomain Proteins/genetics , Animals , CDX2 Transcription Factor , Carcinogenesis , Mice , Mice, Transgenic
6.
Breast Cancer Res ; 18(1): 127, 2016 12 13.
Article En | MEDLINE | ID: mdl-27964754

BACKGROUND: High-resolution 3D imaging of intact tissue facilitates cellular and subcellular analyses of complex structures within their native environment. However, difficulties associated with immunolabelling and imaging fluorescent proteins deep within whole organs have restricted their applications to thin sections or processed tissue preparations, precluding comprehensive and rapid 3D visualisation. Several tissue clearing methods have been established to circumvent issues associated with depth of imaging in opaque specimens. The application of these techniques to study the elaborate architecture of the mouse mammary gland has yet to be investigated. METHODS: Multiple tissue clearing methods were applied to intact virgin and lactating mammary glands, namely 3D imaging of solvent-cleared organs, see deep brain (seeDB), clear unobstructed brain imaging cocktails (CUBIC) and passive clarity technique. Using confocal, two-photon and light sheet microscopy, their compatibility with whole-mount immunofluorescent labelling and 3D imaging of mammary tissue was examined. In addition, their suitability for the analysis of mouse mammary tumours was also assessed. RESULTS: Varying degrees of optical transparency, tissue preservation and fluorescent signal conservation were observed between the different clearing methods. SeeDB and CUBIC protocols were considered superior for volumetric fluorescence imaging and whole-mount histochemical staining, respectively. Techniques were compatible with 3D imaging on a variety of platforms, enabling visualisation of mammary ductal and lobulo-alveolar structures at vastly improved depths in cleared tissue. CONCLUSIONS: The utility of whole-organ tissue clearing protocols was assessed in the mouse mammary gland. Most methods utilised affordable and widely available reagents, and were compatible with standard confocal microscopy. These techniques enable high-resolution, 3D imaging and phenotyping of mammary cells and tumours in situ, and will significantly enhance our understanding of both normal and pathological mammary gland development.


Imaging, Three-Dimensional , Mammary Glands, Animal/diagnostic imaging , Mammary Neoplasms, Animal/diagnostic imaging , Mammary Neoplasms, Animal/pathology , Animals , Female , Fluorescent Antibody Technique , Imaging, Three-Dimensional/methods , Mice , Microscopy, Confocal , Optical Imaging/methods
7.
Gut ; 63(3): 480-93, 2014 Mar.
Article En | MEDLINE | ID: mdl-23585469

OBJECTIVE: Colorectal cancer (CRC) is a major contributor to cancer mortality and morbidity. LIM kinase 2 (LIMK2) promotes tumour cell invasion and metastasis. The objectives of this study were to determine how LIMK2 expression is associated with CRC progression and patient outcome, and to use genetically modified Drosophila and mice to determine how LIMK2 deletion affects gastrointestinal stem cell regulation and tumour development. DESIGN: LIMK2 expression and activity were measured by immunostaining tumours from CRC-prone mice, human CRC cell lines and 650 human tumours. LIMK knockdown in Drosophila or Limk2 deletion in mice allowed for assessment of their contributions to gastrointestinal stem cell homeostasis and tumour development. RESULTS: LIMK2 expression was reduced in intestinal tumours of cancer-prone mice, as well as in human CRC cell lines and tumours. Reduced LIMK2 expression and substrate phosphorylation were associated with shorter patient survival. Genetic analysis in Drosophila midgut and intestinal epithelial cells isolated from genetically modified mice revealed a conserved role for LIMK2 in constraining gastrointestinal stem cell proliferation. Limk2 deletion increased colon tumour size in a colitis-associated colorectal mouse cancer model. CONCLUSIONS: This study revealed that LIMK2 expression and activity progressively decrease with advancing stage, and supports the hypothesis that there is selective pressure for reduced LIMK2 expression in CRC to relieve negative constraints imposed upon gastrointestinal stem cells.


Biomarkers, Tumor/metabolism , Colon/enzymology , Colorectal Neoplasms/enzymology , Intestinal Mucosa/enzymology , Lim Kinases/metabolism , Neoplastic Stem Cells/enzymology , Animals , Biomarkers, Tumor/deficiency , Cell Line, Tumor , Cell Proliferation , Colon/pathology , Colon/physiopathology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Colorectal Neoplasms/physiopathology , DNA Methylation , Disease Progression , Down-Regulation , Drosophila melanogaster , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Intestinal Mucosa/pathology , Intestinal Mucosa/physiopathology , Lim Kinases/deficiency , Mice , Mice, Knockout , Neoplastic Stem Cells/physiology , Reverse Transcriptase Polymerase Chain Reaction , Tissue Array Analysis
8.
PLoS One ; 6(2): e17143, 2011 Feb 15.
Article En | MEDLINE | ID: mdl-21358804

A common goal for potential cancer therapies is the identification of differences in protein expression or activity that would allow for the selective targeting of tumor vs. normal cells. The Ras proto-oncogene family (K-Ras, H-Ras and N-Ras) are amongst the most frequently mutated genes in human cancers. As a result, there has been substantial effort dedicated to determining which pathways are activated by Ras signaling and, more importantly, which of these contribute to cancer. Although the most widely studied Ras-regulated signaling pathway is the Raf/mitogen-activated protein kinase cascade, previous research in model systems has revealed that the Rac1 GTP-binding protein is also required for Ras-induced biological responses. However, what have been lacking are rigorous in vivo Rac1 target validation data and a clear demonstration that in Ras-driven hyperplastic lesions, Rac1 activity is increased. Using a combination of genetically-modified mouse models that allow for the tissue-selective activation or deletion of signaling molecules and an activation-state sensitive Rac1 antibody that detects GTP-bound Rac1, we found that Rac1 contributes to K-Ras induced epidermal papilloma initiation and growth and that Rac1 activity is elevated by oncogenic K-Ras in vivo. Previously, it was not practical to assess Rac1 activation status in the most commonly used format for clinical tumor specimens, formalin-fixed paraffin embedded (FFPE) tissues samples. However, this study clearly demonstrates that Rac1 is essential for K-Ras driven epithelial cell hyperproliferation and that Rac1 activity is elevated in tissues expressing mutant oncogenic K-Ras, while also characterizing the activation-state specific Rac1-GTP antibody as a probe to examine Rac1 activation status in FFPE samples. Our findings will facilitate further research on the status of Rac1 activity in human tumors and will help to define the tumor types of the patient population that could potentially benefit from therapies targeting Rac activation or downstream effector signaling pathways.


Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Epidermis/pathology , Genes, ras/physiology , rac1 GTP-Binding Protein/physiology , Animals , Cell Proliferation , Enzyme Activation/physiology , Epidermis/metabolism , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Genes, ras/genetics , Humans , Integrases/genetics , Integrases/metabolism , Mice , Mice, Transgenic , Mouth Neoplasms/genetics , Mouth Neoplasms/metabolism , Mouth Neoplasms/pathology , Papilloma/genetics , Papilloma/metabolism , Papilloma/pathology , Proto-Oncogene Mas , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Transgenes , rac1 GTP-Binding Protein/metabolism
9.
Cell Res ; 21(4): 666-82, 2011 Apr.
Article En | MEDLINE | ID: mdl-21079653

The central arbiter of cell fate in response to DNA damage is p53, which regulates the expression of genes involved in cell cycle arrest, survival and apoptosis. Although many responses initiated by DNA damage have been characterized, the role of actin cytoskeleton regulators is largely unknown. We now show that RhoC and LIM kinase 2 (LIMK2) are direct p53 target genes induced by genotoxic agents. Although RhoC and LIMK2 have well-established roles in actin cytoskeleton regulation, our results indicate that activation of LIMK2 also has a pro-survival function following DNA damage. LIMK inhibition by siRNA-mediated knockdown or selective pharmacological blockade sensitized cells to radio- or chemotherapy, such that treatments that were sub-lethal when administered singly resulted in cell death when combined with LIMK inhibition. Our findings suggest that combining LIMK inhibitors with genotoxic therapies could be more efficacious than single-agent administration, and highlight a novel connection between actin cytoskeleton regulators and DNA damage-induced cell survival mechanisms.


Actins/metabolism , Gene Expression Regulation , Lim Kinases/metabolism , Signal Transduction , Tumor Suppressor Protein p53/metabolism , rho GTP-Binding Proteins/metabolism , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Chromatin Immunoprecipitation , Cytoskeleton , DNA Damage , Fluorescent Antibody Technique , Gene Knockdown Techniques , Humans , Immunoblotting , Mice , Microarray Analysis , RNA, Small Interfering , Signal Transduction/drug effects , Transcription, Genetic , rho-Associated Kinases/metabolism , rhoC GTP-Binding Protein
...