Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Nat Commun ; 13(1): 2866, 2022 05 23.
Article in English | MEDLINE | ID: mdl-35606354

ABSTRACT

Current therapy against colorectal cancer (CRC) is based on DNA-damaging agents that remain ineffective in a proportion of patients. Whether and how non-curative DNA damage-based treatment affects tumor cell behavior and patient outcome is primarily unstudied. Using CRC patient-derived organoids (PDO)s, we show that sublethal doses of chemotherapy (CT) does not select previously resistant tumor populations but induces a quiescent state specifically to TP53 wildtype (WT) cancer cells, which is linked to the acquisition of a YAP1-dependent fetal phenotype. Cells displaying this phenotype exhibit high tumor-initiating and metastatic activity. Nuclear YAP1 and fetal traits are present in a proportion of tumors at diagnosis and predict poor prognosis in patients carrying TP53 WT CRC tumors. We provide data indicating the higher efficacy of CT together with YAP1 inhibitors for eradication of therapy resistant TP53 WT cancer cells. Together these results identify fetal conversion as a useful biomarker for patient prognosis and therapy prescription.


Subject(s)
Colorectal Neoplasms , Tumor Suppressor Protein p53/metabolism , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Humans , Tumor Suppressor Protein p53/genetics
2.
Cells ; 10(11)2021 10 28.
Article in English | MEDLINE | ID: mdl-34831152

ABSTRACT

BACKGROUND: Previous studies in mice indicated that Paneth cells and c-Kit-positive goblet cells represent the stem cell niche of the small intestine and colon, respectively, partly by supporting Wnt and Notch activation. Whether these cell populations play a similar role in human intestinal cancer remains unexplored. METHODS: We performed histopathological evaluation and immunohistochemical analysis of early colorectal adenomas and carcinoma adenoma from patients at the Hospital del Mar in Barcelona. We then determined the possible correlation between the different parameters analyzed and with patient outcomes. RESULTS: Paneth cells accumulate in a subset of human colorectal adenomas directly associated with Notch and Wnt/ß-catenin activation. Adenoma areas containing Paneth cells display increased vessel density in the lamina propria and higher levels of the stem cell marker EphB2. In an in-house cohort of 200 colorectal adenoma samples, we also observed a significant correlation between the presence of Paneth cells and Wnt activation. Kaplan-Meier analysis indicated that early adenoma patients carrying Paneth cell-positive tumors display reduced disease-free survival compared with patients with Paneth cell-free lesions. CONCLUSIONS: Our results indicate that Paneth cells contribute to the initial steps of cancer progression by providing the stem cell niche to adenoma cells, which could be therapeutically exploited.


Subject(s)
Adenoma/metabolism , Colorectal Neoplasms/pathology , Paneth Cells/pathology , Signal Transduction , beta Catenin/metabolism , Humans , Kaplan-Meier Estimate , Prognosis , Proto-Oncogene Proteins c-kit/metabolism , Receptor, EphB2/metabolism , Receptors, Notch/metabolism , Synaptophysin/metabolism , Wnt Proteins/metabolism
3.
Genome Biol ; 21(1): 284, 2020 11 23.
Article in English | MEDLINE | ID: mdl-33225950

ABSTRACT

BACKGROUND: Adult T cell acute lymphoblastic leukemia (T-ALL) is a rare disease that affects less than 10 individuals in one million. It has been less studied than its cognate pediatric malignancy, which is more prevalent. A higher percentage of the adult patients relapse, compared to children. It is thus essential to study the mechanisms of relapse of adult T-ALL cases. RESULTS: We profile whole-genome somatic mutations of 19 primary T-ALLs from adult patients and the corresponding relapse malignancies and analyze their evolution upon treatment in comparison with 238 pediatric and young adult ALL cases. We compare the mutational processes and driver mutations active in primary and relapse adult T-ALLs with those of pediatric patients. A precise estimation of clock-like mutations in leukemic cells shows that the emergence of the relapse clone occurs several months before the diagnosis of the primary T-ALL. Specifically, through the doubling time of the leukemic population, we find that in at least 14 out of the 19 patients, the population of relapse leukemia present at the moment of diagnosis comprises more than one but fewer than 108 blasts. Using simulations, we show that in all patients the relapse appears to be driven by genetic mutations. CONCLUSIONS: The early appearance of a population of leukemic cells with genetic mechanisms of resistance across adult T-ALL cases constitutes a challenge for treatment. Improving early detection of the malignancy is thus key to prevent its relapse.


Subject(s)
Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Child , DNA Helicases/genetics , Female , Humans , Models, Genetic , Mutation , Nuclear Proteins/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/diagnosis , Recurrence , T-Lymphocytes , Transcription Factors/genetics , Whole Genome Sequencing , Young Adult
4.
Nature ; 583(7815): 265-270, 2020 07.
Article in English | MEDLINE | ID: mdl-32581361

ABSTRACT

Cancers arise through the acquisition of oncogenic mutations and grow by clonal expansion1,2. Here we reveal that most mutagenic DNA lesions are not resolved into a mutated DNA base pair within a single cell cycle. Instead, DNA lesions segregate, unrepaired, into daughter cells for multiple cell generations, resulting in the chromosome-scale phasing of subsequent mutations. We characterize this process in mutagen-induced mouse liver tumours and show that DNA replication across persisting lesions can produce multiple alternative alleles in successive cell divisions, thereby generating both multiallelic and combinatorial genetic diversity. The phasing of lesions enables accurate measurement of strand-biased repair processes, quantification of oncogenic selection and fine mapping of sister-chromatid-exchange events. Finally, we demonstrate that lesion segregation is a unifying property of exogenous mutagens, including UV light and chemotherapy agents in human cells and tumours, which has profound implications for the evolution and adaptation of cancer genomes.


Subject(s)
Chromosome Segregation/genetics , Evolution, Molecular , Genome/genetics , Neoplasms/genetics , Alleles , Animals , DNA Repair , DNA Replication , ErbB Receptors/metabolism , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Male , Mice , Mutation , Neoplasms/pathology , Selection, Genetic , Signal Transduction , Sister Chromatid Exchange , Transcription, Genetic , raf Kinases/metabolism , ras Proteins/metabolism
5.
EMBO Rep ; 21(6): e49708, 2020 06 04.
Article in English | MEDLINE | ID: mdl-32270911

ABSTRACT

The intestinal epithelium is a paradigm of adult tissue in constant regeneration that is supported by intestinal stem cells (ISCs). The mechanisms regulating ISC homeostasis after injury are poorly understood. We previously demonstrated that IκBα, the main regulator of NF-κB, exerts alternative nuclear functions as cytokine sensor in a subset of PRC2-regulated genes. Here, we show that nuclear IκBα is present in the ISC compartment. Mice deficient for IκBα show altered intestinal cell differentiation with persistence of a fetal-like ISC phenotype, associated with aberrant PRC2 activity at specific loci. Moreover, IκBα-deficient intestinal cells produce morphologically aberrant organoids carrying a PRC2-dependent fetal-like transcriptional signature. DSS treatment, which induces acute damage in the colonic epithelium of mice, results in a temporary loss of nuclear P-IκBα and its subsequent accumulation in early CD44-positive regenerating areas. Importantly, IκBα-deficient mice show higher resistance to damage, likely due to the persistent fetal-like ISC phenotype. These results highlight intestinal IκBα as a chromatin sensor of inflammation in the ISC compartment.


Subject(s)
Intestines , Stem Cells , Animals , Intestinal Mucosa , Mice , NF-KappaB Inhibitor alpha/genetics , Phenotype
6.
Nat Cancer ; 1(1): 122-135, 2020 01.
Article in English | MEDLINE | ID: mdl-35121836

ABSTRACT

E3 ligases and degrons, the sequences they recognize in target proteins, are key parts of the ubiquitin-mediated proteolysis system. There are several examples of alterations of these two components of the system that have a role in cancer. Here we uncover the landscape of the contribution of such alterations to tumorigenesis across cancer types. We first systematically identified new instances of degrons across the human proteome by using a random forest classifier and validated the functionality of a dozen of them, exploiting somatic mutations across >7,000 tumors. We detected signals of positive selection across known and new degron instances. Our results reveal that several oncogenes are frequently targeted by mutations that affect the sequence of their degrons or their cognate E3 ubiquitin ligases, causing an abnormal increase in their protein abundance. Overall, an important number of driver mutations across primary tumors affect either degrons or E3-ubiquitin ligases.


Subject(s)
Neoplasms , Ubiquitin , Humans , Mutation , Neoplasms/genetics , Proteolysis , Proteome/genetics , Ubiquitin/genetics , Ubiquitin-Protein Ligases/genetics
8.
Nat Commun ; 9(1): 2992, 2018 07 31.
Article in English | MEDLINE | ID: mdl-30065304

ABSTRACT

Delta ligands regulate Notch signaling in normal intestinal stem cells, while Jagged1 activates Notch in intestinal adenomas carrying active ß-catenin. We used the ApcMin/+ mouse model, tumor spheroid cultures, and patient-derived orthoxenografts to address this divergent ligand-dependent Notch function and its implication in disease. We found that intestinal-specific Jag1 deletion or antibody targeting Jag1 prevents tumor initiation in mice. Addiction to Jag1 is concomitant with the absence of Manic Fringe (MFNG) in adenoma cells, and its ectopic expression reverts Jag1 dependence. In 239 human colorectal cancer patient samples, MFNG imposes a negative correlation between Jag1 and Notch, being high Jag1 in the absence of MFNG predictive of poor prognosis. Jag1 antibody treatment reduces patient-derived tumor orthoxenograft growth without affecting normal intestinal mucosa. Our data provide an explanation to Jag1 dependence in cancer, and reveal that Jag1-Notch1 interference provides therapeutic benefit in a subset of colorectal cancer and FAP syndrome patients.


Subject(s)
Hexosyltransferases/metabolism , Intestinal Neoplasms/metabolism , Intestinal Neoplasms/pathology , Intracellular Signaling Peptides and Proteins/metabolism , Jagged-1 Protein/metabolism , Membrane Proteins/metabolism , Proteins/metabolism , Animals , Biomarkers, Tumor/metabolism , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Proliferation , Epithelial Cells/metabolism , Epithelial Cells/pathology , Glucosyltransferases , Humans , Ligands , Mice , Models, Biological , Prognosis , Receptor, Notch1/metabolism , Signal Transduction , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Stem Cells/metabolism , Transcription, Genetic
9.
Leukemia ; 32(10): 2211-2223, 2018 10.
Article in English | MEDLINE | ID: mdl-29511289

ABSTRACT

Cutaneous T-cell lymphomas (CTCLs) represent different subtypes of lymphoproliferative disorders with no curative therapies for the advanced forms of the disease (namely mycosis fungoides and the leukemic variant, Sézary syndrome). Molecular events leading to CTCL progression are heterogeneous, however recent DNA and RNA sequencing studies highlighted the importance of NF-κB and ß-catenin pathways. We here show that the kinase TAK1, known as essential in B-cell lymphoma, is constitutively activated in CTCL cells, but tempered by the MYPT1/PP1 phosphatase complex. Blocking PP1 activity, both pharmacologically and genetically, resulted in TAK1 hyperphosphorylation at residues T344, S389, T444, and T511, which have functional impact on canonical NF-κB signaling. Inhibition of TAK1 precluded NF-κB and ß-catenin signaling and induced apoptosis of CTCL cell lines and primary Sézary syndrome cells both in vitro and in vivo. Detection of phosphorylated TAK1 at T444 and T344 is associated with the presence of lymphoma in a set of 60 primary human samples correlating with NF-κB and ß-catenin activation. These results identified TAK1 as a potential biomarker and therapeutic target for CTCL therapy.


Subject(s)
Lymphoma, T-Cell, Cutaneous/metabolism , MAP Kinase Kinase Kinases/metabolism , NF-kappa B/metabolism , Signal Transduction/physiology , Skin Neoplasms/metabolism , beta Catenin/metabolism , Animals , Apoptosis/physiology , Cell Line, Tumor , Humans , Mice , Myosin-Light-Chain Phosphatase/metabolism , Phosphorylation/physiology , Receptors, Neuropeptide Y/metabolism , Sezary Syndrome/metabolism
10.
Development ; 142(1): 41-50, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25480918

ABSTRACT

Genetic data indicate that abrogation of Notch-Rbpj or Wnt-ß-catenin pathways results in the loss of the intestinal stem cells (ISCs). However, whether the effect of Notch is direct or due to the aberrant differentiation of the transit-amplifying cells into post-mitotic goblet cells is unknown. To address this issue, we have generated composite tamoxifen-inducible intestine-specific genetic mouse models and analyzed the expression of intestinal differentiation markers. Importantly, we found that activation of ß-catenin partially rescues the differentiation phenotype of Rbpj deletion mutants, but not the loss of the ISC compartment. Moreover, we identified Bmi1, which is expressed in the ISC and progenitor compartments, as a gene that is co-regulated by Notch and ß-catenin. Loss of Bmi1 resulted in reduced proliferation in the ISC compartment accompanied by p16(INK4a) and p19(ARF) (splice variants of Cdkn2a) accumulation, and increased differentiation to the post-mitotic goblet cell lineage that partially mimics Notch loss-of-function defects. Finally, we provide evidence that Bmi1 contributes to ISC self-renewal.


Subject(s)
Intestines/pathology , Polycomb Repressive Complex 1/metabolism , Proto-Oncogene Proteins/metabolism , Receptors, Notch/metabolism , Signal Transduction , Animals , Cell Compartmentation , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cyclin-Dependent Kinase Inhibitor p19/genetics , Cyclin-Dependent Kinase Inhibitor p19/metabolism , DNA Repair , Homeostasis , Immunoglobulin J Recombination Signal Sequence-Binding Protein/deficiency , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Intestines/abnormalities , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Polycomb Repressive Complex 1/deficiency , Polycomb Repressive Complex 1/genetics , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics , Receptors, Notch/deficiency , Transcriptional Activation/genetics , Wnt Proteins/metabolism , beta Catenin/metabolism
11.
Cancer Cell ; 24(2): 151-66, 2013 Aug 12.
Article in English | MEDLINE | ID: mdl-23850221

ABSTRACT

IκB proteins are the primary inhibitors of NF-κB. Here, we demonstrate that sumoylated and phosphorylated IκBα accumulates in the nucleus of keratinocytes and interacts with histones H2A and H4 at the regulatory region of HOX and IRX genes. Chromatin-bound IκBα modulates Polycomb recruitment and imparts their competence to be activated by TNFα. Mutations in the Drosophila IκBα gene cactus enhance the homeotic phenotype of Polycomb mutants, which is not counteracted by mutations in dorsal/NF-κB. Oncogenic transformation of keratinocytes results in cytoplasmic IκBα translocation associated with a massive activation of Hox. Accumulation of cytoplasmic IκBα was found in squamous cell carcinoma (SCC) associated with IKK activation and HOX upregulation.


Subject(s)
Chromatin/metabolism , I-kappa B Proteins/metabolism , Skin Neoplasms/metabolism , Animals , Cell Differentiation/genetics , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Chromatin/genetics , HEK293 Cells , Histones/genetics , Histones/metabolism , Humans , I-kappa B Proteins/genetics , Keratinocytes/cytology , Keratinocytes/metabolism , Mice , Mice, Inbred C57BL , NF-KappaB Inhibitor alpha , Signal Transduction , Skin Neoplasms/genetics , Skin Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...