Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 111
Filtrer
1.
Cell Metab ; 35(1): 134-149.e6, 2023 01 03.
Article de Anglais | MEDLINE | ID: mdl-36528023

RÉSUMÉ

Effective therapies are lacking for patients with advanced colorectal cancer (CRC). The CRC tumor microenvironment has elevated metabolic waste products due to altered metabolism and proximity to the microbiota. The role of metabolite waste in tumor development, progression, and treatment resistance is unclear. We generated an autochthonous metastatic mouse model of CRC and used unbiased multi-omic analyses to reveal a robust accumulation of tumoral ammonia. The high ammonia levels induce T cell metabolic reprogramming, increase exhaustion, and decrease proliferation. CRC patients have increased serum ammonia, and the ammonia-related gene signature correlates with altered T cell response, adverse patient outcomes, and lack of response to immune checkpoint blockade. We demonstrate that enhancing ammonia clearance reactivates T cells, decreases tumor growth, and extends survival. Moreover, decreasing tumor-associated ammonia enhances anti-PD-L1 efficacy. These findings indicate that enhancing ammonia detoxification can reactivate T cells, highlighting a new approach to enhance the efficacy of immunotherapies.


Sujet(s)
Ammoniac , Tumeurs colorectales , Animaux , Souris , Épuisement des cellules T , Lymphocytes T , Tumeurs colorectales/anatomopathologie , Immunothérapie , Microenvironnement tumoral
2.
Gynecol Oncol ; 165(3): 552-559, 2022 06.
Article de Anglais | MEDLINE | ID: mdl-35414426

RÉSUMÉ

OBJECTIVES: The "incessant ovulation" hypothesis links increased risk for tubo-ovarian high-grade serous carcinoma (HGSC) due to more ovulations and reduced risk conferred by pre-menopausal exposures like oral contraceptive use, multiparity, and breastfeeding. However, most women diagnosed with HGSC are postmenopausal, implying age is a major risk factor for HGSC. Our mouse model for HGSC, based on tamoxifen (TAM)-induced somatic inactivation of the Brca1, Trp53, Rb1, and Nf1 (BPRN) tumor suppressor genes in oviductal epithelium, recapitulates key genetic, histopathologic, and biological features of human HGSCs. We aimed to credential the model for future efforts to define biological and risk modification factors in HGSC pathogenesis. METHODS: BPRN mice were treated with TAM to induce tumors at defined ages and parity status. RESULTS: BPRN mice aged 9-months prior to tumor induction had markedly shorter survival than 6-8 week old mice induced to form tumors (median 46.5 weeks versus 61.5 weeks, log-rank test P = 0.0006). No significant differences in cancer phenotypes were observed between multiparous versus nulliparous BPRN mice. However, using a modified tumor model with one wild-type Nf1 allele (BPRNfl/+), nulliparous mice had more advanced tumors than multiparous mice (Mantel-Haenszel Chi-square test of association, P = 0.01). CONCLUSIONS: Our findings show aging is associated with significantly shortened survival post tumor induction in the BRPN model and multiparity delays development and/or progression of HGSC in certain genetic contexts. The findings support relevance of our mouse model to gain mechanistic insights into how known factors exert their protective effects and to test novel approaches for HGSC prevention.


Sujet(s)
Carcinomes , Cystadénocarcinome séreux , Tumeurs de l'ovaire , Vieillissement , Animaux , Transformation cellulaire néoplasique/anatomopathologie , Cystadénocarcinome séreux/anatomopathologie , Modèles animaux de maladie humaine , Femelle , Humains , Souris , Tumeurs de l'ovaire/anatomopathologie , Parité , Grossesse
3.
Oncoimmunology ; 11(1): 2052640, 2022.
Article de Anglais | MEDLINE | ID: mdl-35309733

RÉSUMÉ

Chronic inflammation and oncogenic pathway activation are key-contributing factors in colorectal cancer pathogenesis. However, colorectal intrinsic mechanisms linking these two factors in cancer development are poorly defined. Here, we show that intestinal epithelial cell (IEC)-specific deletion of Dot1l histone methyltransferase (Dot1lΔIEC ) reduced H3K79 dimethylation (H3K79me2) in IECs and inhibited intestinal tumor formation in ApcMin - and AOM-DSS-induced colorectal cancer models. IEC-Dot1l abrogation was accompanied by alleviative colorectal inflammation and reduced Wnt/ß-catenin signaling activation. Mechanistically, Dot1l deficiency resulted in an increase in Foxp3+RORϒ+ regulatory T (Treg) cells and a decrease in inflammatory Th17 and Th22 cells, thereby reducing local inflammation in the intestinal tumor microenvironment. Furthermore, Dot1l deficiency caused a reduction of H3K79me2 occupancies in the promoters of the Wnt/ß-catenin signaling genes, thereby diminishing Wnt/ß-catenin oncogenic signaling pathway activation in colorectal cancer cells. Clinically, high levels of tumor H3K79me2 were detected in patients with colorectal carcinomas as compared to adenomas, and negatively correlated with RORϒ+FOXP3+ Treg cells. Altogether, we conclude that DOT1L is an intrinsic molecular node connecting chronic immune activation and oncogenic signaling pathways in colorectal cancer. Our work suggests that targeting the DOT1L pathway may control colorectal carcinogenesis. Significance: IEC-intrinsic DOT1L controls T cell subset balance and key oncogenic pathway activation, impacting colorectal carcinogenesis.


Sujet(s)
Tumeurs colorectales , Histone-lysine N-methyltransferase , Sous-populations de lymphocytes T , Carcinogenèse/métabolisme , Tumeurs colorectales/anatomopathologie , Facteurs de transcription Forkhead/métabolisme , Histone-lysine N-methyltransferase/génétique , Histone-lysine N-methyltransferase/métabolisme , Humains , Inflammation , Sous-populations de lymphocytes T/métabolisme , Sous-populations de lymphocytes T/anatomopathologie , Microenvironnement tumoral , Voie de signalisation Wnt , bêta-Caténine/génétique , bêta-Caténine/métabolisme
4.
Cancer Cell ; 40(2): 185-200.e6, 2022 02 14.
Article de Anglais | MEDLINE | ID: mdl-34951957

RÉSUMÉ

Microbial dysbiosis is a colorectal cancer (CRC) hallmark and contributes to inflammation, tumor growth, and therapy response. Gut microbes signal via metabolites, but how the metabolites impact CRC is largely unknown. We interrogated fecal metabolites associated with mouse models of colon tumorigenesis with varying mutational load. We find that microbial metabolites from healthy mice or humans are growth-repressive, and this response is attenuated in mice and patients with CRC. Microbial profiling reveals that Lactobacillus reuteri and its metabolite, reuterin, are downregulated in mouse and human CRC. Reuterin alters redox balance, and reduces proliferation and survival in colon cancer cells. Reuterin induces selective protein oxidation and inhibits ribosomal biogenesis and protein translation. Exogenous Lactobacillus reuteri restricts colon tumor growth, increases tumor reactive oxygen species, and decreases protein translation in vivo. Our findings indicate that a healthy microbiome and specifically, Lactobacillus reuteri, is protective against CRC through microbial metabolite exchange.


Sujet(s)
Tumeurs colorectales/métabolisme , Tumeurs colorectales/anatomopathologie , Microbiome gastro-intestinal , Glycéraldéhyde/analogues et dérivés , Oxydoréduction , Propane/métabolisme , Animaux , Marqueurs biologiques , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine , Métabolisme énergétique , Glutathion/métabolisme , Glycéraldéhyde/métabolisme , Glycéraldéhyde/pharmacologie , Interactions hôte-microbes , Humains , Muqueuse intestinale/métabolisme , Muqueuse intestinale/microbiologie , Muqueuse intestinale/anatomopathologie , Métabolomique/méthodes , Métagénomique/méthodes , Souris , Modèles biologiques , Oxydoréduction/effets des médicaments et des substances chimiques , Stress oxydatif , Propane/pharmacologie , Transduction du signal , Tests d'activité antitumorale sur modèle de xénogreffe
5.
Cancer Res ; 81(12): 3309-3318, 2021 06 15.
Article de Anglais | MEDLINE | ID: mdl-33863776

RÉSUMÉ

Studies have shown bacteria influence the initiation and progression of cancers arising in sites that harbor rich microbial communities, such as the colon. Little is known about the potential for the microbiome to influence tumorigenesis at sites considered sterile, including the upper female genital tract. The recent identification of distinct bacterial signatures associated with ovarian carcinomas suggests microbiota in the gut, vagina, or elsewhere might contribute to ovarian cancer pathogenesis. Here, we tested whether altering the microbiome affects tumorigenesis in a mouse model of high-grade serous carcinoma (HGSC) based on conditional oviduct-specific inactivation of the Brca1, Trp53, Rb1, and Nf1 tumor suppressor genes. Cohorts of control (n = 20) and antibiotic-treated (n = 23) mice were treated with tamoxifen to induce tumor formation and then monitored for 12 months. The antibiotic cocktail was administered for the first 5 months of the monitoring period in the treatment group. Antibiotic-treated mice had significantly fewer and less advanced tumors than control mice at study endpoint. Antibiotics induced changes in the composition of the intestinal and vaginal microbiota, which were durable in the fecal samples. Clustering analysis showed particular groups of microbiota are associated with the development of HGSC in this model. These findings demonstrate the microbiome influences HGSC pathogenesis in an in vivo model that closely recapitulates the human disease. Because the microbiome can modulate efficacy of cancer chemo- and immunotherapy, our genetically engineered mouse model system may prove useful for testing whether altering the microbiota can improve the heretofore poor response of HGSC to immunotherapies. SIGNIFICANCE: This study provides strong in vivo evidence for a role of the microbiome in ovarian cancer pathogenesis.


Sujet(s)
Antibactériens/pharmacologie , Transformation cellulaire néoplasique/effets des médicaments et des substances chimiques , Cystadénocarcinome séreux/prévention et contrôle , Modèles animaux de maladie humaine , Microbiote/effets des médicaments et des substances chimiques , Tumeurs de l'ovaire/prévention et contrôle , Oviductes/effets des médicaments et des substances chimiques , Animaux , Transformation cellulaire néoplasique/anatomopathologie , Cystadénocarcinome séreux/microbiologie , Cystadénocarcinome séreux/anatomopathologie , Femelle , Humains , Souris , Tumeurs de l'ovaire/microbiologie , Tumeurs de l'ovaire/anatomopathologie , Oviductes/microbiologie , Oviductes/anatomopathologie
6.
Cancer Res ; 80(4): 877-889, 2020 02 15.
Article de Anglais | MEDLINE | ID: mdl-31806642

RÉSUMÉ

Robust preclinical models of ovarian high-grade serous carcinoma (HGSC) are needed to advance our understanding of HGSC pathogenesis and to test novel strategies aimed at improving clinical outcomes for women with the disease. Genetically engineered mouse models of HGSC recapitulating the likely cell of origin (fallopian tube), underlying genetic defects, histology, and biologic behavior of human HGSCs have been developed. However, the degree to which the mouse tumors acquire the somatic genomic changes, gene expression profiles, and immune microenvironment that characterize human HGSCs remains unclear. We used integrated molecular characterization of oviductal HGSCs arising in the context of Brca1, Trp53, Rb1, and Nf1 (BPRN) inactivation to determine whether the mouse tumors recapitulate human HGSCs across multiple domains of molecular features. Targeted DNA sequencing showed the mouse BPRN tumors, but not endometrioid carcinoma-like tumors based on different genetic defects (e.g., Apc and Pten), acquire somatic mutations and widespread copy number alterations similar to those observed in human HGSCs. RNA sequencing showed the mouse HGSCs most closely resemble the so-called immunoreactive and mesenchymal subsets of human HGSCs. A combined immuno-genomic analysis demonstrated the immune microenvironment of BPRN tumors models key aspects of tumor-immune dynamics in the immunoreactive and mesenchymal subtypes of human HGSC, with enrichment of immunosuppressive cell subsets such as myeloid-derived suppressor cells and regulatory T cells. The findings further validate the BPRN model as a robust preclinical experimental platform to address current barriers to improved prevention, diagnosis, and treatment of this often lethal cancer. SIGNIFICANCE: The acquired gene mutations, broad genomic alterations, and gene expression and immune cell-tumor axis changes in a mouse model of oviductal serous carcinoma closely mirror those of human tubo-ovarian high-grade serous carcinoma.


Sujet(s)
Cystadénocarcinome séreux/génétique , Tumeurs de la trompe de Fallope/génétique , Neurofibromine-1/génétique , Tumeurs de l'ovaire/génétique , Microenvironnement tumoral/immunologie , Animaux , Carcinome endométrioïde/génétique , Carcinome endométrioïde/immunologie , Carcinome endométrioïde/anatomopathologie , Cystadénocarcinome séreux/immunologie , Cystadénocarcinome séreux/anatomopathologie , Modèles animaux de maladie humaine , Tumeurs de la trompe de Fallope/immunologie , Tumeurs de la trompe de Fallope/anatomopathologie , Trompes utérines/anatomopathologie , Femelle , Régulation de l'expression des gènes tumoraux , Humains , Souris , Souris transgéniques , Cellules myéloïdes suppressives/immunologie , Tumeurs de l'ovaire/immunologie , RNA-Seq , Lymphocytes T régulateurs/immunologie
7.
Sci Rep ; 9(1): 19180, 2019 12 16.
Article de Anglais | MEDLINE | ID: mdl-31844152

RÉSUMÉ

Cancer cells in culture rely on glutamine as an anaplerotic substrate to replenish tricarboxylic acid (TCA) cycle intermediates that have been consumed. but it is uncertain whether cancers in vivo depend on glutamine for anaplerosis. Here, following in vivo infusions of [13C5]-glutamine in mice bearing subcutaneous colon cancer xenografts, we showed substantial amounts of infused [13C5]-glutamine enters the TCA cycle in the tumors. Consistent with our prior observation that colorectal cancers (CRCs) with oncogenic mutations in the phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic (PIK3CA) subunit are more dependent on glutamine than CRCs with wild type PIK3CA, labeling from glutamine to most TCA cycle intermediates was higher in PIK3CA-mutant subcutaneous xenograft tumors than in wild type PIK3CA tumors. Moreover, using orthotopic mouse colon tumors estalished from human CRC cells or patient-derived xenografts, we demonstrated substantial amounts of infused [13C5]-glutamine enters the TCA cycle in the tumors and tumors utilize anaplerotic glutamine to a greater extent than adjacent normal colon tissues. Similar results were seen in spontaneous colon tumors arising in genetically engineered mice. Our studies provide compelling evidence CRCs utilizes glutamine to replenish the TCA cycle in vivo, suggesting that targeting glutamine metabolism could be a therapeutic approach for CRCs, especially for PIK3CA-mutant CRCs.


Sujet(s)
Cycle citrique , Tumeurs colorectales/métabolisme , Glutamine/métabolisme , Animaux , Isotopes du carbone/sang , Phosphatidylinositol 3-kinases de classe I/génétique , Tumeurs colorectales/sang , Femelle , Glutamine/sang , Cellules HCT116 , Humains , Cinétique , Souris nude , Mutation/génétique , Tissu sous-cutané/anatomopathologie , Spécificité du substrat , Tests d'activité antitumorale sur modèle de xénogreffe
8.
Curr Biol ; 29(21): R1140-R1142, 2019 11 04.
Article de Anglais | MEDLINE | ID: mdl-31689402

RÉSUMÉ

A recent paper demonstrates how tissue context impacts the breast cancer cell phenotype. Loss of the E-cadherin tumor suppressor protein enhanced cell invasion, but inhibited multiple steps in metastatic spread due to the accumulation of reactive oxygen species and induction of apoptosis.


Sujet(s)
Tumeurs du sein , Apoptose , Cadhérines , Humains , Invasion tumorale , Protéines suppresseurs de tumeurs
9.
Neoplasia ; 21(10): 1051-1062, 2019 10.
Article de Anglais | MEDLINE | ID: mdl-31521947

RÉSUMÉ

Treatment of locally advanced rectal cancer includes chemotherapy, radiation, and surgery but patient responses to neoadjuvant treatment are variable. We have shown that rectal tumors are comprised of multiple genetically distinct sub-clones. Unique sub-clones within tumors may harbor mutations which contribute to inter-patient variation in response to neoadjuvant chemoradiotherapy (nCRT). Analysis of the influence of nCRT on the extent and nature of intra-tumoral genetic heterogeneity in rectal cancer may provide insights into mechanisms of resistance. Locally advanced rectal cancer patients underwent pre-treatment biopsies. At the time of surgery, tissue from the treated tumor was obtained and analyzed. Pre- and post-treatment specimens were subjected to whole exome and confirmatory deep sequencing for somatic mutations. Copy number variation was assessed using OncoScan SNP arrays. Genomic data were analyzed using PyClone to identify sub-clonal tumor population following nCRT. Alterations that persisted or were enriched in the post-treatment tumor specimen following nCRT were defined for each patient. Thirty-two samples were obtained from ten patients. PyClone identified 2 to 10 genetic sub-clones per tumor. Substantial changes in the proportions of individual sub-clones in pre- versus post-treatment tumor material were found in all patients. Resistant sub-clones recurrently contained mutations in TP53, APC, ABCA13, MUC16, and THSD4. Recurrent copy number variation was observed across multiple chromosome regions after nCRT. Pathway analysis including variant alleles and copy number changes associated with resistant sub-clones revealed significantly altered pathways, especially those linked to the APC and TP53 genes, which were the two most frequently mutated genes. Intra-tumoral heterogeneity is evident in pre-treatment rectal cancer. Following treatment, sub-clonal populations are selectively modified and enrichment of a subset of pre-treatment sub-clones is seen. Further studies are needed to define recurrent alterations at diagnosis that may contribute to resistance to nCRT.


Sujet(s)
Antinéoplasiques/pharmacologie , Évolution clonale/effets des médicaments et des substances chimiques , Évolution clonale/génétique , Hétérogénéité génétique , Tumeurs du rectum/génétique , Adulte , Sujet âgé , Antinéoplasiques/usage thérapeutique , Marqueurs biologiques tumoraux , Chimioradiothérapie , Résistance aux médicaments antinéoplasiques , Femelle , Humains , Mâle , Adulte d'âge moyen , Mutation , Traitement néoadjuvant , Grading des tumeurs , Stadification tumorale , Tumeurs du rectum/métabolisme , Tumeurs du rectum/anatomopathologie , Tumeurs du rectum/thérapie , Transduction du signal , Résultat thérapeutique ,
10.
Cancer Lett ; 462: 1-11, 2019 Oct 10.
Article de Anglais | MEDLINE | ID: mdl-31351087

RÉSUMÉ

Colorectal cancer (CRC) remains one of the leading causes of mortality worldwide. Drug repositioning is a promising approach for new cancer therapies, as it provides the opportunity to rapidly advance potentially promising agents into clinical trials. The FDA-approved anti-helminthic drug rafoxanide was recently reported to antagonize the oncogenic function of the BRAF V600E mutant protein, commonly found in CRCs, as well as to inhibit the proliferation of skin cancer cells. These observations prompted us to investigate the potential anti-cancer effects of rafoxanide in CRC models. We found rafoxanide inhibited proliferation in CRC cells, but not in normal colonic epithelial cells. Rafoxanide's anti-proliferative action was associated with marked reduction in cyclin D1 protein levels and accumulation of cells in the G0/G1 phase. These effects relied on selective induction of the endoplasmic reticulum stress (ERS) response in CRC cells and were followed by caspase-dependent cell death. Systemic administration of rafoxanide to Apcmin/+ mice induced to develop CRCs caused ERS activation, proliferation inhibition and apoptosis induction in the neoplastic cells. Collectively, our data suggest rafoxanide might be repurposed as an anti-cancer drug for the treatment of CRC.


Sujet(s)
Antihelminthiques antinématodes/pharmacologie , Tumeurs du côlon/prévention et contrôle , Tumeurs colorectales/traitement médicamenteux , Stress du réticulum endoplasmique/effets des médicaments et des substances chimiques , Rafoxanide/pharmacologie , Sujet âgé , Animaux , Apoptose , Oxyde de diméthyl-diazène/toxicité , Cancérogènes/toxicité , Prolifération cellulaire , Tumeurs du côlon/induit chimiquement , Tumeurs du côlon/métabolisme , Tumeurs du côlon/anatomopathologie , Tumeurs colorectales/métabolisme , Tumeurs colorectales/anatomopathologie , Femelle , Humains , Mâle , Souris , Cellules cancéreuses en culture
11.
J Biol Chem ; 294(32): 12077-12090, 2019 08 09.
Article de Anglais | MEDLINE | ID: mdl-31213529

RÉSUMÉ

Unlike most other tissues, the colon epithelium is exposed to high levels of H2S derived from gut microbial metabolism. H2S is a signaling molecule that modulates various physiological effects. It is also a respiratory toxin that inhibits complex IV in the electron transfer chain (ETC). Colon epithelial cells are adapted to high environmental H2S exposure as they harbor an efficient mitochondrial H2S oxidation pathway, which is dedicated to its disposal. Herein, we report that the sulfide oxidation pathway enzymes are apically localized in human colonic crypts at the host-microbiome interface, but that the normal apical-to-crypt gradient is lost in colorectal cancer epithelium. We found that sulfide quinone oxidoreductase (SQR), which catalyzes the committing step in the mitochondrial sulfide oxidation pathway and couples to complex III, is a critical respiratory shield against H2S poisoning. H2S at concentrations ≤20 µm stimulated the oxygen consumption rate in colon epithelial cells, but, when SQR expression was ablated, H2S concentrations as low as 5 µm poisoned cells. Mitochondrial H2S oxidation altered cellular bioenergetics, inducing a reductive shift in the NAD+/NADH redox couple. The consequent electron acceptor insufficiency caused uridine and aspartate deficiency and enhanced glutamine-dependent reductive carboxylation. The metabolomic signature of this H2S-induced stress response mapped, in part, to redox-sensitive nodes in central carbon metabolism. Colorectal cancer tissues and cell lines appeared to counter the growth-restricting effects of H2S by overexpressing sulfide oxidation pathway enzymes. Our findings reveal an alternative mechanism for H2S signaling, arising from alterations in mitochondrial bioenergetics that drive metabolic reprogramming.


Sujet(s)
Métabolisme énergétique , Sulfure d'hydrogène/métabolisme , Mitochondries/métabolisme , Lignée cellulaire , Prolifération cellulaire/effets des médicaments et des substances chimiques , Côlon/cytologie , Côlon/métabolisme , Côlon/anatomopathologie , Tumeurs du côlon/métabolisme , Tumeurs du côlon/anatomopathologie , Cystéine/composition chimique , Cystéine/métabolisme , Métabolisme énergétique/effets des médicaments et des substances chimiques , Humains , Sulfure d'hydrogène/composition chimique , Sulfure d'hydrogène/pharmacologie , NAD/composition chimique , Oxydoréduction , Consommation d'oxygène/effets des médicaments et des substances chimiques , Quinone reductases/antagonistes et inhibiteurs , Quinone reductases/génétique , Quinone reductases/métabolisme , Interférence par ARN , Petit ARN interférent/métabolisme
12.
Lab Invest ; 99(10): 1454-1469, 2019 10.
Article de Anglais | MEDLINE | ID: mdl-31148594

RÉSUMÉ

Somatic APC (adenomatous polyposis coli), TP53, KRAS mutations are present in roughly 80%, 60%, and 40%, respectively, of human colorectal cancers (CRCs). Most TP53 mutant alleles in CRCs encode missense mutant proteins with loss-of-function (LOF) of p53's transcriptional activity and dominant negative (DN) effects on wild-type p53 function. Missense mutant p53 proteins have been reported to exert gain-of-function (GOF) effects in cancer. We compared the phenotypic effects of the common human cancer-associated TP53 R273H missense mutation to p53 null status in a genetically engineered mouse CRC model. Inactivation of one allele of Apc together with activation of a Kras mutant allele in mouse colon epithelium instigated development of serrated and hyperplastic epithelium and adenomas (AK mice). Addition of a Trp53R270H or Trp53null mutant allele to the model (AKP mice) led to markedly shortened survival and increased tumor burden relative to that of AK mice, including adenocarcinomas in AKP mice. Comparable life span and tumor burden were seen in AKP mice carrying Trp53R270H or Trp53null alleles, along with similar frequencies of spontaneous metastasis to lymph nodes, lung, and liver. The fraction of adenocarcinomas with submucosa or deeper invasion was higher in AKP270/fl mice than in AKPfl/fl mice, but the incidence of adenocarcinomas per mouse did not differ significantly between AKPfl/fl and AKP270/fl mice. In line with their comparable biological behaviors, mouse primary tumors and tumor-derived organoids with the Trp53R270H or Trp53null alleles had highly similar gene expression profiles. Human CRCs with TP53 R273 missense mutant or null alleles also had essentially homogeneous gene expression patterns. Our findings indicate the R270H/R273H p53 mutant protein does not manifest definite GOF biological effects in mouse and human CRCs, suggesting possible GOF effects of mutant p53 in cancer phenotypes are likely allele-specific and/or context-dependent.


Sujet(s)
Adénocarcinome/génétique , Tumeurs colorectales/génétique , Protéine p53 suppresseur de tumeur/génétique , Animaux , Carcinogenèse , Évolution de la maladie , Transition épithélio-mésenchymateuse , Expression des gènes , Humains , Souris transgéniques , Mutation faux-sens , Invasion tumorale , Métastase tumorale
13.
J Pathol ; 249(2): 206-214, 2019 10.
Article de Anglais | MEDLINE | ID: mdl-31131879

RÉSUMÉ

Most high-grade serous carcinomas are thought to arise from Fallopian tube epithelium (FTE), but some likely arise outside of the tube, perhaps from ectopic tubal-type epithelium known as endosalpingiosis. Importantly, the origin of endosalpingiosis is poorly understood. The proximity of the tubal fimbriae to the ovaries has led to the proposal that disruptions in the ovarian surface that occur during ovulation may allow detached FTE to implant in the ovary and form tubal-type glands and cysts. An alternative model suggests that cells present in ectopic locations outside the Müllerian tract retain the capacity for multi-lineage differentiation and can form glands with tubal-type epithelium. We used double transgenic Ovgp1-iCreERT2 ;R26RLSL-eYFP mice, which express an eYFP reporter protein in OVGP1-positive tissues following transient tamoxifen (TAM) treatment, to track the fate of oviductal epithelial cells. Cohorts of adult mice were given TAM to activate eYFP expression in oviductal epithelium, and ovaries were examined at time points ranging from 2 days to 12 months post-TAM. To test whether superovulation might increase acquisition of endosalpingiosis, additional cohorts of TAM-treated mice underwent up to five cycles of superovulation and ovaries were examined at 1, 6, and 12 months post-TAM. Ovaries were sectioned in their entirety to identify endosalpingiosis. Immunohistochemical staining for PAX8, tubulin, OVGP1, and eYFP was employed to study endosalpingiosis lesions. Ovarian endosalpingiosis was identified in 14.2% of TAM-treated adult mice. The endosalpingiotic inclusion glands and cysts were lined by secretory and ciliated cells and expressed PAX8, tubulin, OVGP1, and eYFP. Neither age nor superovulation was associated with a significant increase in endosalpingiosis. Endosalpingiosis was also occasionally present in the ovaries of pre-pubertal mice. The findings imply that ovarian endosalpingiosis in the mouse does not likely arise as a consequence of detachment and implantation of tubal epithelium and other mechanisms may be relevant. © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Sujet(s)
Lignage cellulaire , Cellules épithéliales/anatomopathologie , Tumeurs de la trompe de Fallope/anatomopathologie , Trompes utérines/anatomopathologie , Tumeurs kystiques, mucineuses et séreuses/anatomopathologie , Tumeurs de l'ovaire/anatomopathologie , Ovaire/anatomopathologie , Animaux , Protéines bactériennes/biosynthèse , Protéines bactériennes/génétique , Suivi cellulaire/méthodes , Cellules épithéliales/métabolisme , Tumeurs de la trompe de Fallope/génétique , Tumeurs de la trompe de Fallope/métabolisme , Trompes utérines/métabolisme , Femelle , Glycoprotéines/génétique , Integrases/génétique , Protéines luminescentes/biosynthèse , Protéines luminescentes/génétique , Souris transgéniques , Tumeurs kystiques, mucineuses et séreuses/génétique , Tumeurs kystiques, mucineuses et séreuses/métabolisme , Tumeurs de l'ovaire/génétique , Tumeurs de l'ovaire/métabolisme , Ovaire/métabolisme , Facteur de transcription PAX-8/métabolisme , ARN non traduit/génétique , Superovulation , Tubuline/métabolisme
14.
Hum Pathol ; 87: 103-114, 2019 05.
Article de Anglais | MEDLINE | ID: mdl-30716341

RÉSUMÉ

Colorectal cancers (CRCs) initiate through distinct mutations, including in APC pathway components leading to tubular adenomas (TAs); in BRAF, with epigenetic silencing of CDX2, leading to serrated adenomas (SAs); and in the DNA mismatch repair machinery driving microsatellite instability (MSI). Transformation through the APC pathway involves loss of the hormone GUCA2A that silences the tumor-suppressing receptor GUCY2C. Indeed, oral hormone replacement is an emerging strategy to reactivate GUCY2C and prevent CRC initiation and progression. Moreover, retained expression by tumors arising from TAs has established GUCY2C as a diagnostic and therapeutic target to prevent and treat metastatic CRC. Here, we defined the potential role of the GUCA2A-GUCY2C axis and its suitability as a target in tumors arising through the SA and MSI pathways. GUCA2A hormone expression was eliminated in TAs, SAs, and MSI tumors compared to their corresponding normal adjacent tissues. In contrast to the hormone, the tumor-suppressing receptor GUCY2C was retained in TA and MSI tumors. Surprisingly, GUCY2C expression was nearly eliminated in SAs, reflecting loss of the transcription factor CDX2. Changes in the GUCA2A-GUCY2C axis in human SAs and MSI tumors were precisely recapitulated in genetic mouse models. These data reveal the possibility of GUCA2A loss silencing GUCY2C in the pathophysiology of, and oral hormone replacement to restore GUCY2C signaling to prevent, MSI tumors. Also, they highlight the potential for targeting GUCY2C to prevent and treat metastases arising from TA and MSI tumors. In contrast, loss of GUCY2C excludes patients with SAs as candidates for GUCY2C-based prevention and therapy.


Sujet(s)
Adénomes/génétique , Tumeurs colorectales/génétique , Récepteurs des entérotoxines/génétique , Adénomes/anatomopathologie , Adulte , Sujet âgé , Animaux , Tumeurs colorectales/anatomopathologie , Femelle , Humains , Mâle , Souris , Souris transgéniques , Adulte d'âge moyen , Transduction du signal
15.
Lab Invest ; 99(1): 128-137, 2019 01.
Article de Anglais | MEDLINE | ID: mdl-30177831

RÉSUMÉ

The Wnt/ß-catenin signaling pathway is dysregulated in different types of neoplasms including colorectal cancer (CRC). Aberrant activation of this signaling pathway is a key early event in the development of colorectal neoplasms, and is mainly caused by loss of function mutations in Adenomatous Polyposis Coli (APC), and less frequently by ß-catenin stabilization mutations via missense or interstitial genomic deletions in CTNNB1. In this study, we have defined an immunohistochemical algorithm to dissect Wnt pathway alterations in formalin-fixed and paraffin-embedded neoplastic tissues. Basically, consecutive sections of tumor specimens were stained by immunohistochemistry with two different monoclonal antibodies against ß-catenin: one (anti-active ß-catenin antibody) recognizes hypo-phosphorylated ß-catenin and the other recognizes the total pool of ß-catenin. We validated the strategy in the HCT116 CRC cell line which has an in-frame deletion of ß-catenin serine 45, and then studied human tumor microarrays containing colon adenomas, CRCs, solid pseudopapillary neoplasms of the pancreas as well as the whole tissue sections of CRCs, desmoid fibromatosis, and pilomatrixoma of the skin. In some tumors, we found strong ß-catenin cytoplasmic and/or nuclear staining with the total ß-catenin antibody but no staining with the anti-active ß-catenin antibody. This was inferred to be an altered/mutant ß-catenin staining pattern. All six colon adenomas of the 126 total adenomas studied for the altered/mutant ß-catenin staining pattern had presumptively pathogenic point mutations or deletions in CTNNB1. Four of 10 CRCs with the alterated/mutant ß-catenin staining pattern studied in depth, from 181 total CRCs from tissue microarray, had pathogenic CTNNB1 mutations. The frequencies of CTNNB1 alterations in non-colonic tumors with altered/mutant ß-catenin staining ranged between 46 and 100%. Our results demonstrate that the immunohistochemical approach described here can detect oncogenic forms of ß-catenin in primary tissue samples and can also highlight other tumors with presumptive novel defects activating the Wnt/ß-catenin pathway.


Sujet(s)
Immunohistochimie/méthodes , Tumeurs/génétique , Voie de signalisation Wnt , bêta-Caténine/génétique , Polypes coliques/composition chimique , Cellules HCT116 , Humains , Tumeurs/composition chimique
16.
Trans Am Clin Climatol Assoc ; 129: 56-62, 2018.
Article de Anglais | MEDLINE | ID: mdl-30166699

RÉSUMÉ

Colorectal cancers (CRCs) harbor accumulated defects in key signaling pathways that regulate cell phenotypes, including proliferation, survival, metabolism, and differentiation. To study the functional contributions of the accumulated molecular defects in CRC, we have developed approaches to inactivate selected tumor suppressor and/or activate oncogenes in mouse colon epithelium. Conditional inactivation of the CDX2 tumor suppressor protein in conjunction with oncogenic activation of the BRAF protein promotes development of serrated glandular benign and malignant tumors in the mouse colon. The mouse tumors share significant morphological and molecular relationships with the 8% to 10% of human CRCs that manifest serrated morphology at diagnosis. The gene and protein expression patterns in the mouse tumors have informed understanding of the relationships between benign and malignant human serrated colon tumors. Our findings are consistent with prior work suggesting that perhaps upwards of one-third of human CRCs may arise from a precursor lesion with serrated morphology rather than a conventional adenoma.


Sujet(s)
Marqueurs biologiques tumoraux/génétique , Transformation cellulaire néoplasique/génétique , Tumeurs colorectales/génétique , Tumeurs expérimentales/génétique , Animaux , Marqueurs biologiques tumoraux/métabolisme , Transformation cellulaire néoplasique/métabolisme , Transformation cellulaire néoplasique/anatomopathologie , Tumeurs colorectales/métabolisme , Tumeurs colorectales/anatomopathologie , Régulation de l'expression des gènes tumoraux , Prédisposition génétique à une maladie , Humains , Souris , Mutation , Tumeurs expérimentales/métabolisme , Tumeurs expérimentales/anatomopathologie , Phénotype , Pronostic , Facteurs de risque , Transduction du signal
17.
J Biol Chem ; 293(8): 2675-2686, 2018 02 23.
Article de Anglais | MEDLINE | ID: mdl-29348167

RÉSUMÉ

Rhodanese domains are structural modules present in the sulfurtransferase superfamily. These domains can exist as single units, in tandem repeats, or fused to domains with other activities. Despite their prevalence across species, the specific physiological roles of most sulfurtransferases are not known. Mammalian rhodanese and mercaptopyruvate sulfurtransferase are perhaps the best-studied members of this protein superfamily and are involved in hydrogen sulfide metabolism. The relatively unstudied human thiosulfate sulfurtransferase-like domain-containing 1 (TSTD1) protein, a single-domain cytoplasmic sulfurtransferase, was also postulated to play a role in the sulfide oxidation pathway using thiosulfate to form glutathione persulfide, for subsequent processing in the mitochondrial matrix. Prior kinetic analysis of TSTD1 was performed at pH 9.2, raising questions about relevance and the proposed model for TSTD1 function. In this study, we report a 1.04 Å resolution crystal structure of human TSTD1, which displays an exposed active site that is distinct from that of rhodanese and mercaptopyruvate sulfurtransferase. Kinetic studies with a combination of sulfur donors and acceptors reveal that TSTD1 exhibits a low Km for thioredoxin as a sulfane sulfur acceptor and that it utilizes thiosulfate inefficiently as a sulfur donor. The active site exposure and its interaction with thioredoxin suggest that TSTD1 might play a role in sulfide-based signaling. The apical localization of TSTD1 in human colonic crypts, which interfaces with sulfide-releasing microbes, and the overexpression of TSTD1 in colon cancer provide potentially intriguing clues as to its role in sulfide metabolism.


Sujet(s)
Modèles moléculaires , NADP/métabolisme , Protéines tumorales/métabolisme , Thioredoxin-disulfide reductase/métabolisme , Thiorédoxines/métabolisme , Thiosulfate sulfurtransferase/métabolisme , Substitution d'acide aminé , Animaux , Biocatalyse , Domaine catalytique , Côlon/enzymologie , Côlon/métabolisme , Côlon/anatomopathologie , Tumeurs colorectales/enzymologie , Tumeurs colorectales/métabolisme , Tumeurs colorectales/anatomopathologie , Cristallographie aux rayons X , Bases de données de protéines , Humains , Muqueuse intestinale/enzymologie , Muqueuse intestinale/métabolisme , Muqueuse intestinale/anatomopathologie , Mutation , Protéines tumorales/composition chimique , Protéines tumorales/génétique , Conformation des protéines , Motifs et domaines d'intéraction protéique , Protéines recombinantes/composition chimique , Protéines recombinantes/métabolisme , Protéines de Saccharomyces cerevisiae/composition chimique , Protéines de Saccharomyces cerevisiae/génétique , Protéines de Saccharomyces cerevisiae/métabolisme , Spécificité du substrat , Thioredoxin-disulfide reductase/composition chimique , Thiorédoxines/composition chimique , Thiorédoxines/génétique , Thiosulfate sulfurtransferase/composition chimique , Thiosulfate sulfurtransferase/génétique
18.
JCI Insight ; 3(2)2018 01 25.
Article de Anglais | MEDLINE | ID: mdl-29367463

RÉSUMÉ

Pancreatic cancer is characterized by nearly universal activating mutations in KRAS. Among other somatic mutations, TP53 is mutated in more than 75% of human pancreatic tumors. Genetically engineered mice have proven instrumental in studies of the contribution of individual genes to carcinogenesis. Oncogenic Kras mutations occur early during pancreatic carcinogenesis and are considered an initiating event. In contrast, mutations in p53 occur later during tumor progression. In our model, we recapitulated the order of mutations of the human disease, with p53 mutation following expression of oncogenic Kras. Further, using an inducible and reversible expression allele for mutant p53, we inactivated its expression at different stages of carcinogenesis. Notably, the function of mutant p53 changes at different stages of carcinogenesis. Our work establishes a requirement for mutant p53 for the formation and maintenance of pancreatic cancer precursor lesions. In tumors, mutant p53 becomes dispensable for growth. However, it maintains the altered metabolism that characterizes pancreatic cancer and mediates its malignant potential. Further, mutant p53 promotes epithelial-mesenchymal transition (EMT) and cancer cell invasion. This work generates new mouse models that mimic human pancreatic cancer and expands our understanding of the role of p53 mutation, common in the majority of human malignancies.


Sujet(s)
Carcinogenèse/génétique , Carcinome du canal pancréatique/génétique , Tumeurs du pancréas/génétique , Protéine p53 suppresseur de tumeur/génétique , Animaux , Carcinome du canal pancréatique/anatomopathologie , Lignée cellulaire tumorale , Modèles animaux de maladie humaine , Évolution de la maladie , Humains , Souris , Souris transgéniques , Mutation , Invasion tumorale/génétique , Invasion tumorale/anatomopathologie , Pancréas/anatomopathologie , Tumeurs du pancréas/anatomopathologie , Protéines proto-oncogènes p21(ras)/génétique
19.
Clin Cancer Res ; 24(9): 2214-2224, 2018 05 01.
Article de Anglais | MEDLINE | ID: mdl-29203589

RÉSUMÉ

Purpose: Recent studies have highlighted the existence of subclones in tumors. Lymph nodes are generally the first location of metastasis for most solid epithelial tumors, including colorectal cancer. We sought to understand the genetic origin of lymph node metastasis in colorectal cancer by evaluating the relationship between colorectal cancer subclones present in primary tumors and lymph nodes.Experimental Design: A total of 33 samples from seven colorectal cancers, including two or three spatially disparate regions from each primary tumor and one to four matched lymph nodes for each tumor, underwent next-generation whole-exome DNA sequencing, Affymetrix OncoScan SNP arrays, and targeted deep confirmatory sequencing. We performed mapping between SNPs and copy number events from the primary tumor and matched lymph node samples, allowing us to profile heterogeneity and the mutational origin of lymph node metastases. The computational method PyClone was used to define subclones within each tumor. The method Clonality Inference in Tumors Using Phylogeny (CITUP) was subsequently used to infer phylogenetic relationships among subclones.Results: We found that there was substantial heterogeneity in mutations and copy number changes among all samples from any given patient. For each patient, the primary tumor regions and matched lymph node metastases were each polyclonal, and the clonal populations differed from one lymph node to another. In some patients, the cancer cell populations in a given lymph node originated from multiple distinct regions of a tumor.Conclusions: Our data support a model of lymph node metastatic spread in colorectal cancer whereby metastases originate from multiple waves of seeding from the primary tumor over time. Clin Cancer Res; 24(9); 2214-24. ©2017 AACRSee related commentary by Gerlinger, p. 2032.


Sujet(s)
Évolution clonale , Tumeurs colorectales/étiologie , Tumeurs colorectales/anatomopathologie , Noeuds lymphatiques/anatomopathologie , Adulte , Sujet âgé , Cartographie chromosomique , Évolution clonale/génétique , Biologie informatique/méthodes , Variations de nombre de copies de segment d'ADN , Femelle , Analyse de profil d'expression de gènes , Hétérogénéité génétique , Séquençage nucléotidique à haut débit , Humains , Métastase lymphatique , Mâle , Adulte d'âge moyen , Modèles biologiques , Stadification tumorale , Phylogenèse , Polymorphisme de nucléotide simple , Transcriptome
20.
Oncotarget ; 8(55): 94330-94344, 2017 Nov 07.
Article de Anglais | MEDLINE | ID: mdl-29212231

RÉSUMÉ

ZBP-89 (Zfp148, ZNF148) is a Kruppel-type zinc-finger family transcription factor that binds to GC-rich DNA elements. Earlier studies in cell lines demonstrated that ZBP-89 cooperates with Wnt ß-catenin signaling by inducing ß-catenin gene expression. Since ß-catenin levels are normally highest at the crypt base, we examined whether ZBP-89 is required for stem cell maintenance. Lineage-tracing using a Zfp148CreERT2 transgenic line demonstrated expression in both intestine and colonic stem cells. Deleting the Zfp148 locus in the colon using the Cdx2NLSCreERT2 transgene, reduced the size and number of polyps formed in the Apc-deleted mice. Since colon polyps form in the presence of butyrate, a short chain fatty acid that suppresses cell growth, we examined the direct effect of butyrate on colon organoid survival. Butyrate induced senescence of colon organoids carrying the Apc deletion, only when Zfp148 was deleted. Using quantitative PCR and chromatin immunoprecipitation, we determined that butyrate treatment of colon cell lines suppressed ZNF148 gene expression, inducing CDKN2a (p16Ink4a ) gene expression. Collectively, Zfp148 mRNA is expressed in CBCs, and is required for stem cell maintenance and colonic transformation. Butyrate induces colonic cell senescence in part through suppression of ZBP-89 gene expression and its subsequent occupancy of the CDKN2A promoter.

SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...